Fact-checked by Grok 2 weeks ago

Bacteriocin

Bacteriocins are ribosomally synthesized or proteins produced by various , primarily to inhibit or kill closely related or similar bacterial strains as a natural competitive strategy in microbial ecosystems. These compounds are typically small, ranging from less than 5 to over 30 kDa, and exhibit specificity toward target while the producing strain is protected by self-immunity mechanisms. First identified in the early through studies on colicins from , bacteriocins have since been recognized for their role in bacterial antagonism and their potential as alternatives to traditional antibiotics. Bacteriocins are produced by a wide array of , including Gram-positive species like (Lactococcus lactis, Lactobacillus plantarum) and Gram-negative ones such as , with production occurring during late-exponential to early-stationary growth phases under conditions influenced by nutrient availability and cell density. They are synthesized via ribosomal pathways, often encoded on plasmids or chromosomal genes, and secreted extracellularly through dedicated transporters like ATP-binding cassette (ABC) systems or the general pathway, sometimes involving post-translational modifications such as lanthionine formation. systems vary but commonly divide them into three main classes: Class I (modified peptides like lantibiotics, e.g., , <5 kDa and heat-stable), Class II (unmodified or minimally modified small peptides, <10 kDa, heat-stable, subdivided into pediocin-like, two-peptide, and circular types), and Class III (larger, heat-labile proteins >30 kDa, either lytic or non-lytic). Recent updates propose a more detailed framework with 12 subclasses for Class I ribosomally synthesized and post-translationally modified peptides (RiPPs) and three for Class II. The mechanisms of action for bacteriocins primarily involve targeting the cell envelope of susceptible , such as forming pores in cytoplasmic membranes leading to ion leakage and , binding to lipid II to inhibit synthesis, or disrupting essential cellular processes like protein synthesis. These actions are often receptor-mediated, with specificity determined by ionic interactions or docking to surface proteins, rendering them effective against Gram-positive pathogens like and , though some, like microcins, target Gram-negatives. In applications, bacteriocins serve as biopreservatives in food industries (e.g., , approved by the FAO/WHO in 1969 for use in over 50 countries), therapeutics against multidrug-resistant infections such as MRSA, and or immunomodulators in for conditions like bovine . Emerging research highlights their prospects in for , aquaculture growth promotion, and microbiome , with over 8,000 publications underscoring their growing impact by 2023.

Overview

Definition and Characteristics

Bacteriocins are ribosomally synthesized or proteins produced by , which exhibit bactericidal or bacteriostatic activity primarily against closely related strains through a narrow of action. These molecules are secreted extracellularly and often include dedicated immunity mechanisms that protect the producing bacterium from self-killing. Key characteristics of bacteriocins include a wide range of molecular weights, typically under 5 for smaller peptide forms and exceeding 5–10 for larger protein variants, influencing their and . Many bacteriocins demonstrate heat , particularly the smaller peptides that retain activity after exposure to temperatures up to 100°C for short durations, whereas larger proteins tend to be more heat-labile. They often possess cationic and amphiphilic properties, enhancing solubility in aqueous environments and facilitating interaction with target bacterial membranes. Post-translational modifications, such as the formation of lanthionine bridges in certain peptides, contribute to their structural rigidity and potency. In contrast to broad-spectrum antibiotics, which are typically secondary metabolites derived from fungi or semi-synthetic sources and affect a wide range of microorganisms, bacteriocins display high specificity for bacterial competitors, frequently employing receptor-mediated uptake and mechanisms that minimize off-target effects. This targeted nature reduces the likelihood of widespread development compared to traditional antibiotics. From an evolutionary perspective, bacteriocins play a crucial role in microbial ecology by serving as weapons in chemical warfare among bacterial populations, enabling producers to secure ecological niches, limit competitor growth, and modulate community dynamics for resource acquisition.

Historical Discovery

The discovery of bacteriocins began in 1925 when Belgian microbiologist André Gratia identified antimicrobial activity produced by one strain of Escherichia coli that inhibited the growth of a related strain, marking the first observation of what would later be termed colicins. This finding arose during efforts to explore bacterial antagonism as a means to combat infections, predating the widespread use of antibiotics. Gratia's work laid the groundwork for recognizing bacteriocins as proteinaceous substances with strain-specific toxicity, though the term "bacteriocin" itself was not coined until later. In the mid-20th century, French researchers advanced the understanding of colicins as proteinaceous toxins. In 1946, André Gratia and Pierre Frédéricq coined the term "colicin" to describe these substances and demonstrated their protein nature through enzymatic digestion experiments, while also noting their narrow spectrum of activity against related E. coli strains. Throughout the , Frédéricq and colleagues further characterized colicins by identifying multiple types (e.g., colicins E and K) based on their receptor specificity and bactericidal effects, establishing them as models for studying bacterial antagonism. These efforts highlighted colicins as plasmid-borne traits, influencing early genetic studies on bacterial inheritance. The 1960s and 1970s saw the expansion of bacteriocin research beyond Gram-negative bacteria to include Gram-positive producers, with a focus on lantibiotics. Nisin, first isolated in 1928 from Lactococcus lactis (then Streptococcus lactis) during studies of milk fermentation inhibitors, gained recognition as a prototypical lantibiotic after its structure—featuring lanthionine bridges—was partially elucidated in the 1950s and fully detailed in 1971. Reviews in this era, such as Tagg et al. (1976), emphasized bacteriocins from Gram-positive lactic acid bacteria, broadening the field to include diverse antimicrobial peptides with applications in food preservation. Key milestones in the 1980s involved genetic analyses revealing plasmid-encoded production and regulation of bacteriocins, particularly in lactic acid bacteria. Studies demonstrated that genes for bacteriocin synthesis, export, and immunity were often clustered on plasmids, enabling conjugal transfer and facilitating industrial strain engineering. By the 1990s, standardized naming conventions and classifications emerged, with Klaenhammer (1993) proposing a system dividing Gram-positive bacteriocins into Class I (lanthionine-containing, like nisin), Class II (small heat-stable non-lantibiotics), and Class III (large heat-labile proteins), providing a framework for ongoing research.

Biosynthesis and Production

Biosynthetic Mechanisms

Bacteriocins are primarily ribosomally synthesized and proteins produced by , involving the translation of precursor encoded by structural genes within biosynthetic gene clusters. These precursors typically consist of an N-terminal leader and a C-terminal core region that undergoes modification to yield the mature bacteriocin. However, leaderless bacteriocins, a subclass primarily among II , lack the leader and are directly translated as the mature form, exported via the general Sec-dependent pathway. For instance, in lantibiotics such as , the structural gene nisA (analogous to lanA in general ) encodes a 57-amino-acid pre, where the leader sequence directs subsequent processing while the core forms the active structure. Post-translational modifications are crucial for bacteriocin maturation, particularly in modified classes like lantibiotics, where specific enzymatic steps confer stability and activity. In class I lantibiotics, dehydration of serine and residues to dehydroalanine (Dha) and dehydrobutyrine (Dhb) is catalyzed by LanB dehydratases, followed by intramolecular cyclization where thiols attack these unsaturated to form lanthionine (Ala-Lan-Ala) or methyllanthionine (Abu-MeLan-Ala) thioether bridges, mediated by zinc-dependent LanC cyclases. The leader is then cleaved by dedicated , such as the Ser/Thr LanP (e.g., NisP for ), typically at a specific recognition site like Gly-Ala-(Xxx)₂-Arg, releasing the mature only after full modification. These processes ensure the formation of rigid, heat-stable structures, as exemplified by 's five interlocking rings. While most bacteriocins rely on ribosomal synthesis, non-ribosomal peptide synthetase (NRPS) pathways are rare and typically associated with distinct classes rather than canonical bacteriocins, though some hybrid systems may incorporate NRPS-like modules in specialized producers. Export of mature bacteriocins occurs via dedicated systems to prevent intracellular toxicity, often involving ATP-binding cassette () transporters that couple secretion with final processing. For example, in many Gram-positive bacteriocins like , bifunctional transporters such as LanT (e.g., NisT) facilitate leader removal during translocation across the , while dedicated complexes like AS-48EFGH handle circular bacteriocins such as enterocin AS-48. Alternatively, some unmodified bacteriocins, such as certain class peptides, utilize the general Sec-dependent secretory pathway for . These mechanisms ensure efficient production and self-immunity in the host bacterium.

Genetic Regulation

The genetic regulation of bacteriocin production is primarily orchestrated through clustered gene operons that coordinate the expression of structural, modification, transport, immunity, and regulatory components. In many bacteriocin systems, such as the well-studied nis operon in Lactococcus lactis responsible for nisin biosynthesis, the genes are organized into divergent transcriptional units: the nisABTCPRIFEG cluster includes the structural gene nisA for the pre-nisin precursor, modification genes (nisB and nisC) for post-translational alterations, transport genes (nisT and nisP for secretion and proteolysis), immunity genes (nisI and nisFEG for producer protection), and regulatory genes (nisR and nisK forming a two-component system). This operon structure ensures synchronized expression, with promoters driving polycistronic transcription to optimize resource allocation during production phases. Similar organization is observed in other lantibiotics and class II bacteriocins, where regulatory elements like promoters and terminators maintain tight control over the entire biosynthetic pathway. A key regulatory mechanism is , which links bacteriocin production to producer cell density, preventing wasteful expression in low-population environments. In nisin-producing strains, the mature itself serves as an autoinducer, to the histidine kinase sensor NisK, which phosphorylates the response regulator NisR to activate transcription of the nis operon at a threshold concentration corresponding to high cell density. This density-dependent induction is conserved across many bacteriocin systems, including those in streptococci where competence-stimulating (CSPs) or BlpC pheromones regulate blp and com operons, coordinating bacteriocin release with population-level behaviors like formation or development. Such circuits enhance ecological fitness by synchronizing deployment only when competitors are likely present. Immunity to self-produced bacteriocins is genetically encoded within the same operons to protect the producer, typically through dedicated immunity proteins expressed constitutively or inducibly. For instance, in colicin E2 systems of Escherichia coli, the immunity gene cei encodes a small protein that binds the colicin in the periplasm, neutralizing its nuclease activity and preventing host cell death; this gene is co-transcribed with the structural cea and lysis cel genes but under separate promoter control for balanced expression. Immunity genes like nisI in the nisin cluster produce lipoprotein anchors that sequester the lantibiotic on the cell surface, ensuring producer survival during high-yield production phases. Bacteriocin genes are predominantly encoded on plasmids, facilitating horizontal transfer and rapid dissemination among bacterial populations, though chromosomal integration occurs in some cases for stable inheritance. -borne loci, such as those for colicins on group B plasmids in , include mobility elements like transposons that promote conjugal spread, enhancing the "selfish genetic element" nature of these systems. In contrast, chromosomal encoding is seen in bacteriocins like mutacin II from , where integration into the genome provides heritable protection without reliance on plasmid stability. This dual encoding strategy reflects evolutionary trade-offs between mobility and persistence in diverse microbial niches.

Classification

Methods of Classification

Bacteriocins are systematically classified using multiple criteria to account for their diversity, including the producing organism, molecular weight, biosynthesis pathways, structural features, mechanisms of action, and genetic characteristics. The primary distinction is based on the Gram status of the producer, separating bacteriocins from Gram-positive bacteria (e.g., lactic acid bacteria producing lantibiotics) and Gram-negative bacteria (e.g., colicins from Escherichia coli). This approach reflects differences in cell wall architecture and export mechanisms, with Gram-positive bacteriocins often being smaller and more heat-stable. Molecular weight serves as a key physicochemical criterion, categorizing bacteriocins into small peptides (<5 , such as lantibiotics) and larger proteins (>30 , such as bacteriolysins). Biosynthesis methods further refine this, distinguishing ribosomally synthesized and post-translationally modified peptides (RiPPs, predominant in most classes) from rare non-ribosomally synthesized ones. Structural attributes, including modifications like lanthionine bridges in lantibiotics or cyclic lasso formations, versus unmodified linear peptides, provide additional granularity. Classification by killing mechanisms highlights functional diversity, including pore-forming activity that disrupts , inhibition of cell wall synthesis (e.g., targeting II), and enzymatic actions like or cell wall degradation. Genetic and phylogenetic approaches analyze structures, in biosynthetic gene clusters, and evolutionary relationships via tools like antiSMASH, revealing horizontal gene transfer patterns. Early schemes evolved from colicin-focused classifications in the , emphasizing Gram-negative producers, to broader systems like Klaenhammer's framework for bacteriocins, which used molecular weight, , and modifications to define three main classes. Modern multi-criteria systems, refined in the and , integrate genomic data and structural insights for a more comprehensive .

Gram-Negative Bacteriocins

Gram-negative bacteriocins represent a diverse group of proteins and peptides produced by , primarily targeting closely related species to confer competitive advantages in microbial communities. Unlike their Gram-positive counterparts, these bacteriocins often face unique challenges in export due to the complex outer membrane of Gram-negative producers, leading to specialized secretion mechanisms. They are broadly classified into small peptide-based microcins, larger modular colicin-like proteins, and phage tail-like tailocins, each exhibiting distinct structural and functional properties. Microcins are the smallest subclass of Gram-negative bacteriocins, typically under 10 kDa, and are highly stable peptides produced mainly by such as . These ribosomally synthesized undergo post-translational modifications, such as cyclization or conjugation, enhancing their stability and activity. A prominent example is microcin J25, a lasso-structured that inhibits bacterial by mimicking DNA in the transcription initiation complex, thereby blocking synthesis in sensitive cells. Another key microcin, B17, targets to induce DNA damage. Microcins are frequently encoded by or chromosomal clusters that include genes for production, self-immunity, and export, often via ABC transporter-dependent type I systems or the TolC outer channel. Their host range is generally narrow, affecting related enterobacteria like and , which underscores their role in niche competition within the gut microbiome. Colicin-like bacteriocins, in contrast, are larger proteins ranging from 20 to 90 kDa, featuring a modular architecture with distinct domains for receptor recognition, translocation across the outer membrane, and cytotoxic activity. Produced predominantly by E. coli and related species, these bacteriocins are plasmid-encoded and include genes for the toxin, immunity protein, and lysis components to facilitate release. For instance, colicin E1 forms voltage-gated pores in the inner membrane, leading to depolarization and cell death in target bacteria. Translocation typically exploits host uptake systems like the TonB-dependent transporters or Tol proteins, allowing entry without dedicated secretion machinery in some cases. This subclass exhibits high specificity, killing only strains with compatible receptors, which limits their spectrum but enhances precision in ecological interactions. Examples extend beyond E. coli to klebicins in Klebsiella and similar proteins in Yersinia. Tailocins, also known as R- and F-type pyocins in certain contexts, are high-molecular-weight, phage tail-like nanostructures produced by various Gram-negative bacteria, notably Pseudomonas aeruginosa. These non-contractile (F-type) or contractile (R-type) assemblies lack phage heads and genetic material, consisting instead of a tail tube, sheath, baseplate, and receptor-binding tail fibers that recognize lipopolysaccharide (LPS) on target cells. Upon binding, R-type tailocins contract to inject the tube into the membrane, disrupting ion balance and causing rapid lysis. Encoded chromosomally in operons responsive to SOS signals like DNA damage, tailocins are released through producer cell lysis mediated by holin-like proteins. Pyocins such as S1 and R1 from P. aeruginosa exemplify this group, with narrow host ranges confined to closely related strains, enabling targeted killing in polymicrobial environments like biofilms. Their production imposes a fitness cost on producers due to lysis, balancing virulence with ecological benefits. A hallmark of Gram-negative bacteriocins is their frequent plasmid-based encoding, which facilitates horizontal transfer and rapid dissemination within populations, though chromosomal loci are common for tailocins. Export mechanisms vary: type I () and type II (Sec-dependent) secretion for microcins and colicins, while tailocins rely on lytic release. Collectively, these features contribute to their species-specific activity, with host ranges often restricted to the producer's or closer, minimizing off-target effects in diverse microbial habitats.

Gram-Positive Bacteriocins

Bacteriocins produced by , primarily such as and species, as well as and species, are diverse agents that target other Gram-positive microbes through disruption or enzymatic degradation. These bacteriocins are typically ribosomally synthesized and classified into four main classes based on structural complexity, size, and post-translational modifications, with a focus on their heat stability and modification status. This classification helps distinguish their biosynthesis pathways and mechanisms, though overlaps exist due to emerging variants. Class I bacteriocins, known as lantibiotics, are small peptides under 5 that undergo extensive post-translational modifications, including the formation of lanthionine and methyllanthionine bridges that create rigid ring structures essential for stability and activity. These modifications involve dehydration of serine/threonine residues followed by intramolecular thioether bond formation with cysteines, rendering them heat-stable and resistant to . Lantibiotics are subdivided into subclasses based on structure: subclass Ia includes elongated, cationic s like nisin A from , which features five thioether rings (including lanthionine and methyllanthionine types) that facilitate pore formation in target membranes; subclass Ib comprises globular, often anionic peptides such as mersacidin from , which inhibit synthesis by binding lipid II intermediates. A further subclass, Type II lantibiotics, consists of two-component systems like lacticin 3147 from , where synergistic peptide pairs enhance potency through cooperative membrane insertion. Class II bacteriocins are unmodified or minimally modified peptides ranging from 3 to 10 , characterized by their heat stability and cationic nature, which promotes interaction with negatively charged bacterial membranes. Subclass IIa, the pediocin-like bacteriocins, are linear peptides with a conserved in their N-terminal , exhibiting strong anti-Listeria activity; a representative example is pediocin PA-1 from , which forms pores by targeting the phosphotransferase system in sensitive cells. Subclass IIb includes synergistically acting two-peptide bacteriocins, such as lactococcin G from , where the α and β peptides (each around 40 residues) must be present in equimolar ratios to disrupt effectively. Other subclasses encompass leaderless (IIc) and miscellaneous linear forms, broadening the diversity within this heat-stable group. Class III bacteriocins consist of large, heat-labile proteins exceeding 30 , often functioning as enzymes that degrade target cell components rather than forming pores. These are further divided into IIIa (bacteriolytic, with muramidase or activity) and IIIb (non-lytic, interfering with ); for instance, megacin A-216 from is a approximately 50 protein with activity that disrupts membranes in sensitive strains, leading to cell death. Another example, lysostaphin from , is a zinc-dependent that hydrolyzes the pentaglycine cross-bridges in staphylococcal cell walls. Class IV bacteriocins represent a smaller, more heterogeneous group of complex macromolecules that incorporate or moieties, making them sensitive to enzymatic but effective in membrane perturbation. These include glycosylated peptides like glycocin F from plantarum, where O-linked glycosylation enhances stability and solubility, or lipidated forms, enhancing their interaction with target membranes. Though less common, these modifications distinguish Class IV from simpler peptide classes and highlight evolutionary adaptations in producer strains like .

Mechanisms of Action

General Modes of Action

Bacteriocins exert their effects through diverse mechanisms that primarily target the cell envelope or intracellular processes of susceptible , leading to or growth inhibition. These modes of action can be broadly categorized into membrane disruption and intracellular targeting, with effects often modulated by concentration. For instance, many bacteriocins form pores or disrupt , causing leakage and dissipation of the proton motive force, while others translocate into the to inhibit essential enzymes or degrade nucleic acids. A primary mode involves disruption via formation, which leads to and leakage of cellular ions such as . This can occur through the barrel-stave model, where amphipathic helices aggregate to form a transmembrane , as seen in colicin A, or the carpet model, in which peptides cover the surface, inducing detergent-like without deep insertion, exemplified by pediocin PA-1 binding to mannose phosphotransferase systems. Such disruptions collapse the across the , halting ATP synthesis and causing rapid . Intracellular targeting represents another key mechanism, where bacteriocins inhibit vital cellular processes after translocation. DNase and RNase activities degrade DNA or RNA, respectively; for example, colicin E7 acts as a DNase to cleave double-stranded DNA, while colicin E3 functions as an RNase targeting 16S rRNA to block protein synthesis. Additionally, some bacteriocins interfere with cell wall synthesis by blocking peptidoglycan precursors, as in colicin M, which hydrolyzes lipid-linked intermediates in the periplasm, or nisin, which binds lipid II to prevent its incorporation into the cell wall. These actions often result in energy dissipation without immediate lysis, starving the cell of essential metabolites. The lethality of bacteriocins is frequently dose-dependent, exhibiting bacteriostatic effects at low concentrations by partially disrupting or metabolic pathways, and bactericidal effects at higher levels through complete pore formation or extensive damage. For instance, demonstrates nanomolar inhibitory activity that escalates with dose, enhancing permeabilization against Gram-positive targets. This variability underscores the bacteriocins' efficiency as targeted antimicrobials, often requiring only a single to kill a sensitive in pore-forming cases.

Target Specificity and Immunity

Bacteriocins achieve selective targeting primarily through receptor-mediated binding to specific components on the surface of susceptible bacterial cells, which restricts their activity to closely related strains and minimizes off-target effects. In , the lantibiotic exemplifies this specificity by binding to lipid II, an essential precursor in , thereby docking onto the synthesis machinery and facilitating pore formation or inhibition of assembly. Similarly, in , colicins such as colicin N utilize outer membrane porins like OmpF as receptors; the receptor-binding domain of colicin N interacts with multiple sites on the OmpF trimer, enabling initial attachment and subsequent translocation across the outer membrane. This precise receptor interaction ensures that bacteriocins primarily affect phylogenetically similar competitors, enhancing their role in microbial niche competition without broadly disrupting unrelated microbial communities. To prevent self-intoxication, bacteriocin-producing cells express dedicated immunity proteins encoded by genes typically located adjacent to the bacteriocin structural genes in the same . These proteins confer protection by directly binding to the bacteriocin molecule, often sequestering its toxic domain and preventing interaction with the producer's cellular targets; for instance, the immunity protein for E3 tightly associates with the colicin in the , blocking its activity with femtomolar affinity. In pediocin-like bacteriocins from Gram-positive producers, the immunity protein forms a complex that inserts into the membrane pore formed by the bacteriocin, effectively blocking ion leakage and maintaining cellular integrity. This mechanism is highly specific, as immunity proteins generally do not protect against non-cognate bacteriocins, underscoring the tailored nature of producer self-protection. Beyond dedicated immunity proteins, additional self-immunity strategies in bacteriocin producers include modifications to receptor sites or active export systems that reduce intracellular accumulation of the . For example, some producers alter their receptor proteins, such as mutations in phosphotransferase systems that prevent binding of class IIa bacteriocins while preserving essential functions. ABC transporters, often involved in bacteriocin secretion, also contribute to immunity by rapidly exporting the from the producer cell, as seen in lantibiotic systems where the transporter recognizes and expels both the bacteriocin and its associated immunity components. These layered defenses ensure robust protection without compromising the producer's fitness. The specificity and immunity mechanisms of bacteriocins reflect ongoing co-evolution between toxin production, , and self-protection genes, driving microbial in competitive environments. Immunity genes have co-evolved tightly with bacteriocin loci, often through of operons containing both toxin and immunity elements, allowing rapid adaptation to selective pressures from rival bacteriocins. In response, target evolve via receptor mutations or accessory immunity factors, which in turn selects for bacteriocin variants with altered binding specificities, as evidenced in studies of where toxin-immunity pairs diversify to counter emerging resistances. This arms-race dynamic promotes genetic mosaicism in bacterial genomes, enhancing in polymicrobial communities.

Applications

Food Preservation

Bacteriocins play a significant role in by inhibiting spoilage organisms and pathogens, particularly in perishable products like and meat. , a lantibiotic produced by , is the most widely used bacteriocin and has been affirmed as (GRAS) by the U.S. (FDA) since 1988 for applications in pasteurized cheese spreads, , and canned foods. It is effective against , including and species, at concentrations up to 250 ppm (250 mg/kg) in finished products, where it extends by preventing outgrowth of these contaminants during storage. Other bacteriocins, such as pediocin PA-1 produced by , have been approved by the FDA for use in meat products, including sausages and ready-to-eat meats, to control growth and reduce spoilage. Commercial formulations like ALTA 2431, containing pediocin PA-1, are applied at levels that achieve reductions in pathogen counts in vacuum-packaged meats stored at temperatures. In dairy applications, enterocins from species, such as enterocin A and B, are utilized through bacteriocin-producing starter cultures in cheese and production, enhancing inhibition of unwanted without direct addition of purified peptides. Bacteriocins often exhibit synergistic effects when combined with hurdle technologies, such as low , mild heat treatments, or high-pressure processing, which broaden their and lower required dosages for efficacy. For instance, combined with organic acids like at 5.0 enhances inhibition of spores in canned , achieving greater log reductions than either alone. The (EFSA) has approved (E 234) for use in various foods at levels up to 25 mg/kg in heat-treated meats and 12.5 mg/kg in unripened cheeses, while pediocin benefits from qualified presumption of safety status for producing strains. As of 2024, the FDA granted GRAS status to a pediocin PA-1 analog for expanded use in , , and products. However, challenges include potential flavor alterations from producing strains in non-fermented foods and the risk of resistance development in target bacteria, necessitating careful formulation and monitoring.

Agricultural and Environmental Uses

Bacteriocins have emerged as promising agents for controlling plant diseases in agriculture, offering targeted antimicrobial activity against phytopathogens without the broad-spectrum effects of chemical pesticides. For instance, a class II bacteriocin produced by Bacillus velezensis HN-Q-8 has demonstrated efficacy against potato common scab caused by Streptomyces species, reducing disease severity in potato tubers. In engineering, bacteriocins from such as play a key role in modulating microbial communities to protect roots from pathogenic invasion. These peptides inhibit competitor bacteria in the soil microbiome, promoting the colonization of beneficial microbes that enhance nutrient uptake and disease resistance in crops like and cereals. Studies have shown that bacteriocin-producing strains can improve growth and stress tolerance in . Beyond plant protection, bacteriocins contribute to environmental bioremediation by aiding in pollutant degradation. In hydrocarbon-contaminated soils, purified bacteriocins from Lactobacillus acidophilus have been applied, achieving up to 89% degradation of petroleum hydrocarbons over 7 days. This approach enhances the efficiency of natural attenuation processes in marine and terrestrial environments. In aquaculture, bacteriocins provide an effective strategy against Vibrio species, major pathogens in fish and shellfish farming that cause vibriosis and significant economic losses. Probiotic strains producing bacteriocins, such as pediocin from Pediococcus or plantaricin from Lactobacillus plantarum, inhibit V. harveyi and V. alginolyticus by pore formation in their cell membranes, reducing mortality rates in tilapia and shrimp in tank trials. Field applications in pond systems during the 2010s demonstrated sustained pathogen control without residue accumulation, supporting healthier fish stocks. The primary advantages of bacteriocins in these contexts lie in their eco-friendly profile as biodegradable, host-specific antimicrobials that degrade rapidly in and , posing minimal risk to non-target organisms compared to synthetic pesticides. Field trials of Bacillus-derived bacteriocins have reported reductions in disease incidence alongside improved microbial diversity, underscoring their role in for resilient agricultural and environmental systems.

Human Health Relevance

Role in Microbiome Dynamics

Bacteriocins play a crucial role in maintaining the balance of bacterial communities within human-associated microbiomes by selectively inhibiting pathogenic or competing bacteria while sparing beneficial commensals. Produced by various commensal species, these antimicrobial peptides facilitate microbial competition and colonization resistance, thereby shaping the composition and diversity of microbiomes in sites such as the gut, vagina, oral cavity, and skin. This ecological function helps prevent dysbiosis, a state of microbial imbalance often associated with disease, by promoting stable, health-associated communities. In the vaginal microbiome, Lactobacillus species dominate the healthy and produce bacteriocins that inhibit pathogens like , a key contributor to . Approximately 80% of tested vaginal Lactobacillus strains exhibit bacteriocin activity against G. vaginalis, helping to maintain Lactobacillus dominance and reduce pathogen overgrowth. For instance, lactocin 160 from Lactobacillus rhamnosus targets G. vaginalis, supporting microbial stability and preventing . Within the gut microbiome, colonic bacteriocins contribute to shaping microbiota diversity by modulating interactions among resident bacteria, particularly in contexts like (IBD). Bacteriocins such as microcin J25 from alter gut microbiota composition, attenuating inflammation and improving barrier function in models of , thereby enhancing resistance to . Studies indicate that bacteriocin-producing , including those from , correct gut in IBD by promoting beneficial taxa and inhibiting pathogens, which supports overall microbial diversity. In the oral and microbiomes, bacteriocins inhibit formation and , aiding in the prevention of conditions like dental caries and wound infections. Oral bacteriocins, such as lantibiotics from , target cariogenic species like , limiting development on teeth and maintaining a balanced oral community. On the , bacteriocins like lugdunin from suppress growth at wound sites, reducing infection risk and promoting healing while preserving commensal diversity; similarly, Lactobacillus plantarum-derived bacteriocins aid in suppressing S. aureus infections in wounds. Ecologically, bacteriocins enhance resistance across by selectively eliminating susceptible competitors, with low bacteriocin activity linked to increased infection susceptibility. For example, lantibiotic-producing bacteria bolster microbiome resilience against pathogens like Klebsiella pneumoniae and Clostridium difficile, preventing sustained ; reduced bacteriocin production in commensals has been associated with higher rates of gastrointestinal and skin infections. This selective action underscores bacteriocins' role in fostering stable microbial ecosystems without broad disruption.

Therapeutic and Clinical Potential

Bacteriocins have emerged as promising alternatives to traditional antibiotics, particularly in combating multidrug-resistant (MDR) pathogens. , a well-studied lantibiotic produced by , exhibits potent activity against , including MDR strains such as methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE), by forming pores in their cell membranes and disrupting lipid II-dependent synthesis. Derivatives of , engineered for enhanced and potency, have shown improved against these pathogens in preclinical models, reducing minimum inhibitory concentrations while maintaining low to host cells. For , colicin-like bacteriocins such as pyocins from and klebicins from and related species offer therapeutic potential by depolarizing the inner membrane or degrading nucleic acids in pathogens like MDR P. aeruginosa and K. pneumoniae, with in vivo studies demonstrating reduced bacterial loads in infection models without significant off-target effects. Clinical applications of bacteriocins remain limited but encouraging, with advancing furthest in human trials. A small randomized clinical study in 2008 evaluated topical for treating staphylococcal in nursing mothers, reporting complete resolution of clinical symptoms in treated participants by day 14, compared to persistent infection in controls, with no adverse effects observed. Preclinical studies, including fecal models as of 2023, have shown that selectively depletes while preserving beneficial bacteria in models of gut infections. These efforts highlight bacteriocins' role as targeted antimicrobials in topical and gastrointestinal settings, though broader systemic trials are needed. Despite their promise, bacteriocins face key challenges in clinical translation, including poor stability due to proteolytic , narrow spectra limiting broad utility, and potential from repeated peptide exposure. To address these, strategies such as fusion with stabilizing domains or delivery vehicles—like nisin fused to cell-penetrating peptides—have enhanced and spectrum, enabling activity against both Gram-positive and Gram-negative targets in animal models. Beyond antibacterial uses, bacteriocins show synergies in antiviral and anticancer therapies. Nisin and related lantibiotics inhibit indirectly by modulating host microbiomes and enhancing immune responses, with in vitro studies demonstrating synergy with antivirals against enveloped viruses like and through membrane disruption. In cancer, bacteriocins such as induce in tumor cells (e.g., and lines). These multifaceted effects underscore bacteriocins' potential as adjuvants in multimodal therapies.

Research and Resources

Emerging Research Directions

Recent advancements in bacteriocin research since 2020 have leveraged and for prediction of novel bacteriocins, enhancing discovery efficiency. Tools like BAGEL4, updated to include expanded databases of ribosomally synthesized and post-translationally modified peptides (RiPPs), have enabled comprehensive genome mining by identifying structural genes, immunity proteins, and transport mechanisms in bacterial operons. For instance, a 2024 analysis of urobiome isolates using BAGEL4 identified 80 putative bacteriocin gene clusters, including 53 novel variants across classes I, II, and III, demonstrating its utility in uncovering diverse from understudied microbiomes. Concurrently, studies on producers have revealed significant untapped potential; a 2024 investigation mined 430 genomes to discover 757 aureocin A53-like leaderless bacteriocins, with 97 from , leading to the synthesis and validation of two novel peptides, arcanocin and arachnicin, active against . These findings underscore 's role as a prolific source of leaderless bacteriocins, particularly from gut and oral microbiomes, addressing gaps in phylum-specific diversity. In , efforts to engineer broad-spectrum bacteriocin variants have advanced through modular genetic circuits and cell-free expression systems. A 2025 study utilized principles—such as abstraction and standardization—to develop multiplexed bacteriocin platforms, producing cocktails of peptides like microcin V for targeted activity against multidrug-resistant pathogens. Complementing this, -based strategies have activated silent clusters encoding bacteriocins by rewiring endogenous regulation. For example, CRISPR interference and activation (CRISPRi/a) tools inserted synthetic promoters into silent biosynthetic clusters (BGCs) in species, yielding significantly increased production of secondary metabolites from silent BGCs, without off-target effects. These approaches facilitate the unlocking of cryptic bacteriocin pathways in and other producers, expanding the repertoire of engineered s. Clinical translation of bacteriocins faces significant gaps, particularly in systemic applications, with limited human trials due to challenges like proteolytic instability, potential , and insufficient toxicity data from preclinical models. Only a handful of trials, such as those evaluating for (NCT02928042), have progressed beyond animal studies, highlighting the need for more robust pharmacokinetic and safety assessments. Research has shifted toward combination therapies to overcome these hurdles, especially against biofilms; a 2020 formulation combining garvicin KS, micrococcin P1, and penicillin G eradicated methicillin-resistant Staphylococcus aureus (MRSA) biofilms by synergistically reducing viability by over 90% in clinical isolates, outperforming individual components. Looking ahead, plant-based expression systems offer a promising avenue for scalable bacteriocin production, bypassing microbial fermentation limitations. Transgenic and Solanum lycopersicum expressing genes for plantaricin, enteriocin, and leucocin yielded active peptides that inhibited plant pathogens like and , with stable inheritance across generations and potential for cost-effective manufacturing. Additionally, bacteriocins may extend to antiviral applications through microbiome ; probiotic-derived bacteriocins from species enhance gut immunity by inhibiting viral replication, such as against and herpesviruses, and restoring microbiota balance, as evidenced by reduced viral loads in preclinical models, though direct clinical efficacy remains unexplored.

Databases and Computational Tools

Databases and computational tools play a crucial role in the , , and of bacteriocins, enabling researchers to identify novel peptides from genomic and metagenomic data without relying solely on experimental validation. These resources facilitate the mining of bacterial genomes for biosynthetic clusters (BGCs) associated with bacteriocin , support sequence-based searches, and integrate structural and functional annotations to advance discovery in and . The database, specifically its version 4 (), serves as a comprehensive resource primarily for Gram-positive bacteriocins and ribosomally synthesized and post-translationally modified peptides (RiPPs). It includes curated databases of core peptides and () motifs for identifying class I and class II bacteriocins, along with tools for genome mining that analyze user-uploaded DNA sequences to detect potential bacteriocin operons. was released in 2018 and updated by 2023 to expand its core peptide database to approximately 500 RiPPs (class I), 230 unmodified peptides (class II), and additional categories such as peptides and sactipeptides, enhancing its utility for rapid screening of bacterial (meta-)genomes. The tool's interface allows visualization of gene clusters and searches against its databases, making it widely adopted for bacteriocin prospecting. BACTIBASE is an open-access database focused on the structural and functional characterization of bacteriocins across all major classes, encompassing both Gram-positive and Gram-negative producers. It provides manually curated annotations for 177 bacteriocins (as of the 2010 second release), including sequence data, physicochemical properties (e.g., molecular weight, ), and activity spectra, with tools for and similarity searches to aid in classification and evolutionary studies. Established in 2007 and updated in its second release in 2010, BACTIBASE incorporates experimental data on producer strains, target organisms, and mechanisms, serving as a key platform for comparative analysis and de novo identification. Its emphasis on validated entries distinguishes it from purely predictive tools, supporting detailed functional annotation. Other notable resources include AntiSMASH, a versatile platform for detecting BGCs involved in production, including bacteriocins as RiPPs. AntiSMASH scans genomic sequences using rule-based and HMM-based detection modules to identify clusters encoding lanthipeptides, sactipeptides, and other bacteriocin types, with version 8.0 (released in 2025) introducing enhanced analyses for chemistry, enzymology, and regulation to improve prediction accuracy across bacterial and fungal genomes. Complementing this, the MIBiG repository standardizes information on experimentally characterized BGCs for s, including bacteriocins, with version 4.0 (2024) curating over 2,500 entries that link genomic contexts to known bioactivities, facilitating cross-referencing with tools like AntiSMASH for validation. These resources collectively enable and integration of bacteriocin data into broader pipelines. Recent computational advances have introduced (ML) models for de novo bacteriocin prediction, particularly those integrating metagenomic data to uncover hidden diversity in uncultured microbiomes. For instance, tools like BPAGS (2024) employ and supervised ML algorithms, such as random forests, trained on BACTIBASE and datasets to predict bacteriocin sequences based on physicochemical properties, achieving high in identifying novel class II peptides from environmental samples. Similarly, the 2025 study in Cell Genomics utilized meta-omics integration with ML-guided prioritization to predict and synthesize 26 class II bacteriocins from human gut metagenomes, demonstrating the potential of for scalable discovery in complex microbial communities. These ML approaches, often web-accessible, outperform traditional homology-based methods by handling sequence variability and incorporating metagenomic context, though they require validation against curated databases to minimize false positives.

References

  1. [1]
    Bacteriocins: An Overview of Antimicrobial, Toxicity, and Biosafety ...
    Bacteriocins are a group of antimicrobial peptides produced by bacteria, capable of controlling clinically relevant susceptible and drug-resistant bacteria.
  2. [2]
    Bacteriocins: potentials and prospects in health and agrifood systems
    Apr 25, 2024 · Bacteriocins have been generally defined as ribosomally synthesized antimicrobial peptide molecules that can either be enzymatically modified or ...<|control11|><|separator|>
  3. [3]
    Bacteriocins from Lactic Acid Bacteria. A Powerful Alternative ... - NIH
    Bacteriocins are peptides synthesized by bacteria that can inhibit the growth of other bacteria and fungi, parasites, and viruses. Lactic acid bacteria (LAB) ...
  4. [4]
    Bacteriocins: Properties and potential use as antimicrobials - PMC
    Most bacteriocins have two main characteristics that distinguish them from traditional antibiotics: Bacteriocins are ribosomally synthesized and have a ...
  5. [5]
    Bacteriocin diversity, function, discovery and application as ... - NIH
    May 10, 2024 · Bacteriocins are a subset of bacterially produced, ribosomally synthesized antimicrobial peptides that have been studied for almost a century.
  6. [6]
    Bacteriocin - an overview | ScienceDirect Topics
    Bacteriocin is defined as a heterogeneous group of bioactive bacterial peptides or proteins that are ribosomally synthesized and exhibit antimicrobial activity ...
  7. [7]
    Bacteriocins, Antimicrobial Peptides from Bacterial Origin
    Bacteriocins exhibit antimicrobial activity with variable spectrum depending on the peptide, which may target several bacteria.
  8. [8]
    Colicin Biology - PMC - PubMed Central
    Colicins are proteins produced by and toxic for some strains of Escherichia coli ... The narrow target range of colicins has been shown by Fredericq to be ...Missing: Pierre | Show results with:Pierre
  9. [9]
  10. [10]
    After a century of nisin research - where are we now?
    Nisin is one of the oldest known antimicrobial compounds. In 1928, the same year that Alexander Fleming discovered penicillin, Rogers and Whittier (Rogers 1928) ...Abstract · Introduction · Genome mining and new nisin... · Nisin bioengineering to...
  11. [11]
  12. [12]
  13. [13]
    Lantibiotics: structure, biosynthesis and mode of action
    The prototype LAB bacteriocin, nisin, was first discovered in 1928, when Rogers [5] observed metabolites of Streptococcus lactis (now reclassified as ...
  14. [14]
    Mechanistic Understanding of Lanthipeptide Biosynthetic Enzymes
    Lanthipeptides are ribosomally synthesized and post-translationally modified peptides (RiPPs) that display a wide variety of biological activities, ...
  15. [15]
  16. [16]
    Multifaceted ABC transporters associated to microcin and ...
    This review specifically focuses on the different classes of ABC transporters that are involved in either export and/or maturation of microcins and bacteriocins ...
  17. [17]
    Circular Bacteriocins: Biosynthesis and Mode of Action - ASM Journals
    Oct 17, 2014 · Circular bacteriocins are synthesized as linear precursors, containing a leader sequence of variable size which is cleaved off during maturation ...
  18. [18]
  19. [19]
    Genes responsible for nisin synthesis, regulation and immunity form ...
    The nisZBTCIPRKFEG gene cluster, responsible for nisin biosynthesis, immunity and regulation, consists of two operons, nisZBTCIPRK and nisFEG.
  20. [20]
    Genetics of Bacteriocins Biosynthesis by Lactic Acid Bacteria
    In many LAB bacteriocin production is. “quorum-sensing” mechanisms controlled. Quorum-sensing systems control plethora of different important biological ...
  21. [21]
    Regulation of the nisin operons in Lactococcus lactis N8 - PubMed
    These results showed that the nisZBTCIPRK operon is positively autoregulated and that the nisFEG operon is in the same regulon. Publication types. Research ...Missing: nis | Show results with:nis
  22. [22]
    Quorum-Sensing Regulation of the Production of Blp Bacteriocins in ...
    This locus contains all the genetic information required for the production of bacteriocin and is regulated at the transcriptional level by a QS mechanism in ...
  23. [23]
    Regulation of Bacteriocin Production in Streptococcus mutans by the ...
    The results presented above have shown that expression of several genes related to bacteriocin synthesis is controlled by the comCDE quorum-sensing system.
  24. [24]
    CsrA and its regulators control the time-point of ColicinE2 release in ...
    Apr 25, 2018 · The ColicinE2 operon consists of three genes: cea (the colicin activity gene), cei (the immunity gene) and cel (the lysis gene) (Fig.
  25. [25]
    Function of Lactococcus lactis Nisin Immunity Genes nisI and ...
    Nisin-producing Lactococcus lactisstrains show a high degree of resistance to the action of nisin, which is based upon expression of the self-protection ...<|control11|><|separator|>
  26. [26]
    The role of bacteriocins as selfish genetic elements - PMC - NIH
    Some are encoded on plasmids and can be horizontally transferred via mobile genetic elements [4]. Bacteriocin genetic systems are composed of a bacteriocin gene ...
  27. [27]
    Complete Sequence of the Enterocin Q-Encoding Plasmid pCIZ2 ...
    In many cases, bacteriocin production is a plasmid-encoded trait; however, bacteriocin genes have been also found on the chromosome and mobile genetic elements, ...
  28. [28]
    Overview of Global Trends in Classification, Methods of Preparation ...
    This paper summarizes information about the division of bacteriocins into classes (Gram-negative bacteria, Gram-positive bacteria, and archaea).
  29. [29]
    Classification and Multi-Functional Use of Bacteriocins in Health ...
    This review aims to synthetize the information available nowadays regarding bacteriocins, and their classification, while also providing an insight into the ...
  30. [30]
  31. [31]
    Bacteriocin: A natural approach for food safety and food security
    In 1993 Klaenhammer categorized bacteriocins into four classes (Klaenhammer, 1993). In this grouping, class I is lantibiotics portrayed by thermostable ...
  32. [32]
  33. [33]
  34. [34]
    Revisiting the Multifaceted Roles of Bacteriocins - PubMed Central
    Feb 14, 2024 · Among gram-negative bacteria, colicins (30–80 kDa) and microcins (1–10 kDa) from E. coli have been studied most extensively, although other ...Missing: tailocins | Show results with:tailocins
  35. [35]
  36. [36]
  37. [37]
    The therapeutic potential of bacteriocins as protein antibiotics
    Apr 21, 2017 · This review discusses the state of the art regarding the use for therapeutic purposes of two types of Gram-negative bacteriocins: colicin-like bacteriocins ( ...
  38. [38]
    The world of phage tail-like bacteriocins: State of the art and ...
    Among the bacteriocins originating from Gram-negative bacteria, colicins, produced mainly by Escherichia coli, and microcins are the important ones (Benítez ...
  39. [39]
  40. [40]
  41. [41]
    Bacteriocins, Antimicrobial Peptides from Bacterial Origin - MDPI
    Bacteriocins Produced by Gram-Positive Bacteria (BGPB) BGPBs are currently classed into four different classes [30,32]: Class I is also named lantibiotics and ...Bacteriocins, Antimicrobial... · 4. Mechanism Of Action · 4.1. Antimicrobial...
  42. [42]
    Overview of Global Trends in Classification, Methods of Preparation ...
    Aug 28, 2020 · This paper summarizes information about the division of bacteriocins into classes (Gram-negative bacteria, Gram-positive bacteria, and archaea).2.1. Cell Type... · 2.1. 1. Bacteriocins From... · 2.1. 2. Bacteriocins From...
  43. [43]
    Lactic Acid Bacteria and Their Bacteriocins: Classification ... - MDPI
    Class IIb bacteriocins require the synergistic activity of two complementary peptides in order to exert antimicrobial activity, examples being plantaricin A, ...Missing: glycosylated | Show results with:glycosylated
  44. [44]
    Structural basis of lantibiotic recognition by the nisin resistance ...
    Jan 4, 2016 · Nisin is the most prominent member and contains five crucial lanthionine rings. Some clinically relevant bacteria express membrane-associated ...
  45. [45]
    Bacteriocins of lactic acid bacteria: extending the family
    Feb 10, 2016 · In addition to the three bacteriocin classes proposed in the early 1990s (Klaenhammer 1993), our broad genome mining analysis of LAB shows ...
  46. [46]
    Bacteriocin from Bacillus megaterium ATCC 19213 - PubMed
    A bacteriocin produced by Bacillus megaterium ATCC 19213 was identified, purified, and compared with megacin A from B. megaterium 216.
  47. [47]
    Innovative perspectives on bacteriocins: advances in classification ...
    Bacteriocins are antimicrobial peptides that are produced by bacteria and can kill or inhibit different other bacterial strains, and therefore can be used in ...Class I Bacteriocins: Ripps... · Novel Classification · Bacteriocin In Food...
  48. [48]
    Colicin Biology | Microbiology and Molecular Biology Reviews
    Below is a merged summary of the modes of action of colicins, combining all the information from the provided segments into a concise yet comprehensive response. To retain all details efficiently, I will use a table in CSV format for the core data, followed by additional narrative details and quotes that don’t fit neatly into the table. This approach ensures maximum density and clarity while preserving all the information.
  49. [49]
    Colicins and Microcins Produced by Enterobacteriaceae - MDPI
    Colicins kill target cells through three different mechanisms: (a) by making voltage-dependent channels in the inner membrane of the target bacteria, (b) by a ...
  50. [50]
    Lipid II is an intrinsic component of the pore induced by nisin in ...
    However, it was not known whether Lipid II functions only as a receptor to recruit nisin to bacterial membranes, thus increasing its specificity for bacterial ...Missing: bacteriocin | Show results with:bacteriocin
  51. [51]
    Bifurcated binding of the OmpF receptor underpins import ... - PubMed
    Jul 3, 2020 · We demonstrate here that ColN uses OmpF both as its receptor and translocator. This dual function is achieved by ColN having multiple distinct OmpF-binding ...Missing: specificity | Show results with:specificity
  52. [52]
    Common mechanisms of target cell recognition and immunity for ...
    In bacteriocin-producing cells (e.g., B244), the cognate immunity protein is tightly associated with the bacteriocin-bound receptor proteins to protect cells ...Results · Formation Of Protein Complex... · Bacteriocin Concentration...
  53. [53]
    Structural Basis of the Immunity Mechanisms of Pediocin-like ...
    Jun 15, 2022 · The C-terminal loop of the immunity protein then inserts into the pore and blocks leakage induced by bacteriocins. Elucidation of the toxicity ...
  54. [54]
    Subclass IId bacteriocins targeting mannose phosphotransferase ...
    Sep 3, 2024 · Finally, we propose that while the bacteriocins from separate groups display specific binding patterns to Man-PTS, the general mechanism of ...Missing: specificity | Show results with:specificity
  55. [55]
    Self‐immunity to antibacterial peptides by ABC transporters - Smits
    Oct 11, 2020 · In this review, we have tried to capture the 'diverse' export systems ... bacteriocin ABC transporters carry out proteolytic processing of ...
  56. [56]
    Current Knowledge of the Mode of Action and Immunity Mechanisms ...
    [12,44,45]. Genes coding for bacteriocins and those coding for immunity proteins are usually located in the same cluster and are often co-regulated. This review ...
  57. [57]
    The evolution of spectrum in antibiotics and bacteriocins - PNAS
    Sep 13, 2022 · This broad-spectrum of activity has its evolutionary roots in microbial warfare, where antibiotics can provide a competitive edge.
  58. [58]
    Epidemiological dynamics of bacteriocin competition and antibiotic ...
    Oct 5, 2022 · This modelling approach reflects observed variation in the absence/presence of toxin and immunity genes in pneumococcal genomes [4–8,11,12].
  59. [59]
    21 CFR 184.1538 -- Nisin preparation. - eCFR
    The current good manufacturing practice level is the quantity of the ingredient that delivers a maximum of 250 parts per million of nisin in the finished ...
  60. [60]
    Expanding Layers of Bacteriocin Applications: From Food ... - MDPI
    For example, the MicroGARDTM series and ALTA 2431, both containing pediocin PA-1, are approved by the FDA. The use of bacteriocin-producing cultures as ...
  61. [61]
    Biopreservation strategies using bacteriocins to control meat ...
    Pediocin PA-1 reduced the counts of L. monocytogenes inoculated in raw chicken meat from 5 log CFU/g to 3.8 log CFU/g when stored at 4 ºC for 1 month, however, ...
  62. [62]
    Application of Bacteriocins and Protective Cultures in Dairy Food ...
    Apr 9, 2018 · Bacteriocins can be applied to dairy foods on a purified/crude form or as a bacteriocin-producing LAB as a part of fermentation process or as adjuvant culture.
  63. [63]
    Safety of nisin (E 234) as a food additive in the light of new ... - EFSA
    Dec 11, 2017 · ... maximum level of 12 mg/kg) and in heat-treated meat products (at maximum level of 25 mg/kg) would not be of safety concern. © European Food ...Missing: concentration | Show results with:concentration
  64. [64]
    Combined antimicrobial effect of bacteriocins with other hurdles of ...
    Jun 15, 2022 · Combination of bacteriocins with other hurdles markedly improve food safety. Synergistic effects are observed between bacteriocin and other hurdles.
  65. [65]
    Bacteriocin: A natural approach for food safety and food security - PMC
    Oct 24, 2022 · The usage of bacteriocin-producing strains for the preservation of non-fermented foods is carried out if they do not impart flavors or any bad ...
  66. [66]
    [PDF] BACTERIOCINS ACTIVE AGAINST PLANT PATHOGENIC BACTERIA
    spp., Pseudomonas spp., and Bacillus spp., to combat potato common scab (Eckwall and Schottel, ... by water stress using Bradyrhizobium japonicum and thuricin-17 ...
  67. [67]
    Bacteriocins from the rhizosphere microbiome – from an agriculture ...
    This review focuses on the bacteriocins produced by plant-rhizosphere colonizers and plant-pathogenic bacteria, that might have uses in agriculture, veterinary ...
  68. [68]
    Bacteriocins from the rhizosphere microbiome - NIH
    This review focuses on the bacteriocins produced by plant-rhizosphere colonizers and plant-pathogenic bacteria, that might have uses in agriculture, veterinary, ...Missing: microcins | Show results with:microcins
  69. [69]
    The efficiency of purified bacteriocin from Lactobacillus acidophilus ...
    Thus, it can be concluded that bacteriocin bioremediation is an efficient and cost-effective biological treatment for eliminating petroleum hydrocarbons ...
  70. [70]
    Use of Probiotic Bacteria and Bacteriocins as an Alternative to ... - NIH
    Overview of bacteriocin effects in fish health or against aquaculture pathogenic bacteria. Aquatic Specie, Bacteriocin, Pathogen or Challenge, Clinical Impact ...
  71. [71]
  72. [72]
    A PGPR-Produced Bacteriocin for Sustainable Agriculture - NIH
    Jul 7, 2020 · This review focuses on the effect of thuricin 17 as a microbe to plant signal that assists crop plants in managing stress and making agricultural systems more ...
  73. [73]
    The potential of bacteriocins and bacteriophages to control bacterial ...
    The use of phage-based products is seen as an eco-friendly and sustainable alternative to chemical treatments, particularly in organic and environmentally ...
  74. [74]
    The microbiome-shaping roles of bacteriocins - PubMed
    Jun 1, 2021 · Many commensal bacteria produce small antibacterial molecules termed bacteriocins that have the capacity to eliminate specific colonizing pathogens.
  75. [75]
    Defense factors of vaginal lactobacilli - PubMed
    Bacteriocin activity was tested on 4 strains of Gardnerella vaginalis. Approximately 80% of the lactobacilli tested produced bacteriocin that inhibited growth ...Missing: microbiome | Show results with:microbiome
  76. [76]
  77. [77]
    Recombinant antimicrobial peptide microcin J25 alleviates DSS ...
    Apr 2, 2021 · MccJ25 effectively attenuates inflammation and improves disrupted barrier function, and the MccJ25-modified gut microbiota plays a central role in this process.Missing: shaping | Show results with:shaping
  78. [78]
    Bacteriocin-Producing Probiotic Lactic Acid Bacteria in ... - PubMed
    May 16, 2022 · In this review, we have discussed the ability of bacteriocin-producing probiotic lactic acid bacteria in the modulation of gut microbiota correcting dysbiosis.
  79. [79]
  80. [80]
  81. [81]
    Lactobacillus plantarum USM8613 Aids in Wound Healing and ...
    Our present study illustrates the potential of L. plantarum USM8613 in aiding wound healing, suppressing of S. aureus infection at wound sites and promoting ...
  82. [82]
    Lantibiotic-producing bacteria impact microbiome resilience and ...
    May 9, 2025 · Lantibiotic-mediated dysbiosis results in sustained loss of colonization resistance against Klebsiella pneumoniae and Clostrioides difficile ...
  83. [83]
    Demystifying Bacteriocins of Human Microbiota by Genome Guided ...
    Unlike antibiotics, bacteriocins selectively inhibit a spectrum of competent bacteria and are said to safeguard gut commensals, reducing the chance of dysbiosis ...
  84. [84]
    Biomedical applications of nisin | Journal of Applied Microbiology
    Nisin is explored for treating drug-resistant infections, oral diseases, and as a cancer therapeutic, showing potential in biomedical applications.
  85. [85]
    Bio-Engineered Nisin with Increased Anti-Staphylococcus and ...
    From a clinical perspective, nisin has already shown great promise in treating infectious mastitis in lactating mothers [48]. Notably, a significant reduction ...
  86. [86]
    The therapeutic potential of bacteriocins as protein antibiotics - NIH
    Bacteriocins, potent protein toxins, show potential as antibiotics for Gram-negative infections, with encouraging in vivo results and low resistance, but more ...
  87. [87]
    The Bacteriocin Nisin, an Effective Agent for the Treatment of ...
    No clinical signs of mastitis were observed among the women of the nisin group on day 14, whereas they persisted throughout the study in the women of the ...<|control11|><|separator|>
  88. [88]
    Modulation of the gut microbiome with nisin | Scientific Reports
    May 16, 2023 · We show that orally-ingested nisin survives transit through the porcine gastrointestinal tract intact (as evidenced by activity and molecular weight ...
  89. [89]
    Current challenges and development strategies of bacteriocins ...
    Dec 14, 2024 · Bacteriocin-producing LAB strains play a beneficial role in improving the flavor and preservation properties of fermented foods (Wang et al., ...Abstract · CURRENT CHALLENGES OF... · IMPROVEMENT STRATEGIES...
  90. [90]
    Design of Recombinant Bacteriocin Fusion Protein and Evaluation ...
    Aug 4, 2025 · ... fusion protein can lead to develop a potent therapeutic drug against breast cancer. ... In Silico Evaluation of Two Targeted Chimeric Proteins ...<|control11|><|separator|>
  91. [91]
    Probiotic-Based Bacteriocin: Immunity Supplementation Against ...
    Jul 25, 2022 · Bacteriocins possess numerous health benefits and exhibit antagonistic activity against enteric pathogens and immunobiotics, thereby inhibiting viral ...
  92. [92]
    Emerging lactic acid bacteria bacteriocins as anti-cancer and anti ...
    Sep 15, 2024 · It has been demonstrated that some bacteriocins, such as pediocin and nisin, can boost the immune system. This is accomplished by activating a ...
  93. [93]
    An exploratory in silico analysis of bacteriocin gene clusters in ... - NIH
    Mar 27, 2024 · BAGEL4 scans the bacterial genome for putative bacteriocin open reading frames (ORFs). It searches for the structural bacteriocin gene, but also ...
  94. [94]
    Discovery and synthesis of leaderless bacteriocins from the ...
    Oct 15, 2024 · This research highlights the biosynthetic potential of Actinomycetota in terms of leaderless bacteriocin production and describes the first antimicrobial ...<|control11|><|separator|>
  95. [95]
    Multiplexed bacteriocin synthesis to combat and prevent ... - Nature
    Aug 19, 2025 · Bacteriocins are underexplored yet promising candidates to combat antimicrobial resistance (AMR) and enable targeted therapy due to their ...
  96. [96]
    Activating natural product synthesis using CRISPR interference and ...
    We describe a novel approach to activate silent BGCs through rewiring endogenous regulation using synthetic gene regulators based upon CRISPR-Cas.Missing: bacteriocin | Show results with:bacteriocin
  97. [97]
    Evaluating the Translational Potential of Bacteriocins as an ... - NIH
    Most clinical trials investigating the ability of bacteriocins as an alternative therapy against S. aureus involve nisin. As S. aureus is the predominant ...
  98. [98]
    A bacteriocin-based antimicrobial formulation to effectively disrupt ...
    Dec 2, 2020 · We show that the combination between garvicin KS, micrococcin P1, and penicillin G potently inhibit cell viability within S. aureus biofilms by causing severe ...
  99. [99]
    Plant-produced bacteriocins inhibit plant pathogens and confer ...
    Jul 25, 2021 · This study highlights that plants can be used as biofactories for AMP production and that the expression of bacteriocins in planta may offer new opportunities ...
  100. [100]
    Probiotic-Based Bacteriocin: Immunity Supplementation Against ...
    Jul 26, 2022 · Researchers studied that enhancing the body's immune system by employing probiotic-based bacteriocin helps in fighting against viral diseases ( ...
  101. [101]
    BAGEL4: a user-friendly web server to thoroughly mine RiPPs and ...
    May 21, 2018 · The bacteriocin database has been updated and now contains almost 500 RiPPs (class 1, see Supplementary Figure S1), 230 unmodified bacteriocins ...
  102. [102]
    BADASS: BActeriocin-Diversity ASsessment Software - PMC
    Jan 20, 2023 · BAGEL4 was updated and now has about 500 Ribosomally synthesized and post-translationally modified peptides (RiPPs) (class 1), 230 non-modified ...
  103. [103]
    BACTIBASE second release: a database and tool platform for ...
    Jan 27, 2010 · BACTIBASE is an integrated open-access database designed for the characterization of bacterial antimicrobial peptides, commonly known as bacteriocins.
  104. [104]
    BACTIBASE: a new web-accessible database for bacteriocin ...
    A new and original database BACTIBASE that contains calculated or predicted physicochemical properties of 123 bacteriocins produced by both Gram-positive and ...
  105. [105]
    antiSMASH 8.0: extended gene cluster detection capabilities ... - NIH
    Apr 25, 2025 · antiSMASH 8.0: extended gene cluster detection capabilities and analyses of chemistry, enzymology, and regulation
  106. [106]
    MIBiG 4.0: advancing biosynthetic gene cluster curation through ...
    Dec 9, 2024 · Here, we describe MIBiG version 4.0, an extensive update to the data repository and the underlying data standard.
  107. [107]
    BPAGS: a web application for bacteriocin prediction via feature ...
    Jan 9, 2024 · A web application for predicting bacteriocin was created in this study, utilizing a machine learning approach.
  108. [108]
    Systematically investigating and identifying bacteriocins in the ...
    Sep 2, 2025 · Guided by meta-omics analysis, we synthesized 26 hypothetical bacteriocins from gut commensal species, with 16 showing antibacterial activities.