Next Article in Journal
Profile of PKS and NRPS Gene Clusters in the Genome of Streptomyces cellostaticus NBRC 12849T
Next Article in Special Issue
Health and Bioactive Compounds of Fermented Foods and By-Products
Previous Article in Journal
From Shallow to Deep Bioprocess Hybrid Modeling: Advances and Future Perspectives
Previous Article in Special Issue
Valorization of Fermented Food Wastes and Byproducts: Bioactive and Valuable Compounds, Bioproduct Synthesis, and Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Certain Fermented Foods and Their Possible Health Effects with a Focus on Bioactive Compounds and Microorganisms

1
Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey
2
Department of Food Safety and Quality, Lithuanian University of Health Sciences, Tilzes 18, LT-47181 Kaunas, Lithuania
3
Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes 18, LT-47181 Kaunas, Lithuania
4
CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associ ado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
5
LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
6
ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
7
Department of Seafood Processing Technology, Cukurova University, Balcalı, Adana 01330, Turkey
8
Biotechnology Research and Application Center, Cukurova University, Adana 01330, Turkey
*
Authors to whom correspondence should be addressed.
Fermentation 2023, 9(11), 923; https://doi.org/10.3390/fermentation9110923
Submission received: 12 September 2023 / Revised: 25 September 2023 / Accepted: 29 September 2023 / Published: 24 October 2023
(This article belongs to the Special Issue Health and Bioactive Compounds of Fermented Foods and By-Products)

Abstract

:
Fermented foods refer to beverages or foods made by carefully regulated microbial growth and the enzymatic conversion of dietary components. Fermented foods have recently become more popular. Studies on fermented foods suggest the types of bacteria and bioactive peptides involved in this process, revealing linkages that may have impacts on human health. By identifying the bacteria and bioactive peptides involved in this process, studies on fermented foods suggest relationships that may have impressions on human health. Fermented foods have been associated with obesity, cardiovascular disease, and type 2 diabetes. In this article, fermented dairy products, vegetables and fruits, legumes, meats, and grains are included. Two elements in particular are emphasized when discussing the fermentation of all of these foods: bioactive chemicals generated during fermentation and microorganisms involved during fermentation. Organic acids, bioactive peptides, conjugated linoleic acid, biogenic amines, isoflavones, phytoestrogens, and nattokinase are a few of the bioactive compounds included in this review. Also, certain bacteria such as Lactobacillus, Bifidobacterium, Streptococcus, and Bacillus species, which are utilized in the fermentation process are mentioned. The effects of both substances including anti-fungal and antioxidant properties; the modulation of intestinal microbiota; anti-inflammatory, antidiabetes, anti-obesity, anticancer, and antihypertension properties; and the protection of cognitive function are explained in this review.

1. Introduction

Fermentation is a food processing technique, and its origins date back many centuries. The existence of fermented products has been demonstrated to be started in India, Iraq, and Egypt in the years BC [1]. The definition of fermentation according to the International Scientific Probiotic and Prebiotic Association (ISAPP) is “foods made through desired microbial growth and enzymatic conversions of food components”. Fermentation has been used by humans for as long as recorded history to preserve and modify food, resulting in more stable and varied food with distinctive organoleptic, sensory, and functional features [2]. Due to their distinctive flavors, fermented foods are being produced and consumed in greater quantities. There has also been a scientific concentration on the health benefits of fermented foods and their components [3]. In the fermentation procedure, microorganisms, specifically bacteria, yeasts, and mycelial fungus, as well as their enzymes, produce fermented foods. Milk, cereals, vegetables, fruits, legumes, meats, and products are food groups used in fermentation [4].
Food fermentation’s main purposes are to increase food safety and lengthen shelf life; additionally, fermented foods have grown to be known for their positive effects on health [5]. Foods that have undergone fermentation may produce bioactive compounds as byproducts of the process, and fermented foods may contain live microorganisms that have health benefits [6]. The main metabolites and microorganisms involved in food fermentation may be divided into categories: alcohol, carbon dioxide (from yeast), propionic acid (from Propionibacterium freudenreichii), lactic acid (from lactic acid bacteria (LAB) from genera such as Lactobacillus and Streptococcus), acetic acid (from Acetobacter), ammonia, and fatty acids (from Bacillus and molds) [7]. The metabolites produced by the fermenting organisms limit the expansion of spoilage, and pathogenic organisms during food fermentation extend the shelf life of perishable foods [8]. During fermentation, macronutrients are broken down, and digestion is facilitated. Many fermented foods include probiotic-potential bacteria in them [5]. Probiotics are defined in the FAO/WHO report as “Live microorganisms which when administered in adequate amounts confer a health benefit on the host” [9]. According to the ISAPP, these concentrations of probiotics can vary daily from 100 million to over a trillion CFU. The majority have been studied at concentrations of 1 to 10 billion CFU/d [10]. Fermented foods may serve as probiotic carriers, effectively delivering the probiotic to the host and conferring health advantages [11]. Although there may be a fermentation process involved, consumed fermented foods may not contain live bacteria. The term “probiotic” is only used when a product has clearly shown health advantages brought about by the action of well-defined and characterized living microorganisms [2]. The metabolic activity of microorganisms during fermentation results in a number of biochemical alterations that have impacts on the nutritive and bioactive qualities of fermented foods. The bioactive components showing health benefits include exopolysaccharides, bioactive peptides, phenolic compounds, short-chain fatty acids (SCFAs), conjugated linoleic acid (CLA), and γ-aminobutyric acids (GABAs) [12]. Fermented foods and their components can have many health effects such as antioxidant, antidiabetes, anti-inflammatory, anti-hypercholesterolemic, and microbiota modulation effects [13,14,15,16].
This review focuses on the advantages of bioactive compounds for health and the probiotic microorganisms that occur in some foods during fermentation or are derived from fermented foods. This article focuses on fermented foods that are frequently consumed in the food groups of dairy, fruits, vegetables, meats, cereals, and legumes. Some fermented foods and the health effects of the components that occur during fermentation, as well as the health effects of probiotic microorganisms in fermented foods, are included. In summary, this article seeks to investigate the possible health effects of fermented foods by focusing on the following: (i) the general effects of fermented foods on health, (ii) the compounds that occur in bioactive components during fermentation and their health effects, and (iii) the health effects of bacteria with probiotic properties that are contained in or isolated from fermented foods.

2. Fermented Dairy Products

Milk is an important source of macro- and micronutrients. Protein, conjugated linoleic acid, calcium, riboflavin, and phosphorus are macro- and micronutrients that are commonly found in milk. These nutrients have impacts on health and diseases [17]. Milk proteins (whey and casein) have positive effects on satiety and body weight control; have hypotensive, antimicrobial, anti-inflammatory, anticancer, and antioxidant effects; and cause insulin release and glucose regulation [18]. Many fermented products such as yogurt, cheese, and kefir are obtained via the fermentation of milk. In the production of fermented milk products, LAB play a crucial role [19]. Milk fermentation using yeasts, propionibacteria, and LAB may result in the synthesis or increase in the number of bioactive compounds that show some health benefits. These include vitamins, CLA, exopolysaccharides (EPSs), GABAs, bioactive peptides, and oligosaccharides [20]. For example, lactic acid bacteria and propionibacteria can increase the amounts of B12 and folic acid in fermented milk products [21,22,23]. In addition, the fermentation of lactic acid in milk reduces the amount of lactose, which may make fermented dairy products tolerable for people with lactose intolerance [24]. The bioactive components and health effects of kefir, yogurt, and cheese, which are widely consumed fermented dairy products, are examined in detail below.

2.1. Kefir

Kefir is an acidic alcoholic fermented dairy product with a creamy consistency and a slightly acidic taste, originating from the Balkans, Eastern Europe, and the Caucasus. Traditionally, kefir is produced using cow, sheep, goat, or buffalo milk [25]. Kefir grains are used as a starter in the production of kefir. The bacteria and yeasts commonly found in kefir grains are Lactobacillus kefiranofaciens, Lacticaseibacillus paracasei, Lactiplantibacillus plantarum, Lactobacillus acidophilus, Lactobacillus delbrueckii subsp. bulgaricus, Kluyveromyces marxianus ssp. Marxianus Candida kefyr, Saccharomyces cerevisiae, and Saccharomyces unisporus [26]. Kefir is thought to include more than 300 distinct microbial species [27]. The microorganisms present in kefir grains may vary depending on the source of kefir, climatic conditions, geographical origin, substrate used in the fermentation process, and production methods [28,29,30,31].
Kefir exerts its positive effects on health through whole kefir, kefir grains, lactic acid bacteria, yeasts, organic acids, polysaccharides (kefiran and exopolysaccharides), and various other metabolites [32]. As a result of fermentation, there is an increase in lactic acid and antioxidant activity in kefir compared to normal milk [33]. In addition, Propionibacterium freudenreichii bacteria in kefir grains can cause an increase in B12 and folate levels [22]. Studies on kefir, kefir grains, and kefir components (lactic acid bacteria, organic acids, bioactive peptides, and polysaccharides) have shown that they have antihypertensive [34], anticancer [35], antioxidant [36], anti-inflammatory [37], antidiabetic [38], and hypocholesterolemic effects [39] in addition to effects on bone health [40], cognitive function [41], and microbiota modulation [42].
The bacteria and yeasts identified in kefir can have positive effects on health. Kluyveromyces marxianus is one of the yeasts in kefir. Its strain, obtained from kefir, has been shown to remain alive in the digestive system [43]. Kluyveromyces marxianus B0399 supplementation decreased proinflammatory cytokines (tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, macrophage inflammatory protein-1 (MIP-1) α, IL-12, IL-8, interferon (IFN)-γ) and increased SCFAs (acetate and propionate). Although there was no change in the total bacterial count, the Bifidobacterium genus count increased [44]. Kluyveromyces marxianus A4 and A5 supplementation showed good adhesion in Caco-2 cells. Kluyveromyces marxianus A4 increased Bacteroidetes, Bacteroidales, and Bacteroides, and Kluyveromyces marxianus A5 increased Corynebacteriales and Corynebacterium [45]. In another study, a high concentration of Kluyveromyces marxianus A5 decreased IL-6 [46]. In this direction, kefir is effective in immune response and colonic microbiota modulation. The lactic acid bacteria obtained from kefir have immunomodulatory and antioxidant effects [39,47].
As a result of fermentation in kefir, kefiran and exopolysaccharides are formed. Polysaccharides have been described to have anticancer, anti-inflammatory, antioxidant, anti-atherosclerosis, and microbiota modulation effects [42,48,49,50,51,52,53,54,55,56,57,58,59,60,61]. In the study by Bengoa et al. [62], Lacticaseibacillus paracasei exopolysaccharides isolated from kefir increased the fecal total SCFA, propionic acid, and butyric acid levels [62]. Lim et al. [63] found that kefir exopolysaccharides reduced intracellular lipid accumulation and enhanced the abundance of Akkermansia spp. in feces. Since there are many studies on the effects of kefir and its fermented components on health, Table 1 summarizes the components and their effects.

2.2. Yogurt

One of the products of the lactic acid fermentation of milk is yogurt. The lactose in its content is converted into lactic acid by bacteria. In this way, it can be tolerated in the case of lactose intolerance. It also has benefits for health due to its protein content, vitamins such as riboflavin, minerals such as calcium, and metabolites that result from fermentation [82]. One of the components in yogurt that may have a positive effect on health is the CLA content. As a result of the increase in fermentation time, the CLA level in yogurt may increase [83]. The CLA contents of natural yogurt, probiotic yogurt, and Greek yogurt obtained from goat milk were found to be 3.28 ± 0.10 mg/g fat, 4.07 ± 0.08 mg/g fat, and 4.19 ± 0.14 mg/g fat, respectively [84]. The CLA contents of cow, sheep, and goat milk yogurts were found to be 0.128–1.501, 0.405–1.250, and 0.433–0.976 g CLA/100 g fat, respectively [85]. In another study, 0.24–0.45 g/100 g fat was found in cow yogurt, and 0.47–0.76 g/100 g fat was found in sheep yogurt. When the effect of storage time on the CLA level was evaluated, the CLA level increased significantly in yogurt obtained from sheep milk after 14 days of storage, while it decreased in yogurt obtained from cow milk [86]. When yogurts from Polish markets were evaluated, the highest CLA content was found in bio yogurt. Probiotics and natural yogurt did not differ in the CLA content [87]. CLA has positive effects on obesity, cancer, cardiovascular diseases, bone health, and immune response [88].
Studies on the consumption of conventional/probiotic yogurt have evaluated its microbiota modulation [89], hypocholesterolemic [90], antidiabetic, antioxidant [91], and anti-obesity effects [92]. It was found that visceral fat decreased, and the abundances of Streptococcus thermophilus and Bifidobacterium animalis subsp. Lactis species increased in individuals who consumed yogurt. A correlation was observed between Bifidobacterium animalis subsp. Lactis and increased fecal 3-hydroxyoctanoic acid contents [89]. Yogurt supplementation (220 g/day) decreased fasting insulin, insulin resistance, intrahepatic lipid, hepatic fat fraction, serum lipopolysaccharide (LPS), fibroblast growth factor 21, triglycerides, TNF-α, total cholesterol, glutathione peroxidase (GPH-Px), and superoxide dismutase (SOD). In addition, it regulated the microbiota composition [91]. Hasegawa et al. [92] reported that yogurt supplementation in obese mice resulted in decreased the levels of Homeostasis Model Assessment of Insulin Resistance (HOMA-IR), serum TNF-α, plasma LPS binding protein, and colonic LPS expression, and altered the diversity of cecal microbiota. In addition, body weight gain was reduced in obese mice [92]. Compared to no snack consumption, yogurt consumption reduced afternoon hunger and delayed the desire to start the next meal. Although this effect was observed for all yogurts, the highest and most significant effect was observed in yogurt containing 24 g of protein [93].
When the effects of probiotic and conventional yogurts were evaluated, the total cholesterol and total cholesterol/high-density lipoprotein cholesterol (HDL-C) ratio decreased as a result of consuming both yogurts. Both yogurts showed hypocholesterolemic effects [90]. In the study by Rezazadeh et al. [94], probiotic yogurt decreased blood glucose, insulin, HOMA-IR, Quantitative Insulin Sensitivity Calculation Index (QUICKI), vascular cell adhesion molecule cell (VCAM)-1, and plasminogen activator inhibitor (PAI)-1 values [94]. Various Lactobacillus and Bifidobacterium species in probiotic yogurts have been shown to lower cholesterol; regulate plasma glucose levels; have antioxidant, anti-inflammatory, and microbiota modulation effects; and have positive effects on cancer and ulcerative colitis [95,96,97,98,99,100]. El-Dein et al. [101] compared fermented yogurt with Lactiplantibacillus plantarum KU985438 or Lacticaseibacillus rhamnosus KU985439 and found that Lacticaseibacillus rhamnosus KU985439 provided a greater reduction in blood glucose, triglycerides, total lipids, total cholesterol, triglycerides, NF-κB expression, and lipid peroxidation [101]. Similarly, Gu et al. [15] found that probiotic yogurt (S. thermophilus ST447, Lactobacillus acidophilus NCFM, Lacticaseibacillus rhamnosus GG, and B. lactis HN019) decreased blood glucose, glycated hemoglobin (HbA1C), HOMA-IR, insulin, low-density lipoprotein cholesterol (LDL-C), and LPS levels, and increased PYY in mice. Yogurt increased the levels of butyric and acetic acids and Lactobacillus and Streptococcus bacterial species [15]. In addition, probiotic yogurts have been shown to have a positive effect on diarrhea in adults and children [102,103]. There are also studies showing that probiotics and conventional yogurt are not effective in glucose control in cardiovascular risk factors, diabetes, and obesity [104,105]. The effect of storage time on bacterial counts in probiotic yogurts was also evaluated. Lacticaseibacillus casei, which has probiotic properties in yogurt, remained at more than 108 CFU/g at the end of the 21 days of storage [106]. In another study, the Lactobacillus bulgaricus content was found to be 8.13 log cfu/g on the 1st day of storage and 7.51 log cfu/g on the 28th day [107]. In the study by Mari lopez et al. [108], the number of S. thermophilus decreased between 1.8 and 3.5 log during storage. Although the probiotic bacteria content decreased, they maintained a content of ≥107 cfu/mL at the end of 3 weeks. The vitality of probiotic bacteria in yogurts varied; Lactobacillus acidophilus ≥ 107 cfu/mL was maintained for 35 days, Lacticaseibacillus casei was maintained for 7 days, and Limosilactobacillus reuteri was maintained for 14 days [108].
Bioactive peptides, which are one of the components formed by the fermentation process in yogurts, can show antioxidant, antibacterial, angiotensin-converting enzyme (ACE) inhibitor, opioid antagonist, antihypertensive, and immunomodulatory effects [109,110,111]. When three groups of yogurts were used as starter cultures, Lactobacillus acidophilus 20552 ATCC and Lactobacillus helveticus CH5 were evaluated; Lactobacillus acidophilus 20552 ATCC and Lactobacillus helveticus CH5 had variable proteolytic activity. However, the peptides obtained from yogurt containing Lactobacillus helveticus CH5 showed the highest antioxidant and antimicrobial effects. All yogurts showed antimicrobial activity against Escherichia coli [109]. In another study, the addition of Lactobacillus helveticus CH 5 to yogurt increased the ACE inhibitory effect compared to normal yogurt. In particular, it supported the formation of bioactive αS1-casein (CN) f(24–32) and β-CN f(193–209) peptides [112]. The β-CN (94–123) peptide fraction in yogurt may provide intestinal homeostasis by increasing the expression of intestinal mucin (Muc2 and Muc4) and antibacterial factors (lysozyme and rDefa5) depending on the dose [113].
Storage time is one of the factors affecting the level of bioactive peptides in yogurt. The proteolytic activity of yogurt increased significantly after 14 days of storage and showed ACE-1 activity, antithrombotic activity, cholesterol-lowering activity, and antioxidant activity. All of these activities were found to be the highest in yogurt that was stored in a cold environment for 14 days with skim milk powder and trypsin added [114]. Heydari et al. [115] showed that proteolysis, antimutagenic, and 2,2-diphenyl-1-picrylhydrazyl (DPPH) antioxidant activity were found to be the highest at the end of the 28th day in yogurt that was obtained using Saccharomyces thermophilus + Lactobacillus bulgaricus + Iranian strain of Bifidobacterium lactis species [115]. The enrichment of yogurt with whey proteins, the proteinase and peptidase activities of the added coculture (probiotic bacteria), and the addition of trypsin were effective in the formation and increase in the amount of peptides with antihypertensive, antioxidant, antimicrobial, hypocholesterolemic, antimutagenic, and cholesterol-lowering effects [114,115]. Macro- and micronutrients, bioactive peptides, and probiotic bacteria species all have roles in the health benefits of yogurt.

2.3. Cheese

Cheese has been produced and consumed for many years. There are 1500 cheese varieties defined in the world. The microorganism content of cheese varies based on the milk used, cheese type, and production [116]. The pH changes that occur during the production and ripening of cheese are heavily influenced by both lactic acid bacteria and yeasts [117]. A meta-analysis study evaluating cheese consumption and its health effects found that cheese consumption (especially 40 g/day) had neutral to moderate benefits for human health, and was moderately inversely connected with all-cause mortality, cardiovascular disease, coronary heart disease, and stroke incidence. It also showed a negative association with the risks of type 2 diabetes and dementia. It is emphasized that these effects are due to the nutrients and bioactive components in cheese [118]. Hu et al. [119] also found that cheese consumption was connected with a reduced risk of type 2 diabetes, coronary artery disease, ischemic stroke, heart failure, and hypertension [119]. There were also inconclusive results showing no connection between cheese consumption and cardiovascular disease risk, and unclear results suggesting that it may be associated with increased and decreased risks. It was suggested that the potential effect of cheese on cardiovascular disease may be due to its calcium content, high protein content, fermentation, and fatty acid content (CLA) [120].
One of the components of cheese that have positive effects on health is bioactive peptides. The type of milk used during cheese ripening, starter culture, and native milk microbiota affect the bioactive peptides formed. Bioactive peptides are composed of certain protein fragments and offer various advantages for regulating bodily processes [121,122,123].
Bioactive peptides in different types of cheese have been shown to have antioxidant, antihypertensive (ACE inhibitory), antimicrobial, and dipeptidyl-peptidase-IV (DPP-IV) inhibitory activity effects [123,124,125,126,127]. In one study, Lactobacillus helveticus A1, which releases the peptide as a key factor in ACE inhibition, was the strain with the strongest ACE inhibitory activity [128]. The number of peptides with angiotensin-converting enzyme (ACE), dipeptidyl peptidase-IV (DPP-IV), and antioxidant activity increased with ripening [129]. The proteolysis of cheese for more than 90 days resulted in increased antioxidant activity. Bioactive peptides derived from αs1-casein and β-CN were detected in cheese. The radical scavenging activity, reducing power, chelate capacity, and ACE inhibition effects of cheese extract derived from these peptides were revealed [124]. In another study on bioactive peptides obtained from whey, peptides were found to have antioxidant and ACEI inhibitory effects [125]. The use of ultrasonic, high-pressure, and microwave pretreatments to the milk used in cheese making affected proteolysis both during cheese making and ripening. These treatments increased the ACE inhibitory activity and antioxidant (total flavonoids, total phenolics, total antioxidants, and DPPH radical scavenging activity) activity of the cheese [130]. The consumption of 30 g/day of Grana Padano cheese reduced blood pressure (systolic and diastolic pressure) in people with hypertension after two months [131]. Helal et al. [123] used six cheese varieties and found that Gouda cheese showed the highest antioxidant, ACE-inhibitory and DPP-IV-inhibitory activity. The findings suggested that 10–20 g of Gouda cheese, 50–100 g of Domiati, and 100 g of Edam cheese might be sufficient to exhibit an antihypertensive effect [123]. The salt content, packaging type, and storage time of the cheeses can also affect ACE inhibitor and antioxidant activities. The highest antioxidant activity was found on the first day of vacuum packaging, at a 1% NaCl concentration, and decreased with the increasing storage time. The highest ACE inhibitor activity and peptide concentration were also found on the seventh day of vacuum packaging with no added salt or 1% NaCl [132]. In another study in which the effect of storage time on bioactive peptides in cheeses was evaluated, an increase in the amount of some bioactive peptides in cheeses occurred in long storage times. At 90 days of storage, αS1-CN f(24–32) peptides increased in vacuum packaging and αS1-CN f(1–16–32) and αS1-CN f(17–22) peptides increased in modified atmosphere packaging [133].
The other compound of cheese associated with health is conjugated linoleic acid. The CLA content in cheeses was found to be 0.44 to 1.04 g/100 g of fat in the study by Donmez et al. [134], and 7.5 to 7.9 mg/g of fat in the study by Luna et al. [134]. It was shown that there is an increase in the CLA levels during cheese ripening, but increased storage time decreases the concentration [135]. CLA may have antidiabetic, anticancer, anticarcinogenic, anti-atherosclerotic, antihypertensive, and endothelial function effects [136,137,138].
The development of biogenic amines may occur in cheese as a result of the bioactivities of some microorganisms. Histamine, cadaverine, putrescine, spermine, spermidine, and tyramine biogenic amines have been detected in different cheese types. High levels of biogenic amine consumption have some drawbacks. For example, histamine has effects such as nausea, vomiting, diarrhea, and stomach upset, while tyramine may cause hypertensive effects and may have a negative relationship with monoamine oxidase inhibitors (MAOIs) [139]. It was emphasized that Lacticaseibasillus casei 4a and 5b isolated from cheese reduced tyramine and histamine accumulation, and therefore may be suitable to be used as co-cultures in order to lessen the amount of biogenic amines [140]. In addition, some Lactobacillus species isolated from cheese may have probiotic, antimicrobial, and antioxidant effects [141,142,143].
It is emphasized that the potential health effects of cheese are due to bioactive peptides, conjugated linoleic acid, calcium, bacteria with probiotic properties, and some prebiotic effects. Due to these components, it is stated that it may be effective in conditions such as blood pressure, diabetes, cardiovascular diseases, and diabetes [144].

3. Fermented Meats

Red meat is any unprocessed mammalian muscle flesh, including frozen or minced meat (such as cattle, veal, pork, or lamb). Meat that has undergone salting, fermenting, smoking, curing, or other methods so as to improve preservation or flavor is well known as processed meat. Pork or beef are typically found as processed meats [145]. The majority of the time, it is agreed that meat and its products provide excellent and high biologic values of proteins, B group vitamins, minerals, trace elements, and some other bioactive components [146].
The process of fermentation is passed down from generation to generation [4]. Different substances are produced by the fermentation technique, which is also preferred in meat products. Carboxylic acids, lactic acid, aldehydes, pyruvic acid, alcohols, and ketones are just a few of the substances that are created during this transition [147]. The fermentation of meat products involves the use of starting cultures and live organisms. These organisms are responsible for carrying out fermentation, reducing pathogenic bacteria, and ensuring the development of appropriate organoleptic qualities in the manufacturing of fermented meat products [148,149,150]. The fermentation process makes use of many types of bacteria and yeast [151]. Listeria monocytogenes levels are reduced, and the food safety and shelf life of fermented meat products are improved as a consequence of the bacteria of lactic acid in meats [152]. Nham is a fermented sausage that is only found in Thailand. The bacteria of lactic acid are utilized in this sausage type. Foods have antibacterial properties thanks to the bacteriocins and organic acids produced during fermentation [153]. Gram-positive cocci (Staphylococcus carnosus and Staphylococcus xylosus), yeast (Debaryomyces hansenii and Candida famata), and mold (Penicillium nalgiovense and Penicillium camambertii) are also utilized in fermentation in addition to lactic acid bacteria [147]. Latilactobacillus sakei, Weissella, Staphylococcus equorum, Debaryomyces hansenii, Kurtzmaniella zeylanoides, Wickerhamomyces subpelliculosus, and Zygosaccharomyces rouxii are the predominant bacteria found in Portuguese fermented sausages. Nitrogen compounds, acids, alcohols, aliphatic hydrocarbons, aldehydes, lactones, pyrans, ketones, terpenoids, esters, sulfur compounds, aromatic hydrocarbons, phenols, and furans are formed as volatile organic compounds as a result of microbiological reactions in these fermented sausages [154]. The most often isolated bacterial species in salami are Lactobacillus and Staphylococcus, but the Gammaproteobacteria phylum, Moraxellaceae family, Acinetobacter, Pseudomonas, Carnobacterium, and Enterococcus are also present [155].
The enzymatic hydrolysis and fermentation of starting cultures produce bioactive peptides. Both meat products and the gastrointestinal system after intake contain these peptides [156]. In microbial activities that take place in fermented meat products, biogenic amines are produced that are important for food safety and quality. The biogenic amines putrescine, histamine, cadaverine, and tyramine, as well as tryptamine and bphenylethylamine, were found in a study on fermented sausages [157,158]. Although the Bifidobacterium longum species, which participates in the fermentation process and has probiotic properties as well [159], inhibits the creation of cadaverine from biogenic amines, and an increase in these amine species also leads to toxicity and the formation of N-nitrosa compounds [160,161]. When nitrites are present, biogenic amines can be transformed into nitrosamines. Secondary amines and NO can combine to generate far more durable carcinogenic nitrosamines than primary amines. Salami samples contain N-Nitrosodimethylamine, N-Nitrosopyrrolidine, N-Nitrosodipropylamine, and N-Nitrosomethylethylamine kinds [162]. N-nitrosoethylmethylamine, N-nitrosodimethylamine, N-nitrosopiperidine, N-nitrosopyrrolidine, N-nitrosodiethylamine, N-nitrosodi-n-propylamine, and N-nitrosomorpholine are the principal nitrosamines found within the majority of products of fermented meat [161,163]. Figure 1 shows the changes throughout the meat fermentation.
In the ripening of fermented sausages, Lactobacillus rhamnosus CTC1679 predominates, and it briefly colonizes the gastrointestinal system [164]. The lactic acid bacteria Pediococcus pentosaceus KL14, KL10, KL11, and KL14 that are isolated from fermented pork flesh have high radical scavenging abilities. The ouperoxide dismutase activity is also strong in some lactic acid species [165]. Strong proteolysis that occurs during fermentation may eventuate in the occurrence of peptides that have ACE inhibitor and antioxidant properties. Belgian samples have stronger radical scavenging activity and a ferric reducing impression, while Belgian and Spanish dry fermented sausages exhibit an ACE inhibitory effect [166]. Carcinogenic, teratogenic, and mutagenic effects can be caused by N-nitrosamines that are produced during the fermentation of meat products [167]. Table 2 lists the effects of fermented meat and certain bioactive compounds on health.
As for fermented fish products, there are more studies examining the health effects of fish rather than fermented fish. However, studies examining the effects of fermented fish products on health are not sufficient. In a study conducted this year (2023), it was suggested that novel dipeptidyl peptidase-IV inhibitory peptides (D4IPs) discovered in fermented mandarin fish may alleviate the active amino acid sequences of type 2 diabetes mellitus [168]. And it is stated that Lactiplantibacillus plantarum, Pediococcus pentosaceus, Pediococcus acidilactici, Pediococcus lolii, Enterococcus hirae, and Enterococcus lactis among the lactic acid bacteria isolated from Shindal, a traditional fermented fish food, have probiotic properties [169].
Table 2. Effects of fermented meat and certain bioactive compounds on health.
Table 2. Effects of fermented meat and certain bioactive compounds on health.
Fermented FoodsCertain Bioactive CompoundsEffects on HealthReferences
Intestine
Fermented mutton jerkyx3-2b Lactiplantibacillus plantarum and composite bacteriaPurine content of fermented mutton jerky by x3-2b Lactobacillus plantarum and composite bacteria
In vitro digestion, decreasing purine content by 37x-3 Pediococcus pentosaceus
[170]
Cured beef-Gastric protein carbonylation
Colonic Ruminococcaceae
Cecal propionate
TBARs and diacetyl in feces
Levels of cecal butyrate, fecal phenol, dimethyl disulfide
Level of fecal carbon disulfide
Colonic Ruminococcaceae
[171]
Fermented sausageEnterococcus faecium CRL 183Lactobacillus spp. in ascending colon, transverse colon, and descending colon
Bacteroides spp. in descending colon
Enterobacteriaceae in transverse colon and descending colon
Colonic ammonium ions
Butyric acid concentration in transverse colon, ascending colon, and descending colon
Concentration of propionic acids in ascending colon and transverse colon
Concentration of acetic acid in ascending colon, transverse colon, and descending colon
[172]
Fermented sausage-Release of free iron in digestive system
Concentration of gastric N-nitrosamine
[173]
Fermented sausageEnterococcus faecium S27Transfer of tetracycline resistance determinant (tet(M)) to E. faecium and Enterococcus faecalis 
Transfer of Enterococcus faecium’s streptomycin resistance
[174]
Fermented sausageBologna sausage (a)
Dry fermented sausage (b)
Calcium transporter in Caco-2 cells: in (a) , in (b) [175]
Fermented salamiPlant extracts Phenol and p-cresol in colon
Acetate, propionate, butyrate in colon
Enterobacteriaceae 
Bifidobacteriaceae
[176]
Fermented fishStaphylococcus sp. DBOCP6Non-hemolytic and non-pathogenic effects against broad and narrow spectrum antibiotics
Ability to adhere to the intestinal wall
[177]
Cardiovascular diseases and ACE-I inhibitory effects
--Cardiovascular disease risk,
stroke risk
Total mortality risk
[178]
Salami
Sausage
Cardiovascular disease risk [179]
-Cardiovascular disease risk [180]
-Total stroke incidence
No association between ischemic stroke and coronary heart disease mortality
[181]
Bacon
Sausage
-Cardiovascular death risk
Ischemic heart disease risk
[182]
Dry-cured pork ham-Levels of total cholesterol, LDL, basal glucose [183]
Semi-dry fermented camel sausageLactiplantibacillus plantarum KX881772Inhibition of ACE
Cytotoxicity activity towards Caco-2 cell line
α-amylase inhibition
α-glucosidase inhibition
[184]
Fermented pork sausageStaphylococcus simulans NJ201
Lactiplantibacillus plantarum CD101
ACE inhibition
Superoxide anion scavenging activities
Ferric-reducing antioxidant activity
[185]
Dry fermented camel sausageStaphylococcus xylosus and Lactiplantibacillus plantarum
Staphylococcus caarnosus and Latilactobacillus sakei
Staphylococcus xylosus and Lactobacillus pentosus
Antioxidant capacity by <3 kDa peptides
Maximum ACE inhibition by <3 kDa peptides
Maximum ACE inhibition in sausages with S. xylosus and L. plantarum
[186]
Dry-cured ham-ACE inhibition
Radical scavenging activity
PAF-AH inhibitory effect
[187]
Fermented meat-Antioxidant activity against OH-radical by GlnTyr-Pro [188]
Dry-fermented sausageStarter culture (P200S34) and protease (EPg222)ACE inhibition
Antioxidant activity
[189]
-Risk of cardiovascular mortality, stroke, myocardial infarction via reduction in processed meat [190]
-Risk of all-mortality cause and cardiometabolic disease via lower consumption [191]
-Risk of heart failure [192]
Cancer
-Risk of colon cancer, rectal cancer, breast cancer, lung cancer, and colorectal cancer [193]
Ham
Sausage
Bacon
-Breast cancer risk [194]
-Weak positive association with breast cancer[195]
-Breast cancer risk with diet rich in processed meat [196]
Ham
Sausage
Bacon
-Gastric cancer risk [197]
Ham
Sausage
Bacon
-Colorectal cancer risk [198]
-Colorectal cancer risk with lower consumption [199]
-Colorectal cancer risk with lower consumption [200]
-Colorectal cancer risk [201]
Ham
Sausage
Bacon
-Colorectal cancer risk [202]
Ham
Sausage
Bacon
-Colorectal cancer risk [203]
-Colorectal adenoma risk [204]
Ham-Risk of renal cell carcinoma
Risk of bladder cancer
[205]
Ham
Sausage
Bacon
-Bladder cancer risk [206]
Ham
Sausage
Bacon
-Minimal connection to kidney cancer risk [207]
Ham
Salami
Sausage
Bacon
-No association with gliomas[208]
Risk of hepatocellular carcinoma [209]
Other diseases
-Risk of type 2 diabetes [210]
Bacon
Salami
Sausages
-Risk of diabetes as well as stroke and coronary heart disease [211]
-Risk of type 2 diabetes [212]
-Type 2 diabetes risk [213]
-Gestational diabetes mellitus risk [214]
-No change in Crohn’s disease flares[215]
-Risk of mortality via increase in consumption [216]
-Mortality risk of all causes (except cancer) and cardiovascular-caused mortality [217]
-Depression risk [218]
N-NitrosodimethylamineNo change in glioma[219]
DiethylnitrosamineProbability of hepatocarcinogenesis[220]
: increased, : decreased, LDL: low-density lipoprotein, ACE: angiotensin-converting enzyme, kDa: kilodalton, PAF-AH: platelet-activating factor acetulhydrolase, Gln Tyr-Pro: Glycine Tyrosine-Proline.

4. Fermented Vegetables and Fruits

Fruits and vegetables have important health effects due to their contents of fiber, vitamins, minerals, phenolic compounds (flavonoids, sulfur compounds, phytoestrogens, and monoterpenes), and bioactive peptides [221]. Their consumption can contribute to the prevention of many chronic diseases such as diabetes, cardiovascular diseases, and cancer [222,223]. It has been reported that the daily consumption of five servings of vegetables and fruits can reduce mortality in diseases [224]. Another way of consuming vegetables and fruits is in their fermented form. Resulting from the lactic acid fermentation of vegetables such as cucumbers, cabbages, capers, carrots, and tomatoes, different products such as kimchi, pickles, turnips, Pak-Gard-Dong, and Dhamuoi are obtained [31,225]. In addition, fermentation has recently been emphasized for the utilization of waste and by-products of vegetables and fruits (pineapple peel, orange peel, mango seed, etc.) [226].
As a result of fermentation, the riddance of anti-nutritional factors; the formation of metabolites with positive effects (bioactive peptides and exopolysaccharides); the improvement in bioavailability through the hydrolysis of polymers (esters of phenolic compounds); increased vitamins, minerals, and phenolic compounds; and the presence of bacteria with probiotic properties and prebiotic effects lead to positive effects on health [221].

4.1. Fermented Vegetables

Vegetables are a significant part of a healthy diet. Low vegetable consumption leads to negative health effects [227]. One of the ways vegetables are consumed is in their fermented form. Mostly lactic acid and alkaline fermentation occurs when vegetables are fermented [228]. Lactic acid fermentation can occur in vegetables when conditions are suitable (anaerobic conditions, suitable temperature, and humidity and salt concentrations). The products created as a result of vegetable fermentation vary between nations. For example, in Europe, sauerkraut is formed based on fermentation of cabbage, while in Korea, kimchi is formed as a result of fermentation of cabbage, green onions, etc. [229]. This section focuses on kimchi and sauerkraut, which have more scientific data on bioactive components and health effects.
A traditional Korean vegetable dish, kimchi (kimchi cabbage), is produced through the fermentation of radish, cucumber, and other vegetables by lactic acid bacteria [230]. Kimchi contains fiber, vitamins (ascorbic acid, etc.), minerals, 3-(4′-Hydroxyl-3′,5′-dimethoxyphenyl) propionic acid (HDMPPA), capsaicin, allyl compounds, isothiocyanate, indole compounds, and thiocyanate [231,232]. In another study using kimchi methanol extract (HDMPPA, quercetin, ascorbic acid, and capsaicin) and kimchi bioactive components, antioxidative (nuclear factor (erythroid-derived 2)-like 2 (Nrf2), SOD1, and GPx increased) and anti-inflammatory (NF-κB, inducible nitric oxide synthase (iNOS), and cyclooxygenase 2 (COX-2) decreased) activities were found to improve cognitive function in mice with amyloid beta (Aβ)25-35-induced Alzheimer’s [233]. The active ingredient of kimchi, HDMPPA, has shown antioxidant, anti-inflammatory, and anti-atherosclerotic effects by lowering cholesterol; reducing cyclooxygenase-2 and ROS levels, lipid peroxidation, and lipid accumulation; and suppressing NF-κB, mitogen-activated protein kinase (MAPK), and phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathways and oxidative stress [234,235,236,237].
Bacteria isolated from kimchi have probiotic, anti-inflammatory, antioxidant, anti-obesity, antidiabetic, antimicrobial, and immune system effects [238,239,240,241,242,243,244,245,246] (Table 3). Not only the bacteria and metabolites derived from kimchi, but also the dietary consumption of kimchi have been shown to have positive effects [247]. Dietary kimchi consumption had a positive effect on C26 adenocarcinoma-induced cancer cachexia by inhibiting IL-6, inhibiting lipolysis, and increasing lipogenesis. It has also been shown to improve cachexia-induced muscle atrophy and reduce NF-κB, extracellular signal-regulated kinase ½ (ERK ½) activation, AKT, mammalian target of rapamycin (mTOR), and PI3K catabolism levels. It also decreased tumor size and tumor mass [247].
Kim et al. [252] showed that by altering the gut–brain axis, kimchi lowered obesity-related neuroinflammation. Kimchi reduced adipose tissue gain, serum free fatty acids, monocyte chemoattractant protein-1 (MCP-1), and TNF-α levels. It also reduced hypothalamic neuroinflammation and hypothalamic apoptotic protein expression. It was effective in the improvement of gout dysbiosis. Claudin-5, occludin, total short-chain fatty acids, acetate levels, and Akkermansia muciniphila colonization increased [252]. There are studies suggesting that kimchi may have a positive effect on colon adenoma, irritable bowel syndrome (IBS), and obesity through its effect on the microbiota composition [253,254,255]. In the case of IBS, kimchi consumption was reported to improve IBS symptoms by increasing fiber intake, controlling immunity, and inhibiting harmful intestinal enzyme activity (β-glucosidase and β-glucuronidase) [253]. Kimchi consumption may also affect diabetes by reducing insulin resistance and HbA1c; increasing insulin sensitivity, QUICKI, and β-cell function; and improving glucose tolerance [256,257]. According to all of this research, kimchi may benefit health due to its bioactive ingredients and probiotic microorganisms.
Sauerkraut is another fermented vegetable product that has been studied. Similar to kimchi, the bacteria isolated from sauerkraut, exopolysaccharides, and the bioactive compounds formed as a result of fermentation have shown antioxidant, antibacterial, and immunomodulatory effects [248]. It was also revealed that the Lactiplantibacillus plantarum strain isolated from sauerkraut may have a probiotic effect. Lactiplantibacillus plantarum S4-1 decreased the total cholesterol, triglyceride, and LDL-C contents [251]. The fermentation of sauerkraut resulted in the formation of ascorbigen, indole-3-carbinol, and the degradation of glucosinolates [258]. Ascorbigen may show antioxidant properties [259], while indole-3-carbinol can show antioxidant, anti-inflammatory, anti-obesity, antidiabetic, anti-atherosclerotic, anticancer, antihypertensive, and neuroprotective effects [260].
In adolescents and adults, high servings of raw/cooked cabbage/sauerkraut (>3 servings/week) were associated with a lower risk of breast cancer than low servings (≤1.5 servings per week) [261]. The consumption of pasteurized and unpasteurized sauerkraut improved the irritable bowel syndrome symptom severity score in individuals with irritable bowel syndrome, and the consumption of unfermented sauerkraut increased sauerkraut-associated LAB (Lactiplantibacillus plantarum and Levilactobacillus brevis) in feces [262].
Similar studies were carried out in fermented cucumber products with kimchi and sauerkraut. It was revealed that the bioactive peptides formed in lactic acid-fermented cucumber showed an ACE inhibitory effect [263]. When the change in the free amino acid profile of cucumbers as a result of fermentation was analyzed, the highest amino acids in fresh cucumbers were glutamine, GABA, arginine, citrulline, and asparagine. As a result of fermentation, the concentrations of leucine, isoleucine, methionine, lysine, phenylalanine, histidine, tyrosine, proline, and ornithine amino acids increased, while the glutamine, GABA, and aminoadipic acid concentrations decreased. Increasing the salt concentration (6%) increased the arginine concentration [264].
Plant-based foods contain micronutrients, macronutrients, and bioactive components as well as anti-nutrient components. These alleged anti-nutrients, which also include tannins, phytoestrogens, lectins, oxalates, and phytates, are thought to limit the absorption of important nutrients. However, it has also been suggested that it may have health-promoting effects [265]. It has been shown that lectin, phytate, tannin, and oxalate levels decrease as a result of fermentation compared to fresh produce [266,267,268]. As a result of lactic acid fermentation in the African nightshade plant, the tannin level decreased by 76.27–92.88%, and the oxalate level decreased by 77.33–90% [267]. Similarly, as a result of the lactic acid fermentation of white cabbage sprouts, the amounts of phytate, tannin, and oxalate decreased by 42, 66, and 53%, respectively [269].

4.2. Fermented Fruits

Fruits show positive effects on health due to their fiber, low energy density, vitamin/mineral (potassium, vitamin C, etc.), and phytochemical (polyphenols and carotenoids) contents [270]. There are studies evaluating the health effects of the fermentation of many fruits. Studies on fermentation in fruits such as apples, mangoes, papayas, lemons, and citrus have been carried out [271,272,273,274,275]. Fermented fruits can be made from fruits mainly based on lactic acid and acetic acid fermentation [276,277]. The use of lemon-fermented products resulting from the fermentation of lemon with Lactobacillus OPC1 decreased the total triglycerides and total cholesterol in the liver and regulated the lipid metabolism and gut microbiota in rats [275]. It was revealed that fermented papaya products may exhibit immunomodulatory, antioxidant, anticancer, anti-inflammatory, antidiabetic, and antidyslipidemic properties. Fermented papaya decreased pro-inflammatory cytokines and pro-oxidant components [271]. Lactobacillus acidophilus (BCRC14079)-fermented mango peel decreased Aβ accumulation, a neuronal protective product, by inhibiting oxidative stress and increasing BDNF expression in neural cells [273].
The fermentation of fruits using selected probiotic strains resulted in beneficial sensory and health effects. Yang et al. [276] found that the fermentation of apple juice with Lactobacillus acidophilus, Lacticaseibacillus casei, and Lactiplantibacillus plantarum bacteria increased the antioxidant and antibacterial capacities of apple juice. The total amino acid content and lactic acid content increased, while the total phenolic acid content decreased. The gallic acid, protocatechuic acid, and catechin concentrations increased with fermentation, but the total phenolic acid content decreased with the effect of storage [276]. As a result of the fermentation of cherry juice using nine Lactobacillus strains, Lactobacillus acidophilus 150 and Limosilactobacillus fermentum DT41 fermentations increased the polyphenol contents compared to the baseline [278]. Dragon fruit fermentation (Lactiplantibacillus plantarum FBS05) increased antibacterial and antioxidant activities [279]. Cirlini et al. [280] evaluated the organic acid content in elderberry fruit and found lactic acid as the main organic acid. Malic acid found in the fruit before fermentation decreased with fermentation, the amount of citric acid varied according to the bacteria, and tartaric acid was also found in fermented juices [280]. Kiwifruit juice fermentation (Lactobacillus acidophilus 85 (La85), Lactobacillus helveticus 76 (Lh76), and Lactiplantibacillus plantarum 90 (Lp90)) resulted in the formation of protocatechuic acid and catechins. Protocatechuic acid was the highest in Lh76 fermentation. These compounds have antioxidant effects. Caffeic acid was not detected as a result of fermentation [281]. Wu et al. [282] showed that the fermentation of blueberry and blackberry juices with Lactiplantibacillus plantarum, Bifidobacterium bifidum, and Streptococcus thermophilus resulted in a decrease in the anthocyanin levels. Lactiplantibacillus plantarum has a higher capacity to metabolize phenolic acids and organic acids. The highest lactic acid, syringic acid, and antioxidant capacity were exhibited as a result of fermentation with this microorganism [282]. The pH, acidity, total and reducing sugars, organic acid, and total phenolic contents of blueberry juice were significantly altered via probiotic fermentation, which had an effect on the juice’s physicochemical, anti-inflammatory, antibacterial, and antidiabetic qualities [283].
Fruit vinegars are among other fermented products obtained from fruits. Two different fermentations can occur in vinegar: alcoholic and acetic acid fermentation. The microbiota leading to vinegar production varies. Acetic acid can be produced in large quantities by the Acetobacter and Komagataeibacter species [277]. Vinegar is obtained from many fruits such as grapes, apples, pomegranates, and blueberries. Polyphenols and organic acids, especially acetic acid, can show beneficial effects in fruit vinegar [284]. Bioactive components such as catechins, p-hydroxybenzoic acid, gallic acid, syringic acid, caffeic acid, p-coumaric acid, and chlorogenic acid have been provided in different kinds of vinegar [285,286]. Apple cider vinegar has been found to have hypocholesterolemic, antidiabetic [287,288,289], antioxidant [288,290], antimicrobial, anti-inflammatory [291], and anti-obesity effects [290] and improve cognitive function [292] reproductive function, and liver function [293]. Apple cider vinegar may exert antidiabetic effects by decreasing HOMA-IR, fasting blood glucose, HOMAβ, and OUICKI levels [287,288,289,293]. It may be effective in hypercholesterolemia by improving the total cholesterol, triglyceride, LDL cholesterol, total cholesterol/HDL-C, and LDL-C/HDL-C levels [287,289,290,293], and in non-alcoholic fatty liver disease by improving hepatic enzymes and reducing steatosis and inflammation in the liver [289,293]. It also has effects on neurodegeneration, ovulation, and obesity. The impact of apple cider vinegar on neurodegeneration was studied by Tripathi et al. [292]. As a result of vinegar consumption in mice, a decrease in the Morris water maze escape time, an increase in the time spent in the target quadrant, a decrease in acetylcholinesterase (AChE) and Malondialdehyde (MDA) levels, and an increase in glutathione (GSH) and SOD levels were found. Thanks to these effects, it was emphasized that it may be effective in dementia and Alzheimer’s, and that this effect in apple cider vinegar would be related to its polyphenol, flavonoid, and organic acid contents [292]. Shams et al. [293] found that apple cider vinegar increases estradiol levels and may have a positive effect on ovarian reserve by increasing the number of primordial and primary follicles [293]. Vinegar may work in obesity by reducing body weight and food intake and delaying gastric emptying [290,294]. Intervention with different vinegars (pomegranate, prickly pear, or apple) decreased body weight; body weight gain; total visceral adipose tissue; mesenteric, epididymal, and perirenal fat; total cholesterol; plasma cardiac biomarkers (creatine kinase-MB isoenzyme (CK-MB), lactate dehydrogenase (LDH), alanine aminotransferase (ALT), and aspartate aminotransferase (AST)); plasma inflammatory markers (CRP, homocysteine, and fibrinogen); plasma and visceral adipose tissue leptin; and TNF-α levels and increased adiponectin levels in obese rats. All of these results suggest that obesity may affect cardiometabolic symptoms [295].
Another fermented fruit product is wine. The fermentation of grape in wine is a process that involves yeast and LAB acting together. Two common fermentation processes are emphasized: Alcoholic fermentation with yeasts, and malolactic fermentation occurs as a result of bacteria [296]. In the analysis of wine, catechins, p-coumaric acid, resveratrol, rutin, quercetin, myricetin, anthocyanin, tannins, flavan-3-ol, and phenolic acids (caffeic acid, ellagic acid, syringic acids, 2.5-dihydroxybenzoic acid, vanillic acid, and ferulic acid) are found [297,298,299]. There is a significant relationship between the polyphenol content and antioxidant activity in wines [297]. Other compounds formed as a result of fermentation in red wine are melatonin [300,301] and hydroxytyrosol [297,302]. Red wine polyphenols have antioxidant [303,304,305], antibacterial [306], anti-inflammation, anticancer [307,308,309,310,311,312,313], antidiabetic [308], antithrombotic, antidepressant [314,315], and neuroprotective effects [309]; are involved in the regulation of bone mineral density [316], microbiota modulation [305,310,317], and micro RNA regulation [308]; effect adipose tissue, hypocholesterolemia [318], and the regulation of endothelial function [319]; and have anti-obesity effects [320,321,322] (Figure 2). In addition, polyphenols have also shown positive effects on ulcerative colitis through microbiota modulation (Akkermansia increase), anti-inflammatory effects, the inhibition of the PI3K/Akt pathway and the hypoxia-inducible factor-1 (HIF-1α)- T helper 17 (Th17) pathway, and the reduction in vascular endothelial growth factor A (VEGFA) [323,324,325].
Melatonin, one of the other molecules in red wine, has anti-inflammatory, anti-inflammation, immunomodulatory, antioxidant, antiapoptotic, maternal/fetal health, cardiovascular, neuroinflammation, and respiratory health effects [326,327,328,329]. Hydroxytyrosol may be effective in cardiovascular disease (CVD), Parkinson’s disease, Alzheimer’s disease, diabetes, metabolic syndrome, cancer, and osteoporosis with the enhancement of AMP-activated protein kinase (AMPK), Sirtuin 1 (SIRT-1) signaling pathways, antioxidant and anti-inflammatory effects, decreased mitochondrial dysfunction, increased epigenetic regulation, and anticancer and anti-hypocholesterolemic effects [330,331,332,333].

5. Fermented Legumes

Plants of the Leguminosae family make up legumes. Peas, chickpeas, lentils, soybeans, and peas are examples of edible legumes [334]. Because of the important and nourishing bioactive compounds that legumes contain, they are essential for human nutrition [335]. They are all rich in protein, fat, carbohydrate, and minerals [334,336]. Additionally, they have a lot of fiber, vitamins from the B group, and useful phytochemicals with biological effects [335]. Galactooligosaccharides, lectins, saponins, and tannins are examples of readily accessible non-nutrient molecules that are common sources of protease inhibitors [337]. They are significant sources of isoflavones with estrogen-like properties and effects on calcium [338].
Due to the inclusion of non-nutrient components, legumes have a limited potential to be digested and bioavailable. It is suggested to use soaking, boiling, or fermentation to improve digestion and bioavailability. Thus, especially with fermentation, non-nutritious foods are changed into substances with nutritional value [339]. Antioxidant chemicals produced by the fermentation of soybeans include furanones, peptides, 3-hydroxyanthranilic acid, and melanoidins. Additionally, bioactive substances with anti-inflammatory effects include isoflavone, butyric acid, 2S albumin, α-linolenic acid, soy sauce polysaccharides, and glycones [340]. The fermentation process uses microorganisms to catalyze metabolic reactions and produce bioactive chemicals [341]. A traditional Korean dish, cheonggukjang, is produced in the wake of the fermentation of soybeans. Bioactive compounds not found in raw soybeans are produced during fermentation as a result of isoflavones such as phenolic acids, genistein, phytic acids, saponins, daidzein, and trypsin inhibitors [342,343].
Legumes are fermented using starter cultures composed of lactic acid bacteria. Examples of lactic acid bacteria include Lactiplantibacillus plantarum subsp. Plantarum, Streptococcus thermophilus, Leuconostoc mesenteroides Lactobacillus acidophilus, and Lactobacillus delbrueckii ssp. bulgaricus Lacticaseibacillus casei. In fermented legumes, these bacteria both perform fermentation and provide a source of probiotics [344]. The most significant fermented legumes employed in soy products are the bacteria of lactic acid, which are the starting bacteria used for the occurrence of soy milk and tofu [345]. Fermented soybeans contain Bacillus species as well the bacteria of lactic acid, and Bacillus genome sequences have been discovered in these foods [346].
There are many products made from fermented legumes. Natto, fermented soy milk, and tempeh have been a part of traditional Asian meals for generations, but they are now available for consumption all over the world [347]. Miso, a Japanese fermented delicacy made from the Aspergillus oryza mold of soybeans, is one of these fermented legume foods [348]. Another fermented soy substance called jang is a common Korean food. Numerous common yeast species, including Debaryomyces hansenii, Hyphopichia burtonii, and Jang Saccharomycopsis fibuligera, are abundant there [349]. Bacillus spp. species are discovered. The following kinds of bacteria are involved in the fermentation process: Bacillus fusiformis Bacillus sphaericus, Bacillus amyloliquefaciens, Bacillus megaterium, Bacillus licheniformis Bacillus cereus, Bacillus badius, and Bacillus pumilus [350]. Ugba, a traditional food made at home in Africa, can be counted among the fermented legumes [351]. Pediococcus pentosaceus and Pichia kudriavzevii TY1322 are isolated in Swedish fermented legume beverages. While the TY1322 of these species is effective in reducing phytates, it is predicted that it can be put to use as a starter culture in various legume fermentations [352].
Fermentation improves digestibility and produces new bioactive chemicals that are helpful to health. It has links to cancer, diabetes, inflammation, antioxidants, and diabetes [353]. The health-protective molecule, GABA, which rises with fermentation, is a substance [354]. It has an impact on enhancing cell viability and preventing oxidative damage [355]. One of the legumes that receives a lot of attention globally is soybean, which, along with its fermentation, has a regulating effect on the stool microbiota [356]. Increases in the catalase, superoxide dismutase, and glutathione peroxidase levels, along with a decrease in reactive oxygen species and pro-inflammatory cytokines (NF-κB, IL-β, COX-2, and TNF-α), give fermented soy products anti-inflammatory effects [357]. Additionally, it affects vital enzymes that are part of the hypoglycemic processes, including α-amylase and α-glucosidase [358]. The primary bioactive ingredient in fermented soybeans, daizein, has been linked to diseases such insulin resistance, obesity, and dyslipidemia [359]. Table 4 provides an overview of the health benefits of fermented legumes.

6. Fermented Cereals

Cereals are edible grains or seeds from the Gramineae family. The group of grains includes rye, oats, barley, maize, triticale, millet, and sorghum. The two most significant cereal crops worldwide are wheat and rice [398]. Many people consume rice as a fundamental food, especially in portions of Asia, Latin America, and Africa [399]. Rice is one of the most widely consumed grains. With regard to minerals, dietary fiber, zinc, protein, lipids, complex vitamins of vitamin B, and vitamin E, cereals play significant roles in our nutrition. However, beneficial phytochemicals such phytic acid, phenolic compounds, and gamma-oryzanol have significant roles [400].
Cereals are also best processed through fermentation, a time-honored technique [401]. The fermenting method is becoming more and more popular due to the growing interest in dietary consumption and nutrition [402]. In Africa, foods made from fermented grains are used as staple foods [403]. Among the most common grains utilized in fermentation are wheat, corn, teff, sorghum, and millet [404]. It is possible to give examples of regionally fermented grain-based dishes like Mawè and Ogi [405]. Utilizing Streptococcus thermophilus during fermentation enhances texture and flavor while also increasing volatile chemicals (diacetyl and acetoin) [406]. The fermentation process decreases the moisture and carbohydrate contents while increasing the total protein and ash contents in corn beverages fermented with Lactobacillus bulgaricus and Streptococcus thermophilus [407]. The traditional Peruvian drink, “Chicha de siete semillas”, is fermented using Streptococcus macedonicus and Leuconoctoc lactis. This fermented cuisine contains a lot of GABAs and is made from grains, pseudograins, and legumes. Streptococcus macedonicus is typically chosen for maize preparation if corn is to be used as a grain source [408]. Amahewu is another type of fermented grain. Amahewu is a fermented oatmeal or beverage made from corn that is mostly enjoyed in South Africa. Depending on the graft type, the type of maize, and the present fermentation circumstances, Amahewu’s nutritional and sensory qualities may change [409]. Bacillus, Arthrobacter, Lactobacillus, Ilyobacter, Clostridium, and Lactococcus are only a few of the numerous and distinct microbial species that are abundant in the fermented rice-based beverage, Chokot, made in India [410]. A popular fermented beverage made from grains called boza is enjoyed in many Balkan nations. Boza is rich in lactic acid bacteria, including Pediococcus parvulus, Lactobacillus parabuchneri, Limosilolactobacillus fermentum, Lactobacillus coryniformis, and Lactobacillus buchneri. Other types of microbiota found in boza, however, include yeasts such Pichia fermentans, Pichia norvegensis, Pichia guilliermondii, and Torulaspora spp. [411]. Boza, a grain-based food, is likewise high in putrescine, spermidine, and tyramine [412]. It has health impacts in addition to enhancing the functional and nutritive value of fermented grain products and satisfying the demands of contemporary consumers for health-promoting products [413]. Table 5 contains a list of the consequences of fermented cereals on health.

7. The Other Side of Fermented Foods

Dairy products, vegetables and fruits, legumes, meats, and grains are among the fermentable food groups. Microorganisms that play roles in fermentation and the bioactive compounds released during fermentation have antioxidant, anti-fungal, and antidiabetes effects; are involved in the protection of cognitive function and the regulation of intestinal microbiota; and have anti-inflammatory, antihypertension, anticancer, and anti-obesity effects (Figure 3).
Although fermentation has positive impacts on health, another aspect of it needs to be examined. One of the aspects that should be emphasized is the biogenic amines contained in some fermented products. Biogenic amines are compounds that exist especially in fermented meat products and increase with fermentation [430]. Biogenic amines such as spermidine and cadaverine cause an increase in N-nitrosodimethylamine and N-nitrosopiperidine levels [431]. The nitrosamines belong to the carcinogen group. While the most prevalent forms of nitrosamines in meat are N-nitrosodimethylamine and N-nitrosopiperidine [432], the International Agency for Research on Cancer categorizes N-nitrosopiperidine as group 2B, while N-nitrosodimethylamine is classified as group 2A [433].
Another component found in fermented foods that has been linked to health is biogenic amines. Histamine, tyramine, putrescine, and cadaverine are the biogenic amines that are most frequently observed [434]. The bacterial decarboxylation of the appropriate amino acids using substrate-specific decarboxylase enzymes is the primary method used to create biogenic amines in food. For example, histamine is formed from the amino acid histidine via histidine decarboxylase, while cadaverine is formed from lysine [435]. They can occur in many fermented foods such as cheese, sauerkraut and another vegetables, soybean, meat, fish, beer, wine, etc. [434,435]. Biogenic amines have many roles in the body such as protein, hormone, and nucleic acid syntheses, blood pressure control, and the promotion of cell growth. However, excessive intake may have toxic effects. Many symptoms such as food poisoning, headache, and sweating can be seen [436]. Biogenic amine formation and an increase in the amount can be prevented by paying attention to the storage temperature of foods, packaging processes, natural components, and appropriate starter culture selection [437]. There are studies on starter cultures, especially in fermented foods. The use of Bacillus polymyxa as a starter culture during salted fish fermentation and Lactobacillus plantarum as a starter culture during miso fermentation reduced biogenic amines [438,439].
Another unhealthy compound that can be found in some fermented foods is salt. High salt consumption has negative health effects. It is therefore important to reduce salt consumption. However, reducing salt in fermented foods may pose a problem with regard to food safety, texture, and flavor [440]. While ensuring that this reduces salt intake, it may cause the development of pathogenic microorganisms. Reducing the salts used may result in increased yeast, Enterobacteriaceae, and microbial growths [441]. Microbial growth can occur due to the decrease in water activity. As a result of the increase in some microorganisms, the formation of biogenic amines and nitrosamines may increase [430,442,443,444,445,446]. In addition, there is an increase, especially in the NDMA type, due to the replacement of sodium salt with potassium salt [447]. There are different practices for reducing sodium salt in some traditional fermented foods to change the diffusion and dissolution state by improving the physical form of the sodium salt used. Vacuum curing technology, ultrasound technology, high pressure technology, and microwave technology are used to obtain low-sodium products [440].
The future growth of the fermented food sector is made possible by the lowering of sodium and nitrosamines in traditional fermented foods. Studies on fermented foods show heterogeneous characteristics. This makes it difficult to compile the studies on a fermented food and to make a general evaluation of the positive and negative properties of that fermented food. In addition, studies on the amounts of fermented foods consumed by people need to be increased. In this way, responses to the effects of the amounts of fermented foods consumed on humans can be evaluated.
The regulatory effects of many fermented foods on health are explained in this article. These foods have a large place in our daily diets. However, their “consumption amount” is not determined by legal regulations. However, the lack of production standards (industrial type, homemade type, etc.) of fermented foods will cause both difficulty and inability in achieving homogenization, especially in surveillance and determining microorganism species differences and by-products due to such microorganisms.

8. Conclusions

The earliest food processing technique to have developed alongside human civilization is fermentation. The foods not only stayed fresh for a very long time, but they also developed new sensory qualities like flavors and smells. With the development of the food sector, fermented goods are becoming more and more popular. Fermented foods fall into significant food groups in human nutrition. Food groups that can be fermented include dairy products, cereals, legumes, fruits and vegetables, meats, and grains. In addition to internationally popular fermented foods like kefir, yogurt, cheese, fruit vinegar, and wine, traditional fermented pickles like kimchi and sauerkraut are also referred to as “pickles”. In addition, there are more regionally specific traditional fermented foods that can be pointed out including the following: meats such as nham; cereals such as mawè, jalebi, borde, kunu-zaki, kounou, togwa, bhaati jaanr, ogi, chicha de siete semillas, amahewu, chokot, and boza; and legumes such as cheonggukjang, natto, miso, jang, ugba, doenjang, koji, and meju. In the fermentation of foods, two significant components can be identified: (i) bioactive substances generated during fermentation and (ii) microorganisms involved during fermentation. Among the bioactive substances are organic acids, bioactive peptides, exopolysaccharides, conjugated linoleic acid, biogenic amines, isoflavones, phytoestrogens, nattokinase, and N-nitrosamines. Microorganisms, which are another factor in the realization of fermentation, come to the fore in different ways in different foods. Microorganisms such as Lactobacillus, Bifidobacterium, Streptococcus, and Bacillus species are regarded as probiotics in the fermentation process. Microorganisms that are efficient in the fermentation process and the bioactive substances they produce have impacts on health. Antidiabetes, anticancer, antioxidant, anti-inflammatory, antihypertension, and anti-fungal effects; the regulation of intestinal microbiota; the protection of cognitive function; and anti-obesity activities are just a few of fermented foods’ effects. Along with the positive effects of microorganisms and bioactive compounds in fermented foods on health, studies should be increased to elucidate the mechanisms for the effects of these foods on health. Studies have generally been carried out on isolated bioactive compounds and/or microorganisms. However, it is substantial to increase studies that evaluate the intake of fermented foods as a complex, rather than as a single component in human nutrition, and their interactions with each other. In future studies, the amount and duration of fermented foods in human nutrition should be evaluated.

Author Contributions

Conceptualization, D.A.; writing—original draft preparation, G.D. and E.Ç.; writing—review and editing, G.D., E.Ç., D.A., E.B., J.M.F.R. and F.Ö.; visualization, G.D. and E.Ç.; supervision, D.A., J.M.F.R., E.B. and F.Ö. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to acknowledge the COST Action 18101 SOURDOMICS Sourdough biotechnology network towards novel, healthier and sustainable food and bioprocesses (https://sourdomics.com/; https://www.cost.eu/actions/CA18101/, accessed on 28 July 2023), where the author F.Ö. is the Leader of working group 8, “Food safety, healthpromoting, sensorial perception and consumers’ behavior”; the author E.B. is the Vice Chair and leader of working group 6, “Project design and development innovative prototypes of products and smallscale processing technologies”; and the author J.M.R. is the Chair and Grant Holder Scientific Representative and is supported by COST (European Co-operation in Science and Technology) (https://www.cost.eu/, accessed on 28 July 2023). COST is a funding agency for research and innovation networks. The author, J.M.R., also acknowledges the Universidade Católica Portuguesa, CBQF (Centro de Biotecnologia e Química Fina) Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal, as well as the support from LA/P/0045/2020 (ALiCE) and UIDB/00511/2020-UIDP/00511/2020 (LEPABE) funded by national funds through FCT/MCTES (PIDDAC).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ray, R.; Joshi, V. Fermented Foods: Past, Present and Future. In Microorganisms and Fermentation of Traditional Foods; CRC Press: Boca Raton, FL, USA, 2014. [Google Scholar]
  2. Marco, M.L.; Sanders, M.E.; Gänzle, M.; Arrieta, M.C.; Cotter, P.D.; De Vuyst, L.; Hill, C.; Holzapfel, W.; Lebeer, S.; Merenstein, D. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on fermented foods. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 196–208. [Google Scholar] [PubMed]
  3. Annunziata, G.; Arnone, A.; Ciampaglia, R.; Tenore, G.C.; Novellino, E. Fermentation of foods and beverages as a tool for increasing availability of bioactive compounds. Focus on short-chain fatty acids. Foods 2020, 9, 999. [Google Scholar] [CrossRef]
  4. Melini, F.; Melini, V.; Luziatelli, F.; Ficca, A.G.; Ruzzi, M. Health-Promoting Components in Fermented Foods: An Up-to-Date Systematic Review. Nutrients 2019, 11, 1189. [Google Scholar] [CrossRef]
  5. Leeuwendaal, N.K.; Stanton, C.; O’Toole, P.W.; Beresford, T.P. Fermented Foods, Health and the Gut Microbiome. Nutrients 2022, 14, 1527. [Google Scholar] [CrossRef] [PubMed]
  6. Mathur, H.; Beresford, T.P.; Cotter, P.D. Health Benefits of Lactic Acid Bacteria (LAB) Fermentates. Nutrients 2020, 12, 1679. [Google Scholar] [CrossRef] [PubMed]
  7. Marco, M.L.; Heeney, D.; Binda, S.; Cifelli, C.J.; Cotter, P.D.; Foligné, B.; Gänzle, M.; Kort, R.; Pasin, G.; Pihlanto, A. Health benefits of fermented foods: Microbiota and beyond. Curr. Opin. Biotechnol. 2017, 44, 94–102. [Google Scholar]
  8. Terefe, N.S. Food Fermentation. In Reference Module in Food Science; Elsevier: Amsterdam, The Netherlands, 2016. [Google Scholar]
  9. FAO; WHO. Report of a Joint FAO/WHO Expert Consultation on Evaluation of Health and Nutritional Properties of Probiotics in Food including Powder Milk with Live Lactic Acid Bacteria. pp. 1–29. Available online: https://www.fao.org/3/y6398e/y6398e.pdf (accessed on 11 September 2023).
  10. International Scientific Association for Probiotics and Prebiotics, ISAPP. Probiotics: Dispelling Myths; ISAPP: Sacramento, CA, USA, 2018. [Google Scholar]
  11. Ilango, S.; Antony, U. Probiotic microorganisms from non-dairy traditional fermented foods. Trends Food Sci. Technol. 2021, 118, 617–638. [Google Scholar] [CrossRef]
  12. Diez-Ozaeta, I.; Astiazaran, O.J. Fermented foods: An update on evidence-based health benefits and future perspectives. Food Res. Int. 2022, 156, 111133. [Google Scholar] [CrossRef]
  13. Jaiswal, S.; Pant, T.; Suryavanshi, M.; Antony, U. Microbiological diversity of fermented food Bhaati Jaanr and its antioxidant and anti-inflammatory properties: Effect against colon cancer. Food Biosci. 2023, 55, 102822. [Google Scholar] [CrossRef]
  14. Papadimitriou, C.G.; Vafopoulou-Mastrojiannaki, A.; Silva, S.V.; Gomes, A.-M.; Malcata, F.X.; Alichanidis, E. Identification of peptides in traditional and probiotic sheep milk yoghurt with angiotensin I-converting enzyme (ACE)-inhibitory activity. Food Chem. 2007, 105, 647–656. [Google Scholar] [CrossRef]
  15. Gu, Y.; Li, X.; Chen, H.; Sun, Y.; Yang, L.; Ma, Y.; Yong Chan, E.C. Antidiabetic effects of multi-species probiotic and its fermented milk in mice via restoring gut microbiota and intestinal barrier. Food Biosci. 2022, 47, 101619. [Google Scholar] [CrossRef]
  16. Khakhariya, R.; Sakure, A.A.; Maurya, R.; Bishnoi, M.; Kondepudi, K.K.; Padhi, S.; Rai, A.K.; Liu, Z.; Patil, G.B.; Mankad, M.; et al. A comparative study of fermented buffalo and camel milk with anti-inflammatory, ACE-inhibitory and anti-diabetic properties and release of bio active peptides with molecular interactions: In vitro, in silico and molecular study. Food Biosci. 2023, 52, 102373. [Google Scholar] [CrossRef]
  17. Tunick, M.H.; Van Hekken, D.L. Dairy Products and Health: Recent Insights. J. Agric. Food Chem. 2015, 63, 9381–9388. [Google Scholar] [CrossRef] [PubMed]
  18. Nongonierma, A.B.; FitzGerald, R.J. Bioactive properties of milk proteins in humans: A review. Peptides 2015, 73, 20–34. [Google Scholar] [CrossRef] [PubMed]
  19. Shiby, V.K.; Mishra, H.N. Fermented milks and milk products as functional foods—A review. Crit. Rev. Food Sci. Nutr. 2013, 53, 482–496. [Google Scholar] [CrossRef]
  20. Fernández, M.; Hudson, J.A.; Korpela, R.; de los Reyes-Gavilán, C.G. Impact on human health of microorganisms present in fermented dairy products: An overview. Biomed. Res. Int. 2015, 2015, 412714. [Google Scholar] [CrossRef]
  21. Lin, M.Y.; Young, C.M. Folate levels in cultures of lactic acid bacteria. Int. Dairy J. 2000, 10, 409–413. [Google Scholar] [CrossRef]
  22. Van Wyk, J.; Witthuhn, R.C.; Britz, T.J. Optimisation of vitamin B12 and folate production by Propionibacterium freudenreichii strains in kefir. Int. Dairy J. 2011, 21, 69–74. [Google Scholar] [CrossRef]
  23. Hugenschmidt, S.; Schwenninger, S.M.; Lacroix, C. Concurrent high production of natural folate and vitamin B12 using a co-culture process with Lactobacillus plantarum SM39 and Propionibacterium freudenreichii DF13. Process Biochem. 2011, 46, 1063–1070. [Google Scholar] [CrossRef]
  24. Ibrahim, S.A.; Gyawali, R.; Awaisheh, S.S.; Ayivi, R.D.; Silva, R.C.; Subedi, K.; Aljaloud, S.O.; Anusha Siddiqui, S.; Krastanov, A. Fermented foods and probiotics: An approach to lactose intolerance. J. Dairy Res. 2021, 88, 357–365. [Google Scholar] [CrossRef]
  25. Prado, M.R.; Blandón, L.M.; Vandenberghe, L.P.; Rodrigues, C.; Castro, G.R.; Thomaz-Soccol, V.; Soccol, C.R. Milk kefir: Composition, microbial cultures, biological activities, and related products. Front. Microbiol. 2015, 6, 1177. [Google Scholar] [CrossRef]
  26. Azizi, N.F.; Kumar, M.R.; Yeap, S.K.; Abdullah, J.O.; Khalid, M.; Omar, A.R.; Osman, M.A.; Mortadza, S.A.S.; Alitheen, N.B. Kefir and Its Biological Activities. Foods 2021, 10, 1210. [Google Scholar] [CrossRef]
  27. Rosa, D.D.; Dias, M.M.S.; Grześkowiak, Ł.M.; Reis, S.A.; Conceição, L.L.; Peluzio, M. Milk kefir: Nutritional, microbiological and health benefits. Nutr. Res. Rev. 2017, 30, 82–96. [Google Scholar] [CrossRef] [PubMed]
  28. Garofalo, C.; Ferrocino, I.; Reale, A.; Sabbatini, R.; Milanović, V.; Alkić-Subašić, M.; Boscaino, F.; Aquilanti, L.; Pasquini, M.; Trombetta, M.F.; et al. Study of kefir drinks produced by backslopping method using kefir grains from Bosnia and Herzegovina: Microbial dynamics and volatilome profile. Food Res. Int. 2020, 137, 109369. [Google Scholar] [CrossRef] [PubMed]
  29. Gao, J.; Gu, F.; Abdella, N.H.; Ruan, H.; He, G. Investigation on culturable microflora in Tibetan kefir grains from different areas of China. J. Food Sci. 2012, 77, M425–M433. [Google Scholar] [CrossRef]
  30. Zhou, J.; Liu, X.; Jiang, H.; Dong, M. Analysis of the microflora in Tibetan kefir grains using denaturing gradient gel electrophoresis. Food Microbiol. 2009, 26, 770–775. [Google Scholar] [CrossRef]
  31. Altay, F.; Karbancıoglu-Güler, F.; Daskaya-Dikmen, C.; Heperkan, D. A review on traditional Turkish fermented non-alcoholic beverages: Microbiota, fermentation process and quality characteristics. Int. J. Food Microbiol. 2013, 167, 44–56. [Google Scholar] [CrossRef]
  32. Bourrie, B.C.; Willing, B.P.; Cotter, P.D. The Microbiota and Health Promoting Characteristics of the Fermented Beverage Kefir. Front. Microbiol. 2016, 7, 647. [Google Scholar] [CrossRef]
  33. Yirmibesoglu, S.; Tefon Öztürk, B. Comparing microbiological profiles, bioactivities, and physicochemical and sensory properties of donkey milk kefir and cow milk kefir. Turk. J. Vet. Anim. Sci. 2020, 44, 774–781. [Google Scholar] [CrossRef]
  34. Aires, R.; Gobbi Amorim, F.; Côco, L.Z.; da Conceição, A.P.; Zanardo TÉ, C.; Taufner, G.H.; Nogueira, B.V.; Vasquez, E.C.; Melo Costa Pereira, T.; Campagnaro, B.P.; et al. Use of kefir peptide (Kef-1) as an emerging approach for the treatment of oxidative stress and inflammation in 2K1C mice. Food Funct. 2022, 13, 1965–1974. [Google Scholar] [CrossRef]
  35. Maalouf, K.; Baydoun, E.; Rizk, S. Kefir induces cell-cycle arrest and apoptosis in HTLV-1-negative malignant T-lymphocytes. Cancer Manag. Res. 2011, 3, 39–47. [Google Scholar] [CrossRef] [PubMed]
  36. Erdogan, F.S.; Ozarslan, S.; Guzel-Seydim, Z.B.; Kök Taş, T. The effect of kefir produced from natural kefir grains on the intestinal microbial populations and antioxidant capacities of Balb/c mice. Food Res. Int. 2019, 115, 408–413. [Google Scholar] [CrossRef] [PubMed]
  37. Ton, A.M.M.; Campagnaro, B.P.; Alves, G.A.; Aires, R.; Côco, L.Z.; Arpini, C.M.; Guerra, E.O.T.; Campos-Toimil, M.; Meyrelles, S.S.; Pereira, T.M.C.; et al. Oxidative Stress and Dementia in Alzheimer’s Patients: Effects of Synbiotic Supplementation. Oxidative Med. Cell Longev. 2020, 2020, 2638703. [Google Scholar] [CrossRef]
  38. Bellikci-Koyu, E.; Sarer-Yurekli, B.P.; Akyon, Y.; Aydin-Kose, F.; Karagozlu, C.; Ozgen, A.G.; Brinkmann, A.; Nitsche, A.; Ergunay, K.; Yilmaz, E.; et al. Effects of Regular Kefir Consumption on Gut Microbiota in Patients with Metabolic Syndrome: A Parallel-Group, Randomized, Controlled Study. Nutrients 2019, 11, 2089. [Google Scholar] [CrossRef]
  39. Karaffová, V.; Mudroňová, D.; Mad’ar, M.; Hrčková, G.; Faixová, D.; Gancarčíková, S.; Ševčíková, Z.; Nemcová, R. Differences in Immune Response and Biochemical Parameters of Mice Fed by Kefir Milk and Lacticaseibacillus paracasei Isolated from the Kefir Grains. Microorganisms 2021, 9, 831. [Google Scholar] [CrossRef]
  40. Chen, H.L.; Tung, Y.T.; Chuang, C.H.; Tu, M.Y.; Tsai, T.C.; Chang, S.Y.; Chen, C.M. Kefir improves bone mass and microarchitecture in an ovariectomized rat model of postmenopausal osteoporosis. Osteoporos. Int. 2015, 26, 589–599. [Google Scholar] [CrossRef]
  41. Malta, S.M.; Batista, L.L.; Silva, H.C.G.; Franco, R.R.; Silva, M.H.; Rodrigues, T.S.; Correia, L.I.V.; Martins, M.M.; Venturini, G.; Espindola, F.S.; et al. Identification of bioactive peptides from a Brazilian kefir sample, and their anti-Alzheimer potential in Drosophila melanogaster. Sci. Rep. 2022, 12, 11065. [Google Scholar] [CrossRef]
  42. Hamet, M.F.; Medrano, M.; Pérez, P.F.; Abraham, A.G. Oral administration of kefiran exerts a bifidogenic effect on BALB/c mice intestinal microbiota. Benef. Microbes 2016, 7, 237–246. [Google Scholar] [CrossRef]
  43. Youn, H.Y.; Kim, D.H.; Kim, H.J.; Bae, D.; Song, K.Y.; Kim, H.; Seo, K.H. Survivability of Kluyveromyces marxianus Isolated from Korean Kefir in a Simulated Gastrointestinal Environment. Front. Microbiol. 2022, 13, 842097. [Google Scholar] [CrossRef]
  44. Maccaferri, S.; Klinder, A.; Brigidi, P.; Cavina, P.; Costabile, A. Potential probiotic Kluyveromyces marxianus B0399 modulates the immune response in Caco-2 cells and peripheral blood mononuclear cells and impacts the human gut microbiota in an in vitro colonic model system. Appl. Env. Microbiol. 2012, 78, 956–964. [Google Scholar] [CrossRef]
  45. Youn, H.Y.; Kim, H.J.; Kim, D.H.; Jang, Y.S.; Kim, H.; Seo, K.H. Gut microbiota modulation via short-term administration of potential probiotic kefir yeast Kluyveromyces marxianus A4 and A5 in BALB/c mice. Food Sci. Biotechnol. 2023, 32, 589–598. [Google Scholar] [CrossRef] [PubMed]
  46. Youn, H.Y.; Kim, D.H.; Kim, H.J.; Jang, Y.S.; Song, K.Y.; Bae, D.; Kim, H.; Seo, K.H. A Combined In Vitro and In Vivo Assessment of the Safety of the Yeast Strains Kluyveromyces marxianus A4 and A5 Isolated from Korean Kefir. Probiotics Antimicrob. Proteins 2023, 15, 129–138. [Google Scholar] [CrossRef]
  47. Tang, W.; Li, C.; He, Z.; Pan, F.; Pan, S.; Wang, Y. Probiotic Properties and Cellular Antioxidant Activity of Lactobacillus plantarum MA2 Isolated from Tibetan Kefir Grains. Probiotics Antimicrob. Proteins 2018, 10, 523–533. [Google Scholar] [CrossRef] [PubMed]
  48. Serafini, F.; Turroni, F.; Ruas-Madiedo, P.; Lugli, G.A.; Milani, C.; Duranti, S.; Zamboni, N.; Bottacini, F.; van Sinderen, D.; Margolles, A.; et al. Kefir fermented milk and kefiran promote growth of Bifidobacterium bifidum PRL2010 and modulate its gene expression. Int. J. Food Microbiol. 2014, 178, 50–59. [Google Scholar] [CrossRef] [PubMed]
  49. Jenab, A.; Roghanian, R.; Ghorbani, N.; Ghaedi, K.; Emtiazi, G. The Efficacy of Electrospun PAN/Kefiran Nanofiber and Kefir in Mammalian Cell Culture: Promotion of PC12 Cell Growth, Anti-MCF7 Breast Cancer Cells Activities, and Cytokine Production of PBMC. Int. J. Nanomed. 2020, 15, 717–728. [Google Scholar] [CrossRef] [PubMed]
  50. Elsayed, E.; Farooq, M.; Dailin, D.; El-Enshasy, H.; Othman, N.; Malek, R.; Danial, E.N.; Wadaan, M. In vitro and in vivo biological screening of kefiran polysaccharide produced by Lactobacillus kefiranofaciens. Biomed. Res. 2017, 28, 594–600. [Google Scholar]
  51. Bahari, A.; Shahabi-Ghahfarrokhi, I.; Koolivand, D. Kefiran ameliorates malfunctions in primary and functional immune cells caused by lipopolysaccharides. Int. J. Biol. Macromol. 2020, 165, 619–624. [Google Scholar] [CrossRef]
  52. Vinderola, G.; Perdigón, G.; Duarte, J.; Farnworth, E.; Matar, C. Effects of the oral administration of the exopolysaccharide produced by Lactobacillus kefiranofaciens on the gut mucosal immunity. Cytokine 2006, 36, 254–260. [Google Scholar] [CrossRef]
  53. Radhouani, H.; Gonçalves, C.; Maia, F.R.; Oliveira, J.M.; Reis, R.L. Biological performance of a promising Kefiran-biopolymer with potential in regenerative medicine applications: A comparative study with hyaluronic acid. J. Mater. Sci. Mater. Med. 2018, 29, 124. [Google Scholar] [CrossRef]
  54. Hasheminya, S.-M.; Dehghannya, J. Novel ultrasound-assisted extraction of kefiran biomaterial, a prebiotic exopolysaccharide, and investigation of its physicochemical, antioxidant and antimicrobial properties. Mater. Chem. Phys. 2020, 243, 122645. [Google Scholar] [CrossRef]
  55. Uchida, M.; Ishii, I.; Inoue, C.; Akisato, Y.; Watanabe, K.; Hosoyama, S.; Toida, T.; Ariyoshi, N.; Kitada, M. Kefiran reduces atherosclerosis in rabbits fed a high cholesterol diet. J. Atheroscler. Thromb. 2010, 17, 980–988. [Google Scholar] [CrossRef]
  56. Riaz Rajoka, M.S.; Mehwish, H.M.; Fang, H.; Padhiar, A.A.; Zeng, X.; Khurshid, M.; He, Z.; Zhao, L. Characterization and anti-tumor activity of exopolysaccharide produced by Lactobacillus kefiri isolated from Chinese kefir grains. J. Funct. Foods 2019, 63, 103588. [Google Scholar] [CrossRef]
  57. Wang, X.; Tian, J.; Zhang, X.; Tang, N.; Rui, X.; Zhang, Q.; Dong, M.; Li, W. Characterization and Immunological Activity of Exopolysaccharide from Lacticaseibacillus paracasei GL1 Isolated from Tibetan Kefir Grains. Foods 2022, 11, 3330. [Google Scholar] [CrossRef]
  58. You, X.; Yang, L.; Zhao, X.; Ma, K.; Chen, X.; Zhang, C.; Wang, G.; Dong, M.; Rui, X.; Zhang, Q.; et al. Isolation, purification, characterization and immunostimulatory activity of an exopolysaccharide produced by Lactobacillus pentosus LZ-R-17 isolated from Tibetan kefir. Int. J. Biol. Macromol. 2020, 158, 408–419. [Google Scholar] [CrossRef] [PubMed]
  59. You, X.; Li, Z.; Ma, K.; Zhang, C.; Chen, X.; Wang, G.; Yang, L.; Dong, M.; Rui, X.; Zhang, Q.; et al. Structural characterization and immunomodulatory activity of an exopolysaccharide produced by Lactobacillus helveticus LZ-R-5. Carbohydr. Polym. 2020, 235, 115977. [Google Scholar] [CrossRef] [PubMed]
  60. Xiao, L.; Xu, D.; Tang, N.; Rui, X.; Zhang, Q.; Chen, X.; Dong, M.; Li, W. Biosynthesis of exopolysaccharide and structural characterization by Lacticaseibacillus paracasei ZY-1 isolated from Tibetan kefir. Food Chem. Mol. Sci. 2021, 3, 100054. [Google Scholar] [CrossRef]
  61. Zhang, J.; Zhao, X.; Jiang, Y.; Zhao, W.; Guo, T.; Cao, Y.; Teng, J.; Hao, X.; Zhao, J.; Yang, Z. Antioxidant status and gut microbiota change in an aging mouse model as influenced by exopolysaccharide produced by Lactobacillus plantarum YW11 isolated from Tibetan kefir. J. Dairy Sci. 2017, 100, 6025–6041. [Google Scholar] [CrossRef] [PubMed]
  62. Bengoa, A.A.; Dardis, C.; Gagliarini, N.; Garrote, G.L.; Abraham, A.G. Exopolysaccharides from Lactobacillus paracasei Isolated from Kefir as Potential Bioactive Compounds for Microbiota Modulation. Front. Microbiol. 2020, 11, 583254. [Google Scholar] [CrossRef]
  63. Lim, J.; Kale, M.; Kim, D.H.; Kim, H.S.; Chon, J.W.; Seo, K.H.; Lee, H.G.; Yokoyama, W.; Kim, H. Antiobesity Effect of Exopolysaccharides Isolated from Kefir Grains. J. Agric. Food Chem. 2017, 65, 10011–10019. [Google Scholar] [CrossRef]
  64. Brasil, G.A.; Silva-Cutini, M.A.; Moraes, F.S.A.; Pereira, T.M.C.; Vasquez, E.C.; Lenz, D.; Bissoli, N.S.; Endringer, D.C.; de Lima, E.M.; Biancardi, V.C.; et al. The benefits of soluble non-bacterial fraction of kefir on blood pressure and cardiac hypertrophy in hypertensive rats are mediated by an increase in baroreflex sensitivity and decrease in angiotensin-converting enzyme activity. Nutrition 2018, 51–52, 66–72. [Google Scholar] [CrossRef]
  65. Khoury, N.; El-Hayek, S.; Tarras, O.; El-Sabban, M.; El-Sibai, M.; Rizk, S. Kefir exhibits anti-proliferative and pro-apoptotic effects on colon adenocarcinoma cells with no significant effects on cell migration and invasion. Int. J. Oncol. 2014, 45, 2117–2127. [Google Scholar] [CrossRef]
  66. Liu, J.R.; Wang, S.Y.; Lin, Y.Y.; Lin, C.W. Antitumor activity of milk kefir and soy milk kefir in tumor-bearing mice. Nutr. Cancer 2002, 44, 183–187. [Google Scholar] [CrossRef] [PubMed]
  67. Zeng, X.; Jia, H.; Zhang, X.; Wang, X.; Wang, Z.; Gao, Z.; Yuan, Y.; Yue, T. Supplementation of kefir ameliorates azoxymethane/dextran sulfate sodium induced colorectal cancer by modulating the gut microbiota. Food Funct. 2021, 12, 11641–11655. [Google Scholar] [CrossRef]
  68. Ben Taheur, F.; Mansour, C.; Mechri, S.; Skhiri, S.S.; Jaouadi, B.; Mzoughi, R.; Chaieb, K.; Zouari, N. Does probiotic Kefir reduce dyslipidemia, hematological disorders and oxidative stress induced by zearalenone toxicity in wistar rats? Toxicon X 2022, 14, 100121. [Google Scholar] [CrossRef]
  69. Grishina, A.; Kulikova, I.; Alieva, L.; Dodson, A.; Rowland, I.; Jin, J. Antigenotoxic effect of kefir and ayran supernatants on fecal water-induced DNA damage in human colon cells. Nutr. Cancer 2011, 63, 73–79. [Google Scholar] [CrossRef] [PubMed]
  70. Yılmaz, İ.; Dolar, M.E.; Özpınar, H. Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turk. J. Gastroenterol. 2019, 30, 242–253. [Google Scholar] [CrossRef]
  71. Albuquerque Pereira, M.F.; Morais de Ávila, L.G.; Ávila Alpino, G.C.; Dos Santos Cruz, B.C.; Almeida, L.F.; Macedo Simões, J.; Ladeira Bernardes, A.; Xisto Campos, I.; de Oliveira Barros Ribon, A.; de Oliveira Mendes, T.A.; et al. Milk kefir alters fecal microbiota impacting gut and brain health in mice. Appl. Microbiol. Biotechnol. 2023, 107, 5161–5178. [Google Scholar] [CrossRef] [PubMed]
  72. Ostadrahimi, A.; Taghizadeh, A.; Mobasseri, M.; Farrin, N.; Payahoo, L.; Beyramalipoor Gheshlaghi, Z.; Vahedjabbari, M. Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: A randomized double-blind placebo-controlled clinical trial. Iran. J. Public Health 2015, 44, 228–237. [Google Scholar]
  73. Kakisu, E.J.; Abraham, A.G.; Pérez, P.F.; De Antoni, G.L. Inhibition of Bacillus cereus in milk fermented with kefir grains. J. Food Prot. 2007, 70, 2613–2616. [Google Scholar] [CrossRef]
  74. Iraporda, C.; Abatemarco Júnior, M.; Neumann, E.; Nunes, Á.C.; Nicoli, J.R.; Abraham, A.G.; Garrote, G.L. Biological activity of the non-microbial fraction of kefir: Antagonism against intestinal pathogens. J. Dairy Res. 2017, 84, 339–345. [Google Scholar] [CrossRef]
  75. Amorim, F.G.; Coitinho, L.B.; Dias, A.T.; Friques, A.G.F.; Monteiro, B.L.; Rezende, L.C.D.; Pereira, T.M.C.; Campagnaro, B.P.; De Pauw, E.; Vasquez, E.C.; et al. Identification of new bioactive peptides from Kefir milk through proteopeptidomics: Bioprospection of antihypertensive molecules. Food Chem. 2019, 282, 109–119. [Google Scholar] [CrossRef]
  76. Ebner, J.; Arslan, A.A.; Fedorova, M.; Hoffmann, R.; Küçükçetin, A.; Pischetsrieder, M. Peptide profiling of bovine kefir reveals 236 unique peptides released from caseins during its production by starter culture or kefir grains. J. Proteom. 2015, 117, 41–57. [Google Scholar] [CrossRef]
  77. Quirós, A.; Hernández-Ledesma, B.; Ramos, M.; Amigo, L.; Recio, I. Angiotensin-Converting Enzyme Inhibitory Activity of Peptides Derived from Caprine Kefir. J. Dairy Sci. 2005, 88, 3480–3487. [Google Scholar] [CrossRef]
  78. Chen, Y.H.; Chen, H.L.; Fan, H.C.; Tung, Y.T.; Kuo, C.W.; Tu, M.Y.; Chen, C.M. Anti-Inflammatory, Antioxidant, and Antifibrotic Effects of Kefir Peptides on Salt-Induced Renal Vascular Damage and Dysfunction in Aged Stroke-Prone Spontaneously Hypertensive Rats. Antioxidants 2020, 9, 790. [Google Scholar] [CrossRef]
  79. de Lima, M.; da Silva, R.A.; da Silva, M.F.; da Silva, P.A.B.; Costa, R.; Teixeira, J.A.C.; Porto, A.L.F.; Cavalcanti, M.T.H. Brazilian Kefir-Fermented Sheep’s Milk, a Source of Antimicrobial and Antioxidant Peptides. Probiotics Antimicrob. Proteins 2018, 10, 446–455. [Google Scholar] [CrossRef]
  80. Miao, J.; Liu, G.; Ke, C.; Fan, W.; Li, C.; Chen, Y.; Dixon, W.; Song, M.; Cao, Y.; Xiao, H. Inhibitory effects of a novel antimicrobial peptide from kefir against Escherichia coli. Food Control 2016, 65, 63–72. [Google Scholar] [CrossRef]
  81. Tu, M.Y.; Han, K.Y.; Chang, G.R.; Lai, G.D.; Chang, K.Y.; Chen, C.F.; Lai, J.C.; Lai, C.Y.; Chen, H.L.; Chen, C.M. Kefir Peptides Prevent Estrogen Deficiency-Induced Bone Loss and Modulate the Structure of the Gut Microbiota in Ovariectomized Mice. Nutrients 2020, 12, 3432. [Google Scholar] [CrossRef] [PubMed]
  82. Yanni, A.E.; Kartsioti, K.; Karathanos, V.T. The role of yoghurt consumption in the management of type II diabetes. Food Funct. 2020, 11, 10306–10316. [Google Scholar] [CrossRef]
  83. Qing, J.; Peng, C.; Chen, H.; Li, H.; Liu, X. Small molecule linoleic acid inhibiting whey syneresis via interact with milk proteins in the fermentation of set yogurt fortified with c9,t11-conjugated linoleic acid. Food Chem. 2023, 429, 136849. [Google Scholar] [CrossRef] [PubMed]
  84. Paszczyk, B.; Czarnowska-Kujawska, M.; Klepacka, J.; Tońska, E. Health-Promoting Ingredients in Goat’s Milk and Fermented Goat’s Milk Drinks. Animals 2023, 13, 907. [Google Scholar] [CrossRef]
  85. Serafeimidou, A.; Zlatanos, S.; Laskaridis, K.; Sagredos, A. Chemical characteristics, fatty acid composition and conjugated linoleic acid (CLA) content of traditional Greek yogurts. Food Chem. 2012, 134, 1839–1846. [Google Scholar] [CrossRef] [PubMed]
  86. Serafeimidou, A.; Zlatanos, S.; Kritikos, G.; Tourianis, A. Change of fatty acid profile, including conjugated linoleic acid (CLA) content, during refrigerated storage of yogurt made of cow and sheep milk. J. Food Compos. Anal. 2013, 31, 24–30. [Google Scholar] [CrossRef]
  87. Paszczyk, B.; Czarnowska-Kujawska, M. Fatty Acid Profile, Conjugated Linoleic Acid Content, and Lipid Quality Indices in Selected Yogurts Available on the Polish Market. Animals 2022, 12, 96. [Google Scholar] [CrossRef]
  88. Dilzer, A.; Park, Y. Implication of conjugated linoleic acid (CLA) in human health. Crit. Rev. Food Sci. Nutr. 2012, 52, 488–513. [Google Scholar] [CrossRef]
  89. Le Roy, C.I.; Kurilshikov, A.; Leeming, E.R.; Visconti, A.; Bowyer, R.C.E.; Menni, C.; Falchi, M.; Koutnikova, H.; Veiga, P.; Zhernakova, A.; et al. Yoghurt consumption is associated with changes in the composition of the human gut microbiome and metabolome. BMC Microbiol. 2022, 22, 39. [Google Scholar] [CrossRef]
  90. Sadrzadeh-Yeganeh, H.; Elmadfa, I.; Djazayery, A.; Jalali, M.; Heshmat, R.; Chamary, M. The effects of probiotic and conventional yoghurt on lipid profile in women. Br. J. Nutr. 2010, 103, 1778–1783. [Google Scholar] [CrossRef]
  91. Chen, Y.; Feng, R.; Yang, X.; Dai, J.; Huang, M.; Ji, X.; Li, Y.; Okekunle, A.P.; Gao, G.; Onwuka, J.U.; et al. Yogurt improves insulin resistance and liver fat in obese women with nonalcoholic fatty liver disease and metabolic syndrome: A randomized controlled trial. Am. J. Clin. Nutr. 2019, 109, 1611–1619. [Google Scholar] [CrossRef]
  92. Hasegawa, Y.; Pei, R.; Raghuvanshi, R.; Liu, Z.; Bolling, B.W. Yogurt Supplementation Attenuates Insulin Resistance in Obese Mice by Reducing Metabolic Endotoxemia and Inflammation. J. Nutr. 2023, 153, 703–712. [Google Scholar] [CrossRef]
  93. Douglas, S.M.; Ortinau, L.C.; Hoertel, H.A.; Leidy, H.J. Low, moderate, or high protein yogurt snacks on appetite control and subsequent eating in healthy women. Appetite 2013, 60, 117–122. [Google Scholar] [CrossRef]
  94. Rezazadeh, L.; Gargari, B.P.; Jafarabadi, M.A.; Alipour, B. Effects of probiotic yogurt on glycemic indexes and endothelial dysfunction markers in patients with metabolic syndrome. Nutrition 2019, 62, 162–168. [Google Scholar] [CrossRef]
  95. Wongrattanapipat, S.; Chiracharoenchitta, A.; Choowongwitthaya, B.; Komsathorn, P.; La-Ongkham, O.; Nitisinprasert, S.; Tunsagool, P.; Nakphaichit, M. Selection of potential probiotics with cholesterol-lowering properties for probiotic yoghurt production. Food Sci. Technol. Int. 2022, 28, 353–365. [Google Scholar] [CrossRef] [PubMed]
  96. Asgharian, H.; Homayouni-Rad, A.; Mirghafourvand, M.; Mohammad-Alizadeh-Charandabi, S. Effect of probiotic yoghurt on plasma glucose in overweight and obese pregnant women: A randomized controlled clinical trial. Eur. J. Nutr. 2020, 59, 205–215. [Google Scholar] [CrossRef]
  97. Mazani, M.; Nemati, A.; Amani, M.; Haedari, K.; Mogadam, R.A.; Baghi, A.N. The effect of probiotic yoghurt consumption on oxidative stress and inflammatory factors in young females after exhaustive exercise. J. Pak. Med. Assoc. 2018, 68, 1748–1754. [Google Scholar]
  98. Mirjalili, M.; Salari Sharif, A.; Sangouni, A.A.; Emtiazi, H.; Mozaffari-Khosravi, H. Effect of probiotic yogurt consumption on glycemic control and lipid profile in patients with type 2 diabetes mellitus: A randomized controlled trial. Clin. Nutr. ESPEN 2023, 54, 144–149. [Google Scholar] [CrossRef] [PubMed]
  99. Odamaki, T.; Kato, K.; Sugahara, H.; Xiao, J.Z.; Abe, F.; Benno, Y. Effect of probiotic yoghurt on animal-based diet-induced change in gut microbiota: An open, randomised, parallel-group study. Benef. Microbes 2016, 7, 473–484. [Google Scholar] [CrossRef] [PubMed]
  100. Del Carmen, S.; de Moreno de LeBlanc, A.; LeBlanc, J.G. Development of a potential probiotic yoghurt using selected anti-inflammatory lactic acid bacteria for prevention of colitis and carcinogenesis in mice. J. Appl. Microbiol. 2016, 121, 821–830. [Google Scholar] [CrossRef]
  101. Negm El-Dein, A.; Ezzat, A.; Aly, H.F.; Awad, G.; Farid, M. Lactobacillus-fermented yogurt exerts hypoglycemic, hypocholesterolemic, and anti-inflammatory activities in STZ-induced diabetic Wistar rats. Nutr. Res. 2022, 108, 22–32. [Google Scholar] [CrossRef]
  102. Velasco, M.; Requena, T.; Delgado-Iribarren, A.; Peláez, C.; Guijarro, C. Probiotic Yogurt for the Prevention of Antibiotic-associated Diarrhea in Adults: A Randomized Double-blind Placebo-controlled Trial. J. Clin. Gastroenterol. 2019, 53, 717–723. [Google Scholar] [CrossRef]
  103. Fox, M.J.; Ahuja, K.D.; Robertson, I.K.; Ball, M.J.; Eri, R.D. Can probiotic yogurt prevent diarrhoea in children on antibiotics? A double-blind, randomised, placebo-controlled study. BMJ Open 2015, 5, e006474. [Google Scholar] [CrossRef]
  104. Barengolts, E.; Smith, E.D.; Reutrakul, S.; Tonucci, L.; Anothaisintawee, T. The Effect of Probiotic Yogurt on Glycemic Control in Type 2 Diabetes or Obesity: A Meta-Analysis of Nine Randomized Controlled Trials. Nutrients 2019, 11, 671. [Google Scholar] [CrossRef]
  105. Ivey, K.L.; Hodgson, J.M.; Kerr, D.A.; Thompson, P.L.; Stojceski, B.; Prince, R.L. The effect of yoghurt and its probiotics on blood pressure and serum lipid profile; a randomised controlled trial. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 46–51. [Google Scholar] [CrossRef]
  106. Bandiera, N.S.; Carneiro, I.; da Silva, A.S.; Honjoya, E.R.; de Santana, E.H.; Aragon-Alegro, L.C.; de Souza, C.H. Viability of probiotic Lactobacillus casei in yoghurt: Defining the best processing step to its addition. Arch. Latinoam. Nutr. 2013, 63, 58–63. [Google Scholar]
  107. Sah, B.N.P.; Vasiljevic, T.; McKechnie, S.; Donkor, O.N. Effect of refrigerated storage on probiotic viability and the production and stability of antimutagenic and antioxidant peptides in yogurt supplemented with pineapple peel. J. Dairy Sci. 2015, 98, 5905–5916. [Google Scholar] [CrossRef] [PubMed]
  108. Mani-López, E.; Palou, E.; López-Malo, A. Probiotic viability and storage stability of yogurts and fermented milks prepared with several mixtures of lactic acid bacteria. J. Dairy Sci. 2014, 97, 2578–2590. [Google Scholar] [CrossRef] [PubMed]
  109. Taha, S.; El Abd, M.; De Gobba, C.; Abdel-Hamid, M.; Khalil, E.; Hassan, D. Antioxidant and antibacterial activities of bioactive peptides in buffalo’s yoghurt fermented with different starter cultures. Food Sci. Biotechnol. 2017, 26, 1325–1332. [Google Scholar] [CrossRef] [PubMed]
  110. Aloğlu, H.S.; Oner, Z. Determination of antioxidant activity of bioactive peptide fractions obtained from yogurt. J. Dairy Sci. 2011, 94, 5305–5314. [Google Scholar] [CrossRef]
  111. Jin, Y.; Yu, Y.; Qi, Y.; Wang, F.; Yan, J.; Zou, H. Peptide profiling and the bioactivity character of yogurt in the simulated gastrointestinal digestion. J. Proteom. 2016, 141, 24–46. [Google Scholar] [CrossRef]
  112. Giacometti Cavalheiro, F.; Parra Baptista, D.; Domingues Galli, B.; Negrão, F.; Nogueira Eberlin, M.; Lúcia Gigante, M. High protein yogurt with addition of Lactobacillus helveticus: Peptide profile and angiotensin-converting enzyme ACE-inhibitory activity. Food Chem. 2020, 333, 127482. [Google Scholar] [CrossRef]
  113. Plaisancié, P.; Claustre, J.; Estienne, M.; Henry, G.; Boutrou, R.; Paquet, A.; Léonil, J. A novel bioactive peptide from yoghurts modulates expression of the gel-forming MUC2 mucin as well as population of goblet cells and Paneth cells along the small intestine. J. Nutr. Biochem. 2013, 24, 213–221. [Google Scholar] [CrossRef]
  114. Abd El-Fattah, A.; Sakr, S.; El-Dieb, S.; Elkashef, H. Developing functional yogurt rich in bioactive peptides and gamma-aminobutyric acid related to cardiovascular health. LWT 2018, 98, 390–397. [Google Scholar] [CrossRef]
  115. Heydari, S.; Hosseini, S.E.; Mortazavian, A.M.; Taheri, S. Extraction of bioactive peptides produced in probiotic yoghurt and determination of their biological activities. Int. Dairy J. 2023, 139, 105544. [Google Scholar] [CrossRef]
  116. Bintsis, T. Yeasts in different types of cheese. AIMS Microbiol. 2021, 7, 447–470. [Google Scholar] [CrossRef] [PubMed]
  117. Fröhlich-Wyder, M.T.; Arias-Roth, E.; Jakob, E. Cheese yeasts. Yeast 2019, 36, 129–141. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, M.; Dong, X.; Huang, Z.; Li, X.; Zhao, Y.; Wang, Y.; Zhu, H.; Fang, A.; Giovannucci, E.L. Cheese consumption and multiple health outcomes: An umbrella review and updated meta-analysis of prospective studies. Adv. Nutr. 2023, 14, 1170–1186. [Google Scholar] [CrossRef]
  119. Hu, M.J.; Tan, J.S.; Gao, X.J.; Yang, J.G.; Yang, Y.J. Effect of Cheese Intake on Cardiovascular Diseases and Cardiovascular Biomarkers. Nutrients 2022, 14, 2936. [Google Scholar] [CrossRef]
  120. Hjerpsted, J.; Tholstrup, T. Cheese and Cardiovascular Disease Risk: A Review of the Evidence and Discussion of Possible Mechanisms. Crit. Rev. Food Sci. Nutr. 2016, 56, 1389–1403. [Google Scholar] [CrossRef]
  121. Kurbanova, M.; Voroshilin, R.; Kozlova, O.; Atuchin, V. Effect of Lactobacteria on Bioactive Peptides and Their Sequence Identification in Mature Cheese. Microorganisms 2022, 10, 2068. [Google Scholar] [CrossRef] [PubMed]
  122. Shafique, B.; Murtaza, M.A.; Hafiz, I.; Ameer, K.; Basharat, S.; Mohamed Ahmed, I.A. Proteolysis and therapeutic potential of bioactive peptides derived from Cheddar cheese. Food Sci. Nutr. 2023, 11, 4948–4963. [Google Scholar] [CrossRef]
  123. Helal, A.; Tagliazucchi, D. Peptidomics Profile, Bioactive Peptides Identification and Biological Activities of Six Different Cheese Varieties. Biology 2023, 12, 78. [Google Scholar] [CrossRef]
  124. Álvarez Ramos, L.; Arrieta Baez, D.; Dávila Ortiz, G.; Carlos Ruiz Ruiz, J.; Manuel Toledo López, V. Antioxidant and antihypertensive activity of Gouda cheese at different stages of ripening. Food Chem. X 2022, 14, 100284. [Google Scholar] [CrossRef]
  125. Martín-Del-Campo, S.T.; Martínez-Basilio, P.C.; Sepúlveda-Álvarez, J.C.; Gutiérrez-Melchor, S.E.; Galindo-Peña, K.D.; Lara-Domínguez, A.K.; Cardador-Martínez, A. Production of Antioxidant and ACEI Peptides from Cheese Whey Discarded from Mexican White Cheese Production. Antioxidants 2019, 8, 158. [Google Scholar] [CrossRef] [PubMed]
  126. Timón, M.L.; Andrés, A.I.; Otte, J.; Petrón, M.J. Antioxidant peptides (<3 kDa) identified on hard cow milk cheese with rennet from different origin. Food Res. Int. 2019, 120, 643–649. [Google Scholar] [CrossRef]
  127. Abedin, M.M.; Chourasia, R.; Chiring Phukon, L.; Singh, S.P.; Kumar Rai, A. Characterization of ACE inhibitory and antioxidant peptides in yak and cow milk hard chhurpi cheese of the Sikkim Himalayan region. Food Chem. X 2022, 13, 100231. [Google Scholar] [CrossRef] [PubMed]
  128. Dimitrov, Z.; Chorbadjiyska, E.; Gotova, I.; Pashova, K.; Ilieva, S. Selected adjunct cultures remarkably increase the content of bioactive peptides in Bulgarian white brined cheese. Biotechnol. Biotechnol. Equip. 2015, 29, 78–83. [Google Scholar] [CrossRef] [PubMed]
  129. Helal, A.; Cattivelli, A.; Conte, A.; Tagliazucchi, D. Effect of Ripening and In Vitro Digestion on Bioactive Peptides Profile in Ras Cheese and Their Biological Activities. Biology 2023, 12, 948. [Google Scholar] [CrossRef]
  130. Munir, M.; Nadeem, M.; Mahmood Qureshi, T.; Gamlath, C.J.; Martin, G.J.O.; Hemar, Y.; Ashokkumar, M. Effect of sonication, microwaves and high-pressure processing on ACE-inhibitory activity and antioxidant potential of Cheddar cheese during ripening. Ultrason. Sonochem. 2020, 67, 105140. [Google Scholar] [CrossRef]
  131. Crippa, G.; Zabzuni, D.; Bravi, E.; Piva, G.; De Noni, I.; Bighi, E.; Rossi, F. Randomized, double blind placebo-controlled pilot study of the antihypertensive effects of Grana Padano D.O.P. cheese consumption in mild—Moderate hypertensive subjects. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7573–7581. [Google Scholar] [CrossRef] [PubMed]
  132. Ramírez-Rivas, I.K.; Gutiérrez-Méndez, N.; Rentería-Monterrubio, A.L.; Sánchez-Vega, R.; Tirado-Gallegos, J.M.; Santellano-Estrada, E.; Arevalos-Sánchez, M.M.; Chávez-Martínez, A. Effect of Packaging and Salt Content and Type on Antioxidant and ACE-Inhibitory Activities in Requeson Cheese. Foods 2022, 11, 1264. [Google Scholar] [CrossRef]
  133. Sánchez-Rivera, L.; Recio, I.; Ramos, M.; Gómez-Ruiz, J. Short communication: Peptide profiling in cheeses packed using different technologies. J. Dairy Sci. 2013, 96, 3551–3557. [Google Scholar] [CrossRef]
  134. Donmez, M.; Kemal Seckin, A.; Sagdic, O.; Simsek, B. Chemical characteristics, fatty acid compositions, conjugated linoleic acid contents and cholesterol levels of some traditional Turkish cheeses. Int. J. Food Sci. Nutr. 2005, 56, 157–163. [Google Scholar] [CrossRef]
  135. Laskaridis, K.; Serafeimidou, A.; Zlatanos, S.; Gylou, E.; Kontorepanidou, E.; Sagredos, A. Changes in fatty acid profile of feta cheese including conjugated linoleic acid. J. Sci. Food Agric. 2013, 93, 2130–2136. [Google Scholar] [CrossRef]
  136. Santurino, C.; López-Plaza, B.; Fontecha, J.; Calvo, M.V.; Bermejo, L.M.; Gómez-Andrés, D.; Gómez-Candela, C. Consumption of Goat Cheese Naturally Rich in Omega-3 and Conjugated Linoleic Acid Improves the Cardiovascular and Inflammatory Biomarkers of Overweight and Obese Subjects: A Randomized Controlled Trial. Nutrients 2020, 12. [Google Scholar] [CrossRef]
  137. Koba, K.; Yanagita, T. Health benefits of conjugated linoleic acid (CLA). Obes. Res. Clin. Pr. 2014, 8, e525–e532. [Google Scholar] [CrossRef]
  138. den Hartigh, L.J. Conjugated Linoleic Acid Effects on Cancer, Obesity, and Atherosclerosis: A Review of Pre-Clinical and Human Trials with Current Perspectives. Nutrients 2018, 11, 370. [Google Scholar] [CrossRef]
  139. Omer, A.K.; Mohammed, R.R.; Ameen, P.S.M.; Abas, Z.A.; Ekici, K. Presence of Biogenic Amines in Food and Their Public Health Implications: A Review. J. Food Prot. 2021, 84, 1539–1548. [Google Scholar] [CrossRef] [PubMed]
  140. Herrero-Fresno, A.; Martínez, N.; Sánchez-Llana, E.; Díaz, M.; Fernández, M.; Martin, M.C.; Ladero, V.; Alvarez, M.A. Lactobacillus casei strains isolated from cheese reduce biogenic amine accumulation in an experimental model. Int. J. Food Microbiol. 2012, 157, 297–304. [Google Scholar] [CrossRef]
  141. Ołdak, A.; Zielińska, D.; Rzepkowska, A.; Kołożyn-Krajewska, D. Comparison of Antibacterial Activity of Lactobacillus plantarum Strains Isolated from Two Different Kinds of Regional Cheeses from Poland: Oscypek and Korycinski Cheese. Biomed. Res. Int. 2017, 2017, 6820369. [Google Scholar] [CrossRef]
  142. de Souza, B.M.S.; Borgonovi, T.F.; Casarotti, S.N.; Todorov, S.D.; Penna, A.L.B. Lactobacillus casei and Lactobacillus fermentum Strains Isolated from Mozzarella Cheese: Probiotic Potential, Safety, Acidifying Kinetic Parameters and Viability under Gastrointestinal Tract Conditions. Probiotics Antimicrob. Proteins 2019, 11, 382–396. [Google Scholar] [CrossRef]
  143. Meira, S.M.; Helfer, V.E.; Velho, R.V.; Lopes, F.C.; Brandelli, A. Probiotic potential of Lactobacillus spp. isolated from Brazilian regional ovine cheese. J. Dairy Res. 2012, 79, 119–127. [Google Scholar] [CrossRef] [PubMed]
  144. Summer, A.; Formaggioni, P.; Franceschi, P.; Di Frangia, F.; Righi, F.; Malacarne, M. Cheese as Functional Food: The Example of Parmigiano Reggiano and Grana Padano. Food Technol. Biotechnol. 2017, 55, 277–289. [Google Scholar] [CrossRef]
  145. Smoke, T.; Smoking, I. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. In Red Meat and Processed Meat; International Agency for Research on Cancer: Lyon, France, 2018. [Google Scholar]
  146. Toldrá, F.; Reig, M. Innovations for healthier processed meats. Trends Food Sci. Technol. 2011, 22, 517–522. [Google Scholar] [CrossRef]
  147. Kołożyn-Krajewska, D.; Dolatowski, Z.J. Probiotics in fermented meat products. Acta Sci. Pol. Technol. Aliment. 2009, 8, 61–74. [Google Scholar]
  148. Lücke, F.-K. Fermented meat products. Food Res. Int. 1994, 27, 299–307. [Google Scholar] [CrossRef]
  149. Rodzi, N.A.R.M.; Lee, L.K. Traditional fermented foods as vehicle of non-dairy probiotics: Perspectives in South East Asia countries. Food Res. Int. 2021, 150, 110814. [Google Scholar] [CrossRef]
  150. Leroy, F.; Verluyten, J.; De Vuyst, L. Functional meat starter cultures for improved sausage fermentation. Int. J. Food Microbiol. 2006, 106, 270–285. [Google Scholar] [CrossRef]
  151. Yılmaz, I.; Velioğlu, H. Fermented meat products. In Quality of Meat and Meat Products; Yilmaz, I., Ed.; Transworld Research Network: Trivandrum, India, 2009; pp. 1–16. [Google Scholar]
  152. Lücke, F.-K. Utilization of microbes to process and preserve meat. Meat Sci. 2000, 56, 105–115. [Google Scholar] [CrossRef]
  153. Swetwiwathana, A.; Visessanguan, W. Potential of bacteriocin-producing lactic acid bacteria for safety improvements of traditional Thai fermented meat and human health. Meat Sci. 2015, 109, 101–105. [Google Scholar] [CrossRef]
  154. Belleggia, L.; Ferrocino, I.; Reale, A.; Corvaglia, M.R.; Milanović, V.; Cesaro, C.; Boscaino, F.; Di Renzo, T.; Garofalo, C.; Cardinali, F.; et al. Unfolding microbiota and volatile organic compounds of Portuguese Painho de Porco Preto fermented sausages. Food Res. Int. 2022, 155, 111063. [Google Scholar] [CrossRef]
  155. Settanni, L.; Barbaccia, P.; Bonanno, A.; Ponte, M.; Di Gerlando, R.; Franciosi, E.; Di Grigoli, A.; Gaglio, R. Evolution of indigenous starter microorganisms and physicochemical parameters in spontaneously fermented beef, horse, wild boar and pork salamis produced under controlled conditions. Food Microbiol. 2020, 87, 103385. [Google Scholar] [CrossRef]
  156. López-Pedrouso, M.; Zaky, A.A.; Lorenzo, J.M.; Camiña, M.; Franco, D. A review on bioactive peptides derived from meat and by-products: Extraction methods, biological activities, applications and limitations. Meat Sci. 2023, 204, 109278. [Google Scholar] [CrossRef] [PubMed]
  157. Papavergou, E.J.; Savvaidis, I.N.; Ambrosiadis, I.A. Levels of biogenic amines in retail market fermented meat products. Food Chem. 2012, 135, 2750–2755. [Google Scholar] [CrossRef]
  158. Kukleci, E.; Smulders, F.J.M.; Hamidi, A.; Bauer, S.; Paulsen, P. Prevalence of Foodborne Pathogenic Bacteria, Microbial Levels of Hygiene Indicator Bacteria, and Concentrations of Biogenic Amines in Ready-to-Eat Meat Products at Retail in the Republic of Kosovo. J. Food Prot. 2019, 82, 1135–1140. [Google Scholar] [CrossRef]
  159. Song, M.Y.; Van-Ba, H.; Park, W.S.; Yoo, J.Y.; Kang, H.B.; Kim, J.H.; Kang, S.M.; Kim, B.M.; Oh, M.H.; Ham, J.S. Quality Characteristics of Functional Fermented Sausages Added with Encapsulated Probiotic Bifidobacterium longum KACC 91563. Korean J. Food Sci. Anim. Resour. 2018, 38, 981–994. [Google Scholar] [CrossRef] [PubMed]
  160. Stadnik, J.; J. Dolatowski, Z. Biogenic amines in meat and fermented meat products. Acta Sci. Pol. Technol. Aliment. 2010, 9, 251–263. [Google Scholar]
  161. Ashaolu, T.J.; Khalifa, I.; Mesak, M.A.; Lorenzo, J.M.; Farag, M.A. A comprehensive review of the role of microorganisms on texture change, flavor and biogenic amines formation in fermented meat with their action mechanisms and safety. Crit. Rev. Food Sci. Nutr. 2023, 63, 3538–3555. [Google Scholar] [CrossRef]
  162. Özbay, S. Determination of volatile N-nitrosamines formed in salami cooked by different processes. J. Food Compos. Anal. 2022, 112, 104691. [Google Scholar] [CrossRef]
  163. Herrmann, S.S.; Duedahl-Olesen, L.; Granby, K. Occurrence of volatile and non-volatile N-nitrosamines in processed meat products and the role of heat treatment. Food Control 2015, 48, 163–169. [Google Scholar] [CrossRef]
  164. Jofré, A.; Aymerich, T.; Garriga, M. Probiotic Fermented Sausages: Myth or Reality? Procedia Food Sci. 2015, 5, 133–136. [Google Scholar] [CrossRef]
  165. Łepecka, A.; Szymański, P.; Okoń, A.; Zielińska, D. Antioxidant activity of environmental lactic acid bacteria strains isolated from organic raw fermented meat products. LWT 2023, 174, 114440. [Google Scholar] [CrossRef]
  166. Gallego, M.; Mora, L.; Escudero, E.; Toldrá, F. Bioactive peptides and free amino acids profiles in different types of European dry-fermented sausages. Int. J. Food Microbiol. 2018, 276, 71–78. [Google Scholar] [CrossRef]
  167. Xie, Y.; Geng, Y.; Yao, J.; Ji, J.; Chen, F.; Xiao, J.; Hu, X.; Ma, L. N-nitrosamines in processed meats: Exposure, formation and mitigation strategies. J. Agric. Food Res. 2023, 13, 100645. [Google Scholar] [CrossRef]
  168. Yang, D.; Li, C.; Li, L.; Yang, X.; Chen, S.; Wu, Y.; Feng, Y. Novel insight into the formation and inhibition mechanism of dipeptidyl peptidase-Ⅳ inhibitory peptides from fermented mandarin fish (Chouguiyu). Food Sci. Hum. Wellness 2023, 12, 2408–2416. [Google Scholar] [CrossRef]
  169. Gupta, S.; Mohanty, U.; Majumdar, R.K. Isolation and characterization of lactic acid bacteria from traditional fermented fish product Shidal of India with reference to their probiotic potential. LWT 2021, 146, 111641. [Google Scholar] [CrossRef]
  170. Liu, J.; Sun, X.; Zhang, Y.; Jin, Y.; Sun, L.; Chai, X.; Wang, D.; Su, L.; Zhao, L. The impact of different fermenting microbes on residual purine content in fermented lamb jerky following in vitro digestion. Food Chem. 2023, 405, 134997. [Google Scholar] [CrossRef] [PubMed]
  171. Van Hecke, T.; Vossen, E.; Goethals, S.; Boon, N.; De Vrieze, J.; De Smet, S. In vitro and in vivo digestion of red cured cooked meat: Oxidation, intestinal microbiota and fecal metabolites. Food Res. Int. 2021, 142, 110203. [Google Scholar] [CrossRef]
  172. Roselino, M.N.; Sakamoto, I.K.; Tallarico Adorno, M.A.; Márcia Canaan, J.M.; de Valdez, G.F.; Rossi, E.A.; Sivieri, K.; Umbelino Cavallini, D.C. Effect of fermented sausages with probiotic Enterococcus faecium CRL 183 on gut microbiota using dynamic colonic model. LWT 2020, 132, 109876. [Google Scholar] [CrossRef]
  173. Keuleyan, E.; Bonifacie, A.; Sayd, T.; Duval, A.; Aubry, L.; Bourillon, S.; Gatellier, P.; Promeyrat, A.; Nassy, G.; Scislowski, V.; et al. In vitro digestion of nitrite and nitrate preserved fermented sausages—New understandings of nitroso-compounds’ chemical reactivity in the digestive tract. Food Chem. X 2022, 16, 100474. [Google Scholar] [CrossRef]
  174. Jahan, M.; Zhanel, G.G.; Sparling, R.; Holley, R.A. Horizontal transfer of antibiotic resistance from Enterococcus faecium of fermented meat origin to clinical isolates of E. faecium and Enterococcus faecalis. Int. J. Food Microbiol. 2015, 199, 78–85. [Google Scholar] [CrossRef]
  175. Soto, A.M.; Morales, P.; Haza, A.I.; García, M.L.; Selgas, M.D. Bioavailability of calcium from enriched meat products using Caco-2 cells. Food Res. Int. 2014, 55, 263–270. [Google Scholar] [CrossRef]
  176. Nissen, L.; Casciano, F.; Di Nunzio, M.; Galaverna, G.; Bordoni, A.; Gianotti, A. Effects of the replacement of nitrates/nitrites in salami by plant extracts on colon microbiota. Food Biosci. 2023, 53, 102568. [Google Scholar] [CrossRef]
  177. Borah, D.; Gogoi, O.; Adhikari, C.; Kakoti, B.B. Isolation and characterization of the new indigenous Staphylococcus sp. DBOCP06 as a probiotic bacterium from traditionally fermented fish and meat products of Assam state. Egypt. J. Basic. Appl. Sci. 2016, 3, 232–240. [Google Scholar] [CrossRef]
  178. Iqbal, R.; Dehghan, M.; Mente, A.; Rangarajan, S.; Wielgosz, A.; Avezum, A.; Seron, P.; AlHabib, K.F.; Lopez-Jaramillo, P.; Swaminathan, S.; et al. Associations of unprocessed and processed meat intake with mortality and cardiovascular disease in 21 countries [Prospective Urban Rural Epidemiology (PURE) Study]: A prospective cohort study. Am. J. Clin. Nutr. 2021, 114, 1049–1058. [Google Scholar] [CrossRef]
  179. Bovalino, S.; Charleson, G.; Szoeke, C. The impact of red and processed meat consumption on cardiovascular disease risk in women. Nutrition 2016, 32, 349–354. [Google Scholar] [CrossRef] [PubMed]
  180. Damigou, E.; Kosti, R.I.; Anastasiou, C.; Chrysohoou, C.; Barkas, F.; Adamidis, P.S.; Kravvariti, E.; Pitsavos, C.; Tsioufis, C.; Liberopoulos, E.; et al. Associations between meat type consumption pattern and incident cardiovascular disease: The ATTICA epidemiological cohort study (2002−2022). Meat Sci. 2023, 205, 109294. [Google Scholar] [CrossRef]
  181. de Medeiros, G.; Mesquita, G.X.B.; Lima, S.; Silva, D.F.O.; de Azevedo, K.P.M.; Pimenta, I.; de Oliveira, A.; Lyra, C.O.; Martínez, D.G.; Piuvezam, G. Associations of the consumption of unprocessed red meat and processed meat with the incidence of cardiovascular disease and mortality, and the dose-response relationship: A systematic review and meta-analysis of cohort studies. Crit. Rev. Food Sci. Nutr. 2022, 1–14. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, J.; Hayden, K.; Jackson, R.; Schutte, R. Association of red and processed meat consumption with cardiovascular morbidity and mortality in participants with and without obesity: A prospective cohort study. Clin. Nutr. 2021, 40, 3643–3649. [Google Scholar] [CrossRef] [PubMed]
  183. Montoro-García, S.; Zafrilla-Rentero, M.P.; Celdrán-de Haro, F.M.; Piñero-de Armas, J.J.; Toldrá, F.; Tejada-Portero, L.; Abellán-Alemán, J. Effects of dry-cured ham rich in bioactive peptides on cardiovascular health: A randomized controlled trial. J. Funct. Foods 2017, 38, 160–167. [Google Scholar] [CrossRef]
  184. Ayyash, M.; Liu, S.-Q.; Al Mheiri, A.; Aldhaheri, M.; Raeisi, B.; Al-Nabulsi, A.; Osaili, T.; Olaimat, A. In vitro investigation of health-promoting benefits of fermented camel sausage by novel probiotic Lactobacillus plantarum: A comparative study with beef sausages. LWT 2019, 99, 346–354. [Google Scholar] [CrossRef]
  185. Kong, Y.-w.; Feng, M.-q.; Sun, J. Effects of Lactobacillus plantarum CD101 and Staphylococcus simulans NJ201 on proteolytic changes and bioactivities (antioxidant and antihypertensive activities) in fermented pork sausage. LWT 2020, 133, 109985. [Google Scholar] [CrossRef]
  186. Mejri, L.; Vásquez-Villanueva, R.; Hassouna, M.; Marina, M.L.; García, M.C. Identification of peptides with antioxidant and antihypertensive capacities by RP-HPLC-Q-TOF-MS in dry fermented camel sausages inoculated with different starter cultures and ripening times. Food Res. Int. 2017, 100, 708–716. [Google Scholar] [CrossRef]
  187. Li, H.; Wu, J.; Wan, J.; Zhou, Y.; Zhu, Q. Extraction and identification of bioactive peptides from Panxian dry-cured ham with multifunctional activities. LWT 2022, 160, 113326. [Google Scholar] [CrossRef]
  188. Ohata, M.; Uchida, S.; Zhou, L.; Arihara, K. Antioxidant activity of fermented meat sauce and isolation of an associated antioxidant peptide. Food Chem. 2016, 194, 1034–1039. [Google Scholar] [CrossRef]
  189. Fernández, M.; Benito, M.J.; Martín, A.; Casquete, R.; Córdoba, J.J.; Córdoba, M.G. Influence of starter culture and a protease on the generation of ACE-inhibitory and antioxidant bioactive nitrogen compounds in Iberian dry-fermented sausage “salchichón”. Heliyon 2016, 2, e00093. [Google Scholar] [CrossRef]
  190. Zeraatkar, D.; Han, M.A.; Guyatt, G.H.; Vernooij, R.W.M.; El Dib, R.; Cheung, K.; Milio, K.; Zworth, M.; Bartoszko, J.J.; Valli, C.; et al. Red and Processed Meat Consumption and Risk for All-Cause Mortality and Cardiometabolic Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann. Intern. Med. 2019, 171, 703–710. [Google Scholar] [CrossRef] [PubMed]
  191. Vernooij, R.W.M.; Zeraatkar, D.; Han, M.A.; El Dib, R.; Zworth, M.; Milio, K.; Sit, D.; Lee, Y.; Gomaa, H.; Valli, C.; et al. Patterns of Red and Processed Meat Consumption and Risk for Cardiometabolic and Cancer Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann. Intern. Med. 2019, 171, 732–741. [Google Scholar] [CrossRef] [PubMed]
  192. Cui, K.; Liu, Y.; Zhu, L.; Mei, X.; Jin, P.; Luo, Y. Association between intake of red and processed meat and the risk of heart failure: A meta-analysis. BMC Public Health 2019, 19, 354. [Google Scholar] [CrossRef]
  193. Farvid, M.S.; Sidahmed, E.; Spence, N.D.; Mante Angua, K.; Rosner, B.A.; Barnett, J.B. Consumption of red meat and processed meat and cancer incidence: A systematic review and meta-analysis of prospective studies. Eur. J. Epidemiol. 2021, 36, 937–951. [Google Scholar] [CrossRef]
  194. Anderson, J.J.; Darwis, N.D.M.; Mackay, D.F.; Celis-Morales, C.A.; Lyall, D.M.; Sattar, N.; Gill, J.M.R.; Pell, J.P. Red and processed meat consumption and breast cancer: UK Biobank cohort study and meta-analysis. Eur. J. Cancer 2018, 90, 73–82. [Google Scholar] [CrossRef]
  195. Alexander, D.D.; Morimoto, L.M.; Mink, P.J.; Cushing, C.A. A review and meta-analysis of red and processed meat consumption and breast cancer. Nutr. Res. Rev. 2010, 23, 349–365. [Google Scholar] [CrossRef]
  196. Dandamudi, A.; Tommie, J.; Nommsen-Rivers, L.; Couch, S. Dietary Patterns and Breast Cancer Risk: A Systematic Review. Anticancer. Res. 2018, 38, 3209–3222. [Google Scholar] [CrossRef]
  197. Kim, S.R.; Kim, K.; Lee, S.A.; Kwon, S.O.; Lee, J.K.; Keum, N.; Park, S.M. Effect of Red, Processed, and White Meat Consumption on the Risk of Gastric Cancer: An Overall and Dose–Response Meta-Analysis. Nutrients 2019, 11, 826. [Google Scholar] [CrossRef]
  198. Chan, D.S.; Lau, R.; Aune, D.; Vieira, R.; Greenwood, D.C.; Kampman, E.; Norat, T. Red and processed meat and colorectal cancer incidence: Meta-analysis of prospective studies. PLoS ONE 2011, 6, e20456. [Google Scholar] [CrossRef]
  199. Ubago-Guisado, E.; Rodríguez-Barranco, M.; Ching-López, A.; Petrova, D.; Molina-Montes, E.; Amiano, P.; Barricarte-Gurrea, A.; Chirlaque, M.D.; Agudo, A.; Sánchez, M.J. Evidence Update on the Relationship between Diet and the Most Common Cancers from the European Prospective Investigation into Cancer and Nutrition (EPIC) Study: A Systematic Review. Nutrients 2021, 13, 3582. [Google Scholar] [CrossRef]
  200. Schwingshackl, L.; Schwedhelm, C.; Hoffmann, G.; Knüppel, S.; Laure Preterre, A.; Iqbal, K.; Bechthold, A.; De Henauw, S.; Michels, N.; Devleesschauwer, B.; et al. Food groups and risk of colorectal cancer. Int. J. Cancer 2018, 142, 1748–1758. [Google Scholar] [CrossRef]
  201. Vieira, A.R.; Abar, L.; Chan, D.S.M.; Vingeliene, S.; Polemiti, E.; Stevens, C.; Greenwood, D.; Norat, T. Foods and beverages and colorectal cancer risk: A systematic review and meta-analysis of cohort studies, an update of the evidence of the WCRF-AICR Continuous Update Project. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 1788–1802. [Google Scholar] [CrossRef]
  202. Händel, M.N.; Rohde, J.F.; Jacobsen, R.; Nielsen, S.M.; Christensen, R.; Alexander, D.D.; Frederiksen, P.; Heitmann, B.L. Processed meat intake and incidence of colorectal cancer: A systematic review and meta-analysis of prospective observational studies. Eur. J. Clin. Nutr. 2020, 74, 1132–1148. [Google Scholar] [CrossRef]
  203. Alexander, D.D.; Miller, A.J.; Cushing, C.A.; Lowe, K.A. Processed meat and colorectal cancer: A quantitative review of prospective epidemiologic studies. Eur. J. Cancer Prev. Off. J. Eur. Cancer Prev. Organ. (ECP) 2010, 19, 328–341. [Google Scholar] [CrossRef]
  204. Aune, D.; Chan, D.S.M.; Vieira, A.R.; Navarro Rosenblatt, D.A.; Vieira, R.; Greenwood, D.C.; Kampman, E.; Norat, T. Red and processed meat intake and risk of colorectal adenomas: A systematic review and meta-analysis of epidemiological studies. Cancer Causes Control 2013, 24, 611–627. [Google Scholar] [CrossRef] [PubMed]
  205. Rosato, V.; Negri, E.; Serraino, D.; Montella, M.; Libra, M.; Lagiou, P.; Facchini, G.; Ferraroni, M.; Decarli, A.; La Vecchia, C. Processed Meat and Risk of Renal Cell and Bladder Cancers. Nutr. Cancer 2018, 70, 418–424. [Google Scholar] [CrossRef] [PubMed]
  206. Crippa, A.; Larsson, S.C.; Discacciati, A.; Wolk, A.; Orsini, N. Red and processed meat consumption and risk of bladder cancer: A dose-response meta-analysis of epidemiological studies. Eur. J. Nutr. 2018, 57, 689–701. [Google Scholar] [CrossRef] [PubMed]
  207. Alexander, D.D.; Cushing, C.A. Quantitative assessment of red meat or processed meat consumption and kidney cancer. Cancer Detect. Prev. 2009, 32, 340–351. [Google Scholar] [CrossRef]
  208. Saneei, P.; Willett, W.; Esmaillzadeh, A. Red and processed meat consumption and risk of glioma in adults: A systematic review and meta-analysis of observational studies. J. Res. Med. Sci. Off. J. Isfahan Univ. Med. Sci. 2015, 20, 602–612. [Google Scholar] [CrossRef]
  209. Yu, J.; Liu, Z.; Liang, D.; Li, J.; Ma, S.; Wang, G.; Chen, W. Meat Intake and the Risk of Hepatocellular Carcinoma: A Meta-Analysis of Observational Studies. Nutr. Cancer 2022, 74, 3340–3350. [Google Scholar] [CrossRef] [PubMed]
  210. Schwingshackl, L.; Hoffmann, G.; Lampousi, A.M.; Knüppel, S.; Iqbal, K.; Schwedhelm, C.; Bechthold, A.; Schlesinger, S.; Boeing, H. Food groups and risk of type 2 diabetes mellitus: A systematic review and meta-analysis of prospective studies. Eur. J. Epidemiol. 2017, 32, 363–375. [Google Scholar] [CrossRef] [PubMed]
  211. Micha, R.; Wallace, S.K.; Mozaffarian, D. Red and processed meat consumption and risk of incident coronary heart disease, stroke, and diabetes mellitus: A systematic review and meta-analysis. Circulation 2010, 121, 2271–2283. [Google Scholar] [CrossRef] [PubMed]
  212. Yang, X.; Li, Y.; Wang, C.; Mao, Z.; Zhou, W.; Zhang, L.; Fan, M.; Cui, S.; Li, L. Meat and fish intake and type 2 diabetes: Dose–response meta-analysis of prospective cohort studies. Diabetes Metab. 2020, 46, 345–352. [Google Scholar] [CrossRef]
  213. Zhang, R.; Fu, J.; Moore, J.B.; Stoner, L.; Li, R. Processed and Unprocessed Red Meat Consumption and Risk for Type 2 Diabetes Mellitus: An Updated Meta-Analysis of Cohort Studies. Int. J. Environ. Res. Public Health 2021, 18, 10788. [Google Scholar] [CrossRef]
  214. Mijatovic-Vukas, J.; Capling, L.; Cheng, S.; Stamatakis, E.; Louie, J.; Cheung, N.W.; Markovic, T.; Ross, G.; Senior, A.; Brand-Miller, J.C.; et al. Associations of Diet and Physical Activity with Risk for Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 698. [Google Scholar] [CrossRef]
  215. Albenberg, L.; Brensinger, C.M.; Wu, Q.; Gilroy, E.; Kappelman, M.D.; Sandler, R.S.; Lewis, J.D. A Diet Low in Red and Processed Meat Does Not Reduce Rate of Crohn’s Disease Flares. Gastroenterology 2019, 157, 128–136.e5. [Google Scholar] [CrossRef]
  216. Taneri, P.E.; Wehrli, F.; Roa-Díaz, Z.M.; Itodo, O.A.; Salvador, D.; Raeisi-Dehkordi, H.; Bally, L.; Minder, B.; Kiefte-de Jong, J.C.; Laine, J.E.; et al. Association Between Ultra-Processed Food Intake and All-Cause Mortality: A Systematic Review and Meta-Analysis. Am. J. Epidemiol. 2022, 191, 1323–1335. [Google Scholar] [CrossRef]
  217. Wang, X.; Lin, X.; Ouyang, Y.Y.; Liu, J.; Zhao, G.; Pan, A.; Hu, F.B. Red and processed meat consumption and mortality: Dose-response meta-analysis of prospective cohort studies. Public Health Nutr. 2016, 19, 893–905. [Google Scholar] [CrossRef]
  218. Nucci, D.; Fatigoni, C.; Amerio, A.; Odone, A.; Gianfredi, V. Red and Processed Meat Consumption and Risk of Depression: A Systematic Review and Meta-Analysis. Int. J. Environ. Res. Public Health 2020, 17, 6686. [Google Scholar] [CrossRef] [PubMed]
  219. Michaud, D.S.; Holick, C.N.; Batchelor, T.T.; Giovannucci, E.; Hunter, D.J. Prospective study of meat intake and dietary nitrates, nitrites, and nitrosamines and risk of adult glioma12. Am. J. Clin. Nutr. 2009, 90, 570–577. [Google Scholar] [CrossRef]
  220. Travis, C.C.; McClain, T.W.; Birkner, P.D. Diethylnitrosamine-induced hepatocarcinogenesis in rats: A theoretical study. Toxicol. Appl. Pharmacol. 1991, 109, 289–304. [Google Scholar] [CrossRef]
  221. Septembre-Malaterre, A.; Remize, F.; Poucheret, P. Fruits and vegetables, as a source of nutritional compounds and phytochemicals: Changes in bioactive compounds during lactic fermentation. Food Res. Int. 2018, 104, 86–99. [Google Scholar] [CrossRef] [PubMed]
  222. Jiang, Z.; Sun, T.Y.; He, Y.; Gou, W.; Zuo, L.S.; Fu, Y.; Miao, Z.; Shuai, M.; Xu, F.; Xiao, C.; et al. Dietary fruit and vegetable intake, gut microbiota, and type 2 diabetes: Results from two large human cohort studies. BMC Med. 2020, 18, 371. [Google Scholar] [CrossRef]
  223. Aune, D.; Giovannucci, E.; Boffetta, P.; Fadnes, L.T.; Keum, N.; Norat, T.; Greenwood, D.C.; Riboli, E.; Vatten, L.J.; Tonstad, S. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality-a systematic review and dose-response meta-analysis of prospective studies. Int. J. Epidemiol. 2017, 46, 1029–1056. [Google Scholar] [CrossRef]
  224. Wang, D.D.; Li, Y.; Bhupathiraju, S.N.; Rosner, B.A.; Sun, Q.; Giovannucci, E.L.; Rimm, E.B.; Manson, J.E.; Willett, W.C.; Stampfer, M.J.; et al. Fruit and Vegetable Intake and Mortality: Results from 2 Prospective Cohort Studies of US Men and Women and a Meta-Analysis of 26 Cohort Studies. Circulation 2021, 143, 1642–1654. [Google Scholar] [CrossRef]
  225. Di Cagno, R.; Coda, R.; De Angelis, M.; Gobbetti, M. Exploitation of vegetables and fruits through lactic acid fermentation. Food Microbiol. 2013, 33, 1–10. [Google Scholar] [CrossRef]
  226. Sabater, C.; Ruiz, L.; Delgado, S.; Ruas-Madiedo, P.; Margolles, A. Valorization of Vegetable Food Waste and By-Products through Fermentation Processes. Front. Microbiol. 2020, 11, 581997. [Google Scholar] [CrossRef]
  227. Harris, J.; Tan, W.; Raneri, J.E.; Schreinemachers, P.; Herforth, A. Vegetables for Healthy Diets in Low- and Middle-Income Countries: A Scoping Review of the Food Systems Literature. Food Nutr. Bull. 2022, 43, 232–248. [Google Scholar] [CrossRef]
  228. Irakoze, M.L.; Wafula, E.N.; Owaga, E. Potential Role of African Fermented Indigenous Vegetables in Maternal and Child Nutrition in Sub-Saharan Africa. Int. J. Food Sci. 2021, 2021, 3400329. [Google Scholar] [CrossRef]
  229. Ashaolu, T.J.; Reale, A. A holistic review on Euro-Asian lactic acid bacteria fermented cereals and vegetables. Microorganisms 2020, 8, 1176. [Google Scholar] [PubMed]
  230. Lee, S.J.; Jeon, H.S.; Yoo, J.Y.; Kim, J.H. Some Important Metabolites Produced by Lactic Acid Bacteria Originated from Kimchi. Foods 2021, 10, 2148. [Google Scholar] [CrossRef]
  231. Park, K.Y.; Jeong, J.K.; Lee, Y.E.; Daily, J.W., 3rd. Health benefits of kimchi (Korean fermented vegetables) as a probiotic food. J. Med. Food 2014, 17, 6–20. [Google Scholar] [CrossRef] [PubMed]
  232. Kim, H.J.; Noh, J.S.; Song, Y.O. Beneficial Effects of Kimchi, a Korean Fermented Vegetable Food, on Pathophysiological Factors Related to Atherosclerosis. J. Med. Food 2018, 21, 127–135. [Google Scholar] [CrossRef] [PubMed]
  233. Woo, M.; Kim, M.J.; Song, Y.O. Bioactive Compounds in Kimchi Improve the Cognitive and Memory Functions Impaired by Amyloid Beta. Nutrients 2018, 10, 1554. [Google Scholar] [CrossRef]
  234. Kim, H.J.; Lee, J.S.; Chung, H.Y.; Song, S.H.; Suh, H.; Noh, J.S.; Song, Y.O. 3-(4′-hydroxyl-3′,5′-dimethoxyphenyl)propionic acid, an active principle of kimchi, inhibits development of atherosclerosis in rabbits. J. Agric. Food Chem. 2007, 55, 10486–10492. [Google Scholar] [CrossRef]
  235. Yun, Y.R.; Kim, H.J.; Song, Y.O. Kimchi methanol extract and the kimchi active compound, 3′-(4′-hydroxyl-3′,5′-dimethoxyphenyl)propionic acid, downregulate CD36 in THP-1 macrophages stimulated by oxLDL. J. Med. Food 2014, 17, 886–893. [Google Scholar] [CrossRef] [PubMed]
  236. Noh, J.S.; Kim, H.J.; Kwon, M.J.; Song, Y.O. Active principle of kimchi, 3-(4′-hydroxyl-3′,5′-dimethoxyphenyl)propionic acid, retards fatty streak formation at aortic sinus of apolipoprotein E knockout mice. J. Med. Food 2009, 12, 1206–1212. [Google Scholar] [CrossRef]
  237. Jeong, J.W.; Choi, I.W.; Jo, G.H.; Kim, G.Y.; Kim, J.; Suh, H.; Ryu, C.H.; Kim, W.J.; Park, K.Y.; Choi, Y.H. Anti-Inflammatory Effects of 3-(4′-Hydroxyl-3′,5′-Dimethoxyphenyl)Propionic Acid, an Active Component of Korean Cabbage Kimchi, in Lipopolysaccharide-Stimulated BV2 Microglia. J. Med. Food 2015, 18, 677–684. [Google Scholar] [CrossRef]
  238. Jeon, H.L.; Lee, N.K.; Yang, S.J.; Kim, W.S.; Paik, H.D. Probiotic characterization of Bacillus subtilis P223 isolated from kimchi. Food Sci. Biotechnol. 2017, 26, 1641–1648. [Google Scholar] [CrossRef]
  239. Yu, H.S.; Lee, N.K.; Choi, A.J.; Choe, J.S.; Bae, C.H.; Paik, H.D. Anti-Inflammatory Potential of Probiotic Strain Weissella cibaria JW15 Isolated from Kimchi through Regulation of NF-κB and MAPKs Pathways in LPS-Induced RAW 264.7 Cells. J. Microbiol. Biotechnol. 2019, 29, 1022–1032. [Google Scholar] [CrossRef]
  240. Sohn, H.; Chang, Y.H.; Yune, J.H.; Jeong, C.H.; Shin, D.M.; Kwon, H.C.; Kim, D.H.; Hong, S.W.; Hwang, H.; Jeong, J.Y.; et al. Probiotic Properties of Lactiplantibacillus plantarum LB5 Isolated from Kimchi Based on Nitrate Reducing Capability. Foods 2020, 9, 1777. [Google Scholar] [CrossRef]
  241. Yoon, S.; Cho, H.; Nam, Y.; Park, M.; Lim, A.; Kim, J.H.; Park, J.; Kim, W. Multifunctional Probiotic and Functional Properties of Lactiplantibacillus plantarum LRCC5314, Isolated from Kimchi. J. Microbiol. Biotechnol. 2022, 32, 72–80. [Google Scholar] [CrossRef] [PubMed]
  242. Cheon, M.J.; Lee, N.K.; Paik, H.D. Neuroprotective Effects of Heat-Killed Lactobacillus plantarum 200655 Isolated from Kimchi against Oxidative Stress. Probiotics Antimicrob Proteins 2021, 13, 788–795. [Google Scholar] [CrossRef] [PubMed]
  243. Lim, S.; Moon, J.H.; Shin, C.M.; Jeong, D.; Kim, B. Effect of Lactobacillus sakei, a Probiotic Derived from Kimchi, on Body Fat in Koreans with Obesity: A Randomized Controlled Study. Endocrinol. Metab. 2020, 35, 425–434. [Google Scholar] [CrossRef]
  244. Jang, H.J.; Lee, N.K.; Paik, H.D. Probiotic characterization of Lactobacillus brevis KU15153 showing antimicrobial and antioxidant effect isolated from kimchi. Food Sci. Biotechnol. 2019, 28, 1521–1528. [Google Scholar] [CrossRef]
  245. Kim, K.T.; Yang, S.J.; Paik, H.D. Probiotic properties of novel probiotic Levilactobacillus brevis KU15147 isolated from radish kimchi and its antioxidant and immune-enhancing activities. Food Sci. Biotechnol. 2021, 30, 257–265. [Google Scholar] [CrossRef]
  246. Youn, H.S.; Kim, J.H.; Lee, J.S.; Yoon, Y.Y.; Choi, S.J.; Lee, J.Y.; Kim, W.; Hwang, K.W. Lactobacillus plantarum Reduces Low-Grade Inflammation and Glucose Levels in a Mouse Model of Chronic Stress and Diabetes. Infect. Immun. 2021, 89, e0061520. [Google Scholar] [CrossRef]
  247. An, J.M.; Kang, E.A.; Han, Y.M.; Oh, J.Y.; Lee, D.Y.; Choi, S.H.; Kim, D.H.; Hahm, K.B. Dietary intake of probiotic kimchi ameliorated IL-6-driven cancer cachexia. J. Clin. Biochem. Nutr. 2019, 65, 109–117. [Google Scholar] [CrossRef]
  248. Shankar, T.; Palpperumal, S.; Kathiresan, D.; Sankaralingam, S.; Balachandran, C.; Baskar, K.; Hashem, A.; Alqarawi, A.A.; Abd Allah, E.F. Biomedical and therapeutic potential of exopolysaccharides by Lactobacillus paracasei isolated from sauerkraut: Screening and characterization. Saudi J. Biol. Sci. 2021, 28, 2943–2950. [Google Scholar] [CrossRef] [PubMed]
  249. Xu, X.; Qiao, Y.; Peng, Q.; Shi, B.; Dia, V.P. Antioxidant and Immunomodulatory Properties of Partially purified Exopolysaccharide from Lactobacillus Casei Isolated from Chinese Northeast Sauerkraut. Immunol. Investig. 2022, 51, 748–765. [Google Scholar] [CrossRef] [PubMed]
  250. Xu, X.; Peng, Q.; Zhang, Y.; Tian, D.; Zhang, P.; Huang, Y.; Ma, L.; Dia, V.P.; Qiao, Y.; Shi, B. Antibacterial potential of a novel Lactobacillus casei strain isolated from Chinese northeast sauerkraut and the antibiofilm activity of its exopolysaccharides. Food Funct. 2020, 11, 4697–4706. [Google Scholar] [CrossRef]
  251. Yu, Z.; Zhang, X.; Li, S.; Li, C.; Li, D.; Yang, Z. Evaluation of probiotic properties of Lactobacillus plantarum strains isolated from Chinese sauerkraut. World J. Microbiol. Biotechnol. 2013, 29, 489–498. [Google Scholar] [CrossRef]
  252. Kim, N.; Lee, J.; Song, H.S.; Oh, Y.J.; Kwon, M.S.; Yun, M.; Lim, S.K.; Park, H.K.; Jang, Y.S.; Lee, S.; et al. Kimchi intake alleviates obesity-induced neuroinflammation by modulating the gut-brain axis. Food Res. Int. 2022, 158, 111533. [Google Scholar] [CrossRef] [PubMed]
  253. Kim, H.Y.; Park, E.S.; Choi, Y.S.; Park, S.J.; Kim, J.H.; Chang, H.K.; Park, K.Y. Kimchi improves irritable bowel syndrome: Results of a randomized, double-blind placebo-controlled study. Food Nutr. Res. 2022, 66, 1–12. [Google Scholar] [CrossRef]
  254. Park, J.M.; Lee, W.H.; Seo, H.; Oh, J.Y.; Lee, D.Y.; Kim, S.J.; Hahm, K.B. Fecal microbiota changes with fermented kimchi intake regulated either formation or advancement of colon adenoma. J. Clin. Biochem. Nutr. 2021, 68, 139–148. [Google Scholar] [CrossRef]
  255. Park, S.E.; Kwon, S.J.; Cho, K.M.; Seo, S.H.; Kim, E.J.; Unno, T.; Bok, S.H.; Park, D.H.; Son, H.S. Intervention with kimchi microbial community ameliorates obesity by regulating gut microbiota. J. Microbiol. 2020, 58, 859–867. [Google Scholar] [CrossRef]
  256. An, S.Y.; Lee, M.S.; Jeon, J.Y.; Ha, E.S.; Kim, T.H.; Yoon, J.Y.; Ok, C.O.; Lee, H.K.; Hwang, W.S.; Choe, S.J.; et al. Beneficial effects of fresh and fermented kimchi in prediabetic individuals. Ann. Nutr. Metab. 2013, 63, 111–119. [Google Scholar] [CrossRef]
  257. Islam, M.S.; Choi, H. Antidiabetic effect of Korean traditional Baechu (Chinese cabbage) kimchi in a type 2 diabetes model of rats. J. Med. Food 2009, 12, 292–297. [Google Scholar] [CrossRef]
  258. Palani, K.; Harbaum-Piayda, B.; Meske, D.; Keppler, J.K.; Bockelmann, W.; Heller, K.J.; Schwarz, K. Influence of fermentation on glucosinolates and glucobrassicin degradation products in sauerkraut. Food Chem. 2016, 190, 755–762. [Google Scholar] [CrossRef] [PubMed]
  259. Tai, A.; Fukunaga, K.; Ohno, A.; Ito, H. Antioxidative properties of ascorbigen in using multiple antioxidant assays. Biosci. Biotechnol. Biochem. 2014, 78, 1723–1730. [Google Scholar] [CrossRef]
  260. Amarakoon, D.; Lee, W.J.; Tamia, G.; Lee, S.H. Indole-3-Carbinol: Occurrence, Health-Beneficial Properties, and Cellular/Molecular Mechanisms. Annu. Rev. Food Sci. Technol. 2023, 14, 347–366. [Google Scholar] [CrossRef] [PubMed]
  261. Pathak, D.R.; Stein, A.D.; He, J.P.; Noel, M.M.; Hembroff, L.; Nelson, D.A.; Vigneau, F.; Shen, T.; Scott, L.J.; Charzewska, J.; et al. Cabbage and Sauerkraut Consumption in Adolescence and Adulthood and Breast Cancer Risk among US-Resident Polish Migrant Women. Int. J. Environ. Res. Public Health 2021, 18, 10795. [Google Scholar] [CrossRef]
  262. Nielsen, E.S.; Garnås, E.; Jensen, K.J.; Hansen, L.H.; Olsen, P.S.; Ritz, C.; Krych, L.; Nielsen, D.S. Lacto-fermented sauerkraut improves symptoms in IBS patients independent of product pasteurisation—A pilot study. Food Funct. 2018, 9, 5323–5335. [Google Scholar] [CrossRef]
  263. Fideler, J.; Johanningsmeier, S.D.; Ekelöf, M.; Muddiman, D.C. Discovery and quantification of bioactive peptides in fermented cucumber by direct analysis IR-MALDESI mass spectrometry and LC-QQQ-MS. Food Chem. 2019, 271, 715–723. [Google Scholar] [CrossRef]
  264. Moore, J.F.; DuVivier, R.; Johanningsmeier, S.D. Changes in the free amino acid profile of pickling cucumber during lactic acid fermentation. J. Food Sci. 2022, 87, 599–611. [Google Scholar] [CrossRef]
  265. Petroski, W.; Minich, D.M. Is There Such a Thing as “Anti-Nutrients”? A Narrative Review of Perceived Problematic Plant Compounds. Nutrients 2020, 12, 2929. [Google Scholar] [CrossRef]
  266. Cuadrado, C.; Hajos, G.; Burbano, C.; Pedrosa, M.M.; Ayet, G.; Muzquiz, M.; Pusztai, A.; Gelencser, E. Effect of natural fermentation on the lectin of lentils measured by immunological methods. Food Agric. Immunol. 2002, 14, 41–49. [Google Scholar]
  267. Sangija, F.; Martin, H.; Matemu, A. Effect of lactic acid fermentation on the nutritional quality and consumer acceptability of African nightshade. Food Sci. Nutr. 2022, 10, 3128–3142. [Google Scholar] [CrossRef]
  268. Knez, E.; Kadac-Czapska, K.; Grembecka, M. Effect of Fermentation on the Nutritional Quality of the Selected Vegetables and Legumes and Their Health Effects. Life 2023, 13, 655. [Google Scholar] [CrossRef] [PubMed]
  269. Layla, A.; Syed, Q.A.; Zahoor, T.; Shahid, M. Investigating the role of Lactiplantibacillus plantarum vs. spontaneous fermentation in improving nutritional and consumer safety of the fermented white cabbage sprouts. Int. Microbiol. 2023, 1–12. [Google Scholar] [CrossRef]
  270. Dreher, M.L. Whole Fruits and Fruit Fiber Emerging Health Effects. Nutrients 2018, 10, 1833. [Google Scholar] [CrossRef]
  271. Leitão, M.; Ribeiro, T.; García, P.A.; Barreiros, L.; Correia, P. Benefits of Fermented Papaya in Human Health. Foods 2022, 11, 563. [Google Scholar] [CrossRef]
  272. Cousin, F.J.; Le Guellec, R.; Schlusselhuber, M.; Dalmasso, M.; Laplace, J.M.; Cretenet, M. Microorganisms in Fermented Apple Beverages: Current Knowledge and Future Directions. Microorganisms 2017, 5, 39. [Google Scholar] [CrossRef]
  273. Lee, B.H.; Hsu, W.H.; Hou, C.Y.; Chien, H.Y.; Wu, S.C. The Protection of Lactic Acid Bacteria Fermented-Mango Peel against Neuronal Damage Induced by Amyloid-Beta. Molecules 2021, 26, 3503. [Google Scholar] [CrossRef] [PubMed]
  274. Huang, C.H.; Hsiao, S.Y.; Lin, Y.H.; Tsai, G.J. Effects of Fermented Citrus Peel on Ameliorating Obesity in Rats Fed with High-Fat Diet. Molecules 2022, 27, 8966. [Google Scholar] [CrossRef] [PubMed]
  275. Wu, C.C.; Huang, Y.W.; Hou, C.Y.; Chen, Y.T.; Dong, C.D.; Chen, C.W.; Singhania, R.R.; Leang, J.Y.; Hsieh, S.L. Lemon fermented products prevent obesity in high-fat diet-fed rats by modulating lipid metabolism and gut microbiota. J. Food Sci. Technol. 2023, 60, 1036–1044. [Google Scholar] [CrossRef] [PubMed]
  276. Yang, J.; Sun, Y.; Gao, T.; Wu, Y.; Sun, H.; Zhu, Q.; Liu, C.; Zhou, C.; Han, Y.; Tao, Y. Fermentation and Storage Characteristics of “Fuji” Apple Juice Using Lactobacillus acidophilus, Lactobacillus casei and Lactobacillus plantarum: Microbial Growth, Metabolism of Bioactives and in vitro Bioactivities. Front. Nutr. 2022, 9, 833906. [Google Scholar] [CrossRef]
  277. Yassunaka Hata, N.N.; Surek, M.; Sartori, D.; Vassoler Serrato, R.; Aparecida Spinosa, W. Role of Acetic Acid Bacteria in Food and Beverages. Food Technol. Biotechnol. 2023, 61, 85–103. [Google Scholar] [CrossRef] [PubMed]
  278. Perjéssy, J.; Hegyi, F.; Nagy-Gasztonyi, M.; Zalán, Z. Effect of the lactic acid fermentation by probiotic strains on the sour cherry juice and its bioactive compounds. Food Sci. Technol. Int. 2022, 28, 408–420. [Google Scholar] [CrossRef]
  279. Muhialdin, B.J.; Kadum, H.; Zarei, M.; Meor Hussin, A.S. Effects of metabolite changes during lacto-fermentation on the biological activity and consumer acceptability for dragon fruit juice. LWT 2020, 121, 108992. [Google Scholar] [CrossRef]
  280. Cirlini, M.; Ricci, A.; Galaverna, G.; Lazzi, C. Application of lactic acid fermentation to elderberry juice: Changes in acidic and glucidic fractions. LWT 2020, 118, 108779. [Google Scholar] [CrossRef]
  281. Wang, Z.; Feng, Y.; Yang, N.; Jiang, T.; Xu, H.; Lei, H. Fermentation of kiwifruit juice from two cultivars by probiotic bacteria: Bioactive phenolics, antioxidant activities and flavor volatiles. Food Chem. 2022, 373, 131455. [Google Scholar] [CrossRef] [PubMed]
  282. Wu, Y.; Li, S.; Tao, Y.; Li, D.; Han, Y.; Show, P.L.; Wen, G.; Zhou, J. Fermentation of blueberry and blackberry juices using Lactobacillus plantarum, Streptococcus thermophilus and Bifidobacterium bifidum: Growth of probiotics, metabolism of phenolics, antioxidant capacity in vitro and sensory evaluation. Food Chem. 2021, 348, 129083. [Google Scholar] [CrossRef]
  283. Zhong, H.; Abdullah; Zhao, M.; Tang, J.; Deng, L.; Feng, F. Probiotics-fermented blueberry juices as potential antidiabetic product: Antioxidant, antimicrobial and antidiabetic potentials. J. Sci. Food Agric. 2021, 101, 4420–4427. [Google Scholar] [CrossRef]
  284. Ousaaid, D.; Mechchate, H.; Laaroussi, H.; Hano, C.; Bakour, M.; El Ghouizi, A.; Conte, R.; Lyoussi, B.; El Arabi, I. Fruits Vinegar: Quality Characteristics, Phytochemistry, and Functionality. Molecules 2021, 27, 222. [Google Scholar] [CrossRef]
  285. Bakir, S.; Toydemir, G.; Boyacioglu, D.; Beekwilder, J.; Capanoglu, E. Fruit Antioxidants during Vinegar Processing: Changes in Content and in Vitro Bio-Accessibility. Int. J. Mol. Sci. 2016, 17, 1658. [Google Scholar] [CrossRef]
  286. Budak, H.N.; Guzel-Seydim, Z.B. Antioxidant activity and phenolic content of wine vinegars produced by two different techniques. J. Sci. Food Agric. 2010, 90, 2021–2026. [Google Scholar] [CrossRef]
  287. Hadi, A.; Pourmasoumi, M.; Najafgholizadeh, A.; Clark, C.C.T.; Esmaillzadeh, A. The effect of apple cider vinegar on lipid profiles and glycemic parameters: A systematic review and meta-analysis of randomized clinical trials. BMC Complement. Med. Ther. 2021, 21, 179. [Google Scholar] [CrossRef]
  288. Gheflati, A.; Bashiri, R.; Ghadiri-Anari, A.; Reza, J.Z.; Kord, M.T.; Nadjarzadeh, A. The effect of apple vinegar consumption on glycemic indices, blood pressure, oxidative stress, and homocysteine in patients with type 2 diabetes and dyslipidemia: A randomized controlled clinical trial. Clin. Nutr. ESPEN 2019, 33, 132–138. [Google Scholar] [CrossRef]
  289. Ousaaid, D.; Laaroussi, H.; Bakour, M.; ElGhouizi, A.; Aboulghazi, A.; Lyoussi, B.; ElArabi, I. Beneficial Effects of Apple Vinegar on Hyperglycemia and Hyperlipidemia in Hypercaloric-Fed Rats. J. Diabetes Res. 2020, 2020, 9284987. [Google Scholar] [CrossRef]
  290. Halima, B.H.; Sonia, G.; Sarra, K.; Houda, B.J.; Fethi, B.S.; Abdallah, A. Apple Cider Vinegar Attenuates Oxidative Stress and Reduces the Risk of Obesity in High-Fat-Fed Male Wistar Rats. J. Med. Food 2018, 21, 70–80. [Google Scholar] [CrossRef]
  291. Yagnik, D.; Serafin, V.; Shah, A.J. Antimicrobial activity of apple cider vinegar against Escherichia coli, Staphylococcus aureus and Candida albicans; downregulating cytokine and microbial protein expression. Sci. Rep. 2018, 8, 1732. [Google Scholar] [CrossRef]
  292. Tripathi, S.; Kumari, U.; Mitra Mazumder, P. Ameliorative effects of apple cider vinegar on neurological complications via regulation of oxidative stress markers. J. Food Biochem. 2020, 44, e13504. [Google Scholar] [CrossRef]
  293. Shams, F.; Aghajani-Nasab, M.; Ramezanpour, M.; Fatideh, R.H.; Mohammadghasemi, F. Effect of apple vinegar on folliculogenesis and ovarian kisspeptin in a high-fat diet-induced nonalcoholic fatty liver disease in rat. BMC Endocr. Disord. 2022, 22, 330. [Google Scholar] [CrossRef]
  294. Hlebowicz, J.; Darwiche, G.; Björgell, O.; Almér, L.O. Effect of apple cider vinegar on delayed gastric emptying in patients with type 1 diabetes mellitus: A pilot study. BMC Gastroenterol. 2007, 7, 46. [Google Scholar] [CrossRef]
  295. Bounihi, A.; Bitam, A.; Bouazza, A.; Yargui, L.; Koceir, E.A. Fruit vinegars attenuate cardiac injury via anti-inflammatory and anti-adiposity actions in high-fat diet-induced obese rats. Pharm. Biol. 2017, 55, 43–52. [Google Scholar] [CrossRef] [PubMed]
  296. James, A.; Yao, T.; Ke, H.; Wang, Y. Microbiota for production of wine with enhanced functional components. Food Sci. Hum. Wellness 2023, 12, 1481–1492. [Google Scholar] [CrossRef]
  297. Garaguso, I.; Nardini, M. Polyphenols content, phenolics profile and antioxidant activity of organic red wines produced without sulfur dioxide/sulfites addition in comparison to conventional red wines. Food Chem. 2015, 179, 336–342. [Google Scholar] [CrossRef]
  298. Pintać, D.; Bekvalac, K.; Mimica-Dukić, N.; Rašeta, M.; Anđelić, N.; Lesjak, M.; Orčić, D. Comparison study between popular brands of coffee, tea and red wine regarding polyphenols content and antioxidant activity. Food Chem. Adv. 2022, 1, 100030. [Google Scholar] [CrossRef]
  299. Ju, Y.; Yang, L.; Yue, X.; Li, Y.; He, R.; Deng, S.; Yang, X.; Fang, Y. Anthocyanin profiles and color properties of red wines made from Vitis davidii and Vitis vinifera grapes. Food Sci. Hum. Wellness 2021, 10, 335–344. [Google Scholar] [CrossRef]
  300. Rodriguez-Naranjo, M.I.; Gil-Izquierdo, A.; Troncoso, A.M.; Cantos-Villar, E.; Garcia-Parrilla, M.C. Melatonin is synthesised by yeast during alcoholic fermentation in wines. Food Chem. 2011, 126, 1608–1613. [Google Scholar] [CrossRef]
  301. Viegas, O.; Esteves, C.; Rocha, J.; Melo, A.; Ferreira, I.M.P.L.V.O. Simultaneous determination of melatonin and trans-resveratrol in wine by dispersive liquid–liquid microextraction followed by HPLC-FLD. Food Chem. 2021, 339, 128091. [Google Scholar] [CrossRef]
  302. Álvarez-Fernández, M.A.; Fernández-Cruz, E.; Cantos-Villar, E.; Troncoso, A.M.; García-Parrilla, M.C. Determination of hydroxytyrosol produced by winemaking yeasts during alcoholic fermentation using a validated UHPLC–HRMS method. Food Chem. 2018, 242, 345–351. [Google Scholar] [CrossRef] [PubMed]
  303. Micallef, M.; Lexis, L.; Lewandowski, P. Red wine consumption increases antioxidant status and decreases oxidative stress in the circulation of both young and old humans. Nutr. J. 2007, 6, 27. [Google Scholar] [CrossRef]
  304. Tedesco, I.; Spagnuolo, C.; Russo, G.L.; Russo, M.; Cervellera, C.; Moccia, S. The Pro-Oxidant Activity of Red Wine Polyphenols Induces an Adaptive Antioxidant Response in Human Erythrocytes. Antioxidants 2021, 10, 800. [Google Scholar] [CrossRef]
  305. Wang, P.; Gao, J.; Ke, W.; Wang, J.; Li, D.; Liu, R.; Jia, Y.; Wang, X.; Chen, X.; Chen, F.; et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic. Biol. Med. 2020, 156, 83–98. [Google Scholar] [CrossRef]
  306. Di Lorenzo, A.; Bloise, N.; Meneghini, S.; Sureda, A.; Tenore, G.C.; Visai, L.; Arciola, C.R.; Daglia, M. Effect of Winemaking on the Composition of Red Wine as a Source of Polyphenols for Anti-Infective Biomaterials. Materials 2016, 9, 316. [Google Scholar] [CrossRef]
  307. Chalons, P.; Courtaut, F.; Limagne, E.; Chalmin, F.; Cantos-Villar, E.; Richard, T.; Auger, C.; Chabert, P.; Schini-Kerth, V.; Ghiringhelli, F.; et al. Red Wine Extract Disrupts Th17 Lymphocyte Differentiation in a Colorectal Cancer Context. Mol. Nutr. Food Res. 2020, 64, e1901286. [Google Scholar] [CrossRef]
  308. Mahjabeen, W.; Khan, D.A.; Mirza, S.A. Role of resveratrol supplementation in regulation of glucose hemostasis, inflammation and oxidative stress in patients with diabetes mellitus type 2: A randomized, placebo-controlled trial. Complement. Ther. Med. 2022, 66, 102819. [Google Scholar] [CrossRef] [PubMed]
  309. Wei, R.M.; Zhang, Y.M.; Feng, Y.Z.; Zhang, K.X.; Zhang, J.Y.; Chen, J.; Luo, B.L.; Li, X.Y.; Chen, G.H. Resveratrol ameliorates maternal separation-induced anxiety- and depression-like behaviors and reduces Sirt1-NF-kB signaling-mediated neuroinflammation. Front. Behav. Neurosci. 2023, 17, 1172091. [Google Scholar] [CrossRef]
  310. Martínez-Flórez, S.; Gutiérrez-Fernández, B.; Sánchez-Campos, S.; González-Gallego, J.; Tuñón, M.J. Quercetin attenuates nuclear factor-kappaB activation and nitric oxide production in interleukin-1beta-activated rat hepatocytes. J. Nutr. 2005, 135, 1359–1365. [Google Scholar] [CrossRef]
  311. Ortega, M.G.; Saragusti, A.C.; Cabrera, J.L.; Chiabrando, G.A. Quercetin tetraacetyl derivative inhibits LPS-induced nitric oxide synthase (iNOS) expression in J774A.1 cells. Arch. Biochem. Biophys. 2010, 498, 105–110. [Google Scholar] [CrossRef]
  312. Wu, H.; Chen, L.; Zhu, F.; Han, X.; Sun, L.; Chen, K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins 2019, 11, 731. [Google Scholar] [CrossRef]
  313. Li, J.; Fan, Y.; Zhang, Y.; Liu, Y.; Yu, Y.; Ma, M. Resveratrol Induces Autophagy and Apoptosis in Non-Small-Cell Lung Cancer Cells by Activating the NGFR-AMPK-mTOR Pathway. Nutrients 2022, 14, 2413. [Google Scholar] [CrossRef] [PubMed]
  314. Iban-Arias, R.; Sebastian-Valverde, M.; Wu, H.; Lyu, W.; Wu, Q.; Simon, J.; Pasinetti, G.M. Role of Polyphenol-Derived Phenolic Acid in Mitigation of Inflammasome-Mediated Anxiety and Depression. Biomedicines 2022, 10, 1264. [Google Scholar] [CrossRef]
  315. Ye, S.; Fang, L.; Xie, S.; Hu, Y.; Chen, S.; Amin, N.; Fang, M.; Hu, Z. Resveratrol alleviates postpartum depression-like behavior by activating autophagy via SIRT1 and inhibiting AKT/mTOR pathway. Behav. Brain Res. 2023, 438, 114208. [Google Scholar] [CrossRef]
  316. Wong, R.H.; Thaung Zaw, J.J.; Xian, C.J.; Howe, P.R. Regular Supplementation with Resveratrol Improves Bone Mineral Density in Postmenopausal Women: A Randomized, Placebo-Controlled Trial. J. Bone Min. Res. 2020, 35, 2121–2131. [Google Scholar] [CrossRef]
  317. Inchingolo, A.D.; Malcangi, G.; Inchingolo, A.M.; Piras, F.; Settanni, V.; Garofoli, G.; Palmieri, G.; Ceci, S.; Patano, A.; De Leonardis, N.; et al. Benefits and Implications of Resveratrol Supplementation on Microbiota Modulations: A Systematic Review of the Literature. Int. J. Mol. Sci. 2022, 23, 4027. [Google Scholar] [CrossRef]
  318. Avellone, G.; Di Garbo, V.; Campisi, D.; De Simone, R.; Raneli, G.; Scaglione, R.; Licata, G. Effects of moderate Sicilian red wine consumption on inflammatory biomarkers of atherosclerosis. Eur. J. Clin. Nutr. 2006, 60, 41–47. [Google Scholar] [CrossRef]
  319. Loke, W.M.; Hodgson, J.M.; Proudfoot, J.M.; McKinley, A.J.; Puddey, I.B.; Croft, K.D. Pure dietary flavonoids quercetin and (-)-epicatechin augment nitric oxide products and reduce endothelin-1 acutely in healthy men. Am. J. Clin. Nutr. 2008, 88, 1018–1025. [Google Scholar] [CrossRef]
  320. Huang, Y.; Zhu, X.; Chen, K.; Lang, H.; Zhang, Y.; Hou, P.; Ran, L.; Zhou, M.; Zheng, J.; Yi, L.; et al. Resveratrol prevents sarcopenic obesity by reversing mitochondrial dysfunction and oxidative stress via the PKA/LKB1/AMPK pathway. Aging 2019, 11, 2217–2240. [Google Scholar] [CrossRef]
  321. Su, L.; Zeng, Y.; Li, G.; Chen, J.; Chen, X. Quercetin improves high-fat diet-induced obesity by modulating gut microbiota and metabolites in C57BL/6J mice. Phytother. Res. 2022, 36, 4558–4572. [Google Scholar] [CrossRef] [PubMed]
  322. Moreno-Indias, I.; Sánchez-Alcoholado, L.; Pérez-Martínez, P.; Andrés-Lacueva, C.; Cardona, F.; Tinahones, F.; Queipo-Ortuño, M.I. Red wine polyphenols modulate fecal microbiota and reduce markers of the metabolic syndrome in obese patients. Food Funct. 2016, 7, 1775–1787. [Google Scholar] [CrossRef] [PubMed]
  323. Zhu, F.; Zheng, J.; Xu, F.; Xi, Y.; Chen, J.; Xu, X. Resveratrol Alleviates Dextran Sulfate Sodium-Induced Acute Ulcerative Colitis in Mice by Mediating PI3K/Akt/VEGFA Pathway. Front. Pharmacol. 2021, 12, 693982. [Google Scholar] [CrossRef]
  324. Sabzevary-Ghahfarokhi, M.; Soltani, A.; Luzza, F.; Larussa, T.; Rahimian, G.; Shirzad, H.; Bagheri, N. The protective effects of resveratrol on ulcerative colitis via changing the profile of Nrf2 and IL-1β protein. Mol. Biol. Rep. 2020, 47, 6941–6947. [Google Scholar] [CrossRef] [PubMed]
  325. Yao, J.; Wei, C.; Wang, J.Y.; Zhang, R.; Li, Y.X.; Wang, L.S. Effect of resveratrol on Treg/Th17 signaling and ulcerative colitis treatment in mice. World J. Gastroenterol. 2015, 21, 6572–6581. [Google Scholar] [CrossRef]
  326. Chitimus, D.M.; Popescu, M.R.; Voiculescu, S.E.; Panaitescu, A.M.; Pavel, B.; Zagrean, L.; Zagrean, A.M. Melatonin’s Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules 2020, 10, 1211. [Google Scholar] [CrossRef]
  327. Tu, Y.; Song, E.; Wang, Z.; Ji, N.; Zhu, L.; Wang, K.; Sun, H.; Zhang, Y.; Zhu, Q.; Liu, X.; et al. Melatonin attenuates oxidative stress and inflammation of Müller cells in diabetic retinopathy via activating the Sirt1 pathway. Biomed. Pharmacother. 2021, 137, 111274. [Google Scholar] [CrossRef]
  328. Rehman, S.U.; Ikram, M.; Ullah, N.; Alam, S.I.; Park, H.Y.; Badshah, H.; Choe, K.; Kim, M.O. Neurological Enhancement Effects of Melatonin against Brain Injury-Induced Oxidative Stress, Neuroinflammation, and Neurodegeneration via AMPK/CREB Signaling. Cells 2019, 8, 760. [Google Scholar] [CrossRef]
  329. Kang, J.Y.; Xu, M.M.; Sun, Y.; Ding, Z.X.; Wei, Y.Y.; Zhang, D.W.; Wang, Y.G.; Shen, J.L.; Wu, H.M.; Fei, G.H. Melatonin attenuates LPS-induced pyroptosis in acute lung injury by inhibiting NLRP3-GSDMD pathway via activating Nrf2/HO-1 signaling axis. Int. Immunopharmacol. 2022, 109, 108782. [Google Scholar] [CrossRef]
  330. de Pablos, R.M.; Espinosa-Oliva, A.M.; Hornedo-Ortega, R.; Cano, M.; Arguelles, S. Hydroxytyrosol protects from aging process via AMPK and autophagy; a review of its effects on cancer, metabolic syndrome, osteoporosis, immune-mediated and neurodegenerative diseases. Pharmacol. Res. 2019, 143, 58–72. [Google Scholar] [CrossRef]
  331. Karković Marković, A.; Torić, J.; Barbarić, M.; Jakobušić Brala, C. Hydroxytyrosol, Tyrosol and Derivatives and Their Potential Effects on Human Health. Molecules 2019, 24, 2001. [Google Scholar] [CrossRef]
  332. D’Adamo, S.; Cetrullo, S.; Guidotti, S.; Borzì, R.M.; Flamigni, F. Hydroxytyrosol modulates the levels of microRNA-9 and its target sirtuin-1 thereby counteracting oxidative stress-induced chondrocyte death. Osteoarthr. Cartil. 2017, 25, 600–610. [Google Scholar] [CrossRef]
  333. Wang, W.; Jing, T.; Yang, X.; He, Y.; Wang, B.; Xiao, Y.; Shang, C.; Zhang, J.; Lin, R. Hydroxytyrosol regulates the autophagy of vascular adventitial fibroblasts through the SIRT1-mediated signaling pathway. Can. J. Physiol. Pharmacol. 2018, 96, 88–96. [Google Scholar] [CrossRef] [PubMed]
  334. Bolarinwa, I.; Al-Ezzi, M.; Carew, I.; Muhammad, K. Nutritional Value of Legumes in Relation to Human Health: A Review. Adv. J. Food Sci. Technol. 2019, 17, 72–85. [Google Scholar] [CrossRef]
  335. Cakir, Ö.; Ucarli, C.; TARHAN, Ç.; Pekmez, M.; Turgut-Kara, N. Nutritional and health benefits of legumes and their distinctive genomic properties. Food Sci. Technol. 2019, 39, 1–12. [Google Scholar]
  336. Juárez-Chairez, M.F.; Cid-Gallegos, M.S.; Meza-Márquez, O.G.; Jiménez-Martínez, C. Biological functions of peptides from legumes in gastrointestinal health. A review legume peptides with gastrointestinal protection. J. Food Biochem. 2022, 46, e14308. [Google Scholar] [CrossRef]
  337. Schuster-Gajzágó, I. Nutritional aspects of legumes. In Cultivated Plants, Primarily as Food Sources; Encyclopedia of Food and Agricultural Sciences, Engineering and Technology Resources; Encyclopedia of Life Support System (EOLSS): Abu Dhabi, United Arab Emirates, 2004; Volume 1, pp. 1–7. [Google Scholar]
  338. Kubota, M.; Shimizu, H. Nutrition and bone health. Soybean and soy foods, and bone health. Clin. Calcium 2009, 19, 1514–1519. [Google Scholar] [PubMed]
  339. Garrido-Galand, S.; Asensio-Grau, A.; Calvo-Lerma, J.; Heredia, A.; Andrés, A. The potential of fermentation on nutritional and technological improvement of cereal and legume flours: A review. Food Res. Int. 2021, 145, 110398. [Google Scholar] [CrossRef] [PubMed]
  340. Qiao, Y.; Zhang, K.; Zhang, Z.; Zhang, C.; Sun, Y.; Feng, Z. Fermented soybean foods: A review of their functional components, mechanism of action and factors influencing their health benefits. Food Res. Int. 2022, 158, 111575. [Google Scholar] [CrossRef] [PubMed]
  341. Liu, L.; Chen, X.; Hao, L.; Zhang, G.; Jin, Z.; Li, C.; Yang, Y.; Rao, J.; Chen, B. Traditional fermented soybean products: Processing, flavor formation, nutritional and biological activities. Crit. Rev. Food Sci. Nutr. 2022, 62, 1971–1989. [Google Scholar] [CrossRef] [PubMed]
  342. Kim, I.S.; Hwang, C.W.; Yang, W.S.; Kim, C.H. Current Perspectives on the Physiological Activities of Fermented Soybean-Derived Cheonggukjang. Int. J. Mol. Sci. 2021, 22, 5746. [Google Scholar] [CrossRef]
  343. Kaufman, P.B.; Duke, J.A.; Brielmann, H.; Boik, J.; Hoyt, J.E. A comparative survey of leguminous plants as sources of the isoflavones, genistein and daidzein: Implications for human nutrition and health. J. Altern. Complement. Med. 1997, 3, 7–12. [Google Scholar] [CrossRef]
  344. Cichońska, P.; Ziarno, M. Legumes and Legume-Based Beverages Fermented with Lactic Acid Bacteria as a Potential Carrier of Probiotics and Prebiotics. Microorganisms 2021, 10, 91. [Google Scholar] [CrossRef]
  345. Takagi, A.; Kano, M.; Kaga, C. Possibility of breast cancer prevention: Use of soy isoflavones and fermented soy beverage produced using probiotics. Int. J. Mol. Sci. 2015, 16, 10907–10920. [Google Scholar] [CrossRef]
  346. Kimura, K.; Yokoyama, S. Trends in the application of Bacillus in fermented foods. Curr. Opin. Biotechnol. 2019, 56, 36–42. [Google Scholar] [CrossRef]
  347. Cao, Z.H.; Green-Johnson, J.M.; Buckley, N.D.; Lin, Q.Y. Bioactivity of soy-based fermented foods: A review. Biotechnol. Adv. 2019, 37, 223–238. [Google Scholar] [CrossRef]
  348. Allwood, J.G.; Wakeling, L.T.; Bean, D.C. Fermentation and the microbial community of Japanese koji and miso: A review. J. Food Sci. 2021, 86, 2194–2207. [Google Scholar] [CrossRef] [PubMed]
  349. Jeong, D.M.; Kim, H.J.; Jeon, M.S.; Yoo, S.J.; Moon, H.Y.; Jeon, E.J.; Jeon, C.O.; Eyun, S.I.; Kang, H.A. Genomic and functional features of yeast species in Korean traditional fermented alcoholic beverage and soybean products. FEMS Yeast Res. 2023, 23, foac066. [Google Scholar] [CrossRef]
  350. Owusu-Kwarteng, J.; Parkouda, C.; Adewumi, G.A.; Ouoba, L.I.I.; Jespersen, L. Technologically relevant Bacillus species and microbial safety of West African traditional alkaline fermented seed condiments. Crit. Rev. Food Sci. Nutr. 2022, 62, 871–888. [Google Scholar] [CrossRef] [PubMed]
  351. Ogueke, C.C.; Nwosu, J.N.; Owuamanam, C.I.; Iwouno, J.N. Ugba, the fermented African oilbean seeds; its production, chemical composition, preservation, safety and health benefits. Pak. J. Biol. Sci. PJBS 2010, 13, 489–496. [Google Scholar] [CrossRef] [PubMed]
  352. Labba, I.M.; Andlid, T.; Lindgren, Å.; Sandberg, A.S.; Sjöberg, F. Isolation, identification, and selection of strains as candidate probiotics and starters for fermentation of Swedish legumes. Food Nutr. Res. 2020, 64, 1–13. [Google Scholar] [CrossRef]
  353. Jayachandran, M.; Xu, B. An insight into the health benefits of fermented soy products. Food Chem. 2019, 271, 362–371. [Google Scholar] [CrossRef]
  354. Nikmaram, N.; Dar, B.N.; Roohinejad, S.; Koubaa, M.; Barba, F.J.; Greiner, R.; Johnson, S.K. Recent advances in γ-aminobutyric acid (GABA) properties in pulses: An overview. J. Sci. Food Agric. 2017, 97, 2681–2689. [Google Scholar] [CrossRef]
  355. Das, G.; Paramithiotis, S.; Sundaram Sivamaruthi, B.; Wijaya, C.H.; Suharta, S.; Sanlier, N.; Shin, H.S.; Patra, J.K. Traditional fermented foods with anti-aging effect: A concentric review. Food Res. Int. 2020, 134, 109269. [Google Scholar] [CrossRef]
  356. Belobrajdic, D.P.; James-Martin, G.; Jones, D.; Tran, C.D. Soy and Gastrointestinal Health: A Review. Nutrients 2023, 15, 1959. [Google Scholar] [CrossRef] [PubMed]
  357. Das, D.; Sarkar, S.; Borsingh Wann, S.; Kalita, J.; Manna, P. Current perspectives on the anti-inflammatory potential of fermented soy foods. Food Res. Int. 2022, 152, 110922. [Google Scholar] [CrossRef] [PubMed]
  358. Hu, K.; Huang, H.; Li, H.; Wei, Y.; Yao, C. Legume-Derived Bioactive Peptides in Type 2 Diabetes: Opportunities and Challenges. Nutrients 2023, 15, 1096. [Google Scholar] [CrossRef] [PubMed]
  359. Das, D.; Sarkar, S.; Bordoloi, J.; Wann, S.B.; Kalita, J.; Manna, P. Daidzein, its effects on impaired glucose and lipid metabolism and vascular inflammation associated with type 2 diabetes. BioFactors 2018, 44, 407–417. [Google Scholar] [CrossRef] [PubMed]
  360. Monk, J.M.; Zhang, C.P.; Wu, W.; Zarepoor, L.; Lu, J.T.; Liu, R.; Pauls, K.P.; Wood, G.A.; Tsao, R.; Robinson, L.E.; et al. White and dark kidney beans reduce colonic mucosal damage and inflammation in response to dextran sodium sulfate. J. Nutr. Biochem. 2015, 26, 752–760. [Google Scholar] [CrossRef] [PubMed]
  361. Pang, W.; Wang, D.; Zuo, Z.; Wang, Y.; Sun, W.; Zhang, N.; Zhang, D. Kidney Bean Fermented Broth Alleviates Hyperlipidemic by Regulating Serum Metabolites and Gut Microbiota Composition. Nutrients 2022, 14, 3202. [Google Scholar] [CrossRef]
  362. Georgetti, S.R.; Casagrande, R.; Vicentini, F.T.; Baracat, M.M.; Verri, W.A., Jr.; Fonseca, M.J. Protective effect of fermented soybean dried extracts against TPA-induced oxidative stress in hairless mice skin. BioMed Res. Int. 2013, 2013, 340626. [Google Scholar] [CrossRef]
  363. Yang, J.H.; Byeon, E.H.; Kang, D.; Hong, S.G.; Yang, J.; Kim, D.R.; Yun, S.P.; Park, S.W.; Kim, H.J.; Huh, J.W.; et al. Fermented Soybean Paste Attenuates Biogenic Amine-Induced Liver Damage in Obese Mice. Cells 2023, 12, 822. [Google Scholar] [CrossRef]
  364. Frias, J.; Song, Y.S.; Martínez-Villaluenga, C.; González de Mejia, E.; Vidal-Valverde, C. Immunoreactivity and amino acid content of fermented soybean products. J. Agric. Food Chem. 2008, 56, 99–105. [Google Scholar] [CrossRef]
  365. Ali, N.M.; Yeap, S.K.; Yusof, H.M.; Beh, B.K.; Ho, W.Y.; Koh, S.P.; Abdullah, M.P.; Alitheen, N.B.; Long, K. Comparison of free amino acids, antioxidants, soluble phenolic acids, cytotoxicity and immunomodulation of fermented mung bean and soybean. J. Sci. Food Agric. 2016, 96, 1648–1658. [Google Scholar] [CrossRef]
  366. Das, D.; Sarkar, S.; Dihingia, A.; Afzal, N.U.; Wann, S.B.; Kalita, J.; Dewanjee, S.; Manna, P. A popular fermented soybean food of Northeast India exerted promising antihyperglycemic potential via stimulating PI3K/AKT/AMPK/GLUT4 signaling pathways and regulating muscle glucose metabolism in type 2 diabetes. J. Food Biochem. 2022, 46, e14385. [Google Scholar] [CrossRef]
  367. Sapbamrer, R.; Visavarungroj, N.; Suttajit, M. Effects of dietary traditional fermented soybean on reproductive hormones, lipids, and glucose among postmenopausal women in northern Thailand. Asia Pac. J. Clin. Nutr. 2013, 22, 222–228. [Google Scholar] [CrossRef]
  368. Rim, H.K.; Kim, K.Y.; Ryu, J.G.; Song, Y.H.; Kim, H.H.; Han, J.H.; Jeong, H.J.; Kim, H.M. Alcohol-fermented soybean increases the expression of receptor-interacting protein 2 and IκB kinase β in mouse peritoneal macrophages. J. Med. Food 2011, 14, 1181–1189. [Google Scholar] [CrossRef]
  369. Chen, K.; Luo, H.; Li, Y.; Han, X.; Gao, C.; Wang, N.; Lu, F.; Wang, H. Lactobacillus paracasei TK1501 fermented soybeans alleviate dextran sulfate sodium-induced colitis by regulating intestinal cell function. J. Sci. Food Agric. 2023, 103, 5422–5431. [Google Scholar] [CrossRef] [PubMed]
  370. Bhatt, P.C.; Pathak, S.; Kumar, V.; Panda, B.P. Attenuation of neurobehavioral and neurochemical abnormalities in animal model of cognitive deficits of Alzheimer’s disease by fermented soybean nanonutraceutical. Inflammopharmacology 2018, 26, 105–118. [Google Scholar] [CrossRef] [PubMed]
  371. Koh, Y.C.; Kuo, L.H.; Chang, Y.Y.; Tung, Y.C.; Lo, Y.C.; Pan, M.H. Modulatory Effect of Fermented Black Soybean and Adlay on Gut Microbiota Contributes to Healthy Aging. Mol. Nutr. Food Res. 2023, 67, e2200700. [Google Scholar] [CrossRef] [PubMed]
  372. Lin, C.C.; Wu, P.S.; Liang, D.W.; Kwan, C.C.; Chen, Y.S. Quality, antioxidative ability, and cell proliferation-enhancing activity of fermented black soybean broths with various supplemental culture medium. J. Food Sci. 2012, 77, C95–C101. [Google Scholar] [CrossRef]
  373. Malardé, L.; Vincent, S.; Lefeuvre-Orfila, L.; Efstathiou, T.; Groussard, C.; Gratas-Delamarche, A. A fermented soy permeate improves the skeletal muscle glucose level without restoring the glycogen content in streptozotocin-induced diabetic rats. J. Med. Food 2013, 16, 176–179. [Google Scholar] [CrossRef] [PubMed]
  374. Kim, H.B.; Lee, H.S.; Kim, S.J.; Yoo, H.J.; Hwang, J.S.; Chen, G.; Youn, H.J. Ethanol extract of fermented soybean, Chungkookjang, inhibits the apoptosis of mouse spleen, and thymus cells. J. Microbiol. 2007, 45, 256–261. [Google Scholar]
  375. Lee, D.H.; Kim, M.J.; Ahn, J.; Lee, S.H.; Lee, H.; Kim, J.H.; Park, S.H.; Jang, Y.J.; Ha, T.Y.; Jung, C.H. Nutrikinetics of Isoflavone Metabolites After Fermented Soybean Product (Cheonggukjang) Ingestion in Ovariectomized Mice. Mol. Nutr. Food Res. 2017, 61, 1700322. [Google Scholar] [CrossRef]
  376. Cho, B.O.; Shin, J.Y.; Kim, J.S.; Che, D.N.; Kang, H.J.; Jeong, D.Y.; Jang, S.I. Soybean Fermented with Bacillus amyloliquefaciens (Cheonggukjang) Ameliorates Atopic Dermatitis-Like Skin Lesion in Mice by Suppressing Infiltration of Mast Cells and Production of IL-31 Cytokine. J. Microbiol. Biotechnol. 2019, 29, 827–837. [Google Scholar] [CrossRef]
  377. Choi, J.; Kwon, S.H.; Park, K.Y.; Yu, B.P.; Kim, N.D.; Jung, J.H.; Chung, H.Y. The anti-inflammatory action of fermented soybean products in kidney of high-fat-fed rats. J. Med. Food 2011, 14, 232–239. [Google Scholar] [CrossRef]
  378. Lee, J.H.; Paek, S.H.; Shin, H.W.; Lee, S.Y.; Moon, B.S.; Park, J.E.; Lim, G.D.; Kim, C.Y.; Heo, Y. Effect of fermented soybean products intake on the overall immune safety and function in mice. J. Vet. Sci. 2017, 18, 25–32. [Google Scholar] [CrossRef] [PubMed]
  379. Jang, S.E.; Kim, K.A.; Han, M.J.; Kim, D.H. Doenjang, a fermented Korean soybean paste, inhibits lipopolysaccharide production of gut microbiota in mice. J. Med. Food 2014, 17, 67–75. [Google Scholar] [CrossRef]
  380. Sumi, H.; Hamada, H.; Tsushima, H.; Mihara, H.; Muraki, H. A novel fibrinolytic enzyme (nattokinase) in the vegetable cheese Natto; a typical and popular soybean food in the Japanese diet. Experientia 1987, 43, 1110–1111. [Google Scholar] [CrossRef] [PubMed]
  381. Oba, M.; Rongduo, W.; Saito, A.; Okabayashi, T.; Yokota, T.; Yasuoka, J.; Sato, Y.; Nishifuji, K.; Wake, H.; Nibu, Y.; et al. Natto extract, a Japanese fermented soybean food, directly inhibits viral infections including SARS-CoV-2 in vitro. Biochem. Biophys. Res. Commun. 2021, 570, 21–25. [Google Scholar] [CrossRef] [PubMed]
  382. Uenishi, K. Recommended soy and soy products intake to prevent bone fracture and osteoporosis. Clin. Calcium 2005, 15, 1393–1398. [Google Scholar]
  383. Katsuyama, H.; Ideguchi, S.; Fukunaga, M.; Saijoh, K.; Sunami, S. Usual dietary intake of fermented soybeans (Natto) is associated with bone mineral density in premenopausal women. J. Nutr. Sci. Vitaminol. 2002, 48, 207–215. [Google Scholar] [CrossRef]
  384. Murai, U.; Sawada, N.; Charvat, H.; Inoue, M.; Yasuda, N.; Yamagishi, K.; Tsugane, S. Soy product intake and risk of incident disabling dementia: The JPHC Disabling Dementia Study. Eur. J. Nutr. 2022, 61, 4045–4057. [Google Scholar] [CrossRef]
  385. Sasaki, H.; Pham Thi Ngoc, D.; Nishikawa, M.; Kanauchi, M. Lipopolysaccharide neutralizing protein in Miso, Japanese fermented soybean paste. J. Food Sci. 2020, 85, 2498–2505. [Google Scholar] [CrossRef]
  386. Matsuo, M. Chemical components, palatability, antioxidant activity and antimutagenicity of oncom miso using a mixture of fermented soybeans and okara with Neurospora intermedia. J. Nutr. Sci. Vitaminol. 2006, 52, 216–222. [Google Scholar] [CrossRef]
  387. Nagata, C.; Shimizu, H.; Takami, R.; Hayashi, M.; Takeda, N.; Yasuda, K. Hot flushes and other menopausal symptoms in relation to soy product intake in Japanese women. Climacteric 1999, 2, 6–12. [Google Scholar] [CrossRef]
  388. Uemura, H.; Katsuura-Kamano, S.; Nakamoto, M.; Yamaguchi, M.; Fujioka, M.; Iwasaki, Y.; Arisawa, K. Inverse association between soy food consumption, especially fermented soy products intake and soy isoflavone, and arterial stiffness in Japanese men. Sci. Rep. 2018, 8, 9667. [Google Scholar] [CrossRef]
  389. Nozue, M.; Shimazu, T.; Sasazuki, S.; Charvat, H.; Mori, N.; Mutoh, M.; Sawada, N.; Iwasaki, M.; Yamaji, T.; Inoue, M.; et al. Fermented Soy Product Intake Is Inversely Associated with the Development of High Blood Pressure: The Japan Public Health Center-Based Prospective Study. J. Nutr. 2017, 147, 1749–1756. [Google Scholar] [CrossRef] [PubMed]
  390. Matsuo, M. Low-salt O-miso produced from Koji fermentation of oncom improves redox state and cholesterolemia in rats more than low-salt soybean-miso. J. Nutr. Sci. Vitaminol. 2004, 50, 362–366. [Google Scholar] [CrossRef]
  391. Park, S.; Lee, J.J.; Shin, H.W.; Jung, S.; Ha, J.H. Effect of Soybean and Soybean Koji on Obesity and Dyslipidemia in Rats Fed a High-Fat Diet: A Comparative Study. Int. J. Environ. Res. Public Health 2021, 18, 6032. [Google Scholar] [CrossRef] [PubMed]
  392. Kim, D.H.; Kim, S.A.; Jo, Y.M.; Seo, H.; Kim, G.Y.; Cheon, S.W.; Yang, S.H.; Jeon, C.O.; Han, N.S. Probiotic potential of Tetragenococcus halophilus EFEL7002 isolated from Korean soy Meju. BMC Microbiol. 2022, 22, 149. [Google Scholar] [CrossRef]
  393. Kulprachakarn, K.; Chaipoot, S.; Phongphisutthinant, R.; Paradee, N.; Prommaban, A.; Ounjaijean, S.; Rerkasem, K.; Parklak, W.; Prakit, K.; Saengsitthisak, B.; et al. Antioxidant Potential and Cytotoxic Effect of Isoflavones Extract from Thai Fermented Soybean (Thua-Nao). Molecules 2021, 26, 7432. [Google Scholar] [CrossRef]
  394. Arumugam, S.; Dioletis, E.; Paiva, R.; Fields, M.R.; Weiss, T.R.; Secor, E.R.; Ali, A. Fermented Soy Beverage Q-CAN Plus Consumption Improves Serum Cholesterol and Cytokines. J. Med. Food 2020, 23, 560–563. [Google Scholar] [CrossRef]
  395. Lin, C.Y.; Tsai, Z.Y.; Cheng, I.C.; Lin, S.H. Effects of fermented soy milk on the liver lipids under oxidative stress. World J. Gastroenterol. 2005, 11, 7355–7358. [Google Scholar] [CrossRef] [PubMed]
  396. Biscola, V.; de Olmos, A.R.; Choiset, Y.; Rabesona, H.; Garro, M.S.; Mozzi, F.; Chobert, J.M.; Drouet, M.; Haertlé, T.; Franco, B. Soymilk fermentation by Enterococcus faecalis VB43 leads to reduction in the immunoreactivity of allergenic proteins β-conglycinin (7S) and glycinin (11S). Benef. Microbes 2017, 8, 635–643. [Google Scholar] [CrossRef]
  397. Hwang, J.H.; Wu, S.J.; Wu, P.L.; Shih, Y.Y.; Chan, Y.C. Neuroprotective effect of tempeh against lipopolysaccharide-induced damage in BV-2 microglial cells. Nutr. Neurosci. 2019, 22, 840–849. [Google Scholar] [CrossRef]
  398. McKevith, B. Nutritional aspects of cereals. Nutr. Bull. 2004, 29, 111–142. [Google Scholar] [CrossRef]
  399. Mishra, S.; Mithul Aravind, S.; Charpe, P.; Ajlouni, S.; Ranadheera, C.S.; Chakkaravarthi, S. Traditional rice-based fermented products: Insight into their probiotic diversity and probable health benefits. Food Biosci. 2022, 50, 102082. [Google Scholar] [CrossRef]
  400. Patra, M.; Bashir, O.; Amin, T.; Wani, A.W.; Shams, R.; Chaudhary, K.S.; Mirza, A.A.; Manzoor, S. A comprehensive review on functional beverages from cereal grains-characterization of nutraceutical potential, processing technologies and product types. Heliyon 2023, 9, e16804. [Google Scholar] [CrossRef]
  401. Tsafrakidou, P.; Michaelidou, A.M.; Biliaderis, C.G. Fermented Cereal-based Products: Nutritional Aspects, Possible Impact on Gut Microbiota and Health Implications. Foods 2020, 9, 734. [Google Scholar] [CrossRef]
  402. Goksen, G.; Sugra Altaf, Q.; Farooq, S.; Bashir, I.; Cappozzi, V.; Guruk, M.; Lucia Bavaro, S.; Kumar Sarangi, P. A Glimpse into Plant-based Fermented Products Alternative to Animal Based Products: Formulation, Processing, Health Benefits. Food Res. Int. 2023, 173, 113344. [Google Scholar] [CrossRef]
  403. Hlangwani, E.; Njobeh, P.B.; Chinma, C.E.; Oyedeji, A.B.; Fasogbon, B.M.; Oyeyinka, S.A.; Sobowale, S.S.; Dudu, O.E.; Molelekoa, T.B.J.; Kesa, H.; et al. Chapter 2—African cereal-based fermented products. In Indigenous Fermented Foods for the Tropics; Adebo, O.A., Chinma, C.E., Obadina, A.O., Soares, A.G., Panda, S.K., Gan, R.-Y., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 15–36. [Google Scholar]
  404. Pswarayi, F.; Gänzle, M. African cereal fermentations: A review on fermentation processes and microbial composition of non-alcoholic fermented cereal foods and beverages. Int. J. Food Microbiol. 2022, 378, 109815. [Google Scholar] [CrossRef]
  405. Zannou, O.; Agossou, D.J.; Miassi, Y.; Agani, O.B.; Darino Aisso, M.; Chabi, I.B.; Euloge Kpoclou, Y.; Azokpota, P.; Koca, I. Traditional fermented foods and beverages: Indigenous practices of food processing in Benin Republic. Int. J. Gastron. Food Sci. 2022, 27, 100450. [Google Scholar] [CrossRef]
  406. Kütt, M.-L.; Orgusaar, K.; Stulova, I.; Priidik, R.; Pismennõi, D.; Vaikma, H.; Kallastu, A.; Zhogoleva, A.; Morell, I.; Kriščiunaite, T. Starter culture growth dynamics and sensory properties of fermented oat drink. Heliyon 2023, 9, e15627. [Google Scholar] [CrossRef] [PubMed]
  407. Ramos, P.I.K.; Tuaño, A.P.P.; Juanico, C.B. Microbial quality, safety, sensory acceptability, and proximate composition of a fermented nixtamalized maize (Zea mays L.) beverage. J. Cereal Sci. 2022, 107, 103521. [Google Scholar] [CrossRef]
  408. Rebaza-Cardenas, T.; Montes-Villanueva, N.D.; Fernández, M.; Delgado, S.; Ruas-Madiedo, P. Microbiological and physical-chemical characteristics of the Peruvian fermented beverage “Chicha de siete semillas”: Towards the selection of strains with acidifying properties. Int. J. Food Microbiol. 2023, 406, 110353. [Google Scholar] [CrossRef]
  409. Oyeyinka, A.T.; Siwela, M.; Pillay, K. A mini review of the physicochemical properties of amahewu, a Southern African traditional fermented cereal grain beverage. LWT 2021, 151, 112159. [Google Scholar] [CrossRef]
  410. Bhattacharjee, S.; Sarkar, I.; Sen, G.; Ghosh, C.; Sen, A. Biochemical and Metagenomic sketching of microbial populations in the starter culture of ‘Chokot’, a rice-based fermented liquor of Rabha Tribe in North Bengal, India. Ecol. Genet. Genom. 2023, 29, 100193. [Google Scholar] [CrossRef]
  411. Osimani, A.; Garofalo, C.; Aquilanti, L.; Milanović, V.; Clementi, F. Unpasteurised commercial boza as a source of microbial diversity. Int. J. Food Microbiol. 2015, 194, 62–70. [Google Scholar] [CrossRef]
  412. Yeğin, S.; Üren, A. Biogenic amine content of boza: A traditional cereal-based, fermented Turkish beverage. Food Chem. 2008, 111, 983–987. [Google Scholar] [CrossRef]
  413. Zhang, J.; Liu, M.; Zhao, Y.; Zhu, Y.; Bai, J.; Fan, S.; Zhu, L.; Song, C.; Xiao, X. Recent Developments in Fermented Cereals on Nutritional Constituents and Potential Health Benefits. Foods 2022, 11, 2243. [Google Scholar]
  414. Shangpliang, H.N.J.; Tamang, J.P. Metagenomics and metagenome-assembled genomes mining of health benefits in jalebi batter, a naturally fermented cereal-based food of India. Food Res. Int. 2023, 172, 113130. [Google Scholar] [CrossRef] [PubMed]
  415. Banwo, K.; Oyeyipo, A.; Mishra, L.; Sarkar, D.; Shetty, K. Improving phenolic bioactive-linked functional qualities of traditional cereal-based fermented food (Ogi) of Nigeria using compatible food synergies with underutilized edible plants. NFS J. 2022, 27, 1–12. [Google Scholar] [CrossRef]
  416. Gebre, T.S.; Emire, S.A.; Chelliah, R.; Aloo, S.O.; Oh, D.-H. Isolation, functional activity, and safety of probiotics from Ethiopian traditional cereal-based fermented beverage, “Borde”. LWT 2023, 184, 115076. [Google Scholar] [CrossRef]
  417. Ogunremi, O.R.; Freimüller Leischtfeld, S.; Mischler, S.; Miescher Schwenninger, S. Antifungal activity of lactic acid bacteria isolated from kunu-zaki, a cereal-based Nigerian fermented beverage. Food Biosci. 2022, 49, 101648. [Google Scholar] [CrossRef]
  418. Bayoï, J.R.; Ndegoue, S.V.; Etoa, F.-X. Traditional processing and quality attributes of “kounou”, a fermented indigenous cereal-based beverage from the northern zone of Cameroon. J. Agric. Food Res. 2021, 6, 100209. [Google Scholar] [CrossRef]
  419. Rasheed, H.A.; Tuoheti, T.; Zhang, Y.; Azi, F.; Tekliye, M.; Dong, M. Purification and partial characterization of a novel bacteriocin produced by bacteriocinogenic Lactobacillus fermentum BZ532 isolated from Chinese fermented cereal beverage (Bozai). LWT 2020, 124, 109113. [Google Scholar] [CrossRef]
  420. Wang, C.-Y.; Wu, S.-j.; Fang, J.-Y.; Wang, Y.-P.; Shyu, Y.-T. Cardiovascular and intestinal protection of cereal pastes fermented with lactic acid bacteria in hyperlipidemic hamsters. Food Res. Int. 2012, 48, 428–434. [Google Scholar] [CrossRef]
  421. Oguntoyinbo, F.A.; Narbad, A. Multifunctional properties of Lactobacillus plantarum strains isolated from fermented cereal foods. J. Funct. Foods 2015, 17, 621–631. [Google Scholar] [CrossRef]
  422. Ayyash, M.; Johnson, S.K.; Liu, S.-Q.; Al-Mheiri, A.; Abushelaibi, A. Cytotoxicity, antihypertensive, antidiabetic and antioxidant activities of solid-state fermented lupin, quinoa and wheat by Bifidobacterium species: In-vitro investigations. LWT 2018, 95, 295–302. [Google Scholar] [CrossRef]
  423. Zhu, C.; Guan, Q.; Song, C.; Zhong, L.; Ding, X.; Zeng, H.; Nie, P.; Song, L. Regulatory effects of Lactobacillus fermented black barley on intestinal microbiota of NAFLD rats. Food Res. Int. 2021, 147, 110467. [Google Scholar] [CrossRef] [PubMed]
  424. Sánchez-García, J.; Muñoz-Pina, S.; García-Hernández, J.; Heredia, A.; Andrés, A. Fermented quinoa flour: Implications of fungal solid-state bioprocessing and drying on nutritional and antioxidant properties. LWT 2023, 182, 114885. [Google Scholar] [CrossRef]
  425. Kingamkono, R.; Sjögren, E.; Svanberg, U. Enteropathogenic bacteria in faecal swabs of young children fed on lactic acid-fermented cereal gruels. Epidemiol. Infect. 1999, 122, 23–32. [Google Scholar] [CrossRef] [PubMed]
  426. Liu, Y.; Xue, K.; Iversen, K.N.; Qu, Z.; Dong, C.; Jin, T.; Hallmans, G.; Åman, P.; Johansson, A.; He, G.; et al. The effects of fermented rye products on gut microbiota and their association with metabolic factors in Chinese adults—An explorative study. Food Funct. 2021, 12, 9141–9150. [Google Scholar] [CrossRef]
  427. Ren, R.; Zeng, H.; Mei, Q.; Xu, Z.; Mazhar, M.; Qin, L. Effects of Monascus purpureus-fermented tartary buckwheat extract on the blood lipid profile, glucose tolerance and antioxidant enzyme activities in KM mice. J. Cereal Sci. 2022, 105, 103465. [Google Scholar] [CrossRef]
  428. Salar, R.K.; Purewal, S.S.; Sandhu, K.S. Fermented pearl millet (Pennisetum glaucum) with in vitro DNA damage protection activity, bioactive compounds and antioxidant potential. Food Res. Int. 2017, 100, 204–210. [Google Scholar] [CrossRef]
  429. Ofosu, F.K.; Elahi, F.; Daliri, E.B.-M.; Aloo, S.O.; Chelliah, R.; Han, S.-I.; Oh, D.-H. Fermented sorghum improves type 2 diabetes remission by modulating gut microbiota and their related metabolites in high fat diet-streptozotocin induced diabetic mice. J. Funct. Foods 2023, 107, 105666. [Google Scholar] [CrossRef]
  430. Li, L.; Wang, P.; Xu, X.; Zhou, G. Influence of various cooking methods on the concentrations of volatile N-nitrosamines and biogenic amines in dry-cured sausages. J. Food Sci. 2012, 77, C560–C565. [Google Scholar] [CrossRef]
  431. Drabik-Markiewicz, G.; Dejaegher, B.; De Mey, E.; Kowalska, T.; Paelinck, H.; Vander Heyden, Y. Influence of putrescine, cadaverine, spermidine or spermine on the formation of N-nitrosamine in heated cured pork meat. Food Chem. 2011, 126, 1539–1545. [Google Scholar] [CrossRef]
  432. Gushgari, A.J.; Halden, R.U. Critical review of major sources of human exposure to N-nitrosamines. Chemosphere 2018, 210, 1124–1136. [Google Scholar] [CrossRef] [PubMed]
  433. WHO. Iarc Monographs on the Identification of Carcinogenic Hazards to Humans; WHO: Geneva, Switzerland, 2023; pp. 1–33. [Google Scholar]
  434. Ahmad, W.; Mohammed, G.I.; Al-Eryani, D.A.; Saigl, Z.M.; Alyoubi, A.O.; Alwael, H.; Bashammakh, A.S.; O’Sullivan, C.K.; El-Shahawi, M.S. Biogenic Amines Formation Mechanism and Determination Strategies: Future Challenges and Limitations. Crit. Rev. Anal. Chem. 2020, 50, 485–500. [Google Scholar] [CrossRef] [PubMed]
  435. Doeun, D.; Davaatseren, M.; Chung, M.S. Biogenic amines in foods. Food Sci. Biotechnol. 2017, 26, 1463–1474. [Google Scholar] [CrossRef] [PubMed]
  436. Wójcik, W.; Łukasiewicz, M.; Puppel, K. Biogenic amines: Formation, action and toxicity—A review. J. Sci. Food Agric. 2021, 101, 2634–2640. [Google Scholar] [CrossRef]
  437. Jaguey-Hernández, Y.; Aguilar-Arteaga, K.; Ojeda-Ramirez, D.; Añorve-Morga, J.; González-Olivares, L.G.; Castañeda-Ovando, A. Biogenic amines levels in food processing: Efforts for their control in foodstuffs. Food Res. Int. 2021, 144, 110341. [Google Scholar] [CrossRef]
  438. Lee, Y.C.; Kung, H.F.; Huang, Y.L.; Wu, C.H.; Huang, Y.R.; Tsai, Y.H. Reduction of Biogenic Amines during Miso Fermentation by Lactobacillus plantarum as a Starter Culture. J. Food Prot. 2016, 79, 1556–1561. [Google Scholar] [CrossRef]
  439. Lee, Y.C.; Kung, H.F.; Huang, C.Y.; Huang, T.C.; Tsai, Y.H. Reduction of histamine and biogenic amines during salted fish fermentation by Bacillus polymyxa as a starter culture. J. Food Drug Anal. 2016, 24, 157–163. [Google Scholar] [CrossRef]
  440. Lin, X.; Tang, Y.; Hu, Y.; Lu, Y.; Sun, Q.; Lv, Y.; Zhang, Q.; Wu, C.; Zhu, M.; He, Q.; et al. Sodium Reduction in Traditional Fermented Foods: Challenges, Strategies, and Perspectives. J. Agric. Food Chem. 2021, 69, 8065–8080. [Google Scholar] [CrossRef]
  441. Bautista-Gallego, J.; Rantsiou, K.; Garrido-Fernández, A.; Cocolin, L.; Arroyo-López, F.N. Salt Reduction in Vegetable Fermentation: Reality or Desire? J. Food Sci. 2013, 78, R1095–R1100. [Google Scholar] [CrossRef] [PubMed]
  442. Laranjo, M.; Gomes, A.; Agulheiro-Santos, A.C.; Potes, M.E.; Cabrita, M.J.; Garcia, R.; Rocha, J.M.; Roseiro, L.C.; Fernandes, M.J.; Fraqueza, M.J.; et al. Impact of salt reduction on biogenic amines, fatty acids, microbiota, texture and sensory profile in traditional blood dry-cured sausages. Food Chem. 2017, 218, 129–136. [Google Scholar] [CrossRef]
  443. Dugat-Bony, E.; Sarthou, A.S.; Perello, M.C.; de Revel, G.; Bonnarme, P.; Helinck, S. The effect of reduced sodium chloride content on the microbiological and biochemical properties of a soft surface-ripened cheese. J. Dairy Sci. 2016, 99, 2502–2511. [Google Scholar] [CrossRef]
  444. Laranjo, M.; Gomes, A.; Agulheiro-Santos, A.C.; Potes, M.E.; Cabrita, M.J.; Garcia, R.; Rocha, J.M.; Roseiro, L.C.; Fernandes, M.J.; Fernandes, M.H.; et al. Characterisation of “Catalão” and “Salsichão” Portuguese traditional sausages with salt reduction. Meat Sci. 2016, 116, 34–42. [Google Scholar] [CrossRef] [PubMed]
  445. Zhou, Q.; Zang, S.; Zhao, Z.; Li, X. Dynamic changes of bacterial communities and nitrite character during northeastern Chinese sauerkraut fermentation. Food Sci. Biotechnol. 2018, 27, 79–85. [Google Scholar] [CrossRef] [PubMed]
  446. Shen, Q.; Zeng, X.; Kong, L.; Sun, X.; Shi, J.; Wu, Z.; Guo, Y.; Pan, D. Research Progress of Nitrite Metabolism in Fermented Meat Products. Foods 2023, 12, 1485. [Google Scholar]
  447. Li, F.; Zhuang, H.; Qiao, W.; Zhang, J.; Wang, Y. Effect of partial substitution of NaCl by KCl on physicochemical properties, biogenic amines and N-nitrosamines during ripening and storage of dry-cured bacon. J. Food Sci. Technol. 2016, 53, 3795–3805. [Google Scholar] [CrossRef]
Figure 1. Schematic summary of changes throughout meat fermentation.
Figure 1. Schematic summary of changes throughout meat fermentation.
Fermentation 09 00923 g001
Figure 2. Health effects of red wine components. Red line: suppressive effect, green line: increase, purple line: another effect, miRNA: micro RNA, Sirt 1: Sirtuin 1, NF-kB: nuclear factor kappa B, NGFR: nerve growth factor receptor, AMPK: AMP-activated protein kinase, mTOR: mammalian target of rapamycin, PKA: protein kinase A, LKB1: liver kinase B1, MyD88: myeloid differentiation protein 88, TLR4: Toll-like receptor 4, p-IkB-α: phospho-IkappaB-alpha, PI3K/Akt: phosphatidylinositol 3-kinase/protein kinase B, HIF-1α: hypoxia-inducible factor-1, Th17: T helper 17, HOMA-IR: homeostasis model assessment of insulin resistance, iNOS: inducible nitric oxide synthase, IFN-γ: interferon gamma, TNF-α: tumor necrosis factor alpha, IL: interleukin, CRP: C-reactive protein, NO: nitric oxide, LDL: low-density lipoprotein, HDL-C: high-density lipoprotein cholesterol, VEGFA: vascular endothelial growth factor A, LPS: lipopolysaccharides, GSH: glutathione, SOD: superoxide dismutase, CAT: catalase, TGF-β1: transforming growth factor beta, Apo A1: apoprotein A1, CVD: cardiovascular disease.
Figure 2. Health effects of red wine components. Red line: suppressive effect, green line: increase, purple line: another effect, miRNA: micro RNA, Sirt 1: Sirtuin 1, NF-kB: nuclear factor kappa B, NGFR: nerve growth factor receptor, AMPK: AMP-activated protein kinase, mTOR: mammalian target of rapamycin, PKA: protein kinase A, LKB1: liver kinase B1, MyD88: myeloid differentiation protein 88, TLR4: Toll-like receptor 4, p-IkB-α: phospho-IkappaB-alpha, PI3K/Akt: phosphatidylinositol 3-kinase/protein kinase B, HIF-1α: hypoxia-inducible factor-1, Th17: T helper 17, HOMA-IR: homeostasis model assessment of insulin resistance, iNOS: inducible nitric oxide synthase, IFN-γ: interferon gamma, TNF-α: tumor necrosis factor alpha, IL: interleukin, CRP: C-reactive protein, NO: nitric oxide, LDL: low-density lipoprotein, HDL-C: high-density lipoprotein cholesterol, VEGFA: vascular endothelial growth factor A, LPS: lipopolysaccharides, GSH: glutathione, SOD: superoxide dismutase, CAT: catalase, TGF-β1: transforming growth factor beta, Apo A1: apoprotein A1, CVD: cardiovascular disease.
Fermentation 09 00923 g002
Figure 3. Schematic summary of the effects of fermented foods on health.
Figure 3. Schematic summary of the effects of fermented foods on health.
Fermentation 09 00923 g003
Table 1. Possible health effects of kefir and its components.
Table 1. Possible health effects of kefir and its components.
Bioactive ComponentsHealth EffectsSpecific EffectsReferences
Kefir and kefir grainsAntihypertensiveACE inhibitory activity
Blood pressure
[34]
Mean arterial pressure
Cardiac hypertrophy
TNF-α/IL-10
ACE activity
[64]
AnticancerTGF-α downregulation and TGF-β1 mRNA expression upregulation has antiproliferative effects
Dose-dependent effects:
Transcriptional levels of TGF-α
Transcriptional levels of TGF-β1
Apoptotic cells
[35]
Expression of TGF-α TGF-β1
p53-independent p21 expression
Upregulation in Bax/Bcl-2 ratio
Kefir may induce apoptosis and inhibit proliferation
[65]
Tumor growth 64.8% [66]
The size and the amount of tumor
In colon tissue:
The mRNA expression levels of mRNA of TNF-α, IL-6, and IL-17a
TNF-α, IL-6, and IL-17a
Proliferating cell indicators (Ki67, NF-κB, β-Catenin)
Claudin 1, ZO-1 mRNA, and protein levels
Serum LPS
In feces:
Butyric acid, acetic acid, and propionic acid
Ascomycota/Basidiomycota ratio and Firmicutes/Bacteroidetes ratio
Lactobacillus and Bifidobacterium 
The relative abundance of probiotics
The pathogenic bacteria (Aspergillus, Clostridium sensu stricto, and Talaromyces)
[67]
AntioxidantTAS [36]
Serum levels of ·O2, H2O2, and ONOO/OH 
NO levels
Protein oxidation
p53 expression
DNA fragmentation
Apoptosis
[37]
MDA, CAT , SOD , GPx [68]
DNA damage
Antioxidant capacity of kefir according to milk
[69]
Anti-inflammatoryTNF-α, IL12p70, and IL-8
IL-8/IL-10 and IL-12/IL-10
[37]
TNF-α, IFN-γ [38]
Microbiota modulationBifidobacterium bifidum PRL2010 [48]
Lactobacillus quantity of treatment group for Crohn’s disease
Lactobacillus quantity of treatment group for ulcerative colitis
[70]
Relative abundance of Actinobacteria [38]
Firmicutes/Bacteroidetes ratio, Ascomycota/Basidiomycota ratio
Lactobacillus and Bifidobacterium 
Probiotics’ relative abundance
The pathogenic bacterium (Clostridium sensu stricto, Aspergillus, and Talaromyces)
Clostridium_sensu_stricto_1, Bacteroides, Lachnospiraceae_NK4A136_group, Oscillospiraceae, Desulfovibrio 
Muribaculaceae and Alloprevotella
[67]
Milk kefir had a free radical scavenging activity of 76.640.42%
In the colon: SOD and CAT
Brain butyrate and propionate
Fecal butyrate
Lachnospiraceae and Lachnoclostridium
Relative abundance of Firmicutes
Proteobacteria and Epsilonbacteraeota
[71]
Bone healthPrevented estrogen-deficiency-induced bone loss
Bone volume/total volume
Bone mineral density
Trabecular thickness
Trabecular number
Average cortical elastic moduli, hardness
Trabecular separation
Type I collagen levels
[40]
AntidiabeticInsulin , HOMA-IR [38]
Serum glucose
HbA1c
[72]
Cognitive functionImprovement in performance in the MMSE
Improvement in the memory test
[37]
HypocholesterolemicSerum LDL-C
LDL-C/HDL-C ratio
Serum HDL-C
[39]
Lactic acid bacteriaImmunomodulatoryMucins (MUC-1 and MUC-2) and IgA gene expression [39]
AntioxidantLactiplantibacillus plantarum MA2 had antioxidant potential[47]
Organic acidsAntimicrobialMilk fermentation with kefir grains antagonizes Bacillus cereus through the organic acids (lactic acid and acetic acid) produced during fermentation[73]
Escherichia coli, Salmonella, and Bacillus Cereus pathogenic strains’ growths were inhibited
This related to the concentration of lactic acid
[74]
Bioactive peptidesAntihypertensiveACE activity inhibition[75]
Peptides defined in kefir have previously shown an ACE inhibiting effect[76]
ACE inhibitory activity[77]
AntifibrosisKidney cells
Relative expression of α-SMA)
Relative expression of ET-1
Relative expression of MMCP-1)
Kidney tissues
Protein expression of ET-1
Protein expression of α-SMA
[78]
Anti-inflammatoryPro-inflammatory cytokines [34]
NF-kB protein expression
TGF-β protein expression
NLPR3 protein expression
[78]
AntioxidantTotal antioxidant capacity of the FRAP [41]
ABTS and DPPH radical scavenging activity[79]
ROS production
Lipid peroxidation
[34]
Renal effects:
SOD activity
ROS activity
[78]
Antimicrobial Escherichia coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, Klebsiella pneumoniae ATCC 29665, Bacillus subtilis ATCC 6633, Bacillus cereus ATCC 33019, and Staphylococcus aureus ATCC 6538 growths were inhibited[79]
Increasing the outer and inner membrane permeability of Escherichia coli, causing damage to the cell membrane, and promoting intracellular material leakage[80]
NeuromodulationNeurodegeneration index
Acetylcholinesterase activity
Lower amyloid content
[41]
Bone healthPreventing menopausal osteoporosis
Trabecular number
Trabecular bone volume
Trabecular thickness
Average cortical elastic moduli, hardness
Bone mineral density
Trabecular separation
[81]
Microbiota modulationRestored the abundances of Alloprevotella, Parasutterella, Anaerostipes, Ruminococcus_1, Romboutsia, and Streptococcus genera[81]
Polysaccharide
KefiranAnticancerMCF7 cancer cells , PBMC [49]
Anti-proliferative effect on HeLa and HepG2
Cell viability of HeLa and HepG2
[50]
Anti-inflammatory and immunomodulatory rolesProinflammatory cytokines (NF-kB, IL-1β, TNF-α)
Overexpression of TLR4
[51]
BALB/c mice
Small intestine:
IgA, IL-10, IL-6, IL-12
Serum:
IL-4, IL-6, IL-10
Intestinal fluid:
IL-4, IL-12
Large intestine:
IgA, IgG, IL-6, IL-10, IL-4, IFN, TNF
[52]
Inhibition percentage of nitric oxide radical production[53]
AntioxidantScavenging of superoxide and hydroxyl radicals[53]
DPPH free radicals scavenging activity [54]
Lipid peroxide of βVLDL [55]
Microbiota modulationIntestinal Bifidobacteria [42]
Bifidobacterium bifidum PRL2010 [48]
ExopolysaccharideAnticancerAntitumor activity against colon cancer HT-29 cells
Upregulate the expression of Cyto-c, BAD, BAX, caspase3, caspase8, and caspase9
Downregulate BCl-2
[56]
Anti-inflammatory and immunomodulatory rolesCell viability of the RAW264.7 cells
NO concentration
TNF-α, IL-1β concentration
iNOS concentration
Proliferation and phagocytosis are increased to combat infection and inflammation
[57]
Dose-dependent effects:
Cell viability of the RAW264.7 cells
NO concentration
TNF-α, IL-1β concentration
Enhanced the proliferation, phagocytosis
[58]
Dose-dependent effects:
NO concentration
TNF-α, IL-6, IL-1β, IL-10 concentration
Increasing the activity of acid phosphatase
Enhancing macrophages’ phagocytosis
Viability of macrophages
[59]
AntioxidantGPx 21.55%, SOD 33.14%, CAT 61.09%
Total antioxidant capacity 38.18%
MDA
[61]
Certain scavenging activities:
DPPH free radical scavenging activity
ABTS free radical scavenging activity
Hydroxyl free radical scavenging activity
[60]
Microbiota modulationThe abundance of Flexispira
The abundances of Blautia and Butyricicoccus
Content of SCFA
Content of NO
[61]
Total SCFA
Propionic acid and Butyric acid
Proportion of the genera Victivallis, Acidaminococcus, and Comamonas
Proportion of Enterobacteria
[62]
The abundance of the phyla Bacteroidetes, Verrucomicrobia, and Proteobacteria
The abundance of the Firmicutes and Actinobacteria
The enhanced abundance of Akkermansia spp. in feces
[63]
Anti-obesityLower intracellular lipid accumulation
Epididymal adipose tissue weight 19%
Body weight gain
VLDL-C 36%
[63]
: increased, : decreased, ACE: angiotensin-converting enzyme, TNF-α: tumor necrosis factor alpha, IL: interleukin, TGF-α: transforming growth factor alpha, TGF-β1: transforming growth factor beta, TAS: total antioxidant status, LPS: lipopolysaccharide, CAT: catalase, MDA: malondialdehyde, SOD: superoxide dismutase, GPx: glutathione peroxidase, α-SMA: α-smooth muscle actin, ET-1: endothelial-1, MCP-1: monocyte chemoattractant protein-1, HOMA-IR: homeostasis model assessment of insulin resistance, ABTS: 2,2′-azino-bis(3-ehtylbenzothiazoline-6-sulfonate), DPPH: 2,2-diphenyl-1-picrylhydrazyl, iNOS: inducible nitric oxide synthase, NO: nitric oxide, SCFA: short-chain fatty acid, LDL-C: low-density lipoprotein cholesterol, HDL-C: high-density lipoprotein cholesterol, VLDL-C: very-low-density lipoprotein cholesterol, TLR-4: Toll-like receptor 4, Ig: immunoglobulin, IFN: interferon, DNA: deoxyribonucleic acid, MMSE: mini-mental state examination, HbA1C: glycated hemoglobin, NF-Kb: nuclear factor kappa B, PBMC: peripheral blood mononuclear cells, NLPR3: nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3, HeLa: human cervical cancer cells, HepG2: human liver hepatocarcinoma cells.
Table 3. Possible health effects of microorganisms isolated from some fermented vegetables.
Table 3. Possible health effects of microorganisms isolated from some fermented vegetables.
Fermented VegetablesMicroorganismHealth EffectsSpecific EffectsReferences
Kimchi
Weissella cibaria JW15Anti-inflammatoryProinflammatory cytokines (IL-1β, IL-6, TNF-α)
Nitric oxide, prostaglandin E2, COX-2
IκB-α degradation and MAPKs, NF-κB activation
[239]
Lactiplantibacillus plantarum LB5 (LPLB5)Antioxidant
Anti-inflammatory Antibacterial
Proinflammatory cytokines (IL-1β, IL-6, TNF-α)
Anti-inflammatory cytokines (IL-4, IL-10, IFN-γ)
Escherichia coli O157:H7 Pseudomonas aeruginosa, Listeria monocytogenes, and Staphylococcus aureus 
ABTS radical scavenging activity
[240]
Lactiplantibacillus plantarum LRCC5314Anti-inflammatory
Anti-stress
TNF-α, IL-1β, IFN-γ, NO
Cortisol concentration
Adipocytes:
TG concentration
Adipogenesis-related genes, adiponectin, FAS, PPAR/γ, and C/EBPα, TNF-α, IL-6
[241]
Lactiplantibacillus plantarum 200655Neuroprotective BDNF expression and concentration
BDNF and tyrosine hydroxylase mRNA expression
Apoptosis-related Bax/Bcl-2 ratio
Caspase-3 activity
[242]
Lactobacillus sakeiAnti-obesityBody fat mass
Abdominal visceral fat
Waist circumference
[243]
Levilactobacillus brevis KU15153Antioxidant
Antimicrobial
Escherichia coli ATCC 25922, L. monocytogenes ATCC 15313, S. Typhimurium P99, and S. aureus KCCM 11335
DPPH radical scavenging activity
[244]
Levilactobacillus brevis KU15147Antioxidant
Immune enhancing
NO production, iNOS, TNF-α
Radical scavenging activity of DPPH 38.56%
Radical scavenging activity of ABTS 22.30%
β-carotene bleaching inhibitory activity 23.82%
[245]
Lactiplantibacillus plantarum LRCC5310
Lactiplantibacillus plantarum
LRCC5314
Antidiabetic
Anti-inflammatory
Serum insulin
Fasting blood glucose
Upregulating expression of GLUT 4 and adiponectin
TNF-α, IL-6
Downregulation of Ccl2 and leptin expression
Serum corticosterone
mRNA levels of stress-related genes (Npy, Y2r)
[246]
Sauerkraut
Exopolysaccharides
from Lacticaseibacillus paracasei
AntioxidantTotal antioxidant capacity 76.34%
Hydrogen peroxide scavenging activity 68.65%
DPPH free radical scavenging activity 60.31%
[248]
Exopolysaccharides from Lacticaseibacillus CaseiAntioxidant
Immunomodulatory
Showed dose-dependent effects: Hydrogen peroxide scavenging activity, DPPH free radical scavenging activity, superoxide radicals scavenging activity
In macrophages:
TNF-α, ROS production
NF-κB p65 expression
Expression of the c-jun protein
[249]
Lacticaseibacillus casei NA-2AntibacterialInhibit the growth of Bacillus cereus, Staphylococcus aureus, Escherichia coli O157:H7, and Salmonella typhimurium[250]
Lactiplantibacillus plantarumAntimicrobialEscherichia coli O157 and Shigella flexneri CMCC(B) [251]
: increased, : decreased, IL: interleukin, COX-2: cyclooxygenase 2, IFN: interferon, TNF-α: tumor necrosis factor alpha, MAPK: mitogen-activated protein kinases, ABTS: 2,2′-azino-bis(3-ehtylbenzothiazoline-6-sulfonate), NO: nitric oxide, NF-kB-: nuclear factor kappa B, IκB-α: IkappaB-alpha, iNOS: inducible nitric oxide synthase, BDNF: brain-derived neurotrophic factor, DPPH: 2,2-diphenyl-1-picrylhydrazyl, ROS: reactive oxygen species, GLUT-4: glucose transporter-4, PPAR/γ: peroxisome proliferator-activated receptor gamma, FAS: fatty acid synthase (lipogenic marker), NPY: neuropeptide Y, Y2R: neuropeptide Y receptor 2, CCL-2: C-C motif chemokine ligand 2, C/EBPα: CCAAT/enhancer binding protein alpha.
Table 4. Effects of fermented legumes and certain bioactive compounds on health.
Table 4. Effects of fermented legumes and certain bioactive compounds on health.
Fermented FoodsSpecific FoodsCertain Bioactive CompoundsEffects of HealthReferences
Kidney beansWhite and dark kidney beans-Cecal short-chain fatty acid levels (acetate, butyrate, and propionate), colon crypt height, and MUC1 and Relmβ mRNA expression
Genes of TLR4, MUC1-3, Relmβ
Expressions of IL-6, IFNγ, IL-1β, MCP-1, and TNFα
Levels of serum for IL-17A, TNFα, IL-6, IL-1β, and IFNγ
[360]
Kidney bean fermented broth-With this diet, level of blood lipids (ALT, AST, TG) in hyperlipidemia
With this diet, serum HDL in hyperlipidemia
Firmicutes/Bacterioidetes ratio and pathogenic bacteria
Beneficial bacteria
[361]
SoybeanFermented soybean dried extractsIsoflavin β12-O-tetradecanoylphorbol-13-acetate (TPA-)-induced biochemical alterations in skin
GSH depletion
[362]
Fermented soybean pasteHistamine
Tyramine
Increased hepatic expression of IL-1β and PARP-1
Elevated blood plasma levels of MAO-A, AST/ALT, and CRP
[363]
Fermented soybean products-IgE immunoreactivity [364]
Fermented mung bean
Fermented soybean
-Having cytotoxicity activities opposite to breast cancer MCF-7 cells by arresting the G0/G1 phase, followed by apoptosis
Vaibility and the proliferation of splenocyte
Levels of serum for IL-2 and IFN-γ
[365]
Fermented soybeanAqueous extract of HawaijarGlucose uptake, G6P production, and expressions of pPI3K, pAKT, pAMPK, and GLUT4 [366]
Fermented soybeanIsoflavone
(genistein and daidzein)
Level of progesterone [367]
Alcohol-fermented soybean-p38, iNOS mRNA, JNK, and TNF-α in mouse peritoneal macrophages [368]
Soybean fermented by Lacticaseibacillus paracasei TK1501Lipoteichoic acid (LTA)
Peptidoglycan (PGN)
Via lipoteichoic acid (LTA):
Serum IL-4 and colonic TGF-β1 expression , serum IL-1β and colonic IFN-γ expression , intestinal inflammation , mRNA levels of MUC2
Via peptidoglycan (PGN):
Serum TNF-α and colonic IFN-γ , colonic TGF-β1 expression , mRNA levels of MUC2
[369]
Soybean fermented by Bacillus subtilisMenaquinone-7, daidzin, genistein, glycitin, and nattokinase AChE activity within hippocampus
Protein carbonyl contents in hippocampus
Activity of reduced glutathione, catalase, superoxide dismutase in hippocampus
[370]
Black soybean fermented by Bacillus subtilis-Expression of aging biomarkers (hepatic p16INK4A and GLB1)
Hepatic 8-hydoxy-2′-deoxyguanosine (8-oxodG)
Hepatic levels of IL-6, MCP-1, and IL-10 levels in elder mice
Beneficial microbiomes (Alistipes, Anaeroplasma, Coriobacteriaceae UCG002, and Parvibacter spp.)
[371]
Black soybean and fermented black soybean broth-Antioxidative effect by inhibiting power and ferrous ion chelating
Detroit 551 cell viability
[372]
Fermented soy permeateIsoflavones and α-galactooligosaccharidesMuscle glycogen content [373]
ChungkookjangGenistin
Daidzein
DNA fragmentation
Viability of splenocytes and thymocytes
Apoptosis of splenocytes and thymocytes
[374]
CheonggukjangIntact isoflavones (genistein, daidzein, and glycitein)
Equol 7-glucuronide
Genistein, 3-hydroxygenistein, and 4′-sulfate
Intact isoflavones (genistein, daidzein, glycitein), 3-hydroxygenistein, genistein 4′-sulfate, and equol 7-glucuronide promote osteoblastogenesis via increased ALP activity,
3-hydroxygenistein inhibits osteoclast formation via decreased bone resorption activity
[375]
Cheonggukjang-NF-kB and MAPK activation, IL-4 mRNA expression, IgE expression, and IL-31 mRNA expression in atopic dermatitis [376]
Doenjang
Cheonggukjang
-Activation of redox-sensitive NF-kB
iNOs levels, COX-2
[377]
Doenjang
Cheonggukjang
-Th1-mediated immune responses
Level of IFN-γ
Level of IL-4
Resistance to Listeria monocytogenes infection
[378]
Doenjang-Fecal lipopolysaccharide levels
The amount of Ruminococcaceae, Bifidobacteria, Lachnospiraceae, and Firmicutes 
The amount of Odoribacter_f and Bacterioidetes 
β-glucuronidase and NF-kB activity
TNF- α expression
IL-10 expression
Occludin
[379]
Cheonggukjang (natto)NattokinaseDigestion of fibrin
Digestion of plasmin substrate (H-D-Val-Leu-Lys-Pna (s-2251))
[380]
NattoNatto extract
(Heated-natto extract
or
Unheated-natto extract)
Heated-natto extract,
degradation of Glycoprotein D of BHV-1
Degradation of SARS-CoV-2 receptor-binding domain
Unheated-natto extract,
inhibition of anti-BHV-1 activity by serine protease inhibitor
[381]
NattoVitamin K
Phytoestrogens
Vitamin K, bone health
Phytoestrogens, menopausal disorder, osteoporosis, breast cancer risk
[382]
NattoVitamin K2Maintaining bone stiffness[383]
Natto-In women aged under 60 years, dementia risk [384]
MisoLipopolysaccharide-neutralizing proteinPGD2 production via macrophage cells [385]
Fermented soybeansC-miso (a)
S10-miso (b)
S9O1-miso (c)
Antioxidant effects:
For unheated forms: a > b > c
For heated forms: a > b > c
Antimutagenicity effects:
For unheated forms: a = b > c
For heated forms: a > b > c
[386]
Miso soup, fermented soybeans, houba-misoIsoflavoneHot flush severity[387]
Miso soup, natto, and soybeans-Attenuated arterial stiffness via brachial–ankle pulse wave velocity [388]
Miso and nattoIsoflavonesBlood pressure [389]
Low-salt O-miso-Serum cholesterol
Serum and liver TBARS value
Serum GSH-Px and hepatic catalase
[390]
Soybean koji-Increase in mRNA expression incident to lipogenic genes and weightiness of white adipose tissue
Serum levels of triglyceride, low-density lipoprotein cholesterol, and total cholesterol
Serum levels of high-density lipoprotein cholesterol
Lipid accumulation in the white adipose tissue and liver
[391]
Soy MejuTetragenococcus halophilus EFEL7002May adhere to Caco-2 cells
Protective effect against H2O2-induced epithelial damage
Antioxidant activity in human intestine
Anti-inflammatory effects by inhibiting NO synthase within RAW 264.7 cells
mRNA expressions of IL-6, IL-10, and IL-1β
[392]
Thua-NaoDaidzein
Genistein
MCF-7 and HEK293 cancer cell growth
Amount of viable HepG2 cells
[393]
Fermented soy beverage-Levels of LDL cholesterol and total cholesterol [394]
Soy milkSoy milk powderIsoflavones
3-HAA
TG accumulation and total cholesterol within liver under oxidative stress [395]
Fermented soy milk via Enterococcus faecalis VB43Reduction in conglycinin (7S) and glycinin (11S)Enterococcus faecalis VB43-fermented soy milk may cause less severe allergy reactions in susceptible people[396]
Red beanTempeh
(fermented red bean via Rhizopus and Lactobacillus)
Anthocyanin and GABAROS, pCREB, and iNOS expressions
BDNF expression
[397]
: increased, : decreased, MUC1: colonic mucin 1, MUC2: colonic mucin 2, CREB: cAMP response element binding protein, MUC3: colonic mucin 3, Relmβ: resistin-like molecule beta, TLR4: Toll-like receptor 4, IFN-γ: interferon-γ, AST: aspartate aminotransferase, TG: triglyceride, NF- kB: nuclear factor kB, HDL: high-density lipoprotein, GSH: glutathione, PARP-1: poly (ADP-ribose) polymerase 1, MAO-A: monoamine oxidase A, CRP: C-reactive protein, IL-: interleukin-, TGF-β1: transforming growth factor beta 1, AChE: acetylcholinesterase, SOD: superoxide dismutase, GLB1: galactosidase beta-1, ALP: alkaline phosphatase, MAPK: mitogen-activated protein kinase, MCP-1: monocyte chemoattractant protein-1, Th1: T helper type 1, iNOS: inducible nitric oxide synthase, PGD2: prostaglandin D2, TBARS: thiobarbituric acid reactive substances, H2O2: hydrogen peroxide, LDL: low-density lipoprotein, ROS: reactive oxygen species, NO: nitric oxide, cAMP: cyclic adenosine monophosphate, BDNF: brain-derived neurotrophic factor, 3-HAA: 3-hydroxyanthranilic acid, JNK: c-Jun N-terminal kinase, TNF-α: tumor necrosis factor-α, ALT: alanine aminotransferase.
Table 5. Effects of fermented cereals and certain bioactive compounds on health.
Table 5. Effects of fermented cereals and certain bioactive compounds on health.
Fermented FoodsCertain Bioactive CompoundsEffects of HealthReferences
JalebiLapidilactobacillus bayanensis
Bacillota
Candida glabrata
Lapidilactobacillus dextrinicus
Pichia kudriavzevii
Pediococcus stilesii
Wickerhamomyces anomalus
Gluconobacter japonicus
Probiotic functions[414]
OgiCombination with tigernuts and sesame seedsAntioxidant activity
α-glucosidase enzyme inhibitory activity
[415]
BordeLactic acid bacteria strains (WS07, AM15, and AM20)
Yeast strains (WS15, AA19, AM18, and AM23)
Cholesterol lowering ability [416]
Kunu-zakiLimosilactobacillus fermentum
Leuconostoc citreum
Weissella confusa
Anti-fungal activity [417]
KounouFlavonoids
Polyphenols
Antioxidant activity[418]
Bozai (Boza)Bacteriocin LF-BZ532Antimicrobial spectrum opposite to both Gram-positive and Gram-negative bacteria[419]
Fermented cereal pastesLactobacillusSerum and hepatic cholesterol levels
Ratio of LDL-C to HDL-C
Hepatic LDL receptor and CYP7A1 gene expressions
Activity of superoxide dismutase
Count of coliform and Clostridium perfringens in feces
[420]
Kunu-zaki
Ogi
Lactiplantibacillus plantarum ULAG11
Lactiplantibacillus plantarum ULAG24
Exclusion of Salmonella enterica LT2 via adherence of L. plantarum ULAG24 to HT29 cell line
Stimulation of IFNγ and IL-10 via L. plantarum ULAG24
Expression of amylase via L. plantarum ULAG11
[421]
Fermented quinoa and wheatBifidobacterium breve
Bifidobacterium longum
ACE-inhibition activities
Antioxidant activities
Cytotoxicity activities against Caco-2 cell line
[422]
Fermented barleyLactobacillusHepatic superoxide dismutase activity
Improvement in intestinal microbiota dysbiosis
Bacteroidetes
Firmicutes/Bacteroidetes ratio
[423]
Fermented quinoa flourPleurotus ostreatusACE-I inhibitory [424]
TogwaLactic acidCampylobacter spp., Salmonella spp., ETEC and Shigella spp. [425]
Fermented rye-Romboutsia
Bilophila
Fecal acetic acid
[426]
Fermented Tartary buckwheatMonascus purpureusLiver glycogen content
SOD activity
CAT activity
[427]
Fermented pearl millet flourAspergillus sojaeAntioxidant activity
DNA damage protection activity
[428]
Fermented sorghumPediococcus acidilactici OHFR1Muribaculum, Parabacteroides, and Phocaeicola 
Oscillibcater, Acetatifactor, and Acetivibrio
[429]
Bhaati Jaanr-Proliferation of colon adenocarcinoma cell lines (HT29 and SW480)
Expression of IL-1β, COX-2, IL-6, and TNF-α
[13]
: increased, : decreased, LDL-C: low-density lipoprotein cholesterol, HDL-C: high-density lipoprotein cholesterol, CYP7A1: cholesterol-7α-hydroxylase, IL-: interleukin-, ACE: angiotensin-converting enzyme, ETEC: enterotoxigenic Esherichia coli, SOD: superoxide dismutase, CAT: catalase, DNA: deoxyribose nucleic acid, IFN-γ: interferon-γ, TNF-α: tumor necrosis factor-α.
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

Deveci, G.; Çelik, E.; Ağagündüz, D.; Bartkiene, E.; Rocha, J.M.F.; Özogul, F. Certain Fermented Foods and Their Possible Health Effects with a Focus on Bioactive Compounds and Microorganisms. Fermentation 2023, 9, 923. https://doi.org/10.3390/fermentation9110923

AMA Style

Deveci G, Çelik E, Ağagündüz D, Bartkiene E, Rocha JMF, Özogul F. Certain Fermented Foods and Their Possible Health Effects with a Focus on Bioactive Compounds and Microorganisms. Fermentation. 2023; 9(11):923. https://doi.org/10.3390/fermentation9110923

Chicago/Turabian Style

Deveci, Gülsüm, Elif Çelik, Duygu Ağagündüz, Elena Bartkiene, João Miguel F. Rocha, and Fatih Özogul. 2023. "Certain Fermented Foods and Their Possible Health Effects with a Focus on Bioactive Compounds and Microorganisms" Fermentation 9, no. 11: 923. https://doi.org/10.3390/fermentation9110923

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

Article Metrics

Back to TopTop