Next Article in Journal
Whole-Genome Sequencing to Predict Mycobacterium tuberculosis Drug Resistance: A Retrospective Observational Study in Eastern China
Next Article in Special Issue
Molecular Cloning, Expression Analyses, and Physiological Roles of Cathelicidins in the Bursa of Fabricius of the Japanese Quail, Coturnix japonica
Previous Article in Journal
Preventing Multidrug-Resistant Bacterial Transmission in the Intensive Care Unit with a Comprehensive Approach: A Policymaking Manual
Previous Article in Special Issue
From Gene to Transcript and Peptide: A Deep Overview on Non-Specific Lipid Transfer Proteins (nsLTPs)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evaluating the Translational Potential of Bacteriocins as an Alternative Treatment for Staphylococcus aureus Infections in Animals and Humans

Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(8), 1256; https://doi.org/10.3390/antibiotics12081256
Submission received: 27 June 2023 / Revised: 24 July 2023 / Accepted: 27 July 2023 / Published: 30 July 2023

Abstract

:
Antibiotic resistance remains a global threat to human and animal health. Staphylococcus aureus is an opportunistic pathogen that causes minor to life-threatening infections. The widespread use of antibiotics in the clinical, veterinary, and agricultural setting combined with the increasing prevalence of antibiotic-resistant S. aureus strains makes it abundantly clear that alternatives to antibiotics are urgently needed. Bacteriocins represent one potential alternative therapeutic. They are antimicrobial peptides that are produced by bacteria that are generally nontoxic and have a relatively narrow target spectrum, and they leave many commensals and most mammalian cells unperturbed. Multiple studies involving bacteriocins (e.g., nisin, epidermicin, mersacidin, and lysostaphin) have demonstrated their efficacy at eliminating or treating a wide variety of S. aureus infections in animal models. This review provides a comprehensive and updated evaluation of animal studies involving bacteriocins and highlights their translational potential. The strengths and limitations associated with bacteriocin treatments compared with traditional antibiotic therapies are evaluated, and the challenges that are involved with implementing novel therapeutics are discussed.

1. Introduction

Antibiotic resistance remains a global threat to human health. The World Health Organization predicts that, by 2050, the increase of antibiotic resistance could lead to an estimated 10 million deaths annually, costing approximately USD 100 trillion [1]. Staphylococcus aureus, specifically methicillin-resistant S. aureus (MRSA), is a significant contributor to this trend despite S. aureus being an opportunistic pathogen that only causes infection under certain circumstances. Many S. aureus infections occur in individuals who are already colonized by the bacterium. This is significant because 30–50% of the population is colonized with S. aureus on their skin, in their intestine, or in their nasal cavities; about one third of the population are persistently colonized [2,3]. Certain conditions also significantly increase an individual’s risk of developing a symptomatic S. aureus infection, such as type 1 diabetes [4], immunodeficiencies [5], and undergoing surgery [6] or hemodialysis [7]. Infections vary in presentation based on the site of infection but can include bacteremia, sepsis, toxic shock syndrome, endocarditis, pneumonia, gastroenteritis, meningitis, urinary tract infections, scalded skin syndrome, osteomyelitis, and multiple skin and soft tissue infections (SSTIs) [8]. To this day, S. aureus remains one of the most difficult-to-treat ESKAPE pathogens (Enterococcus faecium, S. aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species).
As patients infected with S. aureus tend to have recurrent infections, antibiotic resistance can quickly occur. Several major classes of antibiotics (e.g., β-lactams, glycopeptides, fluoroquinolones) are now largely ineffective at controlling S. aureus infections. A timeline describing the emergence of antibiotic-resistant strains is depicted in Figure 1. The methods by which S. aureus gains resistance and the impact that these resistance mechanisms have on treatment outcomes have been the focus of extensive research. Historically, the treatment of S. aureus infections primarily involved penicillin G, which is a β-lactam antibiotic. The production of β-lactamase led to the emergence of penicillin-resistant S. aureus (PRSA), and PRSA became widespread by the late 1950s. Synthesis of penicillin G derivatives that were resistant to β-lactamase hydrolysis became a priority, and methicillin, a β-lactam antibiotic that is resistant to hydrolysis by β-lactamase, was synthesized not long after. Methicillin functions similarly to other β-lactams as it targets the penicillin-binding proteins (PBPs) that are used in the formation of peptidoglycan in the cell wall [9]. Unfortunately, methicillin-resistant strains of S. aureus (MRSA) began to appear not long after methicillin was first used to treat S. aureus infections in the clinic [10]. Methicillin resistance occurs through the expression of PBP2a, a PBP that is resistant to methicillin’s mechanism of action and that can take over the cross-linking reactions of the host PBPs [11]. MRSA strains differ genetically from methicillin-sensitive S. aureus (MSSA) isolates by the presence of a large stretch of foreign DNA that includes the mecA gene, which encodes PBP2a. A global rise in MRSA infections and high resistance to all β-lactam antibiotics resulted in multidrug-resistant MRSA clones becoming the most common causative agent of S. aureus infections by the late 1990s [12]. Community-associated MRSA (CA-MRSA) began to spread in the 1990s [13], and livestock-associated MRSA (LA-MRSA) began to spread in the 2000s [14].
Glycopeptides such as vancomycin or teicoplanin are a class of antibiotics that eliminate S. aureus by preventing cell wall synthesis. Vancomycin specifically interferes with late-stage peptidoglycan synthesis, which results in cell death [15]. Glycopeptides and specifically vancomycin have remained the most common treatment for MRSA infections since the 1980s [16]. The use of vancomycin increased as a direct result of the increasing frequency of MRSA infections [17], which led to the emergence of vancomycin-resistant strains. Vancomycin-resistant S. aureus is categorized into vancomycin intermediate-resistance S. aureus (VISA) and vancomycin-resistant S. aureus (VRSA). The major difference between VISA and VRSA is the threshold in the minimum inhibitory concentration (MIC) values (4–8 µg/mL vs. ≥16 µg/mL, respectively). The first VISA strain was isolated from a patient in Japan in 1997 [18], but some studies have suggested that reduced susceptibility to vancomycin in S. aureus isolates dates back to 1987 [19]. The first VRSA strain was isolated from a patient in 2002 in the United States [20]. VISA phenotypes are known to arise from heterogenous populations of S. aureus where most cells have little or no resistance to vancomycin. After exposure to vancomycin, most S. aureus cells are eliminated, but some survive; these survivors persist and ultimately result in the homogenous VISA phenotype [21]. VRSA strains gain vancomycin resistance via the vanA operon on transposon Tn1546, which was originally obtained from a vancomycin-resistant enterococci (VRE) conjugative plasmid [22]. VRSA strains have remained rare, which is probably due to a considerable fitness cost that is associated with vanA in S. aureus [23].
Fluoroquinolones represent some of the most prescribed antibiotics. Fluoroquinolones exert their bactericidal effects by inhibiting the activity of topoisomerase II (gyrase) and topoisomerase IV, which are responsible for DNA superspiralization and respiralization, respectively. By inhibiting these activities, the synthesis of DNA is impaired and the bacterial cells die. Fluoroquinolones are effective against most MSSA infections but few MRSA infections [24]. S. aureus resistance to fluoroquinolones occurs via mutations in the gyrA, gyrB (topoisomerase II), parC (grlA), and are (topoisomerase IV) genes, and these mutations result in the synthesis of proteins with reduced susceptibility or insensitivity to fluoroquinolones [25]. Resistance can also be achieved through the overproduction of the chromosome-encoded proteins that are responsible for the efflux of fluoroquinolones through major facilitator superfamily (MFS) transporters [26].
The continual arms race between antibiotic discovery and the development of antibiotic resistance is a cause for great concern. The issue is exasperated by the high costs of novel antibiotic therapies, the limited effectiveness of antibiotic treatments, and the adverse outcomes for patients [27]. Alternatives to antibiotics are therefore needed to treat the devastating infections that are caused by antibiotic-resistant strains of S. aureus. Some potential therapeutics include bacteriophages, probiotics, prebiotics, and antimicrobial peptides that are produced by other bacteria (i.e., bacteriocins) [28,29,30,31]. The aim of this review is to provide a comprehensive evaluation of bacteriocins and focus on the translational potential of bacteriocins as an alternative therapeutic for S. aureus infections.

2. Bacteriocins

2.1. Background

Bacteriocins are a group of proteins or peptides that are produced by bacteria to kill competitors. They are usually mostly active against phylogenetically closely related strains. Bacteriocins are a diverse group consisting of various sizes, characteristics, producer species, target species, and mechanisms of action. The production of bacteriocins is costly to the producer cell, which is why their production is tightly regulated [32,33,34]. Despite the high cost to the producer cell, several isolates of many bacterial and archaebacterial species produce bacteriocins. It has been estimated that bacteriocin-producing isolates can be found in 30–99% of bacterial species [35,36,37]; therefore, it is believed that bacteriocins enhance the competitiveness of producer cells.
Bacteriocins can be classified into several categories. Both Class I and Class II bacteriocins are heat-stable and have a molecular weight of less than 10 kDa. Class I bacteriocins are post-translationally modified peptides that often result in non-standard amino acids, whereas Class II bacteriocins are not post-translationally modified. Class III bacteriocins, in contrast, are thermo-labile large (>30 kDa) unmodified proteins [37,38,39]. Due to the differences in size and heat lability between Class III bacteriocins and the other classes, some have suggested reclassifying Class III as bacteriolysins [37]. Class I and Class II bacteriocins can be further divided into three and four subclasses, respectively. Class Ia bacteriocins (i.e., the lantibiotics) characteristically contain lanthionine and β-methyl lanthionine, Class Ib comprise carbacylic lantibiotics that contain labyrinthin and labionin, and Class Ic bacteriocins are composed of sactibiotics that usually contain cysteine sulfur to α-carbon linkages [40,41,42]. With respect to the Class II subclasses, Class IIa contain pediocin-like bacteriocins, Class IIb contain two-component systems that form an active complex that is constructed from two different peptides, Class IIc contain circular bacteriocins, and Class IId contain non-pediocin-like bacteriocins [40,42].
The exact method of bacterial killing by bacteriocins varies, but it typically occurs in one of two ways: the bacteriocin interacts with the target cell envelope or it exerts its bactericidal activity inside the target cell, such as by inhibiting the synthesis of essential macromolecules. For example, nisin (nisin A) is a Class Ia bacteriocin, a lantibiotic, and one of the most extensively studied bacteriocins. It has a well-known mechanism of action against a wide spectrum of Gram-positive bacteria, including S. aureus. Nisin A, its biological and synthetic variants, and several other lantibiotics prevent cell wall synthesis and form pores in the target cell by binding to the peptidoglycan precursor lipid II molecules, which results in target cell death [37,43,44]. Interacting with lipid II is a common mechanism of bacterial killing among Class Ia bacteriocins [45,46,47,48], but the mechanism of action and the receptors involved in Class Ib and Class Ic bacteriocin-induced bacterial killing remain unknown [40]. The four Class II subclasses induce membrane permeabilization via pore formation with amphiphilic helical structures that can insert into the target cell membranes without the need for an “anchor” molecule like lipid II, which results in depolarization and target cell death [37]. However, the receptors involved in pore formation by Class II bacteriocins vary. In general, Class IIa bacteriocins utilize mannose permease, Class IIb use undecaprenyl pyrophosphate phosphatise (UppP), Class IIc involve an ABC transporter, and Class IId bacteriocins utilize a metallopeptidase [40]. Class III bacteriocins also result in target cell death by catalyzing the hydrolysis of the cell wall, but the specific receptors that are involved remain unknown [37]. Even though the mechanisms underlying the bactericidal activity of some bacteriocins have been identified, there are many more bacteriocins with mechanisms that have yet to be elucidated.

2.2. The Strengths and Limitations of Bacteriocins

When employed as bactericidal agents, bacteriocins offer several advantages over antibiotics. Bacteriocins can be considered more “natural” to humans because many are produced by bacteria that are present in the foods that have been consumed throughout human history. For example, lactic acid bacteria (LAB) produce a wide range of bacteriocins that can be isolated from food products such as meat and cheese [49]. Bacteriocins are also stable at high temperatures and in environments with extreme pH levels, and some bacteriocins retain bactericidal activity even after autoclavation [50,51]. Compared with antibiotics, many bacteriocins have lower toxicity profiles [52], and some reach their desired effect at lower concentrations (i.e., are more potent) [53]; however, a few studies have observed signs of toxicity in certain cell lines from animal (e.g., mice, rabbits, cows) and human sources [54,55].
Bacteriocins may be produced by chemical synthesis, using cell-free systems, or by fermentation of the naturally producing or engineered bacteria [56]. Historically, the production of bacteriocins involved batch fermentation by a producer strain. Production and purification are often challenging and may be optimized by heterologous bacteriocin expression [57]. As for most antibiotics, the chemical synthesis of bacteriocins is challenging due to their often complex chemistry. Bioengineering has given rise to bacteriocin derivatives with enhanced antimicrobial activity and derivatives that are tailored for therapeutic interventions [28,58,59]. Despite advances in purification techniques and expression systems, the production of bacteriocins in higher quantities as needed for their potential use as therapeutics is still a challenging and expensive task [60,61,62].
Conventional antibiotic therapies tend to have a broad-spectrum nature; thus, commensal bacteria of the gut are often eliminated as unintended consequences of the treatment. The concomitant elimination of non-pathogenic members of the gut microbiome can lead to the overgrowth of opportunistic pathogens. For example, antibiotic exposure is one of the most important risk factors for developing Clostridioides difficile infection (CDI) [63,64]. Bacteriocins have somewhat narrower target spectra than antibiotics, but the mechanisms of action of bacteriocins are not sufficiently species-specific that the killing of non-pathogenic bacteria with potentially important roles for human health can be completely avoided. Furthermore, the narrower target spectrum of bacteriocins can become problematic in the clinic because it requires that clinicians identify the causative agent of infection. Initial antimicrobial therapy of critically ill patients typically includes broad-spectrum antibiotics due to the etiological agent often being unknown at the time, and the early administration of these treatments is an important determinant of hospital mortality [65,66,67,68]. The potential immunogenicity of these bacteriocin therapies is also of concern, especially for the longer peptides.
Bacteriocins can be used to eliminate antibiotic-resistant strains because their mechanism of action differs from antibiotics [28]. However, it is to be anticipated that resistance to bacteriocins will develop or similarly lead to the resistance to antibiotics. Mechanisms of bacteriocin resistance are diverse and include enzymatic inactivation such as via proteolysis, target modifications, and efflux systems, among others. These resistance mechanisms have been elucidated mostly for nisin [69]. For example, acid-adapted Listeria monocytogenes displayed resistance to the bacteriocins nisin and lacticin 3147 [70], while Streptococcus bovis developed nisin resistance [71]. The spread of bacteriocin resistance is even more likely because bacteriocin-producing bacteria commonly have specific producer immunity factors encoded in their biosynthetic cluster; thus, specific resistance factors are already present in nature. The fact that bacteriocins commonly target bacteria that are phylogenetically related also means that such resistance factors may be readily functional once transferred by horizontal gene transfer to target pathogens. Moreover, target bacteria may develop non-specific resistance via other physiological alterations that are often based on random mutations, including the direct degradation of bacteriocins via enzymatic action, mimicry of natural immunity of the bacteriocin producer, and adaption of target cell membrane and/or growth conditions [72,73,74,75]. Finally, cross-resistance, or resistance to both antibiotics and bacteriocins, becomes a genuine and concerning possibility if bacteriocins and antibiotics are employed in combination as a dual therapeutic [76]. Unfortunately, there are a limited number of studies that have investigated the development of bacteriocin resistance in S. aureus [77], but Blake et al. reported a substantial decrease (4- to 32-fold reduction) in nisin susceptibility in small-colony variants of S. aureus. Comparative genome sequencing of a single nisin-resistant colony with a 32-fold increase in MIC values revealed two mutations, and the reintroduction of these mutations in nisin-susceptible strains conferred up to a 16-fold reduction in nisin susceptibility [78]. Thus, bacteriocins may represent a potential alternative to antibiotics for the treatment of S. aureus infections; however, the possibility of S. aureus developing or acquiring resistance cannot be understated.
Being peptides or peptide-like molecules, bacteriocins are generally susceptible to degradation by proteolytic enzymes (proteases), which usually results in a significant loss of antimicrobial activity. The sensitivity to digestive proteases (e.g., trypsin, chymotrypsin, and pepsin) is problematic when considering bacteriocins as potentially orally administered therapeutics [51,56,79,80]. Parenteral administration might avoid degradation of the bacteriocin in the digestive tract, but it cannot circumvent all bacteriocin–protease interactions as the bacteriocin will still be subjected to proteases in the blood [77,81]. Bacteriocins are rarely resistant to proteolytic enzyme degradation, but one study has shown that the bacteriocin BAC1B17 was resistant to multiple proteases, including digestive proteases, for up to two hours with no loss of antimicrobial activity against several MRSA strains [82]. Furthermore, many lantibiotics have increased anti-proteolytic stability due to their characteristic lanthionine bridges, which can be interpreted as an evolutionary adaptation to counteract the enzymatic inactivation by target organisms [83]. Enzymatic degradation dramatically decreases the bioavailability and half-life of bacteriocins, limiting the potential of bacteriocins to serve as a therapeutic alternative to antibiotics.
To combat some of the innate limitations of bacteriocins, mainly the enzymatic degradation issue, delivery systems that use nanoparticles have been developed. Nanoparticles are composed of metals, metal oxides, inorganic matter, organic matter, and/or carbon that are <100 nm in size and have varying dimensionality [84,85,86]. Several in vitro studies involving nisin or other bacteriocins have shown that using nanoparticles to encapsulate bacteriocins can enhance their therapeutic effects (e.g., increasing antimicrobial killing, bioavailability, and half-life) and minimize their degradation. For example, by using a combination of chitosan and alginate as delivery vehicle, nisin-loaded nanoparticles had greater antimicrobial effects against S. aureus than free nisin and better inhibited the growth of S. aureus in milk and cheese [87,88]. Another study found that a nisin-loaded poly(vinyl alcohol)–wheat gluten–zirconia membrane had well-controlled nisin release and that the delivery method enhanced nisin’s antimicrobial activity against S. aureus [89]. In a murine excisional skin infection model, a nisin-containing poly(ethylene oxide) and poly(D, L-lactide) nanofiber (50:50) blend significantly reduced S. aureus burden after seven days compared with the no-nisin control nanofiber dressings [90]. Finally, Liu et al. used nanoparticles to deliver the hydrophobic bacteriocin micrococcin P1 that is produced by a strain of Staphylococcus hominis and achieved a significant reduction in the S. aureus CFU and disease manifestations in systemic mouse and skin infection models [91]. Thus, using nanotechnology to enhance the antimicrobial effects of bacteriocins and mitigate their limitations shows promise, but additional studies are needed to determine toxicity profiles, pharmacokinetics, and potential interactions between the nanoparticles, bacteriocins, and target bacteria.

3. In Vivo Experiments Evaluating the Efficacy of Bacteriocins against S. aureus Infections

3.1. Skin Infection Models

Skin and soft tissue infections (SSTIs) are common bacterial infections that account for ~10% of infection-related hospital admissions in the United States [92,93]. SSTIs vary in severity, ranging from mild superficial infections (e.g., folliculitis, cellulitis) to life-threatening infections such as necrotizing fasciitis and shock due to Staphylococcal scalded skin syndrome. Treatment depends on the infection severity, etiologic agent, and drug susceptibility of the pathogen [94,95,96]. S. aureus is a leading cause of SSTIs, and many of these infections involve drug-resistant strains of S. aureus.
There are several in vivo studies that have investigated the translational potential of bacteriocins to treat S. aureus-induced SSTIs. Van Staden Adu et al. used a mouse wound infection model with imaging of the bioluminescent S. aureus Xen36 to test the antimicrobial activity of the bacteriocins nisin, clausin, and amyloliquecidin compared with mupirocin ointment. The latter is often used as a treatment for S. aureus-induced SSTIs in the clinic. The bioluminescence of S. aureus was significantly reduced in all three bacteriocin treatment groups to a value that was similar to that observed in the mupirocin treatment group [97]. In addition, the bacteriocin-treated mice had smaller wounds on day seven of the experiment with no significant difference in the numbers of S. aureus cells between the bacteriocin and antibiotic treatment groups [97]. Another study involving nisin found that S. aureus burdens were significantly reduced in mice that were treated with a nisin-eluting nanofiber wound dressing compared with the no-nisin control nanofiber dressing [90]. As antimicrobials often become ineffective in single-antimicrobial formations, Ovchinnikov et al. investigated the efficacy of a combination formulation of two bacteriocins (garvicin KS and micrococcin P1) and penicillin G to treat MRSA skin infections in a mouse model compared with fucidin cream, which is commonly used to treat skin infections. The three-component formulation eradicated S. aureus Xen31, a multidrug-resistant bioluminescent strain of MRSA, from skin puncture wounds after four consecutive days of treatment [98]. These results suggest that the addition of bacteriocins to previously ineffective antibiotics such as penicillin G might render these antibiotics effective again. However, any conclusions drawn from the study are limited because the in vivo efficacy of the individually administered penicillin G, garvicin KS, and micrococcin P1 and a bacteriocin-only combination of garvicin KS and micrococcin P1 were not tested. Finally, the recently discovered bacteriocin lugdunin, which is produced by strains of Staphylococcus lugdunensis, showed strong in vitro activity against S. aureus and some other tested Gram-positive pathogens [99]. In a skin infection model, application of lugdunin led to strongly reduced surface-attached S. aureus CFU but only moderately reduced the CFU in deeper skin tissue. These results indicate that an analysis of deeper tissue should generally be included in similar efficacy assessments of topical anti-S. aureus formulae.
Rather than using pure bacteriocin substance, bacteriocin-producing strains can also be topically applied to wound infections using a “probiotic” approach. Liu et al. applied a micrococcin P1-producing S. hominis strain to S. aureus-infected wounds in mice while using the bioluminescent strain Xen 36. The authors reported reduced S. aureus burden and wound closure time when using this approach [91].

3.2. Respiratory Infection Models

One of the many manifestations of S. aureus infection is pneumonia, which is usually seen in patients who are recovering from influenza. Individuals who are colonized on the skin or in the nares are the most at-risk for developing staphylococcal pneumonia [100]. Current treatment includes vancomycin or linezolid for MRSA and oxacillin, nafcillin, or cefazolin for MSSA [101,102]. As resistance to these antibiotics, particularly with respect to linezolid and vancomycin, continues to become more common in healthcare settings, alternative methods of abating S. aureus respiratory infections are needed; two bacteriocins, nisin F and NVB333, have shown some promise in this regard [103,104].
Nisin F, a natural derivative of nisin, is a lantibiotic produced by Lactococcus lactis F10 that has anti-staphylococcal activity in vitro [105]. One study evaluated if nisin F could treat immunosuppressed mice with S. aureus-induced respiratory infections. De Kwaadsteniet et al. found that nisin F prevented lung tissue damage caused by S. aureus in immunosuppressed mice, but nisin F itself had no effect on the healthy, non-immunosuppressed mice, as both treated and untreated mice had no lung tissue damage [104]. Boakes et al. evaluated NVB333 as a potential therapeutic in a murine lung infection model. They challenged mice with S. aureus MRSA UNT084-3 intranasally and gave NVB333 2 and 14 h post infection. NVB333 significantly reduced bacterial loads 26 h after infection compared with vancomycin treatment in a dose-dependent fashion [103]. However, NVB333 could not fully eradicate S. aureus from the lungs.

3.3. Systemic Infection and Other Severe Infection Models

Severe systemic S. aureus infection is often modeled using intraperitoneal injections. After an intraperitoneal challenge, large volumes of bacterial suspensions are quickly absorbed into the body. S. aureus subsequently invades the organs and causes systemic infection. Such models are often used to evaluate the effectiveness of novel therapeutics in preventing severe and rapid-onset infections. Several studies have used intraperitoneal injections, for example, to examine the effectiveness of mutacin 1140, lysostaphin, and lacticin NK34 against systemic MRSA infections.
Mutacin 1140 is a member of the epidermin-type lantibiotics with a broad activity spectrum against Gram-positive bacteria. Like other bacteriocins, mutacin 1140’s clinical applications are limited by its short half-life (e.g., 1.6 h in a rat model) [106]. As it is speculated that the high rate of clearance is associated with enzymatic degradation and attack by nucleophiles [107], Geng et al. evaluated the effects of charged and dehydrated residues on mutacin 1140’s pharmacokinetics after an MRSA intraperitoneal challenge. The alanine substitutions for lysine (K2A) or arginine (R13A) resulted in significantly lower clearances and higher plasma concentrations of mutacin 1140 over time. A single intravenous injection (10 mg/kg) of the K2A and R13A analogs protected 100% of the mice, while a smaller dose (2.5 mg/kg) resulted in a 50% survival compared with a 100% mortality in the vehicle control group. In addition, there was a significant reduction in the bacterial load in the kidneys and livers of the 10 mg/kg analog groups compared with the vehicle control group [107]. While these results are promising, additional studies are needed to determine the most effective dosage, tolerability, toxicology profiles, and efficacy of infection clearance of mutacin 1140 versus standard-of-care antibiotic regiments. Two studies evaluated a Class III bacteriocin, lysostaphin, for the treatment of systemic S. aureus infections compared with the antibiotic vancomycin. Lysostaphin, which degrades the staphylococcal cell wall, was originally isolated from Staphylococcus simulans [108]. Treatment with lysostaphin was shown to be as effective as treatment with vancomycin in MRSA and MSSA rodent neonate infection models [109,110]. While these studies show that lysostaphin is a potent bacteriocin that can clear S. aureus infections, additional studies are needed to determine the extent of lysostaphin’s translational potential. Another study evaluated the efficacy of lacticin NK34 at treating systemic S. aureus infections. Lacticin NK34 is a nisin-like lantibiotic that was isolated from Lactococcus lactis and that has antimicrobial activity against S. aureus and other Gram-positive bacteria [111]. After an intraperitoneal challenge of S. aureus at the minimal lethal dose (MLD), Kim et al. treated mice with lacticin NK34. Lacticin NK34 treatment increased the survival between treatment groups from 5% in the non-treatment group to 72% in the treatment group after seven days [112]. However, the study only used a single strain of S. aureus, and a previous study published by the same laboratory showed that multiple S. aureus strains were not inhibited by lacticin NK34 [111].
Osteomyelitis, an infection of the bones, is commonly caused by staphylococcal species such as S. aureus and S. epidermidis [113]. To prevent orthopedic-related bacterial infections after surgical procedures, polymethylmethacrylate (PMMA) bone cement that is loaded with antibiotics is often used, as it has been shown to reduce the rate of infection in revision hip arthroplasty and primary hip arthroplasty by 40% and 50%, respectively [114,115]. One study evaluated if nisin F could control orthopedic-related bacterial infections when loaded onto cement that was subcutaneously implanted on the back of mice before infection with bioluminescent S. aureus Xen 36. The nisin F-loaded cement prevented S. aureus infection for seven days, and no viable bacterial cells were obtained after the mice were euthanized and the bone cylinders were removed [116].
S. aureus is also one of the most common causes of infective endocarditis, which has a high rate of mortality [117]. In one study, the lantibiotic NAI-107 (“microbisporicin” produced by Microbispora sp.) [118] was investigated by Jabés et al. as a treatment for both S. aureus-induced endocarditis and systemic infection. The authors used a granuloma pouch model with an intraperitoneal injection of a MRSA strain and NAI-107 that was intravenously administered. NAI-107’s bactericidal activity was dose-proportional, and a single 40 mg/kg dose caused a 3-log reduction in viable MRSA in exudates compared with two 20 mg/kg doses at 12 or 24 h, respectively [119]. In the same study, female rats were induced with endocarditis by S. aureus before treatment with NAI-107 or vancomycin (twice/day for five days). The authors found a dose-dependent reduction in the bacterial load, which was significantly lower than that obtained with vancomycin at higher doses. Overall, NAI-107 showed promise in its ability to severe S. aureus infections, although the study did not assess mortality [119].

3.4. Nasal, Intestinal, and Skin Carriage

S. aureus colonizes the anterior nares in about one third of the population [120]. Nasal colonization with S. aureus and especially with MRSA variants is a known risk factor for developing infections, including complications after surgery, and several studies have indicated that nasal S. aureus represent the source of the infectious bacteria [120,121,122,123]. The antibiotic mupirocin is often used to decolonize the nares from S. aureus prior to surgery and is occasionally combined with chlorhexidine body washes; however, given the rising abundance of mupirocin-resistant S. aureus strains, the development of alternative decolonization methods needs to be considered [124,125,126].
Epidermicin NI01 is a novel class II bacteriocin produced by S. epidermidis that exhibits antimicrobial activity against a wide range of pathogens, including MRSA [127]. Epidermicin has been the subject of extensive study [127] even though its exact mechanism of action remains unknown. One study showed that epidermicin NI01 was at least as effective as mupirocin at decolonizing MRSA from the nares of cotton mice [128]. Twice-daily administrations of mupirocin multiple days in a row were required to reduce colonization, while a single dose of epidermicin NI01 resulted in persistent decolonization that lasted for days. Additionally, Sandiford and Upton found that epidermicin NI01 was active against susceptible bacteria at lower concentrations and that exposure to the bacteriocin did not result in the development of resistance in the target bacteria [127]. These results suggest that epidermicin NI01 is a promising antibiotic alternative that can reduce the risk of developing MRSA infections in hospitals, but there are additional alternatives. For example, mersacidin is a lantibiotic that is produced by Bacillus sp. HIL Y-85, 54728. It was shown to inhibit the growth of MRSA and other Gram-positive bacteria in vitro [129]. Kruszewska et al. found that mersacidin eliminated S. aureus nasal colonization after twice-daily applications for three days in a mouse rhinitis model. No signs of systemic infection were present and no S. aureus cells were recovered from the nares of mice treated with mersacidin [130]. Finally, S. lugdunensis that produced the above-mentioned lugdunin but was not a non-producing isogenic S. lugdunensis outcompeted S. aureus when applied to the noses of cotton rats [99], suggesting the potential of a probiotic approach that uses such bacteriocin-producing bacteria to control S. aureus nasal carriage.
It should be noted that recent findings have emphasized the role of the intestinal carriage of S. aureus, as carriers of S. aureus have many more S. aureus in their gut than in their noses [3,131]. Elimination of intestinal carriage by Bacillus subtilis, which produces molecules that inhibit the quorum-sensing system that is essential for S. aureus intestinal colonization [30], have been suggested to control S. aureus intestinal colonization and, due to the key role attributed to the intestine as the source of S. aureus, S. aureus colonization in general [131]. On the other hand, bacteriocin-producing bacteria have been shown to eradicate intestinal pathogens in mouse colonization models where most of the natural microbiome was eradicated or in germ-free mice such as Salmonella sp. by microcin-producing Escherichia coli [132]. However, whether bacteriocins show sufficient target specificity for a “microbiome editing” probiotic approach in healthy people that leaves the microbiome virtually intact—as does the more targeted B. subtilis quorum-quenching approach for S. aureus—is questionable due to the often relatively broad target spectrum of bacteriocins. It may work in specific cases [133], but generally, such approaches should be limited to the treatment of severe disease where microbiome effects are a calculated risk. Possibly, the effects on the nasal microbiome when using S. aureus-targeted bacteriocins for nasal carriage are less problematic than those in a potential gut-targeted bacteriocin-based approach; however, similarly to the traditional mupirocin-based strategies, they suffer from the general problem of recolonization from non-targeted S. aureus sites of colonization, particularly the intestine.
As for the skin, S. aureus does not normally colonize the skin of healthy individuals in considerable numbers, but increased S. aureus colonization is found in affected areas of atopic dermatitis (AD) patients [134]. Therefore, bacteriocin therapy has long been proposed as a means of S. aureus-targeted microbiome editing for AD treatment [135,136]; a recently completed clinical trial demonstrated the safety of a bacteriotherapy approach to treat AD with a bacteriocin-producing Staphylococcus hominis [137]. Furthermore, AD microbiome editing with an even more S. aureus-targeted form is possible with bacteriocins through quorum-quenching approaches in a form similar to that which was outlined above for the gut [138]. Of note, as the staphylococcal quorum-sensing signals are peptides, some have classified them as antimicrobial peptides [136]. However, they are not directly antimicrobial and thus clearly distinguished from bacteriocins.
A summary of in vivo experiments evaluating the efficacy of bacteriocins in S. aureus infections is shown in Table 1.

4. Additional Applications of Bacteriocins against S. aureus

4.1. Industrial Applications

Bacteriocins are used as antimicrobial agents in several sectors of industry. The use of bacteriocins in the preservation of meat, dairy, egg, and vegetable products has been thoroughly investigated, and bacteriocins are assumed to be nontoxic to the consumer [143,144,145]. The most widely studied bacteriocin, nisin (trademarked as NisaplinTM), is used as a natural food protectant when preparing dairy products and canned foods in over 48 countries as it is active against a range of Gram-positive pathogens such as S. aureus and food-borne pathogens such as Listeria monocytogenes and Clostridium botulinum [146,147,148]. Other bacteriocins with anti-S. aureus activity that could be used in food preservation include enterocin AS-48 in skim milk, non-fat soft cheese, and vegetable sausages [149,150,151]; enterocin CCM 4231 in skim milk and yogurt [152]; and bacteriocin CAMT2 in meat products and milk [153,154].
The potential use of bacteriocins in industry is, however, most important in the agricultural sector. Antibiotics used in farm animals account for approximately 73% of all antibiotic use [147,155]; in the United States, that number rises to 80% [156]. Antibiotics are used in agriculture and aquaculture to treat infections in sick livestock animals, as prophylactics to prevent disease in healthy animals, and as growth promoters to improve weight gain and feed conversion [157,158,159]. Approximately 70% of the antibiotics used in farm animals and in human patients are identical or nearly identical [160], making agriculture a significant driving force in the development of drug-resistant pathogens [161]. It is therefore important to identify areas of the agricultural sector that could employ alternative therapies, such as bacteriocins, to minimize antibiotic use.
For example, bacteriocins could be used to treat bovine mastitis, which is predominantly caused by S. aureus [162], in lactating and dry cows. Cao et al. investigated the ability of a nisin-based formulation vs. the antibiotic gentamicin, to which many S. aureus isolates have resistance, to treat bovine clinical mastitis in lactating cows. The nisin formulation offered a similar clinical cure rate to gentamicin [139]. Another study found that a bismuth-based teat seal product containing lacticin 3147 provided protection against S. aureus-induced bovine mastitis [142]. Several other bacteriocins have potential as treatments for S. aureus-induced bovine mastitis based on in vitro data involving S. aureus isolates from clinically presenting dairy cows [163,164]. Bacteriocins could also be used as prophylactics in lieu of antibiotics in the agricultural sector. Ryan et al. incorporated lacticin 3147 into an intramammary teat seal product for dry cows and found robust antimicrobial activity against all strains of streptococci and staphylococci that were tested, which included seventeen strains of S. aureus. The study also investigated a nisin-based intramammary teat seal product, but nisin’s insolubility at the physiological pH makes the nisin-based intramammary teat seal product unsuitable as a prophylactic for preventing bovine mastitis [165].
Bacteriocins have further potential applications in the pharmaceutical industry. For example, fermenticin HV6B is produced by a strain of Lactobacillus fermentum that was originally isolated from the human vagina. In vitro experiments showed that fermenticin HV6B possesses spermicidal activity, results in sperm immobilization, and is antimicrobial against several opportunistic pathogens, including S. aureus [166], but it has been noted that fermenticin HV6B is not suitable for commercial use because of its toxic effects on the native vaginal microbiota [167].

4.2. Bacteriocins against S. aureus in Clinical Trials

Results from pre-clinical animal studies and the successful use of bacteriocins in agriculture suggest that bacteriocins represent a viable alternative to antibiotics for treating S. aureus infections in humans. Unfortunately, to date, there are only a limited number of completed or ongoing clinical trials investigating the efficacy and safety of bacteriocins (Table 2). There are even fewer studies evaluating bacteriocins as a potential therapeutic for S. aureus infections (Table 2), and many studies have yet to publish or make their results available. Table 3 lists all bacteriocins, their structure and modes of action, if known, that were discussed in this review.
Novacta Biosystems Limited (UK) initiated a phase I clinical trial that investigated the safety, tolerability, and pharmacokinetics of single and multiple ascending orally administered doses of NVB302, which has promising in vitro and in vivo activity against MRSA and VRSA strains [103]. In part A of the study, up to five cohorts of eight healthy participants were randomized to receive either a single dose of NVB203 (starting dose: 100 mg) or a placebo. In part B of the study, up to four cohorts of eight healthy participants received a daily dose of NVB203 or a placebo for ten days. The trial was completed in 2012, but the results are not yet available (EudraCT/CTIS: 2011-002703-14).
Most clinical trials investigating the ability of bacteriocins as an alternative therapy against S. aureus involve nisin. As S. aureus is the predominant etiological agent of acute mastitis in humans [168,169], Fernández et al. investigated the usage of a nisin solution in lactating women with clinical signs of staphylococcal mastitis. The participants were randomly assigned to the nisin solution (6 μg/mL) group or control group (a similar solution devoid of nisin), and the solutions were applied to the nipple and mammary areola for two weeks. Compared with day 0, where the staphylococcal counts in the breast milk between the two groups were similar, the counts were significantly lower on day 14 in the nisin group (3.22 ± 0.43 log10 CFU/mL) compared with the control group (5.01 ± 0.21 log10 CFU/mL). In addition, no clinical signs of mastitis were reported for the nisin group on day 14, while the control group had clinical signs for the entire study. Another clinical trial (NCT02928042) investigated the inhibitory effects of nisin and LAB members on the etiological agents of ventilator-associated pneumonia [170]. Several pathogens (i.e., P. aeruginosa, A. baumannii, K. pneumonia, and S. aureus) were obtained from the tracheal aspiration cultures of 80 patients who were treated with mechanical ventilation (>48 h). The probiotic effects of the LAB species on several P. aeruginosa strains were published in 2014 [171], but the results pertaining to nisin’s antimicrobial activity against S. aureus have not been made available. A different clinical trial (NCT02467972) evaluated how the addition of several experimental additives that included nisin could impact hunger, satiety, bowel movements, and colonic bacterial populations. Sixty-one healthy participants were recruited and randomly assigned to one of the experimental groups or the control group. Nisin was added to 18 portions of ready-to-eat frozen soups (3 meals/week over 6 weeks), and changes in colonic bacterial populations were measured using the microbiome with r16S DNA sequencing; however, the results have yet to be published [172].
Table 2. Clinical trials with bacteriocins against S. aureus.
Table 2. Clinical trials with bacteriocins against S. aureus.
Identifier or CitationInterventionProposed Sample Size/EnrollmentPrimary Endpoint(s)StatusResults
Fernández, et al. [173]Application of a nisin solution (6 μg/mL) to the nipple and mammary areola8 lactating women with clinical signs of staphylococcal mastitis Evaluate the clinical signs of mastitis and bacterial loads after 2 weeksCompleteBacterial load in the nisin group was statistically lower than the control group. No clinical signs of staphylococcal mastitis were observed in the nisin group on day 14 of the trial
Clinical Trials.gov
NCT02467972
One of the experimental groups had a dietary supplement of nisin. Nisin was added to ready-to-eat frozen soups (3 times/week with 18 portions in total)61 healthy participants Change in bowel function, colonic bacteria population, and hormonal parameters related to hunger and satiety after six weeksCompleteNot available
Clinical Trials.gov
NCT02928042
Tracheal aspiration-derived pathogens were treated with LAB members and nisin to evaluate their effectiveness at treating ventilator-associated pneumonia80 patients who were mechanically ventilated for at least 48 h were recruited and who had their tracheal aspirate cultures used for the studyThe antimicrobial properties and effects of nisin and LAB members on P. aeruginosa, A. baumannii, K. pneumonia, and S. aureus growth rateCompleteNot available
EudraCT/CTIS
2011-002703-14
Part A: a single dose (100 mg) of NVB302 or a placebo
Part B: Once daily doses of NVB302 or a placebo for ten days
Part A: Up to five cohorts of 8 healthy subjects
Part B: Up to four cohorts of 8 healthy subjects
Assessment of the safety and tolerability of single and multiple oral ascending doses of NVB302CompleteNot available

5. Conclusions

Antibiotic resistance represents an urgent and growing threat to animal and human health worldwide. The arms race between antibiotic discovery and the emergence of resistant organisms is ever-ongoing, but investments into antibiotic development by biotechnology and pharmaceutical industries are slowing down [174]. S. aureus is a major contributor to antibiotic-resistant infections. Infections caused by antibiotic-resistant strains of S. aureus, such as MRSA, result in significant health-related and economic burdens across the clinical and agricultural sectors; thus, there is an urgent need to identify alternative pathways to treat these devastating infections. Bacteriocins represent one such alternative therapy. Bacteriocins are generally considered to be nontoxic and to have a narrower target spectrum than antibiotics, which reduces off-target effects on host mammalian cells and commensal bacteria. Nanotechnology can be used to enhance a bacteriocin’s antimicrobial activity against its target bacteria and minimize its limitations. Despite these benefits, there are few in vivo studies investigating the ability of bacteriocins as a potential therapeutic to treat S. aureus infections.
As of 2021, Benítez-Chao et al. estimate that approximately 50% of studies investigating the antimicrobial action of bacteriocins in murine models lack toxicity and/or biosafety studies [175]. However, toxicity assays and biosafety studies are important precursors for initiating a clinical trial. For the in vitro and in vivo studies to translate into the clinic and for the therapeutic potential of bacteriocins to be maximized, future experiments must include these crucial data. In addition, there is a clear lack of clinical trials that assess systemically administered bacteriocins for the treatment of severe S. aureus infections, which can probably be attributed to the limitations that are associated with systemic administration, such as proteolytic degradation, toxicity, or immunogenicity. These problems may have prevented seemingly promising bacteriocins from proceeding to clinical trials, and the negative outcomes of pre-clinical assessments may not have been published. Even though the efficacy results from many published animal models of infection are promising, additional in vivo studies and clinical trials are needed before bacteriocins can become a truly viable alternative to antibiotics in the treatment of infections with S. aureus.
Table 3. Additional information about bacteriocins of interest.
Table 3. Additional information about bacteriocins of interest.
BacteriocinPeptide SequenceMechanism of ActionReference
Mutacin 1140Antibiotics 12 01256 i001Inhibition of cell wall synthesis, pore formation[107,176,177,178]
Epidermicin NI01MAAFMKLIQFLATLGQKYVSLAWKHKGTILKWINAGQSFEWIYKQIKKLWAR (unmodified)Multi-mode pore formation induced by four-helices[127,179]
Nisin (Nisin A)Antibiotics 12 01256 i002
Nisin F: Nisin A H27N; Nisin Z: Nisin A H27N, I30V
Inhibition of cell wall synthesis, pore formation[105,180,181,182,183]
NAI-107 (Microbisporicin)Antibiotics 12 01256 i003Inhibition of cell wall synthesis[118]
CMB001Antibiotics 12 01256 i004Unknown[141,184]
Garvicin KSThree-component:
GakA, MGAIIKAGAKIVGKGVLGGGASWLGWNVGEKIWK
GakB, MGAIIKAGAKIIGKGLLGGAAGGATYGGLKKIFG
GakC, MGAIIKAGAKIVGKGALTGGGVWLAEKLFGGK
(unmodified)
Unknown[185]
Micrococcin P1Antibiotics 12 01256 i005Inhibition of ribosomal protein synthesis[186]
Lysostaphin26.9 kDa protein in mature formCleaving of pentaglycine bridges in the cell wall[187,188]
NVB333Antibiotics 12 01256 i006Inhibition of cell wall synthesis[103]
ClausinAntibiotics 12 01256 i007Inhibition of cell wall synthesis[47]
Amyloliquecidin Two-component lantibiotic, exact structure unknown, primary amino acid sequences:
CAWYDISCKLGNKGAWCTLTVECQSSCN, TTPSSLPCGVFVTAAFCPSTKCTSSC
Inhibition of cell wall synthesis[48]
MersacidinAntibiotics 12 01256 i008Inhibition of cell wall synthesis[45,189]
Lacticin NK34Antibiotics 12 01256 i009Inhibition of cell wall synthesis[112]
Lacticin 3147Antibiotics 12 01256 i010Inhibition of cell wall synthesis[46,190]
LugduninAntibiotics 12 01256 i011Disrupts membrane potential[99]

Author Contributions

Writing—original draft preparation, L.R.H. and A.R.P; writing—review and editing, M.O.; visualization, A.R.P., J.A.W. and M.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH) (project number ZIA AI000904, provided to M.O.).

Acknowledgments

The authors would like to thank Una Karanovic for assisting in the literature search for this review.

Conflicts of Interest

The authors report no potential conflict of interest.

References

  1. World Health Organization. Antimicrobial Resistance: Global Report on Surveillance; World Health Organization: Geneva, Switzerland, 2014; 256p. [Google Scholar]
  2. Noble, W.C.; Valkenburg, H.A.; Wolters, C.H.L. Carriage of Staphylococcus aureus in random samples of a normal population. Epidemiology Infect. 1967, 65, 567–573. [Google Scholar] [CrossRef] [PubMed]
  3. Acton, D.S.; Plat-Sinnige, M.J.T.; Van Wamel, W.; De Groot, N.; Van Belkum, A. Intestinal carriage of Staphylococcus aureus: How does its frequency compare with that of nasal carriage and what is its clinical impact? Eur. J. Clin. Microbiol. Infect. Dis. 2009, 28, 115–127. [Google Scholar] [CrossRef]
  4. Tuazon, C.U.; Perez, A.; Kishaba, T.; Sheagren, J.N. Staphylococcus aureus Among Insulin-Injecting Diabetic Patients. JAMA 1975, 231, 1272. [Google Scholar] [CrossRef] [PubMed]
  5. Weinke, T.; Schiller, R.; Fehrenbach, F.J.; Pohle, H.D. Association between Staphylococcus aureus nasopharyngeal colonization and septicemia in patients infected with the human immunodeficiency virus. Eur. J. Clin. Microbiol. Infect. Dis. 1992, 11, 985–989. [Google Scholar] [CrossRef]
  6. Kluytmans, J.A.J.W.; Mouton, J.W.; Ijzerman, E.P.F.; Vandenbroucke-Grauls, C.M.J.E.; Maat, A.W.P.M.; Wagenvoort, J.H.T.; Verbrugh, H.A. Nasal Carriage of Staphylococcus aureus as a Major Risk Factor for Wound Infections after Cardiac Surgery. J. Infect. Dis. 1995, 171, 216–219. [Google Scholar] [CrossRef] [PubMed]
  7. Yu, V.L.; Goetz, A.; Wagener, M.; Smith, P.B.; Rihs, J.D.; Hanchett, J.; Zuravleff, J.J. Staphylococcus aureus Nasal Carriage and Infection in Patients on Hemodialysis. N. Engl. J. Med. 1986, 315, 91–96. [Google Scholar] [CrossRef] [PubMed]
  8. Tong, S.Y.C.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus Infections: Epidemiology, Pathophysiology, Clinical Manifestations, and Management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef]
  9. Giesbrecht, P.; Kersten, T.; Maidhof, H.; Wecke, J. Staphylococcal Cell Wall: Morphogenesis and Fatal Variations in the Presence of Penicillin. Microbiol. Mol. Biol. Rev. 1998, 62, 1371–1414. [Google Scholar] [CrossRef]
  10. Chambers, H.F. Methicillin resistance in staphylococci: Molecular and biochemical basis and clinical implications. Clin. Microbiol. Rev. 1997, 10, 781–791. [Google Scholar] [CrossRef]
  11. Hackbarth, C.J.; Kocagoz, T.; Kocagoz, S.; Chambers, H.F. Point mutations in Staphylococcus aureus PBP 2 gene affect penicillin-binding kinetics and are associated with resistance. Antimicrob. Agents Chemother. 1995, 39, 103–106. [Google Scholar] [CrossRef]
  12. DeLeo, F.R.; Chambers, H.F. Reemergence of antibiotic-resistant Staphylococcus aureus in the genomics era. J. Clin. Investig. 2009, 119, 2464–2474. [Google Scholar] [CrossRef]
  13. Otto, M. Understanding the epidemic of community-associated MRSA and finding a cure: Are we asking the right questions? Expert Rev. Anti-Infect. Ther. 2009, 7, 141–143. [Google Scholar] [CrossRef] [PubMed]
  14. Fitzgerald, J.R. Livestock-associated Staphylococcus aureus: Origin, evolution and public health threat. Trends Microbiol. 2012, 20, 192–198. [Google Scholar] [CrossRef] [PubMed]
  15. Barna, J.C.J.; Williams, D.H. The structure and mode of action of glycopeptide antibiotics of the vancomycin group. Annu. Rev. Microbiol. 1984, 38, 339–357. [Google Scholar] [CrossRef]
  16. Maple, P.; Hamilton-Miller, J.; Brumfitt, W. World-wide antibiotic resistance in methicillin-resistant Staphylococcus aureus. Lancet 1989, 333, 537–540. [Google Scholar] [CrossRef] [PubMed]
  17. Levine, D.P. Vancomycin: A History. Clin. Infect. Dis. 2006, 42, S5–S12. [Google Scholar] [CrossRef]
  18. Hiramatsu, K.; Hanaki, H.; Ino, T.; Yabuta, K.; Oguri, T.; Tenover, F.C. Methicillin-resistant Staphylococcus aureus clinical strain with reduced vancomycin susceptibility. J. Antimicrob. Chemother. 1997, 40, 135–136. [Google Scholar] [CrossRef]
  19. Jackson, M.A.; Hicks, R.A. Vancomycin failure in staphylococcal endocarditis. Pediatr. Infect. Dis. J. 1987, 6, 750–751. [Google Scholar] [CrossRef]
  20. Chang, S.; Sievert, D.M.; Hageman, J.C.; Boulton, M.L.; Tenover, F.C.; Downes, F.P.; Shah, S.; Rudrik, J.T.; Pupp, G.R.; Brown, W.J.; et al. Infection with Vancomycin-Resistant Staphylococcus aureus Containing the vanA Resistance Gene. N. Engl. J. Med. 2003, 348, 1342–1347. [Google Scholar] [CrossRef]
  21. Howden, B.P.; Davies, J.K.; Johnson, P.D.R.; Stinear, T.P.; Grayson, M.L. Reduced Vancomycin Susceptibility in Staphylococcus aureus, Including Vancomycin-Intermediate and Heterogeneous Vancomycin-Intermediate Strains: Resistance Mechanisms, Laboratory Detection, and Clinical Implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef]
  22. Arthur, M.; Molinas, C.; Depardieu, F.; Courvalin, P. Characterization of Tn1546, a Tn3-related transposon conferring glycopeptide resistance by synthesis of depsipeptide peptidoglycan precursors in Enterococcus faecium BM4147. J. Bacteriol. 1993, 175, 117–127. [Google Scholar] [CrossRef]
  23. Foucault, M.-L.; Courvalin, P.; Grillot-Courvalin, C. Fitness Cost of VanA-Type Vancomycin Resistance in Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2009, 53, 2354–2359. [Google Scholar] [CrossRef]
  24. Weber, S.G.; Gold, H.S.; Hooper, D.C.; Karchmer, A.; Carmeli, Y. Fluoroquinolones and the Risk for Methicillin-resistant Staphylococcus aureus in Hospitalized Patients. Emerg. Infect. Dis. 2003, 9, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
  25. Lowy, F.D. Antimicrobial resistance: The example of Staphylococcus aureus. J. Clin. Investig. 2003, 111, 1265–1273. [Google Scholar] [CrossRef] [PubMed]
  26. Ding, Y.; Onodera, Y.; Lee, J.C.; Hooper, D.C. NorB, an Efflux Pump in Staphylococcus aureus Strain MW2, Contributes to Bacterial Fitness in Abscesses. J. Bacteriol. 2008, 190, 7123–7129. [Google Scholar] [CrossRef]
  27. Boucher, H.W.; Talbot, G.H.; Bradley, J.S.; Edwards, J.E.; Gilbert, D.; Rice, L.B.; Scheld, M.; Spellberg, B.; Bartlett, J. Bad Bugs, No Drugs: No ESKAPE! An Update from the Infectious Diseases Society of America. Clin. Infect. Dis. 2009, 48, 1–12. [Google Scholar] [CrossRef] [PubMed]
  28. Cotter, P.D.; Ross, R.; Hill, C. Bacteriocins—A viable alternative to antibiotics? Nat. Rev. Genet. 2012, 11, 95–105. [Google Scholar] [CrossRef]
  29. Piewngam, P.; Otto, M. Probiotics to prevent Staphylococcus aureus disease? Gut Microbes 2019, 11, 94–101. [Google Scholar] [CrossRef]
  30. Piewngam, P.; Zheng, Y.; Nguyen, T.H.; Dickey, S.W.; Joo, H.-S.; Villaruz, A.E.; Glose, K.A.; Fisher, E.L.; Hunt, R.L.; Li, B.; et al. Pathogen elimination by probiotic Bacillus via signalling interference. Nature 2018, 562, 532–537. [Google Scholar] [CrossRef]
  31. Pulingam, T.; Parumasivam, T.; Gazzali, A.M.; Sulaiman, A.M.; Chee, J.Y.; Lakshmanan, M.; Chin, C.F.; Sudesh, K. Antimicrobial resistance: Prevalence, economic burden, mechanisms of resistance and strategies to overcome. Eur. J. Pharm. Sci. 2021, 170, 106103. [Google Scholar] [CrossRef]
  32. Ebner, P.; Reichert, S.; Luqman, A.; Krismer, B.; Popella, P.; Götz, F. Lantibiotic production is a burden for the producing staphylococci. Sci. Rep. 2018, 8, 7471. [Google Scholar] [CrossRef] [PubMed]
  33. Maldonado-Barragán, A.; West, S.A. The cost and benefit of quorum sensing-controlled bacteriocin production in Lactobacillus plantarum. J. Evol. Biol. 2019, 33, 101–111. [Google Scholar] [CrossRef] [PubMed]
  34. Heilbronner, S.; Krismer, B.; Brötz-Oesterhelt, H.; Peschel, A. The microbiome-shaping roles of bacteriocins. Nat. Rev. Genet. 2021, 19, 726–739. [Google Scholar] [CrossRef] [PubMed]
  35. Klaenhammer, T.R. Bacteriocins of lactic acid bacteria. Biochimie 1988, 70, 337–349. [Google Scholar] [CrossRef] [PubMed]
  36. Riley, M.A. Molecular mechanisms of bacteriocin evolution. Annu. Rev. Genet. 1998, 32, 255–278. [Google Scholar] [CrossRef]
  37. Cotter, P.D.; Hill, C.; Ross, R.P. Bacteriocins: Developing innate immunity for food. Nat. Rev. Genet. 2005, 3, 777–788. [Google Scholar] [CrossRef]
  38. Jack, R.W.; Tagg, J.R.; Ray, B. Bacteriocins of gram-positive bacteria. Microbiol. Rev. 1995, 59, 171–200. [Google Scholar] [CrossRef]
  39. Klaenhammer, T.R. Genetics of bacteriocins produced by lactic acid bacteria. FEMS Microbiol. Rev. 1993, 12, 39–85. [Google Scholar] [CrossRef]
  40. Kumariya, R.; Garsa, A.K.; Rajput, Y.; Sood, S.; Akhtar, N.; Patel, S. Bacteriocins: Classification, synthesis, mechanism of action and resistance development in food spoilage causing bacteria. Microb. Pathog. 2019, 128, 171–177. [Google Scholar] [CrossRef]
  41. Mathur, H.; Rea, M.; Cotter, P.; Hill, C.; Ross, R. The Sactibiotic Subclass of Bacteriocins: An Update. Curr. Protein Pept. Sci. 2015, 16, 549–558. [Google Scholar] [CrossRef]
  42. Mokoena, M.P. Lactic Acid Bacteria and Their Bacteriocins: Classification, Biosynthesis and Applications against Uropathogens: A Mini-Review. Molecules 2017, 22, 1255. [Google Scholar] [CrossRef] [PubMed]
  43. Wiedemann, I.; Breukink, E.; van Kraaij, C.; Kuipers, O.P.; Bierbaum, G.; de Kruijff, B.; Sahl, H.-G. Specific Binding of Nisin to the Peptidoglycan Precursor Lipid II Combines Pore Formation and Inhibition of Cell Wall Biosynthesis for Potent Antibiotic Activity. J. Biol. Chem. 2001, 276, 1772–1779. [Google Scholar] [CrossRef]
  44. Hsu, S.-T.D.; Breukink, E.; Tischenko, E.; Lutters, M.A.G.; De Kruijff, B.; Kaptein, R.; Bonvin, A.M.; Van Nuland, N.A. The nisin–lipid II complex reveals a pyrophosphate cage that provides a blueprint for novel antibiotics. Nat. Struct. Mol. Biol. 2004, 11, 963–967. [Google Scholar] [CrossRef]
  45. Brötz, H.; Bierbaum, G.; Leopold, K.; Reynolds, P.E.; Sahl, H.-G. The Lantibiotic Mersacidin Inhibits Peptidoglycan Synthesis by Targeting Lipid II. Antimicrob. Agents Chemother. 1998, 42, 154–160. [Google Scholar] [CrossRef]
  46. Wiedemann, I.; Bottiger, T.; Bonelli, R.R.; Wiese, A.; Hagge, S.O.; Gutsmann, T.; Seydel, U.; Deegan, L.; Hill, C.; Ross, P.; et al. The mode of action of the lantibiotic lacticin 3147—A complex mechanism involving specific interaction of two peptides and the cell wall precursor lipid II. Mol. Microbiol. 2006, 61, 285–296. [Google Scholar] [CrossRef] [PubMed]
  47. Bouhss, A.; Al-Dabbagh, B.; Vincent, M.; Odaert, B.; Aumont-Nicaise, M.; Bressolier, P.; Desmadril, M.; Mengin-Lecreulx, D.; Urdaci, M.C.; Gallay, J. Specific Interactions of Clausin, a New Lantibiotic, with Lipid Precursors of the Bacterial Cell Wall. Biophys. J. 2009, 97, 1390–1397. [Google Scholar] [CrossRef]
  48. Van Staden, A.D.P. In Vitro and In Vivo Characterization of Amyloliquecidin, a Novel Two-Component Lantibiotic Produced by Bacillus Amyloliquefaciens; Stellenbosch University: Stellenbosch, South Africa, 2015. [Google Scholar]
  49. Cleveland, J.; Montville, T.J.; Nes, I.F.; Chikindas, M.L. Bacteriocins: Safe, natural antimicrobials for food preservation. Int. J. Food Microbiol. 2001, 71, 1–20. [Google Scholar] [CrossRef] [PubMed]
  50. Perez, R.H.; Zendo, T.; Sonomoto, K. Novel bacteriocins from lactic acid bacteria (LAB): Various structures and applications. Microb. Cell Factories 2014, 13, S3. [Google Scholar] [CrossRef]
  51. Jiang, H.; Zou, J.; Cheng, H.; Fang, J.; Huang, G. Purification, Characterization, and Mode of Action of Pentocin JL-1, a Novel Bacteriocin Isolated from Lactobacillus pentosus, against Drug-Resistant Staphylococcus aureus. BioMed Res. Int. 2017, 2017, 7657190. [Google Scholar] [CrossRef]
  52. Soltani, S.; Hammami, R.; Cotter, P.D.; Rebuffat, S.; Said, L.B.; Gaudreau, H.; Bédard, F.; Biron, E.; Drider, D.; Fliss, I. Bacteriocins as a new generation of antimicrobials: Toxicity aspects and regulations. FEMS Microbiol. Rev. 2021, 45, fuaa039. [Google Scholar] [CrossRef]
  53. Newstead, L.L.; Varjonen, K.; Nuttall, T.; Paterson, G.K. Staphylococcal-Produced Bacteriocins and Antimicrobial Peptides: Their Potential as Alternative Treatments for Staphylococcus aureus Infections. Antibiotics 2020, 9, 40. [Google Scholar] [CrossRef] [PubMed]
  54. Cox, C.R.; Coburn, P.S.; Gilmore, M.S. Enterococcal Cytolysin: A Novel Two Component Peptide System that Serves as a Bacterial Defense Against Eukaryotic and Prokaryotic Cells. Curr. Protein Pept. Sci. 2005, 6, 77–84. [Google Scholar] [CrossRef] [PubMed]
  55. Vaucher, R.d.A.; Gewehr, C.d.C.V.; Correa, A.P.F.; Sant‘anna, V.; Ferreira, J.; Brandelli, A. Evaluation of the immunogenicity and in vivo toxicity of the antimicrobial peptide P34. Int. J. Pharm. 2011, 421, 94–98. [Google Scholar] [CrossRef] [PubMed]
  56. Hols, P.; Ledesma-García, L.; Gabant, P.; Mignolet, J. Mobilization of Microbiota Commensals and Their Bacteriocins for Therapeutics. Trends Microbiol. 2019, 27, 690–702. [Google Scholar] [CrossRef] [PubMed]
  57. Mesa-Pereira, B.; Rea, M.C.; Cotter, P.D.; Hill, C.; Ross, R.P. Heterologous Expression of Biopreservative Bacteriocins with a View to Low Cost Production. Front. Microbiol. 2018, 9, 1654. [Google Scholar] [CrossRef] [PubMed]
  58. Field, D.; Gaudin, N.; Lyons, F.; O’Connor, P.M.; Cotter, P.D.; Hill, C.; Ross, R.P. A Bioengineered Nisin Derivative to Control Biofilms of Staphylococcus pseudintermedius. PLoS ONE 2015, 10, e0119684. [Google Scholar] [CrossRef]
  59. Field, D.; Connor, P.M.O.; Cotter, P.D.; Hill, C.; Ross, R.P. The generation of nisin variants with enhanced activity against specific Gram-positive pathogens. Mol. Microbiol. 2008, 69, 218–230. [Google Scholar] [CrossRef]
  60. Sabo, S.D.S.; Lopes, A.M.; Santos-Ebinuma, V.D.C.; Rangel-Yagui, C.D.O.; Oliveira, R.P.D.S. Bacteriocin partitioning from a clarified fermentation broth of Lactobacillus plantarum ST16Pa in aqueous two-phase systems with sodium sulfate and choline-based salts as additives. Process. Biochem. 2018, 66, 212–221. [Google Scholar] [CrossRef]
  61. da Silva Sabo, S.; Vitolo, M.; González, J.M.D.; de Souza Oliveira, R.P. Overview of Lactobacillus plantarum as a promising bacteriocin producer among lactic acid bacteria. Food Res. Int. 2014, 64, 527–536. [Google Scholar] [CrossRef]
  62. Guyonnet, D.; Fremaux, C.; Cenatiempo, Y.; Berjeaud, J.M. Method for Rapid Purification of Class IIa Bacteriocins and Comparison of Their Activities. Appl. Environ. Microbiol. 2000, 66, 1744–1748. [Google Scholar] [CrossRef]
  63. Deshpande, A.; Pasupuleti, V.; Thota, P.; Pant, C.; Rolston, D.D.K.; Sferra, T.J.; Hernandez, A.V.; Donskey, C.J. Community-associated Clostridium difficile infection and antibiotics: A meta-analysis. J. Antimicrob. Chemother. 2013, 68, 1951–1961. [Google Scholar] [CrossRef] [PubMed]
  64. Wilcox, M.H.; Mooney, L.; Bendall, R.; Settle, C.D.; Fawley, W.N. A case-control study of community-associated Clostridium difficile infection. J. Antimicrob. Chemother. 2008, 62, 388–396. [Google Scholar] [CrossRef] [PubMed]
  65. Kollef, M.H. Broad-Spectrum Antimicrobials and the Treatment of Serious Bacterial Infections: Getting It Right up Front. Clin. Infect. Dis. 2008, 47 (Suppl. 1), S3–S13. [Google Scholar] [CrossRef]
  66. Kollef, M.H.; Sherman, G.; Ward, S.; Fraser, V.J. Inadequate Antimicrobial Treatment of Infections. Chest 1999, 115, 462–474. [Google Scholar] [CrossRef]
  67. Mitchell, D.H.; Howden, B.P. Diagnosis and management of Staphylococcus aureus bacteraemia. Intern. Med. J. 2005, 35, S17–S24. [Google Scholar] [CrossRef]
  68. Rhee, C.; Kadri, S.S.; Dekker, J.P.; Danner, R.L.; Chen, H.-C.; Fram, D.; Zhang, F.; Wang, R.; Klompas, M.; For The Cdc Prevention Epicenters Program. Prevalence of Antibiotic-Resistant Pathogens in Culture-Proven Sepsis and Outcomes Associated With Inadequate and Broad-Spectrum Empiric Antibiotic Use. JAMA Netw. Open 2020, 3, e202899. [Google Scholar] [CrossRef]
  69. Barbosa, A.A.T.; de Melo, M.R.; da Silva, C.M.R.; Jain, S.; Dolabella, S.S. Nisin resistance in Gram-positive bacteria and approaches to circumvent resistance for successful therapeutic use. Crit. Rev. Microbiol. 2021, 47, 376–385. [Google Scholar] [CrossRef]
  70. Van Schaik, W.; Gahan, C.G.; Hill, C. Acid-adapted Listeria monocytogenes displays enhanced tolerance against the lantibiotics nisin and lacticin 3147. J. Food Prot. 1999, 62, 536–539. [Google Scholar] [CrossRef]
  71. Mantovani, H.C.; Russell, J.B. Nisin Resistance of Streptococcus bovis. Appl. Environ. Microbiol. 2001, 67, 808–813. [Google Scholar] [CrossRef] [PubMed]
  72. de Freire Bastos, M.D.C.; Coelho, M.L.V.; Da Silva Santos, O.C. Resistance to bacteriocins produced by Gram-positive bacteria. Microbiology 2015, 161, 683–700. [Google Scholar] [CrossRef]
  73. Jarvis, B. Resistance to Nisin and Production of Nisin-Inactivating Enzymes by Several Bacillus Species. J. Gen. Microbiol. 1967, 47, 33–48. [Google Scholar] [CrossRef]
  74. Collins, B.; Guinane, C.M.; Cotter, P.D.; Hill, C.; Ross, R.P. Assessing the Contributions of the LiaS Histidine Kinase to the Innate Resistance of Listeria monocytogenes to Nisin, Cephalosporins, and Disinfectants. Appl. Environ. Microbiol. 2012, 78, 2923–2929. [Google Scholar] [CrossRef]
  75. Dicks, L.M.T.; Dreyer, L.; Smith, C.; Van Staden, A.D. A Review: The Fate of Bacteriocins in the Human Gastro-Intestinal Tract: Do They Cross the Gut–Blood Barrier? Front. Microbiol. 2018, 9, 2297. [Google Scholar] [CrossRef]
  76. Meade, E.; Slattery, M.A.; Garvey, M. Bacteriocins, Potent Antimicrobial Peptides and the Fight against Multi Drug Resistant Species: Resistance is Futile? Antibiotics 2020, 9, 32. [Google Scholar] [CrossRef] [PubMed]
  77. Walsh, L.; Johnson, C.N.; Hill, C.; Ross, R.P. Efficacy of Phage- and Bacteriocin-Based Therapies in Combatting Nosocomial MRSA Infections. Front. Mol. Biosci. 2021, 8, 654038. [Google Scholar] [CrossRef] [PubMed]
  78. Blake, K.L.; Randall, C.P.; O’Neill, A.J. In Vitro Studies Indicate a High Resistance Potential for the Lantibiotic Nisin in Staphylococcus aureus and Define a Genetic Basis for Nisin Resistance. Antimicrob. Agents Chemother. 2011, 55, 2362–2368. [Google Scholar] [CrossRef] [PubMed]
  79. Qin, Y.; Wang, Y.; He, Y.; Zhang, Y.; She, Q.; Chai, Y.; Li, P.; Shang, Q. Characterization of Subtilin L-Q11, a Novel Class I Bacteriocin Synthesized by Bacillus subtilis L-Q11 Isolated From Orchard Soil. Front. Microbiol. 2019, 10, 484. [Google Scholar] [CrossRef] [PubMed]
  80. Zhao, R.; Lu, Y.; Ran, J.; Li, G.; Lei, S.; Zhu, Y.; Xu, B. Purification and characterization of bacteriocin produced by Lactobacillus rhamnosus zrx01. Food Biosci. 2020, 38, 100754. [Google Scholar] [CrossRef]
  81. Ghobrial, O.; Derendorf, H.; Hillman, J.D. Human serum binding and its effect on the pharmacodynamics of the lantibiotic MU1140. Eur. J. Pharm. Sci. 2010, 41, 658–664. [Google Scholar] [CrossRef]
  82. Ansari, A.; Zohra, R.R.; Tarar, O.M.; Qader, S.A.U.; Aman, A. Screening, purification and characterization of thermostable, protease resistant Bacteriocin active against methicillin resistant Staphylococcus aureus (MRSA). BMC Microbiol. 2018, 18, 192. [Google Scholar] [CrossRef]
  83. Twomey, D.; Ross, R.P.; Ryan, M.; Meaney, B.; Hill, C. Lantibiotics produced by lactic acid bacteria: Structure, function and applications. Antonie Van Leeuwenhoek 2002, 82, 165–185. [Google Scholar] [CrossRef] [PubMed]
  84. Sulthana, R.; Archer, A. Bacteriocin nanoconjugates: Boon to medical and food industry. J. Appl. Microbiol. 2020, 131, 1056–1071. [Google Scholar] [CrossRef] [PubMed]
  85. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
  86. Tiwari, J.N.; Tiwari, R.N.; Kim, K.S. Zero-dimensional, one-dimensional, two-dimensional and three-dimensional nanostructured materials for advanced electrochemical energy devices. Prog. Mater. Sci. 2012, 57, 724–803. [Google Scholar] [CrossRef]
  87. Zohri, M.; Alavidjeh, M.S.; Mirdamadi, S.S.; Behmadi, H.; Nasr, S.M.H.; Gonbaki, S.E.; Ardestani, M.S.; Arabzadeh, A.J. Nisin-Loaded Chitosan/Alginate Nanoparticles: A Hopeful Hybrid Biopreservative. J. Food Saf. 2013, 33, 40–49. [Google Scholar] [CrossRef]
  88. Zohri, M.; Alavidjeh, M.S.; Haririan, I.; Ardestani, M.S.; Ebrahimi, S.E.S.; Sani, H.T.; Sadjadi, S.K. A Comparative Study Between the Antibacterial Effect of Nisin and Nisin-Loaded Chitosan/Alginate Nanoparticles on the Growth of Staphylococcus aureus in Raw and Pasteurized Milk Samples. Probiotics Antimicrob. Proteins 2010, 2, 258–266. [Google Scholar] [CrossRef]
  89. Wang, H.; She, Y.; Chu, C.; Liu, H.; Jiang, S.; Sun, M.; Jiang, S. Preparation, antimicrobial and release behaviors of nisin-poly (vinyl alcohol)/wheat gluten/ZrO2 nanofibrous membranes. J. Mater. Sci. 2015, 50, 5068–5078. [Google Scholar] [CrossRef]
  90. Heunis, T.D.J.; Smith, C.; Dicks, L.M.T. Evaluation of a Nisin-Eluting Nanofiber Scaffold To Treat Staphylococcus aureus-Induced Skin Infections in Mice. Antimicrob. Agents Chemother. 2013, 57, 3928–3935. [Google Scholar] [CrossRef]
  91. Liu, Y.; Liu, Y.; Du, Z.; Zhang, L.; Chen, J.; Shen, Z.; Liu, Q.; Qin, J.; Lv, H.; Wang, H.; et al. Skin microbiota analysis-inspired development of novel anti-infectives. Microbiome 2020, 8, 85. [Google Scholar] [CrossRef]
  92. Miller, L.G.; Eisenberg, D.F.; Liu, H.; Chang, C.-L.; Wang, Y.; Luthra, R.; Wallace, A.; Fang, C.; Singer, J.; Suaya, J.A. Incidence of skin and soft tissue infections in ambulatory and inpatient settings, 2005–2010. BMC Infect. Dis. 2015, 15, 1–8. [Google Scholar] [CrossRef]
  93. Esposito, S.; Noviello, S.; Leone, S. Epidemiology and microbiology of skin and soft tissue infections. Curr. Opin. Infect. Dis. 2016, 29, 109–115. [Google Scholar] [CrossRef] [PubMed]
  94. Stevens, D.L.; Bisno, A.L.; Chambers, H.F.; Dellinger, E.P.; Goldstein, E.J.C.; Gorbach, S.L.; Hirschmann, J.; Kaplan, S.L.; Montoya, J.G.; Wade, J.C.; et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2014, 59, e10–e52. [Google Scholar] [CrossRef]
  95. Esposito, S.; Bassetti, M.; Concia, E.; De Simone, G.; De Rosa, F.G.; Grossi, P.; Novelli, A.; Menichetti, F.; Petrosillo, N.; Tinelli, M.; et al. Diagnosis and management of skin and soft-tissue infections (SSTI). A literature review and consensus statement: An update. J. Chemother. 2017, 29, 197–214. [Google Scholar] [CrossRef] [PubMed]
  96. Watkins, R.R.; David, M.Z. Approach to the Patient with a Skin and Soft Tissue Infection. Infect. Dis. Clin. North Am. 2021, 35, 1–48. [Google Scholar] [CrossRef] [PubMed]
  97. Staden, A.D.P.v.; Heunis, T.; Smith, C.; Deane, S.; Dicks, L.M.T. Efficacy of Lantibiotic Treatment of Staphylococcus aureus-Induced Skin Infections, Monitored by In Vivo Bioluminescent Imaging. Antimicrob. Agents Chemother. 2016, 60, 3948–3955. [Google Scholar] [CrossRef] [PubMed]
  98. Ovchinnikov, K.V.; Kranjec, C.; Thorstensen, T.; Carlsen, H.; Diep, D.B. Successful Development of Bacteriocins into Therapeutic Formulation for Treatment of MRSA Skin Infection in a Murine Model. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef]
  99. Zipperer, A.; Konnerth, M.C.; Laux, C.; Berscheid, A.; Janek, D.; Weidenmaier, C.; Burian, M.; Schilling, N.A.; Slavetinsky, C.; Marschal, M.; et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature 2016, 535, 511–516. [Google Scholar] [CrossRef]
  100. Francis, J.S.; Doherty, M.C.; Lopatin, U.; Johnston, C.P.; Sinha, G.; Ross, T.; Cai, M.; Hansel, N.N.; Perl, T.; Ticehurst, J.R.; et al. Severe Community-Onset Pneumonia in Healthy Adults Caused by Methicillin-Resistant Staphylococcus aureus Carrying the Panton-Valentine Leukocidin Genes. Clin. Infect. Dis. 2005, 40, 100–107. [Google Scholar] [CrossRef]
  101. Kalil, A.C.; Metersky, M.L.; Klompas, M.; Muscedere, J.; Sweeney, D.A.; Palmer, L.B.; Napolitano, L.M.; O’Grady, N.P.; Bartlett, J.G.; Carratala, J.; et al. Management of Adults With Hospital-acquired and Ventilator-associated Pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin. Infect. Dis. 2016, 63, e61–e111. [Google Scholar] [CrossRef]
  102. Jiang, H.; Tang, R.-N.; Wang, J. Linezolid versus vancomycin or teicoplanin for nosocomial pneumonia: Meta-analysis of randomised controlled trials. Eur. J. Clin. Microbiol. Infect. Dis. 2013, 32, 1121–1128. [Google Scholar] [CrossRef]
  103. Boakes, S.; Weiss, W.J.; Vinson, M.; Wadman, S.; Dawson, M.J. Antibacterial activity of the novel semisynthetic lantibiotic NVB333 in vitro and in experimental infection models. J. Antibiot. 2016, 69, 850–857. [Google Scholar] [CrossRef]
  104. De Kwaadsteniet, M.; Doeschate, K.; Dicks, L. Nisin F in the treatment of respiratory tract infections caused by Staphylococcus aureus. Lett. Appl. Microbiol. 2009, 48, 65–70. [Google Scholar] [CrossRef] [PubMed]
  105. de Kwaadsteniet, M.; Doeschate, K.T.; Dicks, L.M.T. Characterization of the Structural Gene Encoding Nisin F, a New Lantibiotic Produced by a Lactococcus lactis subsp. lactis Isolate from Freshwater Catfish (Clarias gariepinus). Appl. Environ. Microbiol. 2008, 74, 547–549. [Google Scholar] [CrossRef] [PubMed]
  106. Ghobrial, O.G.; Derendorf, H.; Hillman, J.D. Pharmacokinetic and pharmacodynamic evaluation of the lantibiotic MU1140. J. Pharm. Sci. 2010, 99, 2521–2528. [Google Scholar] [CrossRef] [PubMed]
  107. Geng, M.; Ravichandran, A.; Escano, J.; Smith, L. Efficacious Analogs of the Lantibiotic Mutacin 1140 against a Systemic Methicillin-Resistant Staphylococcus aureus Infection. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef]
  108. Bastos, M.D.C.D.F.; Coutinho, B.G.; Coelho, M.L.V. Lysostaphin: A Staphylococcal Bacteriolysin with Potential Clinical Applications. Pharmaceuticals 2010, 3, 1139–1161. [Google Scholar] [CrossRef]
  109. Oluola, O.; Kong, L.; Fein, M.; Weisman, L.E. Lysostaphin in Treatment of Neonatal Staphylococcus aureus Infection. Antimicrob. Agents Chemother. 2007, 51, 2198–2200. [Google Scholar] [CrossRef]
  110. Placencia, F.X.; Kong, L.; E Weisman, L. Treatment of Methicillin-Resistant Staphylococcus aureus in Neonatal Mice: Lysostaphin Versus Vancomycin. Pediatr. Res. 2009, 65, 420–424. [Google Scholar] [CrossRef]
  111. Lee, N.-K.; Park, Y.-L.; Kim, H.-W.; Park, Y.-H.; Rhim, S.-L.; Kim, J.-M.; Kim, J.-M.; Nam, H.-M.; Jung, S.-C.; Paik, H.-D. Purification and Characterization of Lacticin NK34 Produced by Lactococcus lactis NK34 against Bovine Mastitis. Korean J. Food Sci. Anim. Resour. 2008, 28, 457–462. [Google Scholar] [CrossRef]
  112. Kim, S.; Shin, S.; Koo, H.; Youn, J.-H.; Paik, H.-D.; Park, Y. In vitro antimicrobial effect and in vivo preventive and therapeutic effects of partially purified lantibiotic lacticin NK34 against infection by Staphylococcus species isolated from bovine mastitis. J. Dairy Sci. 2010, 93, 3610–3615. [Google Scholar] [CrossRef]
  113. Kavanagh, N.; Ryan, E.J.; Widaa, A.; Sexton, G.; Fennell, J.; O’Rourke, S.; Cahill, K.C.; Kearney, C.J.; O’Brien, F.J.; Kerrigan, S.W. Staphylococcal Osteomyelitis: Disease Progression, Treatment Challenges, and Future Directions. Clin. Microbiol. Rev. 2018, 31, e00084-17. [Google Scholar] [CrossRef]
  114. Van de Belt, H.; Neut, D.; Schenk, W.; van Horn, J.R.; van der Mei, H.C.; Busscher, H.J. Infection of orthopedic implants and the use of antibiotic-loaded bone cements: A review. Acta Orthop. Scand. 2001, 72, 557–571. [Google Scholar] [CrossRef] [PubMed]
  115. Parvizi, J.; Saleh, K.J.; Ragland, P.S.; Pour, A.E.; Mont, M.A. Efficacy of antibiotic-impregnated cement in total hip replacement. Acta Orthop. 2008, 79, 335–341. [Google Scholar] [CrossRef] [PubMed]
  116. van Staden, A.; Brand, A.; Dicks, L. Nisin F-loaded brushite bone cement prevented the growth of Staphylococcus aureus in vivo. J. Appl. Microbiol. 2012, 112, 831–840. [Google Scholar] [CrossRef] [PubMed]
  117. Murray, R.J. Staphylococcus aureus infective endocarditis: Diagnosis and management guidelines. Intern. Med. J. 2005, 35, S25–S44. [Google Scholar] [CrossRef]
  118. Castiglione, F.; Lazzarini, A.; Carrano, L.; Corti, E.; Ciciliato, I.; Gastaldo, L.; Candiani, P.; Losi, D.; Marinelli, F.; Selva, E.; et al. Determining the Structure and Mode of Action of Microbisporicin, a Potent Lantibiotic Active Against Multiresistant Pathogens. Chem. Biol. 2008, 15, 22–31. [Google Scholar] [CrossRef]
  119. Jabés, D.; Brunati, C.; Candiani, G.; Riva, S.; Romanó, G.; Donadio, S. Efficacy of the New Lantibiotic NAI-107 in Experimental Infections Induced by Multidrug-Resistant Gram-Positive Pathogens. Antimicrob. Agents Chemother. 2011, 55, 1671–1676. [Google Scholar] [CrossRef]
  120. Wertheim, H.F.; Melles, D.C.; Vos, M.C.; van Leeuwen, W.; van Belkum, A.; Verbrugh, H.A.; Nouwen, J.L. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect. Dis. 2005, 5, 751–762. [Google Scholar] [CrossRef]
  121. Bode, L.G.M.; Kluytmans, J.A.J.W.; Wertheim, H.F.L.; Bogaers, D.; Vandenbroucke-Grauls, C.M.J.E.; Roosendaal, R.; Troelstra, A.; Box, A.T.A.; Voss, A.; van der Tweel, I.; et al. Preventing Surgical-Site Infections in Nasal Carriers of Staphylococcus aureus. N. Engl. J. Med. 2010, 362, 9–17. [Google Scholar] [CrossRef]
  122. Muñoz, P.; Hortal, J.; Giannella, M.; Barrio, J.; Rodríguez-Créixems, M.; Pérez, M.; Rincón, C.; Bouza, E. Nasal carriage of S. aureus increases the risk of surgical site infection after major heart surgery. J. Hosp. Infect. 2008, 68, 25–31. [Google Scholar] [CrossRef]
  123. von Eiff, C.; Becker, K.; Machka, K.; Stammer, H.; Peters, G. Nasal Carriage as a Source of Staphylococcus aureus Bacteremia. N. Engl. J. Med. 2001, 344, 11–16. [Google Scholar] [CrossRef]
  124. Lee, A.S.; Macedo-Vinas, M.; François, P.; Renzi, G.; Schrenzel, J.; Vernaz, N.; Pittet, D.; Harbarth, S. Impact of Combined Low-Level Mupirocin and Genotypic Chlorhexidine Resistance on Persistent Methicillin-Resistant Staphylococcus aureus Carriage After Decolonization Therapy: A Case-control Study. Clin. Infect. Dis. 2011, 52, 1422–1430. [Google Scholar] [CrossRef]
  125. Hetem, D.; Bonten, M. Clinical relevance of mupirocin resistance in Staphylococcus aureus. J. Hosp. Infect. 2013, 85, 249–256. [Google Scholar] [CrossRef] [PubMed]
  126. Desroches, M.; Potier, J.; Laurent, F.; Bourrel, A.-S.; Doucet-Populaire, F.; Decousser, J.-W.; Archambaud, M.; Aubert, G.; Biendo, M.; Blanchard-Marche, G.; et al. Prevalence of mupirocin resistance among invasive coagulase-negative staphylococci and methicillin-resistant Staphylococcus aureus (MRSA) in France: Emergence of a mupirocin-resistant MRSA clone harbouring mupA. J. Antimicrob. Chemother. 2013, 68, 1714–1717. [Google Scholar] [CrossRef]
  127. Sandiford, S.; Upton, M. Identification, Characterization, and Recombinant Expression of Epidermicin NI01, a Novel Unmodified Bacteriocin Produced by Staphylococcus epidermidis That Displays Potent Activity against Staphylococci. Antimicrob. Agents Chemother. 2012, 56, 1539–1547. [Google Scholar] [CrossRef] [PubMed]
  128. Halliwell, S.; Warn, P.; Sattar, A.; Derrick, J.P.; Upton, M. A single dose of epidermicin NI01 is sufficient to eradicate MRSA from the nares of cotton rats. J. Antimicrob. Chemother. 2016, 72, 778–781. [Google Scholar] [CrossRef]
  129. Niu, W.W.; Neu, H.C. Activity of mersacidin, a novel peptide, compared with that of vancomycin, teicoplanin, and daptomycin. Antimicrob. Agents Chemother. 1991, 35, 998–1000. [Google Scholar] [CrossRef]
  130. Kruszewska, D.; Sahl, H.-G.; Bierbaum, G.; Pag, U.; Hynes, S.O.; Ljungh, Å. Mersacidin eradicates methicillin-resistant Staphylococcus aureus (MRSA) in a mouse rhinitis model. J. Antimicrob. Chemother. 2004, 54, 648–653. [Google Scholar] [CrossRef] [PubMed]
  131. Piewngam, P.; Khongthong, S.; Roekngam, N.; Theapparat, Y.; Sunpaweravong, S.; Faroongsarng, D.; Otto, M. Probiotic for pathogen-specific Staphylococcus aureus decolonisation in Thailand: A phase 2, double-blind, randomised, placebo-controlled trial. Lancet Microbe 2023, 4, e75–e83. [Google Scholar] [CrossRef]
  132. Sassone-Corsi, M.; Nuccio, S.-P.; Liu, H.; Hernandez, D.; Vu, C.T.; Takahashi, A.A.; Edwards, R.A.; Raffatellu, M. Microcins mediate competition among Enterobacteriaceae in the inflamed gut. Nature 2016, 540, 280–283. [Google Scholar] [CrossRef] [PubMed]
  133. Kommineni, S.; Bretl, D.J.; Lam, V.; Chakraborty, R.; Hayward, M.; Simpson, P.; Cao, Y.; Bousounis, P.; Kristich, C.J.; Salzman, N.H. Bacteriocin production augments niche competition by enterococci in the mammalian gastrointestinal tract. Nature 2015, 526, 719–722. [Google Scholar] [CrossRef] [PubMed]
  134. Leyden, J.J.; Marples, R.R.; Kligman, A.M. Staphylococcus aureus in the lesions of atopic dermatitis. Br. J. Dermatol. 1974, 90, 525. [Google Scholar] [CrossRef] [PubMed]
  135. Valenta, C.; Bernkop-Schnürch, A.; Rigler, H.P. The Antistaphylococcal Effect of Nisin in a Suitable Vehicle: A Potential Therapy for Atopic Dermatitis in Man. J. Pharm. Pharmacol. 1996, 48, 988–991. [Google Scholar] [CrossRef] [PubMed]
  136. Joshi, A.A.; Vocanson, M.; Nicolas, J.-F.; Wolf, P.; Patra, V. Microbial derived antimicrobial peptides as potential therapeutics in atopic dermatitis. Front. Immunol. 2023, 14, 1125635. [Google Scholar] [CrossRef]
  137. Nakatsuji, T.; Hata, T.R.; Tong, Y.; Cheng, J.Y.; Shafiq, F.; Butcher, A.M.; Salem, S.S.; Brinton, S.L.; Spergel, A.K.R.; Johnson, K.; et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat. Med. 2021, 27, 700–709. [Google Scholar] [CrossRef]
  138. Williams, M.R.; Costa, S.K.; Zaramela, L.S.; Khalil, S.; Todd, D.A.; Winter, H.L.; Sanford, J.A.; O’neill, A.M.; Liggins, M.C.; Nakatsuji, T.; et al. Quorum sensing between bacterial species on the skin protects against epidermal injury in atopic dermatitis. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef]
  139. Cao, L.; Wu, J.; Xie, F.; Hu, S.; Mo, Y. Efficacy of Nisin in Treatment of Clinical Mastitis in Lactating Dairy Cows. J. Dairy Sci. 2007, 90, 3980–3985. [Google Scholar] [CrossRef]
  140. Lepak, A.J.; Marchillo, K.; Craig, W.A.; Andes, D.R. In Vivo Pharmacokinetics and Pharmacodynamics of the Lantibiotic NAI-107 in a Neutropenic Murine Thigh Infection Model. Antimicrob. Agents Chemother. 2015, 59, 1258–1264. [Google Scholar] [CrossRef] [PubMed]
  141. Karczewski, J.; Brown, C.M.; Maezato, Y.; Krasucki, S.P.; Streatfield, S.J.; Karczewski, J.; Brown, C.M.; Maezato, Y.; Krasucki, S.P.; Streatfield, S.J. Efficacy of a novel lantibiotic, CMB001, against MRSA. J. Antimicrob. Chemother. 2021, 76, 1532–1538. [Google Scholar] [CrossRef]
  142. Twomey, D.; Wheelock, A.; Flynn, J.; Meaney, W.; Hill, C.; Ross, R. Protection Against Staphylococcus aureus Mastitis in Dairy Cows Using a Bismuth-Based Teat Seal Containing the Bacteriocin, Lacticin 3147. J. Dairy Sci. 2000, 83, 1981–1988. [Google Scholar] [CrossRef]
  143. Khan, I.; Oh, D.-H. Integration of nisin into nanoparticles for application in foods. Innov. Food Sci. Emerg. Technol. 2016, 34, 376–384. [Google Scholar] [CrossRef]
  144. Ng, Z.J.; Abu Zarin, M.; Lee, C.K.; Tan, J.S. Application of bacteriocins in food preservation and infectious disease treatment for humans and livestock: A review. RSC Adv. 2020, 10, 38937–38964. [Google Scholar] [CrossRef]
  145. Verma, D.K.; Thakur, M.; Singh, S.; Tripathy, S.; Gupta, A.K.; Baranwal, D.; Patel, A.R.; Shah, N.; Utama, G.L.; Niamah, A.K.; et al. Bacteriocins as antimicrobial and preservative agents in food: Biosynthesis, separation and application. Food Biosci. 2022, 46, 101594. [Google Scholar] [CrossRef]
  146. Negash, A.W.; Tsehai, B.A. Current Applications of Bacteriocin. Int. J. Microbiol. 2020, 2020, 1–7. [Google Scholar] [CrossRef]
  147. Mbandlwa, P.; Doyle, N.; Hill, C.; Stanton, C.; Ross, R.P. Bacteriocins: Novel Applications in Food, and Human and Animal Health, in Encyclopedia of Dairy Sciences, 3rd ed.; McSweeney, P.L.H., McNamara, J.P., Eds.; Academic Press: Oxford, UK, 2022; pp. 46–54. [Google Scholar] [CrossRef]
  148. Rilla, N.; Martínez, B.; Rodriguez, A. Inhibition of a Methicillin-Resistant Staphylococcus aureus Strain in Afuega’l Pitu Cheese by the Nisin Z–Producing Strain Lactococcus lactis subsp. lactis IPLA 729. J. Food Prot. 2004, 67, 928–933. [Google Scholar] [CrossRef] [PubMed]
  149. Grande, M.J.; López, R.L.; Abriouel, H.; Valdivia, E.; Ben Omar, N.; Maqueda, M.; Martínez-Cañamero, M.; Gálvez, A. Treatment of Vegetable Sauces with Enterocin AS-48 Alone or in Combination with Phenolic Compounds To Inhibit Proliferation of Staphylococcus aureus. J. Food Prot. 2007, 70, 405–411. [Google Scholar] [CrossRef] [PubMed]
  150. Muñoz, A.; Ananou, S.; Gálvez, A.; Martínez-Bueno, M.; Rodríguez, A.; Maqueda, M.; Valdivia, E. Inhibition of Staphylococcus aureus in dairy products by enterocin AS-48 produced in situ and ex situ: Bactericidal synergism with heat. Int. Dairy J. 2007, 17, 760–769. [Google Scholar] [CrossRef]
  151. Burgos, M.J.G.; Pulido, R.P.; Aguayo, M.D.C.L.; Gálvez, A.; Lucas, R. The Cyclic Antibacterial Peptide Enterocin AS-48: Isolation, Mode of Action, and Possible Food Applications. Int. J. Mol. Sci. 2014, 15, 22706–22727. [Google Scholar] [CrossRef]
  152. Lauková, A.; Czikková, S.; Dobránsky, T.; Burdová, O. Inhibition ofListeria monocytogenesand Staphylococcus aureus by enterocin CCM 4231 in milk products. Food Microbiol. 1999, 16, 93–99. [Google Scholar] [CrossRef]
  153. An, J.; Zhu, W.; Liu, Y.; Zhang, X.; Sun, L.; Hong, P.; Wang, Y.; Xu, C.; Xu, D.; Liu, H. Purification and characterization of a novel bacteriocin CAMT2 produced by Bacillus amyloliquefaciens isolated from marine fish Epinephelus areolatus. Food Control. 2015, 51, 278–282. [Google Scholar] [CrossRef]
  154. Wu, Y.; An, J.; Liu, Y.; Wang, Y.; Ren, W.; Fang, Z.; Sun, L.; Gooneratne, R. Mode of action of a novel anti-Listeria bacteriocin (CAMT2) produced by Bacillus amyloliquefaciens ZJHD3-06 from Epinephelus areolatus. Arch. Microbiol. 2018, 201, 61–66. [Google Scholar] [CrossRef] [PubMed]
  155. European Centre for Disease Prevention and Control (ECDC); European Food Safety Authority (EFSA); European Medicines Agency (EMA). ECDC/EFSA/EMA second joint report on the integrated analysis of the consumption of antimicrobial agents and occurrence of antimicrobial resistance in bacteria from humans and food-producing animals. EFSA J. 2017, 15, 4872–5007. [Google Scholar] [CrossRef]
  156. Hollis, A.; Ahmed, Z. Preserving Antibiotics, Rationally. N. Engl. J. Med. 2013, 369, 2474–2476. [Google Scholar] [CrossRef]
  157. Muurinen, J.; Richert, J.; Wickware, C.L.; Richert, B.; Johnson, T.A. Swine growth promotion with antibiotics or alternatives can increase antibiotic resistance gene mobility potential. Sci. Rep. 2021, 11, 1–13. [Google Scholar] [CrossRef]
  158. Morris, C.; Helliwell, R.; Raman, S. Framing the agricultural use of antibiotics and antimicrobial resistance in UK national newspapers and the farming press. J. Rural. Stud. 2016, 45, 43–53. [Google Scholar] [CrossRef]
  159. Ben Lagha, A.; Haas, B.; Gottschalk, M.; Grenier, D. Antimicrobial potential of bacteriocins in poultry and swine production. Vet. Res. 2017, 48, 22. [Google Scholar] [CrossRef] [PubMed]
  160. Roy, S.M.; Riley, M.A.; Crabb, J.H. Treating Bovine Mastitis with Nisin: A Model for the Use of Protein Antimicrobials in Veterinary Medicine. In The Bacteriocins: Current Knowledge and Future Prospects; Caister Academic Press: Poole, UK, 2016; pp. 127–140. [Google Scholar] [CrossRef]
  161. Holmes, A.H.; Moore, L.S.P.; Sundsfjord, A.; Steinbakk, M.; Regmi, S.; Karkey, A.; Guerin, P.J.; Piddock, L.J.V. Understanding the mechanisms and drivers of antimicrobial resistance. Lancet 2016, 387, 176–187. [Google Scholar] [CrossRef]
  162. Bradley, A. Bovine Mastitis: An Evolving Disease. Vet. J. 2002, 164, 116–128. [Google Scholar] [CrossRef]
  163. Coelho, M.L.V.; dos Santos Nascimento, J.; Fagundes, P.C.; Madureira, D.J.; de Oliveira, S.S.; de Paiva Brito, M.A.V.; de Freire Bastos, M.D.C. Activity of staphylococcal bacteriocins against Staphylococcus aureus and Streptococcus agalactiae involved in bovine mastitis. Res. Microbiol. 2007, 158, 625–630. [Google Scholar] [CrossRef]
  164. Barboza-Corona, J.E.; de la Fuente-Salcido, N.; Alva-Murillo, N.; Ochoa-Zarzosa, A.; López-Meza, J.E. Activity of bacteriocins synthesized by Bacillus thuringiensis against Staphylococcus aureus isolates associated to bovine mastitis. Vet. Microbiol. 2009, 138, 179–183. [Google Scholar] [CrossRef]
  165. Ryan, M.P.; Meaney, W.J.; Ross, R.P.; Hill, C. Evaluation of Lacticin 3147 and a Teat Seal Containing This Bacteriocin for Inhibition of Mastitis Pathogens. Appl. Environ. Microbiol. 1998, 64, 2287–2290. [Google Scholar] [CrossRef]
  166. Kaur, B.; Balgir, P.P.; Mittu, B.; Kumar, B.; Garg, N. Biomedical Applications of Fermenticin HV6b Isolated from Lactobacillus fermentum HV6b MTCC10770. BioMed Res. Int. 2013, 2013, 1–8. [Google Scholar] [CrossRef]
  167. Ahmad, V.; Khan, M.S.; Jamal, Q.M.S.; Alzohairy, M.A.; Al Karaawi, M.A.; Siddiqui, M.U. Antimicrobial potential of bacteriocins: In therapy, agriculture and food preservation. Int. J. Antimicrob. Agents 2017, 49, 1–11. [Google Scholar] [CrossRef] [PubMed]
  168. Angelopoulou, A.; Field, D.; Ryan, C.A.; Stanton, C.; Hill, C.; Ross, R.P. The microbiology and treatment of human mastitis. Med Microbiol. Immunol. 2018, 207, 83–94. [Google Scholar] [CrossRef] [PubMed]
  169. Jiménez, E.; de Andrés, J.; Manrique, M.; Pareja-Tobes, P.; Tobes, R.; Martínez-Blanch, J.F.; Codoñer, F.M.; Ramón, D.; Fernández, L.; Rodríguez, J.M. Metagenomic Analysis of Milk of Healthy and Mastitis-Suffering Women. J. Hum. Lact. 2015, 31, 406–415. [Google Scholar] [CrossRef] [PubMed]
  170. Inhibition Effects of Probiotics on Pathogens Associated with VAP. Available online: https://ClinicalTrials.gov/show/NCT02928042 (accessed on 1 June 2023).
  171. Alexandre, Y.; Le Berre, R.; Barbier, G.; Le Blay, G. Screening of Lactobacillus spp. for the prevention of Pseudomonas aeruginosa pulmonary infections. BMC Microbiol. 2014, 14, 107. [Google Scholar] [CrossRef]
  172. Functional Ingredients: Effect in Gastrointestinal System. Available online: https://ClinicalTrials.gov/show/NCT02467972 (accessed on 1 June 2023).
  173. Fernández, L.; Delgado, S.; Herrero, H.; Maldonado, A.; Rodríguez, J.M. The Bacteriocin Nisin, an Effective Agent for the Treatment of Staphylococcal Mastitis during Lactation. J. Hum. Lact. 2008, 24, 311–316. [Google Scholar] [CrossRef]
  174. Årdal, C.; Balasegaram, M.; Laxminarayan, R.; McAdams, D.; Outterson, K.; Rex, J.H.; Sumpradit, N. Antibiotic development—Economic, regulatory and societal challenges. Nat. Rev. Microbiol. 2020, 18, 267–274. [Google Scholar] [CrossRef]
  175. Benítez-Chao, D.F.; León-Buitimea, A.; Lerma-Escalera, J.A.; Morones-Ramírez, J.R. Bacteriocins: An Overview of Antimicrobial, Toxicity, and Biosafety Assessment by in vivo Models. Front. Microbiol. 2021, 12. [Google Scholar] [CrossRef]
  176. Hillman, J.D.; Novák, J.; Sagura, E.; Gutierrez, J.A.; Brooks, T.A.; Crowley, P.J.; Hess, M.; Azizi, A.; Leung, K.-P.; Cvitkovitch, D.; et al. Genetic and Biochemical Analysis of Mutacin 1140, a Lantibiotic from Streptococcus mutans. Infect. Immun. 1998, 66, 2743–2749. [Google Scholar] [CrossRef] [PubMed]
  177. Ghobrial, O.G.; Derendorf, H.; Hillman, J.D. Pharmacodynamic activity of the lantibiotic MU1140. Int. J. Antimicrob. Agents 2009, 33, 70–74. [Google Scholar] [CrossRef] [PubMed]
  178. Pokhrel, R.; Bhattarai, N.; Baral, P.; Gerstman, B.S.; Park, J.H.; Handfield, M.; Chapagain, P.P. Molecular mechanisms of pore formation and membrane disruption by the antimicrobial lantibiotic peptide Mutacin 1140. Phys. Chem. Chem. Phys. 2019, 21, 12530–12539. [Google Scholar] [CrossRef] [PubMed]
  179. Hammond, K.; Lewis, H.; Halliwell, S.; Desriac, F.; Nardone, B.; Ravi, J.; Hoogenboom, B.W.; Upton, M.; Derrick, J.P.; Ryadnov, M.G. Flowering Poration—A Synergistic Multi-Mode Antibacterial Mechanism by a Bacteriocin Fold. iScience 2020, 23, 101423. [Google Scholar] [CrossRef]
  180. Breukink, E.; Wiedemann, I.; van Kraaij, C.; Kuipers, O.P.; Sahl, H.-G.; de Kruijff, B. Use of the Cell Wall Precursor Lipid II by a Pore-Forming Peptide Antibiotic. Science 1999, 286, 2361–2364. [Google Scholar] [CrossRef]
  181. Sahl, H.G.; Kordel, M.; Benz, R. Voltage-dependent depolarization of bacterial membranes and artificial lipid bilayers by the peptide antibiotic nisin. Arch. Microbiol. 1987, 149, 120–124. [Google Scholar] [CrossRef]
  182. Mulders, J.W.M.; Boerrigter, I.J.; Rollema, H.S.; Siezen, R.J.; de Vos, W.M. Identification and characterization of the lantibiotic nisin Z, a natural nisin variant. Eur. J. Biochem. 1991, 201, 581–584. [Google Scholar] [CrossRef]
  183. Kaletta, C.; Entian, K.D. Nisin, a peptide antibiotic: Cloning and sequencing of the nisA gene and posttranslational processing of its peptide product. J. Bacteriol. 1989, 171, 1597–1601. [Google Scholar] [CrossRef]
  184. Karczewski, J.; Krasucki, S.P.; Asare-Okai, P.N.; Diehl, C.; Friedman, A.; Brown, C.M.; Maezato, Y.; Streatfield, S.J. Isolation, Characterization and Structure Elucidation of a Novel Lantibiotic from Paenibacillus sp. Front. Microbiol. 2020, 11. [Google Scholar] [CrossRef]
  185. Ovchinnikov, K.V.; Chi, H.; Mehmeti, I.; Holo, H.; Nes, I.F.; Diep, D.B. Novel Group of Leaderless Multipeptide Bacteriocins from Gram-Positive Bacteria. Appl. Environ. Microbiol. 2016, 82, 5216–5224. [Google Scholar] [CrossRef] [PubMed]
  186. Ciufolini, M.A.; Lefranc, D. Micrococcin P1: Structure, biology and synthesis. Nat. Prod. Rep. 2010, 27, 330–342. [Google Scholar] [CrossRef]
  187. Heinrich, P.; Rosenstein, R.; Böhmer, M.; Sonner, P.; Götz, F. The molecular organization of the lysostaphin gene and its sequences repeated in tandem. Mol. Genet. Genom. 1987, 209, 563–569. [Google Scholar] [CrossRef] [PubMed]
  188. A Recsei, P.; Gruss, A.D.; Novick, R.P. Cloning, sequence, and expression of the lysostaphin gene from Staphylococcus simulans. Proc. Natl. Acad. Sci. 1987, 84, 1127–1131. [Google Scholar] [CrossRef] [PubMed]
  189. Bierbaum, G.; Brã¶Tz, H.; Koller, K.-P.; Sahl, H.-G. Cloning, sequencing and production of the lantibiotic mersacidin. FEMS Microbiol. Lett. 1995, 127, 121–126. [Google Scholar] [CrossRef] [PubMed]
  190. Martin, N.I.; Sprules, T.; Carpenter, M.R.; Cotter, P.D.; Hill, C.; Ross, R.P.; Vederas, J.C. Structural Characterization of Lacticin 3147, a Two-Peptide Lantibiotic with Synergistic Activity. Biochemistry 2004, 43, 3049–3056. [Google Scholar] [CrossRef]
Figure 1. Emergence of antibiotic-resistant S. aureus strains.
Figure 1. Emergence of antibiotic-resistant S. aureus strains.
Antibiotics 12 01256 g001
Table 1. In vivo experiments evaluating the efficacy of bacteriocins in S. aureus infections.
Table 1. In vivo experiments evaluating the efficacy of bacteriocins in S. aureus infections.
BacteriocinProducing StrainTarget StrainsModel OrganismIn Vivo DemonstrationReference
Mutacin 1140Streptococcus mutansS. aureus (ATCC
25923 and ATCC 33591)
BALB/c mouseSystemic infection[107]
Epidermicin NI01S. epidermidisMRSA ATCC 43300SPF Cotton ratNasal carriage[128]
Nisin (Nisin A)Lactococcus lactisS. aureus Xen36Nude mouseSkin infection[97]
BALB/c mouse[90]
Nisin FLactococcus lactis F10S. aureus K
S. aureus Xen36
Wistar rat
BALB/c mouse
Respiratory infection
Osteomyelitis
[104,116]
Nisin ZLactococcus lactisUndetermined S. aureus strainsCowMastitis[139]
NAI-107Microbispora ATCC PTA-5024S. aureus 1524
S. aureus (ATCC 29213, USA200, 307109, MW2, USA300, ATCC 25923, 6538P, Smith, WIS-1)
SD rat
ICR mouse
Endocarditis[119]
Intramuscular infection[140]
CMB001 Paenibacillus kyungheensisS. aureus USA300ICR mouseIntramuscular infection[141]
Garvicin KSLactococcus garvieae KS1546S. aureus Xen31BALB/c mouseSkin infection[98]
Micrococcin P1Staphylococcus equorum WS 2733S. aureus Xen31BALB/c mouseSkin infection[98]
LysostaphinStaphylococcus simulansS. aureus USA300Neonatal Wistar mouse
Neonatal FVB mouse
Systemic infection
Systemic infection
[109]
[110]
NVB333Actinoplanes liguriaeS. aureus UNT103-3, S. aureus ATCC 33591, MRSA UNT084-3SPF CD-1 mouseIntramuscular infection
Respiratory infection
[103]
ClausinAlkalihalobacillus clausiiS. aureus Xen36Nude mouseSkin Infection[97]
AmyloliquecidinBacillus velezensisS. aureus Xen36Nude mouseSkin infection[97]
MersacidinBacillus sp. strain HIL Y-85,54728.S. aureus 99308BALB/c mouseNasal carriage[130]
Lacticin NK34Lactococcus lactisS. aureus 69ICR mouseSystemic infection[112]
Lacticin 3147Lactococcus lactis subsp. lactis DPC3147Undetermined S. aureus strainsCowMastitis[142]
LugduninStaphylococcus lugunensisS. aureus USA3000Cotton ratSkin infection[99]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Heinzinger, L.R.; Pugh, A.R.; Wagner, J.A.; Otto, M. Evaluating the Translational Potential of Bacteriocins as an Alternative Treatment for Staphylococcus aureus Infections in Animals and Humans. Antibiotics 2023, 12, 1256. https://doi.org/10.3390/antibiotics12081256

AMA Style

Heinzinger LR, Pugh AR, Wagner JA, Otto M. Evaluating the Translational Potential of Bacteriocins as an Alternative Treatment for Staphylococcus aureus Infections in Animals and Humans. Antibiotics. 2023; 12(8):1256. https://doi.org/10.3390/antibiotics12081256

Chicago/Turabian Style

Heinzinger, Lauren R., Aaron R. Pugh, Julie A. Wagner, and Michael Otto. 2023. "Evaluating the Translational Potential of Bacteriocins as an Alternative Treatment for Staphylococcus aureus Infections in Animals and Humans" Antibiotics 12, no. 8: 1256. https://doi.org/10.3390/antibiotics12081256

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

Article Metrics

Back to TopTop