Next Article in Journal
Effect of pH on the Stability of Dairy Beverages Stabilized with Soluble Soybean Polysaccharides
Previous Article in Journal
Soybean Soluble Polysaccharides: Composition, Structure, and Protein Stabilization Mechanism in Acidic Milk Drinks
Previous Article in Special Issue
Influence of Onion Peel Extract on the Dough Characteristics of High-Gluten Wheat Flour and the Quality of Bread
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synergistic Effects of Anthocyanin-Enriched Morus alba L. Extract and Vitamin C: Promising Nutraceutical Ingredients in Functional Food Development for Neuroprotection

1
Faculty of Medicine, Mahasarakham University, Mahasarakham 44000, Thailand
2
School of Biotechnology, Institute of Agricultu ral Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
3
State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
*
Author to whom correspondence should be addressed.
Foods 2025, 14(21), 3630; https://doi.org/10.3390/foods14213630 (registering DOI)
Submission received: 2 October 2025 / Revised: 16 October 2025 / Accepted: 23 October 2025 / Published: 24 October 2025

Abstract

Oxidative stress-induced mitochondrial dysfunction and apoptosis are critical factors in the pathogenesis of neurodegenerative diseases. This study investigated the synergistic neuroprotective effects of anthocyanin-enriched Morus alba L. extract combined with vitamin C (MAC) against hydrogen peroxide (H2O2)-induced oxidative stress in SH-SY5Y neuronal cells. Exposure to H2O2 triggered excessive reactive oxygen species (ROS) production and apoptosis, whereas treatment with MAC markedly alleviated these effects. Biochemical analyses revealed that MAC significantly reduced malondialdehyde (MDA) and enhanced the activities of antioxidant enzymes, including catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px), thereby contributing to improved redox balance. Furthermore, MAC modulated apoptosis-related signaling by upregulating extracellular signal-regulated kinase (ERK), cAMP response element-binding protein (CREB), and the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2), while downregulating the pro-apoptotic protein Bcl-2-associated X (BAX) and cleaved caspase-3. Collectively, these findings demonstrate that MAC acts synergistically as a promising nutraceutical ingredient, supporting the development of functional foods for the prevention or mitigation of oxidative stress-related neurodegenerative disorders.

1. Introduction

Neurodegenerative diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD), represent a growing global health concern due to their increasing prevalence in aging populations. As of 2023, more than 55 million people worldwide are living with dementia, with Alzheimer’s disease accounting for approximately 60–70% of cases, a number projected to reach 78 million by 2030 and 139 million by 2050 [1]. Similarly, Parkinson’s disease affects over 10 million people globally, with incidence rising rapidly in older adults [2]. These disorders are characterized by progressive neuronal loss, cognitive and/or motor decline, and reduced quality of life, placing a substantial socioeconomic burden on families, caregivers, and healthcare systems [3]. The rising prevalence, limited treatment options, and irreversible nature of neuronal damage underscore the urgent need for research into neuroprotective strategies that can mitigate disease progression or delay onset.
One of the key pathological mechanisms involved in neurodegeneration is oxidative stress, which leads to cellular injury through excessive production of ROS [4,5,6]. Oxidative stress disrupts redox homeostasis, damages cellular macromolecules, impairs mitochondrial function, and ultimately triggers apoptosis [7,8]. In particular, hydrogen peroxide (H2O2) is a major contributor to oxidative damage and is widely used as a model oxidant in neuronal cell studies [9,10,11]. The mitogen-activated protein kinase (MAPKs) signaling cascade, including extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), is activated in response to oxidative stress and modulates the balance between pro-apoptotic proteins such as Bcl-2-associated X protein (BAX) and anti-apoptotic proteins such as B-cell lymphoma 2 (Bcl-2), ultimately influencing mitochondrial integrity and caspase-3 activation [12,13,14]. Moreover, oxidative stress can suppress cAMP response element-binding protein (CREB), a key factor in neuronal survival and plasticity [15].
Given the critical role of oxidative stress and apoptosis in neurodegenerative diseases, natural compounds with antioxidant and anti-apoptotic properties have received considerable attention as potential neuroprotective agents [16,17]. Anthocyanins, a subclass of flavonoids found in various plant species, exhibit potent antioxidant, anti-inflammatory, and neuroprotective effects [18,19]. Morus alba L., commonly known as mulberry fruit, is particularly rich in anthocyanins and has demonstrated promising pharmacological activities, including free radical scavenging and protection against neuronal injury [11,20]. Vitamin C (ascorbic acid) was selected as a complementary compound because it functions as a water-soluble antioxidant capable of neutralizing ROS and supporting enzymatic antioxidant defenses [21,22]. In addition, it stabilizes anthocyanins against degradation and oxidation during first-pass metabolism, thereby enhancing their bioavailability. Moreover, physiological transport mechanisms enable vitamin C to reach the central nervous system, underscoring its relevance not only for neuroprotection in vitro and in vivo but also for potential clinical applications and practical use in functional foods and nutraceuticals [23]. Despite these complementary properties, the synergistic potential of anthocyanins and vitamin C in mitigating oxidative stress-induced neuronal injury remains largely unexplored, providing a clear rationale for the present study.
Therefore, the present study aimed to investigate the combined neuroprotective effects of anthocyanin-enriched Morus alba L. extract and vitamin C in SH-SY5Y human neuroblastoma cells exposed to H2O2-induced oxidative stress. We specifically examined their effects on oxidative biomarkers, endogenous antioxidant enzyme activities, and key apoptotic signaling pathways, including p-ERK, ERK, CREB, Bcl-2, BAX, and cleaved caspase-3. Our findings highlight the synergistic action of these natural food-derived compounds in protecting neuronal cells and support their potential application as functional food ingredients for the dietary management or prevention of oxidative stress-related neurodegenerative disorders.

2. Materials and Methods

2.1. Reagents and Chemicals

Morus alba L. (mulberry fruits var. Chiangmai) were obtained from the Queen Sirikit Department of Sericulture Center, located at 296 Moo 13, Ban That Subdistrict, Phen District, Udon Thani Province, Thailand (17.02411° N, 102.99020° E). A voucher specimen was deposited under the reference number 61,001 for authentication and future reference. To minimize variability of the plant matrix, all samples were collected from the same geographical location and harvested at a similar stage of ripeness and season. Vitamin C (ascorbic acid; C6H8O6; MW: 176.12; CAS No. 50-81-7), MTT reagent, dimethyl sulfoxide (DMSO), and the Immobilon® Forte Western HRP substrate were purchased from Merck KGaA (Darmstadt, Germany).
All cell culture reagents, including Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum (FBS), penicillin-streptomycin, non-essential amino acids (NEAA), CM-H2DCFDA (5-(and-6)-carboxy-2′,7′-dichlorofluorescein diacetate), and N-PER™ neuronal protein extraction buffer, were obtained from Thermo Fisher Scientific (Waltham, MA, USA).
For Western blotting, SDS-polyacrylamide gels, nitrocellulose membranes, and 0.1% Tween-20 were supplied by Bio-Rad (Hercules, CA, USA). All primary and secondary antibodies, including those specific to p-ERK, ERK, CREB, Bcl-2, BAX, cleaved caspase-3, and β-actin, were obtained from Cell Signaling Technology (Danvers, MA, USA).

2.2. Extraction of Anthocyanin-Enriched Morus alba L. Extract

The dried fruits of Morus alba L. were thoroughly cleaned and oven-dried at 60 °C for 48 h (Memmert GmbH, Schwabach, Germany) to prevent microbial growth and preserve heat-sensitive phytochemicals. The dried samples were then ground into a fine powder using a laboratory grinder. To obtain an anthocyanin-enriched extract, the powdered material was macerated in a 50% hydroalcoholic solution (ethanol/water, v/v) at room temperature for 72 h with occasional stirring. This solvent system was selected to enhance the extraction efficiency of polar phenolic compounds, particularly anthocyanins, while maintaining compound stability due to the mild temperature and absence of strong acids or bases.
After maceration, the mixture was centrifuged at 3000 rpm for 10 min to separate the solids, and the supernatant was filtered through Whatman No. 1 filter paper. The filtrate was concentrated using a rotary evaporator under reduced pressure at 40 °C (Heidolph Instruments GmbH, Schwabach, Germany) to remove ethanol. The remaining aqueous extract was then freeze-dried (lyophilized) to preserve bioactive compounds and obtain a stable powdered extract for further analysis. The extraction method was adapted from previously established protocols for anthocyanin isolation from pigmented plant sources [24].
The anthocyanin-enriched Morus alba L. extract used in this study was obtained from the same voucher specimen (No. 61001) as previously characterized in our earlier work [11,24]. Total anthocyanin content was determined using the pH differential spectrophotometric method, with absorbance measured at 520 and 700 nm in buffer systems at pH 1.0 and 4.5, and expressed as cyanidin-3-glucoside (C3G) equivalents. The extract contained a total anthocyanin content of 270.33 ± 4.19 µg C3G per mg of extract [11]. Consistent with previous chromatographic data, C3G was identified as the predominant anthocyanin, as confirmed by the HPLC profile obtained from the same botanical source at the Queen Sirikit Department of Sericulture Center, Udon Thani Province [11,24,25]. In addition, external studies have demonstrated that Morus alba L. typically exhibits high anthocyanin concentrations, with C3G consistently reported as the major component [26]. These findings support the consistency of our extract’s phytochemical characteristics with established anthocyanin profiles in Morus alba L. and reinforce its reliability for biological evaluation.

2.3. Determination of Optimal Ratio and Synergistic Effects of Morus alba L. Extract Combined with Vitamin C

Non-toxic concentrations of Morus alba L. extract and vitamin C were first determined using SH-SY5Y cell viability assays. Morus alba L. extract and vitamin C were tested at 0–1000 µg/mL, and concentrations just below their cytotoxic thresholds were selected for subsequent experiments: 31.25 µg/mL for Morus alba L. extract and 62.5 µg/mL for vitamin C, maintaining >80% cell viability.
To evaluate synergistic effects, the half-maximal effective concentrations (EC50) of Morus alba L. extract (21.30 µg/mL) and vitamin C (43.30 µg/mL) were determined and used as reference doses (1X). Combination treatments were prepared at 0.125X, 0.25X, 0.5X, 1X, and 2X relative to these EC50 values.
The interactions between Morus alba L. extract and vitamin C were analyzed using the SynergyFinder platform (version R-3.10.3.), based on the Loewe additivity model, suitable for compounds with overlapping mechanisms of action. The original dose–response data used as input for the analysis are provided in Supplementary Table S1. SynergyFinder generates a dose–response matrix and calculates quantitative synergy scores, which are visualized as 2D contour plots and 3D landscapes, allowing identification of concentrations that exhibit synergistic interactions.
To confirm the synergistic effect and select the optimal ratios, the Combination Index (CI) was calculated using the Chou–Talalay equation:
CI = [D1/Dx1] + [D2/Dx2]
where D1 and D2 are the doses of Morus alba L. extract and vitamin C in combination required to achieve a given effect, and Dx1 and Dx2 are the doses of the same agents alone required to achieve the same effect. CI values indicate the type of interaction:
CI < 1: synergism
CI = 1: additive effect
CI > 1: antagonism
Based on the synergy analysis and CI values, the 1X:1X and 1X:0.5X combinations (Morus alba L. extract: vitamin C) exhibited the strongest synergistic neuroprotective effects against H2O2-induced cytotoxicity. These combinations were designated as MAC1 (1X:1X) and MAC2 (1X:0.5X) for subsequent experiments.

2.4. Cell Culture and Treatment

Cell culture and treatment were performed as previously described in our established protocol. SH-SY5Y neuroblastoma cells (ATCC, CRL-2266) were maintained in DMEM supplemented with 10% fetal bovine serum, 1% penicillin-streptomycin, and 1% non-essential amino acids at 37 °C in a humidified incubator with 5% CO2. For experiments, cells were pretreated with varying concentrations of the combination for 24 h, followed by exposure to 200 µM H2O2 for 24 h with or without the test compounds. This concentration and exposure duration were selected based on preliminary dose–response experiments to induce moderate oxidative stress without excessive cell death, allowing evaluation of neuroprotective effects [11,27].

2.5. Cell Viability Assay

Cell viability was assessed using the MTT assay as previously reported [28]. Briefly, after treatment, MTT reagent was added to each well, and the plates were incubated to allow for formazan crystal formation. The crystals were then solubilized in DMSO, and absorbance was measured at 570 nm. Cell viability was expressed as a percentage relative to the control.

2.6. Protein Measurement

Total protein concentration in SH-SY5Y cell lysates was determined using the NanoDrop™ 2000/2000c spectrophotometer (Thermo Fisher Scientific). Briefly, cell lysates were appropriately diluted with buffer, and the absorbance at 280 nm was measured according to the manufacturer’s instructions. Each sample was analyzed in triplicate to ensure accuracy and reproducibility. Protein concentrations were calculated directly from the absorbance readings and used to normalize the results of lipid peroxidation, antioxidant enzyme activity assays, and Western blot analysis. NanoDrop was selected for its small sample requirement, rapid direct measurement, and compatibility with downstream assays.

2.7. Evaluation of Lipid Peroxidation

Lipid peroxidation was assessed by measuring MDA levels using a modified thiobarbituric acid reactive substances (TBARS) assay, as detailed in our previous study [29]. Briefly, cell lysates were processed, and the absorbance of the MDA–TBA complex was measured at 532 nm. Results were calculated based on a standard curve and expressed as nanograms of MDA per milligram of protein.

2.8. Evaluation of Intracellular Reactive Oxygen Species

Intracellular ROS levels were determined using the fluorescent probe CM-H2DCFDA, as described in our previous study [30]. Briefly, SH-SY5Y cells were treated with test compounds, incubated with CM-H2DCFDA, and then exposed to H2O2. Fluorescence intensity was measured at 488 nm excitation and 520 nm emission to assess ROS accumulation.

2.9. Assessment of Antioxidant Enzyme Activities

To evaluate cellular antioxidant defense mechanisms, the activities of CAT, SOD, and GSH-Px were measured in SH-SY5Y cell lysates, following methods previously described in our study [31].
CAT activity was assessed based on its ability to decompose H2O2, with the remaining H2O2 quantified via reaction with potassium permanganate and absorbance measured at 490 nm.
SOD activity was determined by its capacity to inhibit the reduction in cytochrome C in a xanthine–xanthine oxidase system, with absorbance monitored at 415 nm.
GSH-Px activity was measured through the rate of glutathione (GSH) oxidation in the presence of H2O2, followed by the reaction with DTNB, and absorbance was recorded at 412 nm.
Enzyme activities were calculated using standard curves prepared with purified enzymes and expressed as units per milligram of protein.

2.10. Western Blotting Analysis

Western blotting was performed to examine key proteins involved in apoptosis and survival pathways, including p-ERK, ERK, CREB, Bcl-2, BAX, and cleaved caspase-3, as described in detail in our previous study [32]. SH-SY5Y cells were lysed in protein extraction buffer, and total protein concentrations were determined. Equal amounts of protein were separated by SDS-PAGE, transferred to polyvinylidene fluoride (PVDF) membranes, and probed with specific primary antibodies. After incubation with HRP-conjugated secondary antibodies, protein bands were detected using chemiluminescence and quantified with Image Lab software. Protein expression levels were normalized to β-actin and expressed relative to the control group.

2.11. Statistical Analysis

Data were expressed as mean ± SEM. Group comparisons were analyzed using one-way ANOVA followed by Tukey’s post hoc test. Prior to ANOVA, the assumptions of normality and homogeneity of variance were evaluated using the Shapiro–Wilk test and Levene’s test, respectively. A p-value < 0.05 was considered statistically significant. All analyses were performed using SPSS Statistics v21.0 (IBM Corp., Armonk, NY, USA).

3. Results

3.1. Determination of Non-Toxic Doses of Morus alba L. Extract and Vitamin C

Figure 1 shows the effects of various concentrations of Morus alba L. extract and vitamin C (0–1000 µg/mL) on SH-SY5Y neuronal cell viability. Exposure to Morus alba L. extract significantly reduced cell viability at 62.5 µg/mL (p < 0.01 vs. control) and at higher concentrations. Similarly, vitamin C significantly decreased cell viability starting at 125 µg/mL (p < 0.001 vs. control) and at subsequent higher doses.
For subsequent experiments, concentrations just below the cytotoxic threshold were selected, specifically 31.25 µg/mL for Morus alba L. extract and 62.5 µg/mL for vitamin C. These doses maintained greater than 80% cell viability, indicating they are safe and suitable for treatment in SH-SY5Y cells.

3.2. Determination of EC50 Values for Morus alba L. Extract and Vitamin C

After identifying the optimal non-toxic concentrations of Morus alba L. extract (31.25 µg/mL) and vitamin C (62.5 µg/mL), we next determined the half-maximal effective concentration (EC50) of each compound in order to establish fixed ratios for combination treatment and evaluation of potential synergistic effects. Dose–response curves were generated using concentration ranges of 0–31.25 µg/mL for Morus alba L. extract and 0–62.5 µg/mL for vitamin C, with inhibitory activity assessed as percentage inhibition. As shown in Figure 2, the EC50 value of Morus alba L. extract was calculated to be 21.30 µg/mL (R2 = 0.994), while that of vitamin C was 43.30 µg/mL (R2 = 0.9948). These results provided the basis for selecting appropriate dose ratios to evaluate the synergistic neuroprotective effects of the combined treatment in subsequent experiments.

3.3. Evaluation of Synergistic Effects of Morus alba L. Extract and Vitamin C

To determine whether Morus alba L. extract and vitamin C act synergistically, combination treatments were analyzed using the SynergyFinder platform based on the Loewe additivity model, which is commonly applied to compounds with overlapping mechanisms of action. The EC50 values were 21.30 µg/mL for Morus alba L. extract and 43.30 µg/mL for vitamin C, which were used as the reference doses (1X). Additional concentrations were tested at 0.125X, 0.25X, 0.5X, 1X, and 2X relative to these EC50 values.
Figure 3A shows a two-dimensional synergy contour plot, while Figure 3B presents the corresponding three-dimensional landscape. The analysis yielded a Loewe synergy score of 22.488, indicating a strong synergistic interaction between Morus alba L. extract and vitamin C in protecting SH-SY5Y cells against H2O2-induced cytotoxicity. The highest synergy was observed around the concentrations corresponding to their respective EC50 values, demonstrating that the combined treatment provides greater protection than either compound alone.

3.4. Identification of Synergistic Dose Combinations of Morus alba L. Extract and Vitamin C

To identify the optimal synergistic doses of Morus alba L. extract and vitamin C, combination index (CI) values were calculated (Table 1), and the dose–response matrix was analyzed (Figure 4). The results showed that the 0.5X:0.5X, 1X:0.5X, and 1X:1X combinations exhibited synergistic interactions. Among these, the 1X:0.5X and 1X:1X combinations provided the highest protective effects, with values of 90.15 and 91.34, respectively.
Based on these findings, the 1X:0.5X and 1X:1X ratios were selected for further investigation of the combined neuroprotective effects of anthocyanin-enriched Morus alba L. extract and vitamin C (MAC) against H2O2-induced oxidative stress in SH-SY5Y cells. These combinations were designated as MAC1 (1X:1X) and MAC2 (1X:0.5X), as both CI values and the dose–response matrix consistently confirmed their strong synergistic efficacy.

3.5. Determination of Neuroprotective Effects of MAC

Following the identification of the two optimal ratios of anthocyanin-enriched Morus alba L. extract and vitamin C (MAC), their neuroprotective effects were evaluated. Figure 5A shows the morphological changes in SH-SY5Y neuronal cells under different treatment conditions. Cells exposed to H2O2 with vehicle exhibited a marked reduction in density, along with evident morphological damage, including cell shrinkage, shortened neurites, nuclear condensation, and the formation of apoptotic bodies. These alterations were accompanied by a significant decrease in cell viability (p < 0.001 vs. control group).
In contrast, cells treated with H2O2 in combination with either MAC1 or MAC2 displayed markedly preserved morphology and significantly higher viability compared with the H2O2 + vehicle group (all p < 0.001). These findings indicate that both MAC1 and MAC2 effectively attenuate H2O2-induced cytotoxicity in SH-SY5Y neuronal cells.

3.6. Effects of MAC on ERK/CREB Signaling Pathway

To explore the possible underlying mechanism of the neuroprotective effects of MAC, we focused on the ERK/CREB signaling pathway, which plays a critical role in neuronal survival and plasticity. The results presented in Figure 6 demonstrate that exposure to H2O2 with vehicle significantly reduced ERK expression (Figure 6B) and CREB expression (Figure 6C) compared with the control group (p < 0.01 and p < 0.001, respectively). In contrast, treatment with MAC1 and MAC2 markedly increased ERK levels (all p < 0.01 vs. H2O2 + vehicle) and CREB levels (all p < 0.001 vs. H2O2 + vehicle).
These findings suggest that MAC protects SH-SY5Y neuronal cells, at least in part, through activation of the ERK/CREB signaling pathway.

3.7. Effect of MAC on Bcl-2 and BAX Expression

Bcl-2 and BAX are key regulators of apoptosis, where Bcl-2 acts as an anti-apoptotic protein and BAX functions as a pro-apoptotic counterpart. The ERK/CREB signaling pathway has been reported to modulate the expression of these apoptotic regulators, thereby influencing neuronal survival. As shown in Figure 7, exposure to H2O2 with vehicle significantly reduced Bcl-2 expression (Figure 7B) while markedly increasing BAX expression (Figure 7C) compared with the control group (all p < 0.001). In contrast, treatment with MAC1 markedly upregulated Bcl-2 and downregulated BAX levels (all p < 0.001 vs. H2O2 + vehicle). Similarly, MAC2 also elevated Bcl-2 expression (p < 0.05 vs. H2O2 + vehicle) and suppressed BAX expression (p < 0.001 vs. H2O2 + vehicle), suggesting that MAC attenuates apoptosis through the regulation of Bcl-2 and BAX, potentially mediated by ERK/CREB signaling.
In addition to individual expression changes, the Bcl-2/BAX ratio was evaluated as an indicator of cellular susceptibility to apoptosis. As shown in Figure 8, the control group exhibited a ratio of 1.61 ± 0.05, consistent with a pro-survival profile. Exposure to H2O2 with vehicle treatment significantly reduced this ratio to 0.65 ± 0.03 (p < 0.001 vs. control), indicating a strong shift toward pro-apoptotic signaling. Treatment with MAC1 restored the ratio to 1.23 ± 0.07 (p < 0.001 vs. H2O2 + vehicle), while MAC2 increased it to 1.04 ± 0.05 (p < 0.05 vs. H2O2 + vehicle). Although both treatments improved the ratio compared with H2O2 alone, MAC1 exerted a more pronounced effect, suggesting greater efficacy in re-establishing the balance between pro- and anti-apoptotic proteins.

3.8. Effect of MAC on Cleaved Caspase-3 Expression

Cleaved caspase-3 is a critical executioner in the apoptotic pathway, activated downstream of mitochondrial regulators such as Bcl-2 and BAX. As illustrated in Figure 9, exposure to H2O2 with vehicle markedly increased cleaved caspase-3 expression compared with the control group (p < 0.001). In contrast, treatment with both MAC1 and MAC2 significantly reduced cleaved caspase-3 levels (all p < 0.05 vs. H2O2 + vehicle). These results suggest that MAC attenuates H2O2-induced apoptotic cell death by suppressing caspase-3 activation, further reinforcing its anti-apoptotic effects through modulation of the ERK/CREB signaling pathway and regulation of the Bcl-2/BAX balance.

3.9. Effect of MAC on Antioxidant Enzyme Activities

Oxidative stress plays a central role in triggering neuronal apoptosis, in part through activation of signaling pathways such as ERK/CREB and modulation of mitochondrial proteins including Bcl-2, BAX, and cleaved caspase-3. To further clarify the antioxidant contribution of MAC, the activities of CAT, SOD, and GSH-Px were evaluated (Figure 10). Exposure to H2O2 with vehicle significantly decreased the activities of CAT, SOD, and GSH-Px compared with the control group (p < 0.001, p < 0.001, and p < 0.01, respectively). In contrast, treatment with MAC1 markedly restored CAT, SOD, and GSH-Px activities (p < 0.01, p < 0.001, and p < 0.05 vs. H2O2 + vehicle, respectively). Similarly, MAC2 also enhanced CAT, SOD, and GSH-Px activities (p < 0.01, p < 0.001, and p < 0.05 vs. H2O2 + vehicle, respectively).
These findings indicate that MAC protects neuronal cells not only by inhibiting apoptosis but also by reinforcing the endogenous antioxidant defense system, thereby reducing oxidative stress and preserving neuronal integrity under H2O2-induced injury.

3.10. Effect of MAC on Intracellular ROS Production

Since excessive accumulation of ROS is a major driver of oxidative stress and downstream apoptotic signaling, we next assessed intracellular ROS levels in SH-SY5Y neuronal cells (Figure 11). Cells exposed to H2O2 with vehicle showed a marked increase in ROS generation compared with the control group (p < 0.001). Treatment with both MAC1 and MAC2 significantly attenuated ROS accumulation (all p < 0.001 vs. H2O2 + vehicle).
These results are consistent with the enhanced activities of antioxidant enzymes observed earlier, supporting the notion that MAC alleviates oxidative damage by reducing ROS overproduction, thereby contributing to its neuroprotective effects against H2O2-induced cytotoxicity.

3.11. Effect of MAC on Lipid Peroxidation (MDA Levels)

To further evaluate oxidative damage, we measured MDA, a marker of lipid peroxidation (Figure 12). Exposure to H2O2 with vehicle resulted in a significant elevation of MDA levels compared with the control group (p < 0.001). In contrast, treatment with either MAC1 or MAC2 markedly reduced MDA accumulation (all p < 0.001 vs. H2O2 + vehicle).
These findings, together with the observed reductions in ROS and enhancements in antioxidant enzyme activities, indicate that MAC effectively mitigates oxidative damage by limiting lipid peroxidation and restoring redox homeostasis in SH-SY5Y neuronal cells.

4. Discussion

Oxidative stress is one of the most important contributors to neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease, where oxidative stress-induced mitochondrial dysfunction and dysregulated apoptotic signaling converge to drive progressive neuronal loss [33,34]. Our model using H2O2-induced oxidative stress is a classical paradigm that reproduces these mechanisms. In SH-SY5Y cells, H2O2 exposure led to excessive ROS accumulation, elevated lipid peroxidation, and suppression of endogenous antioxidant defenses, accompanied by activation of apoptotic pathways through Bcl-2/BAX imbalance, cytochrome c release, and cleaved caspase-3, together with dysregulation of the ERK/CREB signaling pathway. These results are in line with previous studies demonstrating that oxidative stress simultaneously promotes mitochondrial apoptosis and inhibits survival signaling, thereby accelerating neuronal injury.
H2O2-induced ROS accumulation caused mitochondrial dysfunction by disrupting the mitochondrial membrane potential and altering the balance of pro- and anti-apoptotic proteins [35]. Under physiological conditions, a high Bcl-2/BAX ratio stabilizes the mitochondrial outer membrane, preventing permeabilization. Conversely, when BAX exceeds Bcl-2, the membrane becomes destabilized, leading to the release of cytochrome c into the cytosol. Once released, cytochrome c binds to apoptotic protease-activating factor-1 (Apaf-1), promoting apoptosome formation. This multiprotein complex recruits and activates caspase-9, which in turn cleaves and activates executioner caspase-3. Activated caspase-3 initiates proteolytic degradation of structural and regulatory proteins, culminating in the execution phase of apoptosis [36,37]. Our results demonstrated that MAC treatment suppressed this cascade by restoring the Bcl-2/BAX ratio, preventing cytochrome c release, and reducing caspase-3 activation. These findings are consistent with established models of mitochondrial apoptosis in neurodegeneration.
In addition to apoptosis, ROS accumulation disrupted the ERK/CREB signaling pathway, which plays a pivotal role in synaptic plasticity and neuronal resilience [38,39,40]. Normally, ERK activation phosphorylates CREB, enabling transcription of anti-apoptotic and survival-promoting genes, including Bcl-2 [41]. In our model, H2O2 exposure suppressed ERK/CREB activity, leading to reduced Bcl-2 expression and increased susceptibility to apoptosis. The downregulation of CREB impairs transcription of anti-apoptotic genes, tipping the balance toward pro-apoptotic signaling. In this context, the decrease in Bcl-2 further destabilizes mitochondrial integrity, amplifying the apoptotic cascade [41,42]. Our results showed that MAC treatment reactivated ERK/CREB signaling, enhanced Bcl-2 expression, and thereby interrupted the feed-forward cycle of ROS-induced apoptosis. These observations support the idea that modulation of ERK/CREB is a critical mechanism by which MAC confers neuroprotection.
H2O2-induced ROS also promoted lipid peroxidation, a critical process in neurodegeneration [43,44]. Peroxidized lipids not only compromise membrane integrity but also generate toxic aldehydes such as MDA, which propagate oxidative damage by amplifying protein misfolding, DNA damage, and mitochondrial injury [45,46]. These effects further exacerbate mitochondrial apoptosis, as described above. Moreover, oxidative stress impaired enzymatic antioxidant defenses, as indicated by decreased activities of CAT, SOD, and GSH-Px, which accelerated lipid peroxidation and redox imbalance [43,47,48]. Our results revealed that MAC treatment significantly enhanced these endogenous antioxidant enzymes, reduced lipid peroxidation, and restored redox homeostasis. By reinforcing antioxidant defenses, MAC indirectly protected mitochondrial membranes, thereby limiting apoptotic activation and supporting neuronal survival.
Interestingly, the results showed that anthocyanin-enriched Morus alba L. extract and vitamin C exhibit strong synergism, as confirmed by both the combination index and the Loewe additivity model, which quantitatively demonstrated that the combined effect exceeded the predicted additive effect of each agent alone. Our results further indicate that the equimolar combination MAC1 (1X:1X) provides stronger neuroprotective effects than the sub-equimolar MAC2 (1X:0.5X), reflecting key mechanistic differences between the two ratios [11,27,49,50]. MAC1 maintains an optimal balance between anthocyanins and vitamin C, allowing effective regeneration of oxidized anthocyanins by vitamin C and stabilization of vitamin C by anthocyanins, thereby sustaining the redox cycle [51]. In contrast, the lower vitamin C concentration in MAC2 may be insufficient to fully regenerate anthocyanins, reducing ROS scavenging, antioxidant enzyme activation, and apoptosis suppression. Furthermore, anthocyanins from Morus alba L. are structurally diverse molecules capable of scavenging multiple ROS, chelating transition metals, and interacting with membranes, but their maximal efficacy depends on adequate vitamin C levels. Under physiological conditions, the stability and bioavailability of both anthocyanins and vitamin C are limited by factors such as pH, metabolism, and enzymatic degradation, which further supports the use of equimolar combinations in future in vivo or clinical studies [52,53,54,55].
In a translational context, our findings indicate that anthocyanin-enriched Morus alba L. extract combined with vitamin C has strong synergistic potential, supporting its application in functional food formulations or dietary supplements aimed at protecting neurons from oxidative stress-related neurodegeneration and age-associated damage. Future studies should validate these effects in primary neuronal cultures and in vivo models, clarify pharmacokinetic properties and bioavailability, and evaluate interactions with other redox-sensitive pathways. Collectively, this work provides a foundation for developing next-generation functional foods that target oxidative stress, apoptosis, and mitochondrial dysfunction to preserve neuronal function and support overall brain health.

5. Conclusions

In conclusion, the combination of anthocyanin-enriched Morus alba L. extract and vitamin C (MAC) exerts potent synergistic neuroprotective effects against H2O2-induced oxidative stress in SH-SY5Y neuronal cells. MAC effectively reduces ROS accumulation, limits lipid peroxidation, enhances endogenous antioxidant enzyme activities, and suppresses apoptosis through modulation of the ERK/CREB signaling pathway and Bcl-2/BAX-caspase-3 axis, as shown in Figure 13. The superior efficacy of the MAC1 (1X:1X) ratio highlights the importance of optimal stoichiometric combinations to maximize complementary mechanisms of action. These findings support the concept that multi-targeted interventions combining dietary polyphenols and classical antioxidants can provide enhanced neuronal protection compared with single-agent approaches. Overall, this study identifies MAC as a promising nutraceutical candidate, paving the way for the development of functional foods aimed at preventing or mitigating oxidative stress-related neurodegenerative disorders.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/foods14213630/s1. Figure S1: Western blot analysis of p-ERK expression; Figure S2: Western blot analysis of ERK expression; Figure S3: Western blot analysis of CREB expression; Figure S4: Western blot analysis of Bcl-2 expression; Figure S5: Western blot analysis of BAX expression; Figure S6: Western blot analysis of cleaved caspase-3 expression; Figures S7–S9: Western blot analysis of β-actin expression; Table S1: Original input data for dose–response and synergy analysis of anthocyanin-enriched Morus alba L. extract and vitamin C.

Author Contributions

Conceptualization, N.P., N.M., J.J., K.A., P.N., G.B. and Y.H.; Methodology, N.P. and N.M.; Validation, N.P.; Formal Analysis, N.P.; Investigation, N.P.; Resources, N.P., N.M. and P.N.; Data Curation, N.P.; Writing—Original Draft Preparation, N.P.; Writing—Review and Editing, N.P., N.M., J.J., K.A., G.B. and Y.H.; Visualization, N.P.; Supervision, N.P.; Project Administration, N.P.; Funding Acquisition, N.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research project was financially supported by Thailand Science Research and Innovation (TSRI).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed in this study are included in the article and its Supplementary Material. Further information can be obtained from the corresponding authors upon request.

Acknowledgments

The authors gratefully acknowledge the financial support provided by (i) Thailand Science Research and Innovation (TSRI), Thailand, and (ii) Mahasarakham University (MSU), Thailand. The authors also wish to express their appreciation to the Faculty of Medicine, Mahasarakham University, Thailand, for providing the facilities and resources essential to this research, and to Nankai University, China, for their valuable collaboration.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. World Health Organization. Global Status Report on the Public Health Response to Dementia; World Health Organization: Geneva, Switzerland, 2021; Available online: https://www.who.int/publications/i/item/9789240033245 (accessed on 17 July 2025).
  2. Dorsey, E.R.; Bloem, B.R. The Parkinson Pandemic—A Call to Action. JAMA Neurol. 2018, 75, 9–10. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, S.; Jiang, Y.; Yang, A.; Meng, F.; Zhang, J. The Expanding Burden of Neurodegenerative Diseases: An Unmet Medical and Social Need. Aging Dis. 2024, 16, 2937–2952. [Google Scholar] [CrossRef]
  4. Manoharan, S.; Guillemin, G.J.; Abiramasundari, R.S.; Essa, M.M.; Akbar, M.; Akbar, M.D. The Role of Reactive Oxygen Species in the Pathogenesis of Alzheimer’s Disease, Parkinson’s Disease, and Huntington’s Disease: A Mini Review. Oxid. Med. Cell. Longev. 2016, 2016, 8590578. [Google Scholar] [CrossRef]
  5. Chong, Z.Z.; Souayah, N. Oxidative Stress: Pathological Driver in Chronic Neurodegenerative Diseases. Antioxidants 2025, 14, 696. [Google Scholar] [CrossRef]
  6. Li, J.; O, W.; Li, W.; Jiang, Z.-G.; Ghanbari, H.A. Oxidative Stress and Neurodegenerative Disorders. Int. J. Mol. Sci. 2013, 14, 24438–24475. [Google Scholar] [CrossRef]
  7. Fields, M.; Marcuzzi, A.; Gonelli, A.; Celeghini, C.; Maximova, N.; Rimondi, E. Mitochondria-Targeted Antioxidants, an Innovative Class of Antioxidant Compounds for Neurodegenerative Diseases: Perspectives and Limitations. Int. J. Mol. Sci. 2023, 24, 3739. [Google Scholar] [CrossRef]
  8. Guo, C.; Sun, L.; Chen, X.; Zhang, D. Oxidative Stress, Mitochondrial Damage and Neurodegenerative Diseases. Neural Regen. Res. 2013, 8, 2003–2014. [Google Scholar] [CrossRef]
  9. Zhang, H.; Li, Y.; Xun, Y.; Liu, H.; Wei, C.; Wang, H.; Yang, X.; Yuan, S.; Liu, N.; Xiang, S. Polydatin Protects Neuronal Cells from Hydrogen Peroxide Damage by Activating CREB/Ngb Signaling. Mol. Med. Rep. 2022, 25, 9. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, Q.; Yu, M.; Tian, Z.; Cui, Y.; Deng, D.; Rong, T.; Liu, Z.; Song, M.; Li, Z.; Ma, X.; et al. Exogenous Glutathione Protects IPEC-J2 Cells against Oxidative Stress through a Mitochondrial Mechanism. Molecules 2022, 27, 2416. [Google Scholar] [CrossRef] [PubMed]
  11. Mairuae, N.; Palachai, N.; Noisa, P. The Neuroprotective Effects of the Combined Extract of Mulberry Fruit and Mulberry Leaf against Hydrogen Peroxide-Induced Cytotoxicity in SH-SY5Y Cells. BMC Complement. Med. Ther. 2023, 23, 117. [Google Scholar] [CrossRef]
  12. Yue, J.; López, J.M. Understanding MAPK Signaling Pathways in Apoptosis. Int. J. Mol. Sci. 2020, 21, 2346. [Google Scholar] [CrossRef]
  13. Lee, K.I.; Fang, K.M.; Kuo, C.Y.; Huang, C.F.; Liu, S.H.; Liu, J.M.; Lai, W.C.; Chang, K.C.; Su, C.C.; Chen, Y.W. Roles of Oxidative Stress/JNK/ERK Signals in Paraquat-Triggered Hepatic Apoptosis. Curr. Res. Toxicol. 2024, 6, 100155. [Google Scholar] [CrossRef]
  14. Hollville, E.; Romero, S.E.; Deshmukh, M. Apoptotic Cell Death Regulation in Neurons. FEBS J. 2019, 286, 3276–3298. [Google Scholar] [CrossRef]
  15. Wang, L.; Nie, Q.; Gao, M.; Yang, L.; Xiang, J.W.; Xiao, Y.; Liu, F.Y.; Gong, X.D.; Fu, J.L.; Wang, Y.; et al. The Transcription Factor CREB Acts as an Important Regulator Mediating Oxidative Stress-Induced Apoptosis by Suppressing αB-Crystallin Expression. Aging 2020, 12, 13594–13617. [Google Scholar] [CrossRef]
  16. Ullah, R.; Khan, M.; Shah, S.A.; Saeed, K.; Kim, M.O. Natural Antioxidant Anthocyanins—A Hidden Therapeutic Candidate in Metabolic Disorders with Major Focus in Neurodegeneration. Nutrients 2019, 11, 1195. [Google Scholar] [CrossRef]
  17. Lee, J.Y.; Wong, C.Y.; Koh, R.Y.; Lim, C.L.; Kok, Y.Y.; Chye, S.M. Natural Bioactive Compounds from Macroalgae and Microalgae for the Treatment of Alzheimer’s Disease: A Review. Yale J. Biol. Med. 2024, 97, 205–224. [Google Scholar] [CrossRef]
  18. Zaa, C.A.; Marcelo, Á.J.; An, Z.; Medina-Franco, J.L.; Velasco-Velázquez, M.A. Anthocyanins: Molecular Aspects on Their Neuroprotective Activity. Biomolecules 2023, 13, 1598. [Google Scholar] [CrossRef]
  19. Rębas, E. Role of Flavonoids in Protecting against Neurodegenerative Diseases—Possible Mechanisms of Action. Int. J. Mol. Sci. 2025, 26, 4763. [Google Scholar] [CrossRef]
  20. Sabuj, M.S.S.; Islam, M.S.; Akanda, M.R.; Lee, J.; Kim, R.; Lee, S.H.; Kim, Y.J.; Oh, J.M.; Ro, H.G.; Kim, I.S.; et al. Macluraparishin C Enhances Neuroprotection against Oxidative Stress-Induced Neurodegeneration by Activating the Antioxidant/MAPK Signaling Pathway. Mol. Neurobiol. 2025, 62, 13736–13747. [Google Scholar] [CrossRef]
  21. Lee, K.H.; Cha, M.; Lee, B.H. Neuroprotective Effect of Antioxidants in the Brain. Int. J. Mol. Sci. 2020, 21, 7152. [Google Scholar] [CrossRef]
  22. Covarrubias-Pinto, A.; Acuña, A.I.; Beltrán, F.A.; Torres-Díaz, L.; Castro, M.A. Old Things New View: Ascorbic Acid Protects the Brain in Neurodegenerative Disorders. Int. J. Mol. Sci. 2015, 16, 28194–28217. [Google Scholar] [CrossRef]
  23. Harrison, F.E.; May, J.M. Vitamin C Function in the Brain: Vital Role of the Ascorbate Transporter SVCT2. Free Radic. Biol. Med. 2009, 46, 719–730. [Google Scholar] [CrossRef]
  24. Palachai, N.; Wattanathorn, J.; Muchimapura, S.; Thukham-Mee, W. Antimetabolic Syndrome Effect of Phytosome Containing the Combined Extracts of Mulberry and Ginger in an Animal Model of Metabolic Syndrome. Oxid. Med. Cell. Longev. 2019, 2019, 5972575. [Google Scholar] [CrossRef]
  25. Wattanathorn, J.; Kawvised, S.; Thukham-Mee, W. Encapsulated Mulberry Fruit Extract Alleviates Changes in an Animal Model of Menopause with Metabolic Syndrome. Oxid. Med. Cell. Longev. 2019, 2019, 5360560. [Google Scholar] [CrossRef]
  26. Chen, T.; Shuang, F.-F.; Fu, Q.-Y.; Ju, Y.-X.; Zong, C.-M.; Zhao, W.-G.; Zhang, D.-Y.; Yao, X.-H.; Cao, F.-L. Evaluation of the Chemical Composition and Antioxidant Activity of Mulberry (Morus alba L.) Fruits from Different Varieties in China. Molecules 2022, 27, 2688. [Google Scholar] [CrossRef]
  27. Khongrum, J.; Mairuae, N.; Thanchomnang, T.; Zhang, M.; Bai, G.; Palachai, N. Synergistic Neuroprotection through Epigenetic Modulation by Combined Curcumin-Enriched Turmeric Extract and L-Ascorbic Acid in Oxidative Stress-Induced SH-SY5Y Cell Damage. Foods 2025, 14, 892. [Google Scholar] [CrossRef]
  28. Mairuae, N.; Noisa, P.; Palachai, N. Phytosome-Encapsulated 6-Gingerol- and 6-Shogaol-Enriched Extracts from Zingiber officinale Roscoe Protect against Oxidative Stress-Induced Neurotoxicity. Molecules 2024, 29, 6046. [Google Scholar] [CrossRef]
  29. Wattanathorn, J.; Palachai, N.; Thukham-Mee, W.; Muchimapura, S. Memory-Enhancing Effect of a Phytosome Containing the Combined Extract of Mulberry Fruit and Ginger in an Animal Model of Ischemic Stroke with Metabolic Syndrome. Oxid. Med. Cell. Longev. 2020, 2020, 3096826. [Google Scholar] [CrossRef]
  30. Mairuae, N.; Palachai, N.; Noisa, P. An Anthocyanin-Rich Extract from Zea mays L. var. ceratina Alleviates Neuronal Cell Death Caused by Hydrogen Peroxide-Induced Cytotoxicity in SH-SY5Y Cells. BMC Complement. Med. Ther. 2024, 24, 162. [Google Scholar] [CrossRef]
  31. Mairuae, N.; Palachai, N. Synergistic Antioxidant Effects of C3G-Enriched Oryza sativa L. cv. RD83 Extract and α-Tocopherol against H2O2-Induced Oxidative Stress in SH-SY5Y Cells. Int. J. Mol. Sci. 2025, 26, 6490. [Google Scholar] [CrossRef]
  32. Palachai, N.; Supawat, A.; Kongsui, R.; Klimaschewski, L.; Jittiwat, J. Galangin’s Neuroprotective Role: Targeting Oxidative Stress, Inflammation, and Apoptosis in Ischemic Stroke in a Rat Model of Permanent Middle Cerebral Artery Occlusion. Int. J. Mol. Sci. 2025, 26, 1847. [Google Scholar] [CrossRef]
  33. Yan, M.H.; Wang, X.; Zhu, X. Mitochondrial Defects and Oxidative Stress in Alzheimer Disease and Parkinson Disease. Free Radic. Biol. Med. 2013, 62, 90–101. [Google Scholar] [CrossRef]
  34. urcău, M.C.; Andronie-Cioara, F.L.; Jurcău, A.; Marcu, F.; Ţiț, D.M.; Pașcalău, N.; Nistor-Cseppentö, D.C. The Link between Oxidative Stress, Mitochondrial Dysfunction and Neuroinflammation in the Pathophysiology of Alzheimer’s Disease: Therapeutic Implications and Future Perspectives. Antioxidants 2022, 11, 2167. [Google Scholar] [CrossRef]
  35. Vlasova, Y.A.; Zakharova, I.O.; Avrova, N.F. The Effects of Alpha-Tocopherol and H2O2 on the Mitochondrial Membrane Potential and Bax/Bcl-xL Ratio in PC12 Cells. Neurochem. J. 2016, 10, 318–322. [Google Scholar] [CrossRef]
  36. Lin, T.-K.; Cheng, C.-H.; Chen, S.-D.; Liou, C.-W.; Huang, C.-R.; Chuang, Y.-C. Mitochondrial Dysfunction and Oxidative Stress Promote Apoptotic Cell Death in the Striatum via Cytochrome c/Caspase-3 Signaling Cascade Following Chronic Rotenone Intoxication in Rats. Int. J. Mol. Sci. 2012, 13, 8722–8739. [Google Scholar] [CrossRef]
  37. Zhang, L.; Liu, Y.; Li, J.Y.; Li, L.Z.; Zhang, Y.L.; Gong, H.Y.; Cui, Y. Protective Effect of Rosamultin against H2O2-Induced Oxidative Stress and Apoptosis in H9c2 Cardiomyocytes. Oxid. Med. Cell. Longev. 2018, 2018, 8415610. [Google Scholar] [CrossRef]
  38. Massaad, C.A.; Klann, E. Reactive Oxygen Species in the Regulation of Synaptic Plasticity and Memory. Antioxid. Redox Signal. 2011, 14, 2013–2054. [Google Scholar] [CrossRef]
  39. Khezri, M.R.; Yousefi, K.; Esmaeili, A.; Ghasemnejad-Berenji, M. The Role of ERK1/2 Pathway in the Pathophysiology of Alzheimer’s Disease: An Overview and Update on New Developments. Cell. Mol. Neurobiol. 2023, 43, 177–191. [Google Scholar] [CrossRef]
  40. Kim, S.K.; Kwon, Y.J.; Seo, E.B.; Lee, H.S.; Sohn, J.O.; Shin, H.M.; Kim, S.J.; Ye, S.K. Neuroprotective Effects of STAT3 Inhibitor on Hydrogen Peroxide-Induced Neuronal Cell Death via the ERK/CREB Signaling Pathway. Neurochem. Res. 2024, 50, 52. [Google Scholar] [CrossRef] [PubMed]
  41. Fu, X.; Feng, Y.; Shao, B.; Zhang, Y. Activation of the ERK/CREB/Bcl-2 Pathway Protects Periodontal Ligament Stem Cells against Hydrogen Peroxide-Induced Oxidative Stress. Mol. Med. Rep. 2019, 19, 3649–3657. [Google Scholar] [CrossRef]
  42. Guo, K.M.; Li, W.; Wang, Z.H.; He, L.C.; Feng, Y.; Liu, H.S. Low-Dose Aspirin Inhibits Trophoblast Cell Apoptosis by Activating the CREB/Bcl-2 Pathway in Pre-Eclampsia. Cell Cycle 2022, 21, 2223–2238. [Google Scholar] [CrossRef]
  43. Olufunmilayo, E.O.; Gerke-Duncan, M.B.; Holsinger, R.M.D. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants 2023, 12, 517. [Google Scholar] [CrossRef] [PubMed]
  44. Shichiri, M. The Role of Lipid Peroxidation in Neurological Disorders. J. Clin. Biochem. Nutr. 2014, 54, 151–160. [Google Scholar] [CrossRef]
  45. Allowitz, K.; Taylor, J.; Harames, K.; Yoo, J.; Baloch, O.; Ramana, K.V. Oxidative Stress-Mediated Lipid Peroxidation-Derived Lipid Aldehydes in the Pathophysiology of Neurodegenerative Diseases. Curr. Neuropharmacol. 2025, 23, 671–685. [Google Scholar] [CrossRef]
  46. Cheng, J.; Wang, F.; Yu, D.F.; Wu, P.F.; Chen, J.G. The Cytotoxic Mechanism of Malondialdehyde and Protective Effect of Carnosine via Protein Cross-Linking/Mitochondrial Dysfunction/Reactive Oxygen Species/MAPK Pathway in Neurons. Eur. J. Pharmacol. 2011, 650, 184–194. [Google Scholar] [CrossRef]
  47. Chen, Z.; Xing, T.; Li, J.; Zhang, L.; Jiang, Y.; Gao, F. Hydrogen Peroxide-Induced Oxidative Stress Impairs Redox Status and Damages Aerobic Metabolism of Breast Muscle in Broilers. Poult. Sci. 2021, 100, 918–925. [Google Scholar] [CrossRef]
  48. Afzal, S.; Abdul Manap, A.S.; Attiq, A.; Albokhadaim, I.; Kandeel, M.; Alhojaily, S.M. From Imbalance to Impairment: The Central Role of Reactive Oxygen Species in Oxidative Stress-Induced Disorders and Therapeutic Exploration. Front. Pharmacol. 2023, 14, 1269581. [Google Scholar] [CrossRef]
  49. García-Fuente, A.; Vázquez, F.; Viéitez, J.M.; Alonso, F.J.G.; Martín, J.I.; Ferrer, J. CISNE: An Accurate Description of Dose-Effect and Synergism in Combination Therapies. Sci. Rep. 2018, 8, 4964. [Google Scholar] [CrossRef] [PubMed]
  50. Caesar, L.K.; Cech, N.B. Synergy and Antagonism in Natural Product Extracts: When 1 + 1 Does Not Equal 2. Nat. Prod. Rep. 2019, 36, 869–888. [Google Scholar] [CrossRef] [PubMed]
  51. Enaru, B.; Drețcanu, G.; Pop, T.D.; Stǎnilǎ, A.; Diaconeasa, Z. Anthocyanins: Factors Affecting Their Stability and Degradation. Antioxidants 2021, 10, 1967. [Google Scholar] [CrossRef]
  52. Sadowska-Bartosz, I.; Bartosz, G. Antioxidant Activity of Anthocyanins and Anthocyanidins: A Critical Review. Int. J. Mol. Sci. 2024, 25, 12001. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, L.; Wang, Y.; Cao, Y.; Wang, F.; Li, F. Review: Enhancing the Bioavailability and Stability of Anthocyanins for the Prevention and Treatment of Central Nervous System-Related Diseases. Foods 2025, 14, 2420. [Google Scholar] [CrossRef] [PubMed]
  54. Leong, S.Y.; Burritt, D.J.; Hocquel, A.; Penberthy, A.; Oey, I. The Relationship between the Anthocyanin and Vitamin C Contents of Red-Fleshed Sweet Cherries and the Ability of Fruit Digests to Reduce Hydrogen Peroxide-Induced Oxidative Stress in Caco-2 Cells. Food Chem. 2017, 227, 404–412. [Google Scholar] [CrossRef]
  55. Xue, H.; Zhao, J.; Wang, Y.; Shi, Z.; Xie, K.; Liao, X.; Tan, J. Factors Affecting the Stability of Anthocyanins and Strategies for Improving Their Stability: A Review. Food Chem. X 2024, 24, 101883. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of Morus alba L. extract and vitamin C on SH-SY5Y cell viability. Cells were treated with 0–1000 µg/mL of each compound for 24 h. Data are presented as mean ± SEM (n = 3). For Morus alba L. extract, ** p < 0.01 and *** p < 0.001 vs. control (0 µg/mL); for vitamin C, ### p < 0.001 vs. control (0 µg/mL).
Figure 1. Effect of Morus alba L. extract and vitamin C on SH-SY5Y cell viability. Cells were treated with 0–1000 µg/mL of each compound for 24 h. Data are presented as mean ± SEM (n = 3). For Morus alba L. extract, ** p < 0.01 and *** p < 0.001 vs. control (0 µg/mL); for vitamin C, ### p < 0.001 vs. control (0 µg/mL).
Foods 14 03630 g001
Figure 2. EC50 values for Morus alba L. extract and vitamin C. (A) Dose–response curve generated using 0–31.25 µg/mL for Morus alba L. extract. (B) Dose–response curve generated using 0–62.5 µg/mL for vitamin C.
Figure 2. EC50 values for Morus alba L. extract and vitamin C. (A) Dose–response curve generated using 0–31.25 µg/mL for Morus alba L. extract. (B) Dose–response curve generated using 0–62.5 µg/mL for vitamin C.
Foods 14 03630 g002
Figure 3. Synergistic effects of Morus alba L. extract and vitamin C. (A) Two-dimensional synergy contour plot and (B) three-dimensional synergy landscape generated using the SynergyFinder platform based on the Loewe additivity model. Concentrations were tested at 0.125X, 0.25X, 0.5X, 1X, and 2X relative to the EC50 values (21.30 µg/mL for Morus alba L. extract and 43.30 µg/mL for vitamin C).
Figure 3. Synergistic effects of Morus alba L. extract and vitamin C. (A) Two-dimensional synergy contour plot and (B) three-dimensional synergy landscape generated using the SynergyFinder platform based on the Loewe additivity model. Concentrations were tested at 0.125X, 0.25X, 0.5X, 1X, and 2X relative to the EC50 values (21.30 µg/mL for Morus alba L. extract and 43.30 µg/mL for vitamin C).
Foods 14 03630 g003
Figure 4. Dose–response matrix showing the synergistic interactions of Morus alba L. extract and vitamin C.
Figure 4. Dose–response matrix showing the synergistic interactions of Morus alba L. extract and vitamin C.
Foods 14 03630 g004
Figure 5. Neuroprotective effects of MAC on SH-SY5Y cells exposed to H2O2. (A) Micrographs illustrating cell morphology at 10× magnification. (B) Cell viability measurements expressed as mean ± SEM (n = 4). *** p < 0.001 vs. control; ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Figure 5. Neuroprotective effects of MAC on SH-SY5Y cells exposed to H2O2. (A) Micrographs illustrating cell morphology at 10× magnification. (B) Cell viability measurements expressed as mean ± SEM (n = 4). *** p < 0.001 vs. control; ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Foods 14 03630 g005
Figure 6. Effects of MAC on the ERK/CREB signaling pathway in SH-SY5Y cells exposed to H2O2. (A) Representative Western blots of ERK and CREB. (B,C) Quantification of ERK and CREB expression, respectively. Data are means ± SEM (n = 4). ** p < 0.01 and *** p < 0.001 vs. control; ## p < 0.01 and ###p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Figure 6. Effects of MAC on the ERK/CREB signaling pathway in SH-SY5Y cells exposed to H2O2. (A) Representative Western blots of ERK and CREB. (B,C) Quantification of ERK and CREB expression, respectively. Data are means ± SEM (n = 4). ** p < 0.01 and *** p < 0.001 vs. control; ## p < 0.01 and ###p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Foods 14 03630 g006
Figure 7. Effects of MAC on Bcl-2 and BAX expression in SH-SY5Y cells exposed to H2O2. (A) Representative Western blots of Bcl-2 and BAX. (B,C) Quantification of Bcl-2 and BAX expression, respectively. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; # p < 0.05 and ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Figure 7. Effects of MAC on Bcl-2 and BAX expression in SH-SY5Y cells exposed to H2O2. (A) Representative Western blots of Bcl-2 and BAX. (B,C) Quantification of Bcl-2 and BAX expression, respectively. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; # p < 0.05 and ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Foods 14 03630 g007
Figure 8. Effects of MAC on the Bcl-2/BAX ratio in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; # p < 0.05 and ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Figure 8. Effects of MAC on the Bcl-2/BAX ratio in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; # p < 0.05 and ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Foods 14 03630 g008
Figure 9. Effects of MAC on cleaved caspase-3 expression in SH-SY5Y cells exposed to H2O2. (A) Representative Western blots of cleaved caspase-3. (B) Quantification of cleaved caspase-3 expression. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; # p < 0.05 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Figure 9. Effects of MAC on cleaved caspase-3 expression in SH-SY5Y cells exposed to H2O2. (A) Representative Western blots of cleaved caspase-3. (B) Quantification of cleaved caspase-3 expression. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; # p < 0.05 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X.
Foods 14 03630 g009
Figure 10. Effects of MAC on antioxidant enzyme activities in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). ** p < 0.01 and *** p < 0.001 vs. control; # p < 0.05, ## p < 0.01 and ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X; CAT: catalase; SOD: superoxide dismutase; GSH-Px: glutathione peroxidase.
Figure 10. Effects of MAC on antioxidant enzyme activities in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). ** p < 0.01 and *** p < 0.001 vs. control; # p < 0.05, ## p < 0.01 and ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X; CAT: catalase; SOD: superoxide dismutase; GSH-Px: glutathione peroxidase.
Foods 14 03630 g010
Figure 11. Effects of MAC on intracellular ROS production in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X; ROS: reactive oxygen species.
Figure 11. Effects of MAC on intracellular ROS production in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; ### p < 0.001 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X; ROS: reactive oxygen species.
Foods 14 03630 g011
Figure 12. Effects of MAC on MDA levels in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; ## p < 0.01 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X; MDA: malondialdehyde.
Figure 12. Effects of MAC on MDA levels in SH-SY5Y cells exposed to H2O2. Data are means ± SEM (n = 4). *** p < 0.001 vs. control; ## p < 0.01 vs. H2O2 + vehicle. H2O2: hydrogen peroxide; MAC1: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:1X; MAC2: anthocyanin-enriched Morus alba L. extract combined with vitamin C at 1X:0.5X; MDA: malondialdehyde.
Foods 14 03630 g012
Figure 13. Schematic diagram summarizing the synergistic neuroprotective effects of anthocyanin-enriched Morus alba L. extract combined with vitamin C against H2O2-induced oxidative stress in SH-SY5Y neuronal cells. H2O2: hydrogen peroxide; ROS: reactive oxygen species; MDA: malondialdehyde; CAT: catalase; SOD: superoxide dismutase; GSH-Px: glutathione peroxidase; ERK: extracellular signal-regulated kinase; CREB: cAMP response element-binding protein; Bcl-2: B-cell lymphoma 2; BAX: Bcl-2-associated X; Apaf-1: apoptotic protease activating factor 1; MAC: anthocyanin-enriched Morus alba L. extract combined with vitamin C.
Figure 13. Schematic diagram summarizing the synergistic neuroprotective effects of anthocyanin-enriched Morus alba L. extract combined with vitamin C against H2O2-induced oxidative stress in SH-SY5Y neuronal cells. H2O2: hydrogen peroxide; ROS: reactive oxygen species; MDA: malondialdehyde; CAT: catalase; SOD: superoxide dismutase; GSH-Px: glutathione peroxidase; ERK: extracellular signal-regulated kinase; CREB: cAMP response element-binding protein; Bcl-2: B-cell lymphoma 2; BAX: Bcl-2-associated X; Apaf-1: apoptotic protease activating factor 1; MAC: anthocyanin-enriched Morus alba L. extract combined with vitamin C.
Foods 14 03630 g013
Table 1. Combination index values of Morus alba L. extract and vitamin C.
Table 1. Combination index values of Morus alba L. extract and vitamin C.
Ratios (Dose Level)% InhibitionCombination Index ValueInteraction
Morus alba L. ExtractVitamin C
0.125X0.125X50.03 ± 0.611.58 ± 0.05Antagonism
0.250.125X41.11 ± 1.791.43 ± 0.06Moderate antagonism
0.5X0.125X22.36 ± 0.710.90 ± 0.03Nearly additive effect
1X0.125X20.29 ± 0.691.16 ± 0.04Slight antagonism
2X0.125X17.35 ± 0.821.01 ± 0.05Nearly additive effect
0.125X0.2545.31 ± 0.621.41 ± 0.03Moderate antagonism
0.250.2538.31 ± 1.081.36 ± 0.06Moderate antagonism
0.5X0.2520.91 ± 1.180.89 ± 0.06Slight synergism
1X0.2518.49 ± 1.071.04 ± 0.05Nearly additive effect
2X0.2520.89 ± 1.061.23 ± 0.07Moderate antagonism
0.125X0.5X43.15 ± 0.471.34 ± 0.02Moderate antagonism
0.250.5X35.94 ± 0.501.25 ± 0.02Moderate antagonism
0.5X0.5X11.20 ± 0.580.46 ± 0.02Synergism
1X0.5X9.85 ± 1.120.61 ± 0.08Synergism
2X0.5X20.31 ± 1.481.23 ± 0.11Moderate antagonism
0.125X1X41.23 ± 1.321.28 ± 0.04Moderate antagonism
0.251X30.04 ± 0.571.05 ± 0.02Nearly additive effect
0.5X1X17.45 ± 0.870.74 ± 0.04Moderate synergism
1X1X8.66 ± 0.980.52 ± 0.06Synergism
2X1X18.73 ± 0.731.09 ± 0.05Nearly additive effect
0.125X2X44.23 ± 0.821.37 ± 0.03Moderate antagonism
0.252X31.60 ± 2.021.11 ± 0.07Slight antagonism
0.5X2X23.46 ± 1.911.02 ± 0.09Nearly additive effect
1X2X19.64 ± 1.371.12 ± 0.08Slight antagonism
2X2X20.63 ± 1.291.17 ± 0.07Slight antagonism
Data are presented as means ± SEM (n = 3).
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

Mairuae, N.; Jittiwat, J.; Apaijit, K.; Noisa, P.; Bai, G.; Hou, Y.; Palachai, N. Synergistic Effects of Anthocyanin-Enriched Morus alba L. Extract and Vitamin C: Promising Nutraceutical Ingredients in Functional Food Development for Neuroprotection. Foods 2025, 14, 3630. https://doi.org/10.3390/foods14213630

AMA Style

Mairuae N, Jittiwat J, Apaijit K, Noisa P, Bai G, Hou Y, Palachai N. Synergistic Effects of Anthocyanin-Enriched Morus alba L. Extract and Vitamin C: Promising Nutraceutical Ingredients in Functional Food Development for Neuroprotection. Foods. 2025; 14(21):3630. https://doi.org/10.3390/foods14213630

Chicago/Turabian Style

Mairuae, Nootchanat, Jinatta Jittiwat, Kwanjit Apaijit, Parinya Noisa, Gang Bai, Yuanyuan Hou, and Nut Palachai. 2025. "Synergistic Effects of Anthocyanin-Enriched Morus alba L. Extract and Vitamin C: Promising Nutraceutical Ingredients in Functional Food Development for Neuroprotection" Foods 14, no. 21: 3630. https://doi.org/10.3390/foods14213630

APA Style

Mairuae, N., Jittiwat, J., Apaijit, K., Noisa, P., Bai, G., Hou, Y., & Palachai, N. (2025). Synergistic Effects of Anthocyanin-Enriched Morus alba L. Extract and Vitamin C: Promising Nutraceutical Ingredients in Functional Food Development for Neuroprotection. Foods, 14(21), 3630. https://doi.org/10.3390/foods14213630

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