Next Article in Journal
A Novel Extracellular Catalase Produced by the Antarctic Filamentous Fungus Penicillium Rubens III11-2
Next Article in Special Issue
Fermented Beverages Revisited: From Terroir to Customized Functional Products
Previous Article in Journal
Engineering an Artificial Pathway to Improve the Bioconversion of Lysine into Chiral Amino Alcohol 2-Hydroxycadaverine Using a Semi-Rational Design
Previous Article in Special Issue
Use of Non-Saccharomyces Yeast to Enhance the Acidity of Wines Produced in a Warm Climate Region: Effect on Wine Composition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fermented Beverages Revisited: From Terroir to Customized Functional Products

by
Spiros Paramithiotis
1,*,
Jayanta Kumar Patra
2,
Yorgos Kotseridis
3 and
Maria Dimopoulou
4
1
Department of Biological Applications and Technology, University of Ioannina, 45110 Ioannina, Greece
2
Research Institute of Integrative Life Sciences, Dongguk University-Seoul, Goyangsi 10326, Republic of Korea
3
Department of Food Science & Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
4
Department of Wine, Vine and Beverage Sciences, University of West Attica, 12243 Athens, Greece
*
Author to whom correspondence should be addressed.
Fermentation 2024, 10(1), 57; https://doi.org/10.3390/fermentation10010057
Submission received: 25 November 2023 / Revised: 20 December 2023 / Accepted: 12 January 2024 / Published: 14 January 2024

Abstract

:
Fermented beverages have been a constant companion of humans throughout their history. A wide range of products have been developed with time, depending on the availability of raw materials and ambient conditions. Their differentiation was based on the specific characteristics of each product, resulting from the cultivation of different varieties and the variability of environmental conditions and agricultural practices, collectively described by the term ‘terroir’ that was developed in winemaking. The health benefits that have been associated with their consumption, which include the control of blood pressure and glycemic control, along with immunomodulatory, hypocholesterolemic, hepatoprotective, and antiproliferative activities, directed their re-discovery that occurred over the last few decades. Thus, the dynamics of the microbial communities of fermented beverages during fermentation and storage have been thoroughly assessed. The functional potential of fermented beverages has been attributed to the chemical composition of the raw materials and the bioconversions that take place during fermentation and storage, due to the metabolic capacity of the driving microbiota. Thus, the proper combination of raw materials with certain microorganisms may allow for the modulation of the organoleptic properties, as well as enrichment with specific functional ingredients, enabling targeted nutritional interventions. This plasticity of fermented beverages is their great advantage that offers limitless capabilities. The present article aims to critically summarize and present the current knowledge on the microbiota and functional potential of fermented beverages and highlight the great potential of these products.

1. Introduction

Fermented beverages are produced since antiquity, with wine being the most characteristic example. They can be classified according to the nature of the raw materials, or the type of fermentation employed. According to the first, two classes of fermented beverages are distinguished: plant-based and dairy. The plant-based ones can be further subdivided into cereal-based products, such as boza, cheka, pozol, kvass, the various types of beer, etc. [1,2,3,4,5,6]; fruit-based products, such as the various types of wine, cider, gilaburu, etc. [7,8,9,10,11]; and herbal-based products, such as kombucha [12,13]. As far as fermented dairy beverages are concerned, several types have been described, such as kefir, kumis, viili, acidophilus milk, etc. [14], each with a unique history, production procedure and microecosystem composition. If the predominant type of fermentation is taken as a criterion, fermented beverages can be classified into acidic, alcoholic, and mixed fermented products; kefir and kombucha can serve as examples of acidic beverages, wine of alcoholic, and boza of mixed fermented beverages.
The term ‘terroir’ has been developed and is currently in use in wine technology. It indicates a specific geographical area along with the associated grape cultivars and oenological practices, which altogether define the specific features of the produced wines. Nowadays, the health benefits that accompany food consumption have also been at the epicenter of consumer interest. This trend has led to the intensive study of fermented beverages throughout the world, their functional ingredients as well as the molecular mechanisms that are implicated. Apart from grape wine, two fermented beverages have also been distinguished for their functional potential, namely kefir and kombucha.
Thus, moderate wine consumption has been correlated with reduced risk of cardiovascular events [15,16], neurodegenerative diseases [17,18], and type 2 diabetes [19,20], as well as systolic blood pressure decrease [21,22,23], improvement of the gastrointestinal tract function [24,25,26] and the main symptoms of fibromyalgia [27]. These have all been principally attributed to the phenolic compounds that it contains. Kombucha consumption has been correlated with a series of effects, such as antidiabetic, antihypertensive, antimicrobial, antiproliferative, hepatoprotective, hypocholesterolemic and immunomodulatory [28,29,30,31,32,33,34,35,36,37]. As in the case of grape wine, these activities have been attributed to kombucha phenolic content. Finally, kefir consumption has been correlated with a series of effects, including antiproliferative and immunomodulatory capacity, effective glycemic control, prevention and treatment of atherosclerosis and liver damage, as well as control of blood pressure [38,39,40,41,42,43,44]. These activities have been attributed to the amino acid and peptide content of kefir.
The present study aimed to comprehensively and critically present the microbiota of fermented beverages, the functional properties that have been associated with the consumption of the most prominent ones, along with the underlying molecular mechanisms, as well as the strategies that have been employed towards their customization, in order to summarize the current knowledge and facilitate the identification of research gaps.

2. The Microbiota of Traditional Fermented Beverages

In Table 1, the microbiota of traditional fermented beverages around the world is presented. Three aspects are immediately noticed: (1) the diversity of the raw materials employed, (2) the diversity of the microorganisms implicated in each type of fermentation, and (3) some products have attracted scientific attention and are therefore heavily studied, while the microecosystem of others has been less intensively assessed.
The availability of raw materials combined with suitable environmental conditions has led to the development of a wide range of fermented beverages, some of which are considered as characteristic of certain geographical areas. Indeed, products such as kombucha and bhaati jaanr, which are based on tea and rice, respectively, have Asian origins; products based on maize, such as pozol, chicha, and atole agrio originate from America, whereas milk and cereal-based fermented beverages are abundant in Africa and Europe. Despite the current globalization of production, traditional knowledge may still be derived from the cradle in which each product was originally developed.
In the majority of the cases, the microbial consortium that drives fermentation of the products reported in Table 1 consists of yeasts and bacteria, and more specifically, lactic and acetic acid bacteria. This can be attributed to the composition of the raw materials and the conditions used for fermented product manufacture but is biased by the number of samples analyzed and the identification methodology employed. In general, the higher the number of samples analyzed, the more likely is to enrich the biodiversity already reported in the literature. A wide range of techniques are currently available, the correct application of which may lead to reliable identification at species or subspecies level. The specific nutritional and growth requirements of each microorganism, at strain level, define the nature of the relationships that will be developed within the microecosystem. Despite the large number of studies, at least in the case of kombucha and kefir, the trophic relationships between the microorganisms are still far from being understood. The only exception seems to be wine must fermentation. In that case, the requirements and the capacity of the implicated yeast species have been extensively studied, and this knowledge is already considered as established. On the other hand, research is currently focused on understanding the physiological attributes of the lactic acid bacteria that carry out malolactic fermentation [45].
Table 1. The microbiota of representative traditional fermented beverages around the world.
Table 1. The microbiota of representative traditional fermented beverages around the world.
ProductMain IngredientMicrobiotaReferences
KombuchaSweetened black or green teaLAB: O. oeni, Lq. nagelii
AAB: A. aceti, A. musti, A. okinawensis, A. pasteurianus, A. peroxydans, A. senegalensis, A. tropicalis, A. xylinoides, A. xylinum, Ga. europaeus, Ga. hansenii, Ga. intermedius, Ga. xylinus, Gb. oxydans, Kb. europaeus, Kb. hansenii, Kb. intermedius, Kb. rhaeticus, Kb. saccharivorans, Kb. xylinus
Yeasts: Br. anomalus, Br. bruxellensis, Br. lambicus, Ca. albicans, Ca. boidinii, Ca. colleculosa, Ca. guilliermondii, Ca. kefyr, Ca. krusei, Ca. sake, Ca. stellata, De. anomala, De. bruxellensis, H. valbyensis, K. marxianus, Kz. unispora, Lh. fermentati, Pi. fermentans, Pi. membranifaciens, R. mucilaginosa, S. cerevisiae, S. uvarum, Sd. ludwigii, Sz. pombe, T. delbrueckii, Z. bailii, Z. lentus, Z. parabaillii, Zt. fiorentina
[12,13,46,47,48,49,50,51,52,53,54]
KefirMilk LAB: E. durans, E. faecalis, La. casei, La. paracasei, Lp. plantarum, Lb. acidophilus, Lb. amylovorus, Lb. delbrueckii, Lb. crispatus, Lb. helveticus, Lb. kefiranofaciens, Lc. lactis, Lt. buchneri, Lt. kefiri, Lt. parabuchneri, Lt. parakefiri, Le. parakefiri, Ln. mesenteroides, Ln. paramesenteroides, Ln. pseudomesenteroides, Lq. uvarum, Lq. satsumensis, Lv. brevis, Str. durans, Str. thermophilus
AAB: A. aceti, A. fabarum, A. lovaniensis, A. okinawensis, A. orientalis, A. rancens, A. syzygii, Gb. frateurii, Gb. japonicus
Yeasts: Br. anomalus, Ca. colliculosa, Ca. inconspicua, Ca. kefyr, Ca. krusei, Ca. lambica, Ca. maris, De. anomala, K. lactis, K. marxianus, Kz. aerobia, Kz. exigua, Kz. kefir, Kz. unispora, Lh. meyersii, M. guilliermondii, Pi. kudriavzevii, Pi. guilliermondii, S. cerevisiae, S. fragilis, S. turicensis, T. delbrueckii
[55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81]
Wine Fruits Yeasts: Au. pullulans, Ca. fermentati, Ca. intermedia, Ca. parapsilopsis, Ca. pulcherrima, Ca. quercitrusa, Ca. zemplinina, H. uvarum, H. guillermondii, H. uvarum, H. valbyensis, I. occidentalis, I. orientalis, I. terricola, Kc. apiculata, Lh. thermotolerans, Pi. fermentans, R. graminis, R. mucilaginosa, S. bayanus, S. cerevisiae, S. italicus, S. pastorianus, S. uvarum, Sd. ludwigii, Sz. pombe, T. delbrueckii, T. globispora, Y. lipolytica, Z. bailii, Z. fermentati[9,10,82,83,84,85,86]
Apple ciderApples Yeasts: H. osmophila, H. uvarum, H. valbyensis, M. pulcherrima, Pi. guillermondii, S. bayanus, S. cerevisiae[8]
Amabere amaruranu Milk LAB: Ln. mesenteroides, Lp. plantarum, Str. thermophilus 
Yeasts: Ca. albicans, Ca. famata, S. cerevisiae, Tr. mucoides
[87]
Andean chichaCereals Yeasts: H. guiermondii, H. opuntiae, H. uvarum, Ko. ohmeri, R. slooffiae, Mz. guillermondii, Pi. kluyveri, Pi. kudriavzevii, R. mucilaginosa, Wi. anomalus, S. cerevisiae, Y. lipolytica[88]
Atole agrioMaize LAB: Ag. composti, E. hirae, La. casei, La. paracasei, La. rhamnosus, Lc. lactis, Lc. piscium, Li. aviarius, Ln. garlicum, Ln. mesenteroides, Ln. pseudomesenteroides, Lo. coryniformis, Lp. fabifermentans, Lp. paraplantarum, Lp. pentosus, Lp. plantarum, Lt. curvatus, Lv. brevis, P. pentosaceus, P. stilesii, W. cibaria, W. confusa, W. hellenica, W. paramesenteroides, Str. equinus
AAB: A. estunensis, A. indonesiensis, A. pasteurianus, A. tropicalis, Gb. frateurii
[89]
Bacaba chichaOenocarpus bacabaLAB: E. durans, E. hirae, Ln. lactis 
Yeasts: Pi. caribbica, Pi. guillermondii
[90]
Bhaati jaanrRice LAB: Lo. bifermentans, P. pentosaceus 
Yeasts: Ca. glabrata, Pi. anomala, S. cerevisiae, Sp. fibuligera
[91]
Bili biliSorghumYeasts: Ca. melibiosica, Cr. albidius, D. hansenii, De. bruxelensis, K. marxianus, R. mucilaginosa, S. cerevisiae, T. delbrueckii[92]
BordeCereals LAB: Lv. brevis, P. pentosaceus, W. confusa, W. viridescens[93]
BozaCereals LAB: Fr. sanfransiscensis, La. casei, La. paracasei, Lb. acidophilus, Lc. lactis, Li. salivarius, Lm. fermentum, Ln. amelibiosum, Ln. mesenteroides, Ln. paramesenteroides, Ln. pseudomesenteroides, Lo. coryniformis, Lp. plantarum, Lt. buchneri, Lt. parabuchneri, Lt. sakei, P. parvulus, W. confusa
Yeasts: Ca. glabrata, Ca. tropicalis, Pi. fermentans, Pi. guillermondii, Pi. norvegensis, S. cerevisiae, S. uvarum
[94,95,96]
Burukutu Sorghum LAB: Lb. acidopilus, Lc. lactis, Lm. fermentum, Lp. plantarum, Lv. brevis
Yeasts: S. cerevisiae
[97]
Chicha Maize LAB: Lm. fermentum, Lp. plantarum, W. cibaria, Str. alactolyticus, Str. luteciae 
AAB: A. okinawensis
[98]
IkigageSorghumYeasts: Ca. humilis, Ca. inconspicua, Ca. magnolia, S. cerevisae, I. orientalis
LAB: Le. buchneri, Lm. fermentum
[99]
Gilaburu European cranberry LAB: La. casei, La. pantheris, Le. buchneri, Le. parabuchneri, Ln. pseudomesenteroides, Lp. plantarum, Lv. brevis, Sc. harbinensis[11]
MahewuCereals LAB: E. hermanniensis, E. lactis, Fu. rossiae, Lc. lactis, Lm. fermentum, Ln. holzapfelii, Ln. pseudomesenteroides, Lp. plantarum, P. pentosaceus, W. cibaria, W. confusa
Yeasts: Ca. glabrata, S. cerevisiae
[100]
Fermented masauZiziphus mauritianaLAB: Cb. divergens, Le. hilgardii, Li. agilis, Lo. bifermentans, Lm. fermentum, Lp. plantarum, W. minor
Yeasts: Ca. glabrata, H. opuntiae, I. orientalis, Pi. fabianii, S. cerevisiae, Sp. fibuligera
[7]
PitoCerealsYeasts: Ca. tropicalis, Ha. anomala, K. africanus, Kc. apiculata, S. cerevisiae, Sz. pombe, T. delbrueckii[1]
Pozol Maize LAB: C. alimentarius, E. saccharolyticus, La. casei, Lb. delbrueckii, Lc. lactis, Lm. fermentum, Lp. plantarum, Str. bovis, Str. suis
Yeasts: Ca. guilliermondii, Cs. cladosporioides, D. hansenii, Ge. candidum, K. lactis, Pe. fellutanum, Ph. fimeti, Ph. glomerata, R. minuta, R. mucilaginosa
[2,101,102]
PulqueAgave spp.LAB: Fr. sanfranciscensis, Lb. acetotolerans, Lb. acidophilus, Lb. delbrueckii, Lc. lactis, Le. hilgardii, Le. kefiri, Ln. citreum, Ln. gasocomitatum, Ln. kimchi, Ln. mesenteroides, Ln. pseudomesenteroides, Lp. plantarum, P. urinaeequi, Se. paracollinoides, Str. deviesei
AAB: A. aceti, A. malorum, A. orientalis, A. pomorum, Gb. οxydans
Yeasts: Ca. parapsilosis, Ca. valida, Cl. lusitaniae, D. carsonii, H. uvarum, Ge. candidum, K. lactis, K. marxianus, Pi. guilliermondii, Pi. membranifaciens, R. mucilaginosa, S. bayanus, S. cerevisiae, S. pastorianus, T. delbrueckii
[103,104,105,106]
AAB: Acetic acid bacteria; LAB: lactic acid bacteria. A.: Acetobacter; Ag.: Agrilactobacillus; Au.: Aureobasidium; Br.: Brettanomyces; C.: Companilactobacillus; Ca.: Candida; Cb.: Carnobacterium; Cl.: Clavispora; Cr.: Cryptococcus; Cs.: Cladosporium; D.: Debaryomyces; De.: Dekkera; E.: Enterococcus; Fr.: Fructilactobacillus; Fu.: Furfurilactobacillus; Ga: Gluconacetobacter; Gb., Gluconobacter; Ge.: Geotrichum; I.: Issatschenkia; Kb.: Komagataeibacter; H.: Hanseniaspora; Ha.: Hansenula; K.: Kluyveromyces; Kc.: Kloeckera; Ko.: Kodamaea; Kz.: Kazachstania; La.: Lacticaseibacillus; Lb.: Lactobacillus; Lc.: Lactococcus; Le.: Lentilactobacillus; Lh., Lachancea; Li.: Ligilactobacillus; Lm.: Limosilactobacillus; Ln.: Leuconostoc; Lo.: Loigolactobacillus; Lp.: Lactiplantibacillus; Lq. Liquorilactobacillus; Lt.: Latilactobacillus; Lv.: Levilactobacillus; M.: Metschnikowia; Mz.: Meyerozyma; O.: Oenococcus; P.: Pediococcus; Pe.: Penicillium; Ph.: Phoma; Pi.: Pichia; R.: Rhodotorula; S.: Saccharomyces; Sc.: Schleiferilactobacillus; Sd.: Saccharomycodes; Se.: Secundilactobacillus; Sp.: Saccharomycopsis; Str.: Streptococcus; Sz.: Schizosaccharomyces; T.: Torulospora; Tr.: Trichosporon; W.: Weissella; Wi.: Wickeramomyces; Y.: Yarrowia; Z.: Zygosaccharomyces; Za.: Zygoascus; Zt.: Zygotorulaspora.

3. Functional Properties of Fermented Beverages

The consumption of fermented beverages has been associated with a series of functional properties. These have been attributed to the chemical composition of the raw materials employed and the bioconversions that take place during the fermentation process. Thus, the same functional properties may be assigned to the same or different bioactive compounds, the formation of which depends upon the metabolic capacity of the microorganisms that constitute the driving micro-community. In the next paragraphs, the functional properties of wine, kombucha and kefir, the most studied fermented beverages, are summarized.

3.1. Functional Properties of Wine

A series of functional properties have been associated with wine and are mainly attributed to the phenolic compounds that it contains. The type and amount of phenolic compounds depend upon factors such as grape variety, environmental conditions, agricultural practices and winemaking technology [107]. In general, the major phenolic compounds of wine are distinguished into flavonoids and non-flavonoids. Flavonols (quercetin, kaempferol and myricetin), anthocyanins (cyanin, petunin, peonin and malvin), and flavan-3-ols (catechin, epicatechin, gallocatechin, procyanidins and condensed tannins) belong to the first category, while phenolic acids (hydroxybenzoic and hydroxycinnamic acids), volatile phenols (ethyl phenol, vinyl phenol, guaiacol, etc.), and stilbenes (resveratrol and its polymers) belong to the second one [108,109]. The total amount of flavonoids in white and red wines has been reported to range between 25–30 and 700–1000 mg of gallic acid equivalent (GAE/L), respectively. Catechins and soluble tannins are quantitatively the most abundant classes of compounds in white and red wines, respectively. On the other hand, the total amount of non-flavonoids has been reported to range between 160–260 and 230–500 mg GAE/L in white and red wines, respectively, with cinnamates being, in both cases, the most abundant class of compounds, with approximately 150 mg of GAE/L [108]. The bioavailability of the aforementioned compounds is a key issue as it affects their biological function. In general, wine phenolic compounds are only partially bioavailable, not only because of their chemical structure but also due to the biotransformations that take place during digestion [110]; it has been reported to range between 2–25% [111,112,113].
Excessive alcohol consumption has been correlated with an increased incidence of disease [114]. On the contrary, moderate alcohol consumption, particularly wine, seems to have a protective role on human health [115]. The concept of moderate or low-risk wine consumption has been exhaustively debated [115,116]. From a quantitative point of view, the consumption of up to 25–40 g alcohol per day for males and 13–25 g for females is generally accepted as moderate [117]. A range of health benefits have been associated with moderate wine consumption, such as lowering the risk of cardiovascular disease and neurodegenerative disease development, protection against type 2 diabetes, and generally life span prolongation. The capacity of wine to confer these health benefits has been assessed through in vitro, in vivo and clinical studies. The next paragraphs focus on the underlying mechanisms that validate the latter.
An association between moderate wine consumption and reduced risk of cardiovascular events, even among persons with established heart diseases, has been reported [15,118]. This has been attributed to the modulation of circulating cholesterol and anti-platelet activity of alcohol and to the antioxidant, anti-inflammatory and anti-platelet activities of the phenolic compounds [119,120]. The antioxidant activity is expressed through free radical scavenging and through the upregulation of Nrf2, which in turn induces antioxidant gene expression [121]. Regarding their anti-inflammatory activities, a series of mechanisms have been proposed, such as switching off the NF-κB pathway [122], blocking oxysterol-related NOX1 induction [123], suppression of NLRP3 inflammasome activation [124], suppression of the JAK/STAT inflammatory pathway and modulation of Nrf2 activity [125], as well as decrease in IL-1β, IL-6 and IL-8 secretion [126,127]. Finally, the capacity of red wine to inhibit thrombin, ADP- and PAF-induced platelet aggregation has been reported [128,129,130,131,132,133] and attributed to ethanol and polyphenols, particularly quercetin, tyrosol and trans-resveratrol [132,133]. In addition, the inhibition of PAF biosynthesis by tyrosol and resveratrol has also been reported in U-937 cells under inflammatory conditions [134].
An association between moderate wine consumption and reduction in the risk of neurodegenerative diseases has also been developed [17]. The mode through which wine consumption may affect the onset of Alzheimer’s and Parkinson’s diseases has been extensively studied. In the first case, protection may take place through the antioxidant and anti-inflammatory activities of wine, as well as through more specific functions such as the modulation of secretase enzymes, the enhancement of amyloid clearance, the inhibition of amyloid aggregation and the prevention of tau protein hyperphosphorylation [135,136,137]. Indeed, the activation of α-secretase activity by 6% Cabernet Sauvignon, myricetin, quercetin, resveratrol, and caffeic acid [138,139,140,141,142,143], along with the inhibition of BACE1 activity by resveratrol and some of its oligomers, epicatechin, myricetin, quercetin, kaempherol and caffeic acid [142,143,144,145,146,147,148,149,150,151,152,153,154] and inhibition of γ-secretase by resveratrol, oxy-resveratrol, and piceatannol [155,156], have been reported. In addition, amyloid accumulation and aggregation seem to be prevented by a variety of mechanisms. Amyloid clearance or the induction of degradation mechanisms, activities that have been reported for resveratrol [157,158,159] and quercetin [160], have been reported to prevent amyloid accumulation. On the other hand, resveratrol, quercetin, and grape seed pro-anthocyanidin consisting of catechin, epicatechin, and epicatechin gallate, have presented an anti-aggregation capacity [161,162,163,164,165]. Finally, the inhibition of tau protein hyperphosphorylation has been reported for resveratrol, quercetin and caffeic acid [166,167,168,169]. Regarding Parkinson’s disease, protection may take place through the antioxidant activity and neuroprotective effects of resveratrol, which seem to be related to its SIRT-activating potential [170,171,172,173,174], and quercetin, which seems to be related to the induction of the PKD1/CREB/BDNF axis [175,176]. In addition, other constituents such as caffeic acid, gallic acid and catechins have also been reported to contribute to the aforementioned activities [177,178,179].
The association between wine consumption and a reduced risk of type 2 diabetes has been repeatedly reported [19,20]. This association was further improved by Ma et al. [180], which highlighted that this protective action takes place when moderate alcohol drinking, especially wine, takes place with meals. The mode of action includes the inhibition of α-amylase and α-glucosidase activities, the inhibition of sodium-dependent glucose transporter 1 (SGLT1) and the activation of 5-adenosine monophosphate-activated protein kinase (AMPK) [181,182,183,184]. Regarding diabetic patients, wine consumption has been reported to attenuate insulin resistance, with no effect on vascular reactivity and nitric oxide production [185], to reduce the diastolic blood pressure and total cholesterol but not glucose parameters and other cardiovascular risk factors [186], and to reduce the risks of cardiovascular events and all-cause mortality [187]. In addition, the initiation of red wine consumption has been associated with increased high-density lipoprotein cholesterol (HDL-C) and apolipoprotein(a)1 level and decreased the ratio of total cholesterol to HDL-C [188]. In addition, wine consumption improved glycemic control, in terms of fasting plasma glucose, homeostatic model assessment of insulin resistance and hemoglobin A1c, but only in patients carrying the alcohol dehydrogenase alleles [ADH1B*1], i.e., the slow ethanol metabolizers.
Several other health benefits have been correlated with moderate wine consumption, such as the decrease in systolic blood pressure [21,22,23], the improvement of gastrointestinal tract function [24,25,26] and the improvement of the main symptoms of fibromyalgia [27]. However, further research is still necessary in order to identify the responsible molecular mechanisms.

3.2. Functional Properties of Kombucha

Kombucha is commercially available as a non-alcoholic beverage; therefore, the ethanol content should not exceed 0.5% alcohol by volume (ABV). However, in many cases, this limit is not respected [189,190,191] and the production of kombucha with ethanol content as high as 5.83 mg/mL has been reported [49]. A series of functional properties, such as antioxidant, antiproliferative, immunomodulatory, antihypertensive, antidiabetic, hypocholesterolemic, hepatoprotective and antimicrobial, have been attributed to kombucha. In all cases, the functional properties have been attributed, at least partially, to specific compounds that are either present in the raw materials or are formed during fermentation. Therefore, every factor that affects the production of the raw materials or the fermentation procedure is expected to affect the functional properties of the final product, to a greater or lesser degree [32,192,193,194,195,196]. Below, a short description of studies assessing these properties is offered.
The antioxidant capacity of kombucha has been principally attributed to the presence of phenolic compounds. The phenolic concentration and diversity in black-tea kombucha have been reported to be greater than those of green-tea kombucha [35]. This has been attributed to the processing steps that are necessary for black tea production. During these steps, the concentration of theaflavins and thearubigins increases, and they become the main polyphenols in black tea [197]. During fermentation, these compounds are subjected to enzymatic or chemical biotransformation, resulting in the formation of a wealth of lower-molecular-weight phenolic compounds. Indeed, Cardoso et al. [35] identified 126 phenolic compounds in the samples of green and black-tea kombucha that they analyzed, of which, 50 compounds were common, 75 were unique to black-tea kombucha, and only one, namely verbascoside, was unique to green-tea kombucha. Interestingly, the occurrence of five phenolic compounds in green-tea kombucha and 30 phenolic compounds in black-tea kombucha was solely assigned to the fermentation process.
The antioxidant activity of kombucha is usually assessed in vitro through free-radical scavenging assays such as DPPH, FRAP, ABTS, MCA, Curpac, etc. [35,198,199]. In vivo studies are generally lacking; only a few are currently available, the main findings of which are described in the following lines. Dipti et al. [200] used a lead acetate solution to induce oxidative stress on male albino (Sprague Dawley) rats and studied the antioxidant effect of black-tea kombucha. The results of kombucha oral administration included the reduction of DNA damage and lipid peroxidation, as well as the increase of glutathione level and GPx activity. Yang et al. [201] used mice from the Institute of Cancer Research and fed them with a hypocholesterolemic diet (HCD) combined with 66 mL Kg−1 DW of traditional kombucha tea (TKT) or modified kombucha tea (MKT) (sweetened black tea fermented with Gluconoacetobacter sp. strain A4) or 60 mg Kg−1 DW of D-saccharic acid-1,4-lactone (DSL) for 12 weeks. The antioxidant activity, measured as total antioxidant capacity (TAOC), superoxide dismutase (SOD) and malonaldehyde (MDA) was assessed in the serum after the end of the 12 weeks of the treatment. The hypocholesterolemic diet resulted in a statistically significant decrease in TAOC and SOD and an increase in MDA values. These values were restored when the HCD was supplemented with TKT, MKT, or DSL. Vazquez-Cabral et al. [202] measured the antioxidant activities of kombucha and a kombucha analog (KAO) made of Quercus resinosa leaves against the oxidative damage caused by H2O2 in activated THP-1 human monocyte cells and reported the capacity of KAO to decrease oxidative stress. Finally, Gaggia et al. [49] studied the capacity of kombucha made from Aspalathus linearis leaves, fermented for 7 and 14 days, to decrease oxidative stress in L929 mouse fibroblasts caused by H2O2. When the treatment with the kombucha preceded the H2O2 application, the cell viability was partially restored by both products. When the treatment with the kombucha followed the application of H2O2, only the kombucha fermented for 14 days was able to restore the viability of the cells.
The antioxidant capacity of kombucha has also been reported to result in hypocholesterolemic and antidiabetic effects. Indeed, kombucha administration has been reported to decrease total and LDL cholesterol in rabbits and mice fed a high-cholesterol diet, as well as attenuate histological effects, such as lesions in the intima [30,34,201,202,203]. As far as the antidiabetic effect is concerned, this is attributed not only to the antioxidant capacity of kombucha, which addresses the oxidative stress caused by diabetes, but also to the reduction in blood glucose and the increase in plasma insulin, which have been reported as the effects of kombucha administration in experimental rats with induced diabetic consequences [31,33,204,205].
The antiproliferative capacity of kombucha has been demonstrated in vitro using human cancer cell lines. More precisely, the cytotoxicity against lung carcinoma (A549), osteosarcoma (U2OS), renal carcinoma (786-O), ileocecal colorectal adenocarcinoma (HCT8), colorectal adenocarcinoma (CACO-2), rhabdomyosarcoma (RD), cervix carcinoma (Hep2c), prostate cancer (PC-3), colon cancer (HCT-116), breast cancer (MCF-7) and a murine fibroblast (L2OB) has been exhibited [32,35,206,207]. Jayabalan et al. [206] proposed that Dimethyl 2-(2-hydroxy-2-methoxypropylidene) malonate and vitexin may contribute to these cytotoxic effects. Cardoso et al. [35] attributed the higher antiproliferative capacity of green-tea kombucha, compared to that of black-tea kombucha, to the presence of higher concentrations of catechins and verbascoside, the antitumor activity of which has already been reported [208,209]. However, not all cell lines were affected by black-tea kombucha [210]. In addition, Srihari et al. [211] reported that the survival of the prostate cancer cell line (PC-3) decreased after treatment with lyophilized kombucha extract, most likely due to the downregulation of the angiogenesis-associated genes HIF-1α, VEGF, IL-8, COX-2, MMP-2, and MMP-9. Despite these promising results, clinical studies are still lacking.
Strong indications of the immunomodulatory capacity of kombucha have been reported. More precisely, black-tea kombucha administration in male Swiss albino mice with indomethacin-induced stomach ulceration resulted in effective healing, which was attributed to the antioxidant activity and the reduction of gastric acid secretion [28]. The delay in the onset and severity of experimental autoimmune encephalomyelitis induced in female C57BL/6 mice through black-tea kombucha administration was reported by Marzban et al. [212]. In addition, the suppression of TNF-α and IL-6 levels in lipopolysaccharide-stimulated macrophages, as well as the in vitro inhibition of 5-LOX enzyme activity by black-tea kombucha extract, has also been reported [32,201].
As far as the antihypertensive activity is concerned, this is indicated by the detection of ACE inhibitory capacity. Certain flavonoids [213] with certain structural features [214] have an excellent antihypertensive capacity. The ACE inhibitory activity of green- and black-tea kombucha, as well as a series of analogues, has been reported [37,215].
The hepatoprotective activity of kombucha has been repeatedly exhibited in animal models. Indeed, the protective effect of black-tea kombucha against tertiary butyl hydroperoxide-induced cytotoxicity in murine hepatocytes of male albino Swiss mice, by reducing ROS generation, as well as through the inhibition of glutathione depletion and the attenuation of malonaldehyde levels, was reported by Bhattacharya et al. [216]. Abshenas et al. [29] induced hepatotoxicity in male Balb/c mice through acetaminophen treatment. Kombucha consumption for 7 days before acetaminophen treatment reduced its toxicity through the reduction of the serum aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and alkaline phosphatase levels. In addition, the decrease of histopathological changes, such as hepatocellular glycogen storage degeneration and necrosis, mononuclear cell infiltration in the portal area, dilation of central veins and capillarization, was also reported. Acetaminophen was also used by Wang et al. [160] to induce hepatotoxicity in male ICR mice. The administration of traditional black-tea kombucha as well as kombucha fermented only by Gluconoacetobacter sp. strain A4 effectively inhibited the increase of alanine aminotransferase, alkaline phosphatase, triglyceride and malondialdehyde, which were induced by acetaminophen treatment. These positive effects were largely attributed to the D-saccharic acid-1,4-lactone produced by the bacterial strain. Kabiri et al. [217] induced hepatotoxicity in male Wistar rats with thioacetamide and studied the effect of black-tea kombucha. Administration of kombucha for 3 weeks before TAA or after TAA treatment had all biochemical parameters assessed (alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, lactate dehydrogenase, total, LDL and HDL cholesterol, triglycerides, and bilirubin) at comparable levels to the control group, accompanied by normal histology. Hyun et al. [218] induced hepatic steatosis in male C57BLKS and C57BLKS db/db mice through a methionine/choline-deficient diet. Black-tea kombucha administration was reported to reduce the liver weight/body weight ratio to control-group levels through the reduction of fatty acids uptake and triglyceride synthesis, which were also indicated through the application of reverse-transcription quantitative PCR.
The antimicrobial activity of kombucha, as well as a series of analogues, has been extensively assessed. The methods of choice are diffusion and microdilution. The inhibition of the growth of a series of microorganisms, including molds such as Aspergillus flavus and As. niger; yeasts such as Candida albicans, Ca. glabrata, Ca. krusei, and Ca. tropicalis; Gram-positive bacteria such as Alicyclobacillus acidoterrestris, Bacillus cereus, Listeria monocytogenes, Micrococcus luteus, Staphylococcus aureus, and St. epidermidis; and Gram-negative bacteria such as Aeromonas hydrophila, Agrobacterium tumefaciens, Campylobacter jejuni, Enterobacter cloacae, Escherichia coli, Esch. coli O157:H7, Haemophilus influenzae, Klebsiella pneumoniae, Proteus mirabilis, Pr. vulgaris, Pseudomonas aeruginosa, Salmonella Enteritidis, Sa. Typhi, Sa. Typhimurium, Shigella dysenteriae, Sh. sonnei, Vibrio cholerae, and Yersinia enterocolitica has been reported [35,36,207,219,220,221,222,223,224].
The ingredients to which this antimicrobial activity has been primarily attributed are organic acids, mainly acetic acid, as well as a series of compounds with antimicrobial capacity, such as catechins, unsaturated lactones, hydroxylactones and verbascoside [225,226,227]. Qualitative and quantitative differences between different types of kombucha have also been attributed to differences in their chemical composition [35,220,224].
In summary, kombucha seems to possess a wealth of functional properties, which have been assessed either in vitro or using animal models. Further study is still necessary, including clinical trials [228].

3.3. Functional Properties of Kefir

Kefir is another thoroughly studied non-alcoholic beverage. As in the case of kombucha, the limit of 0.5% ABV is not always respected [229], and the production of kefir with as much as 10% ethanol has been reported [230]. Many functional properties have been described for kefir and attributed to its amino acid and peptide content. The type of milk, the proteolytic capacity of the micro-consortium that drives fermentation, and the storage time [231] greatly affect these properties. The most important kefir activities that play an essential role in its functional properties are the antioxidant and anti-inflammatory ones. The first is attributed to a series of enzymatic and non-enzymatic systems. Catalase, superoxide dismutase, and glutathione peroxidase account for the enzymatic ones, while vitamin E and β-carotene—along with peptides resulting from the proteolytic breakdown of casein and amino acids, especially methionine, lysine and tryptophan, which seem to possess higher antioxidant activity than threonine, serine, alanine, valine, isoleucine and phenylalanine—account for the non-enzymatic ones [232,233,234,235]. The anti-inflammatory activity of kefir is evidenced through the reduction of the levels of proinflammatory mediators, such as TNF-a and IL-6β, and the increase of the levels of anti-inflammatory cytokines such as IL-10 [236,237,238,239,240,241,242]. The bacteria themselves [238], their extracellular vesicles (as in the case of Lactobacillus kefiranofaciens subsp. kefirgranum PRCC-1301 [243]), their micro integral membrane protein (such as the one of Lactiplantibacillus plantarum [244]) as well as kefir peptides [44,245] seem to contribute to this activity.
The association between kefir consumption and effective glycemic control through reduction of IL-1β and increase of IL-10 expression, as well as a reduction of fasting glucose and insulin levels and reduction of insulin resistance, has been reported [39,40,239,246,247]. Although the exact mode of action is still to be elucidated, it includes the release of bioactive peptides from caseins, most likely through the proteolytic activity of the kefir microbiota [247]. These bioactive peptides may also include ACE-inhibitory ones [38,248], which contribute to the effective control of blood pressure. Indeed, a series of studies have highlighted the antihypertensive capacity of kefir and indicated that ACE-inhibitory activity is among the most important mechanisms [249,250,251]. Kefir has also been reported to have an immunomodulatory capacity [43] and act towards the prevention and treatment of atherosclerosis [44] and liver damage [41], mostly due to its antioxidant and anti-inflammatory activities. In addition, it may also act as a psychobiotic due to the occurrence of LAB capable of producing gamma-aminobutyric acid, a major inhibitory neurotransmitter of the mammalian central nervous system [252], combined with anti-inflammatory activities [253,254].
The anti-carcinogenic capacity of kefir has been assessed in a variety of human cancer cell lines, such as the gastric AGS and SGC7901, mammary MCF-7, myeloid leukemia HL60/AR, colorectal Caco-2 and HT-29, etc. [255,256,257,258,259]. In the majority of cases, inhibition of proliferation and apoptosis induction were reported, mediated by molecular mechanisms that included downregulation of TGF-α and Bcl-2, decreased polarization of mitochondrial membrane potential, as well as the upregulation of TGF-β1, Bax, caspase 3, caspase 8, caspase 9, etc. [42,256,257,258,260,261,262].

4. Customization of Fermented Beverages

In the previous paragraphs, the importance of raw materials in terms of their chemical composition, as well as the importance of the microbiota involved in fermentation in terms of metabolic capacity, have been highlighted. The functional properties of fermented beverages are achieved through the combination of these two parameters.
Product customization is as old as fermentation itself. The use of starter cultures is a form of customization, which, before the dawn of microbiology, took place through back-slopping. Nowadays, customization is facilitated by a wealth of information regarding the molecular mechanisms of disease and the mode of action of bioactive compounds. The customization of fermented beverages aims at achieving reproducibility, and therefore, the standardization of the product, and enhancing its functional properties. The first is principally achieved through the use of starter cultures. Indeed, the use of defined monocultures or micro-consortia allows the acceleration of the fermentation procedure, as well as the predictability of the outcome [263]. On the other hand, the enhancement of functional properties has been achieved mostly through the use of alternative and/or supplementary raw materials. In Table 2, representative studies on the customization of fermented beverages are exhibited.
The employment of alternative raw materials, as partial or complete replacements of traditional ones, has been extensively exercised in the case of kefir and kombucha beverages. In fact, this strategy has been so extensively employed that it has led to a whole new class of products, namely kefir analogs and kombucha analogs. In both cases, the aim was to meet the consumer needs of nutritionally dense and organoleptically appealing products. Especially the latter seems quite a challenge, taking into consideration the varying taste preferences [274]. Therefore, a variety of raw materials have been employed, resulting in variable nutritional and sensorial outcomes. In the case of kefir analogs, the utilization of fruits, vegetables, and sugar solutions is most commonly reported [275,276,277,278,279,280,281,282], while for the production of kombucha analogs, the employment of herbal infusions and fruits is most frequently encountered [283,284,285,286,287,288,289,290,291,292,293,294,295,296,297]. Apart from using kefir or kombucha cultures to ferment their analogs, the development of fruit- and vegetable-based beverages through the fermentation of substrates not traditionally considered for that purpose, using cultures that could effectively carry out fermentation and enhance their functional potential, has been extensively assessed. Indeed, a series of fruit or vegetable juices and pulps have been subjected to fermentation, principally lactic acid fermentation [298,299,300,301,302,303,304,305,306,307,308]. Apart from the organoleptically interesting products, in the majority of cases, the results exhibited an increase in the concentration of bioactive compounds, such as the total phenolic content, vitamin C, shikimic acid, etc., along with an increase in the antioxidant capacity, which, in some cases, was further verified through in vitro experimentation.
The enhancement of functional properties has also been achieved through the direct addition of compounds or their precursor molecules. An example of the first strategy is the study by Frolova et al. [309], in which pre-dissolved inulin and a vitamin premix consisting of thiamine, riboflavin, pyridoxine, folic acid and niacin were added to the already fermented black-tea kombucha at concentrations corresponding to 100% of the recommended daily intake (RDI) of inulin and 29–44% of the RDI of the vitamins. In addition, an infusion of frozen strawberries and lime leaves was created and added after the primary fermentation, and a secondary one was allowed, at 23 °C, for 24 h. The final product exhibited a 82% DPPH inhibitory activity and it was highly accepted by the sensory evaluation panel. Another example of the direct addition of functional compounds is the study by Shahbazi et al. [292]. In that study, medicinal plants, namely cinnamon, cardamom, and shirazi thyme, were added to the green-tea concoction and allowed to ferment using the same SCOBY. The cinnamon-flavored kombucha exhibited higher antioxidant and antimicrobial activity, as well as better sensorial scores, than the green-tea kombucha that served as the control and the cardamom- and shirazi thyme-flavored ones. Based on these results, cinnamon was used to partially or completely replace green tea. Increasing the cinnamon concentration resulted in an increased total phenolic content and radical scavenging activity and had a variable effect on the minimum inhibitory concentration against the Gram-positive and Gram-negative pathogenic bacteria that were examined; the authors noted that the Gram-negative ones seemed to be more susceptible. Similarly, Ozturk et al. [269] combined black tea leaves with hops (Humulus lupulus L.), madimak (Polygonum cognatum), or hawthorn (Crataegus monogyna) dry leaves and created a concoction that was left to ferment into kombucha. The herbs employed had no additive effect on the antioxidant capacity of the black-tea kombucha that served as the control and all products had a comparable antiproliferative activity against the human colorectal carcinoma cell line HCT116 and the human hepatocellular carcinoma cell line Mahlavu. Both studies, along with many similar ones, can be considered as a non-targeted attempt to improve the functionality of the final product. The term ‘non-targeted’ is used to highlight that the aim of the studies was to enhance the total phenolic content, and therefore, the antioxidant capacity, and not the concentration of a specific compound. In the case of kombucha, an example of such a compound would be epigallocatechin gallate (ECGC), the most studied bioactive compound of green tea. There is a strong indication that ECGC exhibits significant antiproliferative and antihypertensive activity through a variety of mechanisms [310,311]. Therefore, targeting the increase in this compound would create a product with specific capacities. However, research is still necessary in order to verify these actions and elucidate the underlying molecular mechanisms. This targeted approach was employed by Moslemi et al. [267], aiming to enhance the conjugated linoleic acid (CLA) concentration of a whey-based beverage. CLA is a group of linoleic acid isomers, the consumption of which has been correlated with a series of health benefits [312,313]. In that study, walnut oil that was already lipolyzed by endogenous lipases was added to a whey-based formulation, homogenized, and allowed to ferment with commercially available starter and probiotic cultures. The lipolysis of the walnut oil was necessary in order to liberate the esterified linoleic acid and thus enable the microorganisms to use it as a precursor for conjugated linoleic acid synthesis. Although the maximum produced amount of 36 mg/g of fat does not meet the recommended daily intake, this study proved that supplementation with substrates used by microorganisms for the production of bioactive compounds is an effective strategy and definitely worth further assessment.
Finally, the valorization of market surplus food, especially bread, into fermented beverages with functional potential has also been considered. Indeed, Massa et al. [314] reported the development of a non-alcoholic beverage using Saccharomyces bayanus 995, a SCOBY, or water kefir grains. The authors proposed a saccharification pre-treatment with Aspergillus oryzae and the supplementation of the thermally treated infusion with 1% w/v multiflora honey. The final product was sensorially evaluated, and the beverage prepared with S. bayanus was more preferred. On the other hand, Nguyen et al. [315] inoculated a sterilized slurry made after the homogenization of finely cut bread and water with La. rhamnosus GG and/or S. cerevisiae CNCM I-3856. Before sterilization, the addition of commercially available zero-calorie sweetener mix and a stabilizer took place. After fermentation at 37 °C for 72 h, the beverage fermented with a consortium of both strains contained the highest amount of amino acids, such as leucine, valine, glycine and GABA, throughout storage at 5 and 30 °C for 6 w. More recently, Siguenza-Andres et al. [316] applied desalting and treatment with a-amylase and glucoamylase to dried and milled surplus bread before inoculation with La. rhamnosus GG or a microconsortium consisting of Bifidobacterium sp., Lb. delbrueckii subsp. bulgaricus, and Streptococcus thermophilus. Fermentation took place at 38 °C for 24 h. The authors reported that the enzyme treatment allowed faster acidification to occur, whereas desalting restricted the maximum rates of growth, pH reduction and acidification.

5. Conclusions

Fermented beverages have a long tradition and a very promising future due to their great capabilities, spanning from their capacity to fit into the mentality of subsequent generations, including the current ‘on the go’ generation, to their customization potential. Especially regarding the latter, the wide range of raw materials that can be used, combined with the metabolic potential of food-grade microorganisms, can give birth to customized products that meet the extensive range of organoleptic preferences and enable targeted nutritional interventions. Although a lot of research is still necessary, this exciting future seems to be within reach.

Author Contributions

Conceptualization, S.P. and M.D.; investigation, S.P., M.D., J.K.P. and Y.K.; resources, S.P., M.D., J.K.P. and Y.K.; data curation, S.P., M.D., J.K.P. and Y.K.; writing—original draft preparation, S.P., M.D., J.K.P. and Y.K.; writing—review and editing, S.P., M.D., J.K.P. and Y.K. 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

The data presented in this study are available in the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sefa-Dedeh, S.; Sanni, A.I.; Tetteh, G.; Sakyi-Dawson, E. Yeasts in the traditional brewing of pito in Ghana. World J. Microbiol. Biotechnol. 1999, 15, 593–597. [Google Scholar] [CrossRef]
  2. Wacher-Rodarte, C.; Canas, A.; Barzana, E.; Lappe, P.; Ulloa, M.; Owens, J.D. Microbiology of Indian and Mestizo pozol fermentations. Food Microbiol. 2000, 17, 251–256. [Google Scholar] [CrossRef]
  3. Van der Aa Kuhle, A.; Jespersen, L.; Glover, R.L.; Diawara, B.; Jakobsen, M. Identification and characterization of Saccharomyces cerevisiae strains isolated from West African sorghum beer. Yeast 2001, 18, 1069–1079. [Google Scholar] [CrossRef]
  4. Osimani, A.; Garofalo, C.; Aquilanti, L.; Milanovic, V.; Clementi, F. Unpasteurised commercial boza as a source of microbial diversity. Int. J. Food Microbiol. 2015, 194, 62–70. [Google Scholar] [CrossRef] [PubMed]
  5. Binitu Worku, B.; Gemede, H.F.; Woldegiorgis, A.Z. Nutritional and alcoholic contents of cheka: A traditional fermented beverage in Southwestern Ethiopia. Food Sci. Nutr. 2018, 6, 2466–2472. [Google Scholar] [CrossRef]
  6. Wang, P.; Wu, J.; Wang, T.; Zhang, Y.; Yao, X.; Li, J.; Wang, X.; Lu, X. Fermentation process optimization, chemical analysis, and storage stability evaluation of a probiotic barley malt kvass. Bioprocess Biosyst. Eng. 2022, 45, 1175–1188. [Google Scholar] [CrossRef]
  7. Nyanga, L.K.; Nout, M.J.R.; Gadaga, T.H.; Theelen, B.; Boekhout, T.; Zwietering, M.H. Yeasts and lactic acid bacteria microbiota from masau (Ziziphus mauritiana) fruits and their fermented fruit pulp in Zimbabwe. Int. J. Food Microbiol. 2007, 120, 159–166. [Google Scholar] [CrossRef] [PubMed]
  8. Valles, B.S.; Bedrinana, R.P.; Tascon, N.F.; Simon, A.Q.; Madrera, R.R. Yeast species associated with the spontaneous fermentation of cider. Food Microbiol. 2007, 24, 25–31. [Google Scholar] [CrossRef]
  9. Duarte, W.F.; Dias, D.R.; de Melo Pereira, G.V.; Gervásio, I.M.; Schwan, R.F. Indigenous and inoculated yeast fermentation of gabiroba (Campomanesia pubescens) pulp for fruit wine production. J. Ind. Microbiol. Biotechnol. 2009, 36, 557–569. [Google Scholar] [CrossRef]
  10. Stringini, M.; Comitini, F.; Taccari, M.; Ciani, M. Yeast diversity during tapping and fermentation of palm wine from Cameroon. Food Microbiol. 2009, 26, 415–420. [Google Scholar] [CrossRef]
  11. Sagdic, O.; Ozturk, I.; Yapar, N.; Yetim, H. Diversity and probiotic potentials of lactic acid bacteria isolated from Gilaburu, a traditional Turkish fermented European cranberrybush (Viburnum opulus L.) fruit drink. Food Res. Int. 2014, 64, 537–545. [Google Scholar] [CrossRef] [PubMed]
  12. Marsh, A.J.; O’Sullivan, O.; Hill, C.; Ross, R.P.; Cotter, P.D. Sequence-based analysis of the bacterial and fungal compositions of multiple kombucha (tea fungus) samples. Food Microbiol. 2013, 38, 171–178. [Google Scholar] [CrossRef] [PubMed]
  13. Ramadani, A.S.; Abulreesh, H.H. Isolation and identification of yeast flora in local kombucha sample: Al Nabtah. J. App. Sci. 2010, 2, 42–51. [Google Scholar]
  14. 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] [PubMed]
  15. Levantesi, G.; Marfisi, R.; Mozaffarian, D.; Franzosi, M.G.; Maggioni, A.; Nicolosi, G.L.; Schweiger, C.; Silletta, M.; Tavazzi, L.; Tognoni, G.; et al. Wine consumption and risk of cardiovascular events after myocardial infarction: Results from the GISSI-Prevenzione trial. Int. J. Cardiol. 2013, 163, 282–287. [Google Scholar] [CrossRef] [PubMed]
  16. Chiva-Blanch, G.; Badimon, L. Benefits and risks of moderate alcohol consumption on cardiovascular disease: Current findings and controversies. Nutrients 2020, 12, 108. [Google Scholar] [CrossRef]
  17. Luceron-Lucas-Torres, M.; Cavero-Redondo, I.; Martinez-Vizcaino, V.; Saz-Lara, A.; Pascual-Morena, C.; Alvarez-Bueno, C. Association between wine consumption and cognitive decline in older people: A systematic review and meta-analysis of longitudinal studies. Front. Nutr. 2022, 9, 863059. [Google Scholar] [CrossRef]
  18. Restani, P.; Fradera, U.; Ruf, J.-C.; Stockley, C.; Teissedre, P.L.; Biella, S.; Colombo, F.; Di Lorenzo, C. Grapes and their derivatives in modulation of cognitive decline: A critical review of epidemiological and randomized-controlled trials in humans. Crit. Rev. Food Sci. Nutr. 2021, 61, 566–576. [Google Scholar] [CrossRef]
  19. Vazquez-Ruiz, Z.; Martinez-Gonzalez, M.A.; Vitelli-Storelli, F.; Bes-Rastrollo, M.; Basterra-Gortari, F.J.; Toledo, E. Effect of dietary phenolic compounds on incidence of type 2 diabetes in the “Seguimiento Universidad de Navarra” (SUN) cohort. Antioxidants 2023, 12, 507. [Google Scholar] [CrossRef]
  20. Baliunas, D.O.; Taylor, B.J.; Irving, H.; Roerecke, M.; Patra, J.; Mohapatra, S.; Rehm, J. Alcohol as a risk factor for type 2 diabetes: A systematic review and meta-analysis. Diabetes Care 2009, 32, 2123–2132. [Google Scholar] [CrossRef]
  21. Karatzi, K.N.; Papamichael, C.M.; Karatzis, E.N.; Papaioannou, T.G.; Aznaouridis, K.A.; Katsichti, P.P.; Stamatelopoulos, K.S.; Zampelas, A.; Lekakis, J.P.; Mavrikakis, M.E. Red wine acutely induces favorable effects on wave reflections and central pressures in coronary artery disease patients. Am. J. Hypertens. 2005, 18, 1161–1167. [Google Scholar] [CrossRef]
  22. Chiva-Blanch, G.; Urpi-Sarda, M.; Ros, E.; Arranz, S.; Valderas-Martinez, P.; Casas, R.; Sacanella, E.; Llorach, R.; Lamuela-Raventos, R.M.; Andres-Lacueva, C.; et al. Dealcoholized red wine decreases systolic and diastolic blood pressure and increases plasma nitric oxide. Circ. Res. 2012, 111, 1065–1068. [Google Scholar] [CrossRef]
  23. Fantin, F.; Bulpitt, C.J.; Zamboni, M.; Cheek, E.; Rajkumar, C. Arterial compliance may be reduced by ingestion of red wine. J. Hum. Hypertens. 2015, 30, 68–72. [Google Scholar] [CrossRef] [PubMed]
  24. Queipo-Ortuno, M.I.; Boto-Ordonez, M.; Murri, M.; Gomez-Zumaquero, J.M.; Clemente-Postigo, M.; Estruch, R.; Cardona Diaz, F.; Andres-Lacueva, C.; Tinahones, F.J. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am. J. Clin. Nutr. 2012, 95, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
  25. Kasicka-Jonderko, A.; Jonderko, K.; Gajek, E.; Piekielniak, A.; Zawislan, R. Sluggish gallbladder emptying and gastrointestinal transit after intake of common alcoholic beverages. J. Physiol. Pharmacol. 2014, 65, 55–66. [Google Scholar]
  26. Moreno-Indias, I.; Sanchez-Alcoholado, L.; Perez-Martinez, P.; Andres-Lacueva, C.; Cardona, F.; Tinahones, F.; Queipo-Ortuno, 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]
  27. Gonzalez-Lopez-Arza, M.V.; Trivino-Palomo, J.V.; Montanero-Fernandez, J.; Garrido-Ardila, E.M.; Gonzalez-Sanchez, B.; Jimenez-Palomares, M.; Rodriguez-Mansilla, J. Benefits of the light consumption of red wine in pain, tender points, and anxiety in women with fibromyalgia: A pilot study. Nutrients 2023, 15, 3469. [Google Scholar] [CrossRef] [PubMed]
  28. Banerjee, D.; Hassarajani, S.A.; Maity, B.; Narayan, G.; Bandyopadhyay, S.K.; Chattopadhyay, S. Comparative healing property of kombucha tea and black tea against indomethacin-induced gastric ulceration in mice: Possible mechanism of action. Food Funct. 2010, 1, 284–293. [Google Scholar] [CrossRef]
  29. Abshenas, J.; Derakhshanfar, A.; Ferdosi, M.H.; Hasanzadeh, S. Protective effect of kombucha tea against acetaminophen-induced hepatotoxicity in mice: A biochemical and histopathological study. Comp. Clin. Pathol. 2012, 21, 1243–1248. [Google Scholar] [CrossRef]
  30. Bellassoued, K.; Ghrab, F.; Makni-Ayadi, F.; van Pelt, J.; Elfeki, A.; Ammar, E. Protective effect of kombucha on rats fed a hypercholesterolemic diet is mediated by its antioxidant activity. Pharm. Biol. 2015, 53, 1699–1709. [Google Scholar] [CrossRef]
  31. Hosseini, S.; Gorjian, M.; Rasouli, L.; Shirali, S. A comparison between the effect of green tea and kombucha prepared from green tea on the weight of diabetic rats. Biosci. Biotechnol. Res. Asia 2015, 12, 141–146. [Google Scholar] [CrossRef]
  32. Villarreal-Soto, S.A.; Beaufort, S.; Bouajila, J.; Souchard, J.P.; Renard, T.; Rollan, S.; Taillandier, P. Impact of fermentation conditions on the production of bioactive compounds with anticancer, anti-inflammatory and antioxidant properties in kombucha tea extracts. Process Biochem. 2019, 83, 44–54. [Google Scholar] [CrossRef]
  33. Zubaidah, E.; Afagni, C.A.; Kalsum, U.; Srianta, I.; Blanc, P.J. Comparison of in vivo antidiabetes activity of snake fruit kombucha, black tea kombucha and metformin. Biocatal. Agric. Biotechnol. 2019, 17, 465–469. [Google Scholar] [CrossRef]
  34. Alaei, Z.; Doudi, M.; Setorki, M. The protective role of kombucha extract on the normal intestinal microflora, high-cholesterol diet caused hypercholesterolemia, and histological structures changes in New Zealand white rabbits. Avicenna J. Phytomed. 2020, 10, 604–614. [Google Scholar] [PubMed]
  35. Cardoso, R.R.; Neto, R.O.; D’Almeida, C.T.S.; Nascimento, T.P.; Pressete, C.G.; Azevedo, L.; Martino, H.S.D.; Cameron, L.C.; Ferreira, M.S.L.; Barros, F.A.R. Kombuchas from green and black teas have different phenolic profile, which impacts their antioxidant capacities, antibacterial and antiproliferative activities. Food Res. Int. 2020, 128, 108782. [Google Scholar] [CrossRef]
  36. Mizuta, A.G.; de Menezes, J.L.; Dutra, T.V.; Ferreira, T.V.; Castro, J.C.; da Silva, C.A.J.; Pilau, E.J.; Junior, M.M.; Filho, A.A.A. Evaluation of antimicrobial activity of green tea kombucha at two fermentation time points against Alicyclobacillus spp. LWT-Food Sci. Technol. 2020, 130, 109641. [Google Scholar] [CrossRef]
  37. Vitas, J.; Vukmanovic, S.; Cakarevic, J.; Popovic, L.; Malbasa, R. Kombucha fermentation of six medicinal herbs: Chemical profile and biological activity. Chem. Ind. Chem. Eng. Q. 2020, 26, 157–170. [Google Scholar] [CrossRef]
  38. Ebner, J.; Asçi Arslan, 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]
  39. Golunuk, S.B.; Oztasan, N.; Sozen, H.; Koca, H.B. Effects of traditional fermented beverages on some blood parameters in aerobic exercises. Biomed. Res. 2017, 28, 9475–9480. [Google Scholar]
  40. 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]
  41. Golli-Bennour, E.E.; Timoumi, R.; Annaibi, E.; Mokni, M.; Omezzine, A.; Bacha, H.; Abid-Essefi, S. Protective effects of kefir against deltamethrin-induced hepatotoxicity in rats. Environ. Sci. Pollut. Res. Int. 2019, 26, 18856–18865. [Google Scholar] [CrossRef]
  42. 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]
  43. Praznikar, Z.J.; Kenig, S.; Vardjan, T.; Cernelic-Bizjak, M.C.; Petelin, A. Effects of kefir or milk supplementation on zonulin in overweight subjects. J. Dairy Sci. 2020, 103, 3961–3970. [Google Scholar] [CrossRef] [PubMed]
  44. Tung, M.C.; Lan, Y.W.; Li, H.H.; Chen, H.L.; Chen, S.Y.; Chen, Y.H.; Lin, C.C.; Tu, M.Y.; Chen, C.M. Kefir peptides alleviate high-fat diet-induced atherosclerosis by attenuating macrophage accumulation and oxidative stress in ApoE knockout mice. Sci. Rep. 2020, 10, 8802. [Google Scholar] [CrossRef]
  45. Paramithiotis, S.; Stasinou, V.; Tzamourani, A.; Kotseridis, Y.; Dimopoulou, M. Malolactic fermentation-theoretical advances and practical considerations. Fermentation 2022, 8, 521. [Google Scholar] [CrossRef]
  46. Belloso-Morales, G.; Hernandez-Sanchez, H. Manufacture of a beverage from cheese whey using a tea fungus fermentation. Rev. Latinoam. Microbiol. 2003, 45, 5. [Google Scholar]
  47. Amarasinghe, H.; Weerakkody, N.; Waisundara, V.Y. Evaluation of physicochemical properties and antioxidant activities of kombucha “tea fungus” during extended periods of fermentation. Food Sci. Nutr. 2018, 6, 659–665. [Google Scholar] [CrossRef] [PubMed]
  48. Barbosa, C.D.; Uetanabaro, A.P.T.; Santos, W.C.R.; Caetano, R.G.; Albano, H.; Kato, R.; Cosenza, G.P.; Azeredo, A.; Goes-Neto, A.; Rosa, C.A.; et al. Microbial–physicochemical integrated analysis of kombucha fermentation. LWT-Food Sci. Tehnol. 2021, 148, 111788. [Google Scholar] [CrossRef]
  49. Gaggia, F.; Baffoni, L.; Galiano, M.; Nielsen, D.S.; Jakobsen, R.R.; Castro-Mejía, J.L.; Bosi, S.; Truzzi, F.; Musumeci, F.; Dinelli, G.; et al. Kombucha beverage from green, black and rooibos teas: A comparative study looking at microbiology, chemistry and antioxidant activity. Nutrients 2019, 11, 1. [Google Scholar] [CrossRef]
  50. Teoh, A.L.; Heard, G.; Cox, J. Yeast ecology of kombucha fermentation. Int. J. Food Microbiol. 2004, 95, 119–126. [Google Scholar] [CrossRef]
  51. Coton, M.; Pawtowski, A.; Taminiau, B.; Burgaud, G.; Deniel, F.; Coulloumme-Labarthe, L.; Fall, A.; Daube, G.; Coton, E. Unraveling microbial ecology of industrial-scale kombucha fermentations by metabarcoding and culture-based methods. FEMS Microbiol. Ecol. 2017, 93, fix048. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, S.; Zhang, L.; Qi, L.; Liang, H.; Lin, X.; Li, S.; Yu, C.; Ji, C. Effect of synthetic microbial community on nutraceutical and sensory qualities of kombucha. Int. J. Food Sci. Technol. 2020, 55, 3327–3333. [Google Scholar] [CrossRef]
  53. Mayser, P.; Fromme, S.; Leitzmann, G.; Grunder, K. The yeast spectrum of the ‘tea fungus kombucha’. Mycoses 1995, 38, 289–295. [Google Scholar] [CrossRef]
  54. Huang, X.; Xin, Y.; Lu, T. A systematic, complexity-reduction approach to dissect the kombucha tea microbiome. eLife 2022, 11, e76401. [Google Scholar] [CrossRef]
  55. Angulo, L.; Lopez, E.; Lema, C. Microflora present in kefir grains of the Galician region (North-West of Spain). J. Dairy Res. 1993, 60, 263–267. [Google Scholar] [CrossRef] [PubMed]
  56. Wyder, M.-T.; Meile, L.; Teuber, M. Description of Saccharomyces turicensis sp. nov., a new species from kefyr. Syst. Appl. Microbiol. 1999, 22, 420–425. [Google Scholar] [CrossRef]
  57. Wyder, M.T.; Spillmann, H.; Puhan, Z. Investigation of the yeast flora in dairy products: A case study of kefyr. Food Technol. Biotechnol. 1997, 35, 299–304. [Google Scholar]
  58. Assadi, M.; Pourahmad, R.; Moazami, N. Use of isolated kefir starter cultures in kefir production. World J. Microbiol. Biotechnol. 2000, 16, 541–543. [Google Scholar] [CrossRef]
  59. Garrote, G.L.; Abraham, A.G.; De Antoni, G.L. Chemical and microbiological characterisation of kefir grains. J. Dairy Res. 2001, 68, 639–652. [Google Scholar] [CrossRef]
  60. Simova, E.; Beshkova, D.; Angelov, A.; Hristozova, T.; Frengova, G.; Spasov, Z. Lactic acid bacteria and yeasts in kefir grains and kefir made from them. J. Ind. Microbiol. Biotechnol. 2002, 28, 1–6. [Google Scholar] [CrossRef]
  61. Dobson, A.; O’Sullivan, O.; Cotter, P.D.; Ross, R.; Hill, C. High-throughput sequence-based analysis of the bacterial composition of kefir and an associated kefir grain. FEMS Microbiol. Lett. 2011, 320, 56–62. [Google Scholar] [CrossRef] [PubMed]
  62. Diosma, G.; Romanin, D.E.; Rey-Burusco, M.F.; Londero, A.; Garrote, G.L. Yeasts from kefir grains: Isolation, identification, and probiotic characterization. World J. Microbiol. Biotechnol. 2013, 30, 43–53. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, Z.; Shi, J.; Yang, X.; Nan, B.; Liu, Y.; Wang, Z. Chemical and physical characteristics and antioxidant activities of the exopolysaccharide produced by Tibetan kefir grains during milk fermentation. Int. Dairy J. 2015, 43, 15–21. [Google Scholar] [CrossRef]
  64. Londero, A.; Hamet, M.F.; De Antoni, G.L.; Garrote, G.L.; Abraham, A.G. Kefir grains as a starter for whey fermentation at different temperatures: Chemical and microbiological characterisation. J. Dairy Res. 2012, 79, 262–271. [Google Scholar] [CrossRef] [PubMed]
  65. Hamet, M.F.; Londero, A.; Medrano, M.; Vercammen, E.; Van Hoorde, K.; Garrote, G.L.; Huys, G.; Vandamme, P.; Abraham, A.G. Application of culture-dependent and culture-independent methods for the identification of Lactobacillus kefiranofaciens in microbial consortia present in kefir grains. Food Microbiol. 2013, 36, 327–334. [Google Scholar] [CrossRef]
  66. Korsak, N.; Taminiau, B.; Leclercq, M.; Nezer, C.; Crevecoeur, S.; Ferauche, C.; Detry, E.; Delcenserie, V.; Daube, G. Short communication: Evaluation of the microbiota of kefir samples using metagenetic analysis targeting the 16S and 26S ribosomal DNA fragments. J. Dairy Sci. 2015, 98, 3684–3689. [Google Scholar] [CrossRef] [PubMed]
  67. Miguel, M.G.C.P.; Cardoso, P.G.; Lago, L.A.; Schwan, R.F. Diversity of bacteria present in milk kefir grains using culture-dependent and culture-independent methods. Food Res. Int. 2010, 43, 1523–1528. [Google Scholar] [CrossRef]
  68. Leite, A.M.O.; Mayoa, B.; Rachid, C.T.; Peixoto, R.S.; Silva, J.T.; Paschoalin, V.M.F.; Delgado, S. Assessment of the microbial diversity of Brazilian kefir grains by PCR-DGGE and pyrosequencing analysis. Food Microbiol. 2012, 31, 215–221. [Google Scholar] [CrossRef]
  69. Zanirati, D.F.; Abatemarco, M.; Cicco Sandesb, S.H.; Nicolia, J.R.; Nunes, A.C.; Neumanna, E. Selection of lactic acid bacteria from Brazilian kefir grains for potential use as starter or probiotic cultures. Anarobe 2015, 32, 70–76. [Google Scholar] [CrossRef]
  70. Magalhães, K.T.; Pereira, G.V.D.M.; Campos, C.R.; Dragone, G.; Schwan, R.F. Brazilian kefir: Structure, microbial communities and chemical composition. Braz. J. Microbiol. 2011, 42, 693–702. [Google Scholar] [CrossRef]
  71. Jianzhong, Z.; Xiaoli, L.; Hanhu, J.; Mingsheng, D. Analysis of the microflora in Tibetan kefir grains using denaturing gradient gel electrophoresis. Food Microbiol. 2009, 26, 770–775. [Google Scholar]
  72. 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, 425–433. [Google Scholar] [CrossRef] [PubMed]
  73. Gao, J.; Gu, F.; He, J.; Xiao, J.; Chen, Q.; Ruan, H.; He, G. Metagenome analysis of bacterial diversity in Tibetan kefir grains. Eur. Food Res. Technol. 2013, 236, 549–556. [Google Scholar] [CrossRef]
  74. Garofalo, C.; Osimani, A.; Milanovic, V.; Aquilanti, L.; De Filippis, F.; Stellato, G.; Di Mauro, S.; Turchetti, B.; Buzzini, P.; Ercolini, D.; et al. Bacteria and yeast microbiota in milk kefir grains from different Italian regions. Food Microbiol. 2015, 49, 123–133. [Google Scholar] [CrossRef] [PubMed]
  75. Yang, X.J.; Fan, M.T.; Shi, J.L.; Dang, B. Isolation and identification of preponderant flora in Tibetan kefir. China Brew. 2007, 171, 52–55. [Google Scholar]
  76. Witthuhn, R.C.; Schoeman, T.; Britz, T.J. Characterisation of the microbial population at different stages of kefir production and kefir grain mass cultivation. Int. Dairy J. 2005, 15, 383–389. [Google Scholar] [CrossRef]
  77. Guzel-Seydim, Z.; Wyffels, J.T.; Seydim, A.C.; Greene, A.K. Turkish kefir and kefir grains: Microbial enumeration and electron microscopic observation. Int. J. Dairy Technol. 2005, 58, 25–29. [Google Scholar] [CrossRef]
  78. Kesmen, Z.; Kacmaz, N. Determination of lactic microflora of kefir grains and kefir beverage by using culture-dependent and culture-independent methods. J. Food Sci. 2011, 76, 276–283. [Google Scholar] [CrossRef]
  79. Kok-Tas, T.; Ekinci, F.Y.; Guzele Seydim, Z.B. Identification of microbial flora in kefir grains produced in Turkey using PCR. Int. J. Dairy Technol. 2012, 65, 126–131. [Google Scholar]
  80. Nalbantoglu, U.; Cakar, A.; Dogan, H.; Abaci, N.; Ustek, D.; Sayood, K.; Can, H. Metagenomic analysis of the microbial community in kefir grains. Food Microbiol. 2014, 41, 42–51. [Google Scholar] [CrossRef]
  81. Yüksekdag, Z.N.; Beyatli, Y.; Aslim, B. Determination of some characteristic coccoid forms of lactic acid bacteria isolated from Turkish kefirs with natural probiotic. Food Sci. Technol. 2004, 37, 663–667. [Google Scholar] [CrossRef]
  82. Satora, P.; Tuszynski, T. Biodiversity of yeasts during plum Wegierka Zwykla spontaneous fermentation. Food Technol. Biotechnol. 2005, 43, 277–282. [Google Scholar]
  83. Chanprasartsuk, O.O.; Prakitchaiwattana, C.; Sanguandeekul, R.; Fleet, G.H. Autochthonous yeasts associated with mature pineapple fruits, freshly crushed juice and their ferments; and the chemical changes during natural fermentation. Bioresour. Technol. 2010, 101, 7500–7509. [Google Scholar] [CrossRef] [PubMed]
  84. Maragatham, C.; Panneerselvam, A. Isolation, identification and characterization of wine-yeast from rotten papaya fruits for wine production. Adv. Appl. Sci. Res. 2011, 2, 93–98. [Google Scholar]
  85. Padilla, B.; Garcia- Fernandez, D.; Gonzalez, B.; Izidoro, I.; Esteve-Zarzoso, B.; Beltran, G.; Mas, A. Yeast biodiversity from DOQ priorat uninoculated fermentations. Front. Microbiol. 2016, 7, 930. [Google Scholar] [CrossRef] [PubMed]
  86. Bougreau, M.; Ascencio, K.; Bugarel, M.; Nightingale, K.; Loneragan, G. Yeast species isolated from Texas High Plains vineyards and dynamics during spontaneous fermentations of Tempranillo grapes. PLoS ONE 2019, 14, e0216246. [Google Scholar] [CrossRef]
  87. Nyambane, B.; Thari, W.M.; Wangoh, J.; Njage, P.M.K. Lactic acid bacteria and yeasts involved in the fermentation of Amabere Amaruranu, a Kenyan fermented milk. Food Sci. Nutr. 2014, 2, 692–699. [Google Scholar] [CrossRef] [PubMed]
  88. Grijalva-Vallejos, N.; Aranda, A.; Matallana, E. Evaluation of yeasts from Ecuadorian Chicha by their performance as starters for alcoholic fermentations in the food industry. Int. J. Food Microbiol. 2020, 317, 108462. [Google Scholar] [CrossRef]
  89. Pérez-Cataluña, A.; Elizaquível, P.; Carrasco, P.; Espinosa, J.; Reyes, D.; Wacher, C.; Aznar, R. Diversity and dynamics of lactic acid bacteria in Atole Agrio, a traditional maize-based fermented beverage from South-Eastern Mexico, analysed by high throughput sequencing and culturing. Antonie Van Leeuwenhoek 2017, 111, 385–399. [Google Scholar] [CrossRef]
  90. Puerari, C.; Magalhães-Guedes, K.T.; Schwan, R.F. Bacaba beverage produced by Umutina Brazilian Amerindians: Microbiological and chemical characterization. Braz. J. Microbiol. 2015, 46, 1207–1216. [Google Scholar] [CrossRef]
  91. Tamang, J.; Thapa, S. Fermentation dynamics during production of Bhaati Jaanr, a traditional fermented rice beverage of the Eastern Himalayas. Food Biotechnol. 2006, 20, 251–261. [Google Scholar] [CrossRef]
  92. Maoura, N.; Mbaiguinam, M.; Nguyen, H.V.; Gaillardin, C.; Pourquie, J. Identification and typing of the yeast strains isolated from Bili Bili, a traditional sorghum beer of Chad. Afr. J. Biotechnol. 2005, 4, 646–656. [Google Scholar]
  93. Abegaz, K. Isolation, characterization and identification of lactic acid bacteria involved in traditional fermentation of Borde, an Ethiopian cereal beverage. Afr. J. Biotechnol. 2007, 6, 1469–1478. [Google Scholar]
  94. Hancioglu, O.; Karapinar, M. Microflora of Boza, a traditional fermented Turkish beverage. Int. J. Food Microbiol. 1997, 35, 271–274. [Google Scholar] [CrossRef] [PubMed]
  95. Moncheva, P.; Chipeva, V.; Kujumdzieva, A.; Ivanova, I.; Dousset, X.; Gocheva, B. The composition of the microflora of Boza, an original Bulgarian beverage. Biotechnol. Biotechnol. Equip. 2003, 17, 164–168. [Google Scholar] [CrossRef]
  96. Gotcheva, V.; Pandiella, S.S.; Angelov, A.; Roshkova, Z.; Webb, C. Monitoring the fermentation of the traditional Bulgarian beverage boza. Int. J. Food Sci. Technol. 2001, 36, 129–134. [Google Scholar] [CrossRef]
  97. Atter, A.; Obiri-Danso, K.; Amoa-Awua, W.K. Microbiological and chemical processes associated with the production of Burukutu a traditional beer in Ghana. Int. Food Res. J. 2014, 21, 1769–1776. [Google Scholar]
  98. Bassi, D.; Orru, L.; Vasquez, J.C.; Cocconcelli, P.S.; Fontana, C. Peruvian chicha: A focus on the microbial populations of this ancient maize-based fermented beverage. Microorganisms 2020, 8, 93. [Google Scholar] [CrossRef]
  99. Lyumugabe, L.; Kamaliza, G.; Bajyana, E.; Thonart, P. Microbiological and physico-chemical characteristics of Rwandese traditional beer “Ikigage”. Afr. J. Biotechnol. 2010, 9, 4241–4246. [Google Scholar]
  100. Pswarayi, F.; Ganzle, M.G. Composition and origin of the fermentation microbiota of Mahewu, a Zimbabwean fermented cereal beverage. Appl. Environ. Microbiol. 2019, 85, e03130-18. [Google Scholar] [CrossRef]
  101. ben Omar, N.; Ampe, F. Microbial community dynamics during production of the Mexican fermented maize dough pozol. Appl. Environ. Microbiol. 2000, 66, 3664–3673. [Google Scholar] [CrossRef] [PubMed]
  102. Escalante, A.; Wacher, C.; Farres, A. Lactic acid bacterial diversity in the traditional Mexican fermented dough pozol as determined by 16S rDNA sequence analysis. Int. J. Food Microbiol. 2001, 64, 21–31. [Google Scholar] [CrossRef] [PubMed]
  103. Estrada-Godina, A.R.; Cruz-Guerrero, A.E.; Lappe, P.; Ulloa, M.; Garcia-Garibay, M.; Gomez-Ruiz, L. Isolation and identification of killer yeasts from Agave sap (aguamiel) and pulque. World J. Microbiol. Biotechnol. 2001, 17, 557–560. [Google Scholar] [CrossRef]
  104. Lappe-Oliveras, P.; Moreno-Terrazas, R.; Arrizon-Gavino, J.; Herrera-Suarez, T.; Garcia-Mendoza, A.; Gschaedler-Mathis, A. Yeasts associated with the production of Mexican alcoholic nondistilled and distilled Agave beverages. FEMS Yeast Res. 2008, 8, 1037–1052. [Google Scholar] [CrossRef] [PubMed]
  105. Escalante, A.; Giles-Gomez, M.; Hernandez, G.; Cordova-Aguilar, M.; Lopez Munguia, A.; Gosset, G.; Bolivar, F. Analysis of bacterial community during the fermentation of pulque, a traditional Mexican alcoholic beverage, using a polyphasic approach. Int. J. Food Microbiol. 2008, 124, 126–134. [Google Scholar] [CrossRef] [PubMed]
  106. Escalante, A.; Rodriguez, M.E.; Martinez, A.; Lopez-Munguia, A.; Bolivar, F.; Gosset, G. Characterization of bacterial diversity in Pulque, a traditional Mexican alcoholic fermented beverage, as determined by 16S rDNA analysis. FEMS Microbiol. Lett. 2004, 235, 273–279. [Google Scholar] [CrossRef]
  107. Sikuten, I.; Stambuk, P.; Andabaka, Z.; Tomaz, I.; Markovic, Z.; Stupic, D.; Maletic, E.; Kontic, J.K.; Preiner, D. Grapevine as a rich source of polyphenolic compounds. Molecules 2020, 25, 5604. [Google Scholar] [CrossRef]
  108. Jackson, R.S. Wine Science, Principles and Applications, 2nd ed.; Academic Press: San Diego, CA, USA, 2000; pp. 270–332. [Google Scholar]
  109. Rentzsch, M.; Wilkens, A.; Winterhalter, P. Non-flavonoid phenolic compounds. In Wine Chemistry and Biochemistry; Moreno-Arribas, M.V., Polo, M.C., Eds.; Springer: New York, NY, USA, 2009; pp. 509–527. [Google Scholar]
  110. Fernandes, I.; Perez-Gregorio, R.; Soares, S.; Mateus, N.; Freitas, V. Wine flavonoids in health and disease prevention. Molecules 2017, 22, 292. [Google Scholar] [CrossRef]
  111. Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Remesy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81, 230–242. [Google Scholar] [CrossRef]
  112. Tian, X.J.; Yang, X.W.; Yang, X.; Wang, K. Studies of intestinal permeability of 36 flavonoids using Caco-2 cell monolayer model. Int. J. Pharm. 2009, 367, 58–64. [Google Scholar] [CrossRef]
  113. Xu, C.; Kong, L.; Tian, Y. Investigation of the phenolic component bioavailability using the in vitro digestion/Caco-2 cell model, as well as the antioxidant activity in Chinese red wine. Foods 2022, 11, 3108. [Google Scholar] [CrossRef] [PubMed]
  114. Poli, A.; Marangoni, F.; Avogaro, A.; Barba, G.; Bellentani, S.; Bucci, M.; Cambieri, R.; Catapano, A.L.; Costanzo, S.; Cricelli, C.; et al. Moderate alcohol use and health: A consensus document. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 487–504. [Google Scholar] [CrossRef] [PubMed]
  115. Hrelia, S.; Di Renzo, L.; Bavaresco, L.; Bernardi, E.; Malaguti, M.; Giacosa, A. Moderate wine consumption and health: A narrative review. Nutrients 2023, 15, 175. [Google Scholar] [CrossRef] [PubMed]
  116. Giacosa, A.; Barale, R.; Bavaresco, L.; Faliva, M.A.; Gerbi, V.; La Vecchia, C.; Negri, E.; Opizzi, A.; Perna, S.; Pezzotti, M.; et al. Mediterranean way of drinking and longevity. Crit. Rev. Food Sci. Nutr. 2016, 56, 635–640. [Google Scholar] [CrossRef]
  117. Poli, A. Is drinking wine in moderation good for health or not? Eur. Heart J. Suppl. 2022, 24, l119–l122. [Google Scholar] [CrossRef]
  118. Jani, B.D.; McQueenie, R.; Nicholl, B.I.; Field, R.; Hanlon, P.; Gallacher, K.I.; Mair, F.S.; Lewsey, J. Association between patterns of alcohol consumption (beverage type, frequency and consumption with food) and risk of adverse health outcomes: A prospective cohort study. BMC Med. 2021, 19, 8. [Google Scholar] [CrossRef]
  119. Fragopoulou, E.; Petsini, F.; Choleva, M.; Detopoulou, M.; Arvaniti, O.S.; Kallinikou, E.; Sakantani, E.; Tsolou, A.; Nomikos, T.; Samaras, Y. Evaluation of anti-inflammatory, anti-platelet and anti-oxidant activity of wine extracts prepared from ten different grape varieties. Molecules 2020, 25, 5054. [Google Scholar] [CrossRef]
  120. Sperkowska, B.; Murawska, J.; Przybylska, A.; Gackowski, M.; Kruszewski, S.; Durmowicz, M.; Rutkowska, D. Cardiovascular effects of chocolate and wine-narrative review. Nutrients 2021, 13, 4269. [Google Scholar] [CrossRef]
  121. Xu, J.; Yang, Y.; Liu, Y.; Cao, S. Natural Nrf2 activators from juices, wines, coffee, and cocoa. Beverages 2020, 6, 68. [Google Scholar]
  122. Magrone, T.; Jirillo, E. Polyphenols from red wine are potent modulators of innate and adaptive immune responsiveness. Proc. Nutr. Soc. 2010, 69, 279–285. [Google Scholar] [CrossRef]
  123. Biasi, F.; Guina, T.; Maina, M.; Cabboi, B.; Deiana, M.; Tuberoso, C.I.; Calfapietra, S.; Chiarpotto, E.; Sottero, B.; Gamba, P.; et al. Phenolic compounds present in Sardinian wine extracts protect against the production of inflammatory cytokines induced by oxysterols in Caco-2 human enterocyte-like cells. Biochem. Pharmacol. 2013, 86, 138–145. [Google Scholar] [CrossRef]
  124. Chang, Y.-P.; Ka, S.-M.; Hsu, W.-H.; Chen, A.; Chao, L.K.; Lin, C.-C.; Hsieh, C.-C.; Chen, M.-C.; Chiu, H.-W.; Ho, C.-L.; et al. Resveratrol inhibits NLRP3 inflammasome activation by preserving mitochondrial integrity and augmenting autophagy. J. Cell Physiol. 2015, 230, 1567–1579. [Google Scholar] [CrossRef] [PubMed]
  125. Nunes, C.; Teixeira, N.; Serra, D.; Freitas, V.; Almeida, L.; Laranjinha, J. Red wine polyphenol extract efficiently protects intestinal epithelial cells from inflammation via opposite modulation of JAK/STAT and Nrf2 pathways. Toxicol. Res. 2016, 5, 53–65. [Google Scholar] [CrossRef] [PubMed]
  126. Chalons, P.; Amor, S.; Courtaut, F.; Cantos-Villar, E.; Richard, T.; Auger, C.; Chabert, P.; Schni-Kerth, V.; Aires, V.; Delmas, D. Study of potential anti-inflammatory effects of red wine extract and resveratrol through a modulation of interleukin-1-beta in macrophages. Nutrients 2018, 10, 1856. [Google Scholar] [CrossRef]
  127. Cornebise, C.; Perus, M.; Hermetet, F.; Valls-Fonayet, J.; Richard, T.; Aires, V.; Delmas, D. Red wine extract prevents oxidative stress and inflammation in ARPE-19 retinal cells. Cells 2023, 12, 1408. [Google Scholar] [CrossRef] [PubMed]
  128. Struck, M.; Watkins, T.; Tomeo, A.; Halley, J.; Bierenbaum, M. Effect of red and white wine on serum lipids, platelet aggregation, oxidation products and antioxidants: A preliminary report. Nutr. Res. 1994, 14, 1811–1819. [Google Scholar] [CrossRef]
  129. Xia, J.; Allenbrand, B.; Sun, G.Y. Dietary supplementation of grape polyphenols and chronic ethanol administration on LDL oxidation and Platelet function in rats. Life Sci. 1998, 63, 383–390. [Google Scholar] [CrossRef] [PubMed]
  130. Muñoz-Bernal, O.A.; de la Rosa, L.A.; Rodrigo-García, J.; Martínez-Ruiz, N.R.; Sáyago-Ayerdi, S.; Rodriguez, L.; Fuentes, E.; Palomo, I.; Alvarez-Parrilla, E. Phytochemical characterization and antiplatelet activity of Mexican red wines and their by-products. S. Afr. J. Enol. Vitic. 2021, 42, 77–90. [Google Scholar] [CrossRef]
  131. Fragopoulou, E.; Antonopoulou, S.; Nomikos, T.; Demopoulos, C.A. Structure elucidation of phenolic compounds from red/white wine with antiatherogenic properties. Biochim. Biophys. Acta 2003, 1632, 90–99. [Google Scholar] [CrossRef]
  132. Fragopoulou, E.; Nomikos, T.; Antonopoulou, S.; Mitsopoulou, C.A.; Demopoulos, C.A. Separation of biologically active lipids from red wine. J. Agric. Food Chem. 2000, 48, 1234–1238. [Google Scholar] [CrossRef]
  133. Xanthopoulou, M.N.; Asimakopoulos, D.; Antonopoulou, S.; Demopoulos, C.A.; Fragopoulou, E. Effect of Robola and Cabernet Sauvignon extracts on platelet activating factor enzymes activity on U937 cells. Food Chem. 2014, 165, 50–59. [Google Scholar] [CrossRef] [PubMed]
  134. Vlachogianni, I.C.; Fragopoulou, E.; Stamatakis, G.M.; Kostakis, I.K.; Antonopoulou, S. Platelet activating factor (PAF) biosynthesis is inhibited by phenolic compounds in U-937 cells under inflammatory conditions. Prostaglandins Other Lipid Mediat. 2015, 121, 176–183. [Google Scholar] [CrossRef] [PubMed]
  135. Silva, P.; Vauzour, D. Wine polyphenols and neurodegenerative diseases: An update on the molecular mechanisms underpinning their protective effects. Beverages 2018, 4, 96. [Google Scholar] [CrossRef]
  136. Reale, M.; Costantini, E.; Jagarlapoodi, S.; Khan, H.; Belwal, T.; Cichelli, A. Relationship of wine consumption with Alzheimer’s disease. Nutrients 2020, 12, 206. [Google Scholar] [CrossRef]
  137. Chinraj, V.; Raman, S. Neuroprotection by resveratrol: A review on brain delivery strategies for Alzheimer’s and Parkinson’s disease. J. Appl. Pharm. Sci. 2022, 12, 1–17. [Google Scholar] [CrossRef]
  138. Porquet, D.; Casadesus, G.; Bayod, S.; Vicente, A.; Canudas, A.M.; Vilaplana, J.; Pelegri, C.; Sanfeliu, C.; Camins, A.; Pallas, M.; et al. Dietary resveratrol prevents Alzheimer’s markers and increases life span in SAMP8. Age 2013, 35, 1851–1865. [Google Scholar] [CrossRef] [PubMed]
  139. Corpas, R.; Grinan-Ferre, C.; Rodriguez-Farre, E.; Pallas, M.; Sanfeliu, C. Resveratrol induces brain resilience against Alzheimer neurodegeneration through proteostasis enhancement. Mol. Neurobiol. 2019, 56, 1502–1516. [Google Scholar] [CrossRef]
  140. Wang, J.; Ho, L.; Zhao, Z.; Seror, I.; Humala, N.; Dickstein, D.L.; Thiyagarajan, M.; Percival, S.S.; Talcott, S.T.; Pasinetti, G.M. Moderate consumption of Cabernet Sauvignon attenuates a beta neuropathology in a mouse model of Alzheimer’s disease. FASEB J. Off. Pub. Fed. Am. Soc. Exp. Biol. 2006, 20, 2313–2320. [Google Scholar]
  141. Martin-Aragon, S.; Jimenez-Aliaga, K.L.; Benedi, J.; Bermejo-Bescos, P. Neurohormetic responses of quercetin and rutin in a cell line over-expressing the amyloid precursor protein (APPswe cells). Phytomed. Inter. J. Phytother. Phytopharmacol. 2016, 23, 1285–1294. [Google Scholar] [CrossRef]
  142. Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Multifunction of myricetin on a beta: Neuroprotection via a conformational change of a beta and reduction of a beta via the interference of secretases. J. Neurosci. Res. 2008, 86, 368–377. [Google Scholar] [CrossRef]
  143. Huang, Y.; Jin, M.; Pi, R.; Zhang, J.; Chen, M.; Ouyang, Y.; Liu, A.; Chao, X.; Liu, P.; Liu, J.; et al. Protective effects of caffeic acid and caffeic acid phenethyl ester against acrolein-induced neurotoxicity in HT22 mouse hippocampal cells. Neurosci. Lett. 2013, 535, 146–151. [Google Scholar] [CrossRef] [PubMed]
  144. Choi, Y.H.; Yoo, M.Y.; Choi, C.W.; Cha, M.R.; Yon, G.H.; Kwon, D.Y.; Kim, Y.S.; Park, W.K.; Ryu, S.Y. A new specific BACE-1 inhibitor from the stembark extract of Vitis vinifera. Planta Med. 2009, 75, 537–540. [Google Scholar] [CrossRef] [PubMed]
  145. Seino, S.; Kimoto, T.; Yoshida, H.; Tanji, K.; Matsumiya, T.; Hayakari, R.; Seya, K.; Kawaguchi, S.; Tsuruga, K.; Tanaka, H.; et al. Gnetin c, a resveratrol dimer, reduces amyloid-beta 1-42 (Aβ42) production and ameliorates Aβ42-lowered cell viability in cultured SH-SY5Y human neuroblastoma cells. Biomed. Res. 2018, 39, 105–115. [Google Scholar] [CrossRef] [PubMed]
  146. Hu, J.; Lin, T.; Gao, Y.; Xu, J.; Jiang, C.; Wang, G.; Bu, G.; Xu, H.; Chen, H.; Zhang, Y.W. The resveratrol trimer miyabenol c inhibits beta-secretase activity and beta-amyloid generation. PLoS ONE 2015, 10, e0115973. [Google Scholar]
  147. Cox, C.J.; Choudhry, F.; Peacey, E.; Perkinton, M.S.; Richardson, J.C.; Howlett, D.R.; Lichtenthaler, S.F.; Francis, P.T.; Williams, R.J. Dietary (-)-epicatechin as a potent inhibitor of βγ-secretase amyloid precursor protein processing. Neurobiol. Aging 2015, 36, 178–187. [Google Scholar] [CrossRef]
  148. Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Flavonols and flavones as BACE-1 inhibitors: Structure-activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim. Biophys. Acta 2008, 1780, 819–825. [Google Scholar] [CrossRef] [PubMed]
  149. De Simone, A.; Mancini, F.; Real Fernandez, F.; Rovero, P.; Bertucci, C.; Andrisano, V. Surface plasmon resonance, fluorescence, and circular dichroism studies for the characterization of the binding of BACE-1 inhibitors. Anal. Bioanal.Chem. 2013, 405, 827–835. [Google Scholar] [CrossRef]
  150. Chakraborty, S.; Kumar, S.; Basu, S. Conformational transition in the substrate binding domain of beta-secretase exploited by NMA and its implication in inhibitor recognition: BACE1-myricetin a case study. Neurochem. Inter. 2011, 58, 914–923. [Google Scholar] [CrossRef]
  151. Jimenez-Aliaga, K.; Bermejo-Bescos, P.; Benedi, J.; Martin-Aragon, S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef]
  152. Lu, J.; Wu, D.M.; Zheng, Y.L.; Hu, B.; Zhang, Z.F.; Shan, Q.; Zheng, Z.H.; Liu, C.M.; Wang, Y.J. Quercetin activates amp-activated protein kinase by reducing PP2C expression protecting old mouse brain against high cholesterol-induced neurotoxicity. J. Pathol. 2010, 222, 199–212. [Google Scholar] [CrossRef]
  153. Cheng, J.; Rui, Y.; Qin, L.; Xu, J.; Han, S.; Yuan, L.; Yin, X.; Wan, Z. Vitamin D combined with resveratrol prevents cognitive decline in SAMP8 mice. Curr. Alzheimer Res. 2017, 14, 820–833. [Google Scholar] [CrossRef] [PubMed]
  154. Eom, T.K.; Ryu, B.; Lee, J.K.; Byun, H.G.; Park, S.J.; Kim, S.K. Beta-secretase inhibitory activity of phenolic acid conjugated chitooligosaccharides. J. Enzym. Inhib. Med. Chem. 2013, 28, 214–217. [Google Scholar] [CrossRef] [PubMed]
  155. Choi, B.; Kim, S.; Jang, B.-G.; Kim, M.-J. Piceatannol, a natural analogue of resveratrol, effectively reduces beta-amyloid levels via activation of alpha-secretase and matrix metalloproteinase-9. J. Funct. Foods 2016, 23, 124–134. [Google Scholar] [CrossRef]
  156. Ohta, K.; Mizuno, A.; Ueda, M.; Li, S.; Suzuki, Y.; Hida, Y.; Hayakawa-Yano, Y.; Itoh, M.; Ohta, E.; Kobori, M.; et al. Autophagy impairment stimulates PS1 expression and gamma-secretase activity. Autophagy 2010, 6, 345–352. [Google Scholar] [CrossRef]
  157. Marambaud, P.; Zhao, H.; Davies, P. Resveratrol promotes clearance of Alzheimer’s disease amyloid-beta peptides. J. Biol. Chem. 2005, 280, 37377–37382. [Google Scholar] [CrossRef]
  158. Harada, N.; Zhao, J.; Kurihara, H.; Nakagata, N.; Okajima, K. Resveratrol improves cognitive function in mice by increasing production of insulin-like growth factor-I in the hippocampus. J. Nutr. Biochem. 2011, 22, 1150–1159. [Google Scholar] [CrossRef]
  159. Deng, H.; Mi, M.T. Resveratrol attenuates Aβ25-35 caused neurotoxicity by inducing autophagy through the TyrRS-PARP1-SIRT1 signaling pathway. Neurochem. Res. 2016, 41, 2367–2379. [Google Scholar] [CrossRef]
  160. Wang, D.M.; Li, S.Q.; Wu, W.L.; Zhu, X.Y.; Wang, Y.; Yuan, H.Y. Effects of long-term treatment with quercetin on cognition and mitochondrial function in a mouse model of Alzheimer’s disease. Neurochem. Res. 2014, 39, 1533–1543. [Google Scholar] [CrossRef]
  161. Ribeiro, C.A.; Saraiva, M.J.; Cardoso, I. Stability of the transthyretin molecule as a key factor in the interaction with a-beta peptide–relevance in Alzheimer’s disease. PLoS ONE 2012, 7, e45368. [Google Scholar] [CrossRef]
  162. Espargaro, A.; Ginex, T.; Vadell, M.D.; Busquets, M.A.; Estelrich, J.; Munoz-Torrero, D.; Luque, F.J.; Sabate, R. Combined in vitro cell-based/in silico screening of naturally occurring flavonoids and phenolic compounds as potential anti-Alzheimer drugs. J. Nat. Prod. 2017, 80, 278–289. [Google Scholar] [CrossRef]
  163. Sabogal-Guaqueta, A.M.; Munoz-Manco, J.I.; Ramirez-Pineda, J.R.; Lamprea-Rodriguez, M.; Osorio, E.; Cardona-Gomez, G.P. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 2015, 93, 134–145. [Google Scholar] [CrossRef] [PubMed]
  164. Ho, L.; Ferruzzi, M.G.; Janle, E.M.; Wang, J.; Gong, B.; Chen, T.Y.; Lobo, J.; Cooper, B.; Wu, Q.L.; Talcott, S.T.; et al. Identification of brain-targeted bioactive dietary quercetin-3-o-glucuronide as a novel intervention for Alzheimer’s disease. FASEB J. Off. Pub. Fed. Am. Soc. Exp. Biol. 2013, 27, 769–781. [Google Scholar] [CrossRef] [PubMed]
  165. Lian, Q.; Nie, Y.; Zhang, X.; Tan, B.; Cao, H.; Chen, W.; Gao, W.; Chen, J.; Liang, Z.; Lai, H.; et al. Effects of grape seed proanthocyanidin on Alzheimer’s disease in vitro and in vivo. Exp. Ther. Med. 2016, 12, 1681–1692. [Google Scholar] [CrossRef] [PubMed]
  166. Jhang, K.A.; Park, J.S.; Kim, H.S.; Chong, Y.H. Resveratrol ameliorates tau hyperphosphorylation at Ser396 site and oxidative damage in rat hippocampal slices exposed to vanadate: Implication of ERK1/2 and GSK-3β signaling cascades. J. Agric. Food. Chem. 2017, 65, 9626–9634. [Google Scholar] [CrossRef] [PubMed]
  167. Shen, X.Y.; Luo, T.; Li, S.; Ting, O.Y.; He, F.; Xu, J.; Wang, H.Q. Quercetin inhibits okadaic acid-induced tau protein hyperphosphorylation through the Ca2+-calpain-p25-CDK5 pathway in HT22 cells. Int. J. Mol. Med. 2018, 41, 1138–1146. [Google Scholar] [CrossRef] [PubMed]
  168. Ali, T.; Kim, M.J.; Rehman, S.U.; Ahmad, A.; Kim, M.O. Anthocyanin-loaded PEG-gold nanoparticles enhanced the neuroprotection of anthocyanins in an Aβ1–42 mouse model of Alzheimer’s disease. Mol. Neurobiol. 2017, 54, 6490–6506. [Google Scholar] [CrossRef]
  169. Sul, D.; Kim, H.S.; Lee, D.; Joo, S.S.; Hwang, K.W.; Park, S.Y. Protective effect of caffeic acid against beta-amyloid-induced neurotoxicity by the inhibition of calcium influx and tau phosphorylation. Life Sci. 2009, 84, 257–262. [Google Scholar] [CrossRef] [PubMed]
  170. Okawara, M.; Katsuki, H.; Kurimoto, E.; Shibata, H.; Kume, T.; Akaike, A. Resveratrol protects dopaminergic neurons in midbrain slice culture from multiple insults. Biochem. Pharmacol. 2007, 73, 550–560. [Google Scholar] [CrossRef]
  171. Wang, H.; Dong, X.; Liu, Z.; Zhu, S.; Liu, H.; Fan, W.; Hu, Y.; Hu, T.; Yu, Y.; Li, Y.; et al. Resveratrol suppresses rotenone-induced neurotoxicity through activation of SIRT1/AKT1 signaling pathway. Anat. Rec. 2018, 301, 1115–1125. [Google Scholar] [CrossRef]
  172. Ferretta, A.; Gaballo, A.; Tanzarella, P.; Piccoli, C.; Capitanio, N.; Nico, B.; Annese, T.; Di Paola, M.; Dell’aquila, C.; De Mari, M.; et al. Effect of resveratrol on mitochondrial function: Implications in Parkin-associated familiar Parkinson’s disease. Biochim. Biophys. Acta 2014, 1842, 902–915. [Google Scholar] [CrossRef]
  173. Wu, Y.; Li, X.; Zhu, J.X.; Xie, W.; Le, W.; Fan, Z.; Jankovic, J.; Pan, T. Resveratrol-activated AMPK/SIRT1/autophagy in cellular models of Parkinson’s disease. Neurosignals 2011, 19, 163–174. [Google Scholar] [CrossRef] [PubMed]
  174. Guo, Y.J.; Dong, S.Y.; Cui, X.X.; Feng, Y.; Liu, T.; Yin, M.; Kuo, S.H.; Tan, E.K.; Zhao, W.J.; Wu, Y.C. Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of α-synuclein via SIRT1-deacetylated LC3. Mol. Nutr. Food Res. 2016, 60, 2161–2175. [Google Scholar] [CrossRef] [PubMed]
  175. Ay, M.; Luo, J.; Langley, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Molecular mechanisms underlying protective effects of quercetin against mitochondrial dysfunction and progressive dopaminergic neurodegeneration in cell culture and MitoPark transgenic mouse models of Parkinson’s disease. J. Neurochem. 2017, 141, 766–782. [Google Scholar] [CrossRef] [PubMed]
  176. Zhu, M.; Han, S.; Fink, A.L. Oxidized quercetin inhibits alpha-synuclein fibrillization. Biochim. Biophys. Acta 2013, 1830, 2872–2881. [Google Scholar] [CrossRef] [PubMed]
  177. Ardah, M.T.; Paleologou, K.E.; Lv, G.; Abul Khair, S.B.; Kazim, A.S.; Minhas, S.T.; Al-Tel, T.H.; Al-Hayani, A.A.; Haque, M.E.; Eliezer, D.; et al. Structure activity relationship of phenolic acid inhibitors of alpha-synuclein fibril formation and toxicity. Front. Aging Neurosci. 2014, 6, 197. [Google Scholar] [CrossRef] [PubMed]
  178. Fazili, N.A.; Naeem, A. Anti-fibrillation potency of caffeic acid against an antidepressant induced fibrillogenesis of human alpha-synuclein: Implications for Parkinson’s disease. Biochimie 2015, 108, 178–185. [Google Scholar] [CrossRef] [PubMed]
  179. Jimenez-Del-Rio, M.; Guzman-Martinez, C.; Velez-Pardo, C. The effects of polyphenols on survival and locomotor activity in Drosophila melanogaster exposed to iron and paraquat. Neurochem. Res. 2010, 35, 227–238. [Google Scholar] [CrossRef]
  180. Ma, H.; Wang, X.; Li, X.; Heianza, Y.; Qi, L. Moderate alcohol drinking with meals is related to lower incidence of type 2 diabetes. Am. J. Clin. Nutr. 2022, 116, 1507–1514. [Google Scholar] [CrossRef]
  181. Xia, X.; Sun, B.; Li, W.; Zhang, X.; Zhao, Y. Anti-diabetic activity phenolic constituents from red wine against α-glucosidase and α-amylase. J. Food Process. Preserv. 2017, 41, e12942. [Google Scholar] [CrossRef]
  182. Welsch, C.A.; Lachance, P.A.; Wasserman, B.P. Dietary phenolic compounds: Inhibition of Na+-dependent d-glucose uptake in rat intestinal brush border membrane vesicles. J. Nutr. 1989, 119, 1698–1704. [Google Scholar] [CrossRef]
  183. Cermak, R.; Landgraf, S.; Wolffram, S. Quercetin glucosides inhibit glucose uptake into brush-border-membrane vesicles of porcine jejunum. Br. J. Nutr. 2004, 91, 849–855. [Google Scholar] [CrossRef] [PubMed]
  184. Gasparrini, M.; Giampieri, F.; Suarez, J.M.A.; Mazzoni, L.; Hernandez, T.Y.F.; Quiles, J.L.; Bullon, P.; Battino, M. AMPK as a new attractive therapeutic target for disease prevention: The role of dietary compounds AMPK and disease prevention. Curr. Drug Tar. 2016, 17, 865–889. [Google Scholar] [CrossRef] [PubMed]
  185. Napoli, R.; Cozzolino, D.; Guardasole, V.; Angelini, V.; Zarra, E.; Matarazzo, M.; Cittadini, A.; Saccà, L.; Torella, R. Red wine consumption improves insulin resistance but not endothelial function in type 2 diabetic patients. Metabolism 2005, 54, 306–313. [Google Scholar] [CrossRef]
  186. Ye, J.; Chen, X.; Bao, L. Effects of wine on blood pressure, glucose parameters, and lipid profile in type 2 diabetes mellitus. Medicine 2019, 98, e15771. [Google Scholar] [CrossRef]
  187. Blomster, J.I.; Zoungas, S.; Chalmers, J.; Li, Q.; Chow, C.K.; Woodward, M.; Mancia, G.; Poulter, N.; Williams, B.; Harrap, S.; et al. The relationship between alcohol consumption and vascular complications and mortality in individuals with type 2 diabetes. Diabetes Care 2014, 37, 1353–1359. [Google Scholar] [CrossRef]
  188. Gepner, Y.; Golan, R.; Harman-Boehm, I.; Henkin, Y.; Schwarzfuchs, D.; Shelef, I.; Durst, R.; Kovsan, J.; Bolotin, A.; Leitersdorf, E.; et al. Effects of initiating moderate alcohol intake on cardiometabolic risk in adults with type 2 diabetes: A 2-year randomized, controlled trial. Ann. Intern Med. 2015, 163, 569–579. [Google Scholar] [CrossRef]
  189. Jang, S.S.; McIntyre, L.; Chan, M.; Brown, P.N.; Finley, J.; Chen, S.X. Ethanol concentration of Kombucha teas in British Columbia, Canada. J. Food Prot. 2021, 84, 1878–1883. [Google Scholar] [CrossRef]
  190. Ivory, R.; Delaney, E.; Mangan, D.; McCleary, B.V. Determination of ethanol concentration in Kombucha beverages: Single-laboratory validation of an enzymatic method, first action method 2019.08. J. AOAC Int. 2021, 104, 422–430. [Google Scholar] [CrossRef]
  191. Rossini, D.; Bogsan, C. Is it possible to brew non-alcoholic Kombucha? Brazilian scenario after restrictive legislation. Fermentation 2023, 9, 810. [Google Scholar] [CrossRef]
  192. Malbasa, R.V.; Loncar, E.S.; Vitas, J.S.; Canadanovic-Brunet, J.M. Influence of starter cultures on the antioxidant activity of kombucha beverage. Food Chem. 2011, 127, 1727–1731. [Google Scholar] [CrossRef]
  193. Fu, C.; Yan, F.; Cao, Z.; Xie, F.; Lin, J. Antioxidant activities of kombucha prepared from three different substrates and changes in content of probiotics during storage. Food Sci. Technol. 2014, 34, 123–126. [Google Scholar] [CrossRef]
  194. Jayabalan, R.; Subathradevi, P.; Marimuthu, S.; Sathishkumar, M.; Swaminathan, K. Changes in free-radical scavenging ability of kombucha tea during fermentation. Food Chem. 2008, 109, 227–234. [Google Scholar] [CrossRef] [PubMed]
  195. Chu, S.-C.; Chen, C. Effects of origins and fermentation time on the antioxidant activities of kombucha. Food Chem. 2006, 98, 502–507. [Google Scholar] [CrossRef]
  196. Bortolomedi, B.M.; Paglarini, C.S.; Brod, F.C.A. Bioactive compounds in kombucha: A review of substrate effect and fermentation conditions. Food Chem. 2022, 385, 132719. [Google Scholar] [CrossRef]
  197. Tanaka, T.; Kouno, I. Oxidation of tea catechins: Chemical structures and reaction mechanism. Food Sci. Technol. Res. 2003, 9, 128–133. [Google Scholar] [CrossRef]
  198. Xia, X.; Dai, Y.; Wu, H.; Liu, X.; Wang, Y.; Yin, L.; Wang, Z.; Li, X.; Zhou, J. Kombucha fermentation enhances the health-promoting properties of soymilk beverage. J. Funct. Foods 2019, 62, 103549. [Google Scholar] [CrossRef]
  199. Gulhan, M.F. A new substrate and nitrogen source for traditional kombucha beverage: Stevia rebaudiana leaves. Appl. Biochem. Biotechnol. 2023, 195, 4096–4115. [Google Scholar] [CrossRef] [PubMed]
  200. Dipti, P.; Yogesh, B.; Kain, A.K.; Pauline, T.; Anju, B.; Sairam, M.; Singh, B.; Mongia, S.S.; Kumar, G.I.; Selvamurthy, W. Lead induced oxidative stress: Beneficial effects of kombucha tea. Biomed. Environ. Sci. 2003, 16, 276–282. [Google Scholar]
  201. Yang, Z.-W.; Ji, B.-P.; Zhou, F.; Li, B.; Luo, Y.; Yang, L.; Li, T. Hypocholesterolaemic and antioxidant effects of kombucha tea in high-cholesterol fed mice. J. Sci. Food Agric. 2009, 89, 150–156. [Google Scholar] [CrossRef]
  202. Vazquez-Cabral, B.D.; Larrosa-Perez, M.; Gallegos-Infante, J.A.; Moreno-Jimenez, M.R.; Gonzalez-Laredo, R.F.; Rutiaga-Quinones, J.G.; Gamboa-Gomez, C.I.; Rocha-Guzman, N.E. Oak kombucha protects against oxidative stress and inflammatory processes. Chem. Biol. Interact. 2017, 272, 1–9. [Google Scholar] [CrossRef]
  203. Aloulou, A.; Hamden, K.; Elloumi, D.; Ali, M.B.; Hargafi, K.; Jaouadi, B.; Ayadi, F.; Elfeki, A.; Ammar, E. Hypoglycemic and antilipidemic properties of kombucha tea in alloxan-induced diabetic rats. BMC Complement. Med. Ther. 2012, 12, 63–72. [Google Scholar] [CrossRef] [PubMed]
  204. Bhattacharya, S.; Gacchui, R.; Sil, P.C. Effect of kombucha, a fermented black tea in attenuating oxidative stress mediated tissue damage in alloxan induced diabetic rats. Food Chem. Toxicol. 2013, 60, 328–340. [Google Scholar] [CrossRef] [PubMed]
  205. Srihari, T.; Karthikesan, K.; Ashokkumar, N.; Satyanarayana, U. Antihyperglycaemic efficacy of kombucha in streptozotocin-induced rats. J. Funct. Foods 2013, 5, 1794–1802. [Google Scholar] [CrossRef]
  206. Jayabalan, R.; Chen, P.N.; Hsieh, Y.S.; Prabhakaran, K.; Pitchai, P.; Marimuthu, S.; Yun, S.E. Effect of solvent fractions of kombucha tea on viability and invasiveness of cancer cells-characterization of dimethyl 2-(2-hydroxy-2-methoxypropylidine)malonate and vitexin. Indian J. Biotechnol. 2011, 10, 75–82. [Google Scholar]
  207. Vitas, J.S.; Cvetanovic, A.D.; Maskovic, P.Z.; Svarc-Gajic, J.V.; Malbasa, R.V. Chemical composition and biological activity of novel types of kombucha beverages with yarrow. J. Funct. Foods 2018, 44, 95–102. [Google Scholar] [CrossRef]
  208. Yang, C.S.; Wang, H. Cancer preventive activities of tea catechins. Molecules 2016, 21, 12. [Google Scholar] [CrossRef] [PubMed]
  209. Zhang, Y.; Yuan, Y.; Wu, H.; Xie, Z.; Wu, Y.; Song, X.; Wang, J.; Shu, W.; Xu, J.; Liu, B.; et al. Effect of verbascoside on apoptosis and metastasis in human oral squamous cell carcinoma. Int. J. Cancer 2018, 143, 980–991. [Google Scholar] [CrossRef]
  210. Cetojevic-Simin, D.D.; Bogdanovic, G.M.; Cvetkovic, D.D.; Velicanski, A.S. Antiproliferative and antimicrobial activity of traditional kombucha and Satureja montana L. kombucha. J. BUON 2008, 13, 395–401. [Google Scholar]
  211. Srihari, T.; Arunkumar, R.; Arunakaran, J.; Satyanarayana, U. Downregulation of signalling molecules involved in angiogenesis of prostate cancer cell line (PC-3) by kombucha (lyophilized). Biomed. Prev. Nutr. 2013, 3, 53–58. [Google Scholar] [CrossRef]
  212. Marzban, F.; Azizi, G.; Afraei, S.; Sedaghat, R.; Seyedzadeh, M.H.; Razavi, A.; Mirshafiey, A. Kombucha tea ameliorates experimental autoimmune encephalomyelitis in mouse model of multiple sclerosis. Food Agric. Immunol. 2015, 26, 782–793. [Google Scholar] [CrossRef]
  213. Nileeka Balasuriya, B.; Vasantha Rupasinghe, H. Plant flavonoids as angiotensin converting enzyme inhibitors in regulation of hypertension. Funct. Food Health Dis. 2011, 5, 172–188. [Google Scholar] [CrossRef]
  214. Guerrero, L.; Castillo, J.; Quiñones, M.; Garcia-Vallvé, S.; Arola, L.; Pujadas, G.; Muguerza, B. Inhibition of angiotensin-converting enzyme activity by flavonoids: Structure-activity relationship studies. PLoS ONE 2012, 7, e49493. [Google Scholar] [CrossRef] [PubMed]
  215. Gamboa-Gomez, C.I.; Gonzalez-Laredo, R.F.; Gallegos-Infante, J.A.; Perez, M.M.L.; Moreno-Jimenez, M.R.; Flores-Rueda, A.G.; Rocha-Guzmam, N.E. Antioxidant and angiotensin-converting enzyme inhibitory activity of Eucalyptus camaldulensis and Litsea glaucescens infusions fermented with kombucha consortium. Food Technol. Biotechnol. 2016, 54, 367–373. [Google Scholar] [CrossRef] [PubMed]
  216. Bhattacharya, S.; Gachhui, R.; Sil, P.C. Hepatoprotective properties of kombucha tea against TBHP-induced oxidative stress via suppression of mitochondria dependent apoptosis. Pathophysiology 2011, 18, 221–234. [Google Scholar] [CrossRef] [PubMed]
  217. Kabiri, N.; Ahangar-Darabi, M.; Rafieian-Kopaei, M.; Setorki, M.; Doudi, M. Protective effect of kombucha tea on liver damage induced by thioacetamide in rats. J. Biol. Sci. 2014, 14, 343–348. [Google Scholar] [CrossRef]
  218. Hyun, J.; Lee, Y.; Wang, S.; Kim, J.; Kim, J.; Cha, J.; Seo, Y.S.; Jung, Y. Kombucha tea prevents obese mice from developing hepatic steatosis and liver damage. Food Sci. Biotechnol. 2016, 25, 861–866. [Google Scholar] [CrossRef]
  219. Al-Mohammadi, A.R.; Ismaiel, A.A.; Ibrahim, R.A.; Moustafa, A.H.; Abou Zeid, A.; Enan, G. Chemical constitution and antimicrobial activity of kombucha fermented beverage. Molecules 2021, 26, 5026. [Google Scholar] [CrossRef]
  220. Greenwalt, C.J.; Ledford, R.A.; Steinkraus, K.H. Determination and characterization of the antimicrobial activity of the fermented tea kombucha. LWT-Food Sci. Technol. 1998, 31, 291–296. [Google Scholar] [CrossRef]
  221. Sreeramulu, G.; Zhu, Y.; Knol, W. Characterization of antimicrobial activity in kombucha fermentation. Acta Biotechnol. 2001, 21, 49–56. [Google Scholar] [CrossRef]
  222. Sreeramulu, G.; Zhu, Y.; Knol, W. Kombucha fermentation and its antimicrobial activity. J. Agric. Food Chem. 2000, 48, 2589–2594. [Google Scholar] [CrossRef]
  223. Battikh, H.; Bakhrouf, A.; Ammar, E. Antimicrobial effect of kombucha analogues. LWT-Food Sci. Technol. 2012, 47, 71–77. [Google Scholar] [CrossRef]
  224. Kaewkod, T.; Bovonsombut, S.; Tragoolpua, Y. Efficacy of Kombucha obtained from green, oolong, and black teas on inhibition of pathogenic bacteria, antioxidation, and toxicity on colorectal cancer cell line. Microorganisms 2019, 7, 700. [Google Scholar] [CrossRef] [PubMed]
  225. Bhattacharya, D.; Bhattacharya, S.; Patra, M.M.; Chakravorty, S.; Sarkar, S.; Chakraborty, W.; Koley, H.; Gachhui, R. Antibacterial activity of polyphenolic fraction of kombucha against enteric bacterial pathogens. Curr. Microbiol. 2016, 3, 885–896. [Google Scholar] [CrossRef] [PubMed]
  226. Winska, K.; Grabarczyk, M.; Maczka, W.; Zarowska, B.; Maciejewska, G.; Aniol, M. Antimicrobial activity of new bicyclic lactones with three or four methyl groups obtained both synthetically and biosynthetically. J. Saudi Chem. Soc. 2016, 22, 363–371. [Google Scholar] [CrossRef]
  227. Milkyas Endale, F.D.; Melaku, Y. Antibacterial and antioxidant phenylpropanoid derivative from the leaves of plantago lanceolate. Nat. Prod. Chem. Res. 2018, 6, 3. [Google Scholar] [CrossRef]
  228. Morales, D. Biological activities of kombucha beverages: The need of clinical evidence. Trends Food Sci. Technol. 2020, 105, 323–333. [Google Scholar] [CrossRef]
  229. Muzaifa, M.; Abubakar, Y.; Nilda, C.; Sapitri, M. Alcohol content and chemical characteristics of fermented beverages in Aceh Province-Indonesia. IOP Conf. Ser. Earth Environ. Sci. 2023, 1241, 012095. [Google Scholar] [CrossRef]
  230. Fiorda, F.A.; Pereira, G.V.M.; Thomaz-Soccol, V.; Rakshit, S.K.; Pagnoncelli, M.G.B.; Vandenberghe, L.P.S.; Soccol, C.R. Microbiological, biochemical, and functional aspects of sugary kefir fermentation—A review. Food Microbiol. 2017, 66, 86–95. [Google Scholar] [CrossRef]
  231. Yilmaz-Ersan, L.; Ozcan, T.; Akpinar-Bayizit, A.; Sahin, S. The antioxidative capacity of kefir produced from goat milk. Int. J. Chem. Eng. Appl. 2016, 7, 22–26. [Google Scholar] [CrossRef]
  232. Suetsuna, K.; Ukeda, H.; Ochi, H. Isolation and characterization of free radical scavenging activities peptides derived from casein. J. Nutr. Biochem. 2000, 11, 128–131. [Google Scholar] [CrossRef]
  233. Arslan, S. A review: Chemical, microbiological and nutritional characteristics of kefir. CyTA-J. Food 2015, 13, 340–345. [Google Scholar] [CrossRef]
  234. Xu, N.; Chen, G.; Liu, H. Antioxidative categorization of twenty amino acids based on experimental evaluation. Molecules 2017, 22, 2066. [Google Scholar] [CrossRef] [PubMed]
  235. Yilmaz-Ersan, L.; Ozcan, T.; Akpinar-Bayizit, A.; Sahin, S. Comparison of antioxidant capacity of cow and ewe milk kefirs. J. Dairy Sci. 2018, 101, 3788–3798. [Google Scholar] [CrossRef] [PubMed]
  236. Hadisaputro, S.; Djokomoeljanto, R.R.; Judiono; Soesatyo, M.H.N.E. The effects of oral plain kefir supplementation on proinflammatory cytokine properties of the hyperglycemia Wistar rats induced by streptozotocin. Acta Med. Indones. 2012, 44, 100–104. [Google Scholar]
  237. Chen, Y.P.; Hsiao, P.J.; Hong, W.S.; Dai, T.Y.; Chen, M.J. Lactobacillus kefiranofaciens M1 isolated from milk kefir grains ameliorates experimental colitis in vitro and in vivo. J. Dairy Sci. 2012, 95, 63–74. [Google Scholar] [CrossRef]
  238. Carasi, P.; Racedo, S.M.; Jacquot, C.; Romanin, D.E.; Serradell, M.A.; Urdaci, M.C. Impact of kefir derived Lactobacillus kefiri on the mucosal immune response and gut microbiota. J. Immunol. Res. 2015, 2015, 361604. [Google Scholar] [CrossRef]
  239. Rosa, D.D.; Grzeskowiak, L.M.; Ferreira, C.L.L.F.; Fonseca, A.C.M.; Reis, B.E.M.; Dias, M.M.; Siqueira, N.P.; Silva, L.L.; Neves, C.A.; Oliveira, L.L.; et al. Kefir reduces insulin resistance and inflammatory cytokine expression in an animal model of metabolic syndrome. Food Funct. 2016, 7, 3390–3401. [Google Scholar] [CrossRef]
  240. Zamberi, N.R.; Abu, N.; Mohamed, N.E.; Nordin, N.; Keong, Y.S.; Beh, B.K.; Zakaria, Z.A.B.; Rahman, N.M.A.N.A.; Alitheen, N.B. The antimetastatic and antiangiogenesis effects of kefir water on murine breast cancer cells. Integr. Cancer Ther. 2016, 15, 53–66. [Google Scholar] [CrossRef]
  241. Seo, M.K.; Park, E.J.; Ko, S.Y.; Choi, E.W.; Kim, S. Therapeutic effects of kefir grain Lactobacillus-derived extracellular vesicles in mice with 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease. J. Dairy Sci. 2018, 101, 8662–8671. [Google Scholar] [CrossRef]
  242. Vieira, L.V.; Sousa, L.M.; Maia, T.A.C.; Gusmao, J.N.F.M.; Goes, P.; Pereira, K.M.A.; Miyajima, F.; Gondim, D.V. Milk Kefir therapy reduces inflammation and alveolar bone loss on periodontitis in rats. Biomed. Pharmacother. 2021, 139, 111677. [Google Scholar] [CrossRef]
  243. Kang, E.A.; Choi, H.I.; Hong, S.W.; Kang, S.; Jegal, H.Y.; Choi, E.W.; Park, B.S.; Kim, J.S. Extracellular vesicles derived from kefir grain Lactobacillus ameliorate intestinal inflammation via regulation of proinflammatory pathway and tight junction integrity. Biomedicines 2020, 8, 522. [Google Scholar] [CrossRef] [PubMed]
  244. Yin, M.; Yan, X.; Weng, W.; Yang, Y.; Gao, R.; Liu, M.; Pan, C.; Zhu, Q.; Li, H.; Wei, Q.; et al. Micro integral membrane protein (MIMP), a newly discovered anti-inflammatory protein of Lactobacillus plantarum, enhances the gut barrier and modulates microbiota and inflammatory cytokines. Cell. Physiol. Biochem. 2018, 45, 474–490. [Google Scholar] [CrossRef]
  245. Chen, H.L.; Hung, K.F.; Yen, C.C.; Laio, C.H.; Wang, J.L.; Lan, Y.W.; Chong, K.Y.; Fan, H.C.; Chen, C.M. Kefir peptides alleviate particulate matter <4 m (PM4.0)-induced pulmonary inflammation by inhibiting the NF-B pathway using luciferase transgenic mice. Sci. Rep. 2019, 9, 11529. [Google Scholar] [PubMed]
  246. Judiono, J.; Hadisaputro, S.; Indranila, K.; Cahyono, B.; Suzery, M.; Widiastuti, Y. Effects of clear kefir on biomolecular aspects of glycemic status of type 2 diabetes mellitus (T2DM) patients in Bandung, West Java [study on human blood glucose, c peptide and insulin]. Funct. Foods Health Dis. 2014, 4, 340–348. [Google Scholar] [CrossRef]
  247. Ostadrahimi, A.; Taghizadeh, A.; Mobasseri, M.; Farrin, N.; Payahoo, L.; Gheshlaghi, Z.B.; 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. [Google Scholar] [PubMed]
  248. 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]
  249. Aihara, K.; Nakamura, Y.; Kajimoto, O.; Hirata, H.; Takahashi, R. Effect of powdered fermented milk with Lactobacillus helveticus on subjects with high-normal blood pressure or mild hypertension. J. Am. Coll. Nutr. 2005, 24, 257–265. [Google Scholar] [CrossRef] [PubMed]
  250. Friques, A.G.F.; Arpini, C.M.; Kalil, I.C.; Gava, A.L.; Leal, M.A.; Porto, M.L.; Nogueira, B.V.; Dias, A.T.; Andrade, T.U.; Pereira, T.M.C.; et al. Chronic administration of the probiotic kefir improves the endothelial function in spontaneously hypertensive rats. J. Transl. Med. 2015, 13, 390. [Google Scholar] [CrossRef]
  251. Brasil, G.A.; Silva-Cutini, M.d.A.; Moraes, F.d.S.A.; Pereira, T.d.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]
  252. Ochoa-de la Paz, L.; Gulias-Canizo, R.; Ruiz-Leyja, E.A.; Sanchez-Castillo, H.; Parodi, J. The role of GABA neurotransmitter in the human central nervous system, physiology, and pathophysiology. Rev. Mex. Neurocienc. 2021, 22, 67–76. [Google Scholar] [CrossRef]
  253. Noori, N.; Bangash, M.Y.; Motaghinejad, M.; Hosseini, P.; Noudoost, B. Kefir protective effects against nicotine cessation-induced anxiety and cognition impairments in rats. Adv. Biomed. Res. 2014, 3, 251. [Google Scholar] [CrossRef] [PubMed]
  254. Van de Wouw, M.; Walsh, A.M.; Crispie, F.; Leuven, L.; Lyte, J.M.; Boehme, M.; Clarke, G.; Dinan, T.G.; Cotter, P.D.; Cryan, J.F. Distinct actions of the fermented beverage kefir on host behaviour, immunity and microbiome gut-brain modules in the mouse. Microbiome 2020, 8, 67. [Google Scholar] [CrossRef] [PubMed]
  255. Chen, C.; Hing, M.C.; Kubow, S. Kefir extracts suppress in vitro proliferation of estrogen-dependent human breast cancer cells but not normal mammary epithelial cells. J. Med. Food 2007, 10, 416–422. [Google Scholar] [CrossRef] [PubMed]
  256. Gao, J.; Gu, F.; Ruan, H.; Chen, Q.; He, J.; He, G. Induction of apoptosis of gastric cancer cells SGC7901 in vitro by a cell-free fraction of Tibetan kefir. Int. Dairy J. 2013, 30, 14–18. [Google Scholar] [CrossRef]
  257. Ghoneum, M.; Felo, N. Selective induction of apoptosis in human gastric cancer cells by Lactobacillus kefiri (PFT), a novel kefir product. Oncol. Rep. 2015, 34, 1659–1666. [Google Scholar] [CrossRef] [PubMed]
  258. Ghoneum, M.; Gimzewski, J. Apoptotic effect of a novel kefir product, PFT, on multidrug-resistant myeloid leukemia cells via a hole-piercing mechanism. Int. J. Oncol. 2014, 44, 830–837. [Google Scholar] [CrossRef]
  259. 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] [PubMed]
  260. Rizk, S.; Maalouf, K.; Baydoun, E. The antiproliferative effect of kefir cell-free fraction on HuT-102 malignant T lymphocytes. Clin. Lymphoma Myeloma Leuk. 2009, 9, S198–S203. [Google Scholar] [CrossRef]
  261. Rizk, S.; Maalouf, K.; Nasser, H.; El-Hayek, S. The pro-apoptotic effect of kefir in malignant T-lymphocytes involves a p53 dependent pathway. Clin. Lymphoma Myeloma Leuk. 2013, 13, S367. [Google Scholar] [CrossRef]
  262. Maalouf, K.; Baydoun, E.; Rizk, S. Kefir induces cell-cycle arrest and apoptosis in HTLV-l-negative malignant T-lymphocytes. Cancer Manag. Res. 2011, 3, 39–47. [Google Scholar]
  263. Laranjo, M. Starter cultures and their role in fermented foods. In Microbial Fermentations in Nature and as Designed Processes; Hurst, C.J., Ed.; John Wiley & Sons: Hoboken, NJ, USA, 2023; pp. 281–292. [Google Scholar]
  264. Zhou, Y.; Wang, R.; Zhang, Y.; Yang, Y.; Sun, X.; Zhang, Q.; Yang, N. Biotransformation of phenolics and metabolites and the change in antioxidant activity in kiwifruit induced by Lactobacillus plantarum fermentation. J. Sci. Food Agric. 2020, 100, 3283–3290. [Google Scholar] [CrossRef] [PubMed]
  265. Isas, A.S.; Escobar, F.; Alvarez-Villamil, E.; Molina, V.; Mateos, R.; Lizarraga, E.; Mozzi, F.; Van Nieuwenhove, C. Fermentation of pomegranate juice by lactic acid bacteria and its biological effect on mice fed a high-fat diet. Food Biosci. 2023, 53, 102516. [Google Scholar] [CrossRef]
  266. Cele, N.P.; Akinola, S.A.; Manhivi, V.E.; Shoko, T.; Remize, F.; Sivakumar, D. Influence of lactic acid bacterium strains on changes in quality, functional compounds and volatile compounds of mango juice from different cultivars during fermentation. Foods 2022, 11, 682. [Google Scholar] [CrossRef] [PubMed]
  267. Moslemi, M.; Moayedi, A.; Khomeiri, M.; Maghsoudlou, Y. Development of a whey-based beverage with enhanced levels of conjugated linoleic acid (CLA) as facilitated by endogenous walnut lipase. Food Chem. X 2023, 17, 100547. [Google Scholar] [CrossRef] [PubMed]
  268. Sales, A.L.; Iriondo-DeHond, A.; DePaula, J.; Ribeiro, M.; Ferreira, I.M.P.L.V.O.; Miguel, M.A.L.; del Castillo, M.D.; Farah, A. Intracellular antioxidant and anti-inflammatory effects and bioactive profiles of coffee cascara and black tea kombucha beverages. Foods 2023, 12, 1905. [Google Scholar] [CrossRef]
  269. Ozturk, B.E.T.; Eroglu, B.; Delik, E.; Cicek, M.; Cicek, E. Comprehensive evaluation of three important herbs for kombucha fermentation. Food Technol. Biotechnol. 2023, 61, 127–137. [Google Scholar] [CrossRef] [PubMed]
  270. Mendonca, G.R.; Pinto, R.A.; Praxedes, E.A.; Abreu, V.K.G.; Dutra, R.P.; Pereira, A.F.; Lemos, T.O.; Reis, A.S.; Pereira, A.L.F. Kombucha based on unconventional parts of the Hibiscus sabdariffa L.: Microbiological, physico-chemical, antioxidant activity, cytotoxicity and sensorial characteristics. Int. J. Gastron. Food Sci. 2023, 34, 100804. [Google Scholar] [CrossRef]
  271. Ziemlewska, A.; Zagorska-Dziok, M.; Niziol-Lukaszewska, Z.; Kielar, P.; Molon, M.; Szczepanek, D.; Sowa, I.; Wojciak, M. In vitro evaluation of antioxidant and protective potential of kombucha-fermented black berry extracts against H2O2-Induced oxidative stress in human skin cells and yeast model. Int. J. Mol. Sci. 2023, 24, 4388. [Google Scholar] [CrossRef]
  272. Amjadi, S.; Armanpour, V.; Ghorbani, M.; Tabibiazar, M.; Soofi, M.; Roufegarinejad, L. Determination of phenolic composition, antioxidant activity, and cytotoxicity characteristics of kombucha beverage containing Echium amoenum. J. Food Meas. Charact. 2023, 17, 3162–3172. [Google Scholar] [CrossRef]
  273. de la Bastida, A.R.; Peiroten, A.; Langa, S.; Rodríguez-Mínguez, E.; Curiel, J.A.; Arques, J.L.; Landete, J.M. Fermented soy beverages as vehicle of probiotic lactobacilli strains and source of bioactive isoflavones: A potential double functional effect. Heliyon 2023, 9, e14991. [Google Scholar] [CrossRef]
  274. Chong, A.Q.; Lau, S.W.; Chin, N.L.; Talib, R.A.; Basha, R.K. Fermented beverage benefits: A comprehensive review and comparison of kombucha and kefir microbiome. Microorganisms 2023, 11, 1344. [Google Scholar] [CrossRef]
  275. Corona, O.; Randazzo, W.; Miceli, A.; Guarcello, R.; Francesca, N.; Erten, H.; Moschetti, G.; Settanni, L. Characterization of kefir-like beverages produced from vegetable juices. LWT-Food. Sci. Technol. 2016, 66, 572–581. [Google Scholar] [CrossRef]
  276. Koh, W.Y.; Utra, U.; Rosma, A.; Effarizah, M.E.; Rosli, W.I.W.; Park, Y.-H. Development of a novel fermented pumpkin-based beverage inoculated with water kefir grains: A response surface methodology approach. Food Sci. Biotechnol. 2018, 27, 525–535. [Google Scholar] [CrossRef] [PubMed]
  277. Laureys, D.; Aerts, M.; Vandamme, P.; De Vuyst, L. Oxygen and diverse nutrients influence the water kefir fermentation process. Food Microbiol. 2018, 73, 351–361. [Google Scholar] [CrossRef]
  278. Laureys, D.; Aerts, M.; Vandamme, P.; De Vuyst, L. The buffer capacity and calcium concentration of water influence the microbial species diversity, grain growth, and metabolite production during water kefir fermentation. Front. Microbiol. 2019, 10, 2876. [Google Scholar] [CrossRef] [PubMed]
  279. Syrokou, M.K.; Papadelli, M.; Ntaikou, I.; Paramithiotis, S.; Drosinos, E.H. Sugary kefir: Microbial identification and technological properties. Beverages 2019, 5, 61. [Google Scholar] [CrossRef]
  280. Verce, M.; De Vuyst, L.; Weckx, S. Shotgun metagenomics of a water kefir fermentation ecosystem reveals a novel Oenococcus species. Front. Microbiol. 2019, 10, 479. [Google Scholar] [CrossRef] [PubMed]
  281. Zannini, E.; Lynch, K.M.; Nyhan, L.; Sahin, A.W.; Riordan, P.O.; Luk, D.; Arendt, E.K. Influence of substrate on the fermentation characteristics and culture-dependent microbial composition of water kefir. Fermentation 2022, 9, 28. [Google Scholar] [CrossRef]
  282. Tavares, P.P.L.G.; Mamona, C.T.P.; Nascimento, R.Q.; dos Anjos, E.A.; de Souza, C.O.; Almeida, R.C.d.C.; Mamede, M.E.d.O.; Magalhães-Guedes, K.T. Non-conventional sucrose-based substrates: Development of non-dairy kefir beverages with probiotic potential. Fermentation 2023, 9, 384. [Google Scholar] [CrossRef]
  283. Velicanski, A.; Cvetkovic, D.; Markov, S.; Tumbas, V.; Savatovic, S. Antimicrobial and antioxidant activity of lemon balm kombucha. Acta Period. Technol. 2007, 38, 165–172. [Google Scholar] [CrossRef]
  284. Sirirat, D.; Jelena, P. Bacterial inhibition and antioxidant activity of kefir produced from Thai jasmine rice milk. Biotechnology 2010, 9, 332–337. [Google Scholar] [CrossRef]
  285. Yavari, N.; Assadi, M.M.; Larijani, K.; Moghadam, M.B. Response surface methodology for optimization of glucuronic acid production using kombucha layer on sour cherry juice. Aust. J. Basic Appl. Sci. 2010, 4, 3250–3256. [Google Scholar]
  286. Liamkaew, D.R.; Chattrawanit, J.; Danvirutai, D.P. Kombucha production by combinations of black tea and apple juice. Prog. Appl. Sci. Technol. 2016, 6, 139–146. [Google Scholar]
  287. Pure, A.E.; Pure, M.E. Antioxidant and antibacterial activity of kombucha beverages prepared using banana peel, common nettles and black tea infusions. Appl. Food Biotechnol. 2016, 3, 125–130. [Google Scholar]
  288. Watawana, M.I.; Jayawardena, N.; Gunawardhana, C.B.; Waisundara, V.Y. Enhancement of the antioxidant and starch hydrolase inhibitory activities of king coconut water (Cocos nucifera var. Aurantiaca) by fermentation with kombucha ‘tea fungus’. Int. J. Food Sci. Technol. 2016, 51, 490–498. [Google Scholar] [CrossRef]
  289. Ayed, L.; Ben Abid, S.; Hamdi, M. Development of a beverage from red grape juice fermented with the kombucha consortium. Ann. Microbiol. 2017, 67, 111–121. [Google Scholar] [CrossRef]
  290. Ayed, L.; Hamdi, M. Manufacture of a beverage from cactus pear juice using “tea fungus” fermentation. Ann. Microbiol. 2015, 65, 2293–2299. [Google Scholar] [CrossRef]
  291. Zubaidah, E.; Yurista, S.; Rahmadani, N.R. Characteristic of physical, chemical, and microbiological kombucha from various varieties of apples. IOP Conf. Ser. Earth Environ. Sci. 2017, 131, 012040. [Google Scholar] [CrossRef]
  292. Shahbazi, H.; Gahruie, H.H.; Golmakani, M.-T.; Eskandari, M.H.; Movahedi, M. Effect of medicinal plant type and concentration on physicochemical, antioxidant, antimicrobial, and sensorial properties of kombucha. Food Sci. Nutr. 2018, 6, 2568–2577. [Google Scholar] [CrossRef]
  293. Vitas, J.; Malbasa, R.; Grahovac, J.; Loncar, E. The antioxidant activity of kombucha fermented milk products with stinging nettle and winter savory. Chem. Ind. Chem. Eng. Q. 2013, 19, 129–139. [Google Scholar] [CrossRef]
  294. Abuduaibifu, A.; Tamer, C.E. Evaluation of physicochemical and bioaccessibility properties of goji berry kombucha. J. Food Process Preserv. 2019, 43, e14077. [Google Scholar] [CrossRef]
  295. Sharifudin, S.A.; Ho, W.Y.; Yeap, S.K.; Abdullah, R.; Koh, S.P. Fermentation and characterisation of potential kombucha cultures on papaya-based substrates. LWT-Food Sci. Technol. 2021, 151, 112060. [Google Scholar] [CrossRef]
  296. Majid, A.A.; Suroto, D.A.; Utami, T.; Rahayu, E.S. Probiotic potential of kombucha drink from butterfly pea (Clitoria ternatea L.) flower with the addition of Lactiplantibacillus plantarum subsp. plantarum Dad-13. Biocatal. Agric. Biotechnol. 2023, 51, 102776. [Google Scholar] [CrossRef]
  297. Kilic, G.; Sengun, I.Y. Bioactive properties of Kombucha beverages produced with Anatolian hawthorn (Crataegus orientalis) and nettle (Urtica dioica) leaves. Food Biosci. 2023, 53, 102631. [Google Scholar] [CrossRef]
  298. Ankolekar, C.; Pinto, M.; Greene, D.; Shetty, K. In vitro bioassay based screening of antihyperglycemia and antihypertensive activities of Lactobacillus acidophilus fermented pear juice. Innov. Food Sci. Emerg. Technol. 2012, 13, 221–230. [Google Scholar] [CrossRef]
  299. Bontsidis, C.; Mallouchos, A.; Terpou, A.; Nikolaou, A.; Batra, G.; Mantzourani, I.; Alexopoulos, A.; Plessas, S. Microbiological and chemical properties of chokeberry juice fermented by novel lactic acid bacteria with potential probiotic properties during fermentation at 4 °C for 4 weeks. Foods 2021, 10, 768. [Google Scholar] [CrossRef]
  300. Chen, R.; Chen, W.; Chen, H.; Zhang, G.; Chen, W. Comparative evaluation of the antioxidant capacities, organic acids, and volatiles of papaya juices fermented by Lactobacillus acidophilus and Lactobacillus plantarum. J. Food Qual. 2018, 2018, 9490435. [Google Scholar] [CrossRef]
  301. Chen, X.; Yuan, M.; Wang, Y.; Zhou, Y.; Sun, X. Influence of fermentation with different lactic acid bacteria and in vitro digestion on the change of phenolic compounds in fermented kiwifruit pulps. Int. J. Food Sci. Technol. 2022, 57, 2670–2679. [Google Scholar] [CrossRef]
  302. Filannino, P.; Bai, Y.; Di Cagno, R.; Gobbetti, M.; Ganzle, M.G. Metabolism of phenolic compounds by Lactobacillus spp. during fermentation of cherry juice and broccoli puree. Food Microbiol. 2015, 46, 272–279. [Google Scholar]
  303. Hashemi, S.M.B.; Jafarpour, D. Fermentation of bergamot juice with Lactobacillus plantarum strains in pure and mixed fermentations: Chemical composition, antioxidant activity and sensorial properties. LWT-Food Sci. Technol. 2020, 131, 109803. [Google Scholar] [CrossRef]
  304. Isas, A.S.; Celis, M.S.M.; Correa, J.R.P.; Fuentes, E.; Rodríguez, L.; Palomo, I.; Mozzi, F.; Van Nieuwenhove, C. Functional fermented cherimoya (Annona cherimola Mill.) juice using autochthonous lactic acid bacteria. Food Res. Int. 2020, 138, 109729. [Google Scholar] [CrossRef]
  305. Liu, Y.; Chen, H.; Chen, W.; Zhong, Q.; Zhang, G.; Chen, W. Beneficial effects of tomato juice fermented by Lactobacillus plantarum and Lactobacillus casei: Antioxidation, antimicrobial effect, and volatile profiles. Molecules 2018, 23, 2366. [Google Scholar] [CrossRef]
  306. Pontonio, E.; Montemurro, M.; Pinto, D.; Marzani, B.; Trani, A.; Ferrara, G.; Mazzeo, A.; Gobbetti, M.; Rizzello, C.G. Lactic acid fermentation of pomegranate juice as a tool to improve antioxidant activity. Front. Microbiol. 2019, 10, 1550. [Google Scholar] [CrossRef] [PubMed]
  307. Quan, Q.; Liu, W.; Guo, J.; Ye, M.; Zhang, J. Effect of six lactic acid bacteria strains on physicochemical characteristics, antioxidant activities and sensory properties of fermented orange juices. Foods 2022, 11, 1920. [Google Scholar] [CrossRef]
  308. Yang, W.; Liu, J.; Zhang, Q.; Liu, H.; Lv, Z.; Zhang, C.; Jiao, Z. Changes in nutritional composition, volatile organic compounds and antioxidant activity of peach pulp fermented by Lactobacillus. Food Biosci. 2022, 49, 101894. [Google Scholar] [CrossRef]
  309. Frolova, Y.; Vorobyeva, V.; Vorobyeva, I.; Sarkisyan, V.; Malinkin, A.; Isakov, V.; Kochetkova, A. Development of fermented kombucha tea beverage enriched with inulin and B vitamins. Fermentation 2023, 9, 552. [Google Scholar] [CrossRef]
  310. Tauber, A.L.; Schweiker, S.S.; Levonis, S.M. From tea to treatment; epigallocatechin gallate and its potential involvement in minimizing the metabolic changes in cancer. Nutr. Res. 2020, 74, 23–36. [Google Scholar] [CrossRef]
  311. Parn, K.W.; Ling, W.C.; Chin, J.H.; Lee, S.-K. Safety and efficacy of dietary epigallocatechin gallate supplementation in attenuating hypertension via its modulatory activities on the intrarenal renin–angiotensin system in spontaneously hypertensive rats. Nutrients 2022, 14, 4605. [Google Scholar] [CrossRef] [PubMed]
  312. Benjamin, S.; Spener, F. Conjugated linoleic acids as functional food: An insight into their health benefits. Nutr. Metab. 2009, 6, 36. [Google Scholar] [CrossRef]
  313. Koba, K.; Yanagita, T. Health benefits of conjugated linoleic acid (CLA). Obes. Res. Clin. Pract. 2014, 8, e525–e532. [Google Scholar] [CrossRef]
  314. Massa, A.; Axpe, E.; Atxa, E.; Hernandez, I. Sustainable, carbonated, non-alcoholic beverages using leftover bread. Int. J. Gastron. Food Sci. 2022, 30, 100607. [Google Scholar] [CrossRef]
  315. Nguyen, T.-L.; Toh, M.; Lu, Y.; Ku, S.; Liu, S.-Q. Biovalorization of market surplus bread for development of probiotic-fermented potential functional beverages. Foods 2022, 11, 250. [Google Scholar] [CrossRef]
  316. Siguenza-Andres, T.; Gomez, M.; Rodríguez-Nogales, J.M.; Caro, I. Development of a fermented plant-based beverage from discarded bread flour. LWT-Food Sci. Technol. 2023, 182, 114795. [Google Scholar] [CrossRef]
Table 2. Representative studies on customization of fermented beverages.
Table 2. Representative studies on customization of fermented beverages.
Product Customization Strategy-OutcomeReferences
Fruit-based fermented beverageImprovement in the antioxidant activity of kiwifruit pulp through fermentation with a Lp. plantarum strain. The increase in DPPH and ABTS scavenging activities were correlated with the increase in total phenolic and flavonoid content.[264]
Fruit-based fermented beveragePomegranate juice was fermented with Lp. paraplantarum CRL2051 and Lp. plantarum CRL2030 and administered to C57BL/6 mice fed a high-fat diet. The fermented juice offered protection against weight gain, liver damage, and dyslipidemia.[265]
Fermented mango juiceDifferent mango cultivars were subjected to lactic acid fermentation with two LAB strains, namely Lp. plantarum 75 and Ln. pseudomesenteroides 56. The latter strain improved the retention of carotenoids, while the former enhanced the phenolic content and the antioxidant activity of all mango cultivars.[266]
Whey-based fermented beverageCommercially available probiotic LAB cultures, capable of producing conjugated linoleic acid (CLA), were used to ferment whey that was enriched with walnut oil lipolyzed by endogenous lipases as a source of free linoleic acid. After the optimization of the fermentation conditions, the whey-based beverage, apart from the CLA content, which could reach 36 mg/g fat, also presented a remarkable antioxidant capacity, most likely due to the presence of phenolic compounds and tocopherol in the walnut oil.[267]
Kombucha analogA kombucha analog with the use of coffee (Coffea arabica) by-product infusion, instead of Camellia sinensis infusion, was developed. The antioxidant activity (as estimated through the reduction in intracellular ROS and uric acid concentration in HK-2 model cells) and the anti-inflammatory activity (as estimated through a reduction in NO formation in LPS-induced macrophages of the kombucha analog and black-tea kombucha) were comparable. [268]
Kombucha analogHops (Humulus lupulus L.), madimak (Polygonum cognatum), and hawthorn (Crataegus monogyna) were used to supplement black-tea kombucha, using the same SCOBY. After fermentation, the antioxidant activity of traditional kombucha was higher than that of the ones supplemented with herbs. All kombuchas exhibited comparative antiproliferative capacity against two cancer cell lines, namely HCT116 and Mahlavu.[269]
Kombucha analogHibiscus sabdarifa L. leaves and stems were used to develop kombucha analogs. The products exhibited similar antioxidant capacity and no cytotoxicity against noncancer cells.[270]
Kombucha analogInfusions of blackcurrant (Ribes nigrum), black chokeberry (Aronia melanocarpa), and blueberry (Vaccinium myrtillus) were fermented using the same SCOBY, resulting in products with a significant content of polyphenolic compounds. The kombucha analogs exhibited significant antioxidant activity, assessed both in vitro and with the use of human keratinocytes (HaCaT) and fibroblasts (BJ).[271]
Enhanced kombuchaIncorporation of Echium amoenum in kombucha resulted in a significant increase in the total phenol, anthocyanin, and flavonoid content, as well as the antioxidant activity. The kombucha prepared solely via E. amoenum infusion exhibited enhanced cytotoxicity against the human prostate cancer cell line (PC3) compared to the products containing both black tea and E. amoenum infusions.[272]
Fermented soy beverageCommercially available probiotic strains La. rhamnosus GG and B. longum BB536, along with isolates with probiotic potential, namely B. breve INIA P734, B. longum INIA P132, La. paracasei INIA P272 and La. rhamnosus INIA P344, were used to ferment a commercially available soy beverage. The product obtained with La. rhamnosus GG and La. rhamnosus INIA P344 contained high levels of bioactive isoflavone aglycones. The viability of the strains, along with the bioactive compounds, was maintained during refrigerated storage for 28 d.[273]
B.: Bifidobacterium; La.: Lacticaseibacillus; Ln.: Leuconostoc; Lp.: Lactiplantibacillus.
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

Paramithiotis, S.; Patra, J.K.; Kotseridis, Y.; Dimopoulou, M. Fermented Beverages Revisited: From Terroir to Customized Functional Products. Fermentation 2024, 10, 57. https://doi.org/10.3390/fermentation10010057

AMA Style

Paramithiotis S, Patra JK, Kotseridis Y, Dimopoulou M. Fermented Beverages Revisited: From Terroir to Customized Functional Products. Fermentation. 2024; 10(1):57. https://doi.org/10.3390/fermentation10010057

Chicago/Turabian Style

Paramithiotis, Spiros, Jayanta Kumar Patra, Yorgos Kotseridis, and Maria Dimopoulou. 2024. "Fermented Beverages Revisited: From Terroir to Customized Functional Products" Fermentation 10, no. 1: 57. https://doi.org/10.3390/fermentation10010057

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