You are on page 1of 16

Science of the Total Environment 825 (2022) 154058

Contents lists available at ScienceDirect

Science of the Total Environment


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

Combined antimicrobial effect of bacteriocins with other hurdles of


physicochemic and microbiome to prolong shelf life of food: A review

Guorong Liu , Rong Nie, Yangshuo Liu, Arshad Mehmood
Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Laboratory of Food Quality and Safety, Beijing Engineering and Technology Research Center of
Food Additives, Beijing Technology and Business University (BTBU), Beijing 100048, China

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• Combination of bacteriocins with other


hurdles markedly improve food safety.
• Synergistic effects are observed between
bacteriocin and other hurdles.
• Combination of bacteriocins with other
hurdles could expand the antibacterial
spectrum.
• Combination of bacteriocins with other
hurdles could reduce usage.
• Combination of bacteriocins with other
hurdles could extend shelf life.

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

Article history: Bacteriocins are ribosomally synthesized peptides to inhibit food spoilage bacteria, which are widely used as a kind of
Received 18 October 2021 food biopreservation. The role of bacteriocins in therapeutics and food industries has received increasing attention
Received in revised form 24 January 2022 across a number of disciplines in recent years. Despite their advantages as alternative therapeutics over existing strat-
Accepted 17 February 2022
egies, the application of bacteriocins suffers from shortcomings such as the high isolation and purification cost, narrow
Available online 22 February 2022
spectrum of activity, low stability and solubility and easy enzymatic degradation. Previous studies have studied the
Editor: Vijai Kumar Kumar synergistic or additive effects of bacteriocins when used in combination with other hurdles including physics,
chemicals, and microbes. These combined treatments reduce the adverse effects of chemical additives, extending
Keywords: the shelf life of food products while guaranteeing food quality. This review highlights the advantages and disadvan-
Bacteriocins tages of bacteriocins in food preservation. It then reviews the combined effect and mechanism of different hurdles
Combination and bacteriocins in enhancing food preservation in detail. The combination of bacterioncins and other hurdles provide
Hurdle technologies potential approaches for maintaining food quality and food safety.
Food preservation

1. Introduction (IPHS) (Manea et al., 2017). A report published by the Centers for Disease
Control and Prevention (CDC) documented that annual foodborne illnesses
The ever increasing globalization of food trade and a strive towards lon- in the United States result in around 76 million infections, 325,000 hospi-
ger shelf-lives imply that novel food preservation technologies are needed talizations, and 5000 deaths. Over 90% of deaths are caused by Bacillus ce-
due to trends of convenience and durability to support long-distance trans- reus, Staphylococcus aureus, Listeria monocytogenes, Campylobacter spp.,
port (Elsser-Gravesen and Elsser-Gravesen, 2013). Meanwhile, foodborne Salmonella spp., and Escherichia coli O157:H7 (Scallan et al., 2011; Van
pathogens threat the safety of international public health and safety Cauteren et al., 2017). Consumer demands are varying for low salt, less

⁎ Corresponding author.
E-mail address: liuguorong@th.btbu.edu.cn (G. Liu).

http://dx.doi.org/10.1016/j.scitotenv.2022.154058
0048-9697/© 2022 Elsevier B.V. All rights reserved.
G. Liu et al. Science of the Total Environment 825 (2022) 154058

sugar and fat in food in pursuit of healthy diet while higher pH for milder combined to have an additive or synergistic effect, could be useful to meet
taste, minimal processing for fresher appearance of the food. These de- current consumer demand.
mands result in a friendlier and less adverse environment for unwanted mi- Bacteriocins are natural antibacterial peptides capable of inhibiting the
croorganisms (Elsser-Gravesen and Elsser-Gravesen, 2013). This growth of foodborne pathogens or spoilage bacteria and some of them
significantly affects the industry when maintaining food quality. To guaran- could be potential biopreservatives for food industries, as i) bacteriocins
tee food safety while preserving the nutritions and sensory properties of markedly retard the growth of C. perfringens, E. coli, Salmonella enteritidis,
food, various preservation techniques are developed including heat treat- B. cereus, and L. monocytogenes (Yang et al., 2012); ii) most bacteriocins
ment (heating, boiling, drying, etc.), cold storage (chilling, frozen, etc.), are safe for human microbiome, because they only inhibit sensitive bacteria
modern preservation methods (pasteurization, canning, etc.), chemical pre- but leave beneficial bacteria intact since most bacteriocins can be degraded
servatives and biopreservation to retard the growth of spoilage microorgan- by enzymes in human body; iii) most of foodborne pathogens and spoilage
isms. However, heat treatment and modern preservation methods may microorganisms are not bacteriocin-resistant, and some bacteriocins can in-
change the sensory properties and destroy the nutritients in food (Smelt hibit methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-
and Brul, 2014); chilling to an unsuitable low temperature is detrimental resistant E. faecalis strains (Kruszewska et al., 2004; Millette et al., 2008);
to some foods of plant origin causing chilling injury (Leistner, 2000); chem- iv) bacteriocins have minimal effects on the nutrients and sensory proper-
ical preservatives were proven to cause allergies and catalyzing the forma- ties of food. However, only a few bacteriocins have been commercialized:
tion of carcinogenic end-products (Pisoschi et al., 2018) (Table 1). These i) nisin has been approved by regulatory authorities including the World
contradictory trends and demands put food biopreservation under the spot- Health Organisation (WHO)/Food Development Authority (FDA) in the
light. A “Hurdle Concept”, two or more suitable preservation techniques are USA and the European Food Safety Authority (ESFA) in Europe as a pure

Table 1
Overview on the advantages and disadvantages of current technologies for food preservation.
Type Hurdles Advantages Disadvantages

⟡ Heat resistance of spores


⟡ Cost-effective ⟡ Affect the taste, texture, smell and color of food
Heat
⟡ Widely available ⟡ Do not satisfy demand for fresh-like products
⟡ Energy consumption
⟡ Slow down the chemical and biological processes in
foods ⟡ Rely on mechanical refrigeration systems or ice
Chilling
⟡ Minimal effects on the nutritional values and sensory ⟡ Chilling injury
properties of food products
Physical Pulsed electric ⟡ Energy efficient
⟡ High cost
hurdles field ⟡ Minimal losses of flavor and food quality
⟡ Widely available
High pressure ⟡ Pressure resistance strains
⟡ Do not affect the taste, texture, smell and color of food
processing ⟡ High cost
⟡ Reduce allergic substances
⟡ Stability in physiological solution
⟡ High surface area-to volume ratio ⟡ Complexified combination processing
Nanotechnology ⟡ Easy to synthesize with low production cost ⟡ Unavailability of proper in vivo experimental information
⟡ Non-toxic nature at low concentrations ⟡ Limited research on the toxic nature of the combinations
⟡ Do not cause rapid resistance of bacteria
⟡ Broad antimicrobial spectrum ⟡ Unacceptable organoleptic properties
Essential oils ⟡ Biodegradable ⟡ Complicated content
⟡ GRAS ⟡ Pose hazards to pregnant women
⟡ Disruption of the outer membrane of some G− bacteria ⟡ Toxicity at relevant concentrations
EDTA
⟡ Make G− bacteria sensitive to bacteriocins ⟡ Environment unfriendly
Organic acid ⟡ Wide antimicrobial spectrum ⟡ Affect taste of food
Chemical ⟡ Wide and effective antimicrobial spectrum
Salts of organic ⟡ Side effects to human
hurdles ⟡ Low cost
acid ⟡ Cause concerns of costumers
⟡ High stability
⟡ Antimicrobial system naturally present in raw milk
Lactoperoxidase
⟡ Wide inhibition spectrum ⟡ Less stable under acidic conditions
system
⟡ Resistant to proteolytic enzymes
⟡ Antimicrobial effect of G+ bacteria
Lysozyme ⟡ High resistance of G− bacteria
⟡ Non-toxic and harmless, high safety
⟡ Wide inhibition spectrum
⟡ Water soluble substance ⟡ Poor inhibition of L. monocytogenes
Reuterin
⟡ Active at a wide range of pH values ⟡ High prurition cost
⟡ Resistant to proteolytic and lipolytic enzymes
⟡ Effective inhabitation of G− bacteria ⟡ Toxicity at relevant concentrations
Polymyxin B
Microbial ⟡ High solubility ⟡ Fail to inhabit G+ bacteria
hurdles Surface layer ⟡ Fail to affect G+ bacteria
⟡ Endopeptidase activity against cell wall of G− bacteria
protein ⟡ Hardly extract
⟡ Highly specific
⟡ Narrow host range
⟡ Low cost
Bacteriophages ⟡ Potential for negative consumer perception regarding the use of the terminology
⟡ Do not affect taste, texture, smell and color of food
“viruses” or “virulent phages” in foods
⟡ Widespread in the natural environment
⟡ Effective antimicrobial activity
⟡ GRAS ⟡ Relatively narrow spectrum of antibacterial activity
⟡ Highly specific ⟡ Rapid but not long bactericidal effect
Bacteriocins ⟡ Minimal effects on the nutritional values and sensory ⟡ Low yield and high production and purification cost
properties of food products ⟡ Low solubility and diffusivity in food matrix
⟡ Widespread in the natural environment ⟡ Adverse interactions of bacteriocins and food matrix
⟡ Low toxicity towards eukaryotic cells

2
G. Liu et al. Science of the Total Environment 825 (2022) 154058

bacteriocin preparation; ii) MicroGARDTM range and ALTA 2431 which negative bacteria and archaea (Johnson et al., 2018) (Fig. 1). Among the
both contain pediocin PA-1 have been approved by FDA as bacteriocin con- gram-positive bacteria, lactic acid bacteria (LAB) can produce bacteriocins
taining fermentates; iii) bacteriocins can be used as bacteriocin cultures, in- with most prepositive antimicrobial peptides to preserve food in a natural
cluding the BactofermTM range, HOLDBAC® protective cultures and way. Threfore, LABs are recognized to be “Generally Regarded As Safe
Micocin® (O’Connor et al., 2020). (GRAS)” by the FDA (Chen and Hoover, 2003). Moreover, some LAB bacte-
Only a very few of bacteriocins have been commercialized out of those riocins like enterocin AS-48, enterocin P, paracin 1.7, plantaricin MG and
discovered and preserved in laboratory. Bacteriocins facing an application lactococcin BZ can inhibit the growth of Salmonella which have resistance
difficulty because i) some bacteriocins are known to exhibit a relatively nar- to most bacteriocins (Chalón et al., 2012). Since LAB is generally present
row spectrum of antibacterial activity; ii) enterococcal cytolysin has a wide in devious fermented food, LAB for bacteriocins production could be used
spread cytotoxic activity (Cox et al., 2005) and gram-negative bacteriocins as a star culture without goting through rigorous purification procedure
when producing strains may also produce some endotoxins resulting in del- (Mills et al., 2017). Bacteriocins from Gram-negative bacteria like
eterious effect, thus requiring rigorous purification procedure; iii) the per- microcins are less studied as food preservatives, because they are produced
meability of the bacteriocins in food matrix, the pH of the food matrix, by E. coli which is potentially threatening food safety and has a narrow
enzymes and other adverse interactions of bacteriocins and food matrix in- spectrum of antibacterial activity (Chalón et al., 2012). Only a few bacterio-
fluence the efficacy of pure bacteriocins; iv) it is reported that strains of cins produced by archaea were reported, including sulfolobicins from
L. monocytogenes and S. aureus are becoming resistant to nisin, plantaricin Sulfolobus and halocin from Halogeometricum sp., and most studies are re-
C19 and sakacin A (Gálvez et al., 2007). Therefore, bacteriocins alone is lated to clinical application (O’Connor and Shand, 2002).
not enough to control food safety, let along its high monetary cost (Fig. 1). Bacteriocins are classified based on the chemical structures, types of
To expand the antibacterial spectrum, reduce usage, enhance antibacte- producing organism, physical properties, mode of action, stability and mo-
rial effect and avoid the resistance, the hurdle technology that involves lecular size. Class I bacteriocins are mainly 19–50 amino acids in size,
many physicochemical treatments with the bacteriocin has attracted con- consisting of highly post-translationally modified emanate of the non-
siderable attention. As suggested, hurdle technology could help adjust the standard amino acids (Parada et al., 2007). Class II bacteriocins comprise
level of bacteriocin to maximize the viability loss of the target bacteria, small non-modified peptides that are heat-stable. The Cass II is further di-
thus minimizing the occurance of resistances in food preservation. The hur- vided into pediocin-like bacteriocins (Class IIa), two-peptide unmodified
dles combined with bacteriocins include (i) physical hurdles: temperature bacteriocins (Class IIb), circular bacteriocins (Class IIc), and unmodified,
(Aras et al., 2020), pulsed electric fields (Pol et al., 2001), high-pressure linear, non-pediocin-like bacteriocins (Class IId) (Drider et al., 2006;
processing (Ramaroson et al., 2018), nanotechnology (Sulthana and Oppegård et al., 2007). Class III bacteriocins include heat-labile proteins
Archer, 2021); (ii) chemical hurdles: essential oils (Alves et al., 2016; Du with large-molecular-weight (>30 kDa) (Parada et al., 2007). Class IV bac-
et al., 2019; Shi et al., 2017), EDTA (Ay and Tuncer, 2016; Castellano teriocins which originally refer to antimicrobial peptides containing large
et al., 2011; Liang et al., 2020), organic acids (Zhao et al., 2017), animal- peptides combined with carbohydrates or lipids are cancelled as the com-
derived enzymes (Arqués et al., 2008a, 2008b); (iii) microbial hurdles: plex structures are not thoroughly purified (Güllüce and Karaday, 2013).
other bacteriocins, reuterin, bacteriophages (Duc et al., 2020; Heo et al., The representative member of Class I, II, III are nisin, pediocin and colicin,
2018). Previous research has observed a synergistic effect when bacterio- respectively.
cins are combined with above hurdles, which gets a maximum lethality
against micro-organisms with the minimum damage to the sensory param- 3. Mode of action of bacteriocins
eters of the food.
This paper sets out to describe the combined antimicrobial effect of bac- The main mode of action of bacteriocins is disturbing the cytoplasmic
teriocins with other hurdles as well as strategic usage in food system. In ad- membrane with pores, or causing cell wall degradation (Fig. 2). Apart
dition, this study also preliminary summary their combination mechaism. It from this, bacteriocins may affect living cells in some other ways, such as
is necessary to enhance the study of bacteriocins and other hurdles to facil- microcin B17 inhibits DNA synthesis, colicins D and E5 can increase ex-
itate bacteriocins application in food industries. haustion of tRNA which slows down protein synthesis (Sulthana and
Archer, 2021), microcin J25 inhibit RNA polymerase inhibitor (Lopez
2. Source and classification of bacteriocins et al., 2007). Nevertheless, bacteriocins' modes of action has not been
fully discovered, especially those that are active against Gram-negative bac-
Microbes of different groups demonstrate a widely ranging capabilities teria (Pérez-Ramos et al., 2021). Although nisin and pediocin PA-1 have
bacteriocins production, which can be divided into Gram-positive, Gram- been commercialized as food additives, other bacteriocins like enterocin

Fig. 1. Characteristics of bacteriocins from different group of microbes.

3
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Fig. 2. Antimicrobial mechanism of bacteriocins alone and combined with other technologies.

AS-48 also have the potential to be used as biopreservatives in food based 3.3. Enterocin and its mode of action
on existing studies. The specific mechanism of above bacteriocins are
thereby introduced to compare the different modes between independently The enterocins produced by different strains of Enterococcus faecium
used bacteriocins and the combination of bacteriocins with other hurdles. have different structures. To be specific, enterocin A, P are Class IIa bacte-
riocins, enterocin 1071 is Class IIb bacteriocin, and enterocin AS-48 is
Class IIc bacteriocin. The E. faecium has the potential to be used as start cul-
3.1. Nisin and its mode of action
tures to provide enterocins and organic acid, enhancing the control of
L. monocytogenes in fermented products. Among enterocins, the action of
Nisin is a Class I bacteriocin produced by Lactococcus lactis used in
enterocin AS-48 has been studied in detail. Enterocin AS-48 takes
cheese manufacture. It has effective antimicrobial activity against a wide
cytoplasmatic membrane as the target, destabilizing the membrane to dissi-
range of Gram-positive foodborne pathogens, while its antimicrobial effect
pate the proton motive force and cause cell death. The simulation results
on Gram-negative bacteria is small. Nisin A was first discovered in 1928
suggested that the protein inserted could organize itself to generate stable
and has been applied to commercial food preservation since 1953 till
pores (Sidhu and Nehra, 2019, 2021), while the supportive experiments
today. Besides, FDA approved to use nisin in pasteurized processed cheese
are absent. The action of enterocins in Class IIa bacteriocin is similar to
spreads established the legal precedent in the U.S. for application of bacte-
pediocin. Enterocins of Class IIb bacteriocin act on the membranes of
riocins as food additives (Chen and Hoover, 2003). Since then several nisin
their targets by rendering them permeable to certain cations such as Na+,
variants (nisin Z, F, P, Q, H, U, U2) have been identified. The difference of
K+, Li+ or H+.
structure between nisin A and Nisin Z is just a single amino acid residue at
position 27, which inducing Nisin Z with a higher rate of diffusion and sol-
ubility under neutral pH conditions (Ellis, 2018). However, the antimicro- 4. Combination of bacteriocin with other hurdles for food preserva-
bial effect does not show significant difference. Nisin act by forming a tion
pore in membranes of target cells, leading to leakage of essential small mol-
ecules at end. The C-terminal region of nisin is responsible for the initial in- Compared with preservation methods that use single bacteriocin, hur-
teraction of nisin with the target membrane (Breukink et al., 1998). Firstly, dle technology is a novel and promising technique that controls food safety
nisin is inserted and partially penetrated into the cytoplasmic membrane of with minimal impact. It improves the antimicrobial effect, reduces the
target bacteria (Breukink and de Kruijff, 2006). Then, nisin binds to amounts of preservations used, saves energy, and expands the shelf life
phosphatidylglycerol (universal receptor) by the C-terminal region (Fig. 3). Therefore, we reviewed studies published over the past two de-
(Breukink and de Kruijff, 2006; Giffard et al., 1997). Followed by the mem- cades to know more about the combined effect of bacteriocins with other
brane insertion, subsequent pore formation results in abrupt cell death hurdles against various microorganisms. According to the origin of the hur-
(Breukink and de Kruijff, 2006; Cotter et al., 2005). dles, they could be divided into physical (Table 2), chemical (Table 3) and
microbial hurdles (Table 4).
3.2. Pediocin and its mode of action
4.1. Combination of bacteriocins and physical hurdles against foodborne patho-
Pediocin is Class IIa bacteriocin produced by Pediococcus pentosaceus, gens and spoilage microorganisms
showing extremely strong inhibition activity against L. monocytogenes
which could grow at 4 °C and cause various foodborne illness. Although 4.1.1. Bacteriocins and heat
pediocin PA-1 has been commercialized in food preservatives, it has not Traditional heat processes are the most commonly used and effective
been legally approved yet to be applied in food preservation worldwide way to control foodborne microorganisms. Traditional heat processes inac-
(Settanni and Corsetti, 2008). Like nisin, pediocin also causes target cell tivate microorganisms via protein unfolding and denaturation (Smelt and
death when forming pores or channels, but in a different pore formation Brul, 2014). In traditional canning methods, extensive heat process was
method. The antimicrobial effect of pediocin is caused by its ability of used to destruct all spores or inhibit the grow of spores below 40 °C during
permeabilizing lipid vesicles, but the author think it only occurres in high the storage. To meet the consumer demands for fresh-like products, milder
pediocin concentrations (Chen et al., 1997a, 1997b; Chikindas et al., 1993). heat treatment is adopted as low or middle temperature would not destruct

4
G. Liu et al. Science of the Total Environment 825 (2022) 154058

membrane. However, the outer membrane recovers rapidly, so does the


nisin resistance (Boziaris and Adams, 2001).
Therefore, the combination of bacteriocins and heat treatment makes
spore, which used to be heat-resistant, become sensitive which saves the en-
ergy. Solving the instant antimicrobial effect of bacteriocins can extend the
shelf life of food and broaden the spectrum of bacteriocins. The combina-
tion enhances action effect because heat could injure the outer membrane
allowing bacteriocins enter the cell membrane.

4.1.2. Bacteriocins and chilling


Chilling treatment generally refers to temperatures above 0 °C and slow
down the growth rate of microorganisms as well as the chemical and bio-
logical processes in foods. Current studies only focus on the antibacterial ef-
fects of the cooling rates alone which indicated that very low (<5 °C/min)
and intermediate cooling rates (180–5000 °C /min) induce obviouse cell in-
juries or death, while low (5–180 °C/min) or very high cooling rates
(>5000 °C/min) may help cell survival (Cao-Hoang et al., 2010). Further-
more, sensitizing Gram-negative bacteria to bacteriocins will enhance the
antibacterial effect of the bacteriocins-and-chilling combination, and the
mechanism of chilling makes nisin-resistant L. monocytogenes sensitive to
nisin is described as followed.
The chilling rate is an important factor influencing the sensitivity of
Fig. 3. Combined effect of bacteriocins with other hurdles for food preservation. Gram-negative bacteria to nisin, but the critical rate of chilling is yet to be
found. Rapid chilling and slow chilling from room temperature to 0.5 °C
in the nisin (50, 500, 2500 IU/mL) showed opposite effects on injury and
the heat resistance of spores and vegetative cells. The combination of bac- inactivation of Gram-negative bacteria (S. enteritidis PT4 and PT7, E. coli
teriocins and heat treatment could increase the heat sensitivity of spore, ex- ATCC 25922, P. aeruginosa USCC 2186 and Pseudomonas fragi ATCC
tend the time of antimicrobial effect and broaden the spectrum of 4973), but no effect is observed in the combination of slow chilling. How-
antibacterial activity. ever, rapid chilling shows a dose-dependent increase in lethality with
The combination of bacteriocins and heat treatment could alter the ther- nisin. P. aeruginosa USCC 2186 is the most sensitive in combinations, show-
mal resistance of bacterial spores. Clostridium sporogenes ATCC 7955 spores ing a 1.5–2.5 log reduction. Though reductions resulting from the chilling-
were rapidly inactivated when being treated in 16 μg/mL nisin at 90 °C. In nisin combination is less than the reduction in broth experiments, adding
contrast, when it is only treated at 90 °C, no spores were inactivated. Ac- EDTA to the combination reduces P. aeruginosa USCC 2186 populations
cordingly, percentage of dipicolinic acid (DPA) which contributes to ther- by 1.3 log CFU (Boziaris and Adams, 2000). As for E. coli TG, it is reported
mal resistance as a component of spores release was higher when spores that slow chilling (2 °C/ min, reached 0 °C) does not induce transient sus-
were treated in nisin at 90 °C, relative to a treatment of 90 °C alone ceptibility to nisin. However, E. coli TG in both exponential and stationary
(Hofstetter et al., 2013a, 2013b). Moreover, the divergent effects of nisin growth phases are sensitive to nisin if there is nisin during the rapid chilling
on spore membrane fluidity provide a rationale for their divergent effects (2000 °C/ min, reached 0 °C) or nisin is added a few minutes after the treat-
on heat-induced spore inactivation and DPA release (Hofstetter et al., ment (Cao-Hoang et al., 2008). Subsequently, under an obvious synergistic
2013a, 2013b). Similarly, adding nisin (2000 IU/mL) to skim milk before effect, exponentially growing E. coli TG in 100 IU/mL nisin under rapid
heat processing (103 °C) enhances the control of spores of the mesophile chilling shows a reduction of more than 6 logs, while the stationary grow-
B. cereus T and the thermophile Bacillus stearothermophilus ATCC 12980. ing E. coli TG in 1000 IU/mL nisin with rapid chilling shows a 2 logs reduc-
Nisin enhances the spore sensitivity to heat and the presence of residual tion (Cao-Hoang et al., 2008). Similarly, the survived P. aeruginosa, S.
nisin inhibits the spore survival and growth (Wandling et al., 1999). Enteritidis USCC2186, S. Enteritidis PT4 and PT7 are not sensitive to
The combination of bacteriocins and heat processing could stop the re- nisin that is added a few minutes after the rapid chilling (7.3 °C/ min,
growth of foodborne pathogens and spoilage bacteriocins, thereby extend- reached 0.5 °C) (Boziaris and Adams, 2001).
ing the shelf-life of food product. Ananou et al. observed that enterocin AS- Apart from changing Gram-negative bacteria’ sensitivity to nisin, the
48 (40,60μg/g) alone significantly reduces the viable counts of combination of chilling and bacteriocins make nisin-resistant Gram-
L. monocytogenes CECT 4032, but L. monocytogenes CECT 4032 regrows dur- positive bacteria sensitive to nisin. After long-time low temperature (10
ing the 60 days storage period of cooked ham at 5/15 °C. Meanwhile, °C) treatment, L. monocytogenes Scott A, the nisin-resistant strain, becomes
S. aureus CECT 976 in cooked ham is slightly sensitive to enterocin AS-48 sensitive to nisin due to the modified membrane fluidity. Cell membranes
and might be out of control at 15 °C with 40μg/g enterocin AS-48. While of L. monocytogenes Scott A do adopt a lower gel-fluid phase transition tem-
sublethal heat (60 °C, 2 min) remarkably increased enterocin AS-48 activity perature to maintain sufficient membrane fluidity at 10 °C. Although the
against S. aureus CECT 976 and L. monocytogenes CECT 4032 in cooked ham lower temperature (10 °C) significantly increased the membrane rigidity
(Ananou et al., 2010a). of the lipid vesicles, L. monocytogenes Scott A is still sensitive to nisin after
In addition, proper heat treatment combined bacteriocins would solve a long-term low temperature treatment (Li et al., 2002). In another study,
the problem of narrow spectrum of antibacterial activity. P. aeruginosa when wild-type cells are switched to a lower temperature (4 °C), the
and the two salmonellae are not sensitive to nisin, while heating at 55 °C order of the fatty acids makes the membrane more rigid, reducing nisin
produce transient sensitivity to nisin. Particularly, these strains are only Z's action. Meanwhile, the K+ efflux rates also decreased with droping tem-
sensitive if nisin is present during heating. These strains would normally peratures (Abee et al., 1994). In conclusion, low temperature could make
survive heating in an injured state, while being inactivated by nisin during the membrane more rigid, negatively affecting the nisin's action, while
heating. Furthermore, the increase in hydrophobicity, lipopolysaccharides the sensitivity of target cells to nisin remains unchanged because the anti-
release and permeability can be detected in the heating treatment, indicat- microbial activity is subject to multiple factors. However, the study mainly
ing that heating can produce transient injury to the outer membrane perme- focuses on nisin and chilling instead of researching the antimicrobial effect
ability barrier, so nisin gains a temporary access to the cytoplasmic of other bacteriocins with chilling treatment.

5
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Table 2
Strategic usage of the combination of bacteriocins and physical hurdles.
Type Combination Food Positive effect Target microorganisms Reference
system
bacteriocins other hurdles

C. sporogenes ATCC 7955, Hofstetter


Nisin Heat – Inactivate endospores C. beijerinckii ATCC 8260, et al., 2013a,
C. difficile 3195 2013b
B. cereus T, Wandling
Nisin Heat Skim milk Reduce D values of spores
Heat + B. stearothermophilus ATCC 12980 et al., 1999
bacteriocins Cooked Avoid regrowth of food S. aureus CECT 976, Ananou et al.,
Enterocin AS-48 Sublethal heat
ham pathogens during 60 days L. monocytogenes CECT 4032 2010a,2010b
Show synergistic inhibitory L. innocua ATCC 51742,
White Al-Holy et al.,
Nisin Sublethal heat activity and avoid regrowth of L. innocua ATCC 33090,
chesses 2012
food pathogens L. innocua ATCC 33091
S. enteritidis PT4 and PT7,
Show enhancement of E. coli ATCC 25922, Boziaris and
Nisin Rapid chilling –
antimicrobial effect P. aeruginosa USCC 2186, Adams, 2001
Chilling + P. fragi ATCC 4973
bacteriocins Show synergistic inhibitory
Nisin Rapid chilling – E. coli TG Bi et al., 2018
effect
Long-term treatment with Sensitize nisin-resistant cell to
Nisin – L. monocytogenes Scott A Li et al., 2002
low temperature nisin
Show synergistic inhibitory Pol et al.,
Nisin PEF Skim milk B. cereus IFR-NL94–25
effect 2001
Plused electric Show obviously synergistic Sobrino-Lopez
Enterocin AS-48 + nisin PEF Milk S. aureus CECT 240
fields (PEF) + inhibitory effect et al., 2009
bacteriocins Fresh
Shear stress-moderate Show synergistic inhibitory E. coli K12, Mok et al.,
Nisin apple-kale
electric field effect L. innocua 2020
blend juice
Expand the antibacterial Rodriguez
Nisin HPP Cheese E. coli O157:H7
spectrum of nisin et al., 2005
Expand the antibacterial
High pressure carbon
Nisin – spectrum of nisin and show E. coli O157:H7 NCTC 12900 Bi et al., 2018
dioxide
high synergistic effect
E. coli parent strain MG1655 and
Reduce pressure-resistant E. García-Graells
Nisin HPP Milk pressure-resistant mutants LMM1010,
Coli et al., 1999
LMM1020 and LMM1030
Orange Eliminate the HHP-resistance Alpas and
High-pressure Nisin HPP S. aureus 485
juice of S. Aureus 485 Bozoglu, 2000
processing
Grade A
(HPP) + Nisin Reduce the HHP-resistance of Alpas and
HPP pasteurized S. aureus 485
bacteriocins Pediocin PA-1 S. Aureus 485 Bozoglu, 2000
milk
Low-acid Show synergistic inhibitory S. aureus,
Ananou et al.,
Enterocin AS-48 HPP fermented activity and don't accept L. monocytogenes,
2010a, 2010b
sausage growth of LAB S. Typhimurium
L. monocytogenes CTC1010,
Cooked Avoid regrowth of food Jofré et al.,
Nisin HPP L. monocytogenes CTC1011,
ham pathogens during 3 months 2008
L. monocytogenes CTC1034
Avoid regrowth of food Dallagnol
Lactocin AL705 HPP Pork loin L. innocua 7
pathogens during 40 days et al., 2017
Soluble soybean Fresh L. monocytogenes,
Luo et al.,
Nisin polysaccharide-based tomato Extend shelf life B. subtilis,
2019
nanoparticles juice S. aureus
Large
Zohri et al.,
Nisin Chitosan nanoparticles yellow Extend shelf life S. aureus
2010
croaker
E. coli, Vukomanović
Nisin Gold nanoparticles – Reduce MIC of nisin
P. aeruginosa et al., 2017
Nanotechnology
Bacteriocins produced by
+ bacteriocins S. aureus, Sidhu and
Lactobacillus spp. Silver nanoparticles – Enhance antimicrobial effect
S. flexneri Nehra, 2020
Bac4463 and Bac22
Show higher antimicrobial
García-Toledo
Pediocin Liposome – activity, stability and L. innocua AST-062
et al., 2019
controlled release
Dual coated liposome Enhance the stability and
Gomaa et al.,
Microcin MCCJ25 (polymeric network of – protect the bacteriocin from S. Enteritidis
2017
pectin and whey proteins) enzymatic degradation

4.1.3. Bacteriocins and pulsed electric fields transmembrane potential across their membrane when being applied an
Pulsed electric field (PEF) is a commonly used non-thermal method of electrical field is applied (Yogesh, 2016). PEF has been used with bacterio-
liquid food preservation. According to existing studies, it has the potential cins to increase antimicrobial effect against foodborne pathogens
to be an alternative to heat processing. PEF treatment has been proven to (Calderón-Miranda et al., 1999; Sobrino-Lopez et al., 2009; Terebiznik
inactivate microorganisms while better maintaining the original flavor et al., 2002).
and quality. Besides, as the method is performed at low processing temper- The combination of PEF and bacteriocins leads different results. On the
atures which turns out to be energy efficient. Bacterial cells experience a one hand, the combination of nisin and PEF showed slight synergy effect.

6
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Table 3
Strategic usage of the combination of bacteriocins and chemical hurdles.
Classification Hurdle technology Food system Positive outcomes Target Reference
microorganisms
Bacteriocins Other hurdles

L.
Cinnamaldehyde, monocytogenes
Alves et al.,
Nisin carvacrol, eugenol, and Cow milk Show synergistic inhibitory effect ATCC 15313,
2016
thymol S. aureus ATCC
25923
E. coli O157:
H7, Campion et al.,
Nisin variants Carvacrol Apple juice Expand the antibacterial spectrum
C. sakazakii 2017
NCTC 8155
Show synergistic antimicrobial effect, expand Ghrairi and
Enteriocin A Thyme – E. coli O157:H7
the antibacterial spectrum, decrease MIC of EOs Hani, 2015
Show synergistic antimicrobial effect, decrease 14 strains of S.
Nisin Cinnamaldehyde Milk Shi et al., 2017
MIC of EOs aureus
Grapefruit seed extract, Show antibacterial effect and reduce dosage of L.
Nisin Pork loin Yu et al., 2019
cinnamaldehyde nisin and EOs monocytogenes
E. coli O157:
H7,
L.
Solomakos
monocytogenes
Essential oils + Nisin Thyme Minced beef Increase the shelf life of minced beef et al., 2008a,
Scott A,
bacteriocins 2008b
L.
monocytogenes
Lmk
Minced sheep Reduce unacceptable organoleptic properties of Govaris et al.,
Nisin Oregano essential oil S. Enteritidis
meat EOs 2010
Cooked Show enhanced antimicrobial effect and protect L. Zhao et al.,
Nisin Grape seed extract
shrimps color during prolonged shelf life monocytogenes 2020a,2020b
L.
Bologna Churklam
Nisin Carvacrol Show synergistic inhibitory effect monocytogenes
sausages et al., 2020
10403S
L.
monocytogenes
ATCC 15313,
L.
Grape fruit seed extract Show antibacterial effect and reduce MIC of
Nisin Lettuce monocytogenes Yu et al., 2019
and cinnamaldehyde nisin and EOs
H7962,
L.
monocytogenes
NADC 2045
Frozen
Bacteriocins form L. curvatus Castellano
EDTA ground-beef Expand the antibacterial spectrum E. coli O157:H7
EDTA + CRL705 and L. lactis CRL1109 et al., 2011
patties
bacteriocins
EDTA and Field et al.,
Nisin A – Expand the antibacterial spectrum E. coli
cinnamaldehyde 2017
S. aureus ATCC
29213,
Zhao et al.,
Nisin Citric acid Milk Enhance antimicrobial effect L.
2017
monocytogenes
Organic acid +
ATCC 19115
bacteriocins
Infant Campion et al.,
Nisaplin® Citric acid Enhance antimicrobial effect C. sakazakii
formula 2017
Enhance antimicrobial effect and no color Ajingi et al.,
Nisin Formic acid Potato B. subtilis
affected 2020
Sodium lactate, sodium L. Lungu and
Nisin Turkey breast Enhance antimicrobial effect
diacetate monocytogenes Johnson, 2005
Cold-smoked Enhance antimicrobial effect and reduce MIC of L. Neetoo et al.,
Nisin Sodium lactate
salmon pate nisin and sodium lactate monocytogenes 2008
L.
monocytogenes
CTC1010,
Salts of organic acid
L.
+ bacteriocins Jofré et al.,
Nisin Sodium lactate Cooked ham Show synergistic inhibitory effect monocytogenes
2007
CTC1011,
L.
monocytogenes
CTC1034
Cold-smoked Enhance antimicrobial effect and extend shelf L. Nykänen et al.,
Nisin Sodium lactate
rainbow trout life monocytogenes 2000
L.
Arqués et al.,
Animial-drived Nisin Lactoperoxidase system Skim milk Avoid regrowth of food pathogens monocytogenes
200
enzymes + Scott A
bacteriocins Infant Oshima et al.,
Nisin or lacticin 3147 Lactoperoxidase system Inhibit outgrowth of C. Sakazakii C. sakazakii
formula 2012

(continued on next page)

7
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Table 3 (continued)

Classification Hurdle technology Food system Positive outcomes Target Reference


microorganisms
Bacteriocins Other hurdles

lactic acid Chung and


Nisin Lysozyme Pork loin Prolong the shelf life to 6 weeks
bacteria Hancock, 2000

The addition of nisin (0.04 mg/mL) individually reduces 1.0 log unit of 5.2 log CFU/mL respectively. Nisin (100 IU/mL) treatment individually in-
B. cereus IFR-NL94–25 and the use of PEF treatment (16.7 kV/cm, 20 pulses activates E. coli K12 and L. innocua by 1.0 and 2.6 log CFU/mL, respectively.
of 2 ms duration, 10 min) alone cause about 0.7 log unit, while the combi- The combination of shear stress-moderate electric field and nisin reduces
nation of nisin and PEF leads to a reduction of 1.9 log unit (Pol et al., 2001). E. coli K12 and L. innocua by 5 log units in 5 min, which is less effective
Under individual shear stress-moderate electric field treatment for 10 min, than the method that uses shear stress-moderate electric field before adding
maximum inactivating levels of E. coli K12 and Listeria innocua are 5.4 and nisin as a post-treatment in 0– 10 min (Mok et al., 2020). On the other hand,

Table 4
Strategic usage of the combination of bacteriocins and microbial hurdles.
Type Hurdle technology Food system Positive outcomes Target Reference
microorganisms
Bacteriocins Other hurdles

Nisin Reuterin Raw milk Enhance antimicrobial effect E. coli K12,


L. monocytogenes
ATCC 13932,
E. faecalis NCDC
135,
Serratia marcescens
ATCC 13880,
S. Typhi NCDC Kumar et al.,
Pediocin Reuterin Raw milk Enhance antimicrobial effect 113, 2020
Yersinia
enterocolitica ATCC
130715,
Klebsiella
pneumoniae NCDC
138,
P. acidilactici LB42
Microbial secondary
S. aureus LMGT
metabolites +
3242,
bacteriocins Chi and Holo,
Nisin+garvicin KS Farnesol – Show synergistic antimicrobial effect A. baumannii
2018
B1162,
E. coli LMGT 3704
A. baumannii
B1162,
Show synergistic antimicrobial effect and Chi and Holo,
Garvicin KS Polymyxin B – A. iwoffii B1163,
expend the antibacterial spectrum 2018
A. calcoaceticus
B1165
Show synergistic antimicrobial effect and L. innocua HPB13, Naghmouchi
Nisin Polymyxin B –
expend the antibacterial spectrum E. coli RR1 et al., 2010
S. Newport,
Surface layer S. aureus ATCC Prado-Acosta
Nisin – Show synergistic antimicrobial effect
protein 6538, et al., 2010
B. cereus 6A1
Surface layer Show synergistic antimicrobial effect and
Nisin Chicken meat S. saprophyticus P2 Sun et al., 2017
protein extended the shelf life of chicken meat
S. aureus
ATCC25923, Sheoran and
Enterocin LD3 Plantaricin LD4 – Show synergistic antimicrobial effect
S. Typhimurium Tiwari, 2021
Bacteriocins + ATCC13311
bacteriocins S. aureus LMGT Chi and Holo,
Garvicin KS Nisin – Show synergistic antimicrobial effect
3242 2018
Producer of nisn A Show synergistic antimicrobial effect and L. innocua
Plantaricin Cheese Mills et al., 2017
and lacticin 3147 more resistant to bacteriophage attack DPC6578
Bacteriocin from S.
Phage P4 and A3 – Show synergistic antimicrobial effect C. perfringens Heo et al., 2018
Hyointestinalis B19
Smoked salmon, hake L. monocytogenes Baños et al.,
Enterocin AS-48 Phage P100 Avoid regrowth of food pathogens
and salmon fillets CECT 4032 2016
Show synergistic antimicrobial effect and L. monocytogenes Rodríguez-Rubio
Coagulin C23 Phage FWLLm1 Milk
Bacteriophages + lower rate of resistance development 2000/47 et al., 2015
bacteriocins L. monocytogenes
1/2a,
Phage LM-103 and Fresh-cut melons and L. monocytogenes Leverentz et al.,
Nisin Enhance antimicrobial activity
LMP-102 apples 1/2b, 2003
L. monocytogenes
4b

8
G. Liu et al. Science of the Total Environment 825 (2022) 154058

the combination of enterocin AS-48 (28 AU/mL) and PEF (35 kV/cm, 1200 MPa) (García-Graells et al., 1999). However, whole milk (pH 6.7) showed
μs) did not show any additive or synergistic effect. Despite the addition of protection of both E. coli MG1655 and its pressure-resistant mutants in
enterocin AS-48 (28 AU/mL), nisin (20 IU/mL) individually caused no var- the HPP treatment (300,400,500,600,700 Mpa for 15 min). Based on
iation in S. aureus CECT 240's survival in milk at its natural pH and pH 5.0 of that, in the test of antibacterial effect on the above strains when using
storage at 4 or 22 °C. Moreover PEF (35 kV/cm) treatment in milk at its nat- nisin/HPP used individually and in combination in milk with different con-
ural pH and pH 5.0 for 1200 μs resulted in 3.5 log reductions of S. aureus centration of fat (3.6%, 1.55%, 0.05% fat), it was suggested that the combi-
CECT 240. However, nisin (20 IU/mL) with enterocin AS-48 (28 AU/mL) nation of nisin (400 IU/mL) and HPP (600Mpa) destructed more target
and PEF (35 kV/cm) treatment at pH 6.8 in milk reduced S. aureus CECT microorganism which was positive correlated to fat content of the milk.
24 by 6.3 log units (Sobrino-Lopez et al., 2009). Further studies showed that cyclic treatment (3×10 min) always results
in a much higher inhibition effect (>1000 folds) than continuous treatment
4.1.4. Bacteriocins and high-pressure processing (1×30 min) (García-Graells et al., 1999). Similarly, S. aureus 485 could not
High-pressure processing (HPP) is a non-thermal processing technique be detected in orange juice (pH 3.76), but it survives after HPP treatment
that typically uses water as a pressure transmission medium while preserv- (345 MPa, 50 °C, 5 min) in Grade A pasteurized milk (pH 6.65). To elimi-
ing the food quality (such as functions, sensory, and nutrition) to the max- nate the HPP-resistance of S. aureus 485, nisin in combination with a
imum. HPP is commonly used in the food processing, including meats, fruit pediocin PA-1 (total 5000 AU/mL) and HPP treatment (345 MPa, 50 °C, 5
juices, and guacamole. HPP utilizes isostatic pressure (about 100 to 1000 min) significantly reduced the S. aureus 485 population in Grade A pasteur-
MPa) which is equally transmitted into food matrices, leading to microbial ized milk when being storaged at 25 °C (Alpas and Bozoglu, 2000). In an-
membrane damage (Barba et al., 2017; Gayán et al., 2012; Guerrero- other study, the activity of enterocin AS-48 and HPP when being used
Beltrán et al., 2005; Huang et al., 2017). Microorganisms have varing sen- individually and in combination against S. aureus, L. monocytogenes and S.
sitivities to HPP in an order of Gram-negative bacteria > Gram-positive bac- Typhimurium was investigated in low-acid fermented sausage (fuet) during
teria > bacterial spores. The application of HPP method requires a longer ripening and storage. HPP has a modest effect on L. monocytogenes when
processing time or higher pressure to kill vegetative bacteria that adversely being used individually, but the S. Typhimurium and L. monocytogenes pop-
affects food quality and increases cost (Gayán et al., 2012). To improve the ulation dropped dramatically when HPP was unsed in combination with
HPP method, we need to develop new strategies to kill microorganisms. enterocin AS-48 (Ananou et al., 2010b).
Previous studies show that a combination of HPP and other hurdles, such The combination of HPP treatment and bacteriocins could extend shelf
as bacteriocins, can effectively retard the growth of microorganisms while life of food because bacteriocin cause an immediate bactericidal effect, and
preserving high food quality (Komora et al., 2020; Lee et al., 2003; Pérez- when it was used individually, the effect was less remarkable in the long-
Baltar et al., 2019; Rodriguez et al., 2005). In addition, the combination term storage. The vacuum packed cooked ham slice with nisin (800 AU/
can also reduce the population of pressure-resistant microorganism. g) at 6 °C is the least effective against L. monocytogenes (CTC1010,
The combination of HPP treatment and bacteriocins could broaden the CTC1011 and CTC1034) and after 3-months the population is not signifi-
antibacterial spectrum of bacteriocins. The Gram-negative bacteria, E. coli cantly different from the control group. However, the level of
and Pseudomonas fluorescens, are both nisin-resistant, but HPP sensitizes L. monocytogenes sustains under 10 CFU/g in refrigerated storage (6 °C)
E. coli and P. fluorescens to the action of nisin (500 IU/mL). Further studies after the combined treatment of HPP (600 MPa) and nisin (800 AU/g).
found that adding nisin (500 IU/mL) before HPP treatment (500 MPa for 5 Moreover, these treatments only reduce the population of pathogens but
min) has a synergy effect, while only a small number of P. fluorescens cells did not affect the LAB (Jofré et al., 2008). In another study, HPP treatment
became sensitive to nisin and most of cells were still nisin-resistant. In (600 MPa) alone was not enough to prevent the regrowth of L. innocua 7 in
this report, the L. innocua population decreased due to the combination, cured-cooked pork loin at the 4 °C storaging condition. While a complete in-
but it shows no traits of synergy effect (Black et al., 2008). As for another hibition of L. innocua 7 in pork loin after the combined treatment with
Gram-negative bacteria E. coli O157:H7, it was also resistant to nisin. HPP lactocin AL705(105 AU/mL) and HPP (600 MPa), no cells regrew up to
treatment (300Mpa, 10 °C,10 min) alone results in a reduction of E. coli 40-day storage. Furthermore, cell membrane/wall damage are observed
O157:H7 population (1.3 log units). However, the combined effect of in all treatments, while complete cell lysis is only found in binary combina-
bacteriocin-producing LAB and HPP treatments is much higher (3.4 log tion treatments (Dallagnol et al., 2017).
unit reduction) than HPP treatment individually in cheese. The synergistic On the one hand, the combination of HHP treatment and bacteriocins
effects are widely observed in bacteriocin-producing LAB, including lacticin could broaden the antibacterial spectrum of bacteriocins, inactivate the re-
481 producing strain Lactococcus lactis TAB 24, nisin Z producing strain growth of target cells in long storage period. On the other hand, the
L. lactis TAB 26, nisin Z producing strain L. lactis TAB 26, TAB 57 producing pressure-resistance strains starts to reduce after the combination treatment.
strain L. lactis TAB 57, TAB 7 producing strain E. faecium TAB 7, Enterocin I Moreover, the combination did not inhibit the growth of LAB.
producing strain E. faecalis TAB 52 and enterocin AS-48 producing strain
E. faecalis INIA 4 (Rodriguez et al., 2005). These results indicates that 4.1.5. Bacteriocins and nanotechnology
bacteriocin-producing LAB could be added as additives to the starter to con- Nanotechnology is the manipulation of functional materials and struc-
trol foodborne pathogens. When E. coli O157:H7 NCTC 12900 is treated tures on the nanoscale size (with diameters ranging from 1 to <1000 nm).
under high pressure carbon dioxide (HPCD) at 5 MPa for 40– 60 min before The basic element of nanotechnology is nanoparticles which have unique
adding 200 μL nisin (20,000 IU/mL) for 5 min (HPCD→nisin), the counts of physical, chemical and biological properties on nanoscale. Most nanoparti-
E. coli O157:H7 NCTC 12900 are significantly decreases by about 0.25–1.0 cles do not have antimicrobial effect, while some nanoparticles such as sil-
log10 cycles than using HPCD individually (Bi et al., 2018). Similar results ver nanoparticles show a wide range of antimicrobial action (Sulthana and
are found in the report that the treatment of HPP (276 MPa, 15 min) in- Archer, 2021). Various studies have documented the enhanced antimicro-
duces E. coli sensitive to nisin (Kalchayanand et al., 1998). To gain a bial activity of bacteriocins against food spoilage bacteria when combined
much stronger synergistic effect, the combination of HPCD and nisin with conjugated nanoparticles (Sidhu and Nehra, 2019; Sulthana and
could cause an extra 0.5–3 log10 cycles reduction on E. coli than using Archer, 2021; Vukomanović et al., 2017). Additionally, bacteriocins com-
HPCD individually. The difference is reflected by the sequence of HPCD bined with nanoparticles could encounter drawbacks of bacteriocins,
and nisin treatments. These results suggested that it is important to add bac- which include broading the antimicrobial spectrum of bacteriocins, reduc-
teriocins and other hurdles in a designed order. ing the dosage of bacteriocins, protecting bacteriocins from degradation
The combination of HPP treatment and bacteriocins could reduce with proteolytic enzymes and increasing their survival during long-term
pressure-resistant strains. Garcia-Graells et al. observed that pressure- storage (Sulthana and Archer, 2021).
resistant E. coli LMM1010, LMM1020 and LMM1030 reduced in skim The combination of nanoparticles and bacteriocins extends the shelf life
milk when nisin or lysozyme (400 IU/mL) were added prior to HPP (550 by enhancing antimicrobial effect and prevents regrowth of the survival.

9
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Nisin packed in soluble soybean polysaccharide-based nanoparticles could synergistic and broad-spectrum action against the tested spoilage microor-
inhibit the growth of L. monocytogenes, B. subtilis and S. aureu in tomato ganisms. However, enterocin KT2W2G alone did not show antimicrobial
juice, thereby extend the shelf life (Luo et al., 2019). The combination of activity against 18 spoilage microorganisms from spoiled banana peel
chitosan and nisin preserve large yellow croaker better than chitosan indi- (Klebsiella pneumoniae, Serratia marcescens, E. faecalis, L. lactis subsp. lactis,
vidually. Compared with nisin that is used individually, the chitosan nano- K. variicola, Kodamaea ohmeri, Hanseniaspora opuntiae, Pichia kudriavzevii,
particles with nisin showes double antimicrobial effect against S. aureus P. fermentans, and Candida metapsilosis) (Issouffou et al., 2018). The poly-
(Zohri et al., 2010). Besides, the MIC of nisin packed in nanoparticles de- phenolic compounds (cinnamaldehyde, carvacrol, eugenol, and thymol)
creased from 2 mg/mL to 0.5 mg/mL. The combination of nisin and gold and nisin have synergy effect against pathogens L. monocytogenes ATCC
nanoparticles are capable of disintegrating the wall of Gram-negative bacte- 15313 and S. aureus ATCC 25923 in milk (Alves et al., 2016). Besides, the
ria, thereby inhibiting the growth of E. coli and P. aeruginosa (Vukomanović synergistic interaction of EOs and bacteriocins against L. monocytogenes
et al., 2017). Similarly, the combination of bacteriocins (produced by Lacto- can be found in various food, such as sliced bologna sausages (Churklam
bacillus spp. Bac4463 and Bac22) and silver nanoparticles enhances the ac- et al., 2020), whole, low and skim milk (Bajpai et al., 2014), lettuce (Yu
tivity against S. aureus and Shigella flexneri (Sidhu and Nehra, 2020). et al., 2019), raw pork loin (Yu et al., 2019), shrimp (Zhao et al., 2020b).
The combination of nanoparticles and bacteriocins enhance the stability However, the exact action mechanism of EO and bacteriocins against
of nisin which can be further improved by encapsulating nisin in L. monocytogenes has yet been studied in depth. The underlying mechanism
nanoliposomes at 25– 70 °C and in extreme pH conditions (Taylor et al., of grape seed extract and nisin against L. monocytogenes has been elucidated
2007). Meanwhile, nanoliposomes keep nisin to be antimicrobial active through a metabolomics approach based on nuclear magnetic resonance
for a longer time, Teixeira et al. found that bacteriocin-like substance spectroscopy (NMR) (Zhao et al., 2020a). The combination of nisin and
retained its original antibacterial effect for more than 16 days compared grape seed extract potently reduces the L. monocytogenes population (4.49
with free nisin in the same condition (Teixeira et al., 2008). Similar results log CFU/mL). Confocal laser scanning microscopy results reveals that cell
showed that pediocin encapsulated in liposome has higher capability of membrane permeability varies dramatically and the leakage of protein
inhibiting L. innocua AST-062, remaining stability and controlling release and nucleic acid increases. Furthermore, the results of NMR-based metabo-
of pediocin (García-Toledo et al., 2019). Microcin MCCJ25 encapsulated lomics coupled with multivariate analysis showed lower threonine, ade-
in dual coated liposome (polymeric network of pectin and whey proteins) nine, cysteine, adenosine triphosphate (ATP), and nicotinamide adenine
enhanced the stability, realized the gradual release and protectd the bacte- dinucleotide phosphate (NADP), while γ-aminobutyric acid and lactic
riocin from gastrointestinal enzymes in vitro (Gomaa et al., 2017). acid increases are observed in a nisin and grape seed extract treatment.
Pathway analysis indicates that the nisin and grape seed extract combina-
4.2. Combination of bacteriocins and chemical hurdles against foodborne patho- tion inhibites L. monocytogenes by blocking amino acid biosynthesis, the
gens and spoilage microorganisms TCA cycle (citric acid cycle) and the energy-producing pathway. The com-
bination of nisin and grape seed extract was applied in shrimp and it mark-
4.2.1. Bacteriocins and essential oils edly inhibits the L. monocytogenes population (1.79 log CFU/g) (Zhao et al.,
Essential oils (EOs) are volatile secondary metabolites produced by 2020b).
plants, mainly comprising the volatile and aromatic compounds naturally Specifically, the combination of bacteriocins and EOs could broaden the
present all parts of the plants including bark, stem, flowers, seeds, peel antibacterial spectrum. Three EOs (derived from basil, sage, and ajowan)
and whole plants (Sánchez-González et al., 2011). EOs are widely used in showed greater antibacterial activity against S. aureus ATCC 29213 but
various countries as food preservatives and medicine, cosmetics and per- very marginal effects on E. coli ATCC 25922, while the antimicrobial effect
fumes. EOs and their phytoconstituents have been reported to exert a of the three EOs used in combination with nisin shows synergic activities
broad spectrum of biological activities (such as antifungal, insecticidal, an- with FIC of 0.06,0.03,0.06 against E. coli ATCC 25922, respectively. How-
tibacterial, and antiviral) (Fitzgerald, 2003; Schnitzler et al., 2007). Re- ever, all these combinations showed no interactive effect on S. aureus
search delving into the application of EOs as antimicrobial agents has ATCC 29213 (Mehdizadeh et al., 2016). Furthermore, nisin variants (60
increased due to their wide spectrum of activities and the property of μM) used in the combination with carvacrol EOs (0.03%) reduces by 3
being GRAS (generally recognized as safe) with natural origins log units E. coli O157:H7 and 4 log units C. sakazakii NCTC 8155 compared
(Nedorostova et al., 2009). However, high EO level in food showed unac- to nisin A carvacrol treatment. Furthermore, the nisin variants (30 μM) and
ceptable organoleptic properties. If the level of EO decreases to preserve carvacrol EOs (75 μg/mL) causes complete inactivation of E. coli O157:H7
the color of food, antibacterial activity effect will be undermined. It has in apple juice within 3 h at 25 °C compared to that of the equivalent nisin
been documented that EOs combined with other antimicrobial agents, A combination (Campion et al., 2017). As for enteriocin A, when it was
such as bacteriocins, have an enhanced or synergistic effect while both used individually, the E. coli O157:H7 is not inhibited from growth. How-
the EOs and bacteriocins can be used in lower volume (Alves et al., 2016; ever, the addition of thyme EOs (0.71 μg/mL) and enterocin A (3.4
Churklam et al., 2020; Turgis et al., 2012). Moreover, the combination of μg/mL) showed a synergistic antimicrobial effect against E. coli O157:H7,
bacteriocins and EOs could inhibit the regrowth of survival cells and even and MIC of thyme EOs decreases from 2.2 to 0.71 μg/mL (Ghrairi and
expand the antibacterial spectrum. This section will discuss the combined Hani, 2015).
effect of EOs and bacteriocins against food spoilage organisms. In addition, mixed natural preservative can be more cost-effective in
It is widely known that EOs and bacteriocins act in an additive or syner- terms of food preparation due to smaller dose of natural preservative.
gistic manner to inhibit the growth of foodborne pathogens and spoilage Cinnamaldehyde (CA), an essential oil isolated from cinnamon bark, and
bacteria. The antimicrobial potential of pediocin, nisin, and two bacterio- nisin both have antimicrobial activity against 14 strains of S. aureus,
cins produced from E. faecium (MT 104 and MT 162) and six essential oils while the combination of nisin (8 μg/mL) and CA (0.25 mg/mL) not only
(Cymbopogon nardus, Cinnamomum cassia, Satureja montagna, Origanum exhibits synergistic antimicrobial activity against S. aureus in milk, but
vulgare, Thymus vulgaris, and Brassica hirta) against two spoilage bacteria also reduced the does of nisin used by about 8– 16 folds and the use of
(Pseudomonas putida and L. sakei) and five pathogenic bacteria CA by about 5 folds. Furthermore, the combination of nisin and CA resulted
(L. monocytogenes, S. aureus, S. Typhimurium, B. cereus, and E. coli) is re- in about 54.5% cell membrane-damage, which is nearly 2 folds of the out-
ported in a previous study. The results indicated that O. vulgare EO and come of using the nisin or CA treatment individually. Similar results are
nisin have a synergistic effect on the inhibition of L. monocytogenes, whereas found in the damage of cell wall. Severe demages of cell membrane and
nisin combined with T. vulgaris EO had a synergistic effect on the inhibition cell wall have caused the synergistic effect of the combination against
of S. Typhimurium, while S. montagna EO combined with pediocin had a S. aureus (Shi et al., 2017). Enterocin A combined with thyme EOs showed
synergistic effect on the inhibition of E. coli (Turgis et al., 2012). The bactericidal effect of L. monocytogenes EGDe after 18 h treatment. More-
enterocin KT2W2G and cinnamon mix at the ratio of 6:4 displayed a over, the combination could reduce the MIC of enterocin A (4.57 to 0.9

10
G. Liu et al. Science of the Total Environment 825 (2022) 154058

μg/mL) and thyme EOs (3.6 to 1.2 μg/mL) (Ghrairi and Hani, 2015). The carboxylic acid and tartaric acid are the most commonly used chelating
combination of nisin (5 to 6 ppm), grapefruit seed extract (6 to 8 ppm) agents in the food industry. Many studies have reported that using bacterio-
and cinnamaldehyde (15 to 20 ppm) increases the antibacterial effect cins with chelating agents could extend the shelf life of food products
against L. monocytogenes on pork loin. The level of nisin decreases from (Castellano et al., 2011; Liang et al., 2020; Morsy et al., 2018).
250 ppm to 15 ppm, the level of grapefruit seed extract reduces from To study the effect of two bacteriocin-producing strains (L. curvatus
31.25 ppm to 6 ppm, and the level of cinnamaldehyde fell from 500 ppm CRL705 and L. lactis CRL1109) combining with disodium ethylenediamine-
to 15 ppm (Zhao et al., 2020b). tetraacetic acid (Na2EDTA) against E. coli O157:H7, the contaminated fro-
The combination of EOs and bacteriocins weakens the unacceptable or- zen ground-beef patties at 5 °C over 9 days are used. Both bioprotective
ganoleptic properties of EOs. Most of the time, high level EOs showed unac- cultures (ca. 107 CFU/g) combined with EDTA (18 g/kg of beef) greatly
ceptable organoleptic properties in food, while low level EOs possesses a contain the growth of E. coli. However, the treatments alone failed to inhibit
weak antibacterial activity, and the thyme EO (0.9%) showes unacceptable the growth of E. coli. Furthermore, no significant effect on ground-beef pat-
organoleptic properties in minced meat. To reduce the side effect and con- ties color was produced after the treatment of bioprotective cultures, while
trol foodborne pathogens, studies have investigated the effect of thyme EO a darker color observed on patties in the presence of EDTA (Castellano
and nisin used individually or in combination against E. coli O157:H7 and et al., 2011). However, the combination of chrisin (a commercial nisin
L. monocytogenes Scott A and Lmk. The results show that thyme EO preparation) with EDTA was effective against all Gram-positive bacteria
(0.6%) with nisin (1000 IU/g) exerts an additive effect against the patho- tested at a range of concentrations, but the combination was not effective
gens and extends the shelf life of minced beef storaged at 4 °C (Solomakos against all of the Gram-negative bacteria at the concentrations tested. Re-
et al., 2008a, 2008b). Sensory evaluation showed that the adding 0.6 or cent studies reported that nisin with three commercial chelating resins
0.9% of oregano essential oil (OEO) in minced sheep meat is organolepti- (Dowex MAC-3, Chelex 100, and Lewatit TP260) did not show significant
cally acceptable, and OEO is easier to be accepted at 0.6% than at 0.9%. inhabitation on the growth of Alicyclobacillus acidoterrestris ATCC 49025
OEO enriched with carvacrol and thymol at 0.6%-0.9% and nisin (500 or in apple juice (Herskovitz et al., 2019). Such oppsite results indicate the im-
1000 IU/g) used individually and in combination are studied for activity portance of applications testing in real food matrices.
against S. Enteritidis in minced sheep meat stored at 4–10 °C for 12 days. Organic acids (acetic acid, lactic acid, and citric acid etc.) and sodium
The results reveals that OEO or nisin when used alone fails to kill S. salts of the low molecular weight organic acids (sodium lactate, sodium
Enteritidis, but when used in combination, OEO and nisin exhibits more po- diacetate etc.) have been used as chemical additives to control microbial
tent antimicrobial activity against S. Enteritidis. The author suggested that growth, improve sensory attributes and extend the shelf life of various
OEO has strong a potentail to inactivate S. Enteritidis when being used in food systems including meat, poultry and fish (Figueiredo and Almeida,
combination with nisin (Govaris et al., 2010). In another study, the combi- 2017). The combined use of nisin M21A (78 μg/mL), a bioengineered de-
nation of nisin and grape seed extract (1:1) reduces L. monocytogenes in the rivative of nisin A, and citric acid (1170 μg/mL) against L. monocytogenes
cooked shrimp from 6.1 to 4.3 log CFU/g in 15 min. Though the food color F6854 has a much higher effect than the combination of nisin A (78
is slightly affected at the beginning of storage after the treatment with grape μg/mL) and citric acid (1170 μg/mL) (Zhao et al., 2017). In addition, the
seed extract, it provids an additional protection to color from the degrada- combined use of nisin M21A (0.1 μg/mL) with citric acid (175 μg/mL),
tion during prolonged shelf life. Furthermore, atomic force microscopy nisin A (0.1 μg/mL) with citric acid (175 μg/mL) and Nisaplin® (30
analysis reveals greater morphological changes when the components of μg/mL) with citric acid (15 μg/mL) eradicated the biofilms of
the treatment are used in combination (Zhao et al., 2020a). L. monocytogenes F6854 (Smith et al., 2016). Similar results were observed
The activity of bioengineered nisin variants is usually superior over the in the synergistic interaction of nisin (0.25 mg/mL) and citric acid (16
wild type nisin A equivalent when used in combination with a variety of es- mg/mL) against S. aureus ATCC 29213 and L. monocytogenes ATCC 19115
sential oils. In a study investigated the effect of semi-purified nisin A and in milk (Zhao et al., 2017), and the main action targets of the combination
nisin V along with EOs (trans-cinnamaldehyde, carvacrol, and thymol) were cell wall and membrane. Another study shows that the combined use
against L. monocytogenes, it is noticed that the growth curves for combined of Nisaplin® (10 mg/mL) and citric acid (30 mM) significantly reduced the
treatment with nisin V shows a delayed lag phase comparing with nisin A C. sakazakii count in infant formula (Campion et al., 2017). Except citric
used in combination with EOs (Field et al., 2015). The antimicrobial activ- acid, other organic acids (formic, lactic, cirtic, malic, fumaric and tartaric)
ity of more nisin variants (nisin V and nisin S29A) combined with carvacrol combined with nisin have synergistic interaction to pathogens
EOs is higher than the wild type nisin A-EOs treatments (Campion et al., (P. aeruginosa, E. coli, S. Typhimurium, S. aureus, and S. faecalis) (Ajingi
2017). et al., 2020). Meanwhile, in the evaluation of the antimicrobial potential
In summary, the combination of EOs and bacteriocins not only shows a of formic acid in combination with nisin towards B. subtilis on potato, the
wide range of antimicrobial effect and inactivates the regrowth of results showed that the combination of nisin (0.016 mg/mL) and formic
foodborne pathogens, but also reduces the dosage of bacteriocins and acid (0.25%) inhibited B. subtilis counts while the color changed slightly
weakens unacceptable organoleptic properties of EOs. The potential mech- (Ajingi et al., 2020).
anism of the combination is changing cell membrane permeability and The combined antimicrobial activity of sodium lactate and bacteriocins
causes leakage of protein and nucleic acid in target cells. have recorded different results. For example, Figueiredo and Almeida re-
ported that the it was no difference of the combination of nisin and sodium
4.2.2. Bacteriocins and EDTA/organic acids/salts of organic acids lactate (nisin (0.0012 μg/g) and sodium lactate (0.5 μg/g) and individually
Studies on chelating agents are growing as a topic in food processing re- used in inhibiting L. monocytogenes on ready-to-eat sliced pork ham
search because the agents are able to bind metal ions and stabilize food. (Figueiredo and Almeida, 2017). Similar results were observed by Lung
Chelating agents help bacteriocins to kill Gram-negative pathogens. The cy- and Johnson, who applied nisin (640 AU/mL), sodium lactate (0.06
toplasmic membrane of Gram-negative bacteria is protected by an outer g/mL) and sodium diacetate (0.06 g/mL) in combination against
membrane (OM) composed of a phospholipid bilayer. The lipopolysaccha- L. monocytogenes in turkey breast (Lungu and Johnson, 2005). On the
ride of Gram-negative bacteria layer forms a tight shield barricading bacte- contray, some studies reported that the combination of nisin and sodium
riocins while the divalent cations, particularly Mg2+ and Ca2+ stabilize the lactate significantly decreased the L. monocytogenes count in cooked ham,
anionic lipopolysaccharide layer. If these cations are removed, lipopolysac- cold-smoked salmon pate and cold-smoked rainbow trout (Jofré et al.,
charide molecules will be released from the outer membrane, and the 2007; Neetoo et al., 2008; Nykänen et al., 2000). Since the nisin and potas-
Gram-negative cells will be sensitive to bacteriocins (Martin-Visscher sium sorbate combination exerted a bacteriostatic effect on
et al., 2011). Sequestrants form chelate complexes with polyvalent metal B. sporothermodurans in milk (Aouadhi et al., 2015), the combination of
ions, which can serve a variety of advantages in assuring food safety and nisin, potassium sorbate and lactocin AL705 coult perform potent anti-
quality (Bohrer, 2019). Ethylenediaminetetraacetic acid (EDTA), L. innocua activity (Verdi et al., 2020).

11
G. Liu et al. Science of the Total Environment 825 (2022) 154058

To sum up, the combination of chelating agents and bacteriocins re- depolarization showed that surface disruption of cells could cause superior
moved lipopolysaccharide of Gram-negative cell membrane to make effect of the combination.
Gram-negative bacteria sensitize to bacteriocins. The combination of or- Above all, the combination of animal-derived enzymes and bacteriocins
ganic acid and bacteriocins shows synergistic antimicrobial effect, extend- would change the spectrum of antimicrobial activities. In addition, the an-
ing the shelf life, however, bacteriocins combined with salt of organic timicrobial activities were affected by nutrient condition because proteins
acid showed opposite effects on antibacterial. or fats protect foodborne pathogens.

4.2.3. Bacteriocins and animal-derived enzymes 4.3. Combination of bacteriocins and microbial hurdles against foodborne path-
Lactoperoxidase (EC 1.11.1.7) (LP) is a member of the peroxidase fam- ogens and spoilage microorganisms
ily, a group of natural enzymes in bovine milk. LP can catalyse the oxidation
of certain molecules using H2O2 to generate reactive product with a wide 4.3.1. Bacteriocins and microbial secondary metabolites/other bacteriocins
antimicrobial activity. LP-system (LPS), consisting of LP, SCN− and H2O2, Reuterin (β-hydroxypropionaldehyde) is a small organic compound bio-
is used as natural biopreservatives in food. LPS can kill Gram-negative bac- synthetically produced by L. reuteri during anaerobic fermentation of glyc-
teria such as pseudomonads, coliforms, salmonellae and shigellae, but only erol. It possesses a broad spectrum of antimicrobial activities against
inhibit Gram-positive bacteria like streptococci and lactobacilli foodborne pathogens and spoilage microorganisms. The combination of
(Kussendrager and van Hooijdonk, 2000). Studies have reported that bacte- reuterin (8 AU/mL) and nisin (100 IU/mL) showed a significant synergistic
riocins induce an rapid but not lasting bactericidal effect, while the com- effect on L. monocytogenes and a slight additive effect on S. aureus after 24 h
bined LPS with nisin will contain the regrowth of L. monocytogenes in in milk at 37 °C (Arqués et al., 2004). In another similar study, the combi-
skim milk at 25 or 30 °C (Boussouel et al., 2000; Zapico et al., 1998). In nation of reuterin (150 AU/mL) and nisin (100 IU/mL) and the combina-
comparison, LPS combined with reuterin (2 AU/mL) is less effective than tion of reuterin (100 AU/mL) and pediocin (2185 AU/mL) reduced
that combined with nisin (100 IU/mL) (Arqués et al., 2008). However, coliform counts by 2.2 log CFU/mL and 1.5 log CFU/mL in raw milk within
when strains produced with nisin (L. lactis ATCC 11454, L. lactis ESI 515) 3 h at 37 °C, respectively (Kumar et al., 2020). It was reported that the com-
and strains produced with enterocin 4 (E. faecalis INIA 4) were used in com- bination of reuterin (8 AU/mL) with nisin (100 IU/mL) did not enhance
bination with LPS in raw milk at 4 °C or 8 °C, the counts of L. monocytogenes the antimicrobial effect of reuterin during the storage of UHT skimmed
was reduced significantly (Rodríguez et al., 1997). In addition, the anti- milk at 4 or 8 °C, although the combination effectively killed Campylobacter
Listeria effect of nisin and LSP was compared between being added simulta- jejuni LMG 6629 and Aeromonas hydrophila subsp. hydrophila CECT 839 in
neously and in two steps. The results showed that the effect of nisin and LPS milk.
added in sequence was greater than the effect of being added simulta- Farnesol, a cheap and harmless compound, has been considered a prom-
neously, while the antimicrobial activity depended on the treating time of ising adjuvant for antibiotics. When being used in combination with nisin
first inhibitor and target microorganism. When nisin (200 IU/mL) and and garvicin KS, it showed a synergistic effect against S. aureus LMGT
L. monocytogenes has been interacted for 4 h than LPS was added, it showed 3242, A. baumannii B1162 and E. coli LMGT 3704 (Chi and Holo, 2018).
a bactericidal effect within 48 h and which lasted for 15 days. However, the Polymyxin B is a drug with strong inhibitory effect on Gram-negative bac-
longer treatment of nisin and L. monocytogenes firstly (12h) would lead to teria, but it could be toxic at certain concentrations. Synergistic effect was
re-grow of L. monocytogenes after 200 h of incubation. When LPS added observed in the combination of polymyxin B and garvicin KS, which inhibit
firstly then nisin was added 24 h later, no viable L. monocytogenes cells A. baumannii B1162, A. iwoffii B1163 and A. calcoaceticus B1165. Moreover,
would be detectable within 10 days. When nisin was added 72 h after the combination reduced the resisitance of A. iwoffii B1163 to polymyxin B.
LPS, counts of L. monocytogenes reduced by 8 log units in milk compared Similar synergistic antibacterial effects were observed when nisin
with the control (Boussouel et al., 2000). In another study, when LPS was combinated with polymyxin B (Chi and Holo, 2018). In addition, the com-
added after 3 h and nisin after 5 h, counts of L. monocytogenes decreased bination significantly reduced the MIC of polymyxin B and removed the
by 4.5 log in 24 h (Zapico et al., 1998). However, the adding nisin and toxicity to human. Besides, the usage of nisin considerably decreased in
LPS simultaneously or in two steps do not affect the inhibitory effect of the combination of polymyxin B on the effect of anti-Listeria
E. coli, Acinetobacter, Flavobacterium indologenes and Psychrobacter immobilis (Naghmouchi et al., 2010).
strains in fish (Elotmani and Assobhei, 2004). As for the combined effect The surface layer (S-layer) of Lactobacillus acidophilus ATCC 4356 has an
of bacteriocins (nisin or lacticin 3147) and LPS, C. sakazakii in rehydrated endopeptidase activity against cell wall of Gram-negative bacteria, but it
infant formula was inhibited outgrowth at 37 °C for 8 h (Oshima et al., did not affect Gram-positive bacteria. When S-layer is used with nisin, it
2012). showed synergistically inhibitory effect on the growth of S. enterica serovar
Hen egg-white lysozyme has been the only lysozyme allowed in the Newport, S. aureus ATCC 6538 and B. cereus 6A1. The S-layer causes pepti-
food industry. It can cleave the β-(1,4)-glycosidic bond between N- doglycan of cell wall hydrolysis, thus enhancing the antimicrobial activity
acetylmuramic acid and N-acetylglucosamine of bacterial cell wall peptido- of nisin by enabling it to cross the cell wall then acting on the cell mem-
glycans, leading to the antimicrobial efficiency of Gram-positive bacteria. brane (Prado-Acosta et al., 2010). Another surface layer protein, SlpB, iso-
However, when being used individually, lysozyme exhibits weak inhibitory lated from Lactobacillus crispatus, also showed enhancement to the
effects against Gram-negative bacteria (Wu et al., 2019). A combination of antimicrobial activity of nisin. The combination of nisin and SlpB show syn-
lysozyme and nisin (3:1,w/w) whose concentration is approximately 260 ergistic antibacterial effect against S. saprophyticus and extends the shelf life
μg/cm2 at the surface of vacuum packaged pork loins is effective in of chicken meat. However, the mechanism of SlpB/nisin is different from
inhibiting the growth of lactic acid bacteria at 2 °C for up to 6 weeks the combination of S-layer and nisin. Cell lysis has not been observed
(Nattress, 2003). Chung and Hancock found that the combination of lyso- after any treatment of SlpB/nisin, while membrane damage and structural
zyme and nisin is active against LAB (Chung and Hancock, 2000), while disorganization are observed, enhancing nisin access to cell membrane,
no such interactions were observed in study of Gill and Holley (Gill and which could cause the dramatic release of micro-molecules from the cell
Holley, 2003), so the action is probably because the two studies used differ- (Sun et al., 2017).
ent growth media (MRS and ATP). Moreover, the antimicrobial effect of ly- Additionally, the combination of different bacteriocins is a cost-efficient
sozyme and nisin was enhanced when MRS was diluted to 1/8 (a nutrient- method for food industry by reducing MIC of bacteriocins. The MIC of
deficient environment), while there was no enhanced bactericidal effect in enterocin LD3 in combination was decreased by 8 folds against S. aureus
a pork juice medium under the same conditions (Chung and Hancock, ATCC25923 and S. Typhimurium ATCC13311 and the MIC of plantaricin
2000). Therefore, the nutrient condition of evaluating model is important LD4 in combination reduced by 3 folds against the above target strains.
influencing factors on control food spoilage microorganism. The results of The results of transmission electron microscopy and FTIR spectra showed
scanning electron microscopy and the increase of optical density and cell that the higher disruption of cell membrane in the combined bacteriocin-

12
G. Liu et al. Science of the Total Environment 825 (2022) 154058

treated cells as compared with alone effects, and the two bacteriocins have concentration. Moreover, synergistic effect usually occurs when one tech-
different mode of action which caused synergistic activity against target nology addresses the barriers of another technology while the latter one
cells (Sheoran and Tiwari, 2021). It was reported that garvicin KS and has different mode of action. The combination of bacteriocins and hurdles
nisin acted synergistically against S. aureus, reducing the strains counts extends shelf life of food as the combination increases the stability of bacte-
(Chi and Holo, 2018). Mills et al. constructed a double-bacteriocin- riocins and inhibit the regrowth of food pathogens and spolige bacteria.
producing starters of L. lactis which produced nisin A and lacticin 3147 sta- More importantly, the two catagories of combination of bacteriocins
bly, although the antimicrobial activity was as effective as its single- and other hurdles could broaden the spectrum of antimicrobial activity.
producing strains, while double-bacteriocin-producing strain was more re- The first is the combination of bacteriocins and food preservaties with a
sistant to bacteriophage attack. Interestingly, the double producer com- wide range antimicrobial activities such as EOs, the other is the combina-
bined with the plantaricin producer as start culture in cheese exhibited a tion of bacteriocins and treatment which destroy cell membrane of Gram-
considerable reduction in L. innocua DPC6578 (Mills et al., 2017). negative bacteria such as EDTA. In addition, different adding sequence of
Therefore, the combination of microbial secondary metabolites hurdles and evaluation system will lead to opposite antimicrobial effect.
(reuterin, farnesol, polymyxin B and surface layer protein) and bacteriocins The determination of appropriate adding sequence is lack of adequate
enhanced the antimicrobial effect and reduced dosage in use, thus lowering research, so future investigations will need to be undertaken. There is a con-
the cost of food preservation. In addition, the bacteriocins of different ac- sensus that the evaluation system is an important factor in food safety re-
tion mode will easily produce synergistic effects. search. Furthur work is required to provide greater insight into the effect
of the adverse interactions between bacteriocins and food ingredients.
4.3.2. Bacteriocins and bacteriophages
Bacteriophages or phages are viruses that infect and replicate within the
bacteria, causing host lysis and cell death. These natural killers of bacteria 6. Future perspectives
are widely distributed around us from sewage plants, silage to food process-
ing environments. Assessment has proved that these viruses can be utilized The early results show that the combination of bacteriocins and other
as new tools to reduce levels of foodborne pathogens and spoilage bacteria. food preservation techniques have the potential to be applied in food indus-
For example, the first phage-based product (ListShield™) was approved to try. However, the governing principles of bacteriocins and other hurdles are
control L. monocytogenes in meat and poultry products (Bren, 2007). Until absent and most strategic usages of hurdle technologies have been merely
now, there is no evidence that phages may harm humans or animals blind attempts. The mechanism of the combination is pending deeper inves-
(Endersen et al., 2014). However, bacteriophages or bacteriocins individu- tigation. It shows that the combination of different preservation methods
ally did not contain the regrow of foodborne pathogens. To inhibit the re- may lead to synergistic antimicrobial effect as one technique may mititage
growth of pathogens, the combined bacteriocins with bacteriophages the adverse impact caused by another method, thus enhancing the action.
were used in food. Moreover, the complicated real food matrix protects foodborne pathogens
Although treatment with bacteriocin or phage alone results in rapid an- and spoilage bacteria by reducing the efficiency of bacteriocins. Under-
timicrobial effects against C. perfringens, the survivals of C. perfringens re- standing the interactions and mechanisms will be helpful to find more pre-
grow during the storge. However, when bacteriocin from S. hyointestinalis cise and optimal utilization of bacteriocins in food, while proteomic,
B19 combined with P4 and A3 phages, it resulted in synergistic effect and genomic and transcriptomic tools could facilitate the discovery of underly-
eradicate C. perfringens completely (Heo et al., 2018). Similar results show ing mechanisms. In addition, it is necessary to have experiments in real
that the phage P100 (2.3 × 107 PFU/cm2) and enterocin AS-48 (0.37 food production, since the antimicrobial activity in broth and media gener-
μg/cm2) against eliminate L. monocytogenes CECT 4032 from hake and ally need less dose for preservation. As for target microorganism, there have
salmon fillets from 2 and 1 days, respectively. The treatment of P100/AS- been sufficient studies on foodborne pathogens like L. monocytogenes, while
48 reduce L. monocytogenes CECT 4032 below detection levels from 1 to food spoilage bacteria, a common and main reason for food spoilage, need
15 days in smoked salmon (Baños et al., 2016). In another study, the Listeria further research. In addition to food safety, future research with more focus
phage FWLLm1 (MOI of 100) in combination with bacteriocin coagulin on nutritions and sensory properties of food is therefore suggested. More-
C23 (584 AU/mL) was more effective than treatment alone for the over, more research is needed to better understand the effect on the taste
inactivating or reducing L. monocytogenes counts in milk at 4 °C and texture of food products. Indeed, when applying hurdle technology, se-
(Rodríguez-Rubio et al., 2015). Moreover, study has shown that the combi- quence of addition should be considered because a minor change in the
nation of phages and the bacteriocin nisin enhanced antimicrobial activity adding sequence of bacteriocins and other hurdles would lead to a hugely
by against L. monocytogenes in artificially contaminated fresh-cut melons different antimicrobial activity. Additionally, current research suffers
and apples (Leverentz et al., 2003) and against S. aureus in pasteurized from insufficient studies on the combination of bacteriocin-producing
milk (Martínez et al., 2008), proving that this approach is feasible in food strain and hurdle. However, adding bacteriocin-producing strain directly
biopreservation (Rodríguez-Rubio et al., 2015). can make the isolation and purification process more cost-efficient. There-
As the results show, the combination of bacteriophages and bacteriocins fore, it is of crucial importance to study the fundamental mechanism behind
has enhanced the antimicrobial effect against various food spoilage micro- the hurdle technology which involves bacteriocins. Based on that, it will be
organisms. Due to practical constraints, this paper cannot provide a com- possible to design an effective, environment friendly and practical food
prehensive review of mechanism behind the possible synergy of safety control hurdle concept based strategies in the future.
bacteriocins with bacteriophages. Future studies on this topic is are there-
fore recommended.
CRediT authorship contribution statement
5. Conclusion
No conflict of interest exits in the submission of this manuscript, and
Though bacteriocins as antimicrobial peptides have planty of advan- manuscript is approved by all authors for publication. I would like to de-
tages, there are several drawbacks when bacteriocins is used as clare on behalf of my co-authors that the work described was original re-
biopreservatives individually for the food industries. The combination of search that has not been published previously, and not under
bacterioncins and other hurdles provide an attractive option for maintain- consideration for publication elsewhere, in whole or in part. All the authors
ing food quality and food safety. Briefly, bacteriocins and other hurdles listed have approved the manuscript that is enclosed.
showed synergistic or additive antimicrobial effect with a lower dosage, re- Guorong Liu: Writing- Original draft preparation, Writing- Reviewing
ducing economic cost of food preservation, while mitigating the advese ef- and Editing. Rong Nie: Conceptualization, Data curation. Yangshuo Liu:
fect such as toxicity or unacceptal organoleptic properties caused by high Data curation, Formal analysis. Arshad Mehmood: Conceptualization.

13
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Funding Black, E.P., Linton, M., McCall, R.D., Curran, W., Fitzgerald, G.F., Kelly, A.L., Patterson, M.F.,
2008. The combined effects of high pressure and nisin on germination and inactivation of
bacillus spores in milk. J. Appl. Microbiol. 105, 78–87. https://doi.org/10.1111/j.1365-
This project was funded by National Natural Science Foundation of 2672.2007.03722.x.
China (No. 31871772; No. 31671832); National Key R & D program of Bohrer, B.M., 2019. Sequestrants as a food ingredient. Encyclopedia of Food Chemistry.
Elsevier, pp. 251–255 https://doi.org/10.1016/B978-0-08-100596-5.21610-7.
China (2019YFC1606200); Beijing Natural Science Foundation (Grant Boussouel, N., Mathieu, F., Revol-Junelles, A.-M., Millière, J.-B., 2000. Effects of combinations
No. 6192003); School Level Cultivation Fund of Beijing Technology and of lactoperoxidase system and nisin on the behaviour of listeria monocytogenes ATCC
Business University for Distinguished and Excellent Young Scholars 15313 in skim milk. Int. J. Food Microbiol. 61, 169–175. https://doi.org/10.1016/
S0168-1605(00)00365-2.
(BTBUYP2022); 2021 Postgraduate Scientific Research Ability Improve-
Boziaris, I.S., Adams, M.R., 2000. Transient sensitivity to nisin in cold-shocked gram nega-
ment Project of Beijing Technology and Business University, and Graduate tives. Lett. Appl. Microbiol. 31, 233–237. https://doi.org/10.1046/j.1365-2672.2000.
Innovation Project of School of Food and Health in 2021. 00804.x.
Boziaris, I.S., Adams, M.R., 2001. Temperature shock, injury and transient sensitivity to nisin
in gram negatives. J. Appl. Microbiol. 91, 715–724. https://doi.org/10.1046/j.1365-
2672.2001.01433.x.
Declaration of competing interest Bren, L., 2007. Bacteria-eating virus approved as food additive. FDA Consum. 41, 20–22.
Breukink, E., de Kruijff, B., 2006. Lipid II as a target for antibiotics. Nat. Rev. Drug Discov. 5,
The authors declare that they have no known competing financial inter- 321–323. https://doi.org/10.1038/nrd2004.
Breukink, E., van Kraaij, C., van Dalen, A., Demel, R.A., Siezen, R.J., de Kruijff, B., Kuipers,
ests or personal relationships that could have appeared to influence the
O.P., 1998. The orientation of nisin in membranes. Biochemistry 37, 8153–8162.
work reported in this paper. https://doi.org/10.1021/bi972797l.
Calderón-Miranda, M.L., Barbosa-Cánovas, G.V., Swanson, B.G., 1999. Inactivation of listeria
References innocua in skim milk by pulsed electric fields and nisin. Int. J. Food Microbiol. 51, 19–30.
https://doi.org/10.1016/S0168-1605(99)00069-0.
Campion, A., Morrissey, R., Field, D., Cotter, P.D., Hill, C., Ross, R.P., 2017. Use of enhanced
Abee, T., Rombouts, F.M., Hugenholtz, J., Guihard, G., Letellier, L., 1994. Mode of action of nisin derivatives in combination with food-grade oils or citric acid to control cronobacter
nisin Z against listeria monocytogenes Scott a grown at high and low temperatures. sakazakii and Escherichia coli O157:H7. Food Microbiol. 65, 254–263. https://doi.org/
Appl. Environ. Microbiol. 60, 1962–1968. https://doi.org/10.1128/aem.60.6.1962- 10.1016/j.fm.2017.01.020.
1968.1994. Cao-Hoang, L., Marechal, P.A., Le-Thanh, M., Gervais, P., 2008. Synergistic action of rapid
Ajingi, Y.S., Ruengvisesh, S., Khunrae, P., Rattanarojpong, T., Jongruja, N., 2020. The com- chilling and nisin on the inactivation of Escherichia coli. Appl. Microbiol. Biotechnol.
bined effect of formic acid and nisin on potato spoilage. Biocatal. Agric. Biotechnol. 24, 79, 105–109. https://doi.org/10.1007/s00253-008-1402-9.
101523. https://doi.org/10.1016/j.bcab.2020.101523. Cao-Hoang, L., Dumont, F., Marechal, P.A., Gervais, P., 2010. Inactivation of Escherichia coli
Al-Holy, M.A., Al-Nabulsi, A., Osaili, T.M., Ayyash, M.M., Shaker, R.R., 2012. Inactivation of and lactobacillus plantarum in relation to membrane permeabilization due to rapid chill-
listeria innocua in brined white cheese by a combination of nisin and heat. Food Control ing followed by cold storage. Arch. Microbiol. 192, 299–305. https://doi.org/10.1007/
23, 48–53. https://doi.org/10.1016/j.foodcont.2011.06.009. s00203-010-0555-y.
Alpas, H., Bozoglu, F., 2000. The combined effect of high hydrostatic pressure, heat and bac- Castellano, P., Belfiore, C., Vignolo, G., 2011. Combination of bioprotective cultures with
teriocins on inactivation of foodborne pathogens in milk and orange juice. World EDTA to reduce Escherichia coli O157:H7 in frozen ground-beef patties. Food Control
J. Microbiol. Biotechnol. 16, 387–392. https://doi.org/10.1023/A:1008936607413. 22, 1461–1465. https://doi.org/10.1016/j.foodcont.2011.02.018.
Alves, F.C.B., Barbosa, L.N., Andrade, B.F.M.T., Albano, M., Furtado, F.B., Marques Pereira, Chalón, M.C., Acuña, L., Morero, R.D., Minahk, C.J., Bellomio, A., 2012. Membrane-active
A.F., Rall, V.L.M., Júnior, A.F., 2016a. Short communication: inhibitory activities of the bacteriocins to control salmonella in foods. Food Res. Int. 45, 735–744. https://doi.
lantibiotic nisin combined with phenolic compounds against Staphylococcus aureus org/10.1016/j.foodres.2011.08.024.
and listeria monocytogenes in cow milk. J. Dairy Sci. 99, 1831–1836. https://doi.org/ Chen, H., Hoover, D.G., 2003. Bacteriocins and their food applications. Compr. Rev. Food Sci.
10.3168/jds.2015-10025. Food Saf. 2, 82–100. https://doi.org/10.1111/j.1541-4337.2003.tb00016.x.
Ananou, S., Baños, A., Maqueda, M., Martínez-Bueno, M., Gálvez, A., Valdivia, E., 2010a. Ef- Chen, Y., Ludescher, R.D., Montville, T.J., 1997a. Electrostatic interactions, but not the
fect of combined physico-chemical treatments based on enterocin AS-48 on the control of YGNGV consensus motif, govern the binding of pediocin PA-1 and its fragments to phos-
listeria monocytogenes and Staphylococcus aureus in a model cooked ham. Food Control pholipid vesicles. Appl. Environ. Microbiol. 63, 4770–4777. https://doi.org/10.1128/
21, 478–486. https://doi.org/10.1016/j.foodcont.2009.07.010. aem.63.12.4770-4777.1997.
Ananou, S., Garriga, M., Jofré, A., Aymerich, T., Gálvez, A., Maqueda, M., Martínez-Bueno, Chen, Y., Shapira, R., Eisenstein, M., Montville, T.J., 1997b. Functional characterization of
M., Valdivia, E., 2010b. Combined effect of enterocin AS-48 and high hydrostatic pres- pediocin PA-1 binding to liposomes in the absence of a protein receptor and its relation-
sure to control food-borne pathogens inoculated in low acid fermented sausages. Meat ship to a predicted tertiary structure. Appl. Environ. Microbiol. 63, 524–531. https://doi.
Sci. 84, 594–600. https://doi.org/10.1016/j.meatsci.2009.10.017. org/10.1128/aem.63.2.524-531.1997.
Aouadhi, C., Mejri, S., Maaroufi, A., 2015. Inhibitory effects of nisin and potassium sorbate Chi, H., Holo, H., 2018. Synergistic antimicrobial activity between the broad Spectrum bacte-
alone or in combination on vegetative cells growth and spore germination of bacillus riocin garvicin KS and nisin, farnesol and polymyxin B against gram-positive and gram-
sporothermodurans in milk. Food Microbiol. 46, 40–45. https://doi.org/10.1016/j.fm. negative bacteria. Curr. Microbiol. 75, 272–277. https://doi.org/10.1007/s00284-017-
2014.07.004. 1375-y.
Aras, S., Kabir, M.N., Chowdhury, S., Fouladkhah, A.C., 2020. Augmenting the pressure-based Chikindas, M.L., Ledeboer, A.M., Nissen-Meyer, J., Nes, I.F., Abee, T., Konings, W.N.,
pasteurization of listeria monocytogenes by synergism with nisin and mild heat. IJERPH Venema’, G., 1993. Pediocin PA-1, a bacteriocin from Pediococcus acidilactici PACl.O,
17, 563. https://doi.org/10.3390/ijerph17020563. forms hydrophilic pores in the cytoplasmic membrane of target cells. Appl. Environ.
Arqués, J.L., Fernández, J., Gaya, P., Nuñez, M., Medina, M., Rodrı́guez, E., 2004. Antimicro- Microbiol. 59, 8.
bial activity of reuterin in combination with nisin against food-borne pathogens. Int. Chung, W., Hancock, R.E.W., 2000. Action of lysozyme and nisin mixtures against lactic acid
J. Food Microbiol. 95, 225–229. https://doi.org/10.1016/j.ijfoodmicro.2004.03.009. bacteria. Int. J. Food Microbiol. 60, 25–32. https://doi.org/10.1016/S0168-1605(00)
Arqués, Juan L., Rodríguez, E., Nuñez, M., Medina, M., 2008. Inactivation of gram-negative 00330-5.
pathogens in refrigerated milk by reuterin in combination with nisin or the Churklam, W., Chaturongakul, S., Ngamwongsatit, B., Aunpad, R., 2020. The mechanisms of
lactoperoxidase system. Eur. Food Res. Technol. 227, 77–82. https://doi.org/10.1007/ action of carvacrol and its synergism with nisin against listeria monocytogenes on sliced
s00217-007-0695-8. bologna sausage. Food Control 108, 106864. https://doi.org/10.1016/j.foodcont.2019.
Arqués, J.L., Rodríguez, E., Nuñez, M., Medina, M., 2008. Antimicrobial activity of nisin, 106864.
reuterin, and the lactoperoxidase system on listeria monocytogenes and Staphylococcus Cotter, P.D., Hill, C., Ross, R.P., 2005. Bacteriocins: developing innate immunity for food. Nat.
aureus in cuajada, a semisolid dairy product manufactured in Spain. J. Dairy Sci. 91, Rev. Microbiol. 3, 777–788. https://doi.org/10.1038/nrmicro1273.
70–75. https://doi.org/10.3168/jds.2007-0133. Cox, C., Coburn, P., Gilmore, M., 2005. Enterococcal cytolysin: a novel two component pep-
Ay, H., Tuncer, N., 2016. Combined antimicrobial effect of nisin, carvacrol and EDTA against tide system that serves as a bacterial defense against eukaryotic and prokaryotic cells.
salmonella typhimurium in TSBYE at 4°C and 37°C. Rom. Biotechnol. Lett. 21, 9. CPPS 6, 77–84. https://doi.org/10.2174/1389203053027557.
Bajpai, V.K., Yoon, J.I., Bhardwaj, M., Kang, S.C., 2014. Anti-listerial synergism of leaf essen- Dallagnol, A.M., Barrio, Y., Cap, M., Szerman, N., Castellano, P., Vaudagna, S.R., Vignolo, G.,
tial oil of Metasequoia glyptostroboides with nisin in whole, low and skim milks. Asian 2017. Listeria inactivation by the combination of high hydrostatic pressure and lactocin
Pac J Trop Med 7, 602–608. https://doi.org/10.1016/S1995-7645(14)60102-4. AL705 on cured-cooked pork loin slices. Food Bioprocess Technol. 10, 1824–1833.
Baños, A., García-López, J.D., Núñez, C., Martínez-Bueno, M., Maqueda, M., Valdivia, E., https://doi.org/10.1007/s11947-017-1956-6.
2016. Biocontrol of listeria monocytogenes in fish by enterocin AS-48 and listeria lytic Drider, D., Fimland, G., Héchard, Y., McMullen, L.M., Prévost, H., 2006. The continuing story
bacteriophage P100. LWT Food Sci. Technol. 66, 672–677. https://doi.org/10.1016/j. of class IIa bacteriocins. Microbiol. Mol. Biol. Rev. 70, 564–582. https://doi.org/10.
lwt.2015.11.025. 1128/MMBR.00016-05.
Barba, F.J., Koubaa, M., do Prado-Silva, L., Orlien, V., Sant’Ana, A.de S., 2017. Mild process- Du, H., Li, X., Lu, Z., Bie, X., Zhao, H., Zhang, C., Lu, F., 2019. Effect of plantaricin 163 in com-
ing applied to the inactivation of the main foodborne bacterial pathogens: a review. bination with thymol and surfactin on crucian carp (Carassius auratus). J. Food Prot. 82,
Trends Food Sci. Technol. 66, 20–35. https://doi.org/10.1016/j.tifs.2017.05.011. 1283–1291. https://doi.org/10.4315/0362-028X.JFP-18-439.
Bi, X., Wang, Y., Hu, X., Liao, X., 2018. Decreased resistance of sublethally injured Escherichia Duc, H.M., Son, H.M., Ngan, P.H., Sato, J., Masuda, Y., Honjoh, K., Miyamoto, T., 2020. Iso-
coli O157:H7 to salt, mild heat, nisin and acids induced by high pressure carbon dioxide. lation and application of bacteriophages alone or in combination with nisin against
Int. J. Food Microbiol. 269, 137–143. https://doi.org/10.1016/j.ijfoodmicro.2018.01. planktonic and biofilm cells of Staphylococcus aureus. Appl. Microbiol. Biotechnol.
027. 104, 5145–5158. https://doi.org/10.1007/s00253-020-10581-4.

14
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Ellis, J.-C., 2018. Biomedical applications for bacteriocins in infection and oncology. PhD Johnson, E.M., Jung, Dr.Y.-G., Jin, Dr.Y.-Y., Jayabalan, Dr.R., Yang, Dr.S.H., Suh, J.W., 2018.
ThesisUniversity College Cork. http://hdl.handle.net/10468/8082. Bacteriocins as food preservatives: challenges and emerging horizons. Crit. Rev. Food Sci.
Elotmani, F., Assobhei, O., 2004. In vitro inhibition of microbial flora of fish by nisin and Nutr. 58, 2743–2767. https://doi.org/10.1080/10408398.2017.1340870.
lactoperoxidase system. Lett. Appl. Microbiol. 38, 60–65. https://doi.org/10.1046/j. Kalchayanand, N., Sikes, A., Dunne, C.P., Ray, B., 1998. Factors influencing death and injury
1472-765X.2003.01441.x. of foodborne pathogens by hydrostatic pressure-pasteurization. Food Microbiol. 15,
Elsser-Gravesen, D., Elsser-Gravesen, A., 2013. Biopreservatives. In: Zorn, H., Czermak, P. 207–214. https://doi.org/10.1006/fmic.1997.0155.
(Eds.), Biotechnology of Food and Feed Additives, Advances in Biochemical Engineer- Komora, N., Maciel, C., Pinto, C.A., Ferreira, V., Brandão, T.R.S., Saraiva, J.M.A., Castro, S.M.,
ing/Biotechnology. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 29–49 https:// Teixeira, P., 2020. Non-thermal approach to listeria monocytogenes inactivation in milk:
doi.org/10.1007/10_2013_234. the combined effect of high pressure, pediocin PA-1 and bacteriophage P100. Food
Endersen, L., O’Mahony, J., Hill, C., Ross, R.P., McAuliffe, O., Coffey, A., 2014. Phage therapy Microbiol. 86, 103315. https://doi.org/10.1016/j.fm.2019.103315.
in the food industry. Annu. Rev. Food Sci. Technol. 5, 327–349. https://doi.org/10. Kruszewska, D., Sahl, H.-G., Bierbaum, G., Pag, U., Hynes, S.O., Ljungh, Å., 2004.
1146/annurev-food-030713-092415. Mersacidin eradicates methicillin-resistant Staphylococcus aureus (MRSA) in a
Field, D., Baghou, I., Rea, M., Gardiner, G., Ross, R., Hill, C., 2017. Nisin in combination with mouse rhinitis model. J. Antimicrob. Chemother. 54, 648–653. https://doi.org/10.
cinnamaldehyde and EDTA to control growth of escherichia coli strains of swine origin. 1093/jac/dkh387.
Antibiotics 6, 35. https://doi.org/10.3390/antibiotics6040035. Kumar, N., Kumar, V., Waheed, S.M., Pradhan, D., 2020. Efficacy of reuterin and bacteriocins
Field, D., Daly, K., O’Connor, P.M., Cotter, P.D., Hill, C., Ross, R.P., 2015. Efficacies of nisin a nisin and pediocin on preservation of raw Milk procured from dairy farms. Food Technol.
and nisin V semipurified preparations alone and in combination with plant essential oils Biotechnol. (Online) 58. https://doi.org/10.17113/ftb.58.04.20.6728.
for controlling listeria monocytogenes. Appl. Environ. Microbiol. 81, 2762–2769. https:// Kussendrager, K.D., van Hooijdonk, A.C.M., 2000. Lactoperoxidase: physico-chemical proper-
doi.org/10.1128/AEM.00070-15. ties, occurrence, mechanism of action and applications. Br. J. Nutr. 84, 19–25. https://
Figueiredo, A.C.L., Almeida, R.C.C., 2017. Antibacterial efficacy of nisin, bacteriophage P100 doi.org/10.1017/S0007114500002208.
and sodium lactate against listeria monocytogenes in ready-to-eat sliced pork ham. Braz. Lee, D.U., Heinz, V., Knorr, D., 2003. Effects of combination treatments of nisin and high-
J. Microbiol. 48, 724–729. https://doi.org/10.1016/j.bjm.2017.02.010. intensity ultrasound with high pressure on the microbial inactivation in liquid whole
Fitzgerald, D., 2003. Analysis of the inhibition of food spoilage yeasts by vanillin. Int. J. Food egg. Innovative Food Sci. Emerg. Technol. 4, 387–393. https://doi.org/10.1016/S1466-
Microbiol. 86, 113–122. https://doi.org/10.1016/S0168-1605(03)00059-X. 8564(03)00039-0.
Gálvez, A., Abriouel, H., López, R.L., Omar, N.B., 2007. Bacteriocin-based strategies for food bio- Leistner, L., 2000. Basic aspects of food preservation by hurdle technology. Int. J. Food
preservation. Int. J. Food Microbiol. 120, 51–70. https://doi.org/10.1016/j.ijfoodmicro. Microbiol. 55, 181–186. https://doi.org/10.1016/S0168-1605(00)00161-6.
2007.06.001. Leverentz, B., Conway, W.S., Camp, M.J., Janisiewicz, W.J., Abuladze, T., Yang, M., Saftner,
García-Graells, C., Masschalck, B., Michiels, C.W., 1999. Inactivation of Escherichia coli in R., Sulakvelidze, A., 2003. Biocontrol of Listeria monocytogenes on fresh-cut produce by
Milk by high-hydrostatic-pressure treatment in combination with antimicrobial peptides. treatment with lytic bacteriophages and a bacteriocin. Appl Environ Microbiol 69,
J. Food Prot. 62, 1248–1254. https://doi.org/10.4315/0362-028X-62.11.1248. 4519–4526. https://doi.org/10.1128/AEM.69.8.4519-4526.2003.
García-Toledo, J.A., Torrestiana-Sánchez, B., Martínez-Sánchez, C.E., Tejero-Andrade, J.M., Li, J., Chikindas, M.L., Ludescher, R.D., Montville, T.J., 2002. Temperature- and surfactant-
García-Bórquez, A., Mendoza-García, P.G., 2019. Nanoencapsulation of a bacteriocin induced membrane modifications that Alter listeria monocytogenes nisin sensitivity by
from pediococcus acidilactici ITV26 by microfluidization. Food Bioprocess Technol. 12, different mechanisms. Appl. Environ. Microbiol. 68, 5904–5910. https://doi.org/10.
88–97. https://doi.org/10.1007/s11947-018-2184-4. 1128/AEM.68.12.5904-5910.2002.
Gayán, E., Torres, J.A., Paredes-Sabja, D., 2012. Hurdle approach to increase the microbial in- Liang, Z., Hsiao, H., Jhang, D., 2020. Synergistic antibacterial effect of nisin, ethylenediamine-
activation by high pressure processing: effect of essential oils. Food Eng. Rev. 4, 141–148. tetraacetic acid, and sulfite on native microflora of fresh white shrimp during ice storage.
https://doi.org/10.1007/s12393-012-9055-y. J. Food Saf. 40. https://doi.org/10.1111/jfs.12794.
Ghrairi, T., Hani, K., 2015. Enhanced bactericidal effect of enterocin a in combination with Lopez, F.E., Vincent, P.A., Zenoff, A.M., Salomón, R.A., Farías, R.N., 2007. Efficacy of microcin
thyme essential oils against L. Monocytogenes and E. Coli O157:H7. J. Food Sci. Technol. J25 in biomatrices and in a mouse model of salmonella infection. J. Antimicrob.
52, 2148–2156. https://doi.org/10.1007/s13197-013-1214-5. Chemother. 59, 676–680. https://doi.org/10.1093/jac/dkm009.
Giffard, C.J., Dodd, H.M., Horn, N., Ladha, S., Mackie, A.R., Parr, A., Gasson, M.J., Lungu, B., Johnson, M.G., 2005. Fate of listeria monocytogenes inoculated onto the surface of
Sanders, D., 1997. Structure−function relations of variant and fragment nisins stud- model Turkey frankfurter pieces treated with zein coatings containing nisin, sodium
ied with model membrane systems. Biochemistry 36, 3802–3810. https://doi.org/ diacetate, and sodium lactate at 4°C. J. Food Prot. 68, 855–859. https://doi.org/10.
10.1021/bi962506t. 4315/0362-028X-68.4.855.
Gill, A.O., Holley, R.A., 2003. Interactive inhibition of meat spoilage and pathogenic Luo, L., Wu, Y., Liu, C., Huang, L., Zou, Y., Shen, Y., Lin, Q., 2019. Designing soluble soybean
bacteria by lysozyme, nisin and EDTA in the presence of nitrite and sodium chloride polysaccharides-based nanoparticles to improve sustained antimicrobial activity of nisin.
at 24 °C. Int. J. Food Microbiol. 80, 251–259. https://doi.org/10.1016/S0168-1605 Carbohydr. Polym. 225, 115251. https://doi.org/10.1016/j.carbpol.2019.115251.
(02)00171-X. Manea, L., Buruleanu, L., Rustad, T., Manea, I., Barascu, E., 2017. Overview on the microbio-
Gomaa, A.I., Martinent, C., Hammami, R., Fliss, I., Subirade, M., 2017. Dual coating of lipo- logical quality of some meat products with impact on the food safety and health of peo-
somes as encapsulating matrix of antimicrobial peptides: development and characteriza- ple. 2017 E-Health and Bioengineering Conference (EHB). Presented at the 2017 E-
tion. Front. Chem. 5, 103. https://doi.org/10.3389/fchem.2017.00103. Health and Bioengineering Conference (EHB). IEEE, Sinaia, Romania, pp. 105–108
Govaris, A., Solomakos, N., Pexara, A., Chatzopoulou, P.S., 2010. The antimicrobial effect of https://doi.org/10.1109/EHB.2017.7995372.
oregano essential oil, nisin and their combination against salmonella enteritidis in Martínez, B., Obeso, J.M., Rodríguez, A., García, P., 2008. Nisin-bacteriophage crossresistance
minced sheep meat during refrigerated storage. Int. J. Food Microbiol. 137, 175–180. in Staphylococcus aureus. Int. J. Food Microbiol. 122, 253–258. https://doi.org/10.
https://doi.org/10.1016/j.ijfoodmicro.2009.12.017. 1016/j.ijfoodmicro.2008.01.011.
Guerrero-Beltrán, J.A., Barbosa-Cánovas, G.V., Swanson, B.G., 2005. High hydrostatic pres- Martin-Visscher, L.A., Yoganathan, S., Sit, C.S., Lohans, C.T., Vederas, J.C., 2011. The activity
sure processing of fruit and vegetable products. Food Rev. Int. 21, 411–425. https:// of bacteriocins from carnobacterium maltaromaticum UAL307 against gram-negative
doi.org/10.1080/87559120500224827. bacteria in combination with EDTA treatment: extending the activity of LAB and non-
Güllüce, M., Karaday, M., 2013. Bacteriocins: Promising Natural Antimicrobials, p. 13. LAB bacteriocins. FEMS Microbiol. Lett. 317, 152–159. https://doi.org/10.1111/j.
Heo, S., Kim, M.G., Kwon, M., Lee, H.S., Kim, G.-B., 2018. Inhibition of Clostridium 1574-6968.2011.02223.x.
perfringens using bacteriophages and bacteriocin producing strains. Korean J. Food Sci. Mehdizadeh, T., Hashemzadeh, M.S., Nazarizadeh, A., Neyriz-Naghadehi, M., Tat, M.,
Anim. Resour. 38, 88–98. https://doi.org/10.5851/kosfa.2018.38.1.88. Dorostkar, R., 2016. Chemical Composition and Antibacterial Properties of Ocimum
Herskovitz, J.E., Worobo, R.W., Goddard, J.M., 2019. The role of solid support bound metal basilicum, Salvia officinalis and Trachyspermum ammi Essential Oils Alone and in Com-
chelators on system-dependent synergy and antagonism with nisin. J. Food Sci. 84, bination With Nisin, p. 8.
580–589. https://doi.org/10.1111/1750-3841.14444. Millette, M., Cornut, G., Dupont, C., Shareck, F., Archambault, D., Lacroix, M., 2008. Capacity
Hofstetter, Simmon, Gebhardt, D., Ho, L., Gänzle, M., McMullen, L.M., 2013. Effects of nisin of human nisin- and pediocin-producing lactic acid bacteria to reduce intestinal coloniza-
and reutericyclin on resistance of endospores of clostridium spp. To heat and high pres- tion by vancomycin-resistant enterococci. Appl. Environ. Microbiol. 74, 1997–2003.
sure. Food Microbiol. 34, 46–51. https://doi.org/10.1016/j.fm.2012.11.001. https://doi.org/10.1128/AEM.02150-07.
Hofstetter, S., Winter, R., McMullen, L.M., Gänzle, M.G., 2013. In situ determination of clos- Mills, S., Griffin, C., O’Connor, P.M., Serrano, L.M., Meijer, W.C., Hill, C., Ross, R.P., 2017. A
tridium endospore membrane fluidity during pressure-assisted thermal processing in multibacteriocin cheese starter system, comprising nisin and lacticin 3147 in lactococcus
combination with nisin or reutericyclin. Appl. Environ. Microbiol. 79, 2103–2106. lactis, in combination with plantaricin from lactobacillus plantarum. Appl. Environ.
https://doi.org/10.1128/AEM.03755-12. Microbiol. 83. https://doi.org/10.1128/AEM.00799-17.
Huang, H.-W., Wu, S.-J., Lu, J.-K., Shyu, Y.-T., Wang, C.-Y., 2017. Current status and future Mok, J.H., Pyatkovskyy, T., Yousef, A., Sastry, S.K., 2020. Synergistic effects of shear stress,
trends of high-pressure processing in food industry. Food Control 72, 1–8. https://doi. moderate electric field, and nisin for the inactivation of Escherichia coli K12 and listeria
org/10.1016/j.foodcont.2016.07.019. innocua in clear apple juice. Food Control 113, 107209. https://doi.org/10.1016/j.
Issouffou, C., Suwansri, S., Salaipeth, L., Domig, K.J., Hwanhlem, N., 2018. Synergistic effect foodcont.2020.107209.
of essential oils and enterocin KT2W2G on the growth of spoilage microorganisms iso- Morsy, M.K., Elsabagh, R., Trinetta, V., 2018. Evaluation of novel synergistic antimicrobial ac-
lated from spoiled banana peel. Food Control 89, 260–269. https://doi.org/10.1016/j. tivity of nisin, lysozyme, EDTA nanoparticles, and/or ZnO nanoparticles to control
foodcont.2018.02.019. foodborne pathogens on minced beef. Food Control 92, 249–254. https://doi.org/10.
Jofré, A., Garriga, M., Aymerich, T., 2007. Inhibition of listeria monocytogenes in cooked ham 1016/j.foodcont.2018.04.061.
through active packaging with natural antimicrobials and high-pressure processing. Naghmouchi, K., Drider, D., Baah, J., Teather, R., 2010. Nisin a and polymyxin B as synergistic
J. Food Prot. 70, 2498–2502. https://doi.org/10.4315/0362-028X-70.11.2498. inhibitors of gram-positive and gram-negative bacteria. Probiotics Antimicro. Prot. 2,
Jofré, A., Garriga, M., Aymerich, T., 2008. Inhibition of salmonella sp. Listeria monocytogenes 98–103. https://doi.org/10.1007/s12602-009-9033-8.
and Staphylococcus aureus in cooked ham by combining antimicrobials, high hydrostatic Nattress, F., 2003. Effects of treatment with lysozyme and nisin on the microflora and sensory
pressure and refrigeration. Meat Sci. 78, 53–59. https://doi.org/10.1016/j.meatsci.2007. properties of commercial pork. Int. J. Food Microbiol. 85, 259–267. https://doi.org/10.
06.015. 1016/S0168-1605(02)00545-7.

15
G. Liu et al. Science of the Total Environment 825 (2022) 154058

Nedorostova, L., Kloucek, P., Kokoska, L., Stolcova, M., Pulkrabek, J., 2009. Antimicrobial enhanced antimicrobial spectrum against food-borne pathogens. LWT 139, 110546.
properties of selected essential oils in vapour phase against foodborne bacteria. Food https://doi.org/10.1016/j.lwt.2020.110546.
Control 20, 157–160. https://doi.org/10.1016/j.foodcont.2008.03.007. Smelt, J.P.P.M., Brul, S., 2014. Thermal inactivation of microorganisms. Crit. Rev. Food Sci.
Neetoo, H., Ye, M., Chen, H., 2008. Potential antimicrobials to control listeria monocytogenes Nutr. 54, 1371–1385. https://doi.org/10.1080/10408398.2011.637645.
in vacuum-packaged cold-smoked salmon pâté and fillets. Int. J. Food Microbiol. 123, Smith, M.K., Draper, L.A., Hazelhoff, P.-J., Cotter, P.D., Ross, R.P., Hill, C., 2016. A bioengi-
220–227. https://doi.org/10.1016/j.ijfoodmicro.2008.02.001. neered nisin derivative, M21A, in combination with food grade additives eradicates
Nykänen, A., Weckman, K., Lapveteläinen, A., 2000. Synergistic inhibition of listeria biofilms of listeria monocytogenes. Front. Microbiol. 7. https://doi.org/10.3389/fmicb.
monocytogenes on cold-smoked rainbow trout by nisin and sodium lactate. Int. J. Food 2016.01939.
Microbiol. 61, 63–72. https://doi.org/10.1016/S0168-1605(00)00368-8. Sobrino-Lopez, A., Viedma-Martínez, P., Abriouel, H., Valdivia, E., Gálvez, A., Martin-Belloso,
O’Connor, E., Shand, R., 2002. Halocins and sulfolobicins: the emerging story of archaeal pro- O., 2009. The effect of adding antimicrobial peptides to milk inoculated with Staphylo-
tein and peptide antibiotics. J. Ind. Microbiol. Biotechnol. 28, 23–31. https://doi.org/10. coccus aureus and processed by high-intensity pulsed-electric field. J. Dairy Sci. 92,
1038/sj/jim/7000190. 2514–2523. https://doi.org/10.3168/jds.2008-1996.
O’Connor, P.M., Kuniyoshi, T.M., Oliveira, R.P., Hill, C., Ross, R.P., Cotter, P.D., 2020. Antimi- Solomakos, N., Govaris, A., Koidis, P., Botsoglou, N., 2008a. The antimicrobial effect of thyme
crobials for food and feed; a bacteriocin perspective. Curr. Opin. Biotechnol. 61, essential oil, nisin, and their combination against listeria monocytogenes in minced beef
160–167. https://doi.org/10.1016/j.copbio.2019.12.023. during refrigerated storage. Food Microbiol. 25, 120–127. https://doi.org/10.1016/j.fm.
Oppegård, C., Rogne, P., Emanuelsen, L., Kristiansen, P.E., Fimland, G., Nissen-Meyer, J., 2007.07.002.
2007. The two-peptide class II bacteriocins: structure, production, and mode of action. Solomakos, N., Govaris, A., Koidis, P., Botsoglou, N., 2008b. The antimicrobial effect of thyme
J. Mol. Microbiol. Biotechnol. 13, 210–219. https://doi.org/10.1159/000104750. essential oil, nisin and their combination against Escherichia coli O157:H7 in minced
Oshima, S., Rea, M.C., Lothe, S., Morgan, S., Begley, M., O’Connor, P.M., Fitzsimmons, A., beef during refrigerated storage. Meat Sci. 80, 159–166. https://doi.org/10.1016/j.
Kamikado, H., Walton, R., Ross, R.P., Hill, C., 2012. Efficacy of organic acids, bacterio- meatsci.2007.11.014.
cins, and the lactoperoxidase system in inhibiting the growth of cronobacter spp. In rehy- Sulthana, R., Archer, A.C., 2021a. Bacteriocin nanoconjugates: boon to medical and food in-
drated infant formula. J. Food Prot. 75, 1734–1742. https://doi.org/10.4315/0362- dustry. J. Appl. Microbiol. 131, 1056–1071. https://doi.org/10.1111/jam.14982.
028X.JFP-12-066. Sun, Z., Li, P., Liu, F., Bian, H., Wang, D., Wang, X., Zou, Y., Sun, C., Xu, W., 2017. Synergistic
Parada, J.L., Caron, C.R., Medeiros, A.B.P., Soccol, C.R., 2007. Bacteriocins from lactic acid antibacterial mechanism of the lactobacillus crispatus surface layer protein and nisin on
bacteria: purification, properties and use as biopreservatives. Braz. Arch. Biol. Technol. staphylococcus saprophyticus. Sci. Rep. 7, 265. https://doi.org/10.1038/s41598-017-
50, 512–542. https://doi.org/10.1590/S1516-89132007000300018. 00303-8.
Pérez-Baltar, A., Serrano, A., Bravo, D., Montiel, R., Medina, M., 2019. Combined effect of Taylor, T.M., Gaysinsky, S., Davidson, P.M., Bruce, B.D., Weiss, J., 2007. Characterization of
high pressure processing with enterocins or thymol on the inactivation of listeria antimicrobial-bearing liposomes by ζ-potential, vesicle size, and encapsulation efficiency.
monocytogenes and the characteristics of sliced dry-cured ham. Food Bioprocess Technol. Food Biophysics 2, 1–9. https://doi.org/10.1007/s11483-007-9023-x.
12, 288–297. https://doi.org/10.1007/s11947-018-2212-4. Teixeira, M.L., dos Santos, J., Silveira, N.P., Brandelli, A., 2008. Phospholipid nanovesicles
Pérez-Ramos, A., Madi-Moussa, D., Coucheney, F., Drider, D., 2021. Current knowledge of the containing a bacteriocin-like substance for control of listeria monocytogenes. Innovative
mode of action and immunity mechanisms of LAB-bacteriocins. Microorganisms 9, 2107. Food Sci. Emerg. Technol. 9, 49–53. https://doi.org/10.1016/j.ifset.2007.05.001.
https://doi.org/10.3390/microorganisms9102107. Terebiznik, M., Jagus, R., Cerrutti, P., de Huergo, M.S., Pilosof, A.M.R., 2002. Inactivation of
Pisoschi, A.M., Pop, A., Georgescu, C., Turcuş, V., Olah, N.K., Mathe, E., 2018. An overview of Escherichia coli by a combination of nisin, pulsed electric fields, and water activity reduc-
natural antimicrobials role in food. Eur. J. Med. Chem. 143, 922–935. https://doi.org/10. tion by sodium chloride. J. Food Prot. 65, 1253–1258. https://doi.org/10.4315/0362-
1016/j.ejmech.2017.11.095. 028X-65.8.1253.
Pol, I.E., Mastwijk, H.C., Slump, R.A., Popa, M.E., Smid, E.J., 2001. Influence of food matrix Turgis, M., Vu, K.D., Dupont, C., Lacroix, M., 2012. Combined antimicrobial effect of essential
on inactivation of Bacillus cereus by combinations of nisin, pulsed electric field treat- oils and bacteriocins against foodborne pathogens and food spoilage bacteria. Food Res.
ment, and carvacrol. J. Food Prot. 64, 1012–1018. https://doi.org/10.4315/0362- Int. 48, 696–702. https://doi.org/10.1016/j.foodres.2012.06.016.
028X-64.7.1012. Van Cauteren, D., Le Strat, Y., Sommen, C., Bruyand, M., Tourdjman, M., Da Silva, N.J.,
Prado-Acosta, M., Ruzal, S.M., Allievi, M.C., Palomino, M.M., Sanchez Rivas, C., 2010. Syner- Couturier, E., Fournet, N., de Valk, H., Desenclos, J.-C., 2017. Estimated annual numbers
gistic effects of the lactobacillus acidophilus surface layer and nisin on bacterial growth. of foodborne pathogen-associated illnesses, hospitalizations, and deaths, France,
Appl. Environ. Microbiol. 76, 974–977. https://doi.org/10.1128/AEM.01427-09. 2008–2013. Emerg. Infect. Dis. 23, 1486–1492. https://doi.org/10.3201/eid2309.
Ramaroson, M., Guillou, S., Rossero, A., Rezé, S., Anthoine, V., Moriceau, N., Martin, J.-L., 170081.
Duranton, F., Zagorec, M., 2018. Selection procedure of bioprotective cultures for their Verdi, M.C., Melian, C., Castellano, P., Vignolo, G., Blanco Massani, M., 2020. Synergistic an-
combined use with high pressure processing to control spore-forming bacteria in cooked timicrobial effect of lactocin AL 705 and nisin combined with organic acid salts against
ham. Int. J. Food Microbiol. 276, 28–38. https://doi.org/10.1016/j.ijfoodmicro.2018.04. listeria innocua 7 in broth and a hard cheese. Int. J. Food Sci. Technol. 55, 267–275.
010. https://doi.org/10.1111/ijfs.14302.
Rodríguez, E., Tomillo, J., Nuñez, M., Medina, M., 1997. Combined effect of bacteriocin- Vukomanović, M., Žunič, V., Kunej, Š., Jančar, B., Jeverica, S., Podlipec, R., Suvorov, D., 2017.
producing lactic acid bacteria and lactoperoxidase system activation on listeria Nano-engineering the antimicrobial Spectrum of lantibiotics: activity of nisin against gram
monocytogenes in refrigerated raw milk. J. Appl. Microbiol. 83, 389–395. https://doi. negative bacteria. Sci. Rep. 7, 4324. https://doi.org/10.1038/s41598-017-04670-0.
org/10.1046/j.1365-2672.1997.00243.x. Wandling, L.R., Sheldon, B.W., Foegeding, P.M., 1999. Nisin in milk sensitizes bacillus spores
Rodriguez, E., Arques, J.L., Nuñez, M., Gaya, P., Medina, M., 2005. Combined effect of high- to heat and prevents recovery of survivors†. J. Food Prot. 62, 492–498. https://doi.org/
pressure treatments and bacteriocin-producing lactic acid bacteria on inactivation of 10.4315/0362-028X-62.5.492.
Escherichia coli O157:H7 in raw-Milk cheese. Appl. Environ. Microbiol. 71, Wu, T., Jiang, Q., Wu, D., Hu, Y., Chen, S., Ding, T., Ye, X., Liu, D., Chen, J., 2019. What is new
3399–3404. https://doi.org/10.1128/AEM.71.7.3399-3404.2005. in lysozyme research and its application in food industry? A review. Food Chem. 274,
Rodríguez-Rubio, L., García, P., Rodríguez, A., Billington, C., Hudson, J.A., Martínez, B., 2015. 698–709. https://doi.org/10.1016/j.foodchem.2018.09.017.
Listeriaphages and coagulin C23 act synergistically to kill listeria monocytogenes in milk Yang, E., Fan, L., Jiang, Y., Doucette, C., Fillmore, S., 2012. Antimicrobial activity of
under refrigeration conditions. Int. J. Food Microbiol. 205, 68–72. https://doi.org/10. bacteriocin-producing lactic acid bacteria isolated from cheeses and yogurts. AMB Ex-
1016/j.ijfoodmicro.2015.04.007. press 2, 48. https://doi.org/10.1186/2191-0855-2-48.
Sánchez-González, L., Vargas, M., González-Martínez, C., Chiralt, A., Cháfer, M., 2011. Use of Yogesh, K., 2016. Pulsed electric field processing of egg products: a review. J. Food Sci.
essential oils in bioactive edible coatings: a review. Food Eng. Rev. 3, 1–16. https://doi. Technol. 53, 934–945. https://doi.org/10.1007/s13197-015-2061-3.
org/10.1007/s12393-010-9031-3. Yu, H.H., Song, M.W., Song, Y.J., Lee, N.-K., Paik, H.-D., 2019. Antibacterial effect of a mixed
Scallan, E., Hoekstra, R.M., Angulo, F.J., Tauxe, R.V., Widdowson, M.-A., Roy, S.L., Jones, J.L., natural preservative against listeria monocytogenes on lettuce and raw pork loin. J. Food
Griffin, P.M., 2011. Foodborne illness acquired in the United States—major pathogens. Prot. 82, 2001–2006. https://doi.org/10.4315/0362-028X.JFP-19-026.
Emerg. Infect. Dis. 17, 7–15. https://doi.org/10.3201/eid1701.P11101. Zapico, P., Medina, M., Gaya, P., Nuñez, M., 1998. Synergistic effect of nisin and the
Schnitzler, P., Koch, C., Reichling, J., 2007. Susceptibility of drug-resistant clinical herpes sim- lactoperoxidase system on listeria monocytogenes in skim milk. Int. J. Food Microbiol.
plex virus type 1 strains to essential oils of ginger, thyme, hyssop, and sandalwood. 40, 35–42. https://doi.org/10.1016/S0168-1605(98)00008-7.
Antimicrob. Agents Chemother. 51, 1859–1862. https://doi.org/10.1128/AAC.00426- Zhao, X., Zhen, Z., Wang, X., Guo, N., 2017. Synergy of a combination of nisin and citric acid
06. against Staphylococcus aureus and listeria monocytogenes. Food Addit. Contam., Part A
Settanni, L., Corsetti, A., 2008. Application of bacteriocins in vegetable food biopreservation. Int. 34, 2058–2068. https://doi.org/10.1080/19440049.2017.1366076.
J. Food Microbiol. 121, 123–138. https://doi.org/10.1016/j.ijfoodmicro.2007.09.001. Zhao, X., Chen, L., Wu, J., He, Y., Yang, H., 2020a. Elucidating antimicrobial mechanism of
Sheoran, P., Tiwari, S.K., 2021. Synergistically-acting enterocin LD3 and plantaricin LD4 nisin and grape seed extract against listeria monocytogenes in broth and on shrimp
against gram-positive and gram-negative pathogenic bacteria. Probiotics Antimicro. through NMR-based metabolomics approach. Int. J. Food Microbiol. 319, 108494.
Prot. 13, 542–554. https://doi.org/10.1007/s12602-020-09708-w. https://doi.org/10.1016/j.ijfoodmicro.2019.108494.
Shi, C., Zhang, X., Zhao, X., Meng, R., Liu, Z., Chen, X., Guo, N., 2017. Synergistic interactions Zhao, X., Chen, L., Zhao, L., He, Y., Yang, H., 2020b. Antimicrobial kinetics of nisin and grape
of nisin in combination with cinnamaldehyde against Staphylococcus aureus in pasteur- seed extract against inoculated listeria monocytogenes on cooked shrimps: survival and
ized milk. Food Control 71, 10–16. https://doi.org/10.1016/j.foodcont.2016.06.020. residual effects. Food Control 115, 107278. https://doi.org/10.1016/j.foodcont.2020.
Sidhu, P.K., Nehra, K., 2019. Bacteriocin-nanoconjugates as emerging compounds for enhanc- 107278.
ing antimicrobial activity of bacteriocins. J. King Saud Univ. Sci. 31, 758–767. https:// Zohri, M., Alavidjeh, M.S., Haririan, I., Ardestani, M.S., Ebrahimi, S.E.S., Sani, H.T., Sadjadi,
doi.org/10.1016/j.jksus.2017.12.007. S.K., 2010. A comparative study between the antibacterial effect of nisin and nisin-
Sidhu, P.K., Nehra, K., 2020. Bacteriocin-capped silver nanoparticles for enhanced antimicro- loaded chitosan/alginate nanoparticles on the growth of Staphylococcus aureus in raw
bial efficacy against food pathogens. IET Nanobiotechnol. 14, 245–252. https://doi.org/ and pasteurized milk samples. Probiotics Antimicro. Prot. 2, 258–266. https://doi.org/
10.1049/iet-nbt.2019.0323. 10.1007/s12602-010-9047-2.
Sidhu, P.K., Nehra, K., 2021. Purification and characterization of bacteriocin Bac23 extracted
from lactobacillus plantarum PKLP5 and its interaction with silver nanoparticles for

16

You might also like