Next Article in Journal
Anti-Staphylococcal, Anti-Candida, and Free-Radical Scavenging Potential of Soil Fungal Metabolites: A Study Supported by Phenolic Characterization and Molecular Docking Analysis
Previous Article in Journal
Natural History of Influenza B Virus—Current Knowledge on Treatment, Resistance and Therapeutic Options
Previous Article in Special Issue
Integrated Analysis of Transcriptome and Metabolome Profiles in the Longissimus Dorsi Muscle of Buffalo and Cattle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biosynthesis of Gamma-Aminobutyric Acid (GABA) by Lactiplantibacillus plantarum in Fermented Food Production

by
Massimo Iorizzo
,
Gianluca Paventi
* and
Catello Di Martino
Department of Agricultural, Environmental and Food Sciences, University of Molise, Via De Sanctis, 86100 Campobasso, Italy
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2024, 46(1), 200-220; https://doi.org/10.3390/cimb46010015
Submission received: 27 November 2023 / Revised: 22 December 2023 / Accepted: 25 December 2023 / Published: 26 December 2023
(This article belongs to the Special Issue Molecular Research in Food Science)

Abstract

:
In recent decades, given the important role of gamma-aminobutyric acid (GABA) in human health, scientists have paid great attention to the enrichment of this chemical compound in food using various methods, including microbial fermentation. Moreover, GABA or GABA-rich products have been successfully commercialized as food additives or functional dietary supplements. Several microorganisms can produce GABA, including bacteria, fungi, and yeasts. Among GABA-producing microorganisms, lactic acid bacteria (LAB) are commonly used in the production of many fermented foods. Lactiplantibacillus plantarum (formerly Lactobacillus plantarum) is a LAB species that has a long history of natural occurrence and safe use in a wide variety of fermented foods and beverages. Within this species, some strains possess not only good pro-technological properties but also the ability to produce various bioactive compounds, including GABA. The present review aims, after a preliminary excursus on the function and biosynthesis of GABA, to provide an overview of the current uses of microorganisms and, in particular, of L. plantarum in the production of GABA, with a detailed focus on fermented foods. The results of the studies reported in this review highlight that the selection of new probiotic strains of L. plantarum with the ability to synthesize GABA may offer concrete opportunities for the design of new functional foods.

1. Introduction

Gamma (γ)-Aminobutyric acid (GABA), also named 4-aminobutyric acid, is a four-carbon non-protein amino acid that is widely distributed in an extensive variety of organisms including algae, bacteria, fungi, animals, plants, and cyanobacteria [1,2,3,4,5,6,7].
Although GABA is present in many foods such as fruits, vegetables and grains, its content in them is relatively low [8,9]. As a result, over the years many studies have been devoted to the most suitable strategies to increase the amount of GABA in food [10,11] such as through chemical synthesis [12], plant enrichment [13], or microbial fermentation [3].
Microbial synthesis of GABA may be much more promising than chemical synthesis methods since the former is characterized by high specificity, environmental friendliness and cost-effectiveness [3].
In addition, GABA production by beneficial and pro-technological microorganisms has the potential to increase the functional effect of some fermented foods and beverages [11,14]. So far, various studies have confirmed that several microorganisms like fungi, bacteria, and yeasts have the ability to synthesize GABA [3,15,16].
Lactic acid bacteria (LAB) are ubiquitous microorganisms and are often naturally present in some traditional fermented foods as well. Many LAB species are used as starters in some industrial food fermentations for their pro-technological properties [17,18].
Some LAB species are capable of producing high amounts of GABA [19,20,21] and could be exploited for the production of GABA-fortified foods [14].
Among GABA-producing LAB, Lactiplantibacillus plantarum (formerly Lactobacillus plantarum) is a facultative heterofermentative species with high adaptability to many different conditions, being isolated from various ecological niches including milk, fruit, cereal crops, vegetables, bee bread, fresh meat [22,23,24,25] and fermented foods [26,27]. This bacterial species L. plantarum is a normal inhabitant of the gastro-intestinal tract of insects, fish and mammals, including humans [28,29,30,31,32] and is included in the QPS (Qualified Presumption of Safety) and in GRAS (Generally Recognised as Safe) lists [33,34].
Because of many of its intrinsic properties, numerous strains belonging to this species are proposed as animal and human probiotics [31,32,35,36,37,38,39,40].
L. plantarum is widely used as a starter culture in the fermentation of raw materials from plant and animal origin, where it contributes to enhancing the sensorial quality and shelf life of fermented products [38,39,41,42,43,44]. Some L. plantarum strains also increase the functional properties of various fermented foods by producing a variety of bioactive compounds, including GABA [19,45].
The present review aims, after a preliminary excursus on the function and biosynthesis of GABA, to provide an overview of the current uses of microorganisms and, in particular, of L. plantarum in the production of GABA, with a detailed focus on fermented foods.

2. GABA Function and Metabolism

GABA is produced by bacteria [3,46] fungi [47,48], plants [49,50], vertebrate animals and invertebrates [51,52,53]. Furthermore, Archaea possesses enzyme genes involved in GABA biosynthesis [54,55,56].
Due to this pervasive presence in biological kingdoms and ecosystems, we tend to consider the GABA molecule more as a ubiquitous signaling molecule than as a specific synaptic neurotransmitter [57,58].
GABA-mediated interregnum communication has been observed between algae and invertebrates [59], plants and fungi [60], plants and insects [61], and plants and bacteria [62].
In plants, GABA is an endogenous signaling molecule involved in various physiological and biochemical processes that promote plant growth and development, and mediate responses to abiotic and biotic stresses, including pathogen and insect attacks [1,63,64]. In addition, GABA improves photosynthetic processes, inhibiting the production of reactive oxygen species (ROS), activating antioxidant enzymes, and regulating stomatal opening in case of water stress [65].
In plants, GABA is synthesized from glutamate or arginine and transferred by GABA-permease to mitochondria, where GABA is catabolized by GABA transaminase and succinate semialdehyde dehydrogenase to succinate. The succinate enters the tricarboxylic acid (TCA) cycle to maintain the C/N balance in cells [1].
Over the last several decades GABA has attracted great attention due to its many positive effects on mammalian physiology [10,15,66].
As known, in fact, GABA is the most common inhibitory neurotransmitter in the human central nervous system [67]. Furthermore, besides being an important antidepressant [68], GABA also performs other functions including neuroprotective, anti-inflammatory, antioxidant and antihypertensive effects [66], enhancement of immunity under stress conditions [69], prevention of cancer cell proliferation [70], prevention of diabetic conditions [71], and cholesterol-lowering effect [72].
In mammalian, GABA is synthesized from L-glutamate in the cytoplasm of neuronal and glial cells by the enzyme glutamate decarboxylase (GAD; EC4.1.1.15) using pyridoxal 5′-phosphate (PLP) as an enzyme cofactor [73] (Figure 1). GABA can also be synthesized through deamination and decarboxylation reactions of putrescine, spermine, spermidine, ornithine, and L-glutamine [2].
As for mammalian species, in microorganisms, GABA is produced from L-glutamate through a GAD enzyme-mediated decarboxylation [58] with PLP as a cofactor (Figure 1).
Through a well-established pathway of enzymes known as GABA shunt (Figure 2), some bacteria catabolize GABA [74,75]. The GABA shunt is characterized by a group of enzymes that convert GABA to succinate to fuel the tricarboxylic acid (TCA) cycle in the production of energy and essential metabolic intermediates as carbon skeletons for the cell. In the shunt of GABA, microbial enzymes such as GABA transaminase and succinic semialdehyde dehydrogenase have an optimal pH in the alkaline range of around 8. These observations have led some scholars to advance the hypothesis that the GABA shunt, in addition to representing the link between nitrogen and carbon metabolism, has an important function in the maintenance of pH homeostasis in acidic environments [75,76].
As reported above, microbial GABA synthesis is strictly dependent on the GAD enzyme which is encoded by gadA or gadB genes in bacterial cells. Glutamate is transported into a cell through an antiporter, and then decarboxylation occurs. Finally, the GABA product is secreted from the cell by the glutamate/GABA antiporter, which is encoded by the gadC gene [77].
In recent years, many researchers have studied L. plantarum for its ability to synthesize GABA using the GAD system.
Only Levilactobacillus brevis possesses two GAD genes that produce isozyme GADs among the LAB examined so far [78,79]. The glutamic acid decarboxylase (GAD) system encoded by the gad operon is responsible for glutamate decarboxylation and GABA secretion in bacteria and consists of two important elements: Glu/GABA antiporter gadC and the glutamate decarboxylase enzyme encoded either by gadA, gadB genes [80]. This system converts glutamate into GABA and while doing so consumes protons thus maintaining cytosolic pH homeostasis [79].
Unlike L. brevis, L. plantarum has only one GAD coding enzyme in its genome, the gadB and there may not be a specific glutamate/GABA antiporter (gadC) gene [80].
In a study conducted by Nakatani et al. on the genome of L. plantarum KB1253, it was found that this strain contains two gadB genes coding for glutamate decarboxylase [81].
Many studies showed that L. plantarum can produce appreciable amounts of GABA, so there must be a transporter responsible for transporting glutamate and GABA in and out of the cell. A glutamate/gamma-aminobutyrate transporter family protein coded by the yjeM gene can be the best candidate for such a transporter [82].
Further investigation, conducted by Surachat et al. indicates that L. plantarum is a key GABA-producing species in nature since almost all strains encoded the GAD operon in their genome [83].

3. Production of GABA by Microorganisms

GABA can be obtained not only from natural sources but also through plant enrichment, chemical synthesis, enzymatic process and microbial metabolism [15,84]. Due to the low GABA content in natural animal- and plant- associated food products, high GABA-producing microorganisms are of great importance to produce food-grade GABA and GABA-rich fermented foods via fermentations [85].
The biosynthesis of GABA by microorganisms is safe and eco-friendly and provides the possibility of production of new naturally fermented health-oriented products [16,86].

3.1. Production of GABA by Fungi

Other than bacteria, various yeasts and molds that belong to the kingdom of fungi, have also been reported to as able to produce GABA.
Some Rhizopus oligosporus and Rhizopus oryzae strains have been shown to produce GABA during tempeh fermentation (fermented soybean) [87].
Similarly, Rhizopus monosporus strain 5351 has been reported to increase GABA content in soybean and mung bean [88,89].
Marine yeasts Pichia guilliermondii and Pichia anomala isolated from the Pacific Ocean off Japan have high GABA-producing abilities [90,91].
Actinomucor elegans AS 3.227 has been reported to increase the GABA concentration in sufu (traditional fermented soybean food from China) manufacturing using solid-state fermentation [92].
Glutamic acid decarboxylase has also been identified in yeasts such as Saccharomyces cerevisiae and Kluyveromyces marxianus isolated from fermented products [93,94,95].
Other yeasts belonging to the species Kazachstania unispora, Sporobolomyces carnicolor, Sporobolomyces ruberrimus, Nakazawaea holstiiand, and Pichia scolyti, isolated from wild flowers, also have GAD activity [96].
Aspergillus oryzae NSK is a GABA-generating mold used as a starter culture to ferment rice koji for sake production and soy sauce koji [97,98,99,100,101].
Cai et al. demonstrated that oats fermented by A. oryzae var. effuses 3.2825, A. oryzae 3.5232 and R. oryzae 3.2751 can be recommended as tempeh-like functional foods with higher GABA [102].
In other studies, fermentations by Monascus pilosus IFO 4520 [103] and Monascus purpureus CCRC 31615 [104] increase the content of γ-aminobutyric acid (GABA) in the beni-koji and in the fermented rice.

3.2. Production of GABA by Bacteria

GABA is naturally synthesized by several bacteria. Indeed, not all strains within one species can produce GABA, as the ability depends on the presence of GAD genes and glutamate/GABA antiporter [74].
Bacillus is a commonly reported bacteria that can produce GABA [105,106]. Besides Bacillus, Corynebacterium glutamicum was found to produce endogenous L-glutamate [107], Streptomyces bacillaris and Streptomyces cinereus were reported to increase the GABA content in fermented tea [108]. Otaru et al. have shown that human intestinal Bacteroides are able to synthesize GABA [109].
Recent studies revealed that the increased level of GABA in the human gut could be derived from the ability of the intestinal microbiota or ingested probiotics, such as Bacteroides, bifidobacteria, and some LAB (lactobacilli), to metabolize dietary monosodium glutamate [109,110,111]. Therefore, numerous research has been directed towards isolating and characterizing GABA-producing bacteria to be used as starters for the production of GABA-enriched fermented food [3].
Because of their GRAS status, some LAB are widely used in the production of fermented foods [18] and act as potential probiotic cultures. Actually, in addition to pro-technological functions, LAB also offer beneficial functions such as antioxidant and antimicrobial activities, as well as the formation of bioactive compounds such as GABA [112,113].
Therefore, the use of GABA-producing LAB has been considered a promising possibility in order to increase the nutritional, functional, sensory and technological properties of some fermented food products [10,19,114].
GABA can be biosynthesized by various LAB strains mainly belonging to the genera of Lactobacillus, Lactococcus, Pediococcus, Leuconostoc, Enterococcus, Streptococcus, Weissella, Lacticaseibacillus, Lactiplantibacillus and recently, Levilactobacillus and Secundilactobacillus [21,115,116,117,118,119,120,121].
Nowadays, Lactiplantibacillus plantarum (formerly classified as Lactobacillus plantarum) is among the main LAB species proposed to be used as probiotic starter cultures to produce GABA in the fermented food and beverage industry [35,122,123].

4. Production of GABA by L. plantarum

The production of GABA varies among various LAB strains and is affected by several factors such as pH, fermentation temperature, fermentation time, L-glutamic acid concentration, media additives, and carbon and nitrogen sources [3,85,114,124]. The optimization of these parameters could maximize the amount of GABA contained in some LAB-fermented foods [3,112].
In recent years, many researchers have studied L. plantarum, in particular, for its ability to synthesize GABA in different substrates and growing conditions.
Table 1 summarizes the results of studies investigating the ability of different strains of L. plantarum to produce GABA in different growing media.
The most commonly used culture medium is MRS (de Man, Rogosa and Sharp), a standard substrate designed to promote LAB growth [142]. Monosodium glutamate (MSG), as a source of L-glutamine, is usually supplemented directly into MRS to enhance GABA synthesis from L. plantarum strains [82].
However, the optimal concentration of MSG depends on the bacterial strain. For example, Yogeswara et al. investigated the GABA production from L. plantarum FNCC 260 strain using a wide range of MSG concentrations. The results showed a maximum GABA production (1226 mg/L) by adding 100 mM of MSG to the MRS medium and then incubating at 37 °C for 108 h [131].
In another study, after 18 h at 34 °C, L. plantarum K74 produced 134.52 μg/mL of GABA in MRS broth containing 1% MSG, 212.27 μg/mL of GABA in MRS broth containing 2% MSG, and 234.63 μg/mL of GABA in MRS broth containing 3% MSG [135].
Gomaa et al. examined the effect of MSG and PLP on GABA production from L. brevis and L. plantarum strains, isolated from Egyptian dairy products. The culture medium used was the following composition: 50 g/L glucose; 25 g/L soya peptone; 0.01 g/L MnSO4C4H2O and 2 mL Tween 80. The results of the aforementioned study show that the amount of extracellular GABA produced is proportional to the amounts of MSG and PLP added. Co-culture of L. brevis and L. plantarum produced the highest amount of GABA, 160.57 mM and 224.69 mM, in the presence of 750 M MSG and 200 μM PLP, respectively [143].
Park et al. have obtained high amounts of GABA (19.8 g/L) at 30 °C from L. plantarum EJ2014 using the following culture medium: 100 g/L Yeast extract, 10 g/L dextrose, and 22.5 g/L (w/v) MSG [124].
In a study conducted by Shan et al. L. plantarum NDC75017 produced 3.2 g/kg of GABA, at 30 °C for 48 h, in skimmed milk with 80 mM MSG and 18 μM PLP [144].
As evidenced in all the studies mentioned above, the amount of monosodium glutamate initially available is an important factor in the production of GABA [145].
In fact, as also confirmed in other studies cited below, an initial excessive concentration of MSG may inhibit cell growth or inhibit GABA production due to osmotic stress, while a low concentration of MSG may not meet the requirements of high GABA production [146]. As far as the incubation time is concerned, we have observed that the amount of GABA after reaching the maximum amount after a certain period of time, tends to decrease subsequently. This effect may be caused by a lower availability of precursors (e.g., MSG) but also be linked to degradation, by GABA aminotransferase, of GABA to succinic semialdehyde, which is subsequently converted by succinic semialdehyde dehydrogenase for entry into TCA [11].
Temperature and pH have been reported as the main environmental factors that can modulate gad gene expression [147]. Therefore, adjusting pH and temperature during fermentation is a very effective way to increase microbial GABA production.
LAB employ a complex but efficient combination of different acid resistance systems [148].
Among the various types of tolerance mechanisms to the acidic environment, the GAD system is considered one of the most effective acid mitigation pathways.
In this system, intracellular protons are consumed through decarboxylation of glutamate in the cytoplasm [74].
Shin et al. showed that 40 °C and a pH of 4.5 were the best parameters for the expression of gadB gene encoding GAD from L. plantarum ATCC 14,917 in E. coli BL21 (DE3) [149].
Variation in pH enhances activation of the GAD pathway since it is considered one of the mechanisms that preserve cell homeostasis [150]. Wu et al. evaluated the performance of the GAD pathway in comparison with other acid resistance mechanisms and highlighted how the GAD system is an essential mechanism to maintain metabolic activity under intra- and extracellular acidity [79].
Therefore, the pH of the environment is crucial for the synthesis of GABA. However, it seems that this depends on the bacterial strain [149].
Zhang et al. tested how initial pH affects GABA production by L. plantarum BC114. The best concentration of GABA was detected at pH 5.5, obtaining double the amount of GABA yielded at pH 4.0 [132]. Similar results have been obtained in other studies [129,139,140].
Tajabadi et al. found that after 60 h L. plantarum Taj-Apis362 produces the highest amount of GABA (7.15 mM; 0.74 g/L) at 36 °C in modified MRS: 497.97 mM glutamate, pH 5.31 [129]. Tanamool et al. found that the highest GABA production (15.74 g/L) by L. plantarum L10-11 cultured in MRS with 4% MSG at 30 °C was obtained within 48 h, with a pH range of 5–6 [139].
Very recently, Cai et al. reported that L. plantarum FRT7 after 48 h produced approximately 1.2 g/L in MRS supplemented with 3% MSG and 2 mmol/L of PLP at 40° C with an initial pH of 7.0 [140].
In a recent study conducted by Kim J et al., the optimal conditions for efficient GABA production by L. plantarum FBT215 in modified MRS broth containing 50 mM MSG were investigated. Therefore, the optimal culture temperature for GABA production (103.67 μg/mL) was 37 °C and this efficiency was highest at pH 7.5 and 8.5 and decreased under acidic conditions [123].
Instead, Yogeswara et al. found that GABA production from L. plantarum FNCC 260 was greatly improved under acidic conditions (pH 3.8) in Pigeon pea (Cajanus cajan) milk fermentation [151]. This result is in line with a previous study by Yogeswara et al. where maximum GABA production from L. plantarum FNCC 260 in MRS was observed at pH 4.0 [131].
Regarding the temperature, Yang et al. reported that GAD functionality is directly related to an increase in temperature until it reaches an optimum, after which GAD activity decreases until thermal inactivation [152]. Another study with L. plantarum showed an increase in GAD activity up to 40 °C, achieving optimal GABA production at 35 °C [144].
Importantly, L. plantarum is a mesophilic bacterium with an optimal growth temperature of around 37 °C. This evidence explains why, in all the studies cited in this review, the optimal temperatures for maximum GABA production were in the range of 30–40 °C.

GABA Production by L. plantarum in Fermented Foods

According to the available data, naturally occurring GABA in foods is usually low [85,153]; therefore, the food industry has shown great interest in GABA-enriched foods, through microbial fermentation.
Currently, L. plantarum is a LAB species commonly found in various fermented foods and beverages. Therefore, some food scientists have proposed strains of L. plantarum as starters in single culture (Table 2) or in co-culture with other microbial species (Table 3) to enrich GABA in some traditional or innovative fermented foods, particularly from plant-based sources.
Table 2 summarizes the results obtained from the use of L. plantarum as a single starter in different fermented foods.
Table 2. GABA production by Lactiplantibacillus plantarum (previously Lactobacillus plantarum) in different fermented foods.
Table 2. GABA production by Lactiplantibacillus plantarum (previously Lactobacillus plantarum) in different fermented foods.
MicroorganismIsolation
Source
Fermented FoodGABA
Production
CommentsRefs.
L. plantarum C48cheesebuckwheat, amaranth, chickpea and quinoa flours504 mg/kg in breadGood organoleptic properties of bread enriched of GABA[154]
L. plantarum DSM19463cheesegrape must8.9 g/kg in fermented grape mustIn vitro potential anti-hypertensive effect and dermatological protection.[155]
L. plantarum KB1253picklestomato juice41 mM GABA-enriched fermented tomato juice [156]
L. plantarum KCTC 3105Unknown soya milk 424.67 µg/g DWSoya yogurt with high levels of GABA, produced using a co-culture of L. acidophilus, L. plantarum and L. brevis strains[157]
L. plantarum NDC75017fermented milk12% skim milk + 80 mM MSG314.56 mg/100 gGood flavor and texture of fermented milk-based product [144]
L. plantarum NTU102cabbage pickles8% skim milk + 1% (w/v) MSG629 mg/Ltogether with GABA, production of ACEI was also found, suggesting a possible use of fermented products as potential functional food (hypertension regulation)[158]
L. plantarum C48cheesewholemeal wheat flour100 mg/Klow ACE inhibitory activity (15%) due to synthesis of ACEI [159]
L. plantarum GB01-21 cassava powder80.5 g/L
2.68 g/L h (productivity)
two-step production with Corynebacterium glutamicum G01 (to produce
glutamate) and L. plantarum GB01-21
[152]
L. plantarum Dad-13FNCCpigeon pea milk5.6 g/L The supplementation of sucrose, MSG, and whey isolate significantly increased GABA levels in fermented pigeon pea[151]
L. plantarum NRRL B-59151 FOE and HFOE
(oat)
GABA content: 7.35 mg/100 g in FOE and 8.49 mg/100 g in HFOEFermented oat
demonstrated antidiabetic
effects
[160,161]
Lactobacillus plantarum HU-C2W litchi juice 134 mg/100 mL Fermentation condition: 37 °C for 40 h[141]
L. plantarum DW12fermented red seaweedred seaweed + 1% MSG4 g/LFermentation at 30 °C after 60 days. Substrate composition: red seaweed, cane sugar and potable water in a ratio of 3:1:10, pH 6[162]
L. plantarum DW12fermented red seaweedred seaweed + 0.5% MSG1284 mg/LFermentation at 30 °C after 60 days.
Substrate composition: red seaweed, cane sugar and potable water in a ratio of 3:1:10, pH 6
[163]
L. plantarum DW12fermented red seaweedMCW + 0.5% MSG12.8 mg/100 mLMCW supplemented with 0.5% MSG and 1% sugarcane, pH 6 after 72 h of fermentation[164]
Abbreviations: DW, dry weight; MSG, mono-sodic glutamate; ACEI, angiotensin converting enzyme inhibitor; ACE, angiotensin converting enzyme; FNCC, Food and Nutrition Culture Collection; HFOE, fermented oat + honey; FOE, Fermented Oat; MCW, mature coconut water.
In a recent study [151], it has been proposed a drink prepared from germinated pigeon pea (Cajanus cajan) and fermented using probiotic L. plantarum Dad-13, isolated from dadih, fermented buffalo milk [165]. C. cajan commonly known as pigeon pea, red gram or gungo pea is an important grain legume crop, particularly in rain-fed agricultural regions in the semi-arid tropics, including Asia, Africa and the Caribbean [166].
Additional nutrients such as MSG 1%, whey 4%, and sucrose 3% were added to pigeon pea extract and fermentation was carried out in a closed container at 30 °C for 48 h without shaking. Maximum GABA production (5.6 g/L) was obtained after 12 h of fermentation.
Wang et al. have shown that it is possible to increase the production of GABA in fermented lychee juice by L. plantarum HU-C2W [141]. Litchi (Litchi chinensis Sonn.) is a well-known tropical fruit originating from Asia [167]. After 40 h at 37 °C, a GABA content of 134 mg/100 mL was observed [141].
In various studies, L. plantarum DW12, isolated by Ratanaburee et al. from a fermented red seaweed, has been successfully used as probiotic and starter culture to produce fermented foods and beverages due to its safety aspects and ability to produce GABA [83,162,163,164].
The results obtained in [162] reported that L. plantarum DW12 produces 4 g/L GABA in red seaweed fermentation (red seaweed-cane sugar-potable water = 3:1:10, w/w/v) at 30 °C after 60 days. The red seaweed Gracilaria fisheri is commonly found along the coast of south-east Asian countries and used as a fresh vegetable and as a dried product [168].
In another study conducted by Hayisama-Ae et al., a novel functional beverage was produced from red seaweed Gracilaria fisheri (known as Pom Nang seaweed in Thailand), using L. plantarum DW12 as a starter culture [163]. Fermented red seaweed beverage was produced as follows: red seaweed, cane sugar and potable water in a ratio of 3:1:10 with an addition of 0.5% of MSG and an initial pH of 6.0. After 60 days the fermented red seaweed beverage (FSB) contained 1.28 g/L GABA.
A study conducted by Kantachote et al. aimed to add value to mature coconut water by using the probiotic L. plantarum DW12 for the production of GABA-enriched fermented beverages. Coconut water, with an initial pH of 5.0, was supplemented with 0.5% monosodium glutamate and 1% sugarcane and fermented from L. plantarum DW12. After 48 h, the fermented product contained 128 μg/mL of GABA [164].
Coconut (Cocos nucifera L.) is an important fruit tree found in tropical regions and its fruit can be made into a variety of foods and beverages [169].
Zarei et al. investigated the potential of GABA production by a L. plantarum strain in whey protein beverage [170], building on previous research, in which this strain, isolated from traditional doogh (yogurt, herbs and water) from west region of Iran, have shown a high concentration of GABA production (170.492 ppm) in MRS broth [171]. The best growing conditions that caused the highest GABA production were temperature 37 °C, pH 5.19, glutamic acid 250 mM, and time 72 h. The highest amount of GABA (195.5 ppm) after 30 days of storage was detected in whey protein drinks containing banana concentrate and stored at 25 °C.
L. plantarum NDC75017 (isolated from a traditional fermented dairy product from Inner Mongolia, China) was used as a starter for fermentation at 36°of Skim Milk and 80 mM L-MSG and 18 μM PLP. Under these conditions, GABA production was about 310 mg/100 g [144].
In a study conducted by Di Cagno et al., the use of L. plantarum DSM19463 (formerly L. plantarum C48) for the production of a functional grape-based beverage was evaluated [155]. The grape must, diluted with water, was enriched with yeast extract and 18.4 mM of L-glutamate and left to ferment at 30 °C. After 72 h L. plantarum DSM19463 synthesizes 4.83 mM of GABA [155].
In another study, the L. plantarum C48 has been used in sourdough fermentation [154].
The use of a blend of buckwheat, amaranth, chickpea and quinoa flours (ratio 1:1:5.3:1) subjected to sourdough fermentation by L. plantarum C48 allowed the manufacture of a bread enriched with GABA (504 mg/kg) [159]. The sourdough starter obtained with L. plantarum C48 had GABA concentrations of 12.65, 100.71 and 44.61 mg/kg for white, whole wheat and rye flours, respectively [159].
In another recent study, L. plantarum VL1 was used for the production of Nem Chua (traditionally Vietnamese fermented meat product). Fresh pork without fat was minced and mixed with 5% salt, 20% sugar, and 1% sodium glutamate. L. plantarum VL1, was added to the mixture and after 72 h of fermentation at 37 °C the meat mixture (pH 4.59) contained 1.1 mg/g of GABA [172].
In a study conducted by Nakatani et al. L. plantarum KB1253, isolated from Japanese pickles, is used in GABA-enriched tomato juice production [156]. This strain produces 41.0 mM GABA from 46.8 mM glutamate in tomato juice (pH 4.0, 20°Bx) incubated for 24 h at 35°.
In another study conducted by Rezaei et al., the GABA-producing strain L. plantarum IBRC (10817) was used in the production of a probiotic beverage made from black grapes. After 21 days, the fermented beverage had a concentration of 117.33 mg/L GABA [173].
L. plantarum K16 isolated from kimchi has been used to valorize some agri-food by-products [174], obtained from tomatoes, apples, oranges and green peppers. The agri-food by-products were enriched with 25 g/L of glucose, 12 g/L of yeast extract and 500 mM of MSG. Subsequently, the pH was adjusted to 5.5, and the media were inoculated with L. plantarum K16 and incubated at 34 °C for 96 h. L. plantarum K16 produced the following concentrations of GABA: 1166.81 mg/L, 1280.01 mg/L, 1626.52 mg/L and 1776.75 mg/L in apple, orange, green pepper and tomato by-products, respectively [122].
GABA is an important molecule naturally present in food matrices of plant and animal origin. However, plant-based foods contain a comparatively lower amount of GABA than animal-based foods [8,175].
Considering its potential health benefits, the studies mentioned above have shown that it is possible to increase the amount of GABA not only in some animal products but also in some fermented plant-based foods and beverages, improving their functional properties. In particular, it has been shown that through the use of L. plantarum as a single starter, it has been possible to produce fermented foods from legumes, cereals, fruit juices and some agri-food by-products containing high amounts of GABA.
Besides its use as a single culture, the use of L. plantarum in co-culture (co-fermentation or two-stage fermentation) with other microbial strains belonging to different species is gaining increasing interest. Table 3 summarizes the relevant reports in this field.
In a study conducted by Hussin et al. [146], the effect of different carbohydrates was investigated on enhancing GABA production in yogurt cultured using a mixture of UPMC90 and UPMC91, self-cloned LAB strains (L. plantarum Taj-Apis362, previously isolated from the stomach of honeybee Apis dorsata and engineered by Tajabadi et al. [129,176]). Glucose induced more GABA production (58.56 mg/100 g) compared to inuline, FOS e GOS as prebiotics (34.19–40.51 mg/100 g), and the control sample with added PLP (48.01 mg/100 g) [146].
In other similar study, conducted by Hussin et al., self-cloned and expressed L. plantarum Taj-Apis362 recombinant cells, UPMC90 and UPMC91 were used to improve the GABA production in yogurt. Fermentation of skimmed milk added with glutamate (11.5 mM) after 7.25 h at 39.0 °C produced GABA-rich yogurt (29.96 mg/100 g) [177].
While many studies reported the use of single-strain LAB to generate GABA, only a few reported the production of GABA by co-culturing different bacterial strains [178].
Table 3. GABA production by Lactiplantibacillus plantarum (previously Lactobacillus plantarum) in co-culture with other microbial species.
Table 3. GABA production by Lactiplantibacillus plantarum (previously Lactobacillus plantarum) in co-culture with other microbial species.
L. plantarum StrainsCooperative Species/StrainFood or
Culture Medium
GABA
Production
NotesRefs.
L. plantarum EJ2014B. subtilis HApumpkin1.47%Two-step fermentation[179]
L. plantarum K154B. subtilis HAturmeric (Curcuma longa)/roasted soybean meal mixture + 5% MSG1.78%Two-step fermentation[180]
L. plantarum K154B. subtilis HAdefined medium fortified with glutamate and skim milk4800 µg/mLTwo-step fermentation[181]
L. plantarum K154Leuconostoc mesenteroides SMWater dropwort100 mMTwo-step fermentation [182]
L. plantarum BC114S. cerevisiae SC125mulberry beverage brewing2.42 g/LCo-fermentation[93]
L. plantarum GB01-21C. glutamicum G01cassava powder80.5 g/LTwo-step fermentation[152]
L. plantarum Taj-Apis362Streptococcus thermophilus and Lactobacillus delbrueckii ssp. bulgaricusSkim milk + 2% glucose and 11.5 mM MSG59.0 mg/100 gCo-fermentation[177]
L. plantarum K154Ceriporia laceratabroth fortified with skim milk and 2% MSG15.53 mg/mLTwo-step fermentation[127]
L. plantarum (KCTC 3105)Lactobacillus brevis OPY-1
L. acidophilus
KCCM 40265
Soya milk424.67 µg/gCo-fermentation[157]
L. plantarum L10-11Lactococcus lactis spp. lactis and Lactococcus lactis spp. cremonismilk11.3 mg/100 mLCo-fermentation[183]
L. plantarum JLSC2-6Levilactobacillus brevis YSJ3cauliflower stems35.00 mg/LCo-fermentation[184]
L. plantarum MCM4Lactococcus lactis subsp. lactiswhey-based formulate365.6 mg/100 mLCo-fermentation[185]
L. plantarum DSM749L. brevis
NM101-1
PM 224.69 mMCo-fermentation[143]
L. plantarum C48Lactobacillus paracasei 15N, Streptococcus
thermophilus DPPMAST1, Lactobacillus delbruecki subsp. bulgarigus DPPMALDb5
Milk + 100 or mg/L of olive vegetation water phenolic extract67 mg/LCo-fermentation[186]
Abbreviations: MSG, mono-sodic glutamate; PM, production medium (50 g/L glucose; 25 g/L peptone; 0.01 g/L MnSO4. 4 H2O; 2 mL Tween 80; 200 μM PLP); PLP, pyridoxal 5-phosphate.
In a study carried out by Lim et al., the co-fermentation of turmeric (Curcuma longa)/roasted soybean meal mixture, containing 5% MSG, was optimized to fortify it with bioactive compounds including GABA [180]. Bacillus subtilis HA was used for the first fermentation and L. plantarum K154 isolated from fermented kimchi was used for the second fermentation. The results showed that the amount of GABA increased from 0.01% before fermentation to 1.78% after the second fermentation [128].
In a further study, a two-step fermentation of pumpkin (Cucurbita moschata) was performed using B. subtilis HA and L. plantarum EJ2014, with the aim of producing a novel food ingredient enriched with GABA [179]. Bacillus subtilis HA (KCCM 10775P) strain was isolated from cheonggukjang (traditional Korean fermented soybean) while L. plantarum EJ2014 (KCCM 11545P) was isolated from rice bran [187]. The co-fermented pumpkin contained 1.47% GABA. Bacillus subtilis HA was also used in a two step-fermentation with L. plantarum K154, obtaining a high level of GABA production (about 4800 µg/mL) in a defined medium fortified with glutamate and skim milk [181]. Instead, Yang et al. proposed a two-step method to produce GABA from cassava powder using C. glutamicum G01 and L. plantarum GB01-21 [152]. In this study, glutamic acid was first obtained from cassava powder by saccharification and simultaneous fermentation with C. glutamicum G01, followed by biotransformation of glutamic acid into GABA with resting cells of L. plantarum GB01-21. C. glutamicum G01 was isolated from soil and L. plantarum GB01-21 was obtained through multi-mutagenesis as described in our previous study [188]. After optimizing the reaction conditions (35 °C, pH 7), the maximum concentration of GABA reached 80.5 g/L [152].
In another study, two self-cloned L. plantarum Taj-Apis362 strains possessing high intracellular GAD activity (UPMC90) and high extracellular GAD activity (UPMC91) and a wild-type L. plantarum Taj-Apis362 (UPMC1065) were co-cultured with a starter culture (a mixture of Streptococcus thermophilus and Lactobacillus delbrueckii ssp. bulgaricus) to produce GABA-rich yogurt [129].
The wild-type L. plantarum Taj-Apis362 (UPMC1065) was previously isolated from the stomach of a honeybee Apis dorsata [176] and used as a host for GAD gene overexpression to produce UPMC90 and UPMC91 strains. After 7 h of fermentation at 39.0 °C, the starter co-culture in skim milk with 2% glucose and 11.5 mM glutamate produces 59.00 mg/100 g of GABA.
Water dropwort (Oenanthe javanica DC), a common aquatic perennial plant widely cultivated in most Southeast Asian countries, was co-fermented with Leuconostoc mesenteroides SM and L. plantarum K154 to produce a novel functional food ingredient enriched with GABA (100 mM) [182]. The acidity of the fermented broth, the low concentration of sugar remaining for the second fermentation and the presence of nitrogen sources, stimulated L. plantarum K154 to produce GABA. These data seem to confirm that the production of GABA by bacteria is a bacterial mechanism of response towards acid stress [74].
Woraratphoka et al. used a co-culture of L. plantarum L10-11, Lactococcus lactis spp. lactis and L. lactis spp. cremonis in fresh cheese production [183]. L. plantarum L10-11 which was isolated from Thai fermented fish (Plaa-som) while Lactococcus lactis spp. lactis and L. lactis spp. cremonis they were commercial strains (Lyofast MWO030, SACCO, Italy). After 18 h the fermented milk by single-L10-11 and co-L10-11 contained 1.21 and 11.30 mg/100 mL of GABA, respectively. Thus, this suggested that in the co-culture test, by transforming lactose into lactic acid, the commercial strains decreased the pH value, creating a favorable condition for the enzymatic activity (GAD) of L. plantarum L10-11 that catalyzes the conversion of glutamate to GABA. Therefore, co-fermentation by L. plantarum L10-11 with other LAB strains could possibly increase the rate of GABA production [183].
In a previous study, it was reported that L. plantarum L10-11 was clearly involved in the conversion of MSG to GABA and the highest GABA production was obtained when the initial pH of MRS was in the range of 5.0–6.0 [139].
The data emerging from the above studies confirm that the optimal pH for GABA production by L. plantarum is placed in an acidic pH range of 4–6 [3].
Zhang et al. evaluated the effects on GABA production by co-culture of Levilactobacillus brevis YSJ3 and L. plantarum JLSC2-6. The results indicate that co-culturing these two strains can improve GABA yield (35.00 ± 1.15 mg/L) in fermented cauliflower stems (Brassica oleracea L. var. botrytis) [184].
Functional milk-based beverages enriched with 100 mg/L and 200 mg/L of olive vegetation water phenolic extract (OVWPE) were obtained via fermentation at 40 °C using L. plantarum C48, L. paracasei 15N, S. thermophilus DPPMAST1 and L. delbruecki subsp. bulgarigus DPPMALDb5. The highest amount of GABA (67 mg/L) was detected after 30 days at 4 °C [186].
The results obtained from the above studies have shown that co-culture fermentation using L. plantarum with other bacterial species is a novel technology to improve fermentation quality and promote GABA synthesis. The increase in GABA production by L. plantarum in co-culture with other bacteria may be related to the greater availability of nutrients released by the metabolism of the bacterium used in co-cultures [152,182] which also generates acidic end products of fermentation, which accumulate in the extracellular environment, increasing its acidity and thus promoting GABA synthesis [182,183,184,185].
Other studies, cited below, have shown that some L. plantarum strains improve GABA production even when used in co-culture with fungi.
Co-fermentation of L. plantarum K154 and fungus Ceriporia lacerate efficiently produced GABA (15.53 mg/mL) in a defined medium containing 3% glucose, 3% soybean flour, 0.15% MgSO4, and 5% rice bran for 7 days at 25 °C [127].
The increase in GABA production in co-culture could be related to the fact that C. lacerate, thanks to its enzymatic activities (protease, α-amylase, cellulase, β-1,3-glucanase and phosphatase) [189], increased the availability of nutrients useful for the growth and survival of L. plantarum.
In a study conducted by Zhang et al., S. cerevisiae SC125 and L. plantarum BC114 were used in co-culture to ferment mulberry (Morus alba L.) and produce a functional beverage enriched with GABA [93]. L. plantarum BC114 and S. cerevisiae SC125 were inoculated in pasteurized mulberry substrate with 5 g/L L-glutamate and incubated at 30 °C for 72 h.
Compared to single fermentations with L. plantarum BC114 and S. cerevisiae SC125, which resulted in low GABA production (1.45 g/L and 1.03 g/L, respectively), co-culture produced a higher amount of GABA (2.42 g/L) [93].
The results obtained in this study confirm that the increased ability of L. plantarum to synthesize GABA could be linked to an increased availability of nutrients produced by yeasts, in particular, amino acids [190].
Therefore, co-cultures of selected fungi with GABA-producing strains belonging to L. plantarum species may be a promising approach for the production of GABA-enriched foods, and therefore, this biotechnological application would also merit further scientific investigation.

5. Conclusions and Future Perspectives

In recent decades, consumers’ needs in the field of food production have increased significantly, not only in terms of organoleptic aspects but also in terms of health and well-being. Among the various functional compounds contained in foods, GABA has attracted more and more attention due to its multiple health benefits.
Although GABA is present in many foods such as fruits, vegetables and grains, its content in them is relatively low. In this context, GABA-fortified foods have been significantly considered by researchers for their important biological and functional properties. At present, GABA can be synthesized using different methods, including chemical and enzymatic synthesis, plant enrichment, and microbial production.
The numerous studies conducted on this topic highlighted that GABA production from LAB can play an important role in the food industry. In particular, fermentation by GABA-producing L. plantarum strains can be considered a promising possibility to increase the nutritional, sensory and functional properties of specific fermented foods.
The studies cited in this review have shown that the optimal conditions for GABA are significantly influenced by substrate composition and environmental conditions. Therefore, it is essential to optimize these parameters to improve the production of GABA, according to the production process adopted to obtain a specific fermented food.
Considering that microbial fermentation is an important technology to increase the GABA content in some fermented foods, we believe that the selection of new high-GABA-producing strains belonging to the species L. plantarum should remain a focus of interest in future research because it can offer concrete opportunities for the design of new functional foods.

Author Contributions

Conceptualization, M.I., G.P. and C.D.M.; writing—original draft preparation, M.I., G.P. and C.D.M.; writing—review and editing, M.I., G.P. and C.D.M.; visualization, C.D.M.; supervision, M.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Li, L.; Dou, N.; Zhang, H.; Wu, C. The versatile GABA in plants. Plant Signal. Behav. 2021, 16, 1862565. [Google Scholar] [CrossRef] [PubMed]
  2. Tillakaratne, N.J.K.; Medina-Kauwe, L.; Gibsons, K.M.; Courtwright, K.H.; Summers, J.W. Gamma-aminobutyric acid (GABA) metabolism in mammalian neural and nonneural tissues. Comp. Biochem. Physiol. 1995, 112, 247–263. [Google Scholar] [CrossRef] [PubMed]
  3. Dhakal, R.; Bajpai, V.K.; Baek, K.H. Production of GABA (γ-aminobutyric acid) by microorganisms: A review. Braz. J. Microbiol. 2012, 43, 1230–1241. [Google Scholar] [CrossRef] [PubMed]
  4. Ding, W.; Cui, J.; Zhao, Y.; Han, B.; Li, T.; Zhao, P.; Xu, J.W.; Yu, X. Enhancing Haematococcus pluvialis biomass and γ-aminobutyric acid accumulation by two-step cultivation and salt supplementation. Bioresour. Technol. 2019, 285, 121334. [Google Scholar] [CrossRef] [PubMed]
  5. Shiels, K.; Murray, P.; Saha, S.K. Marine cyanobacteria as potential alternative source for GABA production. Bioresour. Technol. Rep. 2019, 8, 100342. [Google Scholar] [CrossRef]
  6. Boonburapong, B.; Laloknam, S.; Incharoensakdi, A. Accumulation of gamma-aminobutyric acid in the halotolerant cyanobacterium Aphanothece halophytica under salt and acid stress. J. Appl. Phycol. 2016, 28, 141–148. [Google Scholar] [CrossRef]
  7. Jantaro, S.; Kanwal, S. Low-molecular-weight nitrogenous compounds (GABA and polyamines) in blue–green algae. In Algal Green Chemistry; Elsevier: Amsterdam, The Netherlands, 2017; pp. 149–169. [Google Scholar]
  8. Ramos-Ruiz, R.; Poirot, E.; Flores-Mosquera, M. GABA, a non-protein amino acid ubiquitous in food matrices. Cogent Food Agric. 2018, 4, 1534323. [Google Scholar] [CrossRef]
  9. Sun, Y.; Mehmood, A.; Battino, M.; Xiao, J.; Chen, X. Enrichment of gamma-aminobutyric acid in foods: From conventional methods to innovative technologies. Food Res. Int. 2022, 162, 111801. [Google Scholar] [CrossRef]
  10. Diez-Gutiérrez, L.; Vicente, L.S.; Barrón, L.J.R.; del Carmen Villarán, M.; Chavarri, M. Gamma-aminobutyric acid and probiotics: Multiple health benefits and their future in the global functional food and nutraceuticals market. J. Funct. Foods 2020, 64, 103669. [Google Scholar] [CrossRef]
  11. Sahab, N.R.M.; Subroto, E.; Balia, R.L.; Utama, G.L. γ-Aminobutyric acid found in fermented foods and beverages: Current trends. Heliyon 2020, 6, e05526. [Google Scholar] [CrossRef]
  12. Zhu, N.; Wang, T.; Ge, L.; Li, Y.; Zhang, X.; Bao, H. γ-amino butyric acid (GABA) synthesis enabled by copper-catalyzed carboamination of alkenes. Org. Lett. 2017, 19, 4718–4721. [Google Scholar] [CrossRef] [PubMed]
  13. Bai, Q.; Yang, R.; Zhang, L.; Gu, Z. Salt stress induces accumulation of γ–aminobutyric acid in germinated foxtail millet (Setaria italica L.). Cereal Chem. 2013, 90, 145–149. [Google Scholar] [CrossRef]
  14. Diana, M.; Quílez, J.; Rafecas, M. Gamma-aminobutyric acid as a bioactive compound in foods: A review. J. Funct. Foods 2014, 10, 407–420. [Google Scholar] [CrossRef]
  15. Heli, Z.; Hongyu, C.; Dapeng, B.; Yee Shin, T.; Yejun, Z.; Xi, Z.; Yingying, W. Recent advances of γ-aminobutyric acid: Physiological and immunity function, enrichment, and metabolic pathway. Front. Nutr. 2022, 9, 1076223. [Google Scholar] [CrossRef]
  16. Luo, H.; Liu, Z.; Xie, F.; Bilal, M.; Liu, L.; Yang, R.; Wang, Z. Microbial production of gamma-aminobutyric acid: Applications, state-of-the-art achievements, and future perspectives. Crit. Rev. Biotechnol. 2021, 41, 491–512. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, Y.; Wu, J.; Lv, M.; Shao, Z.; Hungwe, M.; Wang, J.; Bai, X.; Xie, J.; Wang, Y.; Geng, W. Metabolism characteristics of lactic acid bacteria and the expanding applications in food industry. Front. Bioeng. Biotechnol. 2021, 9, 612285. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, J.M.; Fehér, C.; Cao, M.; Lu, F.; Jensen, P.R. Editorial: Lactic acid bacteria: Microbial metabolism and expanding applications. Front. Bioeng. Biotechnol. 2021, 9, 794164. [Google Scholar] [CrossRef]
  19. Cui, Y.; Miao, K.; Niyaphorn, S.; Qu, X. Production of gamma-aminobutyric acid from lactic acid bacteria: A systematic review. Int. J. Mol. Sci. 2020, 21, 995. [Google Scholar] [CrossRef]
  20. Rehman, A.; Di Benedetto, G.; Bird, J.K.; Dabene, V.; Vadakumchery, L.; May, A.; Schyns, G.; Sybesma, W.; Mak, T.N. Development of a workflow for the selection, identification and optimization of lactic acid bacteria with high γ-aminobutyric acid production. Sci. Rep. 2023, 13, 13663. [Google Scholar] [CrossRef]
  21. Yogeswara, I.B.A.; Maneerat, S.; Haltrich, D. Glutamate decarboxylase from lactic acid bacteria—A key enzyme in GABA synthesis. Microorganisms 2020, 8, 1923. [Google Scholar] [CrossRef]
  22. Siezen, R.J.; Tzeneva, V.A.; Castioni, A.; Wels, M.; Phan, H.T.K.; Rademaker, J.L.W.; Starrenburg, M.J.C.; Kleerebezem, M.; Molenaar, D.; Van Hylckama Vlieg, J.E.T. Phenotypic and genomic diversity of Lactobacillus plantarum strains isolated from various environmental niches. Environ. Microbiol. 2010, 12, 758–773. [Google Scholar] [CrossRef] [PubMed]
  23. Martino, M.E.; Bayjanov, J.R.; Caffrey, B.E.; Wels, M.; Joncour, P.; Hughes, S.; Gillet, B.; Kleerebezem, M.; van Hijum, S.A.F.T.; Leulier, F. Nomadic lifestyle of Lactobacillus plantarum revealed by comparative genomics of 54 strains isolated from different habitats. Environ. Microbiol. 2016, 18, 4974–4989. [Google Scholar] [CrossRef] [PubMed]
  24. Siezen, R.J.; van Hylckama Vlieg, J.E.T. Genomic diversity and versatility of Lactobacillus plantarum, a natural metabolic engineer. Microb. Cell Factories 2011, 10, S3. [Google Scholar] [CrossRef] [PubMed]
  25. Filannino, P.; De Angelis, M.; Di Cagno, R.; Gozzi, G.; Riciputi, Y.; Gobbetti, M. How Lactobacillus plantarum shapes its transcriptome in response to contrasting habitats. Environ. Microbiol. 2018, 20, 3700–3716. [Google Scholar] [CrossRef] [PubMed]
  26. Testa, B.; Lombardi, S.J.; Tremonte, P.; Succi, M.; Tipaldi, L.; Pannella, G.; Sorrentino, E.; Iorizzo, M.; Coppola, R. Biodiversity of Lactobacillus plantarum from traditional Italian wines. World J. Microbiol. Biotechnol. 2014, 30, 2299–2305. [Google Scholar] [CrossRef] [PubMed]
  27. Iorizzo, M.; Lombardi, S.J.; Macciola, V.; Testa, B.; Lustrato, G.; Lopez, F.; De Leonardis, A. Technological potential of Lactobacillus strains isolated from fermented green olives: In Vitro studies with emphasis on oleuropein-degrading capability. Sci. World J. 2016, 2016, 1917592. [Google Scholar] [CrossRef] [PubMed]
  28. Iorizzo, M.; Pannella, G.; Lombardi, S.J.; Ganassi, S.; Testa, B.; Succi, M.; Sorrentino, E.; Petrarca, S.; De Cristofaro, A.; Coppola, R.; et al. Inter-and intra-species diversity of lactic acid bacteria in Apis mellifera ligustica colonies. Microorganisms 2020, 8, 1578. [Google Scholar] [CrossRef]
  29. Iorizzo, M.; Albanese, G.; Testa, B.; Ianiro, M.; Letizia, F.; Succi, M.; Tremonte, P.; D’andrea, M.; Iaffaldano, N.; Coppola, R. Presence of lactic acid bacteria in the intestinal tract of the mediterranean trout (Salmo macrostigma) in its natural environment. Life 2021, 11, 667. [Google Scholar] [CrossRef]
  30. Park, S.-Y.; Lim, S.-D. Probiotic characteristics of Lactobacillus plantarum FH185 isolated from human feces. Korean J. Food Sci. Anim. Resour. 2015, 35, 615–621. [Google Scholar] [CrossRef]
  31. Iorizzo, M.; Testa, B.; Ganassi, S.; Lombardi, S.J.; Ianiro, M.; Letizia, F.; Succi, M.; Tremonte, P.; Vergalito, F.; Cozzolino, A.; et al. Probiotic properties and potentiality of Lactiplantibacillus plantarum strains for the biological control of chalkbrood disease. J. Fungi 2021, 7, 379. [Google Scholar] [CrossRef]
  32. Iorizzo, M.; Testa, B.; Lombardi, S.J.; Ganassi, S.; Ianiro, M.; Letizia, F.; Succi, M.; Tremonte, P.; Vergalito, F.; Cozzolino, A.; et al. Antimicrobial activity against Paenibacillus larvae and functional properties of Lactiplantibacillus plantarum strains: Potential benefits for honeybee health. Antibiotics 2020, 9, 442. [Google Scholar] [CrossRef] [PubMed]
  33. Koutsoumanis, K.; Allende, A.; Alvarez-Ordóñez, A.; Bolton, D.; Bover-Cid, S.; Chemaly, M.; De Cesare, A.; Hilbert, F.; Lindqvist, R.; Nauta, M.; et al. Update of the list of qualified presumption of safety (QPS) recommended microbiological agents intentionally added to food or feed as notified to EFSA 17: Suitability of taxonomic units notified to EFSA until September 2022. EFSA J. 2023, 21, e07746. [Google Scholar] [PubMed]
  34. Salvetti, E.; O’Toole, P.W. When regulation challenges innovation: The case of the genus Lactobacillus. Trends Food Sci. Technol. 2017, 66, 187–194. [Google Scholar] [CrossRef]
  35. Letizia, F.; Albanese, G.; Testa, B.; Vergalito, F.; Bagnoli, D.; Di Martino, C.; Carillo, P.; Verrillo, L.; Succi, M.; Sorrentino, E.; et al. In vitro assessment of bio-functional properties from Lactiplantibacillus plantarum strains. Curr. Issues Mol. Biol. 2022, 44, 2321–2334. [Google Scholar] [CrossRef] [PubMed]
  36. Iorizzo, M.; Albanese, G.; Letizia, F.; Testa, B.; Tremonte, P.; Vergalito, F.; Lombardi, S.J.; Succi, M.; Coppola, R.; Sorrentino, E. Probiotic potentiality from versatile Lactiplantibacillus plantarum Strains as resource to enhance freshwater fish health. Microorganisms 2022, 10, 463. [Google Scholar] [CrossRef]
  37. Fidanza, M.; Panigrahi, P.; Kollmann, T.R. Lactiplantibacillus plantarum–nomad and ideal probiotic. Front. Microbiol. 2021, 12, 712236. [Google Scholar] [CrossRef]
  38. Yilmaz, B.; Bangar, S.P.; Echegaray, N.; Suri, S.; Tomasevic, I.; Manuel Lorenzo, J.; Melekoglu, E.; Rocha, J.M.J.M.; Ozogul, F.; Lorenzo, J.M.; et al. The impacts of Lactiplantibacillus plantarum on the functional properties of fermented foods: A Review of Current Knowledge. Microorganisms 2022, 10, 826. [Google Scholar] [CrossRef]
  39. Behera, S.S.; Ray, R.C.; Zdolec, N. Lactobacillus plantarum with functional properties: An approach to increase safety and shelf-life of fermented foods. Biomed Res. Int. 2018, 2018, 9361614. [Google Scholar] [CrossRef]
  40. Echegaray, N.; Yilmaz, B.; Sharma, H.; Kumar, M.; Pateiro, M.; Ozogul, F.; Lorenzo, J.M. A novel approach to Lactiplantibacillus plantarum: From probiotic properties to the omics insights. Microbiol. Res. 2023, 268, 127289. [Google Scholar] [CrossRef]
  41. De Leonardis, A.; Testa, B.; Macciola, V.; Lombardi, S.J.; Iorizzo, M. Exploring enzyme and microbial technology for the preparation of green table olives. Eur. Food Res. Technol. 2016, 242, 363–370. [Google Scholar] [CrossRef]
  42. Lombardi, S.J.; Pannella, G.; Iorizzo, M.; Testa, B.; Succi, M.; Tremonte, P.; Sorrentino, E.; Di Renzo, M.; Strollo, D.; Coppola, R. Inoculum strategies and performances of malolactic starter Lactobacillus plantarum M10: Impact on chemical and sensorial characteristics of fiano wine. Microorganisms 2020, 8, 516. [Google Scholar] [CrossRef] [PubMed]
  43. Succi, M.; Pannella, G.; Tremonte, P.; Tipaldi, L.; Coppola, R.; Iorizzo, M.; Lombardi, S.J.; Sorrentino, E. Sub-optimal pH preadaptation improves the survival of Lactobacillus plantarum strains and the malic acid consumption in wine-like medium. Front. Microbiol. 2017, 8, 470. [Google Scholar] [CrossRef] [PubMed]
  44. Koutsoumanis, K.; Allende, A.; Alvarez-Ordóñez, A.; Bolton, D.; Bover-Cid, S.; Chemaly, M.; Davies, R.; De Cesare, A.; Hilbert, F.; Lindqvist, R.; et al. Update of the list of QPS-recommended biological agents intentionally added to food or feed as notified to EFSA 12: Suitability of taxonomic units notified to EFSA until March 2020. EFSA J. 2020, 18, e06174. [Google Scholar] [PubMed]
  45. Seddik, H.A.; Bendali, F.; Gancel, F.; Fliss, I.; Spano, G.; Drider, D. Lactobacillus plantarum and its probiotic and food potentialities. Probiotics Antimicrob. Proteins 2017, 9, 111–122. [Google Scholar] [CrossRef] [PubMed]
  46. Dover, S.; Halpern, Y.S. Genetic analysis of the γ aminobutyrate utilization pathway in Escherichia coli K 12. J. Bacteriol. 1974, 117, 494–501. [Google Scholar] [CrossRef] [PubMed]
  47. Kumar, S.; Punekar, N.S. The metabolism of 4-aminobutyrate (GABA) in fungi. Mycol. Res. 1997, 101, 403–409. [Google Scholar] [CrossRef]
  48. Coleman, S.T.; Fang, T.K.; Rovinsky, S.A.; Turano, F.J.; Moye-Rowley, W.S. Expression of a glutamate decarboxylase homologue is required for normal oxidative stress tolerance in Saccharomyces cerevisiae. J. Biol. Chem. 2001, 276, 244–250. [Google Scholar] [CrossRef] [PubMed]
  49. Steward, F.C.; Thompson, J.F.; Dent, C.E. γ-aminobutyric acid: A constituent of the potato tuber? Science 1949, 110, 439–440. [Google Scholar]
  50. Bouché, N.; Fromm, H. GABA in plants: Just a metabolite? Trends Plant Sci. 2004, 9, 110–115. [Google Scholar] [CrossRef]
  51. Lunt, G.G. GABA and GABA receptors in invertebrates. Semin. Neurosci. 1991, 3, 251–258. [Google Scholar] [CrossRef]
  52. Ffrench-Constant, R.H.; Rocheleau, T.A.; Steichen, J.C.; Chalmers, A.E. A point mutation in a Drosophila GABA receptor confers insecticide resistance. Nature 1993, 363, 449–451. [Google Scholar] [CrossRef] [PubMed]
  53. Usherwood, P.N.; Grundfest, H. Peripheral inhibition in skeletal muscle of insects. J. Neurophysiol. 1965, 28, 497–518. [Google Scholar] [CrossRef] [PubMed]
  54. Tomita, H.; Yokooji, Y.; Ishibashi, T.; Imanaka, T.; Atomia, H. An archaeal glutamate decarboxylase homolog functions as an aspartate decarboxylase and is involved in β-Alanine and coenzyme a biosynthesis. J. Bacteriol. 2014, 196, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  55. Esclapez, J.; Camacho, M.; Pire, C.; Bautista, V.; Vegara, A.; Pedro-Roig, L.; Pérez-Pomares, F.; Martínez-Espinosa, R.M.; Bonete, M.J. Recent advances in the nitrogen metabolism in Haloarchaea and its biotechnological applications. In Biotechnology of Extremophiles; Springer: Berlin/Heidelberg, Germany, 2016; pp. 273–301. [Google Scholar]
  56. Falb, M.; Müller, K.; Königsmaier, L.; Oberwinkler, T.; Horn, P.; von Gronau, S.; Gonzalez, O.; Pfeiffer, F.; Bornberg-Bauer, E.; Oesterhelt, D. Metabolism of halophilic archaea. Extremophiles 2008, 12, 177–196. [Google Scholar] [CrossRef]
  57. Bouché, N.; Lacombe, B.; Fromm, H. GABA signaling: A conserved and ubiquitous mechanism. Trends Cell Biol. 2003, 13, 607–610. [Google Scholar] [CrossRef] [PubMed]
  58. Quillin, S.J.; Tran, P.; Prindle, A. Potential roles for gamma-aminobutyric acid signaling in bacterial communities. Bioelectricity 2021, 3, 120–125. [Google Scholar] [CrossRef] [PubMed]
  59. Morse, D.E.; Hooker, N.; Duncan, H.; Jensen, L. γ-Aminobutyric acid, a neurotransmitter, induces planktonic abalone larvae to settle and begin metamorphosis. Science 1979, 204, 407–410. [Google Scholar] [CrossRef]
  60. Solomon, P.S.; Oliver, R.P. The nitrogen content of the tomato leaf apoplast increases during infection by Cladosporium fulvum. Planta 2001, 213, 241–249. [Google Scholar] [CrossRef]
  61. Bown, A.W.; MacGregor, K.B.; Shelp, B.J. Gamma-aminobutyrate: Defense against invertebrate pests? Trends Plant Sci. 2006, 11, 424–427. [Google Scholar] [CrossRef]
  62. Chevrot, R.; Rosen, R.; Haudecoeur, E.; Cirou, A.; Shelp, B.J.; Ron, E.; Faure, D. GABA controls the level of quorum-sensing signal in Agrobacterium tumefaciens. Proc. Natl. Acad. Sci. USA 2006, 103, 7460–7464. [Google Scholar] [CrossRef]
  63. Zhang, H.; Chen, Y.; Yang, L.; Shen, Y. Regulation of γ-aminobutyric acid on plant growth and development and stress resistance Zhiwu Shengli Xuebao/Plant Physiol. J. 2020, 56, 600–612. [Google Scholar]
  64. Yuan, D.; Wu, X.; Gong, B.; Huo, R.; Zhao, L.; Li, J.; Lü, G.; Gao, H. GABA Metabolism, transport and their roles and mechanisms in the regulation of abiotic stress (hypoxia, salt, drought) resistance in plants. Metabolites 2023, 13, 347. [Google Scholar] [CrossRef] [PubMed]
  65. Ramesh, S.A.; Tyerman, S.D.; Gilliham, M.; Xu, B. γ-Aminobutyric acid (GABA) signalling in plants. Cell. Mol. Life Sci. 2017, 74, 1577–1603. [Google Scholar] [CrossRef] [PubMed]
  66. Ngo, D.H.; Vo, T.S. An updated review on pharmaceutical properties of gamma-aminobutyric acid. Molecules 2019, 24, 2678. [Google Scholar] [CrossRef] [PubMed]
  67. Boonstra, E.; de Kleijn, R.; Colzato, L.S.; Alkemade, A.; Forstmann, B.U.; Nieuwenhuis, S. Neurotransmitters as food supplements: The effects of GABA on brain and behavior. Front. Psychol. 2015, 6, 6–11. [Google Scholar] [CrossRef] [PubMed]
  68. Ghose, S.; Winter, M.K.; McCarson, K.E.; Tamminga, C.A.; Enna, S.J. The GABA B receptor as a target for antidepressant drug action. Br. J. Pharmacol. 2011, 162, 1–17. [Google Scholar] [CrossRef] [PubMed]
  69. Abdou, A.M.; Higashiguchi, S.; Horie, K.; Kim, M.; Hatta, H.; Yokogoshi, H. Relaxation and immunity enhancement effects of γ-Aminobutyric acid (GABA) administration in humans. BioFactors 2006, 26, 201–208. [Google Scholar] [CrossRef]
  70. An, J.J.; Seok, H.; Ha, E.M. GABA-producing Lactobacillus plantarum inhibits metastatic properties and induces apoptosis of 5-FU-resistant colorectal cancer cells via GABAB receptor signaling. J. Microbiol. 2021, 59, 202–216. [Google Scholar] [CrossRef]
  71. Hosseini Dastgerdi, A.; Sharifi, M.; Soltani, N. GABA administration improves liver function and insulin resistance in offspring of type 2 diabetic rats. Sci. Rep. 2021, 11, 23155. [Google Scholar] [CrossRef]
  72. Nikmaram, N.; Dar, B.; Roohinejad, S.; Koubaa, M.; Barba, F.J.; Greiner, R.; Johnson, S.K. Recent advances in γ-aminobutyric acid (GABA) properties in pulses: An overview. J. Sci. Food Agric. 2017, 97, 2681–2689. [Google Scholar] [CrossRef]
  73. Fenalti, G.; Law, R.H.P.; Buckle, A.M.; Langendorf, C.; Tuck, K.; Rosado, C.J.; Faux, N.G.; Mahmood, K.; Hampe, C.S.; Banga, J.P.; et al. GABA production by glutamic acid decarboxylase is regulated by a dynamic catalytic loop. Nat. Struct. Mol. Biol. 2007, 14, 280–286. [Google Scholar] [CrossRef] [PubMed]
  74. Feehily, C.; Karatzas, K.A.G. Role of glutamate metabolism in bacterial responses towards acid and other stresses. J. Appl. Microbiol. 2013, 114, 11–24. [Google Scholar] [CrossRef]
  75. Feehily, C.; O’Byrne, C.P.; Karatzas, K.A.G. Functional γ-aminobutyrate shunt in Listeria monocytogenes: Role in acid tolerance and succinate biosynthesis. Appl. Environ. Microbiol. 2013, 79, 74–80. [Google Scholar] [CrossRef] [PubMed]
  76. Cotter, P.D.; Gahan, C.G.M.; Hill, C. A glutamate decarboxylase system protects Listeria monocytogenes in gastric fluid. Mol. Microbiol. 2001, 40, 465–475. [Google Scholar] [CrossRef] [PubMed]
  77. Ma, D.; Lu, P.; Yan, C.; Fan, C.; Yin, P.; Wang, J.; Shi, Y. Structure and mechanism of a glutamate-GABA antiporter. Nature 2012, 483, 632–636. [Google Scholar] [CrossRef] [PubMed]
  78. Lyu, C.; Zhao, W.; Peng, C.; Hu, S.; Fang, H.; Hua, Y.; Yao, S.; Huang, J.; Mei, L. Exploring the contributions of two glutamate decarboxylase isozymes in Lactobacillus brevis to acid resistance and γ-aminobutyric acid production. Microb. Cell Fact. 2018, 17, 180. [Google Scholar] [CrossRef]
  79. Wu, Q.; Tun, H.M.; Law, Y.-S.; Khafipour, E.; Shah, N.P. Common distribution of gad operon in Lactobacillus brevis and its GadA contributes to efficient GABA synthesis toward cytosolic near-neutral pH. Front. Microbiol. 2017, 8, 206. [Google Scholar] [CrossRef]
  80. Sezgin, E.; Tekin, B. Molecular evolution and population genetics of glutamate decarboxylase acid resistance pathway in lactic acid bacteria. Front. Genet. 2023, 14, 1027156. [Google Scholar] [CrossRef]
  81. Nakatani, Y.; Fukao, M.; Fukaya, T. Genome sequence of Lactobacillus plantarum KB1253, a gamma-aminobutyric acid (GABA) producer used in GABA-enriched tomato juice production. Microbiol. Resour. Announc. 2019, 8, 10–1128. [Google Scholar] [CrossRef]
  82. Valenzuela, J.A.; Flórez, A.B.; Vázquez, L.; Vasek, O.M.; Mayo, B. Production of γ-aminobutyric acid (GABA) by lactic acid bacteria strains isolated from traditional, starter-free dairy products made of raw milk. Benef. Microbes 2019, 10, 579–587. [Google Scholar] [CrossRef]
  83. Surachat, K.; Deachamag, P.; Kantachote, D.; Wonglapsuwan, M.; Jeenkeawpiam, K.; Chukamnerd, A. In silico comparative genomics analysis of Lactiplantibacillus plantarum DW12, a potential gamma-aminobutyric acid (GABA)-producing strain. Microbiol. Res. 2021, 251, 126833. [Google Scholar] [CrossRef] [PubMed]
  84. Xu, N.; Wei, L.; Liu, J. Biotechnological advances and perspectives of gamma-aminobutyric acid production. World J. Microbiol. Biotechnol. 2017, 33, 64. [Google Scholar] [CrossRef] [PubMed]
  85. Hou, D.; Tang, J.; Feng, Q.; Niu, Z.; Shen, Q.; Wang, L.; Zhou, S. Gamma-aminobutyric acid (GABA): A comprehensive review of dietary sources, enrichment technologies, processing effects, health benefits, and its applications. Crit. Rev. Food Sci. Nutr. 2023, 1–23. [Google Scholar] [CrossRef] [PubMed]
  86. Hudec, J.; Kobida, L.; Čanigová, M.; Lacko-Bartošová, M.; Ložek, O.; Chlebo, P.; Mrázová, J.; Ducsay, L.; Bystrická, J. Production of γ-aminobutyric acid by microorganisms from different food sources. J. Sci. Food Agric. 2015, 95, 1190–1198. [Google Scholar] [CrossRef] [PubMed]
  87. Aoki, H.; Uda, I.; Tagami, K.; Furuya, Y.; Endo, Y.; Fujimoto, K. The production of a new tempeh-like fermented soybean containing a high level of γ-aminobutyric acid by anaerobic incubation with Rhizopus. Biosci. Biotechnol. Biochem. 2003, 67, 1018–1023. [Google Scholar] [CrossRef] [PubMed]
  88. Mohd Ali, N.; Mohd Yusof, H.; Long, K.; Yeap, S.K.; Ho, W.Y.; Beh, B.K.; Koh, S.P.; Abdullah, M.P.; Alitheen, N.B. Antioxidant and hepatoprotective effect of aqueous extract of germinated and fermented mung bean on ethanol-mediated liver damage. Biomed Res. Int. 2013, 2013, 693613. [Google Scholar] [CrossRef] [PubMed]
  89. Koh, S.P.; Jamaluddin, A.; Alitheen Mohd-Ali, N.B.; Mohd-Yusof, N.H.; Yeap, S.K.; Long, K. Nutritive value between fermented and germinated soybean: γ-aminobutyric acid, amino acids content and antioxidant properties. Borneo Sci. 2012, 107–116. [Google Scholar]
  90. Masuda, K.; Guo, X.-F.; Uryu, N.; Hagiwara, T.; Watabe, S. Isolation of marine yeasts collected from the pacific ocean showing a high production of γ-aminobutyric acid. Biosci. Biotechnol. Biochem. 2008, 72, 3265–3272. [Google Scholar] [CrossRef]
  91. Guo, X.-F.; Aoki, H.; Hagiwara, T.; Masuda, K.; Watabe, S. Identification of high γ-aminobutyric acid producing marine yeast strains by physiological and biochemical characteristics and gene sequence analyses. Biosci. Biotechnol. Biochem. 2009, 73, 1527–1534. [Google Scholar] [CrossRef]
  92. Ma, Y.; Cheng, Y.; Yin, L.; Wang, J.; Li, L. Effects of processing and NaCl on Angiotensin I-Converting Enzyme inhibitory activity and γ-aminobutyric acid content during sufu manufacturing. Food Bioprocess Technol. 2013, 6, 1782–1789. [Google Scholar] [CrossRef]
  93. Zhang, Q.; Sun, Q.; Tan, X.; Zhang, S.; Zeng, L.; Tang, J.; Xiang, W. Characterization of γ-aminobutyric acid (GABA)-producing Saccharomyces cerevisiae and coculture with Lactobacillus plantarum for mulberry beverage brewing. J. Biosci. Bioeng. 2020, 129, 447–453. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, L.; Yue, Y.; Wang, X.; Dai, W.; Piao, C.; Yu, H. Optimization of fermentation for γ-aminobutyric acid (GABA) production by yeast Kluyveromyces marxianus C21 in okara (soybean residue). Bioprocess Biosyst. Eng. 2022, 45, 1111–1123. [Google Scholar] [CrossRef] [PubMed]
  95. Perpetuini, G.; Tittarelli, F.; Battistelli, N.; Suzzi, G.; Tofalo, R. γ-aminobutyric acid production by Kluyveromyces marxianus strains. J. Appl. Microbiol. 2020, 129, 1609–1619. [Google Scholar] [CrossRef] [PubMed]
  96. Han, S.-M.; Jeon, S.-J.; Lee, H.-B.; Lee, J.-S. Screening of γ-aminobutyric acid (GABA)-producing wild yeasts and their microbiological characteristics. Korean J. Mycol. 2016, 44, 87–93. [Google Scholar]
  97. Ab Kadir, S.; Wan-Mohtar, W.A.A.Q.I.; Mohammad, R.; Abdul Halim Lim, S.; Sabo Mohammed, A.; Saari, N. Evaluation of commercial soy sauce koji strains of Aspergillus oryzae for γ-aminobutyric acid (GABA) production. J. Ind. Microbiol. Biotechnol. 2016, 43, 1387–1395. [Google Scholar] [CrossRef] [PubMed]
  98. Hajar-Azhari, S.; Wan-Mohtar, W.A.A.Q.I.; Ab Kadir, S.; Rahim, M.H.A.; Saari, N. Evaluation of a Malaysian soy sauce koji strain Aspergillus oryzae NSK for γ-aminobutyric acid (GABA) production using different native sugars. Food Sci. Biotechnol. 2018, 27, 479–488. [Google Scholar] [CrossRef] [PubMed]
  99. Wan-Mohtar, W.A.A.Q.I.; Ab Kadir, S.; Halim-Lim, S.A.; Ilham, Z.; Hajar-Azhari, S.; Saari, N. Vital parameters for high gamma-aminobutyric acid (GABA) production by an industrial soy sauce koji Aspergillus oryzae NSK in submerged-liquid fermentation. Food Sci. Biotechnol. 2019, 28, 1747–1757. [Google Scholar] [CrossRef]
  100. Shin Yee, C.; Sohedein, M.N.A.; Poh Suan, O.; Weng Loen, A.W.; Abd Rahim, M.H.; Soumaya, S.; Ilham, Z.; Wan-Mohtar, W.A.A.Q.I. The production of functional γ-aminobutyric acid Malaysian soy sauce koji and moromi using the trio of Aspergillus oryzae NSK, Bacillus cereus KBC, and the newly identified Tetragenococcus halophilus KBC in liquid-state fermentation. Futur. Foods 2021, 4, 100055. [Google Scholar] [CrossRef]
  101. Zareian, M.; Oskoueian, E.; Majdinasab, M.; Forghani, B. Production of GABA-enriched: Idli with ACE inhibitory and antioxidant properties using Aspergillus oryzae: The antihypertensive effects in spontaneously hypertensive rats. Food Funct. 2020, 11, 4304–4313. [Google Scholar] [CrossRef]
  102. Cai, S.; Gao, F.; Zhang, X.; Wang, O.; Wu, W.; Zhu, S.; Zhang, D.; Zhou, F.; Ji, B. Evaluation of γ- aminobutyric acid, phytate and antioxidant activity of tempeh-like fermented oats (Avena sativa L.) prepared with different filamentous fungi. J. Food Sci. Technol. 2014, 51, 2544–2551. [Google Scholar] [CrossRef]
  103. Kono, I.; Himeno, K. Changes in γ-aminobutyric acid content during beni-koji making. Biosci. Biotechnol. Biochem. 2000, 64, 617–619. [Google Scholar] [CrossRef]
  104. Su, Y.-C.; Wang, J.-J.; Lin, T.-T.; Pan, T.-M. Production of the secondary metabolites γ-aminobutyric acid and monacolin K by Monascus. J. Ind. Microbiol. Biotechnol. 2003, 30, 41–46. [Google Scholar] [CrossRef]
  105. Suwanmanon, K.; Hsieh, P.-C. Isolating Bacillus subtilis and optimizing its fermentative medium for GABA and nattokinase production. CYTA J. Food 2014, 12, 282–290. [Google Scholar] [CrossRef]
  106. Wan-Mohtar, W.A.A.Q.I.; Sohedein, M.N.A.; Ibrahim, M.F.; Kadir, S.A.; Suan, O.P.; Loen, A.W.W.; Sassi, S.; Ilham, Z. Isolation, identification, and optimization of γ-aminobutyric acid (GABA)-producing Bacillus cereus Strain KBC from a commercial soy Sauce moromi in submerged-liquid fermentation. Processes 2020, 8, 652. [Google Scholar] [CrossRef]
  107. Shi, F.; Li, Y. Synthesis of γ-aminobutyric acid by expressing Lactobacillus brevis-derived glutamate decarboxylase in the Corynebacterium glutamicum strain ATCC 13032. Biotechnol. Lett. 2011, 33, 2469–2474. [Google Scholar] [CrossRef] [PubMed]
  108. Jeng, K.-C.; Chen, C.-S.; Fang, Y.-P.; Hou, R.C.-W.; Chen, Y.-S. Effect of microbial fermentation on content of statin, GABA, and polyphenols in Pu-erh tea. J. Agric. Food Chem. 2007, 55, 8787–8792. [Google Scholar] [CrossRef] [PubMed]
  109. Otaru, N.; Ye, K.; Mujezinovic, D.; Berchtold, L.; Constancias, F.; Cornejo, F.A.; Krzystek, A.; de Wouters, T.; Braegger, C.; Lacroix, C.; et al. GABA production by human intestinal Bacteroides spp.: Prevalence, regulation, and role in acid stress tolerance. Front. Microbiol. 2021, 12, 656895. [Google Scholar] [CrossRef]
  110. Duranti, S.; Ruiz, L.; Lugli, G.A.; Tames, H.; Milani, C.; Mancabelli, L.; Mancino, W.; Longhi, G.; Carnevali, L.; Sgoifo, A.; et al. Bifidobacterium adolescentis as a key member of the human gut microbiota in the production of GABA. Sci. Rep. 2020, 10, 14112. [Google Scholar] [CrossRef]
  111. Barrett, E.; Ross, R.P.; O’Toole, P.W.; Fitzgerald, G.F.; Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef]
  112. Li, H.; Cao, Y. Lactic acid bacterial cell factories for gamma-aminobutyric acid. Amino Acids 2010, 39, 1107–1116. [Google Scholar] [CrossRef]
  113. Devi, P.B.; Rajapuram, D.R.; Jayamanohar, J.; Verma, M.; Kavitake, D.; Meenachi Avany, B.A.; Rani, P.U.; Ravi, R.; Shetty, P.H. Gamma-aminobutyric acid (GABA) production by potential probiotic strains of indigenous fermented foods origin and RSM based production optimization. LWT 2023, 176, 114511. [Google Scholar] [CrossRef]
  114. Pannerchelvan, S.; Rios-Solis, L.; Faizal Wong, F.W.; Zaidan, U.H.; Wasoh, H.; Mohamed, M.S.; Tan, J.S.; Mohamad, R.; Halim, M. Strategies for improvement of gamma-aminobutyric acid (GABA) biosynthesis via lactic acid bacteria (LAB) fermentation. Food Funct. 2023, 14, 3929–3948. [Google Scholar] [CrossRef] [PubMed]
  115. Siragusa, S.; De Angelis, M.; Di Cagno, R.; Rizzello, C.G.; Coda, R.; Gobbetti, M. Synthesis of γ-aminobutyric acid by lactic acid bacteria isolated from a variety of Italian cheeses. Appl. Environ. Microbiol. 2007, 73, 7283–7290. [Google Scholar] [CrossRef] [PubMed]
  116. Khanlari, Z.; Moayedi, A.; Ebrahimi, P.; Khomeiri, M.; Sadeghi, A. Enhancement of γ-aminobutyric acid (GABA) content in fermented milk by using Enterococcus faecium and Weissella confusa isolated from sourdough. J. Food Process. Preserv. 2021, 45, e15869. [Google Scholar] [CrossRef]
  117. Franciosi, E.; Carafa, I.; Nardin, T.; Schiavon, S.; Poznanski, E.; Cavazza, A.; Larcher, R.; Tuohy, K.M. Biodiversity and γ-aminobutyric acid production by lactic acid bacteria isolated from traditional alpine raw cow’s milk cheeses. Biomed Res. Int. 2015, 2015, 625740. [Google Scholar] [CrossRef] [PubMed]
  118. Monteagudo-Mera, A.; Fanti, V.; Rodriguez-Sobstel, C.; Gibson, G.; Wijeyesekera, A.; Karatzas, K.-A.; Chakrabarti, B. Gamma aminobutyric acid production by commercially available probiotic strains. J. Appl. Microbiol. 2023, 134, lxac066. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, Z.; Wang, Y.; Dong, Y.; Xiang, F.; Zhang, Y.; Zhang, H.; Sun, Y.; Guo, Z. Characterization of two novel pentose-fermenting and GABA-producing species: Levilactobacillus tujiorum sp. nov. and Secundilactobacillus angelensis sp. nov. Isolated from a solid-state fermented zha-chili. Syst. Appl. Microbiol. 2022, 45, 126344. [Google Scholar] [CrossRef]
  120. Amatachaya, A.; Siramolpiwat, S.; Kraisorn, M.; Yasiri, A. Gamma-aminobutyric acid (GABA) producing probiotic Lactiplantibacillus Pentosus isolated from fermented spider plant (pak sian dong) in Thailand. J. Pure Appl. Microbiol. 2023, 17, 354–361. [Google Scholar] [CrossRef]
  121. Garavand, F.; Daly, D.F.; Gómez-Mascaraque, L.G. Biofunctional, structural, and tribological attributes of GABA-enriched probiotic yoghurts containing Lacticaseibacillus paracasei alone or in combination with prebiotics. Int. Dairy J. 2022, 129, 105348. [Google Scholar] [CrossRef]
  122. Diez-Gutiérrez, L.; Vicente, L.S.; Sáenz, J.; Esquivel, A.; Barron, L.J.R.; Chávarri, M. Biosynthesis of gamma-aminobutyric acid by Lactiplantibacillus plantarum K16 as an alternative to revalue agri-food by-products. Sci. Rep. 2022, 12, 18904. [Google Scholar] [CrossRef]
  123. Kim, J.; Lee, M.-H.; Kim, M.-S.; Kim, G.-H.; Yoon, S.-S. Probiotic properties and optimization of gamma-aminobutyric acid production by Lactiplantibacillus plantarum FBT215. J. Microbiol. Biotechnol. 2022, 32, 783–791. [Google Scholar] [CrossRef] [PubMed]
  124. Park, S.J.; Kim, D.H.; Kang, H.J.; Shin, M.; Yang, S.Y.; Yang, J.; Jung, Y.H. Enhanced production of γ-aminobutyric acid (GABA) using Lactobacillus plantarum EJ2014 with simple medium composition. LWT 2021, 137, 110443. [Google Scholar] [CrossRef]
  125. Das, D.; Goyal, A. Antioxidant activity and γ-aminobutyric acid (GABA) producing ability of probiotic Lactobacillus plantarum DM5 isolated from Marcha of Sikkim. LWT 2015, 61, 263–268. [Google Scholar] [CrossRef]
  126. Kim, N.Y.; Kim, S.K.; Ra, C.H. Evaluation of gamma-aminobutyric acid (GABA) production by Lactobacillus plantarum using two-step fermentation. Bioprocess Biosyst. Eng. 2021, 44, 2099–2108. [Google Scholar] [CrossRef] [PubMed]
  127. Lee, E.J.; Lee, S.P. Novel bioconversion of sodium glutamate to γ-amino butyric acid by co-culture of Lactobacillus plantarum K154 in Ceriporia lacerata culture broth. Food Sci. Biotechnol. 2014, 23, 1997–2005. [Google Scholar] [CrossRef]
  128. Park, S.Y.; Lee, J.W.; Lim, S.D. The probiotic characteristics and GABA production of Lactobacillus plantarum K154 isolated from kimchi. Food Sci. Biotechnol. 2014, 23, 1951–1957. [Google Scholar] [CrossRef]
  129. Tajabadi, N.; Ebrahimpour, A.; Baradaran, A.; Rahim, R.A.; Mahyudin, N.A.; Manap, M.Y.A.; Bakar, F.A.; Saari, N. Optimization of γ-aminobutyric acid production by Lactobacillus plantarum Taj-Apis362 from honeybees. Molecules 2015, 20, 6654–6669. [Google Scholar] [CrossRef]
  130. Ly, D.; Mayrhofer, S.; Yogeswara, I.B.A.; Nguyen, T.H.; Domig, K.J. Identification, classification and screening for γ-amino-butyric acid production in lactic acid bacteria from cambodian fermented foods. Biomolecules 2019, 9, 768. [Google Scholar] [CrossRef]
  131. Yogeswara, I.B.A.; Kittibunchakul, S.; Rahayu, E.S.; Domig, K.J.; Haltrich, D.; Nguyen, T.H. Microbial production and enzymatic biosynthesis of γ-aminobutyric acid (GABA) using Lactobacillus plantarum FNCC 260 isolated from indonesian fermented foods. Processes 2021, 9, 22. [Google Scholar] [CrossRef]
  132. Zhang, Q.; Zeng, L.; Tan, X.; Tang, J.; Xiang, W. An efficient γ-aminobutyric acid (GABA) producing and nitrite reducing ability of Lactobacillus plantarum BC114 isolated from Chinese paocai. Food Sci. Technol. Res. 2017, 23, 749–755. [Google Scholar] [CrossRef]
  133. Phuengjayaem, S.; Booncharoen, A.; Tanasupawat, S. Characterization and comparative genomic analysis of gamma-aminobutyric acid (GABA)-producing lactic acid bacteria from Thai fermented foods. Biotechnol. Lett. 2021, 43, 1637–1648. [Google Scholar] [CrossRef] [PubMed]
  134. Zareian, M.; Ebrahimpour, A.; Sabo Mohamed, A.K.; Saari, N. Modeling of glutamic acid production by Lactobacillus plantarum MNZ. Electron. J. Biotechnol. 2013, 16, 12. [Google Scholar] [CrossRef]
  135. Park, S.Y.; Kim, K.S.; Lee, M.K.; Lim, S.D. Physiological characteristics and GABA production of Lactobacillus plantarum K255 isolated from kimchi. Korean J. Food Sci. Anim. Resour. 2013, 33, 595–602. [Google Scholar] [CrossRef]
  136. Lee, H.-J.; Son, J.-Y.; Lee, S.-J.; Lee, H.-S.; Lee, B.-J.; Choi, I.-S.; Sohn, J.H. Production of gamma-aminobutyric acid (GABA) by Lactobacillus plantarum subsp. plantarum B-134 isolated from makgeolli, traditional Korean rice wine. J. Life Sci. 2017, 27, 567–574. [Google Scholar] [CrossRef]
  137. Krongkeha, W. Isolation and identification of GABA-producing lactic acid bacteria from fermented foods. RMUTSB J. 2022, 10, 66–77. [Google Scholar]
  138. Kim, J.; Yoon, Y.-W.; Kim, M.-S.; Lee, M.-H.; Kim, G.-A.; Bae, K.; Yoon, S.-S. Gamma-aminobutyric acid fermentation in MRS-based medium by the fructophilic Lactiplantibacillus plantarum Y7. Food Sci. Biotechnol. 2022, 31, 333–341. [Google Scholar] [CrossRef]
  139. Tanamool, V.; Hongsachart, P.; Soemphol, W. Screening and characterisation of gamma-aminobutyric acid (GABA) producing lactic acid bacteria isolated from Thai fermented fish (Plaa-som) in Nong Khai and its application in Thai fermented vegetables (Som-pak). Food Sci. Technol. 2020, 40, 483–490. [Google Scholar] [CrossRef]
  140. Cai, H.; Li, X.; Li, D.; Liu, W.; Han, Y.; Xu, X.; Yang, P.; Meng, K. Optimization of gamma-aminobutyric acid production by Lactiplantibacillus plantarum FRT7 from Chinese Paocai. Foods 2023, 12, 3034. [Google Scholar] [CrossRef]
  141. Wang, D.; Wang, Y.; Lan, H.; Wang, K.; Zhao, L.; Hu, Z. Enhanced production of γ-aminobutyric acid in litchi juice fermented by Lactobacillus plantarum HU-C2W. Food Biosci. 2021, 42, 101155. [Google Scholar] [CrossRef]
  142. Man, D. De man, rogosa and sharpe (MRS) agar. Prog. Ind. Microbiol. 1995, 34, 362–363. [Google Scholar]
  143. Gomaa, E.Z. Enhancement of γ-amminobutyric acid production by co-culturing of two lactobacilli strains. Asian J. Biotechnol. 2015, 7, 108–118. [Google Scholar] [CrossRef]
  144. Shan, Y.; Man, C.X.; Han, X.; Li, L.; Guo, Y.; Deng, Y.; Li, T.; Zhang, L.W.; Jiang, Y.J. Evaluation of improved γ-aminobutyric acid production in yogurt using Lactobacillus plantarum NDC75017. J. Dairy Sci. 2015, 98, 2138–2149. [Google Scholar] [CrossRef] [PubMed]
  145. Lammens, T.M.; De Biase, D.; Franssen, M.C.R.; Scott, E.L.; Sanders, J.P.M. The application of glutamic acid α-decarboxylase for the valorization of glutamic acid. Green Chem. 2009, 11, 1562. [Google Scholar] [CrossRef]
  146. Hussin, F.S.; Chay, S.Y.; Hussin, A.S.M.; Wan Ibadullah, W.Z.; Muhialdin, B.J.; Abd Ghani, M.S.; Saari, N. GABA enhancement by simple carbohydrates in yoghurt fermented using novel, self-cloned Lactobacillus plantarum Taj-Apis362 and metabolomics profiling. Sci. Rep. 2021, 11, 9417. [Google Scholar] [CrossRef] [PubMed]
  147. Lin, Q.; Li, D.; Qin, H. Molecular cloning, expression, and immobilization of glutamate decarboxylase from Lactobacillus fermentum YS2. Electron. J. Biotechnol. 2017, 27, 8–13. [Google Scholar] [CrossRef]
  148. van de Guchte, M.; Serror, P.; Chervaux, C.; Smokvina, T.; Ehrlich, S.D.; Maguin, E. Stress responses in lactic acid bacteria. Antonie Van Leeuwenhoek 2002, 82, 187–216. [Google Scholar] [CrossRef] [PubMed]
  149. Shin, S.; Kim, H.; Joo, Y.; Lee, S.-J.; Lee, Y.-J.; Lee, S.J.; Lee, D. Characterization of glutamate decarboxylase from Lactobacillus plantarum and Its C-terminal function for the pH dependence of activity. J. Agric. Food Chem. 2014, 62, 12186–12193. [Google Scholar] [CrossRef]
  150. Wang, W.; He, J.; Pan, D.; Wu, Z.; Guo, Y.; Zeng, X.; Lian, L. Metabolomics analysis of Lactobacillus plantarum ATCC 14917 adhesion activity under initial acid and alkali stress. PLoS ONE 2018, 13, e0196231. [Google Scholar] [CrossRef]
  151. Yogeswara, I.B.A.; Kusumawati, I.G.A.W.; Nursini, N.W.; Mariyatun, M.; Rahayu, E.S.; Haltrich, D. Health-promoting role of fermented pigeon pea (Cajanus cajan L (Mill)) milk enriched with γ-aminobutyric acid (GABA) using probiotic Lactiplantibacillus plantarum Dad-13. Fermentation 2023, 9, 587. [Google Scholar] [CrossRef]
  152. Yang, T.; Rao, Z.; Kimani, B.G.; Xu, M.; Zhang, X.; Yang, S.T. Two-step production of gamma-aminobutyric acid from cassava powder using Corynebacterium glutamicum and Lactobacillus plantarum. J. Ind. Microbiol. Biotechnol. 2015, 42, 1157–1165. [Google Scholar] [CrossRef]
  153. Oh, S.-H.; Moon, Y.-J.; Oh, C.-H. γ-aminobutyric acid (GABA) content of selected uncooked foods. Prev. Nutr. Food Sci. 2003, 8, 75–78. [Google Scholar] [CrossRef]
  154. Coda, R.; Rizzello, C.G.; Gobbetti, M. Use of sourdough fermentation and pseudo-cereals and leguminous flours for the making of a functional bread enriched of γ-aminobutyric acid (GABA). Int. J. Food Microbiol. 2010, 137, 236–245. [Google Scholar] [CrossRef] [PubMed]
  155. Di Cagno, R.; Mazzacane, F.; Rizzello, C.G.; De Angelis, M.; Giuliani, G.; Meloni, M.; De Servi, B.; Gobbetti, M. Synthesis of γ-aminobutyric acid (GABA) by Lactobacillus plantarum DSM19463: Functional grape must beverage and dermatological applications. Appl. Microbiol. Biotechnol. 2010, 86, 731–741. [Google Scholar] [CrossRef]
  156. Nakatani, Y.; Fukaya, T.; Kishino, S.; Ogawa, J. Production of GABA-enriched tomato juice by Lactiplantibacillus plantarum KB1253. J. Biosci. Bioeng. 2022, 134, 424–431. [Google Scholar] [CrossRef] [PubMed]
  157. Park, K.B.; Oh, S.H. Production of yogurt with enhanced levels of gamma-aminobutyric acid and valuable nutrients using lactic acid bacteria and germinated soybean extract. Bioresour. Technol. 2007, 98, 1675–1679. [Google Scholar] [CrossRef] [PubMed]
  158. Tung, Y.T.; Lee, B.H.; Liu, C.F.; Pan, T.M. Optimization of culture condition for ACEI and GABA production by lactic acid bacteria. J. Food Sci. 2011, 76, 585–591. [Google Scholar] [CrossRef]
  159. Rizzello, C.G.; Cassone, A.; Di Cagno, R.; Gobbetti, M. Synthesis of angiotensin I-converting enzyme (ACE)-inhibitory peptides and γ-aminobutyric acid (GABA) during sourdough fermentation by selected lactic acid bacteria. J. Agric. Food Chem. 2008, 56, 6936–6943. [Google Scholar] [CrossRef]
  160. Alharbi, H.F.; Algonaiman, R.; Barakat, H. Ameliorative and antioxidative potential of Lactobacillus plantarum-fermented oat (Avena sativa) and fermented oat supplemented with sidr honey against streptozotocin-induced type 2 diabetes in rats. Antioxidants 2022, 11, 1122. [Google Scholar] [CrossRef]
  161. Algonaiman, R.; Alharbi, H.F.; Barakat, H. Antidiabetic and hypolipidemic efficiency of Lactobacillus plantarum fermented oat (Avena sativa) extract in streptozotocin-induced diabetes in rats. Fermentation 2022, 8, 267. [Google Scholar] [CrossRef]
  162. Ratanaburee, A.; Kantachote, D.; Charernjiratrakul, W.; Penjamras, P.; Chaiyasut, C. Enhancement of γ-aminobutyric acid in a fermented red seaweed beverage by starter culture Lactobacillus plantarum DW12. Electron. J. Biotechnol. 2011, 14, 1. [Google Scholar]
  163. Hayisama-ae, W.; Kantachote, D.; Bhongsuwan, D.; Nokkaew, U.; Chaiyasut, C. A potential synbiotic beverage from fermented red seaweed (Gracilaria fisheri) using Lactobacillus plantarum DW12. Int. Food Res. J. 2014, 21, 1789–1796. [Google Scholar]
  164. Kantachote, D.; Ratanaburee, A.; Hayisama-ae, W.; Sukhoom, A.; Nunkaew, T. The use of potential probiotic Lactobacillus plantarum DW12 for producing a novel functional beverage from mature coconut water. J. Funct. Foods 2017, 32, 401–408. [Google Scholar] [CrossRef]
  165. Rahayu, E.S.; Yogeswara, A.; Mariyatun; Windiarti, L.; Utami, T.; Watanabe, K. Molecular characteristics of indigenous probiotic strains from Indonesia. Int. J. Probiotics Prebiotics 2016, 11, 109–116. [Google Scholar]
  166. Fuller, D.Q.; Murphy, C.; Kingwell-Banham, E.; Castillo, C.C.; Naik, S. Cajanus cajan (L.) Millsp. origins and domestication: The South and Southeast Asian archaeobotanical evidence. Genet. Resour. Crop Evol. 2019, 66, 1175–1188. [Google Scholar] [CrossRef]
  167. Yao, P.; Gao, Y.; Simal-Gandara, J.; Farag, M.A.; Chen, W.; Yao, D.; Delmas, D.; Chen, Z.; Liu, K.; Hu, H.; et al. Litchi (Litchi chinensis Sonn.): A comprehensive review of phytochemistry, medicinal properties, and product development. Food Funct. 2021, 12, 9527–9548. [Google Scholar] [CrossRef] [PubMed]
  168. Lewmanomont, K.; Chirapart, A. Biodiversity, cultivation and utilization of seaweeds in Thailand: An overview. In Sustainable Global Resources of Seaweeds Volume 1; Springer International Publishing: Cham, Switzerland, 2022; pp. 91–107. [Google Scholar]
  169. Camargo Prado, F.; De Dea Lindner, J.; Inaba, J.; Thomaz-Soccol, V.; Kaur Brar, S.; Soccol, C.R. Development and evaluation of a fermented coconut water beverage with potential health benefits. J. Funct. Foods 2015, 12, 489–497. [Google Scholar] [CrossRef]
  170. Zarei, F.; Nateghi, L.; Eshaghi, M.R.; Abadi, M.E.T. Production of gamma-aminobutyric acid (GABA) in whey protein drink during fermentation by Lactobacillus plantarum. J. Microbiol. Biotechnol. Food Sci. 2020, 9, 1087–1092. [Google Scholar] [CrossRef]
  171. Zarei, F.; Nateghi, L.; Eshaghi, M.R.; Taj Abadi, M.E. Optimization of gamma-aminobutyric acid production in probiotics extracted from local dairy products in West Region of Iran using MRS broth and whey protein media. Appl. Food Biotechnol. 2018, 5, 233–242. [Google Scholar]
  172. Xuan Phong, H.; Quoc Viet, L.; Minh Chau, L.; Dang Long, B.H.; Thanh, N.N.; Tan Phat, D.; Truong, L.D. Isolation and selection of lactic acid bacteria with the capacity of producing γ-aminobutyric acid (GABA) and antimicrobial activity: Its application in fermented meat product. Curr. Nutr. Food Sci. 2023, 19, 831–837. [Google Scholar] [CrossRef]
  173. Rezaei, M.; Ghasemi, Y.; Sharifan, A.; Bakhoda, H. Producing and analyzing gamma-aminobutyric acid containing probiotic black grape juice using Lactobacillus plantarum plantarum IBRC(10817) and Lactobacillus brevis IBRC(10818). Meas. Food 2022, 8, 100056. [Google Scholar] [CrossRef]
  174. Diez-Gutiérrez, L.; San Vicente, L.; Sáenz, J.; Barron, L.J.R.; Chávarri, M. Characterisation of the probiotic potential of Lactiplantibacillus plantarum K16 and its ability to produce the postbiotic metabolite γ-aminobutyric acid. J. Funct. Foods 2022, 97, 105230. [Google Scholar] [CrossRef]
  175. Lee, X.Y.; Tan, J.S.; Cheng, L.H. Gamma aminobutyric acid (GABA) enrichment in plant-based food—A mini review. Food Rev. Int. 2023, 39, 5864–5885. [Google Scholar] [CrossRef]
  176. Tajabadi, N.; Baradaran, A.; Ebrahimpour, A.; Rahim, R.A.; Bakar, F.A.; Manap, M.Y.A.; Mohammed, A.S.; Saari, N. Overexpression and optimization of glutamate decarboxylase in Lactobacillus plantarum Taj-Apis362 for high gamma-aminobutyric acid production. Microb. Biotechnol. 2015, 8, 623–632. [Google Scholar] [CrossRef] [PubMed]
  177. Hussin, F.S.; Chay, S.Y.; Zarei, M.; Meor Hussin, A.S.; Ibadullah, W.Z.W.; Zaharuddin, N.D.; Wazir, H.; Saari, N. Potentiality of self-cloned Lactobacillus plantarum Taj-Apis362 for enhancing GABA production in yogurt under glucose induction: Optimization and its cardiovascular effect on spontaneous hypertensive rats. Foods 2020, 9, 1826. [Google Scholar] [CrossRef]
  178. Watanabe, Y.; Hayakawa, K.; Ueno, H. Effects of co-culturing lab on GABA production. J. Biol. Macromol. 2011, 11, 3–13. [Google Scholar]
  179. Park, E.-J.; Garcia, C.V.; Youn, S.-J.; Park, C.-D.; Lee, S.-P. Fortification of γ-aminobutyric acid and bioactive compounds in Cucurbita moschata by novel two-step fermentation using Bacillus subtilis and Lactobacillus plantarum. LWT 2019, 102, 22–29. [Google Scholar] [CrossRef]
  180. Lim, J.-S.; Garcia, C.V.; Lee, S.-P. Optimized production of GABA and γ-PGA in a turmeric and roasted soybean mixture co-fermented by Bacillus subtilis and Lactobacillus plantarum. Food Sci. Technol. Res. 2016, 22, 209–217. [Google Scholar] [CrossRef]
  181. Kim, J.E.; Kim, J.S.; Song, Y.C.; Lee, J.; Lee, S.P. Novel bioconversion of sodium glutamate to γ-poly-glutamic acid and γ-amino butyric acid in a mixed fermentation using Bacillus subtilis HA and Lactobacillus plantarum K154. Food Sci. Biotechnol. 2014, 23, 1551–1559. [Google Scholar] [CrossRef]
  182. Kwon, S.-Y.; Garcia, C.V.; Song, Y.-C.; Lee, S.-P. GABA-enriched water dropwort produced by co-fermentation with Leuconostoc mesenteroides SM and Lactobacillus plantarum K154. LWT 2016, 73, 233–238. [Google Scholar] [CrossRef]
  183. Woraratphoka, J.; Innok, S.; Soisungnoen, P.; Tanamool, V.; Soemphol, W. γ-Aminobutyric acid production and antioxidant activities in fresh cheese by Lactobacillus plantarum L10-11. Food Sci. Technol. 2022, 42, e03121. [Google Scholar] [CrossRef]
  184. Zhang, J.; Liu, D.; Zhang, C.; Niu, H.; Xin, X.; Chen, J.; Yi, H.; Liu, D. The impact of Levilactobacillus brevis YSJ3 and Lactiplantibacillus plantarum JLSC2-6 co-culture on gamma-aminobutyric acid yield, volatile and non-volatile metabolites, antioxidant activity, and bacterial community in fermented cauliflower byproducts. Food Chem. 2024, 432, 137169. [Google Scholar] [CrossRef] [PubMed]
  185. Karimian, E.; Moayedi, A.; Khomeiri, M.; Aalami, M.; Mahoonak, A.S. Application of high-GABA producing Lactobacillus plantarum isolated from traditional cabbage pickle in the production of functional fermented whey-based formulate. J. Food Meas. Charact. 2020, 14, 3408–3416. [Google Scholar] [CrossRef]
  186. Servili, M.; Rizzello, C.G.; Taticchi, A.; Esposto, S.; Urbani, S.; Mazzacane, F.; Di Maio, I.; Selvaggini, R.; Gobbetti, M.; Di Cagno, R. Functional milk beverage fortified with phenolic compounds extracted from olive vegetation water, and fermented with functional lactic acid bacteria. Int. J. Food Microbiol. 2011, 147, 45–52. [Google Scholar] [CrossRef] [PubMed]
  187. Kwon, S.Y.; Lee, S.P. Enrichment of gamma-aminobutyric acid (GABA) in old antler extract fermented by Lactobacillus plantarum. Korean J. Food Sci. Technol. 2018, 50, 37–43. [Google Scholar]
  188. Liu, T.-T.; Yang, T.; Zhang, S.; Xia, H.; Rao, Z. Screening, identification and primary optimizing of a strain producing γ-aminobutyric acid from L-glutamic acid. J. Food Sci. Biotechnol. 2010, 29, 742–747. [Google Scholar]
  189. Jang, Y.; Choi, H.E.; Lim, Y.W.; Lee, J.S.; Kim, J.-J. The first report of Ceriporia lacerata (Phanerochaetaceae, Basidiomycota) in Korea. Mycotaxon 2012, 119, 397–403. [Google Scholar] [CrossRef]
  190. Ponomarova, O.; Gabrielli, N.; Sévin, D.C.; Mülleder, M.; Zirngibl, K.; Bulyha, K.; Andrejev, S.; Kafkia, E.; Typas, A.; Sauer, U.; et al. Yeast creates a niche for symbiotic lactic acid bacteria through nitrogen overflow. Cell Syst. 2017, 5, 345–357.e6. [Google Scholar] [CrossRef]
Figure 1. GABA production from L-glutamate by glutamate decarboxylase (GAD) with pyridoxal-5′-phosphate (PLP) as a cofactor.
Figure 1. GABA production from L-glutamate by glutamate decarboxylase (GAD) with pyridoxal-5′-phosphate (PLP) as a cofactor.
Cimb 46 00015 g001
Figure 2. Metabolic pathway of GABA production from the TCA cycle (adapted from Sahab et al. [11]). For higher clarity, this scheme reports only enzymes and relevant substrates/products, omitting coenzymes and other compounds involved in the reactions showed. Abbreviations: PDH, pyruvate dehydrogenase; GDH, glutamate dehydrogenase; GAD, glutamate decarboxylase; GABA-T, GABA transaminase; SSADH, succinic semialdehyde dehydrogenase.
Figure 2. Metabolic pathway of GABA production from the TCA cycle (adapted from Sahab et al. [11]). For higher clarity, this scheme reports only enzymes and relevant substrates/products, omitting coenzymes and other compounds involved in the reactions showed. Abbreviations: PDH, pyruvate dehydrogenase; GDH, glutamate dehydrogenase; GAD, glutamate decarboxylase; GABA-T, GABA transaminase; SSADH, succinic semialdehyde dehydrogenase.
Cimb 46 00015 g002
Table 1. GABA production by Lactiplantibacillus plantarum (previously Lactobacillus plantarum) in different culture media.
Table 1. GABA production by Lactiplantibacillus plantarum (previously Lactobacillus plantarum) in different culture media.
MicroorganismIsolation
Source
Culture MediumGABA
Production
CommentsRefs.
L. plantarum C48cheeseMRS16.0 mg/kgSurvival and GABA production in simulated GI conditions[115]
L. plantarum
CCARM 0067
CCARMCDM≈700 mM (48 h)Anti-proliferative and anti-metastatic activity in HT-29/5FUR cell line[70]
L. plantarum DM5Marcha of SikkimMRS + 100
mM MSG
not quantifiedGABA production has been qualitatively identified by the TLC[125]
L. plantarum KCTC 3103UnknownMRS modified0.67 g/LTwo-stage fermentation: cell grown (stage 1); GABA production (stage 2)[126]
L. plantarum K154kimchibroth fortified with skim milk and 2% MSG15.53 mg/mLCo-culture with Ceriporia lacerata[127]
L. plantarum EJ2014Rice branSM19.8 g/LOptimization of production by the addition of yeast extract[124]
L. plantarum K154kimchiMRS + 30 g/L MSG0.2 g/LPotential probiotic: good resistance to vancomycin and polymyxin B, tolerance to bile juice and low pH[128]
L. plantarum Taj-Apis362honeycomb and stomach of honeybeeMRS + 50 mM MSG7.15 mMculture temperature of 36 °C, initial pH of 5.31 and incubation time of 60 h[129]
L. plantarum 45acambodian fermented foodsMRS + 2% MSG20.34 mMTwo other strains of L. plantarum capable of synthesizing GABA have been identified: 44d (16.47 mM GABA) and 37e (5.63 mM GABA)[130]
L. plantarum FNCC 260indonesian fermented foodsMRS + 25–100 mM MSG809.2 mg/LMSG, PLP, and pyridoxine were shown
to positively affect GABA production
[131]
L. plantarum BC114Sichuan paocai (fermented vegetable)MRS + 20 g/L MSG3.45 g/LL. plantarum BC114
highlighted the ability to produce GABA and reduce nitrates
[132]
L. plantarum LSI2-1Thailand fermented foodGYP + 3% MSG22.94 g/LOnly the gadA as
glutamate decarboxylase
(GAD) was found in the genome
[133]
L. plantarum MNZfermented soybeanMRS3.96 mM6% glucose, 0.7% ammonium nitrate, pH 4.5 and temperature 37 °C.[134]
L. plantarum K255kimchiMRS + 3% MSG821.2 µg/mLthe K255 strain was incubated at 37 °C for 18 h.[135]
L. plantarum FBT215kimchiMRS modified (1% fructose; 2% tryptone, 50 mM MSG)103.7 μg/mLPLP is a major factor influencing
GABA production
[123]
L. plantarum B-134MakgeolliMRS + 3% MSG25 mMoptimum culture condition: 37 °C, pH 5.7 without NaCl[136]
L. plantarum N1-2NhamMRS + 5% MSG0.13 mg/10 gpH of 5.7, without NaCl[137]
L. plantarum Y7kimchiMRS modified (2% fructose, 2% peptone and 175 mM MSG)4.9 μg/mLculture conditions: 37 °C,
pH 6.5, and 48 h.
[138]
L. plantarum L10-11Plaa-somMRS + 4% MSG15.74 g/Laddition of NaCl by up to 7% (w/v)
did not suppress GABA production
[139]
L. plantarum FRT7PaocaiMRS 3% MSG and 2 mmol/L of PLP1158.6 mg/L40 °C; pH of 7.0 for 48 h[140]
L. plantarum HUC2W MRS + 4% MSG3.92 g/Lat 37 °C for 24 h[141]
Abbreviations: MRS, de Man, Rogosa and Sharp medium; GI, gastrointestinal; CCARM, Culture Collection of Antimicrobial Resistant Microbes; CDM, chemical defined medium; HT-29/5FUR, human colon adenocarcinoma cell line (HT-29) resistant to 5-fluorouracil (5-FU); MSG, mono-sodic glutamate; TLC, thin layer chromatography; SM, synthetic medium (consisting of 100 g/L Yeast extract, 10 g/L dextrose, and 22.5 g/L MSG); PLP, pyridoxal 5′-phosphate; GYP, Glucose-yeast extract-peptone; GAD, glutamic decarboxylase.
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

Iorizzo, M.; Paventi, G.; Di Martino, C. Biosynthesis of Gamma-Aminobutyric Acid (GABA) by Lactiplantibacillus plantarum in Fermented Food Production. Curr. Issues Mol. Biol. 2024, 46, 200-220. https://doi.org/10.3390/cimb46010015

AMA Style

Iorizzo M, Paventi G, Di Martino C. Biosynthesis of Gamma-Aminobutyric Acid (GABA) by Lactiplantibacillus plantarum in Fermented Food Production. Current Issues in Molecular Biology. 2024; 46(1):200-220. https://doi.org/10.3390/cimb46010015

Chicago/Turabian Style

Iorizzo, Massimo, Gianluca Paventi, and Catello Di Martino. 2024. "Biosynthesis of Gamma-Aminobutyric Acid (GABA) by Lactiplantibacillus plantarum in Fermented Food Production" Current Issues in Molecular Biology 46, no. 1: 200-220. https://doi.org/10.3390/cimb46010015

Article Metrics

Back to TopTop