Next Article in Journal
Recent Development of Exploring Ferroptosis-Inspired Effect of Iron as a Feasible Strategy for Combating Multidrug Resistant Bacterial Infections
Previous Article in Journal
Harnessing Wild Jackfruit Extract for Chitosan Production by Aspergillus versicolor AD07: Application in Antibacterial Biodegradable Sheets
Previous Article in Special Issue
Association Between Adherence Levels to the EAT-Lancet Diet in Habitual Intake and Selected Gut Bacteria in a Mexican Subpopulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Cardio-Protective Effects of Microencapsulated Probiotic and Synbiotic Supplements on a Myocardial Infarction Model Through the Gut–Heart Axis

by
Doha A. Mohamed
1,*,
Hoda B. Mabrok
1,
Hoda S. El-Sayed
2,
Sherein Abdelgayed
3,4 and
Shaimaa E. Mohammed
1
1
Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo 12622, Egypt
2
Dairy Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo 12622, Egypt
3
Department of Pathobiology (Anatomic Pathology), College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA
4
Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
*
Author to whom correspondence should be addressed.
Appl. Microbiol. 2025, 5(3), 72; https://doi.org/10.3390/applmicrobiol5030072
Submission received: 12 June 2025 / Revised: 23 July 2025 / Accepted: 24 July 2025 / Published: 27 July 2025

Abstract

Myocardial infarction (MI) is an inflammatory disease responsible for approximately 75% of sudden cardiac deaths. In this study, we aimed to evaluate the cardio-protective influence of microencapsulated probiotic and synbiotic dietary supplements in vivo and in molecular docking studies. MI was induced in rats with the injection of isoproterenol (i.p. 67 mg/kg). Plasma lipid profiles and the levels of oxidative stress markers, inflammatory markers, and cardiac enzymes were determined. The expression levels of MMP-7 and IL-1β in the heart muscle were measured. The impact of dietary supplements on fecal bacterial counts was evaluated across all rat groups. A histopathological examination of cardiac tissue was performed. The cardio-protective potential of cyanidin 3-diglucoside 5-glucoside and arabinoxylan was studied using molecular docking. The results demonstrate that all tested dietary supplements induced an improvement in all the biochemical parameters in association with an improvement in myocardial muscle tissue. The mRNA expression levels of MMP-7 and IL-1β were significantly downregulated by all dietary supplements. All dietary supplements increased the fecal counts of probiotic strains. In the molecular docking analysis, cyanidin 3-diglucoside 5-glucoside exhibited binding affinity values of −8.8 and −10 for lactate dehydrogenase (LDH) and Paraoxonase 1 (PON1), respectively. Arabinoxylan showed similar binding affinity (−8.8) for both LDH and PON1. Conclusion: Microencapsulated probiotic and synbiotic dietary supplements demonstrated notable cardio-protective influence in vivo and in molecular docking studies. These supplements may serve as promising candidates for the prevention of myocardial infarction.

Graphical Abstract

1. Introduction

Cardiovascular diseases (CVDs) are a group of disorders that affect the heart and blood vessels. CVDs such as coronary heart disease, atherosclerosis, and myocardial infarction (MI) account for 32% of global deaths and are expected to affect up to 23.3 million people by the year 2030 [1]. MI is an inflammatory disease. It is a common cause of fibrosis, with a high incidence, high mortality rate, and high recurrence rate, accounting for approximately 75% of sudden cardiac deaths [2,3,4,5]. Inflammation and oxidative stress play an important role in the incidence of MI [6]. Therefore, intervention with dietary supplements that possess anti-inflammatory and antioxidant activities is a good strategy for the prevention of MI. Plants have been an important source of natural products since ancient times. Plants contain many phytochemicals as secondary metabolites, such as phenolic compounds, polyphenols, flavonoids, and anthocyanins, which are well known to possess potent antioxidant and anti-inflammatory activities [7,8]. In previous work, an anthocyanin-rich extract of red cabbage (ARERC) showed cardio-protective potency against isoproterenol-induced MI in a rat model through alleviation of inflammation, reducing oxidative stress and protecting cardiac tissue [9]. Red cabbage is a rich source of anthocyanins such as cyanidin 3-diglucoside 5-glucoside and cyanidin 3-glucoside [10].
Consumption of dietary fiber such as psyllium husk has been associated with a reduction in inflammation in healthy individuals and patients suffering from chronic diseases [11,12,13]. Psyllium husk (PH) is one of the main sources of soluble mucilaginous dietary fiber [14]. Arabinoxylan comprises 55–60% of PH polysaccharides. Arabinoxylan exhibits many biological activities, such as hypolipidemic, antioxidant, and anti-diabetic activities [15]. Dietary fiber intake showed a beneficial effect on reducing the risk of CVDs and MI through decreasing the levels of inflammatory markers (C-reactive protein and tumor necrosis factor-α) [12,16]. Gut microbiota are microorganisms, including bacteria, archaea, fungi, and viruses, that live in the digestive tract of humans and play an important role in the immune system, mental health, and offering the body nutrients essential for growth and development [17,18,19,20]. Dietary fiber, which plays the role of a prebiotic, enhances the beneficial effects of the gut microbiota through increasing the production of beneficial compounds such as short-chain fatty acids, resulting in the suppression of inflammatory cytokine production and maintenance of host immune homeostasis [21,22,23,24]. Gut dysbiosis is associated with the incidence of many cardiovascular diseases, such as atherosclerosis, arrhythmia, and MI [5,19]. Gut dysbiosis not only raised the incidence of MI but also influenced cardiac repair after MI [25]. Probiotic and dietary supplements could recover cardiac function and avoid adverse alterations in experimental MI models [2,26,27,28]. Encapsulation of probiotics enhances their effects and protects them from pH, enzymes, bile salts, and other stressful conditions [29]. Also, encapsulation of probiotics with prebiotics such as inulin or polyphenols increases probiotics’ viability in the gastrointestinal tract [30]. Synbiotics exhibited more beneficial effects than probiotics or prebiotics alone, thus exhibiting synergistic effects [31]. The significant impact of dietary fiber—particularly soluble fiber—on improving gut microbiota, along with our previous findings on the cardio-protective effects of ARERC against myocardial infarction, motivated us to investigate the combined use of ARERC and PH, a rich source of soluble dietary fiber, with probiotics for the development of encapsulated synbiotics. Specifically, this study aimed to develop three encapsulated dietary supplements designed to enhance probiotic viability, ensure targeted intestinal release, and achieve synergistic bioactivity. Two of these were synbiotic formulations: one combining a probiotic with an anthocyanin-rich red cabbage extract (Synbiotic I), and the other with psyllium husk (Synbiotic II). The third consisted of an encapsulated probiotic alone. The cardioprotective effects of these formulations were evaluated using a rat model of isoproterenol-induced myocardial infarction, made more clinically relevant through the incorporation of thermally oxidized oil to mimic oxidative stress and better reflect human cardiovascular risk factors. Additionally, the cardio-protective potential of cyanidin 3-diglucoside 5-glucoside and arabinoxylan—key components of ARECR and PH, respectively—was further explored through molecular docking analysis.

2. Materials and Methods

2.1. Materials

Psyllium husk and red cabbage were obtained from local markets in Giza, Egypt. Red cabbage was kept at 4 °C till extraction.

2.2. Animals

Male adult Sprague Dawley rats of weight ranging between 208 and 273 g and aged 10–12 weeks were used. The animals were housed individually in stainless steel cages at a room temperature of 25 ± 2 °C and a relative humidity of about 55%; water and food were given ad libitum. The animals were provided by the animal house of the National Research Centre (NRC), Egypt. This study was carried out as a part of internal project No. 13050203 in the NRC, which was authorized by the Medical Research Ethics Committee, NRC, with approval number 13050203, and followed the recommendations of the National Institutes of Health Guide for the Care and Use of Laboratory Animals (publication No. 85-23, revised 1985).

2.3. Animals’ Diet

Two balanced diets were prepared in the current research study. Both diets were identical in calories and all ingredients (12.5% casein, 68% carbohydrate, 1% vitamin mixture, 3.5% salt mixture, and 5% fiber), except for the oil used: In the first balanced diet, it was palm oil (10%), while the second balanced diet contained thermally oxidized palm oil (10%) [32]. Thermally oxidized oil was incorporated into the diet of myocardial infarction rats to simulate dietary oxidative stress and amplify clinical relevance, making the model more representative of human cardiovascular risk profiles. All constituents were blended for the preparation of both balanced diets to be fed to the experimental rats. Salt and vitamin mixtures were prepared according to AIN-93 [33]. A balanced diet containing thermally oxidized oil was used for feeding the myocardial infarction model rats.

2.4. Extraction of Anthocyanin-Rich Extract from Red Cabbage

Following the washing process, the red cabbage was chopped into small pieces to extract the anthocyanin-rich extract; for extraction, we used 70% ethanol containing 0.5% citric acid, as described by Mohamed et al. [9]. A rotary evaporator was used to evaporate the solvent; then, the resulting extract was stored in the refrigerator until further use.

Total Phenolic, Total Flavonoid, and Total Anthocyanin Content Determination in Anthocyanin-Rich Extract

Phenolic content was assessed colorimetrically in anthocyanin-rich extract using Folin–Ciocalteu reagent. Absorbance was measured at 765 nm using a UV-visible spectrophotometer. The total phenolic content was expressed as gallic acid equivalent (GAE) in mg/100 g extract [9]. Total flavonoid content was determined by aluminum chloride assay in anthocyanin-rich extract and absorbance was measured at 415 nm using a spectrophotometer. The total flavonoid content was expressed as quercetin (QE) equivalent in mg/100 g. Total anthocyanin as cyanidin-3-O-glucoside (Cy3G) was measured [9], and total anthocyanin as cyanidin 3-diglucoside 5-glucoside (Cy3diG-5G) was measured as well. Absorption of the extracts at 530 and 650 nm was measured using a spectrophotometer. The molar absorbance (ε) coefficient used in calculation for cyanidin-3-O-glucosidei was 26,900 and ε for cyanidin 3-diglucoside 5-glucoside was 30,247 L/mol/cm. Total anthocyanin content was expressed as Cy3G equivalent in mg/100 g extract and total anthocyanin content was expressed as Cy3diG-5G equivalent in mg/100 g extract.

2.5. Bacterial Strains

All probiotic strains, including Bifidobacterium bifidum NRRL B-41410, Lactobacillus helveticus CNRZ32, and Lactobacillus casei FEGY 9973, were provided by the Dairy Department at the National Research Centre (Dairy Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki 12622, Cairo, Egypt).

2.6. Preparation of Different Probiotic and Synbiotic Dietary Supplements as Encapsulated Powders

The microencapsulation of probiotic strains was carried out using the freeze-drying technique. Initially, each probiotic strain (Bifidobacterium bifidum NRRL B-41410, Lactobacillus helveticus CNRZ32, and Lactobacillus casei FEGY 9973) was individually activated using MRS broth to achieve high biomass production and incubated anaerobically at 37 °C for 24 h. The cultures were then centrifuged at 6000 rpm for 15 min at 4 °C to harvest the cells. The resulting cell pellets from each strain were then combined and washed with sterile saline solution 0.9%, followed by lyophilization (freeze-drying) at −80 °C under vacuum pressure of 0.1 mbar for 48 h to produce microencapsulated powder, as described by Mahmud et al. [34]. Additionally, a sodium alginate solution was used as the coating material by dissolving 20 g in 1000 mL of sterilized water under continuous stirring until dissolved, then it was sterilized by autoclave for 15 min at 121 °C. The three different microencapsulated probiotics and synbiotics were prepared as detailed below.

2.6.1. The Encapsulated Synbiotic Containing Anthocyanin-Rich Extract of Red Cabbage (Synbiotic I) Dietary Supplement

One liter of sodium alginate solution was fortified with 40 g of probiotic cells and 18 g of the anthocyanin-rich extract of red cabbage. The mixed suspension was stirred well with a magnetic stirrer for 15 min. The final concentration that was orally administered to each rat was 200 mg/kg RBW anthocyanin-rich extract of red cabbage and 108 CFU probiotics in 1 mL of oral solution/day.

2.6.2. The Encapsulated Synbiotic Containing Psyllium Husk (Synbiotic II) Dietary Supplement

One liter of sodium alginate solution was fortified with 40 g probiotic cells and 16 g psyllium husk. The mixed suspension was stirred well using a magnetic stirrer for 15 min. The final concentration that was orally administered to each rat was 100 mg/kg RBW psyllium husk and 108 CFU probiotics in 1 mL of oral solution/day.

2.6.3. The Encapsulated Probiotic Dietary Supplement

One liter of sodium alginate solution was fortified with 40 g probiotic cells only and stirred well with a magnetic stirrer for 15 min. The final concentration that was orally administered to each rat was 108 CFU probiotics in 1 mL of oral solution/day.
Before freezing, the resultant mixtures were homogenized at 9000 rpm for 1 min at 40 °C. Each mixture was frozen at −20 °C for 24 h and then freeze-dried using a lyophilizer at −18 °C for one day; finally, the mixtures were placed in a freeze-drier (Labconco freeze dryer, Console, Kansas City, MO, USA; 0.1 mbar) for 24 h at −80 °C until being converted into microcapsule powders.

2.6.4. Studying the Cardio-Protective Influence of Encapsulated Probiotic and Synbiotic Dietary Supplements in Rat Model of Myocardial Infarction

After acclimatizing for 7 days, thirty male rats were distributed into five groups (six rats/group). The first group was the normal control group, where rats were fed a balanced diet for 21 days. The other four groups were fed a balanced diet containing thermally oxidized oil for 21 days. The second group was the myocardial infarction control (MI) group. Rats of the third group were administered a daily oral dose of encapsulated synbiotic I dietary supplement (200 mg/kg rat body weight and 108 probiotics/rat/day) for 21 days. Rats of the fourth group were orally administered the encapsulated synbiotic II dietary supplement (100 mg/kg rat body weight and 108 probiotics/rat/day) for 21 days. The fifth group of rats was given a daily oral dose of encapsulated probiotic dietary supplement (108 probiotic/rat/day) for 21 days. On the 21st day, a single dose of isoproterenol solution in saline (67 mg/kg) (Santa Cruz Biotechnology, Dallas, TX, USA) was injected subcutaneously [35] into all rats, except those in the normal control group, to induce MI. The body weight was recorded weekly, and the final body weight of the rats was recorded at the end of the experimental period. Blood samples were collected in heparinized tubes from fasting rats. Plasma samples were separated for the determination of the plasma lipid profile (total cholesterol (T-ch), triglycerides (TG), and high-density lipoprotein cholesterol (HDL-Ch)) using colorimetric kits. Low-density lipoprotein cholesterol (LDL-Ch) was calculated (LDL-Ch = Tc-HDL-c- (TG/5)). In addition, very-low-density lipoprotein cholesterol (VLDL-Ch) was also calculated (VLDL-Ch = triglycerides/5). The levels of oxidized LDL (Ox-LDL) (ELISA kit, Catalog # SL0554Ra, Sunlong®, Hangzhou, China) and Paraoxonase 1 (PON1), an enzyme linked to CVD (ELISA kit, Catalog #SL1304Ra, Sunlong®), were determined using an ELISA kit. Plasma SOD (ELISA kit, Catalog # SL1341Ra, Sunlong®) was evaluated as an indicator of antioxidant status. Plasma TNF-α (ELISA kit, Catalog # SL0202Ra Sunlong®) and C-reactive protein (CRP) (ELISA kit, Catalog # SL0202Ra Sunlong®) were determined as inflammatory markers. The plasma activities of the cardiac marker enzymes lactate dehydrogenase (LDH) [36], aminotransferase (ALT and AST) [37], and creatine kinase-MB (CK-MB) [38] were evaluated. Plasma creatinine [39] and urea [40] were evaluated as kidney function indicators. The heart was dissected from all rats after anesthesia and scarification and weighed according to the following formula: Relative heart weight = Absolute heart weight (g) × 100/final body weight (g). The hearts were collected from all rats and fixed in 10% buffered formalin for the preparation of paraffin wax sections for histopathological studies [41].

2.6.5. Evaluation of MMP-7 and IL-1β Gene Expression in Normal and Myocardial Rats

Total RNA was extracted from the frozen hearts with a PureLink®RNA Mini-Kit (ambion®Life technologiesTM, Carlsbad, CA, USA) according to the manufacturer’s protocol. The extracted RNA (1.5 µg) was reverse-transcribed into cDNA using a RevertAid first-strand cDNA synthesis kit (ThermoFisher®invitrogenTM, Wilmington, DE, USA) according to the manufacturer’s protocol. Real-time PCR was performed with a Rotor-Gene®MDx instrument (Qiagen, Düsseldorf, Germany). A total of 1 µL of cDNA as the template was amplified with EVA-Green Master Mix (HOT FIREPol EvaGreen qPCR Mix Plus, Solis BioDyneTM, Tartu, Estonia) in a 20 µL reaction mixture containing 4 µL of EVA-Green Master Mix, 0.25 µM primer pairs (Table 1), and PCR water. The reaction mixture was heated for 2 min at 50 °C and then at 95 °C for 12 min, followed by 45 cycles, including 95 °C for 20 s, 60 °C for 30 s, and 72 °C for 20 s. The final step was performing the melting curve program (60 to 95 °C). The relative expression of matrix metalloproteinase-7 (MMP-7) and interleukin-1 (IL-1β) was calculated using the 2−∆∆CT method [42]. The gene expression levels of MMP-7 and IL-1β were normalized to the expression of the housekeeping gene GAPDH.

2.7. The Microbiological Load in Fecal Samples of Different Groups

The microbiological load in rat fecal samples was determined after the preparation of serial decimal dilutions using 9 mL of sterile NaCl (0.85%); for the first dilution, we used 2% trisodium citrate, followed by normal decimal saline solution. The total probiotic counts were determined using MRS agar, and the plates were incubated at 37 °C/47 h under anaerobic conditions [45]. The total bacterial counts were determined following aerobic incubation based on the plate count agar method at 25 °C/48 h [46]. Coliform groups were detected with Violet Red bile Agar (Difco), and the plates were incubated at 35 °C/24 h according to the FDA [47]. Staphylococci sp. was detected by spreading 0.1 mL of suitable dilution onto the surface of a plate containing Baird Parker agar medium supplemented with egg yolk–potassium tellurite solution (Oxide); the plates were incubated at 37 °C/48 h [47]. The counts of Listeria sp. were performed using Oxford agar base supplemented with listeria supplement (Oxide); a volume of 0.1 mL of suitable dilution was spread on the surface of the plates with the medium and incubated at 35 °C/48 h [47].

2.8. Molecular Docking Study of Cyanidin 3-Diglucoside 5-Glucoside and Arabinoxylan with Lactate Dehydrogenase (LDH) and Paraoxonase 1 (PON1)

Ligand structures were retrieved from the PubChem database and prepared using Avogadro 1.2.0 software [48]. The structures of LDH and PON-1 were retrieved from the UniProt database. The UniProt ID for LDH is P04642, and that for PON-1 is P55159. The proteins (LDH and PON-1) were prepared for docking by using AutoDock Tools 1.5.7 [49]. Molecular docking studies were performed using AutoDock Vina [50] to predict the binding modes and affinity of the compounds for each protein. The docked complexes were visualized and analyzed using BIOVIA Discovery Studio Visualizer 2020 [51].

2.9. Statistical Analysis

Data were evaluated using one-way ANOVA followed by Tukey’s multiple comparison test with the SPSS version 26 statistical program. Differences were considered significant at p ≤ 0.05.

3. Results

3.1. Total Phenolic, Total Flavonoid, and Total Anthocyanin Content in Anthocyanin-Rich Extract

Total phenolic, total flavonoid, and total anthocyanin content is presented in Figure 1. Total phenolic content in anthocyanin-rich extract was 755.457 ± 2.453 mg GAE/100 g extract, while total flavonoid content was 1000.8 ± 3.545 mg QE/100 g extract. Total anthocyanin was measured equivalent to the two major compounds in the red cabbage according to the literature, total anthocyanin as cyanidin-3-O-glucoside (Cy3G) was 724.25 ± 1.254 mg Cy3G/100 g, and total anthocyanin as cyanidin 3-diglucoside 5-glucoside (Cy3diG-5G) was 946.94 ± 1.511 mg Cy3diG-5G/100 g.

3.2. Biochemical Analyses

The plasma lipid profile, oxidized-LDL, Paraoxonase 1 (PON1), and Paraoxonase 1/HDL-ch ratios (PON1/HDL-ch) of different groups are shown in Table 2. The subcutaneous injections of isoproterenol caused a significant increase in plasma TG, T-Ch, LDL-Ch, VLDL-Ch, and Ox-LDL in association with a significant reduction in HDL-Ch and PON1 in MI rats compared with normal rats. The oral administration of different dietary supplements significantly reduced the increment in the plasma lipid profile (T-Ch, TG, LDL-Ch and VLDL-Ch) and Ox-LDL and significantly increased the levels of HDL-Ch and PON1 in comparison with the myocardial infarction group.
Table 3 shows the levels of cardiac markers, creatinine, and urea of all rat groups. The plasma activities of the cardiac enzymes (CK-MB, LDH, ALT, and AST) were significantly increased in the MI group compared with the normal control group. The oral doses of the dietary supplements under study significantly reduced the increase in these cardiac enzymes in comparison with the MI group. The kidney function indicators (creatinine and urea) showed a significant increase in the MI control group compared with the normal control group (Table 3). The administration of different dietary supplements significantly reduced the increase in creatinine and urea in comparison with the MI control group.
In the present study, the inflammatory markers TNF-α and CRP (Table 4) were significantly elevated in the MI group compared with normal rats. SOD, an antioxidant enzyme, significantly decreased in the MI group compared with the normal control group. The oral administration of different dietary supplements caused a significant increase in the antioxidant enzyme SOD in association with a decrement in the inflammatory markers TNF-α and CRP compared with the MI group.

3.3. Impact of MI and Different Dietary Supplements on Nutritional Parameters

The impact of isoproterenol injection and different dietary supplements on relative heart weight, final body weight, and body weight gain are represented in Table 5. The rats in the MI group showed a significant increase in relative heart weight compared with the normal control group. The oral administration of different dietary supplements to rats reduced the increase in relative heart weight. The analysis of the final body weight revealed non-significant differences among all the study groups. The rats orally administered the synbiotic II dietary supplement showed the lowest weight gain among all the experimental groups.

3.4. Impact of MI and Different Treatments on the Gene Expression of MMP-7 and IL-1β Gene in Heart Tissues

The gene expression of MMP-7 and IL-1β was determined with RTPCR analysis (Figure 2). MMP-7 and IL-1β gene expression was significantly upregulated in the hearts of the myocardial infarction control group compared with the normal group. The encapsulated synbiotic I dietary supplement treatment significantly (p < 0.001) reduced the levels of MMP-7 and IL-1β by 91.24 and 72.03%, respectively, compared with the MI control group. The mRNA expression of MMP-7 and IL-1β was significantly (p < 0.001) downregulated by the encapsulated synbiotic II dietary supplement treatment by 92.72 and 70.34%, respectively. The treatment with encapsulated probiotic dietary supplements significantly (p < 0.001) decreased MMP-7 and IL-1β gene expression by 92.95 and 73.21, respectively, compared with the MI control group. The encapsulated probiotic alone or in the form of synbiotic I and II dietary supplement treatments had the significant effect of reversing elevated mRNA expression of MMP-7 and IL-1β.

3.5. The Microbiological Counts in Fecal Samples of Different Experimental Groups

The microbial counts in fecal samples from different groups are presented in Table 6. The counts of the probiotic strains loaded in encapsulated powder were increased in the rat groups that were orally administered different dietary supplements compared with the normal and MI control groups. In addition, encapsulated synbiotic I, containing the anthocyanin-rich extract of red cabbage, and synbiotic II, containing psyllium husk, increased this count more than the other formulations. Generally, the counts ranged between 4.67 and 8.70 log CFU/g, and the highest counts were recorded as 8.53 and 8.70 log CFU/g, without significant differences, in the fecal samples from groups orally administered synbiotics I and synbiotic II, respectively, followed by fecal samples from the encapsulated probiotic dietary supplement group (7.45 log CFU/g).
Conversely, the coliform counts were decreased in the fecal samples from rats given encapsulated synbiotic I, synbiotic II, and probiotics compared with the normal and MI control groups. The coliform counts were recorded as 5.93, 6.97, 4.91, 4.77, and 4.95 log CFU/g, respectively, for the normal, MI, synbiotic I, synbiotic II, and probiotic groups. The highest count of coliforms was recorded in the MI control group, followed by the normal group. Furthermore, the administration of either encapsulated synbiotic I or synbiotic II to rats resulted in the elimination of coliforms according to the counts in the fecal samples.
The same trend of results was noticed in the total bacterial counts. Specifically, diminished total bacterial counts were found in groups given encapsulated synbiotic I and synbiotic II, followed by the encapsulated probiotic, with values of 5.90, 5.96, and 6.88 log CFU/g, respectively. The highest total bacterial counts were reported in the normal and MI control groups without significant differences. The administration of encapsulated synbiotic I and synbiotic II to rats had the effect of reducing the bacterial counts by around 1.88 and 1.82 log cycles, respectively, compared with the MI control group.
In addition, the counts of the strain Staphylococci sp. declined in groups administered both encapsulated antibiotics and the encapsulated probiotic compared with the normal and MI control groups. These decreases were determined to be around 1.92, 1.24, and 1.47 log cycles, respectively, for the encapsulated synbiotic I and II and probiotic groups compared with the MI control groups. Furthermore, the Listeria sp. counts were found to be higher in the normal and MI control groups than in the groups that were orally administered encapsulated synbiotics I and II. The Listeria sp. counts were found to be 5.25, 7.16, 4.59, 4.58, and 4.72 log CFU/g for the normal, MI control, encapsulated synbiotic I, encapsulated synbiotic II, and encapsulated probiotic groups, respectively, with the counts in the latter three being around 2.57, 2.58, and 2.44 lower than that in the MI control group, respectively.

3.6. Histopathological Studies of Cardiac Tissue

The histopathological studies of cardiac tissue showed that the heart tissue of a rat from the normal group (Figure 3a) revealed normal myocardial muscle with normal striation and nucleation (H&E X400) (lesion score: 0). The hearts of rats from the myocardial infarction group (MI control) (Figure 3b) revealed myocardial hyalinosis and Zenker’s necrosis, together with muscle fragmentation, hemorrhage (arrow), and leucocytic cell infiltration (H&E X400) (lesion score: ++++). The hearts of rats orally administered encapsulated synbiotic I (Figure 3c) showed 90% greater lesion regression than the MI control group; the myocardial muscle was apparently healthy, with slight vacuolation (arrow), and the absence of hemorrhage and leucocytic cell infiltration was noticed (H&E X400) (lesion score: +++). The hearts of rats from the group pretreated with encapsulated synbiotic II (Figure 3d) showed 75% more lesion regression than the MI control group (Figure 3); improved myocardial muscle, the absence of hemorrhage, and mild leucocytic cell infiltration were noticed (H&E X400) (lesion score: ++). The hearts of rats from the group pretreated with the encapsulated probiotic (Figure 3e) showed 50% greater lesion regression than the MI control group; improved myocardial muscle, the absence of hemorrhage, and moderate leucocytic cell infiltration were noticed (arrow) (H&E X400) (lesion score: +). Figure 4 is present the improvement% in the heart lesion scoring compared with MI control.

3.7. Molecular Docking Study of Arbinoxylane and Cyanidin 3-Diglucoside 5-Glucoside with Lactate Dehydrogenase (LDH) and Paraoxonase 1 (PON1)

The 3D and 2D interactions and the binding affinity of arabinoxylan, cyanidin 3-diglucoside 5-glucoside, and cyanidin 3-glucoside for LDH and PON1 are shown in Table 7 and Table 8, respectively. The binding affinity (∆G) values of arabinoxylan and cyanidin 3-diglucoside 5-glucoside, −8.8 and 8.9 kcal/mol, respectively, were similar to that of the LDH enzyme. For the PON1 enzyme, cyanidin 3-diglucoside 5-glucoside had the highest binding affinity (−10 kcal/mol), followed by arabinoxylan (−8.8 kcal/mol). Arabinoxylan interacted with the active site of LDH at the GLY9, ASN113, HIS193, THR95, ASN138, ALA98, GLY29, THR248, and GLY246 residues with conventional hydrogen bonds and carbon–hydrogen bonds. Cyanidin-3-diglucoside 5-glucoside interacted with the active site of LDH at the THR248, GLN100, ASN138, ALA98, TYR239, ALA238, and ILE242 residues with Pi-Pi T-shaped, Pi-alkyl, and conventional hydrogen bonds. Arabinoxylan interacted with the active site of PON-1 at the ASP54, ILE170, ILE226, ILE121, GLU56, LEU230, and ILE271 residues with conventional hydrogen bonds and carbon–hydrogen bonds. Cyanidin-3-diglucoside 5-glucoside interacted with the active site of PON-1 at ILE271, GLU56, THR119, ASP169, ILE57, SER272, GLU56, ASP54, PRO275, VAL273, LEU230, ILE117, and IL170 with Pi-alkyl, Pi-anion, conventional hydrogen and carbon–hydrogen bonds.

4. Discussion

Cardiovascular diseases (CVDs) are a group of ailments that impact the heart and blood vessels. According to the World Health Organization, CVDs such as coronary heart disease, atherosclerosis, and MI are responsible for 32% of global deaths and are projected to affect up to 23.3 million individuals by 2030 [1,52]. Dietary habits play an important role in the increment in the prevalence of CVDs. Healthier dietary habits, such as a diet rich in dietary fiber, polyunsaturated fatty acids, and plant protein, as well as vegetables and fruits, exhibit cardio-protective potency, while bad dietary habits, such as the consumption of saturated fat, refined carbohydrates, animal protein (especially red meat), and fast foods, as well as low intake of vegetables and fruits, increase the risk of CVD incidence [53]. Thus, dietary interventions remain an important approach to the primary prevention of CVDs.
In the current research study, myocardial infarction was induced in rats as a model of CVD with the subcutaneous injection of a single dose of isoproterenol (67 mg/kg rat body weight), and the rats were fed a balanced diet containing thermally oxidized oil as a source of free radicals. It was previously reported [32,54] that a diet containing repeatedly heated oil leads to an increase in and the formation of toxic compounds such as triglycerides, dimers, polycyclic aromatic hydrocarbons, and oxidized fatty acids. These toxic compounds and free radicals exert harmful effects on the human body and lead to different chronic diseases, especially CVDs.
In the present study, oxidative stress (an increase in Ox-LDL and a reduction in PON1 and SOD) and inflammation markers (CRP and TNF-α) were elevated after the induction of MI with isoproterenol. The same was true for cardiac enzymes (CK-MB, LDH, AST, and ALT) and kidney function parameters (creatinine and urea). Moreover, the rats with ISO-induced MI showed dyslipidemia, as evidenced by increments in the plasma levels of T-ch, TG, LDL-ch, VLDL-ch, and T-ch/HDL-ch in association with a reduction in HDL-ch. Histopathological changes were observed in the heart tissue of the MI group. The mRNA expression of MMP7 and IL-1β increased in MI rats. The observed results are in agreement with those of previous studies [9,31,55,56,57] in which myocardial infarction was associated with different unhealthy conditions. Hence, these results show that the method for MI induction chosen in this study allowed us to obtain an ideal MI model.
The administration of encapsulated synbiotic I (containing an anthocyanin-rich extract of red cabbage), synbiotic II (containing psyllium husk), and probiotics to rats improves all the biochemical parameters and the gene expression of MMP7 and IL-1β in association with an improvement in the cardiac tissue histopathology to different degrees.
Red cabbage is a rich source of anthocyanins such as cyanidin 3-diglucoside 5-glucoside and cyanidin 3-glucoside [10]. It was previously reported that an anthocyanin-rich extract of red cabbage and psyllium husk exhibited a positive effect against cardiovascular diseases [9,12]. The alcoholic-rich extract of red cabbage contained 618.2 mg Cy3G/100 g total anthocyanins, as shown in our previous study [9], and displayed antioxidant and cardio-protective activities in a rat model of MI [9]. Anthocyanins also previously exhibited antiradical activity by scavenging reactive oxygen and nitrogen-free radicals [58,59].
Psyllium husk (PH) is a rich source of dietary fiber. Psyllium husk is one of the main sources of soluble mucilaginous dietary fiber [14]. Arabinoxylans comprise 55–60% of PH polysaccharides [14]. The consumption of dietary fiber, especially soluble mucilage, has been associated with a reduction in inflammation in healthy individuals and patients suffering from chronic diseases [12,13,60,61]. Dietary fiber intake showed the beneficial effect of reducing the risk of CVDs and MI through a reduction in the levels of inflammatory markers such as CRP and TNF-α [12,16]. The encapsulation of probiotics with the anthocyanin-rich extract of red cabbage (synbiotic I) and psyllium husk (synbiotic I), which are rich in bioactive and natural compounds, increased their protective effects against myocardial infarction.
The increase in relative heart weight in the groups of rats with MI observed in the present study is in accordance with previous studies [9,62,63] and may be due to increases in water, necrosis, and inflammation of cardiac cells [63]. The oral administration of synbiotic I, synbiotic II, or the encapsulated probiotic showed a significant reduction in the elevated relative heart weight compared with the myocardial infarction control group. This enhancement may be related to the improvement in the inflammation and oxidative stress status in the cardiac tissues indicated by the present histopathological results. The reduction in body weight gain in the rat group given an oral dose of encapsulated synbiotic II may be attributed to satiety due to the high viscosity of psyllium husk gel, which slows the transition time of food and promotes a feeling of fullness, which leads to a reduction in body weight gain. The same results were reported by Mohamed et al. [60] when obese rats were given oral doses of chia seed mucilage. Dietary fiber-rich food increases the time of mastication, which leads to the prolongation of oral exposure time and arbitrary satiety sensations. The increment in oral exposure time leads to a reduction in calorie intake and may be favorable to the generation of signals of gastric filling, which have also been shown to promote satiety [63,64].
The production of reactive oxygen species activates the transcription of matrix metalloproteinases (MMPs). MMPs are responsible for extracellular matrix (ECM) degradation and increase the risk of heart failure [65]. Matrix metalloproteinasis7 (MMP7) is regulated by TNF-α and IL-1β. Increasing the expression of TNF-α and IL-1β leads to the up-regulation of MMP7 [66]. MMP7 can play an important role in ECM and left ventricular remodeling and serves as a predominant mechanism for post-MI arrhythmia [67]. Thus, MMPs are considered targets for many cardiovascular diseases. The encapsulated symbiotics and probiotic under study containing the anthocyanin-rich extract of red cabbage, psyllium husk, and probiotics altered the levels of MMP-7, TNF-α, and IL1β, providing an important treatment strategy for MI. Emerging evidence suggests that beneficial shifts in gut microbiota can enhance the production of short-chain fatty acids (SCFAs) such as butyrate, which are known to inhibit the NF-κB signaling pathway—a key regulator of pro-inflammatory cytokines, including IL-1β and matrix metalloproteinases like MMP-7 [68]. Therefore, the observed reductions in MMP-7 and IL-1β in our study may be partially attributed to SCFA-mediated downregulation of inflammatory signaling.
The probiotic strains selected for this study—Bifidobacterium bifidum NRRL B-41410, Lactobacillus helveticus CNRZ32, and Lactobacillus casei FEGY 9973—were chosen based on prior reports demonstrating their potential health benefits. B. bifidum NRRL B-41410 has shown promising immunomodulatory and gut health-promoting properties. L. helveticus CNRZ32 is a well-characterized strain known for its ability to produce bioactive peptides with antihypertensive and anti-inflammatory effects. L. casei FEGY 9973, a locally isolated strain, has been previously evaluated in our laboratory for its probiotic characteristics, including acid and bile tolerance, adhesion capacity, and antimicrobial activity.
The encapsulated dietary supplements containing the anthocyanin-rich extract of red cabbage or psyllium husk and the microencapsulated probiotics can be considered cardio-protective agents against ISO-induced MI in rats. Encapsulated synbiotic I, which contained the anthocyanin-rich extract of red cabbage, was the most promising in exerting a protective effect on the heart muscle, as evidenced by the histopathological studies. In the present study, the microbial counts in fecal samples from different groups—including the normal rats, the MI group, and the rats administered encapsulated synbiotic I or II—revealed that the probiotic strain counts were elevated in the groups receiving dietary supplements compared with the normal and MI groups (Table 6). In addition, encapsulated synbiotics I and II, which contain the anthocyanin-rich extract of red cabbage and psyllium husk, respectively, increased the probiotic counts compared with the other groups. The encapsulation of probiotics enhances their effects and protects them from pH, enzymes, bile salts, and other stressful conditions [29]. Moreover, the different dietary supplements in the form of microcapsule powders containing probiotics, anthocyanin-rich extract of red cabbage, and psyllium husk produced using a lyophilization method, when passing through the gastrointestinal tract, can increase the stability of these natural compounds and probiotics by protecting them from adverse environmental effects because of the incorporation of a protective material such as sodium alginate [69,70,71]. In addition, these results indicate that feeding rats dietary supplements containing encapsulated probiotics promotes bacterial colonization in the gastrointestinal tract, especially in the presence of the anthocyanin-rich extract of red cabbage or psyllium husk. Previous studies demonstrated that the prebiotic impact of an anthocyanin-rich extract of red cabbage or psyllium husk enhanced the survival rate of probiotics and altered the gut microbiome [72,73,74,75,76]. Conversely, the coliform count decreased in the fecal samples of rats given encapsulated synbiotic I (containing the anthocyanin-rich extract of red cabbage), synbiotic II (containing psyllium husk), and probiotics compared with the other groups (normal and MI). The highest count of coliforms was recorded in the MI group, followed by the normal group. Moreover, the oral administration of encapsulated synbiotic I or II to rats eliminated the coliform load in the fecal samples. These results are related to the antagonistic effect of probiotics and show that the prebiotic agents (i.e., the anthocyanin-rich extract of red cabbage and psyllium husk) positively improved the probiotics’ survival and antimicrobial substance production. The beneficial effects of different probiotics combined with prebiotics (synbiotics) on improving rat health and providing protection against diseases are attributed to the ability of probiotics to inhibit pathogenic microorganisms, counteract harmful pathogens, alter the gut microbiome, and modulate the host’s immune response [76,77,78]. The same trend was noticed for the total bacterial counts, with diminished total bacterial counts being found in groups orally administered encapsulated synbiotic I or II, followed by the encapsulated probiotic. In addition, the counts of Staphylococci species were reduced in the groups that received probiotics compared with the normal and MI groups. A decrease was also detected in groups treated with the different dietary supplements compared with the MI control group. Furthermore, the Listeria sp. counts were reported to be higher in both the normal and MI control groups compared with all other groups. The above results of the microbiology load analyses on the fecal samples indicate that feeding rats synbiotic I and II positively enhanced the colonization of probiotics in the intestinal tract in high quantities and reduced the levels of other undesired bacteria. Numerous researchers have investigated probiotic bacteria for their potential to improve gastrointestinal health and support the function of various body organs in rats, including in conditions such as heart attacks. Important mechanisms of probiotics and prebiotics in the treatment of cardiovascular diseases include a reduction in oxidative stress, hypercholesterolemia, and high blood pressure [78,79,80,81,82,83].
The comparable readings observed for synbiotic I, synbiotic II, and the encapsulated probiotic alone may be attributed to the dominant effect of the probiotic component in modulating the measured outcomes. It is possible that, under the conditions of this study, the prebiotic components (arabinoxylan and cyanidin 3-diglucoside 5-glucoside) did not exert a significantly additive or synergistic effect beyond what was achieved by the encapsulated probiotic alone. This may be due to factors such as dose, bioavailability, or limited interaction between the prebiotic and probiotic in vivo. Additionally, encapsulation itself may have enhanced the stability and delivery of the probiotic, thereby maximizing its efficacy across all groups containing it. Therefore, further investigation is warranted to dissect the individual and combined contributions of each synbiotic component.
Based on literature reports, cyanidin-3-diglucoside-5-glucoside is identified as the major anthocyanin in red cabbage [84]; therefore, it was selected as the ligand for molecular docking studies. The molecular docking studies revealed high binding affinity of the major compounds present in psyllium husk (arabinoxylan) and the anthocyanin of red cabbage extract (cyanidin 3-diglucoside 5-glucoside) for LDH and PON1. LDH plays an important role in many chronic diseases, such as cardiovascular diseases. Elevated blood levels of LDH can be considered an indicator of tissue breakdown. In addition, the increment in the blood levels of LDH can be used as an indicator of cardiac inflammation [85]. Natural compounds such as polyphenols, flavonoids, and alkaloids can be used to inhibit the increase in LDH and improve chronic disease status [86]. In the present study, the anthocyanin-rich extract of red cabbage and psyllium husk induced a significant reduction in LDH plasma levels in rats. These inhibitory effects of both prebiotics used in the present research study were also confirmed by the results of the molecular docking study, which showed high binding affinity of the major compounds of the studied prebiotics for the LDH protein.
PON1 is one of the enzymes linked to cardiovascular diseases and plays an important role in the prevention of oxidative stress [87]. PON1 is inversely correlated with cardiovascular diseases [88]. PON1 antioxidant activity was discovered following the observation of the protection of LDL and HDL from oxidation and the release of biologically active oxidized lipids in lipoproteins and arterial cells, which led to the conclusion that this enzyme has antiatherogenic action [86]. In the present research study, all the studied treatments showed a significant increase in PON1 in comparison with the MI control group, which means that the studied antibiotics were effective as cardio-protective agents against isoproterenol-induced MI in rats. In addition, the arabinoxylan and cyanidin 3-diglucoside 5-glucoside showed high binding affinity for PON1 in the molecular docking study, which supported the results of the in-vivo experiment.
Limitations of the study: One of the most important limitations of the current research study is the absence of the 16S rRNA amplicon sequencing of colon contents. In future research, we will address this by incorporating 16S rRNA sequencing, alongside integrated multi-omics such as metabolomics and metagenomics, to analyze the gut microbiome and gain a more comprehensive understanding—particularly of the gut–heart axis. Furthermore, the relatively short duration of this study represents another limitation. Therefore, future studies will be designed with extended experimental periods. In the present study, the administration of synbiotic I and synbiotic II resulted in a significant elevation in plasma PON1 activity and reduction in LDH. This increase and decrease correlate with the molecular docking results. These findings suggest a potential mechanistic link between the high binding affinity observed in silico and the enhanced PON1 activity detected in vivo. However, we acknowledge that further mechanistic studies would be needed to confirm this relationship. We acknowledge the lack of full chemical profiling of anthocyanidin derivatives in the red cabbage extract and recognize that future studies should focus on the isolation and detailed characterization of these compounds.

5. Conclusions

The encapsulated synbiotic and probiotic dietary supplements investigated in this study exhibited cardio-protective influence in a rat model of MI. The cardio-protective potency of these dietary supplements was noticed as an improvement in dyslipidemia, a decrease in the high levels of cardiac enzymes, a reduction in inflammatory markers, and a reversal of the increase in the mRNA expression of MMP-7 and IL-1β. The administration of all dietary supplements improved the gut microbiota profile. In addition, the major compounds in red cabbage anthocyanin and psyllium husk proved to have high binding affinity for LDH and PON1 in the molecular docking studies.

Author Contributions

D.A.M. is the principal investigator of the project, designed the study, prepared the extract, and prepared the manuscript in the final form, including the interpretation of the results. H.B.M. evaluated all PCR analyses, gene expression studies, and molecular docking studies; performed statistical data analysis; and contributed to the preparation of the manuscript. H.S.E.-S. prepared the encapsulated probiotic and synbiotics, studied the fecal bacterial counts, and contributed to the preparation of the manuscript. S.A. performed the histopathological examination of cardiac tissue and contributed to the preparation of the manuscript. S.E.M. performed the animal experiment, evaluated all the biochemical analyses, entered the data on the Excel sheet and contributed to the preparation of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research study was funded by the National Research Centre, Egypt, research project number 13050203, and the APC was funded by D.A.M.

Institutional Review Board Statement

All animal procedures were performed in accordance with the Ethics Committee of the National Research Centre, Cairo, Egypt, with approval number 13050203 3 (6/2/2023) and following the recommendations of the National Institutes of Health Guide for the Care and Use of Laboratory Animals (publication No. 85-23, 1 January 2023, revised 1985).

Data Availability Statement

All data generated or analyzed during this study are included in this published article.

Acknowledgments

The authors express their sincere gratitude for the National Research Centre, Egypt, for funding the study through research project No. 13050203.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. World Health Organization (WHO). Cardiovascular Diseases (CVDs). 2021. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 11 June 2021).
  2. Bonab, S.F.; Tahmasebi, S.; Ghafouri-Fard, S.; Eslami, S. Preventive impact of probiotic supplements on heart injury and inflammatory indices in a rat model of myocardial infarction: Histopathological and gene expression evaluation. APMIS 2025, 133, e13479. [Google Scholar] [CrossRef]
  3. Shi, H.-T.; Huang, Z.-H.; Xu, T.-Z.; Sun, A.-J.; Ge, J.-B. New diagnostic and therapeutic strategies for myocardial infarction via nanomaterials. EBioMedicine 2022, 78, 103968. [Google Scholar] [CrossRef]
  4. Mu, F.; Zhao, J.; Zhao, M.; Lin, R.; Liu, K.; Zhao, S.; Tao, X.; Li, W.; Dai, Q.; Xi, M.; et al. Styrax (Liquidambar orientalis Mill.) promotes mitochondrial function and reduces cardiac damage following myocardial ischemic injury: The role of the AMPK-PGC1α signaling pathway. Pharm. Pharmacol. 2023, 75, 1496–1508. [Google Scholar] [CrossRef]
  5. Xu, H.; Yang, F.; Bao, Z. Gut microbiota and myocardial fibrosis. Eur. J. Pharmacol. 2023, 940, 175355. [Google Scholar] [CrossRef]
  6. Pereira, B.L.B.; Rodrigue, A.; Arruda, F.C.d.O.; Bachiega, T.F.; Lourenço, M.A.M.; Correa, C.R.; Azevedo, P.S.; Polegato, B.F.; Okoshi, K.; Fernandes, A.A.H.; et al. Spondias mombin L. attenuates ventricular remodelling after myocardial infarction associated with oxidative stress and inflammatory modulation. J. Cell. Mol. Med. 2020, 24, 7862–7872. [Google Scholar] [CrossRef]
  7. Pavithra, K.; Uddandrao, V.V.S.; Chandrasekaran, P.; Brahmanaidu, P.; Sengottuvelu, S.; Vadivukkarasi, S.; Saravanan, G. Phenolic fraction extracted from Kedrostis foetidissima leaves ameliorated isoproterenol-induced cardiotoxicity in rats through restoration of cardiac antioxidant status. J. Food Biochem. 2020, 44, e13450. [Google Scholar] [CrossRef]
  8. Palhares, R.M.; Drummond, M.G.; Brasil, B.; Cosenza, G.P.; Brandão, M.D.G.L.; Oliveira, G. Medicinal plants recommended by the world health organization: DNA barcode identification associated with chemical analyses guarantees their quality. PLoS ONE 2015, 10, e0127866. [Google Scholar] [CrossRef]
  9. Doha, M.; Hoda, M.; Sherein, A.; Hagar, E. Cardio-protective potency of anthocyanin-rich extract of red cabbage against isoproterenol-induced myocardial infarction in experimental animals. J. Appl. Pharm. Sci. 2021, 11, 22–30. [Google Scholar] [CrossRef]
  10. Jana, S.; Patel, D.; Patel, S.; Upadhyay, K.; Thadani, J.; Mandal, R.; Das, S.; Devkar, R.; Yenugu, S. Anthocyanin rich extract of Brassica oleracea L. alleviates experimentally induced myocardial infarction. PLoS ONE 2017, 12, e0182137. [Google Scholar] [CrossRef]
  11. Xu, S.; Cai, Y.; Hu, H.; Zhai, C. Correlation of visceral adiposity index and dietary profile with cardiovascular disease based on decision tree modeling: A cross-sectional study of NHANES. Eur. J. Med. Res. 2025, 30, 123. [Google Scholar] [CrossRef]
  12. Shivakoti, R.; Biggs, M.L.; Djoussé, L.; Durda, P.J.; Kizer, J.R.; Psaty, B.; Reiner, A.P.; Tracy, R.P.; Siscovick, D.; Mukamal, K.J. Intake and sources of dietary fiber, inflammation, and cardiovascular disease in older US adults. JAMA Netw. Open 2022, 5, e225012. [Google Scholar] [CrossRef]
  13. Dong, W.; Yang, Z. Association of Dietary Fiber Intake with Myocardial Infarction and Stroke Events in US Adults: A Cross-Sectional Study of NHANES 2011–2018. Front. Nutr. 2022, 9, 936926. [Google Scholar] [CrossRef]
  14. Bakr, A.F.; Farag, M.A. Soluble Dietary Fibers as Antihyperlipidemic Agents: A Comprehensive Review to Maximize Their Health Benefits. ACS Omega 2023, 8, 24680–24694. [Google Scholar] [CrossRef]
  15. Waleed, M.; Saeed, F.; Afzaal, M.; Niaz, B.; Raza, M.A.; Hussain, M.; Tufail, T.; Rasheed, A.; Ateeq, H.; Al Jbawi, E. Structural and nutritional properties of psyllium husk arabinoxylans with special reference to their antioxidant potential. Int. J. Food Prop. 2022, 25, 2505–2513. [Google Scholar] [CrossRef]
  16. Tong, T.Y.N.; Appleby, P.N.; Key, T.J.; Dahm, C.C.; Overvad, K.; Olsen, A.; Tjønneland, A.; Katzke, V.; Kühn, T.; Boeing, H.; et al. The associations of major foods and fibre with risks of ischaemic and haemorrhagic stroke: A prospective study of 418,329 participants in the epic cohort across nine European Countries. Eur. Heart J. 2020, 41, 2632–2640. [Google Scholar] [CrossRef]
  17. Das, S.; Khanna, C.; Singh, S.; Nandi, S.; Verma, R. Impact of human microbiome on health. In Microbial Diversity, Interventions and Scope; Springer: Singapore, 2020; pp. 349–373. [Google Scholar] [CrossRef]
  18. Kim, M.-S.; Kim, Y.; Choi, H.; Kim, W.; Park, S.; Lee, D.; Kim, D.K.; Kim, H.J.; Choi, H.; Hyun, D.-W.; et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut 2020, 69, 283–294. [Google Scholar] [CrossRef]
  19. Ma, J.; Hong, Y.; Zheng, N.; Xie, G.; Lyu, Y.; Gu, Y.; Xi, C.; Chen, L.; Wu, G.; Li, Y.; et al. Gut microbiota remodeling reverses aging-associated inflammation and dysregulation of systemic bile acid homeostasis in mice sex-specifically. Gut Microbes 2020, 11, 1450–1474. [Google Scholar] [CrossRef]
  20. Zhang, Y.; Zhang, S.; Li, B.; Luo, Y.; Gong, Y.; Jin, X.; Zhang, J.; Zhou, Y.; Zhuo, X.; Wang, Z.; et al. Gut microbiota dysbiosis promotes age-related atrial fibrillation by lipopolysaccharide and glucose-induced activation of NLRP3-inflammasome. Cardiovasc. Res. 2022, 118, 785–797. [Google Scholar] [CrossRef]
  21. Cronin, P.; Joyce, S.A.; O’Toole, P.W.; O’Connor, E.M. Dietary fiber modulates the gut microbiota. Nutrients 2021, 13, 1655. [Google Scholar] [CrossRef]
  22. Wastyk, H.C.; Fragiadakis, G.K.; Perelman, D.; Dahan, D.; Merrill, B.D.; Yu, F.B.; Topf, M.; Gonzalez, C.G.; Van Treuren, W.; Han, S.; et al. Gut-microbiota-targeted diets modulate human immune status. Cell 2021, 184, 4137–4153.e14. [Google Scholar] [CrossRef]
  23. Lai, H.; Li, Y.; He, Y.; Chen, F.; Mi, B.; Li, J.; Xie, J.; Ma, G.; Yang, J.; Xu, K.; et al. Effects of dietary fibers or probiotics on functional constipation symptoms and roles of gut microbiota: A double-blinded randomized placebo trial. Gut Microbes 2023, 15, 2197837. [Google Scholar] [CrossRef]
  24. El-Sayed, A.; Aleya, L.; Kamel, M. Microbiota’s role in health and diseases. Environ. Sci. Pollut. Res. 2021, 28, 36967–36983. [Google Scholar] [CrossRef]
  25. Zhao, J.; Cheng, W.; Lu, H.; Shan, A.; Zhang, Q.; Sun, X.; Kang, L.; Xie, J.; Xu, B. High fiber diet attenuate the inflammation and adverse remodeling of myocardial infarction via modulation of gut microbiota and metabolites. Front. Microbiol. 2022, 21, 1046912. [Google Scholar] [CrossRef]
  26. Gan, X.T.; Ettinger, G.; Huang, C.X.; Burton, J.P.; Haist, J.V.; Rajapurohitam, V.; Sidaway, J.E.; Martin, G.; Gloor, G.B.; Swann, J.R.; et al. Probiotic administration attenuates myocardial hypertrophy and heart failure after myocardial infarction in the rat. Circ. Heart Fail. 2014, 7, 491–499. [Google Scholar] [CrossRef]
  27. Mansuri, N.M.; Mann, N.K.; Rizwan, S.; E Mohamed, A.; E Elshafey, A.; Khadka, A.; Mosuka, E.M.; Thilakarathne, K.N.; Mohammed, L. Role of Gut Microbiome in Cardiovascular Events: A Systematic Review. Cureus 2022, 14, e32465. [Google Scholar] [CrossRef]
  28. Shen, R.; Chen, S.; Lei, W.; Shen, J.; Lv, L.; Wei, T. Nonfood Probiotic, Prebiotic, and Synbiotic Use Reduces All-Cause and Cardiovascular Mortality Risk in Older Adults: A Population-Based Cohort Study. J. Nutr. Health Aging 2023, 27, 391–397. [Google Scholar] [CrossRef]
  29. Zheng, D.; Li, R.; An, J.; Xie, T.; Han, Z.; Xu, R.; Fang, Y.; Zhang, X. Prebiotics-Encapsulated Probiotic Spores Regulate Gut Microbiota and Suppress Colon Cancer. Adv. Mater. 2020, 32, e2004529. [Google Scholar] [CrossRef]
  30. Wang, M.; Zhang, Z.; Sun, H.; He, S.; Liu, S.; Zhang, T.; Wang, L.; Ma, G. Research progress of anthocyanin prebiotic activity: A review. Phytomedicine 2022, 102, 154145. [Google Scholar] [CrossRef]
  31. Gu, Q.; Yin, Y.; Yan, X.; Liu, X.; Liu, F.; McClements, D.J. Encapsulation of multiple probiotics, synbiotics, or nutrabiotics for improved health effects: A review. Adv. Colloid Interface Sci. 2022, 309, 102781. [Google Scholar] [CrossRef]
  32. Abdallah, A.A.M.; El-Deen, N.A.M.N.; Neamat-Allah, A.N.F.; El-Aziz, H.I.A. Evaluation of the hematoprotective and hepato-renal protective effects of Thymus vulgaris aqueous extract on thermally oxidized oil-induced hematotoxicity and hepato-renal toxicity. Comp. Clin. Pathol. 2020, 29, 451–461. [Google Scholar] [CrossRef]
  33. Reeves, P.G.; Nielsen, F.H.; Fahey, G.C., Jr. 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, 1939–1951. [Google Scholar] [CrossRef]
  34. Mahmud, S.; Khan, S.; Khan, M.R.; Islam, J.; Sarker, U.K.; Hasan, G.M.M.A.; Ahmed, M. Viability and stability of microencapsulated probiotic bacteria by freeze-drying under in vitro gastrointestinal conditions. J. Food Process. Preserv. 2022, 46, e17123. [Google Scholar] [CrossRef]
  35. Garson, C.; Kelly-Laubscher, R.; Blackhurst, D.; Gwanyanya, A. Lack of cardioprotection by single-dose magnesium prophylaxis on isoprenaline-induced myocardial infarction in adult Wistar rats. Cardiovasc. J. Afr. 2015, 26, 242–249. [Google Scholar] [CrossRef]
  36. Bais, R.; Philcox, M. Approved recommendation on IFCC methods for the measurement of catalytic concentration of enzymes. Part 8. IFCC method for lactate dehydrogenase (l-Lactate: NAD+Oxidoreductase, EC 1.1.1.27). International Federation of Clinical Chemistry (IFCC) . Eur. J. Clin. Chem. Clin. Biochem. 1994, 32, 639–655. [Google Scholar]
  37. Reitman, S.; Frankel, S. A colorimetric method for the determination of serum glutamic oxaloacetic and glutamic pyruvic transaminases. Am. J. Clin. Pathol. 1957, 28, 56–63. [Google Scholar] [CrossRef]
  38. Fiolet, J.; Willebrands, A.; Lie, K.; Ter Welle, H. Determination of creatine kinase isoenzyme MB (CK-MB): Comparison of methods and clinical evaluation. Clin. Chim. Acta 1977, 80, 23–35. [Google Scholar] [CrossRef]
  39. Bartles, H.; Bohmer, M.; Heierli, C. Serum creatinine determination without protein precipitation. Clin. Chim. Acta 1972, 37, 193–197. [Google Scholar] [CrossRef]
  40. Fawcett, J.K.; Scott, J.E. A rapid and precise method for the determination of urea. J. Clin. Pathol. 1960, 13, 156–159. [Google Scholar] [CrossRef]
  41. Bancroft, J.D.; Suvarna, K.; Layton, C. Bancroft’s Theory and Practice of Histological Techniques, 7th ed.; Elsevier: London, UK, 2012; ISBN 978-0-7020-5032-9. [Google Scholar]
  42. Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2–ΔΔCt method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef]
  43. Deschner, J.; Rath-Deschner, B.; Agarwal, S. Regulation of matrix metalloproteinase expression by dynamic tensile strain in rat fibrochondrocytes. Osteoarthr. Cartil. 2006, 14, 264–272. [Google Scholar] [CrossRef]
  44. Khan, H.; Abdelhalim, M.; Alhomida, A.; Al Ayed, M. Transient increase in IL-1β, IL-6 and TNF-α gene expression in rat liver exposed to gold nanoparticles. Genet. Mol. Res. 2013, 12, 5851–5857. [Google Scholar] [CrossRef]
  45. IDF Standard No. 149A; Dairy Starter Cultures of Lactic Acid Bacteria (LAB) Standard of Identity. International Dairy Federation (IDF): Brussels, Belgium, 1997.
  46. American Public Health Association (APHA). Standard Methods for Examination of Dairy Products, 16th ed.; American Public Health Association: Washington, DC, USA, 1994. [Google Scholar]
  47. Food and Drug Administration (FDA). Bacteriological Analytical Manual, 9th ed.; AOAC International: Arlington, VA, USA, 2002.
  48. Hanwell, M.D.; Curtis, D.E.; Lonie, D.C.; Vandermeersch, T.; Zurek, E.; Hutchison, G.R. Avogadro: An advanced semantic chemical editor, visualization, and analysis platform. J. Cheminform. 2012, 4, 17. [Google Scholar] [CrossRef]
  49. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef]
  50. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef]
  51. BIOVIA Discovery Studio. Client, version 20.1; Dassault Systèmes BIOVIA: San Diego, CA, USA, 2020. [Google Scholar]
  52. Gan, Z.H.; Cheong, H.C.; Tu, Y.-K.; Kuo, P.-H. Association between Plant-Based Dietary Patterns and Risk of Cardiovascular Disease: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Nutrients 2021, 13, 3952. [Google Scholar] [CrossRef]
  53. Nestel, P.J.; Mori, T.A. Dietary patterns, dietary nutrients and cardiovascular disease. Rev. Cardiovasc. Med. 2022, 23, 17. [Google Scholar] [CrossRef]
  54. Honerlaw, J.P.; Ho, Y.-L.; Nguyen, X.-M.T.; Cho, K.; Vassy, J.L.; Gagnon, D.R.; O’DOnnell, C.J.; Gaziano, J.M.; Wilson, P.W.; Djousse, L. Fried food consumption and risk of coronary artery disease: The Million Veteran Program. Clin. Nutr. 2020, 39, 1203–1208. [Google Scholar] [CrossRef]
  55. Dianita, R.; Jantan, I.; Amran, A.Z.; Jalil, J. Protective Effects of Labisiapumila var. Alata on Biochemical and Histopathological Alterations of Cardiac Muscle Cells in Isoproterenol-Induced Myocardial Infarction Rats. Molecules 2015, 20, 4746–4763. [Google Scholar] [CrossRef]
  56. Iftikhar, F.; Tauqeer, S.; Farhat, S.; Orakzai, M.; Naz, R.; Rehman, A. Common Risk Factors Involved in the Development of Myocardial Infarction in Adults Younger Than 45 Years of Age. J. Ayub Med Coll. Abbottabad 2022, 34, S995–S999. [Google Scholar] [CrossRef]
  57. Nowowiejska-Wiewióra, A.; Wita, K.; Mędrala, Z.; Tomkiewicz-Pająk, L.; Bujak, K.; Mizia-Stec, K.; Brzychczy, P.; Gąsior, M.; Gąsior, Z.; Kulbat, A.; et al. Dyslipidemia treatment and attainment of LDL-cholesterol treatment goals in patients participating in the Managed Care for Acute Myocardial Infarction Survivors program. Kardiol. Pol. 2023, 81, 359–365. [Google Scholar] [CrossRef]
  58. Duchowicz, P.R.; Szewczuk, N.A.; Pomilio, A.B. QSAR studies of the antioxidant activity of anthocyanins. J. Food Sci. Technol. 2019, 56, 5518–5530. [Google Scholar] [CrossRef]
  59. Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Gaspari, A.; Di Minno, G.; Ritieni, A. Red Wine Consumption and Cardiovascular Health. Molecules 2019, 24, 3626. [Google Scholar] [CrossRef]
  60. Mohamed, D.A.; Mohamed, R.S.; Fouda, K. Anti-inflammatory potential of chia seeds oil and mucilage against adjuvant induced arthritis in obese and non-obese rats. J. Basic Clin. Physiol. Pharmacol. 2020, 31, 20190236. [Google Scholar] [CrossRef]
  61. Mohamed, D.A.; Mohammed, S.E.; Hamed, I.M. Chia seeds oil enriched with phytosterols and mucilage as a cardioprotective dietary supplement towards inflammation, oxidative stress, and dyslipidemia. J. Herbmed Pharmacol. 2022, 11, 83–90. [Google Scholar] [CrossRef]
  62. Derbali, A.; Mnafgui, K.; Affes, M.; Derbali, F.; Hajji, R.; Gharsallah, N.; Allouche, N.; El Feki, A. Cardioprotective effect of linseed oil against isoproterenol-induced myocardial infarction in Wistar rats: A biochemical and electrocardiographic study. J. Physiol. Biochem. 2015, 71, 281–288. [Google Scholar] [CrossRef]
  63. Shikalgar, T.S.; Naikwade, N.S. Evaluation of cardioprotective activity of fulvic acid against isoproterenol induced oxidative damage in rat cardium. Int. Res. J. Pharm. 2018, 9, 71–80. [Google Scholar] [CrossRef]
  64. Rebello, C.J.; O’Neil, C.E.; Greenway, F.L. Dietary fiber and satiety: The effects of oats on satiety. Nutr. Rev. 2016, 74, 131–147. [Google Scholar] [CrossRef]
  65. Lindner, D.; Zietsch, C.; Becher, P.M.; Schulze, K.; Schultheiss, H.-P.; Tschöpe, C.; Westermann, D. Differential Expression of Matrix Metalloproteases in Human Fibroblasts with Different Origins. Biochem. Res. Int. 2012, 2012, 875742. [Google Scholar] [CrossRef]
  66. Tanase, D.M.; Valasciuc, E.; Anton, I.-B.; Gosav, E.M.; Dima, N.; Cucu, A.I.; Costea, C.F.; Floria, D.E.; Hurjui, L.L.; Tarniceriu, C.C.; et al. Matrix Metalloproteinases: Pathophysiologic Implications and Potential Therapeutic Targets in Cardiovascular Disease. Biomolecules 2025, 15, 598. [Google Scholar] [CrossRef]
  67. DeLeon-Pennell, K.Y.; Meschiari, C.A.; Jung, M.; Lindsey, M.L. Matrix Metalloproteinases in Myocardial Infarction and Heart Failure. Prog. Mol. Biol. Transl. Sci. 2017, 147, 75–100. [Google Scholar] [CrossRef]
  68. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  69. El-Sayed, H.S.; El-Sayed, S.M.; Youssef, A.M. Designated functional microcapsules loaded with green synthesis selenium nanorods and probiotics for enhancing stirred yogurt. Sci. Rep. 2022, 12, 14751. [Google Scholar] [CrossRef]
  70. El Sayed, H.S.; Mabrouk, A.M. Encapsulation of probiotics using mixed sodium alginate and rice flour to enhance their survivability in simulated gastric conditions and in UF-Kariesh cheese. Biocatal. Agric. Biotechnol. 2023, 50, 102738. [Google Scholar] [CrossRef]
  71. El-Sayed, S.M.; El-Sayed, H.S.; Youssef, A.M. Recent developments in encapsulation techniques for innovative and high-quality dairy products: Demands and challenges. Bioact. Carbohydr. Diet. Fibre 2024, 31, 100406. [Google Scholar] [CrossRef]
  72. Pan, P.; Lam, V.; Salzman, N.; Huang, Y.-W.; Yu, J.; Zhang, J.; Wang, L.-S. Black raspberries and their anthocyanin and fiber fractions alter the composition and diversity of gut microbiota in F-344 rats. Nutr. Cancer 2017, 69, 943–951. [Google Scholar] [CrossRef]
  73. Jalanka, J.; Major, G.; Murray, K.; Singh, G.; Nowak, A.; Kurtz, C.; Silos-Santiago, I.; Johnston, J.M.; de Vos, W.M.; Spiller, R. The effect of psyllium husk on intestinal microbiota in constipated patients and healthy controls. Int. J. Mol. Sci. 2019, 20, 433. [Google Scholar] [CrossRef]
  74. Aravind, S.M.; Wichienchot, S.; Tsao, R.; Ramakrishnan, S.; Chakkaravarthi, S. Role of dietary polyphenols on gut microbiota, their metabolites and health benefits. Food Res. Int. 2021, 142, 110189. [Google Scholar] [CrossRef]
  75. Martellet, M.C.; Majolo, F.; Ducati, R.G.; de Souza, C.F.V.; Goettert, M.I. Probiotic applications associated with Psyllium fiber as prebiotics geared to a healthy intestinal microbiota: A review. Nutrition 2022, 103, 111772. [Google Scholar] [CrossRef]
  76. Seke, F.; Manhivi, V.E.; Slabbert, R.M.; Sultanbawa, Y.; Sivakumar, D. In Vitro Release of Anthocyanins from Microencapsulated Natal Plum (Carissa macrocarpa) Phenolic Extract in Alginate/Psyllium Mucilage Beads. Foods 2022, 11, 2550. [Google Scholar] [CrossRef]
  77. Wu, H.; Chiou, J. Potential benefits of probiotics and prebiotics for coronary heart disease and stroke. Nutrients 2021, 13, 2878. [Google Scholar] [CrossRef]
  78. Oniszczuk, A.; Oniszczuk, T.; Gancarz, M.; Szymańska, J. Role of gut microbiota, probiotics and prebiotics in the cardiovascular diseases. Molecules 2021, 26, 1172. [Google Scholar] [CrossRef]
  79. Ren, Z.; Hong, Y.; Huo, Y.; Peng, L.; Lv, H.; Chen, J.; Wu, Z.; Wan, C. Prospects of Probiotic Adjuvant Drugs in Clinical Treatment. Nutrients 2022, 14, 4723. [Google Scholar] [CrossRef]
  80. DiRienzo, D.B. Effect of probiotics on biomarkers of cardiovascular disease: Implications for heart-healthy diets. Nutr. Rev. 2014, 72, 18–29. [Google Scholar] [CrossRef]
  81. Tunapong, W.; Apaijai, N.; Yasom, S.; Tanajak, P.; Wanchai, K.; Chunchai, T.; Kerdphoo, S.; Eaimworawuthikul, S.; Thiennimitr, P.; Pongchaidecha, A.; et al. Chronic treatment with prebiotics, probiotics and synbiotics attenuated cardiac dysfunction by improving cardiac mitochondrial dysfunction in male obese insulin-resistant rats. Eur. J. Nutr. 2018, 57, 2091–2104. [Google Scholar] [CrossRef]
  82. Hesari, Z.; Kafshdoozan, K.; Barati, M.; Kokhaei, P.; Andalib, S.; Kiassari, F.T.; Darban, M.; Abdolshahi, A.; Bagheri, B. Lactobacillus paracasei impact on myocardial hypertrophy in rats with heart failure. J. Chem. Health Risks 2020, 10, 67–74. [Google Scholar] [CrossRef]
  83. Delzenne, N.M.; Bindels, L.B.; Neyrinck, A.M.; Walter, J. The gut microbiome and dietary fibres: Implications in obesity, cardiometabolic diseases and cancer. Nat. Rev. Microbiol. 2025, 23, 225–238. [Google Scholar] [CrossRef]
  84. Sror, H.A.M.; Rizk, E.; Azouz, A.; Hareedy, L.A.M. Evaluation of red cabbage anthocyanin pigments and its potential uses as antioxidant and natural food colorants. Arab. Univ. J. Agric. Sci. 2009, 17, 361–372. [Google Scholar] [CrossRef]
  85. Ryu, S.Y.; Kleine, C.-E.; Hsiung, J.-T.; Park, C.; Rhee, C.M.; Moradi, H.; Hanna, R.; Kalantar-Zadeh, K.; Streja, E. Association of lactate dehydrogenase with mortality in incident hemodialysis patients. Nephrol. Dial. Transplant. 2021, 36, 704–712. [Google Scholar] [CrossRef]
  86. Han, J.H.; Lee, E.J.; Park, W.; Ha, K.T.; Chung, H.S. Natural compounds as lactate dehydrogenase inhibitors: Potential therapeutics for lactate dehydrogenase inhibitors-related diseases. Front. Pharmacol. 2023, 14, 1275000. [Google Scholar] [CrossRef]
  87. Djekic, S.; Vekic, J.; Zeljkovic, A.; Kotur-Stevuljevic, J.; Kafedzic, S.; Zdravkovic, M.; Ilic, I.; Hinic, S.; Cerovic, M.; Stefanovic, M.; et al. HDL Subclasses and the Distribution of Paraoxonase-1 Activity in Patients with ST-Segment Elevation Acute Myocardial Infarction. Int. J. Mol. Sci. 2023, 24, 9384. [Google Scholar] [CrossRef]
  88. Leocádio, P.C.L.; Goulart, A.C.; Santos, I.S.; Lotufo, P.A.; Bensenor, I.M.; Alvarez-Leite, J.I. Lower paraoxonase 1 paraoxonase activity is associated witha worse prognosis in patients with non-ST-segment elevation myocardial infarction in long-term follow-up. Coron. Artery Dis. 2022, 33, 515–522. [Google Scholar] [CrossRef]
Figure 1. Total phenolic, total flavonoid, and total anthocyanin content in anthocyanin-rich extract. Values are mean ± SE (n = 3).
Figure 1. Total phenolic, total flavonoid, and total anthocyanin content in anthocyanin-rich extract. Values are mean ± SE (n = 3).
Applmicrobiol 05 00072 g001
Figure 2. The mRNA expression of MMP-7 and IL1-β in the hearts of different experimental groups. The mRNA expression of MMP7 and IL1β is normalized with housekeeping gene (GAPDH), values are represented as means ± SE (n = 6), same letters indicate not-significant difference, and different letters indicate significant difference at the level of 0.05 probability levels.
Figure 2. The mRNA expression of MMP-7 and IL1-β in the hearts of different experimental groups. The mRNA expression of MMP7 and IL1β is normalized with housekeeping gene (GAPDH), values are represented as means ± SE (n = 6), same letters indicate not-significant difference, and different letters indicate significant difference at the level of 0.05 probability levels.
Applmicrobiol 05 00072 g002
Figure 3. Micrographs of rats’ hearts from different groups. (a) Normal control group, (b) MI group, (c) encapsulated synbiotic I group, (d) encapsulated synbiotic II group, (e) encapsulated probiotic group. (H&E X400).
Figure 3. Micrographs of rats’ hearts from different groups. (a) Normal control group, (b) MI group, (c) encapsulated synbiotic I group, (d) encapsulated synbiotic II group, (e) encapsulated probiotic group. (H&E X400).
Applmicrobiol 05 00072 g003
Figure 4. Improvement percent in heart lesion scoring compared with MI control. Values are mean ± SE (n = 4).
Figure 4. Improvement percent in heart lesion scoring compared with MI control. Values are mean ± SE (n = 4).
Applmicrobiol 05 00072 g004
Table 1. Primer sequences used for MMP-7 and IL-1β gene expression analysis.
Table 1. Primer sequences used for MMP-7 and IL-1β gene expression analysis.
Target GenesSequencesRef.
MMP-7FW (5′-TCG GCG GAG ATG CTC ACT-3′)
RW (5′-TGG CAA CAA ACA GGA AGT TCA C-3′)
[43]
IL-1βFW (5′-TGA TGG ATG CTT CCA AAC TG-3′)
RW (5′-GAG CAT TGG AAG TTG GGG TA-3′)
[44]
GAPDHFW (5′-GTATTGGGCGCCTGGTCACC -3′)
RW (5′-CGCTCCTGGAAGATGGTGATGG -3′)
[44]
Table 2. Plasma lipid profile, oxidized-LDL, Paraoxonase 1, and Paraoxonase 1/HDL ratio of different experimental groups.
Table 2. Plasma lipid profile, oxidized-LDL, Paraoxonase 1, and Paraoxonase 1/HDL ratio of different experimental groups.
ParametersNormal ControlMI ControlEncapsulated Synbiotic IEncapsulated Synbiotic IIEncapsulated
Probiotic
TC (mg/dL)82.7 a ± 0.78155.9 b ± 3.3889.3 a ± 2.9885.8 a ± 3.6282.8 a ± 4.38
TG (mg/dL)79.1 a ± 1.19151.4 b ± 2.9888.15 a ± 2.6483.5 a ± 2.6182.64 a ± 2.76
HDL-Ch (mg/dL)48.1 a ± 0.7434.7 b ± 0.7045.6 a ± 1.5546.92 a ± 0.8947.66 a ± 0.33
LDL-Ch (mg/dL)18.8 a ± 1.3190.96 b ± 2.9126.1 a ± 3.9922.8 a ± 2.8417.3 a ± 4.38
VLDL-Ch (mg/dL)15.8 a ± 0.2430.3 b ± 0.5917.6 a ± 0.5316.0 a ± 0.7816.5 a ± 0.55
T-Ch/HDL-Ch1.72 a ± 0.034.50 b ± 0.081.98 a ± 0.121.83 a ± 0.071.74 a ± 0.10
Ox-LDL (pg/mL)90.6 a ± 2.89165.4 b ± 6.2994.8 a ± 3.9892.3 a ± 2.3593.1 a ± 2.82
PON1 (pg/mL)1126.7 a ± 11.95888.7 b ± 14.411120.1 a ± 17.16 1053.9 c ± 13.451083.9 ac ± 13.91
Paraoxonase 1/HDL ratio23.5 a ± 0.4425.7 b ± 0.8223.9 a ± 0.5423.3 c ± 1.0122.8 ac ± 0.28
In the same row: similar letters mean non-significant difference within groups at p < 0.05, values are mean ± SE (n = 6). T-Ch: total cholesterol, HDL-Ch: high-density lipoprotein cholesterol, LDL-Ch: low-density lipoprotein cholesterol, VLDL-Ch: very-low-density lipoprotein cholesterol, TG: triglycerides, Ox-LDL: oxidized-LDL, PON1: Paraoxonase 1.
Table 3. Cardiac marker enzymes and kidney function of different experimental groups.
Table 3. Cardiac marker enzymes and kidney function of different experimental groups.
GroupsCK-MB (U/L)LDH
(U/L)
AST
(IU/L)
ALT
(IU/L)
Urea
(mg/dL)
Creatinine (mg/dL)
Normal control132.7 a ± 6.59169.1 a ± 7.1234.7 a ± 2.0115.2 a ± 1.5432.8 a ± 0.890.71 a ± 0.03
MI control276.8 b ± 7.09263.48 b ± 7.4271.7 b ± 2.9722.3 b ± 1.3347.32 b ± 1.111.18 b ± 0.08
Encapsulated synbiotic I 186.3 c ± 4.01192.2 a ± 4.5136.2 a ± 1.6417.5 ab ± 1.3636.52 a ± 1.710.80 a ± 0.02
Encapsulated synbiotic II172.3 c ± 5.43194.9 a ± 7.0236.7 a ± 1.4918.2 ab ± 1.7936.98 a ± 1.590.77 a ± 0.04
Encapsulated probiotic 163.0 c ± 8.93179.2 a ± 7.7338.2 a ± 1.6017.5 ab ± 1.3634.95 a ± 1.140.75 a ± 0.01
In the same column: similar letters mean non-significant difference within groups at p < 0.05, values are mean ± SE (n = 6). CK-MB: creatine kinase-MB, LDH: lactate dehydrogenase, AST: aspartate aminotransferase, ALT: alanine aminotransferase.
Table 4. Antioxidant and inflammatory markers of different rat groups.
Table 4. Antioxidant and inflammatory markers of different rat groups.
GroupsSOD (U/mL)TNF-α (pg/mL)CRP (ng/mL)
Normal control12.38 b ± 0.2618.8 a ± 0.602.78 a ± 0.09
MI control2.12 a ± 0.1032.8 b ± 0.709.07 b ± 0.25
Encapsulated synbiotic I 8.82 c ± 0.2522.83 c ± 0.485.83 c ± 0.21
Encapsulated synbiotic II8.40 c ± 0.2222.33 c ± 0.765.73 c ± 0.18
Encapsulated probiotic8.08 c ± 0.3620.92 ac ± 0.525.15 c ± 0.16
In the same column: similar letters mean non-significant difference within groups at p < 0.05, values are mean ± SE (n = 6). SOD: superoxide dismutase, TNF-α: tumor necrosis factor-alpha, CRP: C-reactive protein.
Table 5. Nutritional parameters of different experimental groups.
Table 5. Nutritional parameters of different experimental groups.
GroupsInitial Body Weight (g)Final Body Weight
(g)
Body Weight Gain (g)Relative Heart Weight (%)
Normal control231.2 a ± 8.87290.3 a ± 8.6859.2 ad ± 2.150.35 a ± 0.01
MI control231.3 a ± 8.58281.3 a ± 6.7450.0 ac ± 3.200.43 b ± 0.01
Encapsulated synbiotic I 231.2 a ± 5.36285.8 a ± 7.5754.7 ad ± 5.140.37 a ± 0.01
Encapsulated synbiotic II231.3 a ± 2.19275.5 a ± 3.6244.2 bcd ± 4.210.37 a ± 0.02
Encapsulated probiotic 231.2 a ± 2.46287.2 a ± 1.8556.0 ad ± 1.860.35 a ± 0.01
In the same column: similar letters mean non-significant difference within groups at p ≤ 0.05, values are mean ± SE (n = 6).
Table 6. The microbial population of rat feces of the different experimental groups.
Table 6. The microbial population of rat feces of the different experimental groups.
GroupsProbiotic
Counts
Coliforms CountTotal Bacterial
Counts
Staphylococci CountListeria
Counts
Normal control6.46 c ± 0.285.93 b ± 0.227.37 a ± 0.336.11 a ± 0.195.25 b ± 0.21
MI control4.67 d ± 0.116.97 a ± 0.197.78 a ± 0.286.77 a ± 0.207.16 a ± 0.27
Encapsulated synbiotic I 8.53 a ± 0.304.91 c ± 0.255.90 c ± 0.204.85 c ± 0.194.59 c ± 0.19
Encapsulated synbiotic II8.70 a ± 0.284.77 c ± 0.175.96 c ± 0.185.53 b ± 0.254.58 c ± 0.29
Encapsulated probiotic 7.45 b ± 0.184.95 c ± 0.226.88 b ± 0.255.30 b ± 0.174.72 c ± 0.18
The viable count is expressed as log CFU/g. Columns with the same letter are not significant (p ≤ 0.05). Values with different superscript letters in the same column are significantly different at p ≤ 0.05 levels, values are mean ± SE (n = 6).
Table 7. 3D and 2D interactions between each compound with lactate dehydrogenase (LDH).
Table 7. 3D and 2D interactions between each compound with lactate dehydrogenase (LDH).
CompoundBinding Affinity (∆G (kcal/mol))3D2D
Arabinoxylan−8.8Applmicrobiol 05 00072 i001Applmicrobiol 05 00072 i002
The amino acids involved in the interaction of arabinoxylan with LDH were GLY97, ASN113, HIS193, THR95, ASN138, ALA98, GLY29, THR248, and GLY246.
Cyanidin 3-diglucoside 5-glucoside−8.9Applmicrobiol 05 00072 i003Applmicrobiol 05 00072 i004
The amino acids involved in the interaction of cyanidin 3-diglucoside 5-glucoside with LDH were ALA238, GLN100, TYR239, ILE242, ASN138, ALA98, THR248, and ILE242.
Table 8. 3D and 2D interactions between each compound with Paraoxonase 1 (PON1).
Table 8. 3D and 2D interactions between each compound with Paraoxonase 1 (PON1).
CompoundBinding Affinity (∆G (kcal/mol))3D2D
Arabinoxylan−8.8Applmicrobiol 05 00072 i005Applmicrobiol 05 00072 i006
The amino acids involved in the interaction of arabinoxylan with PON1 were ASP54, ILE170, ILE226, GLU56, LEU230, ILE271, and LEU230.
Cyanidin 3-diglucoside 5-glucoside−10Applmicrobiol 05 00072 i007Applmicrobiol 05 00072 i008
The amino acids involved in the interaction of cyanidin 3-diglucoside 5-glucoside with PON1 were ASP54, GLU56, LEU230, ILE271, THR119, ILE170, ASP169, ILE57, GLY232, SER272, PRO59, PRO275, and VAL273.
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

Mohamed, D.A.; Mabrok, H.B.; El-Sayed, H.S.; Abdelgayed, S.; Mohammed, S.E. Cardio-Protective Effects of Microencapsulated Probiotic and Synbiotic Supplements on a Myocardial Infarction Model Through the Gut–Heart Axis. Appl. Microbiol. 2025, 5, 72. https://doi.org/10.3390/applmicrobiol5030072

AMA Style

Mohamed DA, Mabrok HB, El-Sayed HS, Abdelgayed S, Mohammed SE. Cardio-Protective Effects of Microencapsulated Probiotic and Synbiotic Supplements on a Myocardial Infarction Model Through the Gut–Heart Axis. Applied Microbiology. 2025; 5(3):72. https://doi.org/10.3390/applmicrobiol5030072

Chicago/Turabian Style

Mohamed, Doha A., Hoda B. Mabrok, Hoda S. El-Sayed, Sherein Abdelgayed, and Shaimaa E. Mohammed. 2025. "Cardio-Protective Effects of Microencapsulated Probiotic and Synbiotic Supplements on a Myocardial Infarction Model Through the Gut–Heart Axis" Applied Microbiology 5, no. 3: 72. https://doi.org/10.3390/applmicrobiol5030072

APA Style

Mohamed, D. A., Mabrok, H. B., El-Sayed, H. S., Abdelgayed, S., & Mohammed, S. E. (2025). Cardio-Protective Effects of Microencapsulated Probiotic and Synbiotic Supplements on a Myocardial Infarction Model Through the Gut–Heart Axis. Applied Microbiology, 5(3), 72. https://doi.org/10.3390/applmicrobiol5030072

Article Metrics

Back to TopTop