Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

Skip to main content

Nano-encapsulated ferulic acid in sesame protein isolate alleviates acrylamide-induced liver toxicity and genotoxicity in rats via oxidative stress and DNA damage modulation

Abstract

Background

Acrylamide (ACR) induces hepatotoxicity and genotoxicity through oxidative stress and inflammatory processes.

Aims

This study explores the potential of ferulic acid encapsulated in sesame protein isolate (SPI) and its nanoform as a non-toxic, effective therapy for ACR-induced oxidative liver injury in rats.

Methods

SPI was prepared from defatted sesame flour. SPI exposed to ultrasonic waves to obtain nano SPI, and then ferulic acid was added to form capsules. Fourier transforms infrared spectra, scaning electron microscope, and polarizing optical microscope were used in investigating functional groups and surface morphology of both encapsulations respectively. Rats were divided into four groups, each consisting of six animals: normal control, ACR-treated (20 mg/kg/day), sesame protein encapsulated ferulic acid-treated, and sesame protein nano-encapsulated ferulic acid-treated groups. Both encapsulated forms were administered daily in the diet alongside ACR for two weeks. Liver function indices, oxidative stress biomarkers, DNA fragmentation, comet assay, and histopathological and immunohistochemical examinations were performed.

Results

The encapsulation efficiency of the nano-encapsulated form was higher than that of the other forms. Both encapsulated forms significantly improved liver function, elevated levels of GSH, GPx, SOD, and CAT were observed, along with decreased concentrations of MDA, interleukin-6, and tumor necrosis factor-α. The treatments also provided protection against DNA damage and genotoxicity, alleviated histological damage, and reduced liver toxicity and genotoxicity.

Conclusion

Both encapsulated forms, especially the nanoform, significantly mitigated liver toxicity. These findings underscore their potential as effective natural therapies for liver damage caused by ACR, and supporting liver health.

Peer Review reports

Introduction

Acrylamide (ACR), with the molecular formula C3H5NO, is a water-soluble compound commonly used in polymer production [1]. It has been demonstrated that acrylamide forms when carbohydrate-rich foods are subjected to high-temperature cooking (above 120 °C). This process occurs via a Maillard reaction between reducing sugars, such as glucose, and amino acids like asparagine. This process occurs during cooking methods like roasting, grilling, baking, and frying [2]. Consequently, individuals who frequently consume foods cooked at elevated temperatures may ingest approximately 0.5 mg/kg/day of ACR [3].

ACR is also extensively applied across various industries, including cosmetics, paper manufacturing, textiles, wastewater treatment, and printing, and it plays a role in polymers and copolymers production [4]. However, ACR exposure has been linked to numerous toxic effects, particularly hepatotoxicity, and genotoxicity [5].

The primary mechanism underlying ACR’s toxicity is oxidative stress. ACR is metabolized by pathways involving glutathione or cytochrome P450, resulting in the production of glycidamide, a metabolite with higher toxicity than ACR itself [6]. It has been indicated that exogenous antioxidants can hinder glycidamide formation, thus mitigating oxidative damage [7]. Elevated doses of ACR have been shown to disrupt the oxidative balance and enzyme function, increasing oxidative stress and leading to organelle damage, cellular metabolic disruption, DNA fragmentation, and cell death [8].

Research indicates that phenolic compounds may offer protection against oxidative damage and ACR’s associated toxic effects [9, 10]. Ferulic acid (FA), a phytochemical with the molecular structure C10H10O4, is naturally present in the cell walls of various plants, including grains (such as rice, wheat, and oats), nuts, citrus fruits, coffee, tea, and peanuts. FA has been documented to provide multiple health benefits, including antioxidant, anti-apoptotic, hepatoprotective, anti-inflammatory, and anti-carcinogenic properties [11].

The broad range of preventive effects associated with FA is attributed to its potent antioxidative properties. In animal models, it has demonstrated the ability to prevent methotrexate-induced liver toxicity and reduce oxidative stress, inflammation, and apoptosis by enhancing Nrf2/HO-1 signaling pathways [12].

However, FA ‘s bioavailability and stability are limited. Therefore, encapsulating FA within nanocarriers can enhance its delivery and efficacy. Sesame protein is a promising natural biopolymer for encapsulation due to its high binding affinity, biodegradability, and antioxidant properties [13, 14].

Sesame (Sesamum indicum L.), An essential oil seed crop, is a member of the Pedaliaceae family with a high protein content (30–60%), about 50% oil, and a wide variety of phytochemicals, including phenolics, carotenoids, tocopherols, and phytosterols [15,16,17]. Sesame seeds exhibited a wide range of biological properties, including antidiabetic, anticancer, cardioprotective, and antioxidant properties [18]. Sesame protein isolate (SPI) is amphiphilic, enabling the formation of nanoparticles through hydrophobic interactions, enhancing the stability and controlled release of encapsulated bio-actives [19]. SPI was prepared from defatted sesame flour and utilized for human and animal nutrition due to its protein quality [20]. Ultrasound (US) technology has been employed to modify protein structures, reducing particle size and altering functional properties. These modifications have been shown to enhance the encapsulation efficiency and bioactive stability of FA, as demonstrated with soy and peanut proteins [21,22,23]. Encapsulation technology allows bioactive molecules, such as FA, to be enclosed within a protective matrix, improving their stability and controlled release in physiological environments [24].

Therefore, this study aimed to evaluate the therapeutic effects of FA encapsulated in SPI and its nanoform against ACR-induced liver toxicity and genotoxicity in rats. By leveraging sesame protein as a natural encapsulating agent and employing nanotechnology, this research introduces an innovative approach to enhance the stability, and bioavailability of FA, providing a novel and effective strategy to mitigate oxidative stress, genotoxicity, and liver damage caused by ACR.

Materials and methods

Materials

Chemicals

Sesame seed, Sodium alginate (SA), 1,1-Diphenyl-2-picryl-hydrazyl (DPPH•), ABTS 2,2′-azino-bis-(3- ethylbenzothiazoline-6-sulphonic acid), ferulic acid, all chemicals and reagents used in the study were of analytical grade and applied without further purification Acrylamide (Sigma-Aldrich, St. Louis, MO, USA).

Methods

Preparation of sesame protein isolate (SPI)

Sesame seed meal, which was already hydraulic pressed, was subjected to defatting using a soxhlet apparatus and n-hexane as a defatting solvent. The defatted meal was spread to dry and then ground in a coffee mill to obtain a finely divided material suitable for further extraction. Generally, the Sesame seed meal was dissolved in ten volumes of distilled water, and SPI was prepared by the iso precipitation method as described by Mohamed et al. [25]. The dried SPI was saved in the refrigerator until used.

Preparation of SPI capsules and nano SPI capsules

SPI (150 g) was hydrated to 10% w/v in dist. water overnight at room temperature (21˚C). Sodium alginate 10% w/v dissolved in dist. water by gentle magnetic stirring at 60 ℃ for 20 min. for complete hydration. SPI solution was mixed with sodium alginate in a ratio of 9:1 by gentle stirring for 1 h. at pH 7. FA was then added drop by drop to the wall material 1%w/w by gentle stirring for 20 min.

SPI nanoparticles were prepared as described by El-Kholy et al. [26] with some modifications. SPI (150 g) was hydrated to 10% w/v in dist. water overnight at room temperature (21˚C). Nano protein solution was formed using high-speed homogenizer T18 basic (IKA, Wilmington, USA), operating at a speed of 20,000 rpm for 5 min, then ultrasonicated at 720 W power, for 20 min at 25 °C and with 50% pulse using an ultrasonic with a titanium probe (vibra cell, USA). The solution temperature was controlled to be ≤ 35 ◦C in an ice-water bath. Then it followed the same steps in preparation of SPI capsules.

Both capsules were freeze-dried (by CHRIST Alpha 1–4 LD plus, Germany) at-42 °C under a pressure of 10 Pa for 48 h and stored at -4 °C. After freeze-drying, the samples were stored at 4 ℃ until future use.

Characterizations of prepared SPI

Determination of amino acid profile of Sesame protein isolate using HPLC

The amino acid profile was determined as described by Younos & Akl [27]. It was determined using the HPLC-Pico-Tag method, a commercially developed technique by Waters Associates for integrated amino-acid analysis.

Particle size distribution of nano SPI

The colloidal stability properties, including the demonstration of hydrodynamic diameter (HD) based on dynamic light scattering techniques, have been investigated using the Malvern Zeta Sizer Nano ZS Nano instrument with He/Ne laser (i.e., λ = 633 nm) at 173 °C collecting backscatter optics. The colloidal properties of samples were examined by adding about 2 ml of the sample and dispersing with 1 ml dist. H2O for 10 min under ultrasonication before measurements.

Fourier transforms infrared spectra

Functional groups were detected in the range of 400–4000 cm − 1 using (Shimadzu 8400 S) FTIR Spectrophotometer.

Scan electron microscope (SEM)

SEM studying the surface morphology (FEI IN SPECTS Company, Philips, Poland) environmental scanning without coating with a JEOL JEM-2100 electron microscope at 100k x magnification and an acceleration voltage of 120 kV.

Polarizing optical microscope (POM)

The images were taken by Leica DM750P (Leica microsystems. Switzerland) (Optical magnification 400 times, digital magnification 4 times).

Encapsulation efficiency (EE)

EE was determined according to the method established by Siles-S´anchez et al. [28] 45 mg of encapsulated capsules were suspended in 5 ml of water and agitated for 10 min. The supernatant containing non-encapsulated compounds (500 µl) was centrifuged at 3,000xg for 15 min. EE% was calculated using the following equations: EE(%) = 100 - (sum of the supernatant phenolic compounds /sum of the phenolic compounds in extract)×100.

Evaluation of antioxidant activity by the following methods

Radical DPPH scavenging activity

Free radical scavenging capacity was determined using the stable 1,1-Diphenyl-2-picryl-hydrazyl (DPPH•). The approach given by De Ancos et al. [29] was used to calculate DPPH radical scavenging. The decrease of the DPPH radical was detected at 517 nm. The assay was performed in triplicate. The results were represented as percent inhibition of the DPPH using the following equation:

$$\:Inhibition\:\left(\%\right)\hspace{0.17em}=\hspace{0.17em}100\:\times\:\:(A_{control}-A_{sample})/A_{control}$$

Where: A control: the absorbance of the methanolic DPPH solution, A sample: the absorbance of the extract.

ABTS radical scavenging capacity

Free radical scavenging activity was determined by ABTS radical cation decolorization assay according to Re et al. [30]. ABTS·+ radical cation was produced by the reaction between 7 mM ABTS in water and 2.45 mM potassium persulfate (1:1), stored in the dark at room temperature for 12–16 h. before use. ABTS·+ solution was then diluted with methanol to obtain an absorbance of 0.700 at 734 nm. After the addition of 5 µl of sample to 3.995 ml of diluted ABTS·+ solution, the absorbance at 734 nm was measured 30 min after the initial mixing. The ABTS scavenging effect was measured using the following formula:

$$\:Inhibition\:\left(\%\right)\hspace{0.17em}=\hspace{0.17em}100\:\times\:\:(A_{control}-A_{sample})/A_{control}$$

Where: A control: the absorbance of ABTS solution, A ample: the absorbance of the extract.

Animals

In this study, male Sprague Dawley rats aged 4 to 6 weeks and weighing 200–240 g were sourced from the animal facility of the National Nutrition Institute, Egypt. The animals were allowed a one-week acclimatization period, during which they were housed under standardized laboratory conditions. Each rat was individually placed in a polypropylene cage, with the environment maintained at a temperature of 25 °C, a relative humidity of 55%, and a 12-hour light/dark cycle. The rats were provided with unrestricted access to a standard laboratory diet and water throughout the experiment.

Diet composition

A balanced diet was formulated in accordance with the AIN-93 guidelines outlined by Reeves et al. [31]. The composition included 12% protein derived from casein, 10% corn oil, 10% sucrose, 58.5% corn starch, 5% cellulose, 3.5% salt mixture, and 1% vitamin mixture. Both the salt and vitamin mixtures were prepared based on the AIN-93 formulation, as described by Reeves et al. [31].

Ethical statement

This study was approved by the National Nutrition Institute Ethics Committee (ethical approval number: [IN000147]). Furthermore, the research protocol strictly adhered to the guidelines for the care and use of laboratory animals as established by the National Institutes of Health, detailed by Garber et al. [32].

Experimental design

Twenty-four male Sprague-Dawley rats were divided into four groups (6 rats per group) as follows:

  • Group 1: Normal control group. Rats were orally administered normal saline (0.5 mL/rat) and fed a standard balanced diet daily.

  • Group 2: ACR group. Rats were orally administered ACR (20 mg/kg/day) daily for 2 weeks [33]. This has been reported to induce total liver toxicity [34].

  • Group 3: ACR + Sesame protein encapsulated FA. Rats were orally administered 20 mg/kg ACR daily and fed a balanced diet supplemented with 0.1% FA Zhang et al. [35] and 5% sesame protein in encapsulated form.

  • Group 4: ACR + Sesame protein Nano-encapsulated FA. Rats were orally administered 20 mg/kg ACR daily and fed a balanced diet supplemented with 0.1% FA Zhang et al. [35] and 5% sesame protein in nano-encapsulated form.

All rats in the study were consistently provided with the assigned diet and water ad libitum. Throughout the study, body weight and food consumption were monitored on a weekly basis to assess any changes over time.

Growth-related parameters

At the conclusion of the 2-week study, body weight gain and relative liver weight were evaluated using the methodology described by Chapman et al. [36].

Blood sampling and preparation of liver homogenate

The animals were euthanized 24 h after the 14th dose of ACR administration. Thiopental sodium (50 mg/kg) was used to anesthetize the rats prior to euthanasia. Blood samples were collected via decapitation while the animals were under deep anesthesia. Serum was separated by centrifugation at 3000 rpm for 15 min at 4 °C using a Laborezentrifugen 2k15 centrifuge (Sigma, Germany) and stored at -20 °C for subsequent biochemical analyses. Liver tissues were immediately excised, rinsed with ice-cold saline, blotted dry with filter paper, and weighed. The liver was divided into three portions: one portion was snap-frozen in liquid nitrogen and stored at -80 °C for genotoxicity assessments, the second was preserved in 10% (v/v) neutral buffered formalin for histopathological evaluation, and the third, comprising one gram of tissue, was homogenized in ice-cold phosphate-buffered saline (pH 7.4) to prepare a 20% w/v homogenate using an MPW-120 tissue homogenizer (BitLab Medical Instruments, Poland). The homogenate was centrifuged at 4000 rpm for 10 min at 4 °C using a Laboratory Centrifuge 2 K15 (Sigma, Germany), as described by Essa et al. [37]. The resulting supernatant was collected, stored at -80 °C, and used for measuring oxidative stress markers and inflammatory parameters.

Liver injury assessment

The evaluation of hepatic function biomarkers and protein fractions included the following: Serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured using the Reitman and Frankel method 1975 [38]. Alkaline phosphatase (ALP) levels were determined according to the procedure by Bessey [39], while gamma-glutamyl transferase (γ-GT) was assessed as outlined by Szasz [40]. Total protein and albumin levels were quantified using the methods described by Rheinhold [41] and Doumas [42], respectively. Additionally, total and direct bilirubin were measured according to the methodology of Balistreri and Shaw [43]. All assays were performed using commercial kits from Salucea Co., Netherlands.

Assessment of oxidative stress markers

Hepatic malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), reduced glutathione (GSH), and nitric oxide (NO) levels were evaluated spectrophotometrically using the methods outlined by Nair and Turner [44]; Sun [45]; Luck [46]; Rotruck et al. [47]; Jollow [48]; and Montgomery and Dymock [49], respectively. Biochemical analyses were carried out in accordance with the manufacturer’s instructions. The optical density of all measured parameters was determined using a Shimadzu UV-2401 PC spectrophotometer (Australia). Spectrophotometric analyses were conducted in accordance with the guidelines provided with the bio diagnostic kits.

Determination of TNFα and IL-6 in liver tissue

Levels of TNF-α and IL-6 were measured in liver homogenate using the rat TNF-α ELISA kit (Sunlong Biotech Co., Catalog no. SL0722Ra, China) and the IL-6 ELISA kit (Sunlong Biotech Co., Catalog no. SL0411Ra, China), following the sandwich-ELISA method and spectrophotometric analysis as per the instructions provided with the kits.

Histopathological studies

A. Hematoxylin and Eosin (H/E)

Liver specimens were fixed in a 10% formalin saline solution. After dehydration in different grades of ethyl alcohol (100%, 5 min; 95%, 2 min; 80%, 2 min; 70%, 2 min), cleaning in xylol, impregnation, and proper fixing, we embedded the specimens in paraffin wax (5 mm thick pieces). Then, using a rotatory microtome (LEICA RM 2125 UK), specimens were cut with a 5 μm thickness and placed on glass slides. Staining with hematoxylin and eosin (H/E) solutions (G1120, Solarbio, China) for 30 min at 55 ℃ was used to explore the lung’s general histological structure under a light microscope [50].

For Oil Red O Staining, the samples were rinsed with PBS and fixed in 10% buffered formalin, then stained with Oil Red O (0.5 g in 100 mL of isopropanol) for 60 min. After discarding the staining solution, isopropanol was added to the samples to elute the retained dyes [51]. After mounting, the sections were visualized and photographed using an optical microscope with a camera (Olympus, Tokyo, Japan) at 100× or 400× magnification. Slides were examined under a light microscope (Primo star, ZEISS, China). The photos were taken using (Axiocam Erc 5s, ZEISS, China) camera, at the pathology department, faculty of Medicine for Girls, Al Azhar University.

B. Immunohistochemical study

Apoptosis and Caspase-3 assessment Regarding immunohistochemistry stain, liver paraffin slices were deparaffinized and rehydrated on positively charged glass slides. After 30 min of incubation in 0.3% hydrogen peroxide in 100% methanol, endogenous peroxidase was rendered inactive. At room temperature, sections were incubated for 30 min in 5% skim milk. Microwave (700 W) treatment in 10 mM citrate buffer (pH 7.4) for 15 min was used to retrieve the antigen. After that, sections were incubated with anti-rat caspase-3 for an entire night at 4ºC. Sections were treated in secondary antibodies for 30 min at room temperature following PBS washing. After adding 3-diaminobenzidine for two to four minutes, washing in distilled water, then counter-staining with Mayer’s haematoxylin for one minute at room temperature, a brown hue forms [52, 53].

DNA fragmentation assay

DNA extraction was done from liver tissue (200 mg) by using the Zymoresearch Quik-gDNA MiniPrep kit (CAT. NO. D3024) following the manufacturer’s instructions. DNA extraction was quantified as optical density (OD) using a spectrophotometer at 260 nm. DNA fragmentation was detected as 20 µg of DNA sample was loaded on 2% agarose gel electrophoresis (Ultra-pure agarose, electrophoresis grade) with Ethidium Bromide (0.5 µg/ml). DNA visualization and documentation were done under a UV transilluminator using a DNA ladder,100 bp (Jena Bioscience, Germany), and gel documentation system (Biodoc. Analyser (Biometra).

Alkaline comet assay (single-cell gel electrophoresis)

According to the manufacturer’s instructions, a comet assay test was conducted to detect the DNA strand breaks using COMET ASSAY™ PROTOCOLS (Ams Biotechnology, Europe, ltd), CAT. # 4250-050-K (Comet Assay™ Kit). It is a sensitive technique that enables analysis at the single-cell level.

Statistical analysis

Statistical analyses were conducted using SPSS version 25. The data obtained from the animal experiments were expressed as mean ± standard error (SE) and analyzed using a one-way analysis of variance (ANOVA) followed by Duncan’s multiple range test. Statistical significance was established at a threshold of P ≤ 0.05.

Results

Characterizations of SPI

The amino acid profile of SPI showed that Arginine was the most abundant amino acid, followed by Histidine, Aspartic acid, and Glutamic acid, as shown in Fig. 1. The size of soluble prepared nano SPI is represented in Fig. 2, showing a peak with a percentage of 100% at 71.08 ± 47.66 nm.

Fig. 1
figure 1

Amino acid profile of SPI expressed as gram for each 100 gram

Fig. 2
figure 2

The particle size of the prepared nano SPI

Fourier transforms infrared (FTIR) spectroscopy

The most displayed functional groups detected by FTIR spectra of SPI, and nano SPI, SPI encapsulated, and nano SPI encapsulated were represented in Fig. 3. The spectrum of SPI consists of several bands located at 3273, 2962 cm− 1, 1669, 1632 and 1537 cm− 1. The spectrum of the nano form of SPI showed a similar pattern of bands as those of SPI. However, the bands’ intensity was changed. FTIR spectrum of both encapsulated forms displays some difference in frequency and intensity. It shows a broad band at 3279–3732, 2924, 1633 and 1529 cm− 1 which are assigned to the O-H, C-H, C = O, C-N and N-H groups of peptide groups. In nano encapsulated form, the bands’ intensity was changed, but it still shows the same bands at 3276–3707, 2967, 1632, 1531, and 699–407 cm-1 as detected in SPI encapsulated.

Fig. 3
figure 3

Fourier transform infrared spectrophotometer. A(SPI and nano SPI, B(SPI encapsulated and nano SPI encapsulated

The morphological structure of each encapsulated SPI investigated by polarized optical microscope (POM) and scan electron microscope (SEM)

POM images revealed that FA appeared as needle-like crystals as shown in Fig. 4A. Additionally, both types of protein, whether in micro or nano size exhibited a uniform spherical shape (Fig. 4B and C). SPI particles were found to range from 1.91 ± 0.2 μm to 9.50 ± 0.3 μm (Fig. 4B), while SPI nanoparticles ranged from 439 ± 4 nm to 747 ± 8 nm in the dry form as shown in Fig. 2C. The SEM image in Fig. 4D shows that FA appears as lightening crystals surrounded by nano SPI. This also confirms the formation of nano-protein which ranged from approximately 100 ± 5 nm:400 ± 10 nm.

Fig. 4
figure 4

Shows POM image of A) ferulic acid, B) SPI encapsulated, C) nano SPI encapsulated. Optical magnification 400 times, digital magnification 4 times. D) SEM image of nano SPI encapsulated

Encapsulation efficiency (EE) and antioxidant activity of each SPI encapsulated

Encapsulation efficiency (EE) of nano SPI encapsulated 85.35 ± 1.2 is higher than that of SPI encapsulated (67.41 ± 0.8) which illustrates the importance of nano form particles. The Antioxidant activity of the soluble capsules also showed that the percent scavenging activity of DPPH and ABTS percentage of SPI encapsulated is higher in SPI encapsulated (90.22 ± 0.5, 77.36 ± 0.7) than nano SPI encapsulated (77.65 ± 0.4, 68.88 ± 0.3), respectively as shown in Table 1.

Table 1 Encapsulation efficiency (EE) and Antioxidant activity of both capsules

Effect of SPI encapsulated FA and nano SPI encapsulated FA on nutritional parameters, liver function, hepatic oxidative stress, and inflammatory markers

Table 2 Nutritional parameters, liver weight, and liver index of different experimental groups

As shown in Table 2, the ACR group showed significant (p < 0.05) reductions in final body weight, body weight gain, and total food intake by 8.6%, 51%, and 23.1%, respectively, compared to the normal group, alongside a 1.45-fold increase in relative liver weight, indicating hepatic stress. Treatment with SPI encapsulated FAsignificantly improved these parameters, with final body weight, body weight gain, and food intake increasing by 4.8%, 53.5%, and 10.3%, respectively, compared to the ACR group, while relative liver weight decreased by 7.5%. Nano SPI encapsulated FA demonstrated superior ameliorative effects, restoring final body weight, body weight gain, and food intake to near-normal levels (8.3%, 91.9%, and 20.1% improvement, respectively, vs. acrylamide group) and reducing relative liver weight by 16.6%.

Table 3 Liver function parameters of different experimental groups

Table 3 shows that, the ACR group exhibited significant (p < 0.05) disruptions in liver function, with ALT, AST, ALP, and total bilirubin increasing by 3.29-, 3.8-, 3.32-, and 3.32-fold, respectively, compared to the normal group. serum γ-GT was significantly reduced by 54.6%, and albumin decreased by 19%, reflecting hepatic damage. Treatment with SPI encapsulated FA significantly reduced serum ALT (45.7%), AST (39.8%), ALP (56.6%), and total bilirubin (34.9%) while partially restoring albumin (7.4%) and γ-GT (70.0%) compared to the ACR group. On the other hand, Nano SPI encapsulated FA demonstrated greater amelioration, reducing ALT, AST, ALP, and total bilirubin by 55.4%, 51%, 64.5%, and 50.6%, respectively, while restoring albumin and γ-GT to near-normal levels. These findings underscore the enhanced hepatoprotective effects of nano-encapsulation in mitigating ACR-induced toxicity.

Table 4 Hepatic oxidative, antioxidant, and inflammatory mediators of different experimental groups

The ACR group demonstrated significant (p < 0.05) increases in hepatic oxidative stress and inflammatory markers, with MDA levels rising 2.46-fold, NO increasing 2.37-fold, IL-6 elevating 2.52-fold, and TNF-α increasing 2.60-fold compared to the normal group. Antioxidant parameters, including GSH, GPx, catalase, and SOD, were significantly reduced by 52.4%, 59.3%, 56.5%, and 53.5%, respectively, indicating compromised antioxidant defense. Treatment with SPI encapsulated FA significantly mitigated these effects, reducing MDA by 30.3%, NO by 76.5%, IL-6 by 14.8%, and TNF-α by 16.2%, while enhancing GSH, GPx, CAT, and SOD by 40.0%, 51.1%, 41.7%, and 34.3%, respectively, compared to the ACR group. Nano SPI encapsulated FA showed superior ameliorative effects, further reducing hepatic MDA by 44.8%, NO by 80.4%, IL-6 by 36.6%, and TNF-α by 37.15.0%, with greater enhancements in GPx (89.1%), CAT (82.7%), and SOD (76.5%) Table 4. These findings highlight the potent antioxidant and anti-inflammatory properties of nano-encapsulation.

DNA fragmentation in hepatic tissue

Hepatic DNA fragmentation was tested by agarose gel electrophoresis. The results in Fig. 5; Table 5 revealed a highly significant (p < 0.0001) intra-nucleosomal DNA fragmentation in hepatocytes of the ACR group, evidenced by high smear in the DNA band and low OD value at 260 nm, compared to the control and treated groups. In contrast, both treated groups revealed intact DNA, with a minor smear observed in the SPI-FA-treated group. Surprisingly, the sesame nano-encapsulated form of FA recorded a strong protective effect against DNA damage compared to the control group, evidenced by a sharp, clear DNA band and higher OD at 260 nm. The results indicated that nano-encapsulation enhances the protective effect of the sesame protein of FA against ACR-induced genotoxicity.

Comet assay analysis

DNA damage was also detected in rat hepatocytes using an alkaline comet assay. Figure 6 (A, B, C, D) and Table 5 showed a marked increase (p < 0.0001) in Tail length and Tail% (% DNA in tail) among ACR-treated rats, compared to all experimental groups. Treatment with SPI encapsulated FA and nano-SPI encapsulated FA significantly (p < 0.0001) mitigated the genotoxic effect of ACR on hepatocyte DNA by reducing the % DNA in the comet tail length, with % changes reached 56.20% and 57.8, respectively, compared to the ACR group. Nano-SPI-encapsulated FA recorded a superior effect than Sesame protein-encapsulated FA in DNA protection against DNA damage caused by acrylamide exposure.

Fig. 5
figure 5

Agarose gel electrophoresis showed migration and DNA fragmentation patterns for isolated DNA from hepatocytes of all studied groups. M: DNA ladder, Lane 1: Control group DNA, Lane 2: ACR group DNA, Lane 3: SPI encapsulated FA group DNA, Lane 4: Nano-SPI encapsulated FA group DNA

Table 5 OD260 and comet parameters: tail length (µm), tailed %, and untailed% for DNA samples in all experimental groups
Fig. 6
figure 6

Fluorescence photomicrograph of hepatocytes for all the studied groups after single-cell gel electrophoresis (comet assay) process. (A) control group (intact DNA), (B): ACR group (Degenerated DNA; DNA head at the blue arrow and DNA tail at a blue straight line), (C): SPI encapsulated FA-treated group (Degenerated DNA; DNA head at the blue arrow and DNA tail at blue straight line), (D): Nano-SPI encapsulated ferulic acid- treated group (intact DNA), Scale bar 50 nm

Histopathological results

The results of histopathological examination are shown in Fig 7 (A, B1, B2, C, D). There were no structural alterations found in the liver of the control group. Normal hepatic cells with preserved cytoplasm, a large nucleus, and a central vein were present in the liver tissue.

In comparison to the control group, all of the tissue sections taken from the liver of rats given ACR displayed abnormal liver architecture, central vein congestion, fatty degeneration, inflammatory cellular infiltration in the portal area and between hepatocytes, sinusoidal dilatation and congestion, and Kupffer cell hyperplasia.

Additionally, compared to the ACR -treated group, the SPI encapsulated FA group exhibited a reversal of hepatic abnormalities, primarily vacuolar degeneration. Nano -SPI encapsulated FA group exhibited more improvement compared to SPI encapsulated FA form group, hepatic tissue was comparable to control group.

The gross view of treated liver from different studied groups is displayed in Fig. 8(A, B, C, D). Additionally, it displays the findings of histological analyses that use oil red O staining to identify fat deposits. Compared to the control group, the liver of the ARC-treated rats had a greater degree of lipid accumulation that even appears in the gross microscopic picture of liver tissue as a bright greasy enlarged liver. In contrast to the acrylamide group, the SPI encapsulated FA group evidently demonstrated a significant reduction in hepatosteatosis. In contrast to the ARC and ferulic encapsulated groups, the nano -SPI encapsulated FA group showed greater improvement and nearly full recovery from hepatosteatosis.

In reference to the immunohistochemistry analysis and apoptosis assessment, ARC exhibits a robust positive reactivity to caspase-3, compared to the control group. Comparing the SPI encapsulated FA group to the ARC group, the former showed lower caspase immunoreactivity. In contrast to the SPI encapsulated FA group, nano -SPI encapsulated FA -treated hepatic tissues demonstrated no immunoreactivity as seen in Fig. 9 (A, B,C, D).

Fig. 7
figure 7

Photomicrographs of liver slices from (A) control rats reveal normal hepatic tissue histological structure in the form of normal hepatocyte strands (H), nuclei (N), sinusoids (S) and central vein (CV); B1 rats that received acrylamide displaying degenerative alterations and necrotic changes of hepatocytes (yellow arrow heads), and hemorrhage (red arrow), B2 Steatohepatitis of ARC treated rat hepatocytes with fatty degeneration, steatosis up to cell ballooning (black arrow) and scattered inflammatory cell infiltrate with Kupffer cell hyperplasia (200x) (red arrow), rats pre-treated with SPI encapsulated FA group (C) showing improvement of histopathological picture with slight portal congestion (black arrow) and nano-SPI encapsulated FA group (D) showing dramatic improvement with no histological alterations. (Scale bar 50 μm)

Fig. 8
figure 8

Upper panel showing a gross picture of liver from different groups. The lower panel illustrates Oil red O staining of the liver in different experimental groups: (A) control group, (B) ACR treated group showing increased fatty tissue as appeared in gross picture by increase liver brightness and marked oil red oil stain, (C) SPI encapsulated FA group showing less staining, (D) Nano -SPI encapsulated FA group with great improvement and minimal stain

Fig. 9
figure 9

Immunohistochemical staining of the apoptotic marker Caspase − 3 in liver sections from experimental groups (x400), appeared as brown cytoplasmic immunoreactions and counterstained with Hematoxylin.; Control group (A) showing few scanty immunoexpression of Caspase − 3 (arrows). ACR treated group (B) showing strong positive immunoexpression of Caspase − 3 (arrows) within the liver cells indicating increased cell apoptosis. SPI encapsulated FA group (C) showing a few scanty immunoexpression of Caspase − 3 within the hepatic cells. nano -SPI encapsulated FA group (D) showing more decrease in the expression of Caspase − 3 immunoexpression within most hepatic cells Scale bars = 50 μm

Discussion

Sesame protein isolate (SPI), prepared from defatted sesame flour, is utilized for human and animal nutrition due to its high-quality protein content [20]. SPI contains about 89.4 ± 0.36% proteins as SPI (88.98 ± 0.13%) recorded by Mustafa & Hüseyin [53] who reported 88.98 ± 0.13% protein content. The formation of nano-SPI is due to the utilization of ultrasonic waves, which produce high-temperature, high-pressure, and high-frequency vibrations. These vibrations rapidly change the structural and functional properties of proteins through mechanical and cavitation effects. Ultrasonic waves can also disrupt peptide bonds, induce subunit dissociation or aggregation, and obstruct noncovalent interactions between natural protein molecules [21].

The spectrum of SPI was composed of several bands located at 3273, 2962 cm⁻¹ which are assigned to the stretching vibration of OH groups, C-H groups. Bands at 1669, 1632, 1537 cm⁻¹ correspond to peptide group segments (the amide I band C = O stretching, and amide II bands C-N stretching and N-H stretching) respectively [54, 55]. Similar bands have been observed in other types of proteins as whey protein isolates (WPI) [56]. The most sensitive spectral region is the amide I band (1700–1600 cm⁻¹), which is sensitive to protein secondary structural components [57]. The spectrum of SPI nano form showed a similar pattern of bands as those of SPI, but with a change in band intensity. This could be due to the exposure of the protein to ultrasonic waves, which formed nanoparticles with more functional groups than SPI. The FTIR spectrum of both encapsulated forms shows some frequency and intensity differences, suggesting some alterations in the amino acid contributions and protein conformation due to addition of FA and sodium alginate.

SEM and POM confirmed that the ultrasonic waves applied to SPI successfully prepared the SPI nanoparticles. Particles between 1 and 1000 nm in size are known as polymeric nanoparticles (NPs), and they can have active substances either surface-adsorbed onto or trapped inside the polymeric core [58].

Encapsulation efficiency (EE) of nano SPI encapsulated was higher than that of SPI encapsulated, illustrating the importance of nano form particles. Antioxidants for the soluble capsules also showed that the scavenging activity of DPPH and ABTS percentage was higher in SPI encapsulated than in nano-SPI encapsulated form, which correlates with the encapsulation efficiency. This suggests that the release of FA is faster and higher in SPI capsules than nano SPI-capsules. Encapsulation allows for additional coating or surface alterations of the biomaterial in addition to enclosing the agent in its inner structure to protect the encapsulated materials [59].

This study investigated the hepatoprotective effects of FA encapsulated with sesame protein, both in its capsulated and nano forms, in combating ACR-induced liver toxicity. ACR, a widely recognized hydrophilic toxicant present in various foods, is known for its ease of tissue permeability upon ingestion, with the liver particularly vulnerable due to its central role in metabolism [60]. Our results indicate that both FA formulations encapsulated with sesame protein successfully reduce ACR-induced hepatic damage in rats.

ACR exposure compromises antioxidant defenses by promoting reactive oxygen species (ROS) generation, leading to oxidative stress and lipid peroxidation, contributing to cellular damage [61]. Consistent with this, oral administration of ACR in our study significantly elevated serum liver enzymes (AST, ALT, γ-GT, ALP), total and direct bilirubin, MDA, and NO while decreasing antioxidant markers such as GSH, GPx, catalase, and SOD. These findings align with previous studies demonstrating ACR’s role in ROS induction and oxidative stress [62, 63]. The increase in ROS also leads to cell membrane disruption, as evidenced by lipid peroxidation and MDA formation, which exacerbate inflammatory responses [64].

ACR-induced oxidative stress triggers inflammatory pathways, resulting in elevated levels of inflammatory cytokines TNF-α and IL-6, likely due to NF-κB pathway activation—a process supported by prior studies on oxidative stress and inflammation [65, 66]. Similarly, Nan et al. [67] found that ACR-induced liver inflammation elevated TNF-α and IL-1β levels via NF-κB signaling. However, the administration of FA encapsulated with sesame protein, particularly in nano form, significantly decreased these inflammatory cytokines, indicating anti-inflammatory effects.

Both FA formulations improved oxidative stress markers and liver function. Notably, the nano form exhibited enhanced efficacy, significantly raising antioxidant enzyme levels (SOD, GSH-Px, CAT) while lowering MDA and NO levels. This supports previous research showing FA’s antioxidant and anti-inflammatory properties via Nrf2 upregulation and NF-κB downregulation, promoting a balance between oxidative and inflammatory pathways [68]. The phenolic structure of FA further facilitates its free radical scavenging capabilities, enhancing its effectiveness against oxidative stress and liver damage [69].

The role of sesame protein as an encapsulation medium also contributed to the observed hepatoprotective effects. Sesame protein, rich in sulfur-containing amino acids such as methionine, supports antioxidant defenses by potentially preserving antioxidant enzyme activity [70, 71]. A study by Yang et al. [72] showed that sesame protein hydrolysates have antioxidative and anti-inflammatory properties, which likely potentiated FA’s effects on liver enzyme and bilirubin levels in this study. These findings are further corroborated by a study from Wang [73], which illustrates FA derivatives’ ability to restore liver function, lessen liver injury, and alleviate fibrosis. Importantly, our study showed that nanoencapsulation further enhanced FA’s hepatoprotective effects, likely due to improved bioavailability and efficacy, consistent with a previous report on the biocompatibility and efficiency of lipid-based nanoparticles [71].

Histopathological findings in this study proved that liver exposure to ACR induces hepatic toxicity in the form of steatosis and fatty degeneration up to cell ballooning which was in agreement with Liu et al. [74], who reported that exposure to ACR could induce oxidative stress to the liver and disrupt the metabolism of lipids (including cholesterol metabolism, sphingolipid metabolism, glycerophospholipid metabolism and fatty acid β-oxidation metabolism). Other study revealed that ACR induces hepatic tissue alterations that were represented by light microscopy as fatty deposits, congested central vein and cell vacuolization [75].

The herein work claimed that ACR induces hepatic structure deterioration, the microscopical degenerations that show liver damage, such as the increased necrosis, inflammation and hemorrhagic areas were more common and apparent when compared to other groups which was consistent with Donmez et al. [76]. Moreover, our results were in agreement with Abdulal et al. [77] who reported that histopathological liver tissue examination showed that FA ameliorated the appearance of vacuolated cytoplasm, reduced apoptotic nuclei, and necrotic nodules in rat liver. Also, Junhui Yuan et al. found that FA reduced the degree of necrosis and bleeding and alleviated the pathological changes caused by acetaminophen in rat liver [78]. Previous study showed that SPI decreases membrane lipid peroxidation in rats [79]. Also, Li et al. reported that feeding rats with black and white sesame caused recovery from fatty liver which may be partially agree with our study [80].

Inflammation and oxidative stress are crucial for the pathological development of hepatic steatosis, which can lead to cirrhosis, non-alcoholic fatty liver disease, or non-alcoholic steatohepatitis [81]. Sesame derivatives such as SPI also have anti-inflammatory and ameliorate oxidative stress consequently improving liver functions and histopathological picture as well [82].

Caspases, also referred to as cysteine proteases, are members of the interleukin-1β-converting enzyme family and are crucial for inducing apoptosis. The mammalian caspase family comprises 14 members, referred to as caspase-1-14. While some caspases are essential for apoptosis, others are not. The majority of caspases are involved in inflammation, differentiation, proliferation, or cell survival [83]. Either an intrinsic or an extrinsic pathway can trigger apoptosis in animals. Each of these routes activates the downstream effector caspase-3. It is essential to the cell apoptosis execution phase [84]. Oxidative stress may activate caspases, such as caspase-3, which would promote cell death [85].

Yaun et al. results are in line with the present work as Caspase-3 activity detection showed that FA pretreatment decreased the degree of apoptosis significantly in acetaminophen-induced hepatotoxicity in mice [86]. The present results are in accordance with Luo et al. [87] who reported that FA ameliorate diabetes induced hepatosteatosis in rats.

Additionally, by lowering hydroxyl radicals, peroxynitrite, superoxide anion, and nitric oxide, SPI reduced the production of free radicals. In hepatic damage, it preserved GSH and reduced lipid peroxidation [88]. Proteins and peptides derived from sesame seeds have been reported for medicinal properties, including antioxidant [89]. According to Adebisi et al., SPI provides major pharmacological advantages and health benefits for the entire body, particularly the liver [90]. The same author stated that Sesame proteins contain anti-inflammatory, anti-apoptotic and antioxidant ingredients that may support our findings.

Our findings uncover the genotoxic effects of ACR on hepatic DNA and the potential protective role of SPI-encapsulated FA and its nano-encapsulated form. The results revealed significant DNA fragmentation and damage in ACR-exposed hepatocytes, as evidenced by both the agarose gel electrophoresis and the comet assay. Treatment with SPI-encapsulated FA and nanoform significantly mitigated this damage, with nano-encapsulated form exhibiting superior protection. These findings align with the study of Eisenbrand [91] on ACR-induced genotoxicity and the protective effects of antioxidants. The DNA fragmentation observed in the ACR group supports previous studies indicating that ACR and its metabolite glycidamide can induce significant DNA damage. According to Tareke et al. [92], glycidamide forms DNA adducts contributing to mutations and carcinogenesis. Moreover, several studies reported DNA strand breaks and oxidative stress in hepatocytes following ACR exposure [93, 94] aligned with the significant smear pattern observed in the DNA bands and increased tail length in comet assay in the present study. FA is known for its antioxidant and free radical scavenging properties [95].

Previous studies by Ou and Kwok [96] demonstrated that FA reduces oxidative damage by neutralizing ROS and protecting cellular macromolecules. However, its limited bioavailability restricts its efficacy. In this study, the encapsulation of FA within sesame protein significantly enhanced its protective effect, as reflected in the reduced DNA fragmentation and comet assay parameters. These results are consistent with Garav and his colleagues [97] revealed that encapsulating phenolic compounds enhances their stability, bioavailability, and antioxidant capacity, which aligns with our findings. The superior protective effect of nano-encapsulated form compared to encapsulated form can be attributed to the enhanced cellular uptake and prolonged antioxidant activity provided by the nano-encapsulation process. Nano-encapsulation improves the solubility and bioavailability of FA, allowing for more efficient delivery and sustained protection against oxidative stress and DNA damage. Our results are in harmony with the study of Zhang et al. [98] who reported that nano-carrier systems significantly improve the therapeutic efficacy of antioxidants in protecting DNA integrity.

The protective effects of both encapsulated forms can be explained by their ability to reduce oxidative stress and prevent the formation of DNA adducts. The antioxidant activity of FA likely counteracts the ROS generated by ACR metabolism, thereby preventing oxidative damage to DNA. Additionally, sesame protein may contribute to this effect through its own antioxidant properties, as suggested by Kim et al. [99].

Conclusion

This study suggests the potential of using FA encapsulated within SPI, particularly in its nano form, for a protective effect against ACR-induced liver toxicity and genotoxicity. By targeting the oxidative stress induced by ACR, the research indicates potential hepatoprotective and genoprotective effects of this nutraceutical in this model. The nanoencapsulation appears to enhance the bioavailability and antioxidant activity of FA, potentially offering a promising intervention for mitigating liver injuries caused by environmental and dietary ACR exposure.

Study limitation: The use of a rat model may not fully translate to human responses, and the acute duration restricts the understanding of chronic effects of acrylamide exposure. Notably, the study did not explore the impact of varying dosages of the encapsulated FA, which is crucial for determining optimal therapeutic levels. Further research, including dose-response studies, human trials, and detailed mechanistic and pharmacokinetic analyses, is necessary to validate these findings for clinical translation.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Celik F, Cora T, Yigin A. Investigation of genotoxic and cytotoxic effects of acrylamide in HEK293 cell line. J Cancer Prev Curr Res. 2018;9:260–4.

    Article  Google Scholar 

  2. Tareke E, Rydberg P, Karlsson P, Eriksson S, Törnqvist M. Analysis of acrylamide, a carcinogen formed in heated foodstuffs. J Agric Food Chem. 2002;50:4998–5006. https://doi.org/10.1021/jf020302f.

    Article  CAS  PubMed  Google Scholar 

  3. Boon PE, de Mul A, van der Voet H, van Donkersgoed G, Brette M, van Klaveren JD. Calculations of dietary exposure to acrylamide. Mutat Res Genet Toxicol Environ Mutagen. 2005;580:143–55. https://doi.org/10.1016/j.mrgentox.2004.10.014.

    Article  CAS  Google Scholar 

  4. Ahmed MM, Hammad AA, Orabi SH, Elbaz HT, Elweza AE, Tahoun EA, et al. Reproductive injury in male rats from acrylamide toxicity and potential protection by earthworm methanolic extract. Anim (Basel). 2022;12:1723. https://doi.org/10.3390/ani12131723.

    Article  Google Scholar 

  5. Yan F, Wang L, Zhao L, Wang C, Lu Q, Liu R. Acrylamide in food: occurrence, metabolism, molecular toxicity mechanism and detoxification by phytochemicals. Food Chem Toxicol. 2023;175:113696.

    Article  CAS  PubMed  Google Scholar 

  6. Ghanayem BI, Witt KL, Kissling GE, Tice RR, Recio L. Absence of acrylamide-induced genotoxicity in CYP2E1-null mice: evidence consistent with a glycidamide-mediated effect. Mutat Res Fundam Mol Mech Mutagen. 2005;578:284–97.

    Article  CAS  Google Scholar 

  7. Alturfan AA, Tozan-Beceren A, Şehirli AÖ, Demiralp E, Şener G, Omurtag GZ. Resveratrol ameliorates oxidative DNA damage and protects against acrylamide-induced oxidative stress in rats. Mol Biol Rep. 2012;39:4589–96.

    Article  CAS  PubMed  Google Scholar 

  8. Bin-Jumah MN et al. Potential protective effects of Spirulina platensis on liver, kidney, and brain acrylamide toxicity in rats. Environ Sci Pollut Res. 2021.;28(21):26653-26663

  9. Li L, Sun HY, Liu W, Zhao HY, Shao ML. Silymarin protects against acrylamide-induced neurotoxicity via Nrf2 signalling in PC12 cells. Food Chem Toxicol. 2017;102:93–101.

    Article  CAS  PubMed  Google Scholar 

  10. Sengul E, Gelen V, Yildirim S, Cinar İ, Aksu EH. Effects of naringin on oxidative stress, inflammation, some reproductive parameters, and apoptosis in acrylamide-induced testis toxicity in rats. Environ Toxicol. 2023;38(4):798–808.

    Article  CAS  PubMed  Google Scholar 

  11. Kaur R et al. Natural products as sources of multitarget compounds: advances in the development of ferulic acid as multitarget therapeutic. Curr Top Med Chem. 2022.;22(5), 347-365

  12. Mahmoud AM, Hussein OE, Hozayen WG, Bin-Jumah M, Abd El-Twab SM. Ferulic acid prevents oxidative stress, inflammation, and liver injury via upregulation of Nrf2/HO-1 signaling in methotrexate-induced rats. Environ Sci Pollut Res Int. 2020;27(8):7910–21.

    Article  CAS  PubMed  Google Scholar 

  13. Mateo Anson N, van den Berg R, Havenaar R, Bast A, Haenen GR. Bioavailability of ferulic acid is determined by its bioaccessibility. J Cereal Sci. 2009;49:296–300.

    Article  CAS  Google Scholar 

  14. Saif A, et al. Recent advances in protein-based nanoparticles and their applications in the delivery of bioactive compounds. Int J Food Prop. 2023;26:2866–80.

    Article  CAS  Google Scholar 

  15. Demirhan E, Özbek B. Influence of enzymatic hydrolysis on the functional properties of Sesame cake protein. Chem Eng Commun. 2013;200(5):655–66.

    Article  CAS  Google Scholar 

  16. Kanu PJ, Kerui Z, Ming ZH, Haifeng Q, Kanu JB, Kexue Z. Functional properties of Sesame (Sesamum indicum L.) protein isolate as influenced by pH, temperature, time, and ratio of flour to water during its production. Asian J Biochem. 2007;2:289–301. https://doi.org/10.3923/ajb.2007.289.301.

    Article  CAS  Google Scholar 

  17. Chen Y, Lin H, Lin M, Zheng Y, Chen J. Effect of roasting and in vitro digestion on phenolic profiles and antioxidant activity of water-soluble extracts from Sesame. Food Chem Toxicol. 2020;139:111239. https://doi.org/10.1016/j.fct.2020.111239.

    Article  CAS  PubMed  Google Scholar 

  18. Mekky RH, Abdel-Sattar E, Segura-Carretero A, Contreras MD. Phenolic compounds from Sesame cake and antioxidant activity: A new insight for agri-food residues’ significance for sustainable development. Foods. 2019;8(10):432. https://doi.org/10.3390/foods8100432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tang CH. Nanostructured soy proteins: fabrication and applications as delivery systems for bioactives. Food Hydrocoll. 2019;91:92–116. https://doi.org/10.1016/j.foodhyd.2019.01.012.

    Article  CAS  Google Scholar 

  20. Poveda T, Vilcacundo R, Carpio C, Carrillo W. Analysis of Sesame proteins isolate (Sesamum indicum L.) with water and salt treatment. Asian J Pharm Clin Res. 2016;9(3):404–7.

    CAS  Google Scholar 

  21. Arzeni C, Martinez K, Zema P, Arias A, Perez OE, Pilosof AM. Comparative study of high-intensity ultrasound effects on food proteins functionality. J Food Eng. 2012;108(3):463–72. https://doi.org/10.1016/j.jfoodeng.2011.08.018.

    Article  CAS  Google Scholar 

  22. O’Sullivan J, Park M, Beevers J. The effect of ultrasound upon the physicochemical and emulsifying properties of wheat and soy protein isolates. J Cereal Sci. 2016;69:77–84. https://doi.org/10.1016/j.jcs.2016.02.013.

    Article  CAS  Google Scholar 

  23. Zhang QT, Tu ZC, Xiao H, Wang H, Huang XQ, Liu GX, et al. Influence of ultrasonic treatment on the structure and emulsifying properties of peanut protein isolate. Food Bioprod Process. 2014;92(C1):30–7. https://doi.org/10.1016/j.fbp.2013.07.006.

    Article  CAS  Google Scholar 

  24. Malekhosseini P, Alami M, Khomeiri M, Esteghlal S, Nekoei AR, Hosseini SM. Development of casein-based nanoencapsulation systems for delivery of Epigallocatechin gallate and folic acid. Food Sci Nutr. 2019;7(2):519–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mohamed RS, Fouda K, Akl EM. Hepatorenal protective effect of flaxseed protein isolate incorporated in lemon juice against lead toxicity in rats. Toxicol Rep. 2020;7:30–5.

    Article  PubMed  Google Scholar 

  26. El-Kholy WM, Soliman TN, Darwish AMG. Evaluation of date palm pollen (Phoenix dactylifera L.) encapsulation, impact on the nutritional and functional properties of fortified yogurt. PLoS ONE. 2019;14(10):e0222789. https://doi.org/10.1371/journal.pone.0222789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Younos M, Akl E. Evaluation of enzymatic phenolic extract from garden Cress seed meal against aflatoxigenic fungi isolated from eggplant fruits. Egypt J Chem. 2022;65(4):287–99.

    Google Scholar 

  28. Siles-Sánchez MN, Jaime L, Villalva M, Santoyo S. Encapsulation of marjoram phenolic compounds using Chitosan to improve its colon delivery. Foods. 2022;11(22):3657. https://doi.org/10.3390/foods11223657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. De Ancos B, Sgroppo S, Plaza L, Cano MP. Possible nutritional and health-related value promotion in orange juice preserved by high-pressure treatment. J Sci Food Agric. 2002;82(8):790–6. https://doi.org/10.1002/jsfa.1093.

    Article  CAS  Google Scholar 

  30. Re R, Pellegrini N, Proteggente A, Pannala A, Yang M, Rice-Evans C. Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radic Biol Med. 1999;26(9–10):1231–7.

    Article  CAS  PubMed  Google Scholar 

  31. Reeves PG, Nielsen FH, Fahey GC. AIN-93 purified diets for laboratory rodents: final report of the American Institute of nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J Nutr. 1993;123(11):1939–51. https://doi.org/10.1093/jn/123.11.1939.

    Article  CAS  PubMed  Google Scholar 

  32. Garber JC, Wayne Barbee R, Bielitzki JT, et al. Guide for the care and use of laboratory animals. 8th ed. Washington DC: National Academy of Sciences; 2011.

    Google Scholar 

  33. El-Shehawi AM, Sayed S, Hassan MM, Al-Otaibi S, Althobaiti F, Elseehy MM, Soliman M. Taify pomegranate juice (TPJ) abrogates acrylamide-induced oxidative stress through the regulation of antioxidant activity, inflammation, and apoptosis-associated genes. Front Veterinary Sci. 2022;9:833605.

    Article  Google Scholar 

  34. Jiang G, Lei A, Chen Y, Yu Q, Xie J, Yang Y, et al. The protective effects of the ganoderma atrum polysaccharide against acrylamide-induced inflammation and oxidative damage in rats. Food Funct. 2021;12:397–407. https://doi.org/10.1039/D0FO01873B.

    Article  CAS  PubMed  Google Scholar 

  35. Zhang N, Zhou J, Zhao L, Wang O, Zhang L, Zhou F. Dietary ferulic acid ameliorates metabolism syndrome-associated hyperuricemia in rats via regulating uric acid synthesis, glycolipid metabolism, and hepatic injury. Front Nutr. 2022;9:946556.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Chapman DG, Castillo R, Campbell JA. Evaluation of protein in foods: A method for rapid determination of protein efficiency ratio. Can J Biochem Physiol. 1959;37(5):679–86.

    Article  CAS  PubMed  Google Scholar 

  37. Essa HA, Hashim AF, Abdel-Aziz NN, Mohamed FE, Ali AM. Olive and linseed oil Blend‐Based nanoemulsions fortified with ginger extract nutraceutical: mitigating liver fibrosis induced by carbon tetrachloride by regulating oxidative stress and TGF‐β/MMP9 signaling pathway in rats. Mol Nutr Food Res. 2025;69(1):e202400497. https://doi.org/10.1002/mnfr.202400497.

    Article  CAS  PubMed  Google Scholar 

  38. Reitman S, Frankel S. A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. Am J Clin Pathol. 1957;28(1):56–63.

    Article  CAS  PubMed  Google Scholar 

  39. Bessey OA, Lowry OH, Brock MJ. A method for the rapid determination of alkaline phosphatase with five millimeters of serum. J Biol Chem. 1946;164:321–9. https://doi.org/10.1016/S0021-9258(18)43072-4.

    Article  CAS  PubMed  Google Scholar 

  40. Szasz G. A kinetic photometric method for serum gamma-glutamyl transpeptidase. Clin Chem. 1969;15:124–36. https://doi.org/10.1093/clinchem/15.2.124.

    Article  CAS  PubMed  Google Scholar 

  41. Rheinhold JG, Seligron D, editors. Total protein, albumin, and Globulin. Standard methods of clinical chemistry. New York: Academic; 1953. pp. 88–97.

  42. Doumas BT, Watson WA, Biggs HG. Albumin standards and the measurement of serum albumin with Bromcresol green. Clin Chim Acta. 1971;31(1):87–96. https://doi.org/10.1016/S0009-8981(96)06447-9.

    Article  CAS  PubMed  Google Scholar 

  43. Balistreri WF, Shaw LM. Liver function. In: Tietz NW, editor. Fundamentals of clinical chemistry. 3rd ed. Philadelphia: WB Saunders; 1987. pp. 729–61.

    Google Scholar 

  44. Nair V, Turner GA. The thiobarbituric acid test for lipid peroxidation: structure of the adduct with malondialdehyde. Lipids. 1984;19(10):804–5.

    Article  CAS  Google Scholar 

  45. Sun Y, Oberley LW, Li Y. A simple method for clinical assay of superoxide dismutase. Clin Chem. 1988;34(3):497–500.

    Article  CAS  PubMed  Google Scholar 

  46. Luck H. Catalase. In: Bergmeyer HU, editor. Methods of enzymatic analysis. New York: Academic; 1965.

    Google Scholar 

  47. Rotruck JT, Pope AL, Ganther HE, Swanson AB, Hafeman DG, Hoekstra WG. Selenium: biochemical role as a component of glutathione peroxidase. Science. 1973;179(4073):588–90. https://doi.org/10.1126/science.179.4073.588.

    Article  CAS  PubMed  Google Scholar 

  48. Jollow DJ, Mitchell JR, Zampaglione N, Gillette JR. Bromobenzene-induced liver necrosis: protective role of glutathione and evidence for 3,4-bromobenzene oxide as the hepatotoxic metabolite. Pharmacol. 1974;11:151–69.

    Article  CAS  Google Scholar 

  49. Montgomery H, Dymock J. The determination of nitrite in water: colorimetric method of nitric oxide assay. Analyst. 1961;86:414.

    CAS  Google Scholar 

  50. Kiernan J. Histological and histochemical methods: theory and practice. 5th ed. Bloxham: Scion Publishing Ltd; 2015.

    Google Scholar 

  51. Eckle V-S, Buchmann A, Bursch W, Schulte-Hermann R, Schwarz M. Immunohistochemical detection of activated caspases in apoptotic hepatocytes in rat liver. Toxicol Pathol. 2004;32:9–15.

    Article  CAS  PubMed  Google Scholar 

  52. Thapaliya S, Wree A, Povero D, Inzaugarat ME, Berk M, Dixon L, et al. Caspase 3 inactivation protects against hepatic cell death and ameliorates fibrogenesis in a diet-induced NASH model. Dig Dis Sci. 2014;59:1197–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mustafa Y, Hüseyin G. Sesame seed protein: amino acid, functional, and physicochemical profiles. Foods Raw Mater. 2023;11(1):72–83.

    Google Scholar 

  54. Guo Q, Su J, Shu X, Yuan F, Mao L, Liu J, Gao Y. Fabrication, structural characterization and functional attributes of polysaccharide-surfactant-protein ternary complexes for delivery of Curcumin. Food Chem. 2021;337:128019.

    Article  CAS  PubMed  Google Scholar 

  55. Zhang M, Li Z, Dai M, He H, Liang B, Sun C, et al. Fabrication and characterization of chitosan–pea protein isolate nanoparticles. Molecules. 2022;27(20):6913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Gbassi GK, Yolou FS, Sarr SO, Atheba PG, Amin CN, Ake M. Whey proteins analysis in aqueous medium and in artificial gastric and intestinal fluids. Int J Biol Chem Sci. 2012;6(4):1828–37.

    Google Scholar 

  57. Akyuz S, Akyuz T, Celik O, Atak C. FTIR spectroscopy of protein isolates of salt-tolerant soybean mutants. J Appl Spectrosc. 2018;84:1019–23.

    Article  CAS  Google Scholar 

  58. Carreiró F, Oliveira AM, Neves A, Pires B, Nagasamy Venkatesh D, Durazzo A, et al. Polymeric nanoparticles: production, characterization, toxicology, and ecotoxicology. Molecules. 2020;25:3731.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Mustafa AZ. Encapsulation importance in pharmaceutical area, how it is done and issues about herbal extraction. 2015;0–34.

  60. Abou-Seif HS, Hozayen WG, Hashem KS. Thymus vulgaris extract modulates dexamethasone-induced liver injury and restores the hepatic antioxidant redox system. Beni-Suef Univ J Basic Appl Sci. 2019;8:21. https://doi.org/10.1186/s43088-019-0021-0.

    Article  Google Scholar 

  61. Makhlouf H, Saksouk M, Habib J, Chahine R. Determination of antioxidant activity of saffron taken from the flower of Crocus sativus grown in Lebanon. Afr J Biotechnol. 2011;10:8093–100. https://doi.org/10.5897/AJB11.406.

    Article  Google Scholar 

  62. Rahbardar MG, Farmad HC, Hosseinzadeh H, Mehri S. Protective effects of selenium on acrylamide-induced neurotoxicity and hepatotoxicity in rats. Iran J Basic Med Sci. 2021;24(8):1041–9. https://doi.org/10.22038/ijbms.2021.55009.12331.

    Article  Google Scholar 

  63. Cerrah S, Ozcicek F, Gundogdu B, et al. Carvacrol prevents acrylamide-induced oxidative and inflammatory liver damage and dysfunction in rats. Front Pharmacol. 2023;14:1161448.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ybañez-Julca RO, Palacios J, Asunción-Alvarez D, UHPLC-ESI-MS/MS, et al. Q/Orbitrap/MS/MS. 2022;77(3):460–6. https://doi.org/10.1007/s11130-022-01000-2.

    Article  CAS  Google Scholar 

  65. Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2(1):17023–9. https://doi.org/10.1038/sigtrans.2017.23.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Hong Z, Minghua W, Bo N, et al. Rosmarinic acid attenuates acrylamide-induced apoptosis of BRL-3A cells by inhibiting oxidative stress and Endoplasmic reticulum stress. Food Chem Toxicol. 2021;151:112156. https://doi.org/10.1016/j.fct.2021.112156.

    Article  CAS  PubMed  Google Scholar 

  67. Nan B, Yang C, Li L, et al. Allicin alleviated acrylamide-induced NLRP3 inflammasome activation via oxidative stress and Endoplasmic reticulum stress in Kupffer cells and SD rats liver. Food Chem Toxicol. 2021;148:111937. https://doi.org/10.1016/j.fct.2020.111937.

    Article  CAS  PubMed  Google Scholar 

  68. Lampiasi N, Montana G. An in vitro inflammation model to study the Nrf2 and NF-κB crosstalk in presence of ferulic acid as modulator. Immunobiology. 2018;223:349–55.

    Article  CAS  PubMed  Google Scholar 

  69. Shu G, Tang Z, Du H, et al. Effects of dietary ferulic acid supplementation on hepatic injuries in Tianfu broilers challenged with lipopolysaccharide. Toxins (Basel). 2022;14(3):227. https://doi.org/10.3390/toxins14030227.

    Article  CAS  PubMed  Google Scholar 

  70. El-Shehawi AM, Sayed S, Hassan MM, et al. Taify pomegranate juice (TPJ) abrogates acrylamide-induced oxidative stress through the regulation of antioxidant activity, inflammation, and apoptosis-associated genes. Front Vet Sci. 2022;9:833605.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Colovic MB, Vasic VM, Djuric DM, Krstic DZ. Sulphur-containing amino acids: protective role against free radicals and heavy metals. Curr Med Chem. 2018;25(3):324–35.

    Article  CAS  PubMed  Google Scholar 

  72. Yang C, Li L, Ma Z, et al. Hepatoprotective effect of Methyl ferulic acid against carbon tetrachloride-induced acute liver injury in rats. Exp Ther Med. 2018;15(3):2228–38. https://doi.org/10.3892/etm.2017.5678.

    Article  CAS  PubMed  Google Scholar 

  73. Wang YY. Ferulic acid reduces liver fibrosis by inhibiting MAPK and NF-κB/IκBα signaling pathways in rats. Cent South Pharm. 2021;19(12):2489–95.

    Google Scholar 

  74. Liu Y, Wang R, Zheng K, et al. Metabonomics analysis of liver in rats administered with chronic low-dose acrylamide. Xenobiotica. 2020;50:894–905.

    Article  CAS  PubMed  Google Scholar 

  75. Hamdy SM, El-Khayat Z, Farrag AR, Sayed ON, El-Sayed MM, Massoud D. Hepatoprotective effect of raspberry ketone and white tea against acrylamide-induced toxicity in rats. Drug Chem Toxicol. 2022;45(2):722–30.

    Article  CAS  PubMed  Google Scholar 

  76. Donmez DB, Kacar S, Bagci R, et al. Protective effect of carnosic acid on acrylamide-induced liver toxicity in rats: mechanistic approach over Nrf2‐Keap1 pathway. J Biochem Mol Toxicol. 2020;34:e22524.

    Article  CAS  PubMed  Google Scholar 

  77. Abdulal ZA, Altahhan MY, Qindil AF et al. Ferulic acid inhibits tumor proliferation and attenuates inflammation of hepatic tissues in experimentally induced HCC in rats. J Investig Med. 2024;10815589241270488.

  78. Yuan J, Ge K, Mu J, et al. Ferulic acid attenuated acetaminophen-induced hepatotoxicity though down-regulating the cytochrome P450 2E1 and inhibiting toll-like receptor 4 signaling-mediated inflammation in mice. Am J Transl Res. 2016;8:4205.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Biswas A, Dhar P, Ghosh S. Antihyperlipidemic effect of Sesame (Sesamum indicum L.) protein isolate in rats fed a normal and high cholesterol diet. J Food Sci. 2010;75:H274–279.

    Article  CAS  PubMed  Google Scholar 

  80. Li C, Li Y, Ma Y, et al. Effect of black and white Sesame on Lowering blood lipids of rats with hyperlipidemia induced by high-fat diet. Grain Oil Sci Technol. 2020;3:57–63.

    Article  Google Scholar 

  81. Zheng W, Song Z, Li S, et al. Protective effects of Sesamol against liver oxidative stress and inflammation in high-fat diet-induced hepatic steatosis. Nutrients. 2021;13:4484.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Hadipour E, Emami SA, Tayarani-Najaran N, et al. Effects of Sesame (Sesamum indicum L.) and bioactive compounds (sesamin and sesamolin) on inflammation and atherosclerosis: A review. Food Sci Nutr. 2023;11:3729–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Li J, Yuan J. Caspases in apoptosis and beyond. Oncogene. 2008;27:6194–206.

    Article  CAS  PubMed  Google Scholar 

  84. Larsen BD, Sørensen CS. The caspase-activated DNase: apoptosis and beyond. FEBS J. 2017;284:1160–70.

    Article  CAS  PubMed  Google Scholar 

  85. Eskandari E, Eaves CJ. Paradoxical roles of caspase-3 in regulating cell survival, proliferation, and tumorigenesis. J Cell Biol. 2022;221:e202201159.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Ghorbani N, Yaghubi R, Davoodi J, et al. How does caspases regulation play role in cell decisions? Apoptosis and beyond. Mol Cell Biochem. 2024;479:1599–613.

    Article  CAS  PubMed  Google Scholar 

  87. Luo Z, Li M, Yang Q, et al. Ferulic acid prevents nonalcoholic fatty liver disease by promoting fatty acid oxidation and energy expenditure in C57BL/6 mice fed a high-fat diet. Nutrients. 2022;14:2530.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Idowu AO, Famuwagun AA, Fagbemi NT, et al. Antioxidant and enzyme-inhibitory properties of Sesame seed protein fractions and their isolate and hydrolyzate. Int J Food Prop. 2021;24:780–95.

    Article  CAS  Google Scholar 

  89. Liu BL, Chiang PS. Production of hydrolysate with antioxidative activity and functional properties by enzymatic hydrolysis of defatted Sesame (Sesamum indicum L). Int J Appl Sci Eng. 2008;6:73–83.

    Google Scholar 

  90. Adebisi AK, Stephen EC, Chinedu I, et al. Quantification of protein and amino acid composition in some oilseeds. Mol Biol. 2017;2:8–11.

    Google Scholar 

  91. Eisenbrand G. Revisiting the evidence for genotoxicity of acrylamide (AA), key to risk assessment of dietary AA exposure. Arch Toxicol. 2020;94:2939–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Tareke E, Rydberg P, Karlsson P, et al. Analysis of acrylamide, a carcinogen formed in heated foodstuffs. J Agric Food Chem. 2002;50:4998–5006.

    Article  CAS  PubMed  Google Scholar 

  93. Zhang X, Cao J, Jiang L, et al. Protective effect of Hydroxytyrosol against acrylamide-induced cytotoxicity and DNA damage in HepG2 cells. Mutat Res. 2009;664:64–8.

    Article  CAS  PubMed  Google Scholar 

  94. Zhang L, Yang L, Luo Y, et al. Acrylamide-induced hepatotoxicity through oxidative stress: mechanisms and interventions. Antioxid Redox Signal. 2023;38:1122–37.

    Article  CAS  PubMed  Google Scholar 

  95. Khan KA, et al. Ferulic acid: therapeutic potential due to its antioxidant properties, role in plant growth, and stress tolerance. Plant Growth Regul. 2024. https://doi.org/10.1007/s10725-024-01243-w.

    Article  Google Scholar 

  96. Ou S, Kwok KC. Ferulic acid: pharmaceutical functions, preparation, and applications in foods. J Sci Food Agric. 2004;84:1261–9.

    Article  CAS  Google Scholar 

  97. Garavand F, Jalai-Jivan M, Assadpour E, et al. Encapsulation of phenolic compounds within nano/microemulsion systems: A review. Food Chem. 2021;364:130376.

    Article  CAS  PubMed  Google Scholar 

  98. Zhang Z et al. Polyphenols as plant-based nutraceuticals: health effects, encapsulation, nano-delivery, and application. Foods. 2022;11.

  99. Kim JH, et al. Comparative assessment of compositional components, antioxidant effects, and Lignan extractions from Korean white and black Sesame (Sesamum indicum L.) seeds for different crop years. J Funct Foods. 2014;7:495–505.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

None.

Author information

Authors and Affiliations

Authors

Contributions

Hend A. Essa: proposed the main research point of the work. and conducted the animal experiment, biochemical analyses, and their statistical evaluation. Elham Ali: responsible for the methods, results, and statistical analysis of DNA fragmentation and the comet assay. Fatma El Zahraa Abd EL Hakam: responsible for the methods, results, of histopathological and immunohistochemical evaluations. Engy M. Akl: preparation of SPI, and its characterization, formation of both capsules then their evaluation.All authors contributed to writing and reviewing the manuscript.

Corresponding author

Correspondence to Hend A. Essa.

Ethics declarations

Ethical approval

This study was approved by the National Nutrition Institute Ethics Committee (ethical approval number: [IN000147]). Furthermore, the research protocol strictly adhered to the guidelines for the care and use of laboratory animals as established by the National Institutes of Health.

Confirmation that all authors have approved the manuscript for submission

All authors have approved the manuscript for submission.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Essa, H.A., Ali, E., Zahraa Abd EL Hakam, F.E. et al. Nano-encapsulated ferulic acid in sesame protein isolate alleviates acrylamide-induced liver toxicity and genotoxicity in rats via oxidative stress and DNA damage modulation. BMC Pharmacol Toxicol 26, 120 (2025). https://doi.org/10.1186/s40360-025-00946-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40360-025-00946-8

Keywords