You are on page 1of 12

Enzyme and Microbial Technology 143 (2021) 109722

Contents lists available at ScienceDirect

Enzyme and Microbial Technology


journal homepage: www.elsevier.com/locate/enzmictec

A review on preparation and chemical analysis of postbiotics from lactic


acid bacteria
Mehran Moradi a, *, Rahim Molaei a, Jonas T. Guimarães b
a
Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
b
Department of Food Technology, Faculty of Veterinary Medicine, Federal Fluminense University (UFF), Niterói, Rio de Janeiro, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: Postbiotics may be defined as soluble metabolites released by food-grade microorganisms during the growth and
Cell free supernatant fermentation in complex microbiological culture, food or gut. It is rich in high and low molecular weight bio­
Lactobacillus logically active metabolites. There are still gaps concerning these substances, mainly how to use them for food
Probiotics
applications. Although the most recent work on preparation and application of postbiotics from several pro­
Functional food
Bioactive compounds
biotics are very encouraging, the suitability of postbiotics to combat microorganisms that deal with food safety
should be tested mainly by analyzing the chemical composition and conducting antagonistic tests. Consequently,
foods can effectively benefit from an identified postbiotic with a defined effect. This review approached the
recent advances in relation to the preparation of postbiotics from lactic acid bacteria. The function of different
instrumental analysis techniques and factors affecting the chemical composition of postbiotics were also
comprehensively reviewed.

1. Introduction gram-positive and catalase-negative microorganisms. The traditional


LAB are composed of different genera, predominantly Streptococcus,
Currently, people have been associating healthy foods with a lower Lactococcus, Lactobacillus, and Leuconostoc [5]. Lactobacillus spp. and
quantity of additives and chemical substances, which are called “clean Bifidobacterium spp. are the most prominent probiotic bacteria (Fig. 2).
label” products [1], and this awareness highlights the importance of These generally recognized as safe (GRAS) agents are the dominant and
studying natural substances to ensure the food safety and fulfill the beneficial microorganisms of the colon and they are suitable for
consumers’ demand for healthier and natural foods. Antimicrobial amplification in the intestine, without provoking any significant side
agents (AAs) are chemical or natural compounds that can increase safety effects. Other LAB and yeasts are also used as potential probiotic mi­
and extend the shelf life of food commodities by inhibiting the growth or croorganisms (Fig. 2) [6], which can be found in functional products like
eliminating foodborne pathogens and spoilage microorganisms [2]. those manufactured with kefir cultures [7]. Actually, various strains of
These agents comprise substances prepared mainly from four different Lactobacillus are used as primary and secondary (e.g., probiotic and
sources (Fig. 1). AAs of microbial origins in particular have gained protective strains) starters in the production of fermented foods. Lb. casei
credibility over the years since it is a natural and renewable source. and Lb. acidophilus are probiotic strains used in some renowned pro­
Different microorganisms have been used as a source for the production biotics foods (e.g., BIO®, Actimel®, LC1®, and Yakult®). To stimulate
of AAs, among which probiotics are particularly noteworthy due to their beneficial bacteria to grow in the gut and produce their bioactive sub­
capacity to modulate the microbiota of humans and foods, besides its stances, soluble edible oligosaccharide carbohydrates [e.g.,
recognized health benefits and lower risk of infections [3]. Fructo-oligosaccharides (FOS), galacto-oligosaccharides (GOS)] and
The probiotic microorganisms comprise different species, including other compounds, which are commonly known as prebiotics, may also
some lactic acid bacteria (LAB), Bifidobacterium spp., yeasts, and some be added to the food [8].
specific gram-negative strains [4]. LAB is a very heterogeneous group of LAB are capable of producing several substances, among which many

Abbreviations: AA, antimicrobial agents; LAB, lactic acid bacteria; GRAS, generally recognized as safe; Lac., Lactococcus; Lb., Lactobacillus; Leuc., Leuconostoc; L.,
Listeria; Ped., Pediococcus; Ps., Pseudomonas; Salm., Salmonella; Staph., Staphylococcus; Strep., Streptococcus; B., Bacillus; Cl., Clostridium; Ent., Enterococcus; E.,
Escherichia; Kl., Klebsiella; CFS, cell-free supernatant; BLIS, bacteriocin-like inhibitory substances; EPS, exopolysaccharides; MRS, De Man Rogosa and Sharpe.
* Corresponding author.
E-mail address: m.moradi@urmia.ac.ir (M. Moradi).

https://doi.org/10.1016/j.enzmictec.2020.109722
Received 17 August 2020; Received in revised form 25 November 2020; Accepted 28 November 2020
Available online 3 December 2020
0141-0229/© 2020 Elsevier Inc. All rights reserved.
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

The main challenge of using live starter/probiotic cultures directly


on the food is their incompatibility with different matrices and envi­
ronments that hinder their growth and survival in food. As an alterna­
tive, the use of postbiotics avoids the negative interactions between
primary and secondary starters and the food [15]. Therefore, using
postbiotics instead of the living microorganism is possible to take full
advantage of their broad-spectrum antimicrobial activity, the synergis­
tic activities between organic acids and other metabolites, and the high
heat stability of the postbiotics mixture [16].
For industrial applications, postbiotics are recognized as superior to
probiotics due to the defined chemical composition, safety, ease of use
and storage, stability in a broad range of pH and temperature and broad-
spectrum antimicrobial activity [17]. Currently, there are different
commercially available postbiotics with various applications other than
food (Table 2), however, little is known about their methods of prepa­
ration and analysis, and the factors affecting the production of each
postbiotic compound, which may be a barrier to the new researches and
the large-scale application in the food industry. Recently, the applica­
tion of postbiotics mixtures and individual postbiotics in food safety
[16], biotherapy and functional foods [18,19], nutrition [20,21], dis­
Fig. 1. Classification of natural antimicrobial compounds originated from an­
eases prevention [22,23], animal feed and pharmaceutical industries
imal, microbial and plant sources as well as algae and mushroom.
[24], and allergy treatment [25] have been reviewed; however, there is
still a gap in the literature on the steps prior to the application of post­
can inhibit the growth of other microorganisms (Table 1). The AAs of biotics in food.
LAB have significant importance from the point of view of food safety
and quality so that some of them are currently being used by the industry 2. Definition of postbiotics
such as the nisin, which is considered a bacteriocin [9]. Antimicrobial
substances from LAB can be broadly categorized by their molecular Different nomenclatures have been used to describe the fermentation
weight into those that have low molecular weight (H2O2, organic acids, functional compounds produced by food-grade microorganisms [26,27],
acetoin, acetaldehyde, etc.) and those that have a high molecular weight however, postbiotics (metabiotics or biogenics) is the most used today.
(e.g., bacteriocins) [10]. Bacteriocins, organic acids, enzymes, alcohols There is still no consensus on the definition of postbiotics, but most
and low molecular mass substances (e.g., reuterin derived from Lb. studies consider that they must provide any physiological benefits to the
reuteri) are the main metabolites responsible for the antimicrobial action host, which are sometimes similar to the effects provided by the living
of LAB. Bacteriocins are the most studied postbiotic metabolites [11,12]. probiotics [28,21,22]. However, due to the unclear definition and the
The extracellular polymers of microorganisms, known also as exopoly­ wide bioactivity of metabolites produced by probiotic and non-probiotic
saccharides (EPS), have also gained more attention for food applica­ bacteria, the term postbiotic can be defined as any soluble factor (pro­
tions. Although EPS of LAB displays many techno functional ducts/metabolic by-products of microbial metabolisms or substances
applications [13], some biological properties of this novel additive, as produced by the action of LAB on culture/food ingredients; e.g.,
antimicrobial and antioxidant activities, have also been investigated bioactive peptides) secreted by food-grade microorganisms or released
[14]. after cell lysis, during the growth and fermentation in complex

Fig. 2. Some well-known probiotics microorganisms with perceived functional and nutritional benefits in food and human.

2
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

Table 1
The main inhibitory substances produced by different lactic acid bacteria.
Compounds name LAB strains Biological Analytical method Reference
benefit

Organic Acids
Pyruvic acid Lb, Ped Antimicrobial GC-MS [51]
Lactic acid Lb, Ped Antifungal, G+− LC- SPD/ GC-MS [94,95]
Acetic acid Weissella, Lb Antifungal, G+− GC-MS/ LC- SPD [96]
Tartaric acid Lb Antifungal, G+− [94]
Phenylacetic acid Antifungal, G+− MS LCQ and NMR [97]
Benzeneacetic acid Lb Antifungal LC-MS/1H NMR and 13C [57]
NMR
Succinic acid Lb, Ped, Lact Antifungal, G− HPLC-DAD/ GC-MS [51,83]
Phenyllactic acid Weissella, Lb Antifungal, G+− GC-MS [51,68]
Hydrocinnamic acid Weissella, Lb Antifungal GC-MS/ LC-FTMS/ LC-MS/ [68,76,98]
MS
Caffeic acid Weissella, Lb Antifungal HPLC-UV/DAD [60]
4-Hydroxyphenyllactic acid Weissella, Lb, Antifungal LC-FTMS/ GC-MS [51,98]
Ped
Indole-3-lactic acid Weissella, Lb, Antifungal HPLC-UV/ GC-MS [51,99]
Ped
Citric acid Lb, Ped Antifungal, G+− LC- SPD/ GC-MS [51,94]
Malic acid Lb G+− LC- SPD/GC-MS [58,94]
p-Coumaric acid Lb Antifungal, G+− GC-MS/ LC-FTMS [68,100]
Caftaric acid Lb Antifungal LC-MS/MS [76]
Azelaic acid Weissella, Lb Antifungal, G+− GC-MS/ LC-FTMS [68,98]
Propionic acid Lb, Lac, Strep Antifungal, G+− GC-MS/ LC-MS/MS [76,101]
Phenylpyruvic acid Lb Antifungal LC-MS/MS [76]
Vanillic acid Lb, Lac, Strep Antifungal, G− LC-FTMS [98,100]
Formic acid Lb Antifungal, G+− LC-MS [57]
Fatty Acids
2-Hydroxyisocaproic acid Weissella, Lb Antifungal, G+− LC-FTMS/ GC-MS [51,98,
100]
Oleic acid Weissella, Lb, Antifungal, G +−
GC-MS [51,96]
Ped
Butyric acid Lb, Lac, Strep Antifungal, G+− HPLC-DAD [83]
Linoleic acid Lb, L Antifungal, G+− HPLC-MS [102]
6-Octadecenoic acid methyl ester Lb Antimicrobial GC-MS [50]
3-hydroxydecanoic acid Lb Antifungal GC-MS/ LC-FTMS [68,100]
Myristic acid (tertadecanoic acid) Lb Antifungal, G+ GC-MS [101]
Sebacic acid Lb Antifungal GC-MS [101]
3-hydroxydodecanoic acid Lb, Weisella Antifungal LC-FTMS/HPLC [98,103]
3-hydroxytetradecanoic acid (β-Hydroxymyristic acid) Lb Antifungal HPLC [103]
Capric acid Lb, Weisella Antifungal, G+ LC-FTMS [100]
Bacteriocins
Bacteriocin LF-BZ532 Lb G+− RP-HPLC [104]
Bacteriocin SLG10 Lb G+ RP-HPLC [105]
Bacteriocin BMP32r Lb G+ RP-HPLC [106]
Bacteriocin DY4− 2 Lb G− Gel chromatographic, [107]
HPLC
Salivaricin mmaye1 Lb G+− RP-HPLC [108]
Other metabolites
2,4-Di-tert-butylphenol, Heptacosane Weisella Antifungal, G+− GC-MS [96]
1
Phenylalanine Lb G− H-NMR [109]
2-Methyldecane, Pentadecane,2,4-bis(1,1-dimethylethyl)phenol, Dotriacontane Lb Antifungal, G+− GC-MS [50]
Sinapic acid, 2-Deoxycytidine, Cyclo (L-His-L Pro), Phosphoric acid, Cyclo (L-Tyr-L Pro),3,5- Lb Antifungal LC-MS/MS [76]
Di-O-caffeoylquinic acid
Salicylic acid (2-hydroxybenzoic acid) Lb Antifungal, G− LC-FTMS [98]
Diacetyl(2,3-butadione) Lb, Lac, Ped, Antifungal, G+− Headspace GC-MS [110]
Leuc
Acetaldehyde Lb, Lac, Ped, Antifungal, G+− Headspace GC-MS [110]
Leuc
Acetoin Lb, Lac, Ped, Antifungal, G +−
Headspace GC-MS [110]
Leuc
Hydrogen peroxide, carbon dioxide, ethanol LAB Antifungal, G+− HPLC [111]
pyrrolo [1,2-a] pyrazine-1,4-dione Lb Biofilm removal GC-MS [46]
Secretory proteins Weisella G+− MALDI-TOF MS [61]
(trans, trans)-3,4-dihydroxycyclohexane- 1-carboxylic aci Lb G+− NMR [68]
Gallic acid Lb G+ HPLC [112]

*G+− gram positive and negative. Lac: Lactococcus; Lb: Lactobacillus; Leuc: Leuconostoc; Ped: Pediococcus; Ent: Enterococcus. Strep: Streptococcus. B: Bifidobacterium.
GC–MS: Gas Chromatography–Mass Spectrometry; NMR: Nuclear Magnetic Resonance; MALDI-TOF MS: Matrix Assisted Laser Desorption Ionization-Time of Flight
Mass Spectrometry; HPLC: High Performance Liquid Chromatography; RP-HPLC: Reversed-phase HPLC; LC-FTMS: Liquid Chromatography Fourier Transform Mass
Spectrometry; LC–MS/MS: Liquid Chromatography with tandem mass spectrometry; DAD: Diode-Array Detector; ESI-MS: Electrospray Ionization mass Spectrometry.

3
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

Table 2
Commercially available postbiotics.
Name Corresponding microorganism Active ingredient Application Manufacturer

Del-Immune Lb. rhamnosus V Cell wall and DNA fragments Immunomodulator Pure Research
V® Products LLC
Hylak® Ent. faecalis, Lb. acidophilus, Lb. helveticus and E. coli Bacteria-free liquid containing acetic Antimicrobial activity on pathogenic Ratiopharm/
forte acid, short-chain fatty acids, glutamic bacteria in gut Merckle GmbH
acid, formic acid, riboflavin and
glutamine
CytoFlora® Lb. casei, Lb. plantarum, Lb. acidophilus DDS-1, Lb. reuteri, Probiotics tincture containing cell Balances intestinal dysbiosis, creating BioRay Inc,
Lb. salivarius, Lb. rhamnosus, Lb. sporogenes, Lb. bulgaricus lysate of probiotic bacteria an environment for healthy intestinal
Lb. acidophilus, B. bifidum, B. infantis, B. bulgaricus, and flora to colonize
Strep. thermophiles
Aktoflor C Unknown Lysine, lactic acid, succinic acid Reducing intestinal disorder Solopharm
symptoms company
Zakofalk Unknown Inulin and butyrate Immunomodulating Dr. Falk Germany

Lac: Lactococcus; Lb: Lactobacillus; Leuc: Leuconostoc; Ped: Pediococcus; Ent: Enterococcus; Strep: Streptococcus. B: Bifidobacterium.

microbiological cultures (in this case called cell-free supernatant; CFS), containing 1) products from microbial metabolism, 2) metabolites syn­
food or gut [20,26,29,24,30] (Fig. 3). From this perspective, postbiotics thesized by the action of LAB on culture/food ingredients or 3) struc­
prepared from LAB comprise hundreds of individual metabolites used tural substances produced by LAB and used as postbiotics mixture [18,
either as a crude concentrate (extract) or as a semi-purified form. 19]. The quantity and type of products are mainly related to the type of
Postbiotics of probiotic cultures seem to be a rich source of bacte­ bacterial strain, type of culturing medium, bacterial treatment after
riocin and bacteriocin-like inhibitory substances (BLIS) with antago­ propagation, etc. In all published works on the application of postbiotics
nistic activity on major foodborne pathogens. Moreover, in some in the food, no post-propagation treatments were applied. In this case, as
context, postbiotics were defined as cell metabolites and cell wall- illustrated in Fig. 4, postbiotics contains only soluble factors such as
derived substances (exopolysaccharide; EPS, teichoic acids, peptido­ products or metabolic by-products that have been secreted into the
glycans, polar lipids, glycolipids, peptidoglycan, and proteins) [31] of medium during the bacterial growth [30]. However, in some works,
microorganisms of which the cell walls were ruptured following lysis by after propagation, bacterial cells were subjected to lysis by enzymes,
heat treatment, enzymes application or ultrasonic processing [26,32]. thermal, sonication, and high-pressure treatments or a combination of
Cell-derived ingredients generally display immunomodulation and them (Fig. 4) [19,34,35]. These treatments introduce some additional
antiproliferative activity in the host [19]. The term paraprobiotics intracellular metabolites and cell wall- derived materials into the post­
(ghost probiotics) is used to refer to the non-viable or inactivated pro­ biotics mixture and give new functionality to the obtained postbiotics. In
biotic strains, either in intact or ruptured forms [20,33]. Unfortunately, both treated and non-treated postbiotics mixtures, the obtained solution
there is no agreement on the definition of postbiotics, but it generally is centrifuged or ultrafiltrated in order to separate bacterial cells from
defines as biological materials that contain probiotic-derived metabo­ postbiotics metabolites [24].
lites or cell wall-derived materials (e.g., peptides, teichoic acids, surface Regardless of the type of preparation (bacterial lysis or not), at the
protruding molecules, exopolysaccharides, biosurfactants, and plas­ laboratory and industrial level, there are several important points to be
malogens) without the presence of viable microorganisms or cell considered for postbiotics production such as the fermentation medium,
structures [16,18,19]. bacterial propagation, harvesting and concentration methods, preser­
vation, and method of application. Generally, LAB, whether as the pri­
3. Preparation of postbiotics from LAB mary or secondary starter, are used for postbiotics preparation;
however, the tendency to non-starter LAB has greatly increased and
To obtain a postbiotics mixture, in most cases, the researchers pre­ most attention needs to be paid to the safety of final products. Post­
pared a solution called cell-free supernatant, with or without cell lysis, biotics is most commonly prepared by culturing the LAB in culture

Fig. 3. Schematic illustration for postbiotics concept from lactic acid bacteria in microbiological culture media, food and gastrointestinal tract.

4
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

Fig. 4. The five steps of preparing postbiotics (cell free supernatant) mixture form lactic acid bacteria (LAB). 1: resuscitation of LAB, 2: propagation of LAB, 3:
treatment, 4: postbiotics harvesting and 5: postbiotics concentration. MRS stands for De Man, Rogosa and Sharpe.

media [mainly De Man, Rogosa and Sharpe (MRS) broth], followed by sterilized (0.22 μm) postbiotics solution should be divided into three
an extraction step (centrifugation at 4000− 12000 g for 10 min at 4 ◦ C or equal fractions. The antimicrobial function of organic acids can be
dialysis) (Fig. 4) (Table 3) [17,36,37]. Postbiotics, if prepared in MRS, monitored by neutralizing the pH of the postbiotic solution (pH 6.5–7)
produce a bacteria-free and acid fermentate with brown to brownish with NaOH 1 N [43], while the hydrogen peroxide-dependent activity
yellow color [38] (Fig. 5). Recently, the suitability of other alternatives can be checked by the addition of catalase (5 mg/mL or 300 IU/mL) into
for the production of postbiotics at the industrial scale was also reported the second solution (or previously neutralized solution) for 1 h at 37 ◦ C
(Table 3). In one study, different dairy-based ingredients including [43,44,45]. For bacteriocin and BLIS assay, the third solution (or pre­
low-heat milk and milk permeate have optimized based on incubation viously NaOH-catalase- treated solution) should be treated with
time and temperature, as a fermentation medium for Lactobacillus spp. in 1 mg/mL of a proteolytic enzyme (trypsin, α-chymotrypsin, pepsin,
order to prepare antifungal postbiotics solutions (or fermentate) for food papain or proteinase K) at 37 ◦ C for 2 h followed by incubation at 100 ◦ C
application [39]. Due to differences in protein and fat content, post­ for 5 min [46].
biotics mixtures prepared in low-heat milk revealed remarkable anti­
fungal activity. Postbiotics prepared in both substrates had suitable 4. Chemical analysis of postbiotics
compatibility with dairy food applications. Skim milk (10 %) has suc­
cessfully applied for the production of postbiotics of an adjuncts culture From a fundamental point of view, the suitability of postbiotics to
(i.e., DSM-100 H) at an industrial scale [37]. The suitability of some fight harmful microorganisms that deal with food safety should be tested
alternative dairy substrates such as whey and buttermilk as a propaga­ primarily by analyzing their chemical composition and conducting in
tion medium for different commercial probiotics were also investigated vitro antagonistic tests. Metabolomics involve the identification of me­
[40]. tabolites in biological systems, then, it could help to investigate post­
However, postbiotics can be prepared in both liquid and dry (spray- biotic metabolites originated from LAB [47]. The quantity and quality of
dried and lyophilized) forms [4]. Lyophilization process is the dominant postbiotics and molecules harboring antagonistic activities should be
preservation method, although spray drying is also used [37]. However, tested using analytical procedures [29]. In this regard, foods can effec­
the method of application of postbiotics depends on the type of food. tively benefit from an identified postbiotic with defined effects. There is
Generally, pasty lyophilized postbiotics exhibit enhanced antimicrobial much literature available addressing the existence of a wide range of
activity and minimize the increase of moisture in food compared to the metabolites in postbiotics [20]. Accordingly, given the complexity of
postbiotics solution [41]. In addition, spraying of postbiotics on the biological compounds with different polymerization degrees and
surface of food may serve as an efficient alternative in order to inhibit glycosidic bonds, the qualitative and quantitative identification and
the growth of spoilage and pathogen microorganisms [39]. For sterili­ characterization of the postbiotic matrix generally requires sophisti­
zation of postbiotics, the solution is typically filtered with a 0.22 or cated equipment with several concentration/clean up steps.
0.45-μm pore size filter and kept at -20 to − 80 ◦ C until its use. In the case For the qualitative and/or quantitative analysis of postbiotics, a large
of lyophilized and spray dried concentrates, the solution is sterilized number of instrumental techniques and innovative approaches have
before use. A sterile buffer solution (phosphate buffer saline; PBS, been applied (Table 1). The appropriate method is usually selected
0.01 M phosphate, pH 7.2) is used to resuspend pasty postbiotics [42]. based on the analytical goals and the pursued type of characterization
[17]. Herein, we reviewed various analytical techniques used to
examine the chemical composition of postbiotics.
3.1. Modification of postbiotics mixture for antimicrobial application

The antimicrobial performance of each AA in postbiotics can be


elucidated by treating the postbiotics solution. Initially, a filter-

5
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

Table 3
Postbiotic preparation and application from various LAB strains under different conditions and their potential application.
Name of LAB Postbiotic using Medium Culture conditions/postbiotics preparation Application Reference
form

Lb. acidophilus Solution MRS broth At 37 ◦ C for 24 h/ Centrifugation (6000 g for Antibacterial and antibiofilm [113]
15 min) potential
Lb. curvatus BCS35 Solution and MRS broth At 30 ◦ C for 16 h /Centrifugation Food ingredient in fresh fish [36]
lyophilized forms (12,000 rpm, 4 ◦ C, 10 min)
Lactobacillus spp. isolates- Solution in MRS broth At 37 ◦ C overnight /Centrifugation (10,000 g Antibacterial and antibiofilm [114]
Lb. acidophilus neutralized and for 15 min at 4 ◦ C) potential
heat-treated forms
Different LAB Solution MRS broth At 37 ◦ C for 6 h /Centrifugation (9000 rpm, Antibacterial and inhibition of [115]
4 ◦ C, 20 min) biogenic amine production in
vitro
Lb. acidophilus A9, 08 and Solution Buttermilk and whey based At 37 ◦ C for 10 h None [40]
H5, Lb. paracasei A13 media with or without 0.3 %
and LS and Lact. casei yeast extract, with 0.3 % yeast
extract plus 1% glucose
Lb. acidophilus LA5 Lb. casei Solution MRS broth At 37 ◦ C for 48 h in CO2 incubator/ Antimicrobial and biofilm [43]
431 Centrifugation (4000 g for 10 min) removal activity
A LAB with antifungal Lyophilized MRS broth At 37 ◦ C for 24 h /Centrifugation (8500 rpm, Control of aflatoxin producer [50]
activity 4 ◦ C, 20 min) Aspergillus
LAB Lyophilized Low-heat milk and milk Different formulations Antifungal activity on semi- [39]
permeate hard cheese molds
Lb. plantarum Solution in MRS broth At 37 ◦ C for 24 h /Centrifugation (8000 rpm, In vitro reduction of biogenic [116]
neutralized and 4 ◦ C, 10 min) amines
heat-treated forms
Lb. acidophilus LA-5® Solution MRS broth At 37 ◦ C for 48 h, anaerobic /Centrifugation Biosynthesis of carbon dots [117]
(4000 rpm, 4 ◦ C, 10 min)
Lb. rhamnosus Lyophilized MRS broth At 37 ◦ C for 18 h, anaerobic /Centrifugation Development antimicrobial [118]
(8000 rpm, 4 ◦ C, 10 min) packaging
Lb. brevis WK12 and Leuc. Solution MRS broth At 30 ◦ C, and aerobic anaerobic Washing sanitizer in leaf [119]
mesenteroides WK32 /Centrifugation (5000 rpm, 4 ◦ C, 15 min) vegetable
Selected Lactobacillus spp. Lyophilized 10 % Skim milk Laboratory scale production in bioreactor at Functional food ingredients [37]
43 ◦ C with an agitation rate of 200 rev/min
and CO2 sparring (02 L/min) for 48 h/
Centrifugation (4000 rpm, 8 ◦ C, 30 min)
Lb. plantarum Lyophilized MRS broth at 37 ◦ C for 24 h in CO2 incubator/ Development antimicrobial [72]
Centrifugation (4000 g for 10 min) packaging for ground meat
Lb. plantarum Cys5− 4 Solution MRS broth At 32 ◦ C for 30 h /Centrifugation (13,000 g Antibacterial activity in [120]
4 ◦ C for 20 min) artisanal beverages
Lb. actobacillus reuteri, Lb. Solution in MRS broth At 37 ◦ C until reaching stationary phase/ Down-regulate the gene [121]
plantarum, Lb. fermentum neutralized form Centrifugation (10,000 rpm, 4 ◦ C, 10 min) expression of some virulent
factors in multidrug-resistant
foodborne pathogens
Lb. acidophilus (EMCC Lyophilized MRS broth At 37 ◦ C for 48 h /Centrifugation Control of food borne pathogen [122]
1324), B. bifidum (EMCC (10,000 rpm, 10 min) on soft feta cheese
1334) and Lb. plantarum
(EMCC 1845)
Lb. salivarius Lb. acidophilus Lyophilized MRS broth At 37 for 48 h in CO2 incubator / Improving shelf life of ground [38]
Centrifugation(at 4000 g for 10 min) meat

Lb: Lactobacillus; MRS: De Man, Rogosa and Sharpe.

Fig. 5. The appearance of postbiotics mixture in solution (A) and lyophilized (B) forms prepared from Lb. salivarius in De Man, Rogosa and Sharpe (MRS) Broth.

6
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

4.1. Gas chromatography (GC) quantified [59]. In another study, the analysis of the metabolites in LAB
cultures using a liquid chromatography linear ion trap quadrupole
GC is the most frequently used analytical tool for quantitative and Orbitrap hybrid Fourier transform mass spectrometer (LC-FTMS) was
qualitative analysis of free fatty acids, volatile compounds (e.g., diace­ also reported [60]. It is important to note that the method of analysis
tyl, acetoin and dimethyl sulfone, 2-butanone), and organic acids in affects the type and quantity of a unique metabolite in the postbiotics.
postbiotics. Moradi et al. (2019) have utilized the GC–MS approach to For example, GC–MS analysis of postbiotics prepared from W. cibaria
identify the chemical composition of postbiotics derived from Lb. aci­ showed the presence of oleic and palmitic acids while in HPLC analysis,
dophilus LA5, Lb. casei 431 and Lb. salivarius. However, depending on the only lactic and citric acids were determined at the same postbiotics [61].
sample type and detection technique, additional steps are usually
required before GC–MS analysis. For example, the volatile profile of 4.3. Thin layer chromatography (TLC)
postbiotics from Lb. casei was fully characterized through the headspace-
solid phase microextraction (HS-SPME) GC–MS technique and the au­ Despite the development of some advanced chromatography tech­
thors confirmed the presence of 62 compounds, including alcohols, niques, TLC is still used as a routine qualitative test in some laboratories
terpenes, and norisoprenoids, acids, ketones and esters in the postbiotics because it has advantages such as quickness, less tedious, and less costly
[48]. In another study, a GC equipped with a flame ionization detector over other chromatographic techniques. TLC is the most extensively
(GC-FID) was used to study the short-chain fatty acid (SCFA) content of studied method for the isolation and structural determination of bio­
postbiotics of four bacterial strains, Lb. acidophilus, Lb. helveticus, E. coli, surfactants produced by bacterial strains, which are often associated
and Ent. Faecalis [49]. In the standard process of the analysis of SCFA with a spectroscopic or post chromatographic identification method
with GC-FID, the derivation of fatty acids by boron trifluoride-ethanol [62]. A colorimetric approach using Syldatk, ammonium molybdate or
complex is necessary [49]. Ninhydrin reagents (sprayed on the surface of TLC plates) has been
For the quantification of organic acid by GC–MS, the postbiotics applied to identify moieties, lipid moieties, and amino acids, respec­
sample should primarily be esterified with absolute ethanol and sulfuric tively [62]. In addition, the composition of biosurfactant derived from
acid 97 % v/v [50]. In another work, the organic acid profile was Lactobacillus spp. was characterized by TLC followed by post chro­
determined in the postbiotics from Lb. pentosus K34 and Ped. lolli PL24 matographic detection through chromogenic compounds. The results
by GC–MS following methoxime/ tert-butyldimethylsilyl derivatives obtained from TLC confirmed the presence of glycolipids with poly­
[51]. Fatty acids and volatile compounds produced by Lb. rhamnosus saccharides and lipid fractions in postbiotics secreted by Lb. helveticus
with a potential antifungal activity were evaluated using GC-FID and [63]. Moreover, two fractions with antibacterial properties were iso­
GC–MS methods, respectively [52]. A review paper was published lated from the postbiotics of Lb. plantarum IMAU10014 using TLC,
describing the capability of GC–MS as an interesting method to inves­ consisting of 3-phenylacetic acid, benzeneacetic acid, and 2-propenyl
tigate the monomeric composition of several EPS produced by different ester [57].
LAB such as Lac. lactis spp. cremoris, Strep. thermophilus, Lb. delbrueckii
spp. bulgaricus, Lb. helveticus, Lb. sake, Lb. reuteri, Lb. rhamnosus, and 4.4. Spectrophotometric-based analysis
B. longum [53].
The spectrophotometric technique can be used to determine the
4.2. Liquid chromatography (LC) hydrogen peroxide concentration in postbiotics of selected LAB. The
quantity of total protein in postbiotics of LAB can also be determined by
The high-performance liquid chromatography (HPLC) method is also the colorimetric assay by Bradford method, in which the binding of the
one of the most utilized techniques for qualitative and quantitative dye (Coomassie Brilliant Blue G-250) to protein cause the sample color
investigation of the chemical composition of postbiotics from LAB. The to shift from red to blue (the absorption maximum shift from 465 to
widespread application of this assay is related to the high potency, pu­ 595 nm), being the absorbance changes at 595 nm proportional to the
rity, and performance features of HPLC. The technique is highly rec­ amount of protein [64]. Furthermore, according to a standard protocol,
ommended when we need a concomitant analysis of different organic the proteinase activity of the postbiotics is evaluated using spectro­
acids (acetic, malic, lactic, acetic, and other acids) in the postbiotics. The photometric techniques. In this approach, azocasein is used as a chro­
coupling of HPLC with a proper detector like ultraviolet (UV), ultra mogenic substrate, and the activity is expressed in terms of absorbance
violet-diode array detector (UV/DAD) refractive index (RI), mass spec­ increase at 440 nm over time [65]. Colorimetric methods have also been
troscopy (MS), and pulsed electrochemical detection (PED) is the key to applied to quantify EPS at absorption around 480− 900 nm [66] and
success in HPLC performance [54]. Patil et al. (2019) utilized HPLC-RI alkaline phosphatase in the postbiotics of LAB [67].
to determine the presence of various mono- and disaccharides, EPS,
and vitamins in the postbiotics. As well, HPLC-UV/DAD technique was 4.5. Nuclear magnetic resonance (NMR) spectroscopy
successfully employed to examine the antifungal compounds present in
the growth media of Lb. amylovorus DSM19280 [55]. The NMR is another powerful tool that provides crucial information
One of the promising approaches is the use of ultra-performance about the structure, dynamics, and interactions of biological metabolites
liquid chromatography (UPLC), which represents superior separation of postbiotics extract. The most common types of NMR are the proton
and identification capacity for postbiotics owing to the high efficiency, and carbon-13 NMR spectroscopy which can be applied to any type of
resolution, sensitivity, and accuracy along with low use of solvents [29]. sample containing nuclei possessing spin. In particular, NMR measure­
UPLC has been applied in the characterization of intracellular compo­ ment helps to acquire information about postbiotics metabolite from
nents such as enzymes and proteins of Lb. plantarum [56]. In another LAB. Broberg and co-workers showed that postbiotics of Lb. plantarum
study, the qualitative analysis of the antifungal compounds (3-phenyl­ comprises (trans, trans)-3,4-dihydroxycyclohexane- 1-carboxylic acid
lactic acid, benzeneacetic acid, and 2-propenyl ester) present in the Lb. with antibacterial properties, and they highlighted that they failed to
plantarum IMAU10014 postbiotics was performed through UPLC system identify these compounds using another available technique (e.g.,
equipped quadrupole ion trap mass spectrometer [57]. Furthermore, chromatography) [68]. NMR (1H NMR and 13C NMR) has also been
Lee’s group employed UPLC-MS (UPLC-Q-TOF) to identify the cell employed to reveal the presence of phenolic antifungal metabolites
metabolite profiles of Lb. sakei and its culture media at different growth produced by Lb. brevis P68 [69]. Lin and Pan, (2017) utilized 1H NMR
phases [58]. An antifungal organic acid in the binary combinations of and 13C NMR spectroscopy to confirm the presence of 2-(2− 1-ami­
Lactobacillus spp. (Lb. plantarum L244, Lb. harbinensis L172, Lb. rham­ no-1-hydroxyethoxy) ethyl 2-methylpropanoate as an antimicrobial
nosus CIRMBIA1113, and Lb. brevis L128) using LC–Q–TOF were substance in the postbiotics of Lb. plantarum NTU 102 [70].

7
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

In another work, the production of benzeneacetic acid and 2-pro­ 5.2. Culture type and conditions
penyl ester by Lb. plantarum IMAU10014 as an antifungal substance in
the MRS medium were analyzed by 1H NMR and 13C NMR [57]. In The culture composition and incubation conditions can affect me­
addition, the results of NMR quantification of the biosurfactant derived tabolites production by LAB [4]. For example, Lactobacillus spp. do not
from Lb. helveticus, confirmed the presence of several compounds such as produce hydrogen peroxide under anaerobic conditions [75]. A time and
octadecanoic acid as the main lipid consisting of long aliphatic chain cultivation- dependent formation of organic acids by LAB was also re­
and polysaccharides [63]. ported [83]. The production of organic acids (lactic acid, acetic acid,
succinic acid, and formic acid) by different LAB strains in anchovy
infusion broth is significantly higher than in MRS broth [83]. It was also
4.6. Fourier transform infrared (FTIR) spectroscopy found that the postbiotics from different LAB strains prepared in the
low-heat milk revealed significantly higher antifungal activity than
The FTIR represents a potent characterization technique that has those prepared in milk permeate [39]. In a study, 20 g/L of glucose and
been recognized in exploration, classification, and identification of 36.20 g/L of yeast extract were used to supplement a modified MRS
organic and inorganic components. The FTIR technique is a sensitive, medium in order to achieve maximum bacteriocin-inhibitory activity of
rapid, easy to use, well-established, reagent-less, non-destructive, and postbiotics from Lb. plantarum I-UL4 [84]. The addition of non-ionic
cost-effective approach. Recently, FTIR analysis is recommended to get surfactants and emulsifiers, as for instance polysorbate 80, in culture
qualitative information about metabolites of postbiotics [71]. In this media can improve the production and release of proteinaceous post­
sense, Shafipour Yordshahi et al. (2020) investigated lyophilized post­ biotics from Lb. plantarum I-UL4 [85]. Furthermore, supplementation of
biotics from Lb. plantarum using the FTIR technique. Sharma et al. Lb. plantarum medium with linoleic acid resulted in the formation of a
(2014) provided a summarized table of the studies that used the FTIR therapeutic substance called 10-hydroxy-cis-12-octadecenoic acid
technique to characterize the biosurfactants [71]. Also, Trabelsi et al. (HYA) [86]. Due to the synergistic activity of postbiotics and prebiotic,
(2015) monitored the production of EPS during LAB growth using the postbiotics of Lb. plantarum prepared in a reconstituted media contain­
FTIR technique [73]. ing inulin showed higher antimicrobial activity on a spoilage microor­
ganism (Ped. acidilactici) and various pathogens than the postbiotics
without inulin supplementation [87]. Similarly, in culture media sup­
4.7. Other techniques plemented with glycerol, Lb. Reuteri produced reuterin, a potent anti­
microbial agent [88].
As stated, postbiotics is a matrix of complex metabolites with diverse
physicochemical and functional properties, so the extraction and/or 5.3. Co-culturing strategies for postbiotics production
isolation step is inevitable since it allows an accurate quantitative and
qualitative assessment of the metabolite. The dialysis is one of the The evidences tend to support the hypothesis that a mixture of multi-
earliest purification procedure. It seems that the dialysis can help to strains is more effective than a mono-strain. The LAB can regulate the
distinct low molecular mass antimicrobial metabolites (<1 KDa) from production of specific AAs in postbiotics in the presence of beneficial or
high molecular mass compounds (>1 KDa) of Lactobacillus spp. [10]. even pathogenic microorganisms, according to a quorum-sensing
However, the isolation step can be performed by liquid-liquid extraction mechanism [89]. Co-culturing can facilitate the expression of ribo­
(LLE) [58], preparative HPLC [74], TLC [62], solid-phase extraction somal synthesized AAs like bacteriocins in LAB [90], also leading to the
cartridge (SPE) [75], QuEChERS methodology [76], and electrophoresis discovery of novel AAs. Co-culturing of Lac. lactis subsp. lactis with
[77]. Saccharomyces cerevisiae CL01 increased the production of nisin by 85 %
Recently, the efficacy of matrix-assisted laser desorption/ionization [91]. Enhanced production of nisin (by 50 %) by Lac. lactis subsp. lactis
time-of-flight (MALDI-TOF) to identify proteinaceous components of in the presence of Yarrowia lipolytica was also reported in the sugar beet
postbiotics from Lactobacillus spp. has received remarkable interest [77, molasses [92]. Moreover, Lac. lactis subsp. lactis produce active
78]. The application of electrospray ionization mass spectrometry nisin-rich postbiotics in co-culturing with harmful pathogens like as
(ESI-MS) to identify the molecular masses of metabolites produced by L. monocytogenes and Salm. enterica [89].
Lactobacillus spp. was reported [75]. Moreover, two-dimensional gel
electrophoresis (2D-PAGE) has also been proposed for the molecular 5.4. Postbiotics preparation, processing and analysis
weight determination [77] in LAB. Finally, some authors used specific
enzymatic kits to quantify acetic and D-/L-lactic acids [78]. For the application of postbiotics in foods, it will be interesting the
use of different forms of presentation, like lyophilized or dried, however,
5. Factors affecting the chemical composition of postbiotics the processing of extracted postbiotics can influence their composition,
which may decrease the postbiotics activity. For example, lyophilization
5.1. The type of bacterial species promotes the elimination of hydrogen peroxide, which is a factor
responsible for antimicrobial action [93]. The spray drying method also
The strain-dependent fermentation of LAB is widely reported. removes volatile metabolites, as for instance ethanol [75]. There are
Organic acid production by LAB is mainly a genus-specific phenomenon evidences suggesting the antagonistic stability of postbiotic metabolites
and partially specie-specific [79]. For example, the amount of lactic acid over time and storage at different conditions [43]. While, with the in­
produced by Lactobacillus spp. and Bifidobacterium spp. is significantly crease of pH, the antimicrobial performance of postbiotics can nega­
higher than acetic acid production [80]. Strong positive and negative tively be affected [46].
correlations were found between lactic and acetic acid contents of
postbiotics and antimicrobial characteristics [80]. Bacterial strains can 6. Conclusions
also be engineered to secrete unique recombinant metabolites for food,
human, and animal uses [81]. The highest concentration and produc­ This review focused mainly on defining postbiotics and presenting
tivity of lactic acid were reported in batch and continuous fermentation the methods currently used for preparation and chemical analysis of
of LAB, respectively [82]. Özcelik et al. (2016) investigated organic acid postbiotics obtained from LAB. The concept of postbiotics is relatively
production ability of LAB in fish infusion broth and MRS broth and new in the food industry and some current applications such as biofilm
found theproduction of succinic and acetic acids at the highest and removal, biodegradation of food chemical contaminants, and prepara­
lowest levels, respectively. tion of nanomaterials using postbiotics have been proposed (Fig. 6). For

8
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

Fig. 6. Different potential food safety application of postbiotics mixture obtained from lactic acid bacteria.

these purposes, it is required to establish standard methods of chemical Declaration of Competing Interest
analysis. Although several chromatographic (e.g., GC, HPLC, etc.) and
spectrophotometric instrumental methods have been proposed for The authors reported no declarations of interest.
postbiotics, identification of some unknown chemical constituents re­
quires high-tech instruments, additionally, for a proper and complete Acknowledgments
evaluation, the use of more than one instrument/procedure of analysis
has been preferred. The authors wish to acknowledge Urmia University for support.
Moreover, the quantity and quality of metabolites in postbiotics can
be changed by modifying the composition of the culture media. The References
brown to brownish-yellow color of the postbiotics prepared with MRS
broth may restrict the postbiotics mixture utilization in some foods, [1] J.T. Guimarães, C.F. Balthazar, R. Silva, R.S. Rocha, J.S. Graça, E.A. Esmerino, M.
including many dairy products, especially when using high concentra­ C. Silva, A.S. Sant’Ana, M.C.K.H. Duarte, M.Q. Freitas, A.G. Cruz, Impact of
probiotics and prebiotics on food texture, Curr. Opin. Food Sci. 33 (2020) 38–44,
tions. In this case, the individual postbiotic (e.g., bacteriocins, organic https://doi.org/10.1016/j.cofs.2019.12.002.
acids, etc.) is generally preferable to postbiotics mixture. Alternatively, [2] P.M. O’Connor, T.M. Kuniyoshi, R.P.S. Oliveira, C. Hill, R.P. Ross, P.D. Cotter,
in situ-production of postbiotics by LAB with co-culturing with other Antimicrobials for food and feed; a bacteriocin perspective, Curr. Opin.
Biotechnol. 61 (2020) 160–167, https://doi.org/10.1016/j.copbio.2019.12.023.
beneficial microorganisms would be an interesting choice.
[3] B. Chugh, A. Kamal-Eldin, Bioactive compounds produced by probiotics in food
Despite the facts that favor the use of isolated postbiotic metabolites, products, Curr. Opin. Food Sci. 32 (2020) 76–82, https://doi.org/10.1016/j.
some of them present disadvantages for food application. For example, it cofs.2020.02.003.
[4] H.L. Foo, T.C. Loh, N.E. Abdul Mutalib, R.A. Rahim, The myth and therapeutic
is widely accepted that LAB are associated with the production of
potentials of postbiotics, in: J. Faintuch, S.B.T.-M, M. in D. Faintuch Therapy, and
biogenic amines, therefore, it is highly recommended to monitor the other Strategic Applications (Eds.), Microbiome Metabolome Diagnosis, Ther.
exact chemical content of postbiotics before using it in food products. To Other Strateg. Appl., Academic Press, 2019, pp. 201–211, https://doi.org/
encourage the industrial application and to overcome the main obstacles 10.1016/B978-0-12-815249-2.00021-X.
[5] J. Zheng, S. Wittouck, E. Salvetti, C.M.A.P. Franz, H.M.B. Harris, P. Mattarelli, P.
of postbiotic preparation and application found in laboratory condi­ W. O’Toole, B. Pot, P. Vandamme, J. Walter, K. Watanabe, S. Wuyts, G.E. Felis,
tions, such as the high cost and the undesired color appearance, it is M.G. Gänzle, S. Lebeer, A taxonomic note on the genus Lactobacillus: description
recommended to replace the conventional MRS broth with cost-effective of 23 novel genera, emended description of the genus Lactobacillus Beijerinck
1901, and union of Lactobacillaceae and Leuconostocaceae, Int. J. Syst. Evol.
plant- and animal-based by-products (e.g., whey medium), which is an Microbiol. 70 (2020) 2782–2858, https://doi.org/10.1099/ijsem.0.004107.
area in need of further studies [6] S. Fijan, Microorganisms with claimed probiotic properties: an overview of recent
literature, Int. J. Environ. Res. Public Health 11 (2014) 4745–4767, https://doi.
org/10.3390/ijerph110504745.
Author contributions [7] F.S. Erdogan, S. Ozarslan, Z.B. Guzel-Seydim, T. Kök Taş, The effect of kefir
produced from natural kefir grains on the intestinal microbial populations and
Mehran Moradi conceived and designed the review. All authors antioxidant capacities of Balb/c mice, Food Res. Int. 115 (2019) 408–413,
https://doi.org/10.1016/j.foodres.2018.10.080.
contributed to the writing of the manuscript.
[8] P. Markowiak, K. Śliżewska, Effects of probiotics, prebiotics, and synbiotics on
human health, Nutrients. 9 (2017) 1021, https://doi.org/10.3390/nu9091021.
CRediT authorship contribution statement [9] L.J. de Arauz, A.F. Jozala, P.G. Mazzola, T.C. Vessoni Penna, Nisin
biotechnological production and application: a review, Trends Food Sci. Technol.
20 (2009) 146–154, https://doi.org/10.1016/j.tifs.2009.01.056.
Mehran Moradi: Conceptualization, Writing - review & editing. [10] J. Šušković, B. Kos, J. Beganović, A. Pavunc, K. Habjanič, S. Matošić,
Rahim Molaei: Writing - review & editing. Jonas T. Guimarães: Antimicrobial activity – the most important property of probiotic and starter
Writing - review & editing. lactic acid bacteria, Food Technol. Biotechnol. 48 (2010) 296–307.
[11] C.A. O’Bryan, O.K. Koo, M.L. Sostrin, S.C. Ricke, P.G. Crandall, M.G. Johnson,
Characteristics of bacteriocins and use as food antimicrobials in the United States,
in: S.C. Ricke, G.G. Atungulu, C.E. Rainwater, S.H.B.T.-F, F.S.S, A. Park (Eds.),

9
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

Food Feed Saf. Syst. Anal., Academic Press, 2018, pp. 273–286, https://doi.org/ intracellular content from Lactobacillus caseiCRL-431 on the antioxidant
10.1016/B978-0-12-811835-1.00015-4. properties of breast milk: Randomized double-blind controlled trial, LWT. 130
[12] S.M. Razavi Rohani, M. Moradi, T. Mehdizadeh, S.S. Saei-Dehkordi, M. (2020) 109672, https://doi.org/10.1016/j.lwt.2020.109672.
W. Griffiths, The effect of nisin and garlic (Allium sativum L.) essential oil [36] B. Gómez-Sala, C. Herranz, B. Díaz-Freitas, P.E. Hernández, A. Sala, L.M. Cintas,
separately and in combination on the growth of Listeria monocytogenes, LWT - Strategies to increase the hygienic and economic value of fresh fish:
Food Sci. Technol. 44 (2011), https://doi.org/10.1016/j.lwt.2011.07.020. biopreservation using lactic acid bacteria of marine origin, Int. J. Food Microbiol.
[13] M.-C. Gentès, S.L. Turgeon, D. St-Gelais, Impact of starch and exopolysaccharide- 223 (2016) 41–49, https://doi.org/10.1016/j.ijfoodmicro.2016.02.005.
producing lactic acid bacteria on the properties of set and stirred yoghurts, Int. [37] E. Dunand, P. Burns, A. Binetti, C. Bergamini, G.H. Peralta, L. Forzani,
Dairy J. 55 (2016) 79–86, https://doi.org/10.1016/j.idairyj.2015.12.006. J. Reinheimer, G. Vinderola, Postbiotics produced at laboratory and industrial
[14] F. Nehal, M. Sahnoun, S. Smaoui, B. Jaouadi, S. Bejar, S. Mohammed, level as potential functional food ingredients with the capacity to protect mice
Characterization, high production and antimicrobial activity of against Salmonella infection, J. Appl. Microbiol. 127 (2019) 219–229, https://doi.
exopolysaccharides from Lactococcus lactisF-mou, Microb. Pathog. 132 (2019) org/10.1111/jam.14276.
10–19, https://doi.org/10.1016/j.micpath.2019.04.018. [38] M. Moradi, H. Tajik, K. Mardani, P. Ezati, Efficacy of lyophilized cell-free
[15] S.A. Pujato, A. del L Quiberoni, M.C. Candioti, J.A. Reinheimer, D. supernatant of Lactobacillus salivarius (Ls-BU2) on Escherichia coli and shelf life of
M. Guglielmotti, Leuconostoc citreum MB1 as biocontrol agent of Listeria ground beef, Vet. Res. Forum 10 (2019) 193–198.
monocytogenes in milk, J. Dairy Res. 81 (2014) 137–145, https://doi.org/ [39] L. Garnier, J. Mounier, S. Lê, A. Pawtowski, N. Pinon, B. Camier, M. Chatel,
10.1017/S002202991300068X. G. Garric, A. Thierry, E. Coton, F. Valence, Development of antifungal ingredients
[16] M. Moradi, S. Kousheh, H. Almasi, A. Alizadeh, J. Guimarães, N. Yilmaz, A. Lotfi, for dairy products: from in vitro screening to pilot scale application, Food
Postbiotics produced by lactic acid bacteria: the next frontier in food safety, Microbiol. 81 (2019) 97–107, https://doi.org/10.1016/j.fm.2018.11.003.
Compr. Rev. Food Sci. Food Saf. 19 (2020) 3390–3415, https://doi.org/10.1111/ [40] P. Burns, G. Vinderola, F. Molinari, J. Reinheimer, Suitability of whey and
1541-4337.12613. buttermilk for the growth and frozen storage of probiotic Lactobacilli, Int. J. Dairy
[17] C.P. Barros, J.T. Guimarães, E.A. Esmerino, M.C.K.H. Duarte, M.C. Silva, R. Silva, Technol. 61 (2008) 156–164, https://doi.org/10.1111/j.1471-0307.2008.00393.
B.M. Ferreira, A.S. Sant’Ana, M.Q. Freitas, A.G. Cruz, Paraprobiotics and x.
postbiotics: concepts and potential applications in dairy products, Curr. Opin. [41] O.K. Koo, S.M. Kim, S.-H. Kang, Antimicrobial potential of Leuconostoc species
Food Sci. 32 (2020) 1–8, https://doi.org/10.1016/j.cofs.2019.12.003. againstE. coli O157:H7 in ground meat, J. Korean Soc. Appl. Biol. Chem. 58
[18] B.H. Nataraj, S.A. Ali, P.V. Behare, H. Yadav, Postbiotics-parabiotics: the new (2015) 831–838, https://doi.org/10.1007/s13765-015-0112-0.
horizons in microbial biotherapy and functional foods, Microb. Cell Fact. 19 [42] I. Tahiri, M. Desbiens, E. Kheadr, C. Lacroix, I. Fliss, Comparison of different
(2020) 168, https://doi.org/10.1186/s12934-020-01426-w. application strategies of divergicin M35 for inactivation of Listeria monocytogenes
[19] M.J. Hernández-Granados, E. Franco-Robles, Postbiotics in human health: in cold-smoked wild salmon, Food Microbiol. 26 (2009) 783–793, https://doi.
possible new functional ingredients? Food Res. Int. 137 (2020) 109660, https:// org/10.1016/j.fm.2009.05.003.
doi.org/10.1016/j.foodres.2020.109660. [43] M. Koohestani, M. Moradi, H. Tajik, A. Badali, Effects of cell-free supernatant of
[20] A.M.C. Wegh, Y.S. Geerlings, J. Knol, G. Roeselers, C. Belzer, Postbiotics and their Lactobacillus acidophilus LA5 and Lactobacillus casei 431 against planktonic form
potential applications in early life nutrition and beyond, Int. J. Mol. Sci. 20 and biofilm of Staphylococcus aureus, Vet. Res. Forum 9 (2018) 301–306, https://
(2019) 4673, https://doi.org/10.3390/ijms20194673. doi.org/10.30466/vrf.2018.33086.
[21] P. Tomasik, P. Tomasik, Probiotics, non-dairy prebiotics and postbiotics in [44] A. Ołdak, D. Zielińska, A. Łepecka, E. Długosz, D. Kołożyn-Krajewska,
nutrition, Appl. Sci. 10 (2020), https://doi.org/10.3390/app10041470. Lactobacillus plantarum strains isolated from Polish regional cheeses exhibit anti-
[22] J.N. Malagón-Rojas, A. Mantziari, S. Salminen, H. Szajewska, Postbiotics for Staphylococcal activity and selected probiotic properties, Probiotics Antimicrob.
preventing and treating common infectious diseases in children: a systematic Proteins (2019), https://doi.org/10.1007/s12602-019-09587-w.
review, Nutrients. 12 (2020), https://doi.org/10.3390/nu12020389. [45] L. Goudarzi, R. Kermanshahi, Z. Moosavi-Nejad, M.M.S. Dalla, Evaluation of
[23] A. Mantziari, S. Salminen, H. Szajewska, J.N. Malagón-Rojas, Postbiotics against antimicrobial activity of probiotic Lactobacillus strains against growth and urease
pathogens commonly involved in pediatric infectious diseases, Microorganisms. 8 activity of proteus spp, J. Med. Bacteriol. 6 (2017) 31–43. http://jmb.tums.ac.ir/i
(2020), https://doi.org/10.3390/microorganisms8101510. ndex.php/jmb/article/view/335.
[24] P.F. Cuevas-González, A.M. Liceaga, J.E. Aguilar-Toalá, Postbiotics and [46] M. Moradi, K. Mardani, H. Tajik, Characterization and application of postbiotics
Paraprobiotics: from concepts to applications, Food Res. Int. (2020) 109502, of Lactobacillus spp. on Listeria monocytogenes in vitro and in food models, LWT -
https://doi.org/10.1016/j.foodres.2020.109502. Food Sci. Technol. 111 (2019) 457–464, https://doi.org/10.1016/j.
[25] A. Homayouni Rad, L. Aghebati Maleki, H. Samadi Kafil, A. Abbasi, Postbiotics: a lwt.2019.05.072.
novel strategy in food allergy treatment, Crit. Rev. Food Sci. Nutr. (2020) 1–8, [47] S. Chaluvadi, A.T. Hotchkiss, K.L. Yam, Gut microbiota: impact of probiotics,
https://doi.org/10.1080/10408398.2020.1738333. prebiotics, synbiotics, pharmabiotics, and postbiotics on human health, in: R.
[26] C.N. de Almada, C.N. Almada, R.C.R. Martinez, A.S. Sant’Ana, Paraprobiotics: R. Watson, V.R.B.T.-P. Preedy Prebiotics, and Synbiotics (Eds.), Probiotics,
evidences on their ability to modify biological responses, inactivation methods Pebiotics, Synbiotics Bioact. Foods Heal. Promot., Academic Press, 2016,
and perspectives on their application in foods, Trends Food Sci. Technol. 58 pp. 515–523, https://doi.org/10.1016/B978-0-12-802189-7.00036-8.
(2016) 96–114, https://doi.org/10.1016/j.tifs.2016.09.011. [48] A. Ricci, A. Levante, M. Cirlini, L. Calani, V. Bernini, The Influence of viable cells
[27] R. Martín, P. Langella, Emerging health concepts in the probiotics field: and cell-free extracts of Lactobacillus casei on volatile compounds and
streamlining the definitions, Front. Microbiol. 10 (2019) 1047, https://doi.org/ polyphenolic profile of elderberry juice, Front. Microbiol. 9 (2018) 2784, https://
10.3389/fmicb.2019.01047. doi.org/10.3389/fmicb.2018.02784.
[28] T. Teame, A. Wang, M. Xie, Z. Zhang, Y. Yang, Q. Ding, C. Gao, R.E. Olsen, C. Ran, [49] S. Patil, S. Sawant, K. Hauff, G. Hampp, Validated postbiotic screening confirms
Z. Zhou, Paraprobiotics and postbiotics of probiotic Lactobacilli, their positive presence of physiologically-active metabolites, such as short-chain fatty acids,
effects on the host and action mechanisms: a review, Front. Nutr. 7 (2020) 191. amino acids and vitamins in Hylak ® Forte, Probiotics Antimicrob. Proteins 11
https://www.frontiersin.org/article/10.3389/fnut.2020.570344. (2019) 1124–1131.
[29] J.E. Aguilar-Toalá, R. Garcia-Varela, H.S. Garcia, V. Mata-Haro, A.F. González- [50] M.G. Shehata, A.N. Badr, S.A. El Sohaimy, D. Asker, T.S. Awad, Characterization
Córdova, B. Vallejo-Cordoba, A. Hernández-Mendoza, Postbiotics: An evolving of antifungal metabolites produced by novel lactic acid bacterium and their
term within the functional foods field, Trends Food Sci. Technol. 75 (2018) potential application as food biopreservatives, Ann. Agric. Sci. 64 (2019) 71–78,
105–114, https://doi.org/10.1016/j.tifs.2018.03.009. https://doi.org/10.1016/j.aoas.2019.05.002.
[30] J. Żółkiewicz, A. Marzec, M. Ruszczyński, W. Feleszko, Postbiotics—a step [51] J. Yeon, D. Nguyen, Y. Shik, K. Yeol, Y. Seok, Organic a profiling analysis in
beyond pre- and probiotics, Nutrients. 12 (2020), https://doi.org/10.3390/ culture media of lactic acid bacteria by Gas chromatography-mass spectrometry,
nu12082189. Mass Spectrom. Lett. 3 (2012) 74–77, https://doi.org/10.5478/MSL.2012.3.3.74.
[31] M. Pyclik, D. Srutkova, M. Schwarzer, S. Górska, Bifidobacteria cell wall-derived [52] L. Garnier, M. Penland, A. Thierry, M.-B. Maillard, J. Jardin, M. Coton, M. Leyva
exo-polysaccharides, lipoteichoic acids, peptidoglycans, polar lipids and proteins Salas, E. Coton, F. Valence, J. Mounier, Antifungal activity of fermented dairy
– their chemical structure and biological attributes, Int. J. Biol. Macromol. 147 ingredients: identification of antifungal compounds, Int. J. Food Microbiol. 322
(2020) 333–349, https://doi.org/10.1016/j.ijbiomac.2019.12.227. (2020) 108574, https://doi.org/10.1016/j.ijfoodmicro.2020.108574.
[32] J.T. Guimarães, C.F. Balthazar, H. Scudino, T.C. Pimentel, E.A. Esmerino, [53] P. Ruas-Madiedo, C.G. de los Reyes-Gavilán, Methods for the screening, isolation,
M. Ashokkumar, M.Q. Freitas, A.G. Cruz, High-intensity ultrasound: a novel and characterization of exopolysaccharides produced by lactic acid bacteria,
technology for the development of probiotic and prebiotic dairy products, J. Dairy Sci. 88 (2005) 843–856, https://doi.org/10.3168/jds.S0022-0302(05)
Ultrason. Sonochem. 57 (2019) 12–21, https://doi.org/10.1016/j. 72750-8.
ultsonch.2019.05.004. [54] D. Das, A. Goyal, Lactic acid bacteria in food industry, in: T. Satyanarayana, B.
[33] V. Taverniti, S. Guglielmetti, The immunomodulatory properties of probiotic N. Johri (Eds.), Microorg. Sustain. Agric. Biotechnol., Springer, Netherlands,
microorganisms beyond their viability (ghost probiotics: proposal of Dordrecht, 2012, pp. 757–772, https://doi.org/10.1007/978-94-007-2214-9_33.
paraprobiotic concept), Genes Nutr. 6 (2011) 261–274, https://doi.org/10.1007/ [55] C. Axel, B. Röcker, B. Brosnan, E. Zannini, A. Furey, A. Coffey, E.K. Arendt,
s12263-011-0218-x. Application of Lactobacillus amylovorus DSM19280 in gluten-free sourdough
[34] P.F. Cuevas-González, J.E. Aguilar-Toalá, H.S. García, A.F. González-Córdova, bread to improve the microbial shelf life, Food Microbiol. 47 (2015) 36–44,
B. Vallejo-Cordoba, A. Hernández-Mendoza, Protective effect of the intracellular https://doi.org/10.1016/j.fm.2014.10.005.
content from potential probiotic bacteria against oxidative damage induced by [56] J. Wang, W. Hui, C. Cao, R. Jin, C. Ren, H. Zhang, W. Zhang, Proteomic analysis
acrylamide in human erythrocytes, Probiotics Antimicrob. Proteins 12 (2020) of an engineered isolate of Lactobacillus plantarum with enhanced raffinose
1459–1470, https://doi.org/10.1007/s12602-020-09636-9. metabolic capacity, Nat. Publ. Gr. 6 (2016) 31403, https://doi.org/10.1038/
[35] J.N. González-González, B. Vallejo-Cordoba, A.F. González-Córdova, L. srep31403.
M. Beltrán-Barrientos, H.S. García, A. Hernández-Mendoza, Effect of the

10
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

[57] H. Wang, Y. Yan, J. Wang, H. Zhang, W. Qi, Production and characterization of [80] S. Tejero-Sariñena, J. Barlow, A. Costabile, G.R. Gibson, I. Rowland, In vitro
antifungal compounds produced by Lactobacillus plantarumIMAU10014, PLoS One evaluation of the antimicrobial activity of a range of probiotics against pathogens:
7 (2012) e29452, https://doi.org/10.1371/journal.pone.0029452. evidence for the effects of organic acids, Anaerobe. 18 (2012) 530–538, https://
[58] S.B. Lee, Y.K. Rhee, E. Gu, D. Kim, G. Jang, S. Song, J. Lee, B. Kim, H. Lee, doi.org/10.1016/j.anaerobe.2012.08.004.
H. Hong, C. Cho, H. Kim, Mass-based etabolomic analysis of Lactobacillus sakei [81] B. Singh, G. Mal, S.K. Gautam, M. Mukesh, Nutraceuticals from bioengineered
and its growth media at different growth phases, J. Microbiol. Biotechnol. 27 microorganisms, in: B. Singh, G. Mal, S.K. Gautam, M. Mukesh (Eds.), Adv. Anim.
(2017) 925–932. Biotechnol., Springer International Publishing, Cham, 2019, pp. 59–69, https://
[59] M. Leyva Salas, J. Mounier, M.-B. Maillard, F. Valence, E. Coton, A. Thierry, doi.org/10.1007/978-3-030-21309-1_6.
Identification and quantification of natural compounds produced by antifungal [82] S.M. Ameen, G. Caruso, Lactic Acid Bacteria in Food Industry, Springer,
bioprotective cultures in dairy products, Food Chem. 301 (2019) 125260, https:// Netherlands, Dordrecht, 2012, https://doi.org/10.1007/978-94-007-2214-9_33.
doi.org/10.1016/j.foodchem.2019.125260. [83] S. Özcelik, E. Kuley, F. Özogul, Formation of lactic, acetic, succinic, propionic,
[60] B. Brosnan, A. Coffey, E.K. Arendt, A. Furey, The QuEChERS approach in a novel formic and butyric acid by lactic acid bacteria, LWT - Food Sci. Technol. 73
application for the identification of antifungal compounds produced by lactic acid (2016) 536–542, https://doi.org/10.1016/j.lwt.2016.06.066.
bacteria cultures, Talanta. 129 (2014) 364–373, https://doi.org/10.1016/j. [84] M.F. Ooi, N. Mazlan, H.L. Foo, T.C. Loh, R. Mohamad, R.A. Rahim, A. Ariff,
talanta.2014.05.006. Effects of carbon and nitrogen sources on bacteriocin-inhibitory activity of
[61] H.-S. Lim, J.-E. Yeu, S.-P. Hong, M.-S. Kang, Characterization of antibacterial cell- postbiotic metabolites produced byLactobacillus plantarum I-UL4, Malays. J.
free supernatant from oral care probiotic Weissella cibaria, CMU, Molecules. 23 Microbiol. 11 (2015) 176–184.
(2018) 1–13, https://doi.org/10.3390/molecules23081984. [85] H.K. Tan, H.L. Foo, T.C. Loh, N. Banu, M. Alitheen, R. Abdul Rahim, Cytotoxic
[62] D. Sharma, B.S. Saharan, N. Chauhan, S. Procha, S. Lal, Isolation and functional effect of proteinaceous postbiotic metabolites produced by Lactobacillus
characterization of novel biosurfactant produced by Enterococcus faecium, plantarumI-UL4 cultivated in different media composition on MCF-7 breast cancer
Springerplus. 4 (2015) 4. cell, Malays. J. Microbiol. 11 (2015) 207–214.
[63] D. Sharma, B. Saharan, N. Chauhan, A. Bansal, S. Procha, Production and [86] J. Miyamoto, M. Igarashi, K. Watanabe, S. Karaki, H. Mukouyama, S. Kishino,
structural characterization of Lactobacillus helveticus derived biosurfactant, Sci. X. Li, A. Ichimura, J. Irie, Y. Sugimoto, T. Mizutani, T. Sugawara, T. Miki,
World J. 2014 (2014). J. Ogawa, D.J. Drucker, M. Arita, H. Itoh, I. Kimura, Gut microbiota confers host
[64] Y.Y. Hor, M.T. Liong, Dermatologica Sinica use of extracellular extracts of lactic resistance to obesity by metabolizing dietary polyunsaturated fatty acids, Nat.
acid bacteria and bi fi dobacteria for the inhibition of dermatological pathogen Commun. 10 (2019) 4007, https://doi.org/10.1038/s41467-019-11978-0.
Staphylococcus aureus, Dermatologica Sin. 32 (2014) 141–147, https://doi.org/ [87] K.Y. Kareem, F. Hooi Ling, L. Teck Chwen, O. May Foong, S. Anjas Asmara,
10.1016/j.dsi.2014.03.001. Inhibitory activity of postbiotic produced by strains of Lactobacillus plantarum
[65] M.M. Ayyash, F. Sherkat, N.P. Shah, The effect of NaCl substitution with KCl on using reconstituted media supplemented with inulin, Gut Pathog. 6 (2014) 23,
proteinase activities of cell-free extract and cell-free supernatant at different pH https://doi.org/10.1186/1757-4749-6-23.
levels and salt concentrations : lactobacillus acidophilus andLactobacillus casei, [88] V. Cleusix, C. Lacroix, S. Vollenweider, G. Le Blay, Glycerol induces reuterin
J. Dairy Sci. 96 (2013) 40–49, https://doi.org/10.3168/jds.2012-5761. production and decreases Escherichia colipopulation in an in vitro model of
[66] B. Rühmann, J. Schmid, V. Sieber, Methods to identify the unexplored diversity of colonic fermentation with immobilized human feces, FEMS Microbiol. Ecol. 63
microbial exopolysaccharides, Front. Microbiol. 6 (2015) 565, https://doi.org/ (2008) 56–64, https://doi.org/10.1111/j.1574-6941.2007.00412.x.
10.3389/fmicb.2015.00565. [89] S. Abdollahi, M.H. Ghahremani, N. Setayesh, N. Samadi, Listeria monocytogenes
[67] Y.-H. Zhang, D. Xu, J.-Q. Liu, X.-H. Zhao, Enhanced degradation of five and Salmonella enterica affect the expression of nisin gene and its production
organophosphorus pesticides in skimmed milk by lactic acid bacteria and its byLactococcus lactis, Microb. Pathog. 123 (2018) 28–35, https://doi.org/
potential relationship with phosphatase production, Food Chem. 164 (2014) 10.1016/j.micpath.2018.06.024.
173–178, https://doi.org/10.1016/j.foodchem.2014.05.059. [90] P. Chanos, T. Mygind, Co-culture-inducible bacteriocin production in lactic acid
[68] A. Broberg, K. Jacobsson, K. Ström, A. Broberg, K. Jacobsson, K. Stro, J. Schnu, bacteria, Appl. Microbiol. Biotechnol. 100 (2016) 4297–4308, https://doi.org/
Metabolite profiles of lactic acid bacteria in grass silage, Appl. Environ. 10.1007/s00253-016-7486-8.
Microbiol. 73 (2007), https://doi.org/10.1128/AEM.02939-06. [91] C. Liu, B. Hu, Y. Liu, S. Chen, Stimulation of nisin production from whey by a
[69] M.V. Arasu, N.A. Al-Dhabi, T.S. Rejiniemon, K.D. Lee, V.A.J. Huxley, D.H. Kim, mixed culture of Lactococcus lactisand Saccharomyces cerevisiae, Appl. Biochem.
V. Duraipandiyan, P. Karuppiah, K.C. Choi, Identification and characterization of Biotechnol. 131 (2006) 751–761, https://doi.org/10.1385/ABAB:131:1:751.
Lactobacillus brevis P68 with antifungal, antioxidant and probiotic functional [92] M. Ariana, J. Hamedi, Enhanced production of nisin by co-culture of Lactococcus
properties, Indian J. Microbiol. 55 (2015) 19–28, https://doi.org/10.1007/ lactis sub sp. Lactis and Yarrowia lipolytica in molasses based medium,
s12088-014-0495-3. J. Biotechnol. 256 (2017) 21–26, https://doi.org/10.1016/j.jbiotec.2017.07.009.
[70] T. Lin, T. Pan, Characterization of an antimicrobial substance produced by [93] C.S. Prado, W.L.M. Santos, C.R. Carvalho, E.C. Moreira, O. Costa, Antimicrobial
Lactobacillus plantarum NTU 102, J. Microbiol. Immunol. Infect. (2017) 1–9, activity of lactic acid bacteria isolated from Brazilian dry fermented sausages
https://doi.org/10.1016/j.jmii.2017.08.003. againstListeria monocytogenes, Arq. Bras. Med. Veterinária E Zootec. 52 (2000)
[71] S.K. Satpute, G.R. Kulkarni, A.G. Banpurkar, I.M. Banat, Biosurfactant/s from 417–423.
Lactobacilli species: properties, challenges and potential biomedical applications, [94] C. Hui, H. Li, Q. Ren, Y. Zhou, Characterization of antimicrobial activity of three
J. Basic Microbiol. 56 (2016) 1140–1158, https://doi.org/10.1002/ Lactobacillus plantarum strains isolated from Chinese traditional dairy food, Food
jobm.201600143. Sci. Nutr. 7 (2019) 1997–2005, https://doi.org/10.1002/fsn3.1025.
[72] A. Shafipour Yordshahi, M. Moradi, H. Tajik, R. Molaei, Design and preparation [95] U. George-Okafor, U. Ozoani, F. Tasie, K. Mba-Omeje, The efficacy of cell-free
of antimicrobial meat wrapping nanopaper with bacterial cellulose and supernatants from Lactobacillus plantarum Cs andLactobacillus acidophilusATCC
postbiotics of lactic acid bacteria, Int. J. Food Microbiol. 321 (2020) 108561, 314 for the preservation of home-processed tomato-paste, Sci. African. 8 (2020)
https://doi.org/10.1016/j.ijfoodmicro.2020.108561. e00395, https://doi.org/10.1016/j.sciaf.2020.e00395.
[73] I. Trabelsi, S. Ben Slima, H. Chaabane, B.S. Riadh, Purification and [96] W. Pelyuntha, C. Chaiyasut, D. Kantachote, Organic acids and 2,4-Di-tert-butyl­
characterization of a novel exopolysaccharides produced by Lactobacillus sp, Ca6, phenol: major compounds of Weissella confusa WM36 cell-free supernatant
Int. J. Biol. Macromol. 74 (2015) 541–546, https://doi.org/10.1016/j. against growth, survival and virulence of Salmonella typhi, PeerJ. 8 (2020) e8410,
ijbiomac.2014.12.045. https://doi.org/10.7717/peerj.8410.
[74] J. Magnusson, K. Stro, S. Roos, J. Schnu, Broad and complex antifungal activity [97] I. Sajid, K.A. Shaaban, S. Hasnain, Identification, isolation and optimization of
among environmental isolates of lactic acid bacteria, FEMS Microbiol. Lett. 219 antifungal metabolites from the Streptomyces Malachitofuscus ctf9, Braz. J.
(2003) 129–135, https://doi.org/10.1016/S0378-1097(02)01207-7. Microbiol. 42 (2011) 592–604.
[75] H.A. Hartmann, T. Wilke, R. Erdmann, Efficacy of bacteriocin-containing cell-free [98] B. Brosnan, A. Coffey, E.K. Arendt, A. Furey, Rapid identification fungal
culture supernatants from lactic acid bacteria to controlListeria monocytogenesin compounds produced by lactic acid bacteria, by use of the LTQ Orbitrap hybrid
food, Int. J. Food Microbiol. 146 (2011) 192–199, https://doi.org/10.1016/j. FT mass spectrometer, Anal. Bioanal. Chem. 403 (2012) 2983–2995, https://doi.
ijfoodmicro.2011.02.031. org/10.1007/s00216-012-5955-1.
[76] A. Yépez, C. Luz, G. Meca, G. Vignolo, J. Mañes, R. Aznar, Biopreservation [99] A. Guimarães, A. Venancio, L. Abrunhosa, Antifungal effect of organic acids from
potential of lactic acid bacteria from Andean fermented food of vegetal origin, lactic acid bacteria on Penicillium nordicum, Food Addit. Contam. Part A. 35
Food Control 78 (2017) 393–400, https://doi.org/10.1016/j. (2018) 1803–1818, https://doi.org/10.1080/19440049.2018.1500718.
foodcont.2017.03.009. [100] J. Guo, B. Brosnan, A. Furey, E.K. Arendt, P. Murphy, A. Coffey, Antifungal
[77] Y.K. Im, J.Y.W. Hang, K.Y.W. Hang, O.H. Sejong, S.H.K. Im, Characterization of activity of Lactobacillus against Microsporum canis, Microsporum gypseum and
the cholesterol-reducing activity in a cell-free supernatant of Lactobacillus Epidermophyton floccosum, Bioeng. Bugs 3 (2012) 104–113.
acidophilusATCC 43121, Biosci. Biotechnol. Biochem. 72 (2008) 1483–1490, [101] M. Chen, A. Li, M. Sun, Z. Feng, X. Meng, Y. Wang, Optimization of the quenching
https://doi.org/10.1271/bbb.70802. method for metabolomics analysis of Lactobacillus bulgaricus, J. Zheijang Univ.
[78] I.A. Rather, B.J. Seo, V.J.R. Kumar, U. Choi, K. Choi, J.H. Lim, Y. Park, Isolation Sci. B - Biomed. Biotechnol. 15 (2014) 333–342, https://doi.org/10.1631/jzus.
and characterization of a proteinaceous antifungal compound from Lactobacillus B1300149.
plantarum YML007 and its application as a food preservative, Lett. Appl. [102] M. Peng, Z. Tabashsum, P. Patel, C. Bernhardt, M.S. Palermo, Linoleic acids
Microbiol. 57 (2013) 69–76, https://doi.org/10.1111/lam.12077. overproducing Lactobacillus casei limits growth, survival, and virulence of
[79] A. Thierry, F. Valence, S.-M. Deutsch, S. Even, H. Falentin, Y. Le Loir, G. Jan, Salmonella typhimurium and Enterohaemorrhagic Escherichia coli, Front.
V. Gagnaire, Strain-to-strain differences within lactic and propionic acid bacteria Microbiol. 9 (2018) 2663, https://doi.org/10.3389/fmicb.2018.02663.
species strongly impact the properties of cheese–A review, Dairy Sci. Technol. 95 [103] J. Sjögren, J. Magnusson, A. Broberg, J. Schnürer, L. Kenne, Antifungal 3-hydroxy
(2015) 895–918, https://doi.org/10.1007/s13594-015-0267-9. fatty acids from Lactobacillus plantarum MiLAB 14, Appl. Environ. Microbiol. 69
(2003), https://doi.org/10.1128/AEM.69.12.7554-7557.2003, 7554 LP-7557.

11
M. Moradi et al. Enzyme and Microbial Technology 143 (2021) 109722

[104] H.A. Rasheed, T. Tuoheti, Y. Zhang, F. Azi, M. Tekliye, M. Dong, Purification and extended-spectrum β-lactamase producing Klebsiella pneumoniaeand Pseudomonas
partial characterization of a novel bacteriocin produced by bacteriocinogenic aeruginosa, Infect. Drug Resist. 13 (2020) 543–552, https://doi.org/10.2147/IDR.
Lactobacillus fermentum BZ532 isolated from Chinese fermented cereal beverage S235603.
(Bozai), LWT. 124 (2020) 109113, https://doi.org/10.1016/j.lwt.2020.109113. [114] N. Singh, C. Sharma, R.D. Gulhane, N. Rokana, B.P. Singh, A.K. Puniya, S. Attri,
[105] J. Pei, W. Jin, A.M. Abd El-Aty, D.A. Baranenko, X. Gou, H. Zhang, J. Geng, G. Goel, H. Panwar, Inhibitory effects of lactobacilli of goat’s milk origin against
L. Jiang, D. Chen, T. Yue, Isolation, purification, and structural identification of a growth and biofilm formation by pathogens: an in vitro study, Food Biosci. 22
new bacteriocin made by Lactobacillus plantarum found in conventional (2018) 129–138, https://doi.org/10.1016/j.fbio.2018.02.001.
kombucha, Food Control 110 (2020) 106923, https://doi.org/10.1016/j. [115] N. Toy, F. Özogul, Y. Özogul, The influence of the cell free solution of lactic acid
foodcont.2019.106923. bacteria on tyramine production by food borne-pathogens in tyrosine
[106] Z. Qiao, H. Sun, Q. Zhou, L. Yi, X. Wang, Y. Shan, Y. Yi, B. Liu, Y. Zhou, X. Lü, decarboxylase broth, Food Chem. 173 (2015) 45–53, https://doi.org/10.1016/j.
Characterization and antibacterial action mode of bacteriocin BMP32r and its foodchem.2014.10.001.
application as antimicrobial agent for the therapy of multidrug-resistant bacterial [116] C. Xie, H. Wang, S. Deng, X.-L. Xu, The inhibition of cell-free supernatant
infection, Int. J. Biol. Macromol. 164 (2020) 845–854, https://doi.org/10.1016/j. ofLactobacillus plantarum on production of putrescine and cadaverine by four
ijbiomac.2020.07.192. amine-positive bacteria in vitro, LWT - Food Sci. Technol. 67 (2016) 106–111,
[107] X. Lv, H. Ma, M. Sun, Y. Lin, F. Bai, J. Li, B. Zhang, A novel bacteriocin DY4-2 https://doi.org/10.1016/j.lwt.2015.11.028.
produced by Lactobacillus plantarum from cutlassfish and its application as bio- [117] S.A. Kousheh, M. Moradi, H. Tajik, R. Molaei, Preparation of antimicrobial/
preservative for the control of Pseudomonas fluorescens in fresh turbot ultraviolet protective bacterial nanocellulose film with carbon dots synthesized
(Scophthalmus maximus) fillets, Food Control 89 (2018) 22–31, https://doi.org/ from lactic acid bacteria, Int. J. Biol. Macromol. 155 (2020) 216–225, https://doi.
10.1016/j.foodcont.2018.02.002. org/10.1016/j.ijbiomac.2020.03.230.
[108] S.B. Wayah, K. Philip, Purification, characterization, mode of action, and [118] S.C. Beristain-Bauza, E. Mani-López, E. Palou, A. López-Malo, Antimicrobial
enhanced production of Salivaricin mmaye1, a novel bacteriocin from activity and physical properties of protein films added with cell-free supernatant
Lactobacillus salivarius SPW1 of human gut origin, Electron. J. Biotechnol. 35 of Lactobacillus rhamnosus, Food Control 62 (2016) 44–51, https://doi.org/
(2018) 39–47, https://doi.org/10.1016/j.ejbt.2018.08.003. 10.1016/j.foodcont.2015.10.007.
[109] V. Fuochi, M.A. Coniglio, L. Laghi, A. Rescifina, M. Caruso, A. Stivala, P. [119] K.J. Lee, H.W. Park, E.J. Choi, H.H. Chun, Effects of CFSs produced by lactic acid
M. Furneri, Metabolic characterization of supernatants produced by Lactobacillus bacteria in combination with grape seed extract on the microbial quality of ready-
spp. With in vitro anti-legionella activity, Front. Microbiol. 10 (2019) 1403. https to-eat baby leaf vegetables, Cogent Food Agric. 2 (2016) 1268742, https://doi.
://www.frontiersin.org/article/10.3389/fmicb.2019.01403. org/10.1080/23311932.2016.1268742.
[110] S.D. Aunsbjerg, A.H. Honoré, J. Marcussen, P. Ebrahimi, F.K. Vogensen, [120] G.N. Tenea, A. Barrigas, The efficacy of bacteriocin-containing cell-free
C. Benfeldt, T. Skov, S. Knøchel, Contribution of volatiles to the antifungal effect supernatant from Lactobacillus plantarum Cys5-4 to control pathogenic bacteria
of Lactobacillus paracasei in defined medium and yogurt, Int. J. Food Microbiol. growth in artisanal beverages, Int. Food Res. J. 25 (2018) 2131–2137.
194 (2015) 46–53, https://doi.org/10.1016/j.ijfoodmicro.2014.11.007. [121] M. Ramezani, N. Zainodini, H. Hakimi, E. Rezazadeh Zarandi, V. Bagheri,
[111] S. Siedler, R. Balti, A.R. Neves, Bioprotective mechanisms of lactic acid bacteria R. Bahramabadi, M. Zare-Bidaki, Cell-free culture supernatants of
against fungal spoilage of food, Curr. Opin. Biotechnol. 56 (2019) 138–146, Lactobacillimodify the expression of virulence factors genes inStaphylococcus
https://doi.org/10.1016/j.copbio.2018.11.015. aureus, Jundishapur J. Microbiol. 12 (2019) e96806, https://doi.org/10.5812/
[112] G.M. Hamad, N.M. Abdelmotilib, A.M.G. Darwish, A.M. Zeitoun, Commercial jjm.96806.
probiotic cell-free supernatants for inhibition of Clostridium perfringens poultry [122] G. Hamad, W. Botros, E. Hafez, Combination of probiotic filtrates as antibacterial
meat infection in Egypt, Anaerobe. 62 (2020) 102181, https://doi.org/10.1016/j. agent against selected some pathogenic bacteria in milk and cheese, Int. J. Dairy
anaerobe.2020.102181. Sci. Process. 12 (2017) 368–376.
[113] M.A. El-Mokhtar, K.M. Hassanein, A.S. Ahmed, G.F.M. Gad, M.M. Amin, O.F.
E. Hassanein, Antagonistic activities of cell-free supernatants of lactobacilli against

12

You might also like