Next Article in Journal
Genetic Diversity in Three Sinipercine Fishes Based on Mitochondrial D-Loop and COX1 Sequences
Previous Article in Journal
Effects of Micro- and Macroalgae-Supplemented Diets on Growth and Muscle Fibrillar Constitution of Gilthead Seabream, Sparus aurata L., in the Final On-Growing Phase
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microecological Preparations as Antibiotic Alternatives in Cyprinid Aquaculture

1
College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
2
Hydrobiology Department, National Research Centre, Cairo 999060, Egypt
*
Authors to whom correspondence should be addressed.
Fishes 2025, 10(6), 263; https://doi.org/10.3390/fishes10060263
Submission received: 5 March 2025 / Revised: 29 April 2025 / Accepted: 20 May 2025 / Published: 2 June 2025

Abstract

:
Microecological preparations (MPs), encompassing probiotics, prebiotics, synbiotics, and postbiotics, are microbial feed supplements that enhance host health through gut microbiota modulation. Unlike the narrow definition of probiotics (viable microorganisms), MPs constitute a broader category including non-viable microbial derivatives and selectively fermented substrates. Their application in aquaculture significantly reduces antibiotic dependence. Given the industry’s intensification challenges, while meeting global protein demands, high-density aquaculture elevates disease risks, driving prophylactic antibiotic overuse. This practice accelerates antimicrobial resistance (AMR) development, compromising treatment efficacy and causing residual antibiotics in aquatic products. Such residues violate international food safety standards, triggering trade disputes. As sustainable alternatives, MPs operate through multiple mechanisms: the competitive exclusion of pathogens, immune stimulation, and nutrient absorption enhancement. This review examines the patterns of antibiotic abuse and the emergence of AMR in carp aquaculture, evaluates MP-based mitigation strategies from the perspective of antibiotic alternatives, and analyzes the advantages, disadvantages, and application progress of MPs. Based on existing evidence, we propose targeted research priorities for MP optimization, advocating for scientifically guided implementation in commercial cyprinid aquaculture.
Key Contribution: Cyprinid production leads global freshwater aquaculture. However, challenges such as high feed costs, antibiotic residues, and drug resistance raise significant concerns. Antibiotic-free aquaculture presents a promising solution by prioritizing aquatic animal health, protecting human health; and ensuring the production of safe, nutritious, and residue-free aquatic products. Feed additives serve as effective alternatives to antibiotics, offering multiple benefits. This article highlights the importance, current status, and challenges of microecological agents, antibiotic residues, and resistance. It aims to provide valuable insights for fish farmers and regulatory bodies to deepen their understanding and promote the research, development, application, and management of feed additives in aquaculture.

1. Introduction

Cypriniformes represents the most speciose order of freshwater teleosts, encompassing 321 genera and 3268 documented species [1]. Notably, Cyprinidae exhibits the most extensive geographical distribution and taxonomic diversity within this order. Economically significant species within Cyprinidae encompass Mylopharyngodon piceus, Ctenopharyngodon idellus, Hypophthalmichthys molitrix, Aristichthys nobilis, Cyprinus carpio, Carassius auratus, Megalobrama terminalis, and Parabramis pekinensis, which collectively constitute the principal species in commercial aquaculture. Since the 1980s, the aquaculture sector has experienced substantial production growth, concurrently facing escalating challenges from disease outbreaks that correlate with intensive cultivation practices [2,3].
Aquaculture is one of the fastest-growing industries in the world, helping to address food shortages and promote economic development. The Food and Agriculture Organization of the United Nations (FAO) predicts that by 2030, fish production in aquaculture may reach 109 million tons, with Asia being the dominant sector, accounting for 89% of total production [3]. In 2020, antibiotic consumption in global aquaculture reached 99,502 metric tons, with pronounced regional disparities: Asia accounted for 67% of total usage, while Africa represented a negligible share (<1%). Current models project an 8.0% increase in global antibiotic use by 2030, escalating to 107,472 metric tons. These trends underscore urgent challenges, including the proliferation of antimicrobial resistance (AMR) and the ecological risks posed by antibiotic residues in aquatic ecosystems [2]. The escalating dependence on antibiotics in aquaculture poses profound threats to the sector’s sustainable development. Rampant misuse and overuse of these agents have precipitated a critical surge in antimicrobial resistance (AMR), severely undermining the efficacy of therapeutic interventions against bacterial infections in cyprinid species. Key pathogens complicating cyprinid aquaculture include Aeromonas punctata, Corynebacterium columnum, Aeromonas hydrophila, and Aeromonas thermophila, among others, with AMR exacerbating disease management challenges across global aquaculture systems [4,5]. Most of these pathogens are aerobic, Gram-negative bacteria, leading to the widespread use of antibiotics targeting such bacteria. However, the issue of antibiotic residues remains severe [4,6] (Table 1). Globally, the most commonly used antibiotics in aquaculture include tetracyclines, oxalic acid, fluoroquinolones (e.g., sarafloxacin, enrofloxacin), beta-lactams (e.g., amoxicillin), macrolides (e.g., erythromycin), sulfonamides (e.g., sulfamethoxazole), diaminopyrimidines (e.g., omeprazole), and bisphenols (e.g., florfenicol) [7,8]. The types and levels of antibiotic residues in cyprinid fish are similar to the overall residue situation in fish farming. Currently, there is still a lack of systematic analyses and reports on antibiotic residues in cyprinid fish that are accurate to different regions of each country.
On the one hand, antibiotic residues pose significant toxic risks. The primary public health impacts include the development of antimicrobial resistance (AMR) (Figure 1), allergic reactions (e.g., to penicillin), carcinogenicity (e.g., sulfamethoxazole, oxytetracycline, and furazolidone), anaphylactic shock, nephropathy (e.g., gentamicin), mutagenicity, teratogenicity, bone marrow suppression, and disruptions to the normal gut microbiota [9,10].
Table 1. Adverse effects of antibiotic residue on human health.
Table 1. Adverse effects of antibiotic residue on human health.
Side EffectDescriptionReference
AMRThe residues in aquaculture products can lead to the emergence of antibiotic-resistant bacteria.
This will reduce the effectiveness of the antibiotics used to treat human infections, leading to prolonged disease progression and increased healthcare costs.
[11]
Residue accumulationThe continued consumption of residues in fish products can lead to biological accumulation in the human body, which, over time, may increase the risk of chronic health problems, such as organ damage and endocrine disorders.[12]
Allergic reactionsSome people may experience allergic reactions to antibiotic residues, ranging from mild symptoms, such as rash or urticaria, to more severe reactions, such as allergic reactions.
Specific antibiotics, such as penicillin, are more likely to trigger allergic reactions.
[13,14]
ToxicityLong-term exposure to antibiotic residues, especially those that have not been fully metabolized, may have toxic effects on human organs and systems.
For example, certain antibiotics, such as tetracyclines and sulfonamides, can cause kidney or liver damage if consumed in large quantities.
[15,16]
Disruption of gut microbiotaAntibiotic residues can alter the natural balance of gut bacteria, potentially leading to digestive problems, weakened immune responses, and even increased susceptibility to infections.
This kind of destruction is particularly concerning in children, who heavily rely on healthy gut microbiota for normal development.
[17]
Potential carcinogenicitySome antibiotics used in aquaculture, such as nitrofurans and quinolones, have been shown to have carcinogenic properties.
These antibiotics may cause DNA damage or mutations, thereby disrupting normal cell growth.
For example, nitrofuran can form reactive intermediates that bind to cellular macromolecules, leading to potential cancer development.
Similarly, it has been found that certain quinolone drugs increase oxidative stress and interact with enzymes involved in DNA replication, which may lead to chromosomal damage.
[18,19,20,21,22]
Endocrine disruptionSome antibiotics, such as fluoroquinolones and tetracyclines, can act as endocrine disruptors.
Their residues can interfere with hormone regulation and may affect reproductive health, growth, and development.
This kind of destruction may have a significant impact on human fertility and development.
[23]
Drug interactionsAntibiotic residues in food can interact with drugs taken by humans, reducing their effectiveness or causing adverse reactions.
For example, residual tetracycline may reduce the efficacy of certain anticoagulant drugs, thereby increasing the risk of blood clotting.
[24]
Once bacteria develop AMR, they can transfer the resistance to other species and strains through horizontal gene transfer. For example, zoonotic pathogens, such as Streptococcus suis, Pseudomonas aeruginosa, and Vibrio vulnificus, carry extended-spectrum beta-lactamases (ESBLs) and other antimicrobial resistance genes (ARGs), which can spread through the food chain [25]. Humans can become infected with these zoonotic bacteria through contact with aquatic animals, highlighting the transmission of aquaculture-associated bacteria to humans. This greatly complicates the treatment of human diseases such as pneumonia, tuberculosis, septicemia, gonorrhea, and salmonellosis, which are becoming increasingly resistant to common antibiotics [26,27,28]. Unfortunately, cyprinid fish also serve as hosts for zoonotic diseases like salmonellosis, underscoring the urgent need for alternative strategies to control Gram-negative bacterial infections [29].
On the other hand, challenges persist in the detection and regulation of antibiotic residues in food. The detection process typically involves two steps: sample pretreatment and analysis. Pretreatment techniques include solid-phase extraction (SPE) [30], QuEChERS technology [31], matrix solid-phase dispersion extraction (MSPD) [32], Pressure Liquid Extraction (PLE) [33], and microwave-assisted extraction (MAE) [34,35,36]. Common analytical methods include thin-layer chromatography (TLC) [37], high-performance liquid chromatography (HPLC), capillary electrophoresis (CE) [38], biosensors, microbial assays, and immunoassays. However, traditional detection methods face several limitations, including difficulty in identifying diverse antibiotics, limited sensitivity for trace detection, complexity and potential errors in sample pretreatment, and high costs associated with advanced detection equipment. Although techniques like fluorescence polarization and enzyme-linked immunosorbent assay (ELISA) are reliable and sensitive, they are seldom used by testing institutions and local authorities [35]. Moreover, high-resolution mass spectrometry and nuclear magnetic resonance instruments, while offering precise results, are prohibitively expensive, with equipment costs often reaching millions of USD—posing a major barrier to widespread adoption (Figure 2).
Antibiotics at therapeutic levels, for example, are commonly given to groups of fish that share tanks or cages via the oral route for brief periods in aquaculture. Antibiotics have played an essential role in the aquaculture sector in terms of treatment and growth promotion [37]. While antibiotics are administered via parenteral and oral routes in terrestrial animals, the most common method for delivering antibiotics to fish is to mix the antibiotic with the prepared feed. Antibiotics, on the other hand, are not properly metabolized by fish, and they are passed largely unused back into the environment in feces. Of the antibiotics supplied to fish, 75% are excreted into the water [37], and this fact has prompted most countries with a significant aquaculture business to take some control measures through government organizations. For example, since 2006, the European Union (EU) has banned the use of antibiotics for growth promotion to reduce the risk of AMR and ensure food safety [3]. Similarly, the U.S. Food and Drug Administration (FDA) has issued guidelines limiting antibiotic use for growth promotion and mandates veterinary oversight for antibiotic use in aquaculture [1,2,3].
Modern aquaculture best practices now emphasize the therapeutic use of antibiotics strictly under veterinary supervision and only when necessary to treat specific infections. Additionally, comprehensive management strategies are encouraged, such as improving feeding practices, enhancing disease prevention, and adopting alternatives like vaccines and probiotics to minimize antibiotic reliance and lower the risk of drug resistance [3]. This article aims to review recent advancements in the use of feed additives as alternatives to antibiotics in cyprinid fish farming. It seeks to provide valuable insights to help fish farmers and regulatory authorities deepen their understanding and promote the research, development, application, and regulation of feed additives in sustainable aquaculture.

2. The Advantages and Disadvantages of Different Antibiotic Alternatives

Current alternatives to antibiotics in aquaculture include gene editing, vaccination strategies, phage therapy, probiotics, prebiotics, chicken egg yolk antibodies (IgYs), medicinal plants, and the use of “clean seeds” or specific pathogen-free (SPF) stocks as essential components of biosafety strategies [39]. However, the development and application of SPF systems face opposition due to high investment and maintenance costs, requiring skilled personnel, specialized knowledge, and advanced facilities [40]. Moreover, SPF systems have not fundamentally addressed antibiotic dependency, especially in developing countries.
Medicinal plants have long been recognized as natural immunostimulants due to their bioactive properties. As eco-friendly and cost-effective alternatives to antibiotics and synthetic immunoprophylactics in aquaculture, these plants are gaining global attention for their advantages: simple preparation, low cost, and minimal adverse effects on aquatic species and ecosystems. Extensive studies have investigated diverse plant-derived materials in aquatic animals, including herbs, spices, seaweeds, herbal extracts, traditional Chinese medicine formulations, and commercial phytogenic products. These materials can be utilized either as whole plants or specific parts (e.g., roots, leaves, seeds, flowers) or as purified bioactive compounds.
For compound extraction, ethanol and methanol are commonly employed solvents owing to their efficiency in isolating target molecules. However, the choice of extraction solvents and conditions may influence the biological activity of the final product and potentially affect aquatic organisms. Application methods vary flexibly: medicinal plants can be administered individually or synergistically combined with other immunostimulants, either through water immersion, feed additives, or bioenrichment protocols. Both single-dose and combinatorial approaches have demonstrated efficacy, though optimal dosages and treatment durations require further species-specific validation.
The primary roles of medicinal plants in aquaculture include growth promotion, immune enhancement, and antimicrobial/antiviral activity. Despite their potential, the mechanistic pathways underlying these effects remain poorly elucidated. Consequently, most studies emphasize the need for further research before direct industrial application. Critical knowledge gaps include long-term safety assessments, interactions with aquatic microbiomes, and synergistic effects with probiotics. The exploration of plant–microecological agent combinations represents a promising frontier for sustainable aquaculture development [41].
CRISPR-Cas genome editing has emerged as a transformative tool for advancing fish genetics and disease management yet faces critical challenges in aquatic applications. Three fundamental limitations impede progress: (1) Off-target effects from imperfect guide RNA (gRNA) binding, which is particularly impactful in early developmental stages where genetic mosaicism propagates through rapidly dividing cells. (2) Delivery constraints, where conventional mammalian methods require adaptation for aquatic species, with microinjection dominating embryonic stages but lacking the tissue-specificity for adult applications. (3) Validation complexities involving labor-intensive detection methods (WGS, GUIDE-seq) to ensure edit accuracy and safety. Key mitigation strategies include bioengineered solutions like high-fidelity Cas9 variants (SpCas9-HF1/eSpCas9) and advanced delivery systems such as magnetite nanoparticles [42].
Vaccine development shows great promise; however, vaccines are typically suited for large-scale commercial operations and high-value finfish species [43], primarily addressing bacterial or viral infections. Significant challenges remain in developing vaccines for key parasites such as protozoa, crustaceans, amoebas, monogeneans, and worms [43,44] (the detailed discussion is shown in Table 2) [6,45,46,47,48,49,50,51,52,53,54,55,56,57,58]. Although vaccination technology looks promising, its practical application is limited due to serious side effects, such as growth damage, inflammation, visceral fiber adhesion, scar formation, and pigment deposition [43].
In addition, vaccinating fish is labor-intensive, time-consuming, and often results in inconsistent outcomes [18]. Phage therapy offers a targeted approach without harming gut microbiota or surrounding microbial communities. Bacteriophages have been used to control bacterial infections in aquaculture. Numerous studies have shown that bacteriophages are effective against the most destructive bacteria in aquaculture, such as Aeromonas, Edwardsiella, Flavobacterium, Lactococcus, Pseudomonas, Streptococcus, and Vibrio [59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85].
Previously, the application of bacteriophages in aquatic animals has been extensively reviewed, demonstrating their potential as biological control agents against bacterial infections (Table 3). Although bacteriophages are considered alternatives to traditional antibiotics for treating aquatic animal diseases, their practical application in aquaculture should consider key points:
  • The efficacy of bacteriophages must be supported by in vivo experiments.
  • Ensuring the safety of bacteriophages for humans and aquatic animals is non-negotiable.
  • Phage management should be economically feasible.
In theory, phage therapy could disrupt quorum sensing (QS) and biofilm formation by pathogens on solid surfaces, but practical application without impacting beneficial microbes remains uncertain. Furthermore, bacterial hosts may harbor virulence factors encoded by bacteriophages, potentially altering their pathogenicity [39]. Bacteriophages exhibit genetic diversity that enables them to overcome bacterial defenses through mechanisms such as adsorption inhibition, restriction-modification systems, and CRISPR-Cas systems, which can enhance infectivity and expand host ranges [41]. However, bacterial resistance to one phage can lead to susceptibility to others, and some phages have broad host ranges. In some cases, bacteriophages and their bacterial hosts may form mutualistic relationships, meaning phage resistance could promote the emergence of resistant bacterial strains [86]. Moreover, phage therapy’s reliance on live organisms makes it susceptible to point mutations and genetic drift, reducing treatment efficacy [87]. Despite these challenges, bacteriophage-based products—especially for Vibrio species—are already commercially available. However, more research is necessary before phage therapy can be widely adopted in aquaculture.
In contrast, feed additives, such as probiotics and prebiotics, offer broad-spectrum benefits. These additives can combat bacterial diseases and have shown efficacy against viruses, parasites, and protozoa.
Table 2. Overview of types of aquatic vaccines and their implications for aquatic disease resistance.
Table 2. Overview of types of aquatic vaccines and their implications for aquatic disease resistance.
Vaccine TypeAdministration MethodAdvantagesDisadvantagesApplication Examples (Pathogens/Diseases)Host FishSignificant EffectReference
Inactivated vaccineInjection, oral administration.High safety.
Easy production and storage.
Multiple vaccinations are required.
Short duration of immunity.
Salmon Aeromonas and Streptococcus infections.Salmon and tilapia.++[45,46,47]
Attenuated live vaccineInjection and soaking.Long lasting immunity.
Single vaccination is effective.
Potential risk of toxicity reversal.
Poor thermal stability.
Koi herpesvirus, Edwardsiella late-onset.Carp, sea bream.+++[48,49,50]
Subunit vaccineOral administration and injection.High purity and safety.
Targeted key antigens.
Adjuvant is needed to enhance the effect.
Weak immunogenicity.
Fish respiratory enterovirus (PRV), rhabdovirus.Atlantic salmon.++[51,52]
DNA vaccineIntamuscular injection.Inducing cellular immunity.
Rapid production.
Potential risks of genome integration.
Local absorption limitation.
IHNV, SVCV Bullet virus.Salmonidae fish.+++[53,54,55]
Vector vaccineInjection, oral administration.Multivalent antigen delivery.
Mucosal immune activation.
Risk of carrier autoimmune response.Recombinant Delayed Edwardsiella (RAEV).Zebrafish, salmon.++[6,56]
Nanoparticle vaccineOral administration, soaking.Enhance antigen stability. Targeted delivery.Long-term toxicity unknown.
High cost.
Virus-like particles (VLPs), ISCOM.Various freshwater fish.++[57,58]
(++) represents significance; (+++) represents highly significant.
Table 3. Phage trials of pathogens in aquatic animals and seafood.
Table 3. Phage trials of pathogens in aquatic animals and seafood.
PathogenDiseaseAnimal or Seafood SpeciesName of Phage (Morphology) 1Method of Application 2OutcomeReference
Aeromonas hydrophilaSepticemiaCatfish (Pangasianodon hypophthalmus)φ2 and φ5 (Myoviridae)I.P. injectionImproved survival rates from 18.3% to 100%.[59]
Loach (Misgurnus anguillicaudatus)Akh-2 (Siphoviridae)ImmersionReduced cumulative mortality rates from 100% to 56.67%.[60]
Loach (Misgurnus anguillicaudatu)AH1 (n.d.)Infection with phage before injection in fishPathogenicity eliminated following phage infection.[61]
Loach (Misgurnus anguillicaudatu)pAh1-C, pAh6-C (Myoviridae)I.P. injection and oral administration via feedingReduced cumulative mortality rates.
I.P. injection: from 100% to 43.33% using pAh1-C and to 16.67% using pAh6-C.
Oral administration: from 95.83% to 46.67% with pAh1-C and to 26.67% with pAh6-C.
[62]
A. salmonicidaFurunculosisRainbow trout (Oncorhynchus mykiss)PAS-1 (Myoviridae)I.M. injectionIncrease in survival rates from 0% to 26.7%.[62]
Senegalese sole (Solea senegalensis)AS-A (Myoviridae)ImmersionDecrease in cumulative mortality rates from 36% to 0%.[63]
Brook trout (Salvelinus fontinalis)HER 110 (Myoviridae)ImmersionDecrease in total mortality rates from 100% to 10%.[64]
Edwardsiella tardaEdwardsiellosisZebrafish (Danio rerio)ETP-1 (Podoviridae)Immersion prior to bacterial challengeSurvival rates improved from 18% to 68%.[65]
Flavobacterium columnareColumnaris diseaseRainbow trout and zebrafishFCL-2 (Myoviridae)ImmersionImproved survival rates.
Rainbow trout: increased from 8.3% to 50%.
Zebrafish: increased from 0% to 60%.
[66]
F. psychrophilumRainbow trout fry syndrome and cold water diseaseRainbow trout and Atlantic salmon (Salmo salar)1H, 6H (Siphoviridae)I.P. injectionReduced cumulative mortality rates.
Trout: decreased from 80% to 67% (1H), 47% (6H).
Salmon: decreased from 13% to 0% (1H), 6% (6H).
[67]
Lactococcus garvieaeLactococcosisYellowtail (Seriola quinqueradiata)PLgY-16 (Siphoviridae)I.P. injection and oral administration (feeding)I.P. injection: improved survival rates from 45% to 90%.
Oral administration: reduced cumulative mortality from 65% to 10%.
[68]
Pseudomonas plecoglossicidaBacterial hemorrhagic ascites diseaseAyu (Plecoglossus altivelis)PPpW-3 (Myoviridae), PPpW-4 (Podoviridae)Oral administration (feeding)Reduced cumulative mortality rates from 93.3% to 53.3% for PPpW-3, 40.0% for PPpW-4, and 20.0% for PPpW-3/W-4.[69]
Streptococcus agalactiaeStreptococcosisNile tilapia (Oreochromis niloticus)HN48 (n.d.)I.P. injectionImproved survival rates from 0% to 60%.[55]
S. iniaeStreptococcosisJapanese flounder (Paralichthys olivaceus)PSiJ31, 32, 41, 42 (Siphoviridae)I.P. injectionImproved survival rates from 0% to 28 or 33% (combined usage of PSiJ31 and 32); from 0% to 48, 70, or 90% (combined usage of PSiJ31, 32, 41, and 42).[70]
Vibrio anguillarumHemorrhagic septicemiaAtlantic salmonCHOED (n.d.)ImmersionSurvival rates improved from less than 10% to 100%.[71]
VibriosisZebrafish larvaeVP-2 (n.d.)ImmersionCumulative larval mortality rates reduced from 17% to 2%.[72]
V. harveyiLuminescent vibriosisShrimp (Penaeus monodon) larvaeA (Siphoviridae)ImmersionLarval survival rates improved from 17% to 86%.[73]
Shrimp larvaeVHM1, VHM2 (Myoviridae)
VHS1 (Siphoviridae)
ImmersionLarval survival rates improved from 26.6% to 86.6%.[74]
Abalone (Haliotis laevigata)vB_VhaS-tm (Siphoviridae)ImmersionLarval survival rates improved from 0% to 70%.[75]
Black tiger shrimp (Litopenaeus monodon) larvaeVHP6b (Siphoviridae)ImmersionCumulative larval mortality rates reduced from 70% to 20%.[76]
Shrimp larvaeViha10, Viha8 (Siphoviridae)ImmersionLarval survival rates improved from 65% to 88%.[77]
V. parahaemolyticus Blue mussel (Mytilus edulus)VP10 (n.d.)ImmersionReduction in bacterial growth to undetectable levels.[59]
Oyster (Crassostrea gigas)pVp-1 (Siphoviridae)Immersion, surface inoculationReduction in bacterial growth from 106 CFU/g to 10 CFU/g.[78]
Acute hepatopancreatic necrosis diseaseMarine shrimp (P. vannamei)pVp-1 (Siphoviridae)Immersion, oral administrationReduced cumulative mortality rates from 100% to 0%.[79]
V. splendidusVibriosisSea cucumber (Apostichopus japonicus)PVS-1, PVS-2 (Myoviridae)
PVS-3 (Siphoviridae)
Oral administration (feeding) and coelomic injectionSurvival rates increased from 18% to 82% through feeding and from 20% to 80% via coelomic injection.[60]
1 n.d. (not determined); 2 I.P. (intraperitoneal); I.M. (intramuscular).

3. Microecological Agents

3.1. Probiotics, Prebiotic, Synbiotics, and Postbiotics

3.1.1. Probiotics

The term probiotics is derived from the Latin word pro (“for”) and the Greek word biotic (“life”). Lilly and Stillwell first introduced this term in 1965 to describe substances that stimulate microbial growth. The FAO/WHO (2001) defines probiotics as live beneficial microorganisms that, when administered in adequate amounts, confer health benefits to the host by altering its associated microbial population [88]. Lazado and Caipang (2014) demonstrated the practical advantages of probiotics in aquaculture, building upon studies dating back to 1986 [89]. It is essential to understand the full context of this definition. Microbial cultures can only be classified as probiotics if their beneficial effects on the host have been confirmed through clinical studies or in vivo animal experiments. If evidence is limited to in vitro studies without in vivo confirmation, the microorganism is classified as a potential probiotic. Probiotics colonize animal intestines by maintaining or increasing favorable gut microbiota and restore the stability of gut microbiota disrupted by antibiotic treatment.
Probiotics benefit aquatic animals primarily in two ways:
  • Improving Water Quality: they aid in nitrification, denitrification, water quality management, and pathogen control.
  • Enhancing the Intestinal Microenvironment.
Compared to adult fish, juvenile fish possess immature immune systems and primarily depend on innate immunity for pathogen defense [90].
The mechanisms of probiotic action are multifactorial and context-dependent, presenting challenges for comprehensive elucidation (Table 4). Proposed mechanisms include the following:
  • The synthesis of antimicrobial metabolites.
  • Competitive exclusion (nutrient/attachment site competition).
  • The suppression of virulence-related gene expression.
  • Quorum-sensing interference.
  • Water quality modulation.
  • Immune function enhancement.
  • The provision of essential nutrients.
  • The facilitation of digestive enzyme activity [91].
To ensure efficacy and safety, probiotic strains must satisfy rigorous selection criteria:
  • Host species specificity.
  • Non-pathogenicity and genomic stability.
  • Antimicrobial compound production capacity.
  • Immunomodulatory competence.
Merrifield et al. (2010) proposed expanding these criteria to prioritize strains with validated probiotic functionalities, enabling optimized strain selection [92]. Nevertheless, identifying candidates fulfilling all requirements remains challenging, leading to the prevalent use of multi-strain probiotics [93] or synergistic formulations with prebiotics [94]. Combination strategies (e.g., probiotic consortia or synbiotics) generally demonstrate superior efficacy compared to single-strain applications. Critically, the introduction of probiotics harboring mobile antibiotic resistance genes into aquaculture systems is strongly discouraged due to horizontal gene transfer risks [95].
Before using probiotics in aquaculture, it is critical to screen for the presence of antibiotic resistance or virulence genes in mobile genetic elements [96].
The U.S. Food and Drug Administration (FDA) has approved several microorganisms for use in fish feed and aquatic environments, confirming their safety for direct application. In China, probiotics have been used in aquaculture since the 1980s, and their popularity has grown substantially. In recent years, over 100 companies have participated in the probiotic market, producing more than 50,000 tons of commercial probiotic products annually by Qi et al. (2009) [97] and Wang et al. [98]. In 2020, the European Commission authorized the use of Lactococcus lactis CNCM I-4622 (MA 18/5 M), developed by Lallemand Animal Nutrition. This probiotic, approved for aquaculture in the European Union, is widely recognized for improving the digestion of fish and shrimp. Other probiotic microorganisms approved by the European Union (EU) are internationally recognized for enhancing digestive efficiency in aquatic species through probiotic supplementation.
Lactic acid bacteria (LAB), exemplified by strains such as Lactobacillus acidophilus and Lactococcus lactis, are widely employed in aquaculture to enhance intestinal health and immune function in crucian carp (Carassius auratus). Studies indicate that LAB supplementation elevates serum immune biomarkers, including immunoglobulin M (IgM), lysozyme (LYZ), alkaline phosphatase (AKP), and superoxide dismutase (SOD) [99]. These markers correlate with three key immunological benefits:
(1)
IgM-mediated pathogen recognition;
(2)
LYZ-driven nonspecific immunity;
(3)
AKP-SOD antioxidant synergy against oxidative stress.
LAB exert dual antimicrobial mechanisms: the biofilm-mediated competitive exclusion of pathogens and the secretory modulation of mucosal immunity via intestinal epithelial interactions. The transcriptional regulation of inflammatory cytokines (IL-1β↑/IL-10↑) and antiviral factors (IFN-γ/TNF-α) further highlights LAB’s immunomodulatory potential. Recent screening identified LABy11 and y78 strains with enhanced efficacy [100], while interspecies variability in LAB responses necessitates further investigation into gut microbiota dynamics and bioactive compound mechanisms.
Similar findings have been reported for other probiotic strains (Table 5). Li et al. [101] studied the impact of adding Clostridium species to the diets of juvenile carp (Cyprinus carpio). The results showed that fish fed with this supplement experienced greater weight gain and specific growth rates. Furthermore, dietary Clostridium enhanced the mRNA expression of growth-related genes (PepT-1, PepT-2, and IGF-1) and genes associated with the TOR signaling pathway (TOR, 4E-BP2, and S6K1) [102]. In another study, different doses of LAB were added to Carassius auratus diets (5 × 103, 5 × 104, and 5 × 105 CFU/mL) over eight weeks. The results revealed improvements in water quality, reduced nitrogen concentration, better growth performance, increased serum antioxidant capacity, and the heightened activity of protease, digestive enzymes, and amylase.
Escherichia coli, a Gram-negative, rod-shaped bacterium within the family Enterobacteriaceae, is ubiquitously distributed across terrestrial, aquatic, and intestinal ecosystems. Despite its historical notoriety as an opportunistic pathogen (e.g., serotype O157:H7), emerging evidence highlights the probiotic potential of avirulent E. coli variants engineered through virulence gene deletion [103]. The Nissle 1917 strain (EcN), a prototypical non-pathogenic variant, demonstrates two key attributes for aquaculture applications: exceptional thermo-stability in lyophilized feed matrices and broad-spectrum antimicrobial activity mediated by microcin B17 synthesis, showing 82.4% growth inhibition against Pseudomonas aeruginosa ATCC 27,853 [103]. These findings underscore that targeted strain selection can repurpose E. coli from conditional pathogen to a precision biocontrol agent.
This paradigm is further exemplified by Enterobacter asburiae E7, a candidate probiotic isolated from the intestinal microbiota of Cyprinus carpio. Following rigorous safety screening (the absence of hly, stx1, and stx2 virulence factors), dietary supplementation with E7 at 107 CFU/g for 28 days induced significant immunomodulatory effects: the fold upregulation of *IL-1β* and TNF-α mRNA expression in head kidney leukocytes (qPCR, p < 0.01). A 57% reduction in mortality during Aeromonas veronii challenge trials compared to controls was identified [104].
Notably, while no significant growth promotion was observed, these results position E. asburiae E7 as a viable immunostimulant for sustainable cyprinid aquaculture [105].
Table 4. Mechanisms employed by probiotics to confer benefits.
Table 4. Mechanisms employed by probiotics to confer benefits.
DescriptionReference
1.Probiotics compete for resources and receptor binding sites, making it difficult for harmful microorganisms to survive in the intestine. They produce peptides, including anti-microbial peptides, viz., bacteriocins and β-defensins, to limit the pathogenic growth within the host species.[106,107]
2.Probiotics synthesize anti-bacterial substances, such as short chain fatty acids (SCFAs), e.g., propionate, butyrate, acetate, and H2O2. They also produce organic acids that lower the pH of the GIT, preventing harmful microorganisms from growing and supporting the propagation of probiotics. [107]
3.Probiotics improve intestinal barrier function by regulating the expressivity of tight junction proteins, including occludin, zonula occludens, and claudin, promoting defensin and mucin formation and regulating the immune function in the intestine.[108]
4.They affect both innate and adaptive immunity by modulating B- and T-cell, lysozyme, dendritic cell, complement and macrophage activities. Probiotics interact with intestinal epithelial cells, attracting macrophages and mononuclear cells, leading to the increased production of anti-inflammatory cytokines. They also inhibit pro-inhibitory markers by regulating the cytokines.[91]
5.Probiotics have the ability to produce neurotransmitters in the intestine through the gut–brain axis. Some strains can change the amounts of GABA, serotonin, and dopamine, which can have a positive effect on gastrointestinal motility and stress, as well as mood- and behavior-related pathways[109]
6.Probiotics alter the microbiome of the GIT through boosting the diversity of healthy microbes and decreasing the number of harmful ones.[110]
Table 5. More types of probiotics are being used in cyprinid fish.
Table 5. More types of probiotics are being used in cyprinid fish.
Empty CellProbioticsSpeciesReferences
AeromonasAeromonas veroniiGrass carp (Ctenopharyngodon idellus)[111]
Common carp (Cyprinus carpio)[112]
BacillusBacillus amyloliquefaciensCommon carp (Cyprinus carpio)[113]
Bacillus coagulans SCC-19Common carp (Cyprinus carpio)[114]
Bacillus subtilis
B. velezensis R-71003
Grass carp (Ctenopharyngodon idellus)[115]
Common carp (Cyprinus carpio)[116]
CarnobacteriumC. divergensCommon carp (Cyprinus carpio)[117]
EnterococcusEnterococcus faecium L6Grass carp (Ctenopharyngodon idellus)[118]
FlavobacteriumFlavobacterium sasangense BA-3Common carp (Cyprinus carpio)[112]
LactobacillusLactobacillus acidophilusGoldfish (Carassius auratus gibelio)[119]
Lactobacillus delbrueckiiCyprinus carpio Huanghe[120]
Lactobacillus caseiCommon carp (Cyprinus carpio)[121]
Lactobacillus sakeiCommon carp (Cyprinus carpio)[122]
Common carp (Cyprinus carpio)[123]
PediococcusPediococcus acidilactici MA18/5MCommon carp (Cyprinus carpio)[124]
PseudomonasPseudomonas aeruginosa VSG2Common carp
(Cyprinus carpio)
[125]
SaccharomycesSaccharomyces cerevisiae
Common carp (Cyprinus carpio)[126]
Shewanella xiamenensisGrass carp (Ctenopharyngodon idellus)[127]
VibrioVibrio lentus BA-2Common carp (Cyprinus carpio)[112]
Probiotic application in aquaculture faces multifaceted challenges. Technical constraints in mass-producing aquatic-derived strains, compounded by practitioners’ limited knowledge of administration protocols, have led to the predominant use of terrestrial alternatives [128]. Critical research gaps persist regarding
  • Ecological impacts on aquatic microbiomes.
  • Host-specific isolation hurdles from mucosal tissues [104].
  • Underexplored Bacillus-mediated mucosal immunity mechanisms. Environmental parameters (pH, temperature, dissolved oxygen) crucially influence probiotic viability, with alkalinity fluctuations altering metabolic rates [129,130,131].
Metabolic engineering breakthroughs, particularly CRISPR/Cas9 systems [132], enable targeted probiotic optimization through
  • Transporter engineering for enhanced substrate uptake.
  • Pathway refinement via toxic byproduct elimination.
  • Heterologous enzyme integration (cellulase/pectinase for SCFA production [133]).
  • Feedback-resistant overexpression strategies [134].
While probiotics demonstrate safety and eco-compatibility, they remain supplementary to pharmaceutical additives. Urgent priorities include the standardized validation of indigenous strains for disease prevention [105] and rigorous ecological risk assessments [128].
Probiotics like lactic acid bacteria and bifidobacteria lack the enzymes necessary to break down dietary fiber and complex polysaccharides. By introducing genes that encode enzymes such as cellulase, pectinase, and xylanase, these probiotics could metabolize complex substrates more effectively. This enhancement would improve their competitiveness and survival while promoting the production of short-chain fatty acids (SCFAs) [133].
Currently, while probiotics are regarded as safe and environmentally friendly feed additives, they cannot fully replace pharmaceutical additives in aquaculture.

3.1.2. Prebiotic

Prebiotics are non-digestible compounds that can be absorbed by the gut microbiota. They serve as essential nutrients for probiotic growth, selectively stimulating the proliferation and activity of specific probiotic strains. This promotes the growth of beneficial gut bacteria, enhances the gut microenvironment, strengthens host immunity, and reduces the need for antibiotics in aquaculture practices [135]. Common prebiotics primarily include oligosaccharides (composed of 2–10 monosaccharides) and polysaccharides. In aquaculture, widely used prebiotics include inulin and various oligosaccharides. Other prebiotics, such as bifidogenic factors and isomaltose, act as “nutrient media” for beneficial gut bacteria in aquatic animals. These compounds stimulate beneficial bacterial growth, increase their activity, and suppress harmful bacterial populations. Prebiotics can also enhance the activity of various enzymes in aquatic species.
Inulin, extracted from plants like lilies and chicory, is widely used due to its abundance and low cost. However, as of 2022, the application of oligosaccharides in fish farming remains limited.
The mechanisms by which prebiotics enhance the immune system in animals can be categorized into three primary aspects (Figure 3):
  • Regulating the gut microbiota.
  • Strengthening the intestinal mucosal barrier.
  • Directly activating the immune system.
Cells research shows that certain strains of lactic acid bacteria can regulate the maturation of dendritic (DC) cells, enhance NK cell and macrophage activity, and facilitate the differentiation of T-cells into Th1, Th2, or regulatory T-cells (T-reg), thereby improving immune responses. Additionally, prebiotics increase the populations of T and B lymphocytes.
Oligofructose, a water-soluble dietary fiber, has undergone extensive experimentation and has shown no evidence of genetic toxicity [136]. Hoseinifar S.H. (2017) demonstrated that adding short-chain oligofructose to carp feed significantly increased probiotic abundance in the fish’s gut microbiota [137]. Similarly, Dimitroglou A. (2009) reported that fish fed prebiotic-supplemented diets had significantly higher levels of short-chain fatty acids (SCFAs) in their intestines [138]. SCFAs, including acetic acid, propionic acid, and butyric acid, are key regulators of intestinal health. Butyric acid, in particular, nourishes intestinal epithelial cells, serves as a primary energy source for intestinal tissues, and supports immune cell development. Additionally, SCFA metabolites help lower intestinal pH, creating an unfavorable environment for pathogenic bacteria [139]. Therefore, prebiotics play a crucial role in enhancing fish gut health.

3.1.3. Synbiotics

The concept of synbiotics was first proposed by Gibson and Roberfroid in 1995 [140], with practical applications emerging in aquaculture by 1998. Synbiotics are defined as synergistic formulations combining probiotics and prebiotics, designed to selectively enhance the proliferation and metabolic activity of beneficial gut microbiota. This dual-action mechanism not only promotes the intestinal microbial dominance of probiotics but also augments host immunity and pathogen resistance through microbiome modulation [141]. While probiotics and prebiotics have been extensively utilized in aquaculture independently, synbiotics represent a paradigm shift by integrating their complementary functions. Their efficacy stems from the strategic pairing of specific probiotic strains with compatible prebiotic substrates, thereby optimizing microbial colonization and host–microbe crosstalk [142]. This synergy facilitates three key outcomes: enhanced niche competition against pathogens, the selective amplification of endogenous beneficial species, and systemic immunoregulation [143].
Competitive exclusion constitutes a cornerstone of probiotic functionality in gut ecosystems, operating through spatial and nutritional antagonism. Upon intestinal colonization, probiotics adhere to mucosal epithelia, creating a physicochemical microenvironment unfavorable to pathogens [144]. Vine et al. (2004) demonstrated this phenomenon through in vitro models comparing five probiotic strains (e.g., Lactobacillus spp.) against enteropathogens like Aeromonas hydrophila [145]. Furthermore, under nutrient-limiting conditions, probiotics exhibit metabolic flexibility to outcompete pathogens for critical substrates (e.g., iron-siderophores, oligosaccharides) [145,146].
Beyond direct microbial antagonism, synbiotics modulate immune homeostasis through microbiota-derived signals. Probiotic metabolites (e.g., short-chain fatty acids) are prime innate immune effectors—notably enhancing natural killer (NK) cell cytotoxicity by folding and upregulating interleukin-10 (IL-10) production in lymphocytes [147]. These immunostimulatory effects are amplified when combined with prebiotics, which act as vaccine adjuvants by prolonging antigen-presenting cell (APC) activation and antibody titer persistence [148,149]. However, three critical considerations must be addressed in application:
  • Compatibility: prebiotic physicochemical properties (e.g., molecular weight, solubility) must align with antigen delivery systems to prevent epitope masking [148].
  • Dose Optimization: immunomodulatory effects follow U-shaped dose-response curves, requiring empirical determination to avoid immunosuppression (low dose) or inflammatory cascades (high dose) [147].
  • Safety Profiling: chronic exposure risks include microbiota dysbiosis and hepatic xenobiotic metabolism alterations, necessitating longitudinal toxicity assessments [149].
Oral administration (feed addition) is the main approach in the large-scale aquaculture of cyprinid fish, but the survival rate and intestinal colonization efficiency of prebiotics in feed processing need to be considered. The feasibility of injection or immersion vaccination is low, and the production cost of prebiotics needs to be balanced with the degree of vaccine protection efficacy enhancement, especially in low-value-added fish species; this needs to be carefully evaluated. Establish quality control standards for the purity of prebiotics and the concentrations of active ingredients (such as SCFAs) to ensure consistency between batches. Different pathogens (such as bacterial vs. viral) may need to be matched with specific prebiotics (such as bacteriocins targeting bacterial vaccines), and the immune system differences of host species also need to be considered. Finally, long-term use may alter the microbial community structure of aquaculture water bodies or wild populations, and its potential impact on environmental microbial resistance and ecological balance needs to be evaluated. Oligosaccharides, a type of prebiotic, can stimulate the liver to secrete specific proteins that bind with oligosaccharides to form glycoproteins, which subsequently influence the immune system [150]. Dietary supplementation with Bacillus subtilis and/or oligosaccharides has been shown to significantly increase the levels of alkaline phosphatase (AKP), acid phosphatase (ACP), and lysozyme (LZM) in various fish species, including crucian carp [150], carp [151], and grass carp [152]. Moreover, the supplement effectively mitigated the upregulation of inflammatory cytokines (IL-6, IL-8, IL-1β, and TNF-α) and the downregulation of anti-inflammatory cytokines (IL-10 and TGF-β) caused by NaHCO3 exposure. This indicates that NaHCO3 stress disrupts immune homeostasis in crucian carp, leading to intestinal inflammation, whereas synbiotics can enhance immune function and alleviate this inflammation [153]. In this study, the combination addition had the best effect. Research indicates that xylooligosaccharides (XOSs), fermented by gut microbiota, enhance intestinal epithelial barrier integrity and stimulate enterocyte proliferation, thereby promoting gut structural development and nutrient absorption [151]. XOS fermentation by probiotics generates small organic acids (e.g., SCFAs), lowering the intestinal pH and suppressing pathogenic bacterial growth [152]. Notably, SCFAs—classified as postbiotics—play dual roles in immune regulation:
  • Innate Immunity: activate GPR43 to induce neutrophil chemotaxis, functional activation, and the modulation of innate immune cells [152,154].
  • Adaptive Immunity: enhance T-cell and B-cell responses, bolstering mucosal and systemic antibody production, thereby improving disease resistance in fish.
These findings suggest that Bacillus subtilis may synergize with oligosaccharides by producing beneficial metabolites (e.g., SCFAs) during fermentation.
Despite obtaining promising results, the exact mechanisms of synbiotics remain controversial. The interactions between probiotics and native gut microbiota are often explained through general ecological concepts like saprophytism and competition [153].
There are three main types of interactions between probiotic effector molecules and intestinal mucosal receptors.
The first type involves the identification of probiotics, lectins, and carbohydrates on the surface of intestinal mucosa. Many probiotics express lectins as effector molecules, binding probiotics to carbohydrate conjugates on the cell surface, such as probiotic extracellular polysaccharides, capsule polysaccharides, and intestinal adhesion molecules like membrane mucin [155].
The second type involves the interaction between host cell proteins and probiotic surface proteins, such as the binding of probiotic pili to intestinal mucosal mucin [156].
The third type involves hydrophobic interactions between hydrophobic proteins on the surface of probiotics and complementary hydrophobic protein fragments on animal cells, such as the binding of probiotic surface proteins to intestinal mucosal mucins [157].
However, there is still limited systematic research on how probiotics colonize the intestine, alter microbial ecosystems, or interact synergistically with prebiotics.
Additionally, systematic research on the compatibility and synergistic effects between probiotics and prebiotics is lacking. Current selection processes for prebiotics compatible with probiotics are simplistic, limiting their ability to selectively promote probiotic proliferation [158]. There is also a need for deeper investigation into the biological activity of oligosaccharides with varying molecular weights and degrees of polymerization.

3.1.4. Postbiotics

The concept of postbiotics was first introduced in 2013. In 2021, the International Scientific Association for Probiotics and Prebiotics (ISAPP) defined postbiotics in Nature Reviews Gastroenterology & Hepatology as “preparations of non-living microorganisms and/or their components that confer health benefits to the host” [159,160].
Postbiotics are defined as non-viable microbial formulations derived from inactivated microbial cells, bacterial cellular components, and crude metabolic byproducts generated during fermentation processes [161,162]. These compounds are primarily sourced from probiotic microorganisms spanning diverse genera and species, though not all probiotic strains are suitable for postbiotic production, necessitating a stringent selection process. Currently, only microbial strains that pass rigorous safety assessments—including bacteria (e.g., Lactobacillus spp., such as L. plantarum, L. fermentum, and L. rhamnosus; Bifidobacterium spp., like B. bifidum and B. animalis; and Enterococcus, Bacillus, and Pseudomonas strains) and fungi (primarily yeast)—are utilized. Crucially, postbiotics exclude inactivated pathogens, viruses, or isolated microbial metabolites.
Chemically, postbiotics encompass a heterogeneous mixture of bioactive components, including the following [163]:
  • Structural cell elements: e.g., peptidoglycan, teichoic acids, and cellular debris from lysed probiotics.
  • Metabolic products such as short-chain fatty acids (SCFAs: acetate, propionate, butyrate), organic acids (e.g., lactate), enzymes (proteases, lipases), antimicrobial agents (e.g., bacteriocins), vitamins (B-complex, vitamin K), and antioxidants (catalase, superoxide dismutase).
  • Other bioactive molecules, like nucleotides and their derivatives.
The selection of microbial strains for postbiotic synthesis prioritizes safety, stability, and functionality, often favoring those with a safe status. Postbiotics are distinguished from purified microbial metabolites or single-compound extracts, as their efficacy is attributed to synergistic interactions among their complex components. Current research focuses on their applications in gut microbiota modulation, immune enhancement, anti-inflammatory responses, and metabolic regulation, particularly through SCFA-mediated intestinal barrier reinforcement. However, challenges remain in standardizing production protocols and elucidating mechanistic synergies among constituent molecules.
Postbiotics offer similar health benefits to live probiotics but have several advantages:
  • Defined Chemical Structure and Stability: Postbiotics have a clear chemical composition, greater stability, and a longer shelf life [164];
  • High Safety Profile: Since postbiotics are non-living, they cannot acquire virulence genes or transmit antibiotic resistance, reducing the risk of producing harmful metabolites. This makes them safer for consumption, and they are effective at lower doses [165].
  • Lower Risk for Immunocompromised Hosts: Active probiotics require large doses to be effective, which may cause adverse reactions in immunocompromised individuals. Postbiotics, however, are safer and cause minimal impact even in higher amounts [163].
  • Ease of Storage and Transport: Postbiotics do not need to maintain viability, allowing them to remain stable across a wide range of temperatures and pH levels. This makes them easier to store and transport [166].
  • Environmental Resilience: Postbiotics retain their beneficial effects under extreme environmental conditions, such as high temperatures or exposure to digestive enzymes [167].
Postbiotics are mainly related to immune regulatory activity, which can stimulate innate and adaptive immune systems, stimulate the generation and differentiation of immune cells, release inhibitory inflammatory cytokines, and thus exert anti-inflammatory activity. Postbiotics can [168] attach to intestinal epithelial cells and directly regulate immune function; microfold cells located in the epithelium, which are transported to immune cells; bind with dendritic cells, leading to the activation of cascade reactions, stimulating immune responses. Research has shown that adding endogenous elements to feed can promote the growth of aquatic animals by increasing their food intake, protein content, and energy, improving feed utilization efficiency and thereby achieving the goal of promoting growth; increasing the length of intestinal villi in aquatic animals, thus increasing the absorbent surface area, improving nutrient utilization efficiency and thereby enhancing growth performance [169]; and adjusting the quantity of beneficial bacteria in the intestine to maintain the balance of gut microbiota, improve nutrient absorption and feed utilization, and promote body growth [166].
Postbiotics can regulate beneficial gut microbiota, increase intestinal mucosal thickness and villus length, strengthen the body’s natural defense mechanisms, and have an important impact on maintaining gastrointestinal health. For example, heat inactivated Lactobacillus acidophilus reduces diarrhea caused by Escherichia coli through space-occupying competition [169].
Postbiotics, via their bioactive components, regulate intestinal immune responses to boost mucin production. These mucins support symbiotic bacteria by providing adhesion sites and nutrients while inhibiting pathogens, restoring the gut microbiota balance, and preventing dysbiosis-related diseases [170]. A case in point is Stress–Worry-Free Concentration® (SWFC), a postbiotic derived from Cetobacterium somerae and Lactococcus lactis supernatants. A study on Cyprinus carpio fed high-fat diets (HFDs) evaluated SWFC’s effects (0, 0.2, and 0.3 g/kg supplementation over 98 days) [171].
Though growth remained unaffected, SWFC improved health markers:
Skin mucosa: Enhanced C3 and antioxidant capacity (T-AOC, SOD) and reduced lipid peroxidation (MDA) (p < 0.05).
Liver health: The 0.2 g/kg dose significantly lowered serum ALT, AST, and hepatic triglycerides (p < 0.05/0.01), suppressed pro-inflammatory genes (TNF-α, IL-1β) and lipid synthesis genes (ACC, FAS, PPAR-β/γ), and upregulated anti-inflammatory TGF-β.
Gut microbiota: SWFC restored the microbial equilibrium, mitigating HFD-induced oxidative stress and inflammation [165].
In summary, postbiotics like SWFC reduce feed costs, enhance preservation, and improve animal health, offering a sustainable solution for advancing aquaculture productivity and addressing intensive farming challenges.

3.2. Antimicrobial Peptides

Antimicrobial peptides (AMPs) are small molecules with antibacterial properties. Compared to antibiotics, AMPs are less likely to lead to resistance and can be considered potential treatments for antibiotic-resistant bacteria [168]. They promote fish growth, enhance immunity, and increase disease resistance. AMPs offer a promising alternative in the livestock industry to combat antibiotic overuse. Originating from a variety of life forms, they possess antibacterial, antiviral, and antifungal properties. In aquaculture, adding AMPs to fish feed can enhance fish health and improve resistance to pathogens [172] (Figure 4).
Among the most studied AMPs are Hepcidin and Cecropin [173]. Research shows that AMPs can inhibit a variety of pathogens [174], including Aeromonas hydrophila, and improve fish growth and antioxidant response to better understand their effects on fish gut microbiota [175].
Hepcidin’s primary functions include regulating iron metabolism during immune responses by limiting the supply of iron to bacteria, thus restricting pathogen growth; exhibiting direct antibacterial activity to kill pathogens; and modulating host immunity through interactions with the host immune system [176]. In aquaculture, Hepcidin may act as an immune modulator and an antibacterial therapeutic. Studies have shown that fish eggs expressing Hepcidin can serve as a novel feed supplement, enhancing the immunity of zebrafish against pathogenic infections [177]. Although Hepcidin shows potential as an antibiotic replacement, challenges remain in its practical application, such as its instability in terms of hemolysis and protein hydrolysis [178]. For example, in grass carp, Hepcidin inhibits bacterial growth through two mechanisms: damaging bacterial cell membranes and interacting with bacterial genomic DNA (gDNA). Other antimicrobial peptides, such as tetracycline, NK-18, and bacteriocins, also kill bacteria by binding to DNA. Research indicates that Hepcidin-25 (a mature peptide) and Hepcidin-20 (a truncated peptide) exhibit significant bactericidal activity against both Gram-positive and Gram-negative bacteria, partially through membrane disruption and binding to gDNA. The disruption of bacterial cell membranes by grass carp Hepcidin is a new discovery, as other Hepcidins do not exhibit this function. The antimicrobial peptide NK-18 kills bacteria by disrupting cell membranes and binding to DNA [177]. This dual-target strategy makes it difficult for bacteria to develop resistance to these peptides [179]. Therefore, Hepcidin may be a promising candidate for treating bacterial infections. Additionally, iron regulation promotes antibacterial defense by directly killing microorganisms and regulating iron metabolism in grass carp.
One previous study has shown that Hepcidin may also exert strong antibacterial effects in vivo through synergistic interactions with other inducible acute phase response proteins or tissue-specific antibacterial compounds, such as moxorubicin [180]. However, further research is needed to explore whether there are iron-coordinated molecules present in grass carp. As expected, the prophylactic or therapeutic administration of Hepcidin increased the survival rate of grass carp infected with lethal doses of Aspergillus niger, accompanied by a reduction in bacterial numbers in the fish’s foregut and spleen. The therapeutic effect of grass carp Hepcidin was found to be more effective than its preventive use [181].
In the future, it is necessary to continuously study the function of Hepcidin and optimize its expression system to obtain more easily applicable antimicrobial peptides.

4. Conclusions and Future Perspective

Current regulatory frameworks in some regions approve probiotics based solely on preliminary assessments of antibacterial activity and immunostimulatory potential. However, a 2017 U.S. FDA (Silver Spring, MD) inspection revealed >50% of probiotic manufacturers violated protocols, primarily through strain misidentification and microbial contamination, jeopardizing product safety and efficacy [182]. While antibiotic alternatives demonstrate growth-promoting potential, their performance exhibits geographic variability due to divergent farming practices, regional disease profiles, and management heterogeneity.
This review systematically analyzes microecologics (definition, mechanisms, case studies) in cyprinid aquaculture, highlighting safety concerns as the primary constraint for large-scale adoption. We emphasize science-driven development requiring the rigorous validation of novel formulations.
Sustainable aquaculture advancement necessitates integrated strategies beyond singular additives:
  • Precision nutrition.
  • Biosecurity-enhanced management.
  • Water quality optimization.
  • Preventive health protocols.
Such multidisciplinary approaches synergistically enhance host resilience, reduce disease incidence, and minimize therapeutic reliance, ultimately ensuring the eco-friendly production of premium aquatic products. Though certain additives show promise through dual antimicrobial–nutritive functions, realizing their full potential requires collaborative innovation among farmers, nutritionists, and microbiologists. Critical research priorities include
  • Cost–benefit analyses.
  • Dose–response relationships.
  • Environmental fate assessments.
  • Smallholder applicability.
Immunometabolic modulation mechanisms in practical applications that align aquaculture practices with international guidelines for antimicrobial resistance (AMR), management, and the responsible use of feed additives—including antibiotics and their alternatives—are fundamental for reducing the risk of AMR. Research should prioritize fish health with a strong focus on disease prevention. Key areas of focus should include the following:
  • Evaluating the efficacy and safety of feed additives under varying environmental conditions.
  • Assessing the environmental impact and alternative methods for antimicrobial agents.
  • Implementing the active and passive monitoring of drug withdrawal periods, wastewater treatment, residues, and AMR detection.
By focusing on these areas, aquaculture can adopt more sustainable practices, improve fish health, minimize AMR risks, and secure the long-term sustainability of aquaculture.
A major limitation of this article is its exclusive focus on antibiotic residues in freshwater cyprinid fish. The article primarily addresses strategies to improve the internal health of fish and enhance their immunity to achieve antibiotic-free aquaculture. However, it does not provide detailed information on salt-tolerant cyprinid species or discuss the interactions between beneficial bacteria and environmental factors that could improve external water conditions. Additionally, the article does not explore how nutritional strategies could further enhance the immune systems of cyprinid fish.
These gaps will be addressed in future research.

Author Contributions

Conceptualization, R.Q. and H.W.; methodology, R.Q.; software, R.Q.; validation, H.W.; formal analysis, H.W.; investigation, A.Y.G.; resources, Q.C.; data curation, R.Q.; R.Q.: writing—original draft. H.W., A.Y.G. and A.M.Y.: validation, project administration, and writing—review and editing. A.M.Y. and Q.C.: validation, supervision, project administration, funding acquisition, and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Talent Recruitment Program (24030403699) and Provincial Undergraduate Training Program on Innovation and Entrepreneurship (No. S202410626065 and S202510626039).

Data Availability Statement

No data were used for the research described in the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tan, M.; Armbruster, J.W. Phylogenetic classification of extant genera of fishes of the order Cypriniformes (Teleostei: Ostariophysi). Zootaxa 2018, 4476, 6–39. [Google Scholar] [CrossRef] [PubMed]
  2. Mulchandani, R.; Wang, Y.; Gilbert, M.; Van Boeckel, T.P. Global trends in antimicrobial use in food-producing animals: 2020 to 2030. PLOS Glob. Public Health 2023, 3, e0001305. [Google Scholar] [CrossRef] [PubMed]
  3. Wenning, R. The state of world fisheries and aquaculture (sofia) 2020 report. Integr. Environ. Assess. Manag. 2020, 16, 800–801. [Google Scholar]
  4. Atterbury, R.J.; Tyson, J. Predatory bacteria as living antibiotics—Where are we now? Microbiology 2021, 167, 001025. [Google Scholar] [CrossRef]
  5. Hou, Y.H.; Cao, Y.L. Discussion on several common diseases of carp and their prevention and treatment techniques. China Fish. 2023, 79–82. [Google Scholar]
  6. Irshath, A.A.; Rajan, A.P.; Vimal, S.; Prabhakaran, V.-S.; Ganesan, R. Bacterial pathogenesis in various fish diseases: Recent advances and specific challenges in vaccine development. Vaccines 2023, 11, 470. [Google Scholar] [CrossRef]
  7. Rodriguez-Mozaz, S.; Vaz-Moreira, I.; Giustina, S.V.D.; Llorca, M.; Barceló, D.; Schubert, S.; Berendonk, T.U.; Michael-Kordatou, I.; Fatta-Kassinos, D.; Martinez, J.L.; et al. Antibiotic residues in final effluents of European wastewater treatment plants and their impact on the aquatic environment. Environ. Int. 2020, 140, 105733. [Google Scholar] [CrossRef]
  8. Okocha, R.C.; Olatoye, I.O.; Adedeji, O.B. Food safety impacts of antimicrobial use and their residues in aquaculture. Public Health Rev. 2018, 39, 1–22. [Google Scholar] [CrossRef]
  9. Chen, J.; Sun, R.; Pan, C.; Sun, Y.; Mai, B.; Li, Q.X. Antibiotics and food safety in aquaculture. J. Agric. Food Chem. 2020, 68, 11908–11919. [Google Scholar] [CrossRef]
  10. Joseph, A.M.M.; Linda, D.A.K.; Irina, V.P.; Larissa, A.S.; Souadkia, S.; Ibrahim, K.; Milana, S.D. The public health issue of antibiotic residues in food and feed: Causes, consequences, and potential solutions. Vet. World 2022, 15, 662–671. [Google Scholar]
  11. Ben, Y.; Fu, C.; Hu, M.; Liu, L.; Wong, M.H.; Zheng, C. Human health risk assessment of antibiotic resistance associated with antibiotic residues in the environment: A review. Environ. Res. 2018, 169, 483–493. [Google Scholar] [CrossRef] [PubMed]
  12. Puvača, N.; Vapa-Tankosić, J.; Ignjatijević, S.; Carić, M.; Soleša, D.; Soleša, K. Consumer awareness of antimicrobal residues in drinking Water. Ekon. Misao I Praksa-Econ. Thought Pract. 2023, 16, 40–56. [Google Scholar] [CrossRef]
  13. Mirakian, R.; Leech, S.C.; Krishna, M.T.; Richter, A.G.; Huber, P.A.J.; Farooque, S.; Khan, N.; Pirmohamed, M.; Clark, A.T.; Nasser, S.M. Management of allergy to penicillins and other beta-lactams. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2015, 45, 300–327. [Google Scholar] [CrossRef] [PubMed]
  14. Del Pozzo-Magaña, B.R.; Liy-Wong, C. Drugs and the Skin: A concise review of cutaneous adverse drug reactions. Br. J. Clin. Pharmacol. 2024, 90, 1838–1855. [Google Scholar] [CrossRef]
  15. Zhang, C.; Chen, Y.; Chen, S.; Guan, X.; Zhong, Y.; Yang, Q. Occurrence, risk assessment, and in vitro and in vivo toxicity of antibiotics in surface water in China. Ecotoxicol. Environ. Saf. 2023, 255, 114817. [Google Scholar] [CrossRef]
  16. Seo, J.; Kloprogge, F.; Smith, A.M.; Karu, K.; Ciric, L. Antibiotic residues in UK Foods: Exploring the exposure pathways and associated health risks. Toxics 2024, 12, 174. [Google Scholar] [CrossRef]
  17. Ribeiro, C.F.A.; Silveira, G.G.D.O.S.; Cândido, E.D.S.; Cardoso, M.H.; Espínola Carvalho, C.M.; Franco, O.L. Effects of antibiotic treatment on gut microbiota and how to overcome its negative impacts on human health. Acs Infect. Dis. 2020, 6, 2544–2559. [Google Scholar] [CrossRef]
  18. O’Neill, A.J.; Chopra, I. Preclinical evaluation of novel antibacterial agents by microbiological and molecular techniques. Expert Opin. Investig. Drugs 2005, 13, 1045–1063. [Google Scholar] [CrossRef]
  19. Bao, R.; Yang, Y.; Chen, H.; Li, Y. Occurrence, distribution and health risk assessment of quinolone residues in cultured fish in southeast China. J. Environ. Sci. Health Part B Pestic. Food Contam. Agric. Wastes 2024, 59, 714–724. [Google Scholar] [CrossRef]
  20. EFSA Panel on Contaminants in the Food Chain (CONTAM). Scientific Opinion on nitrofurans and their metabolites in food. EFSA J. 2015, 13, 4140. [Google Scholar]
  21. Bush, N.G.; Diez-Santos, I.; Abbott, L.R.; Maxwell, A. Quinolones: Mechanism, Lethality and Their Contributions to antibiotic resistance. Molecules 2020, 25, 5662. [Google Scholar] [CrossRef] [PubMed]
  22. Drlica, K.; Zhao, X. Bacterial death from treatment with fluoroquinolones and other lethal stressors. Expert Rev. Anti-Infect. Ther. 2020, 19, 11–18. [Google Scholar] [CrossRef] [PubMed]
  23. Qi, X.; Yun, C.; Pang, Y.; Qiao, J. The Impact of the Gut Microbiota on the Reproductive and Metabolic Endocrine System. Gut Microbes 2021, 13, 1894070. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, J.; Ying, G.-G.; Deng, W.J. Antibiotic Residues in Food: Extraction, Analysis, and Human Health Concerns. J. Agric. Food Chem. 2019, 67, 7569–7586. [Google Scholar] [CrossRef]
  25. Kumar, A.; Pal, D. Antibiotic resistance and wastewater: Correlation, impact and critical human health challenges. J. Environ. Chem. Eng. 2018, 6, 52–58. [Google Scholar] [CrossRef]
  26. Wang, X.; Li, F.; Hu, X.; Hua, T. Electrochemical advanced oxidation processes coupled with membrane filtration for degrading antibiotic residues: A review on its potential applications, advances, and challenges. Sci. Total Environ. 2021, 784, 146912. [Google Scholar] [CrossRef]
  27. Yang, X.; Chen, Z.; Zhao, W.; Liu, C.; Qian, X.; Zhang, M.; Wei, G.; Khan, E.; Ng, Y.H.; Ok, Y.S. Recent advances in photodegradation of antibiotic residues in water. Chem. Eng. J. 2021, 405, 126806. [Google Scholar] [CrossRef]
  28. Farid, M.U.; Choi, P.J.; Kharraz, J.A.; Lao, J.-Y.; St-Hilaire, S.; Ruan, Y.; Lam, P.K.S.; An, A.K. Hybrid nanobubble-forward osmosis system for aquaculture wastewater treatment and reuse. Chem. Eng. J. 2022, 435, 135164. [Google Scholar] [CrossRef]
  29. Melchiorri, D.; Rocke, T.; Alm, R.A.; Cameron, A.M.; Gigante, V. Addressing urgent priorities in antibiotic development: Insights from WHO 2023 antibacterial clinical pipeline analyses. Lancet. Microbe 2024, 6, 100992. [Google Scholar] [CrossRef]
  30. He, W.; Martin, J.H.; Shaw, P.N.; Lu, X.; Walpole, E.T.; Dimeski, G. A Simple and sensitive LC-MS/MS method for the simultaneous determination of cyclophosphamide and doxorubicin concentrations in human plasma. Curr. Pharm. Anal. 2017, 14, 53–59. [Google Scholar] [CrossRef]
  31. Yu, X.; Liu, H.; Pu, C.; Chen, J.; Sun, Y.; Hu, L. Determination of multiple antibiotics in leafy vegetables using QuEChERS-UHPLC-MS/MS. J. Sep. Sci. 2018, 41, 713–722. [Google Scholar] [CrossRef] [PubMed]
  32. Sun, X.; Wang, J.; Li, Y.; Yang, J.; Jin, J.; Shah, S.M.; Chen, J. Novel dummy molecularly imprinted polymers for matrix solid-phase dispersion extraction of eight fluoroquinolones from fish samples. J. Chromatogr. A 2014, 1359, 1–7. [Google Scholar] [CrossRef] [PubMed]
  33. Jiménez, V.; Rubies, A.; Centrich, F.; Companyó, R.; Guiteras, J. Development and validation of a multiclass method for the analysis of antibiotic residues in eggs by liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2011, 1218, 1443–1451. [Google Scholar] [CrossRef] [PubMed]
  34. Huang, P.; Zhao, P.; Dai, X.; Hou, X.; Zhao, L.; Liang, N. Trace determination of antibacterial pharmaceuticals in fishes by microwave-assisted extraction and solid-phase purification combined with dispersive liquid–liquid microextraction followed by ultra-high performance liquid chromatography-tandem mass spectrometry. J. Chromatogr. B 2016, 1011, 136–144. [Google Scholar]
  35. Dai, T.; Duan, J.; Li, X.; Xu, X.; Shi, H.; Kang, W.; Huang, K. Determination of sulfonamide residues in food by capillary zone electrophoresis with on-line chemiluminescence detection based on an Ag(III) complex. Int. J. Mol. Sci. 2017, 18, 1286. [Google Scholar] [CrossRef]
  36. Desimoni, E. About CCα and CCβ as introduced by the commission decision of 12 August 2002 implementing Council Directive 96/23/EC. Accredit. Qual. Assur. 2004, 9, 724–725. [Google Scholar] [CrossRef]
  37. Liu, K.J. Research on the detection method of residual antibiotics in food. Mod. Food 2024, 30, 95–97. [Google Scholar]
  38. Costa, L.F.; da Silva, G.S.; Freitas, A.S.; da Silva, G.S.; de Sousa, E.R. Application of dispersive liquid-liquid microextraction technique for the analysis of norfloxacin antibiotic in chicken breast (pectoralis major) by high performance liquid chromatography with fluorescence detector. Rev. Virtual Química 2020, 12, 681–692. [Google Scholar] [CrossRef]
  39. Gunning, D.; Maguire, J.; Burnell, G. The Development of sustainable saltwater-based food production systems: A review of established and novel concepts. Water 2016, 8, 598. [Google Scholar] [CrossRef]
  40. Murray, K.N.; Clark, T.S.; Kebus, M.J.; Kent, M.L. Specific Pathogen Free – A review of strategies in agriculture, aquaculture, and laboratory mammals and how they inform new recommendations for laboratory zebrafish. Res. Vet. Sci. 2022, 142, 78–93. [Google Scholar] [CrossRef]
  41. Hai, V.N. The use of medicinal plants as immunostimulants in aquaculture: A review. Aquaculture 2015, 44, 688–696. [Google Scholar]
  42. Marnis, H.; Syahputra, K. Advancing fish disease research through CRISPR-Cas genome editing: Recent developments and future perspectives. Fish Shellfish Immunol. 2025, 160, 110–220. [Google Scholar] [CrossRef] [PubMed]
  43. Brudeseth, B.E.; Wiulsrød, R.; Fredriksen, B.N.; Lindmo, K.; Løkling, K.-E.; Bordevik, M.; Steine, N.; Klevan, A.; Gravningen, K. Status and future perspectives of vaccines for industrialised fin-fish farming. Fish Shellfish Immunol. 2013, 35, 1759–1768. [Google Scholar] [CrossRef] [PubMed]
  44. Seed, K.D. Battling Phages: How bacteria defend against viral attack? PLoS Pathog. 2015, 11, e1004847. [Google Scholar] [CrossRef]
  45. Ma, J.; Bruce, T.J.; Jones, E.M.; Cain, K.D. A review of fish vaccine development strategies: Conventional methods and modern biotechnological approaches. Microorganisms 2019, 7, 569. [Google Scholar] [CrossRef]
  46. Hølvold, L.B.; Myhr, A.I.; Dalmo, R.A. Strategies and hurdles using DNA vaccines to fish. Vet. Res. 2014, 45, 21. [Google Scholar] [CrossRef]
  47. Subasinghe, R.P.; Curry, D.; McGladdery, S.E.; Bartley, D. Recent technological innovations in aquaculture. Rev. State World Aquac. FAO Fish. Circ. 2003, 886, 59–74. [Google Scholar]
  48. Triet, T.H.; Tinh, B.T.T.; Hau, L.V.; Huong, T.V.; Binh, N.-Q. Development and potential use of an Edwardsiella ictaluri wzz mutant as a live attenuated vaccine against enteric septicemia in Pangasius hypophthalmus (Tra catfish). Fish Shellfish Immunol. 2019, 87, 87–95. [Google Scholar] [CrossRef]
  49. Minor, P.D. Live attenuated vaccines: Historical successes and current challenges. Virology 2015, 479–480, 379–392. [Google Scholar] [CrossRef]
  50. Ruiz de Ybáñez, M.R.; Laura, R.; César, F.; Pilar, M.; Eduardo, B.; Silvia, R.; Carlos, M. Monitoring for Anguillicoloides crassus, Anguillid herpesvirus 1, aquabirnavirus EVE and rhabdovirus EVEX in the European eel population of southern Spain. J. Fish Dis. 2023, 46, 417–431. [Google Scholar] [CrossRef]
  51. Björklund, H.V.; Higman, K.H.; Kurath, G. The glycoprotein genes and gene junctions of the fish rhabdoviruses spring viremia of carp virus and hirame rhabdovirus: Analysis of relationships with other rhabdoviruses. Virus Res. 1996, 42, 65–80. [Google Scholar] [CrossRef] [PubMed]
  52. Siwicki, A.K.; Pozet, F.; Morand, M.; Kazuń, B.; Trapkowska, S.; Małaczewska, J. Influence of methisoprinol on the replication of rhabdoviruses isolated from carp (Cyprinus carpio) and catfish (Ictalurus melas): In vitro study. Pol. J. Vet. Sci. 2003, 6, 47–50. [Google Scholar] [PubMed]
  53. Stone, D.M.; Ahne, W.; Denham, K.L.; Dixon, P.F.; Liu, C.Y.; Sheppard, A.M.; Taylor, G.R.; Way, K. Nucleotide sequence analysis of the glycoprotein gene of putative spring viraemia of carp virus and pike fry rhabdovirus isolates reveals four genogroups. Dis. Aquat. Org. 2003, 53, 203–210. [Google Scholar] [CrossRef] [PubMed]
  54. Coll, J.M. Herpesvirus Infection induces both specific and heterologous antiviral antibodies in carp. Front. Immunol. 2018, 9, 39. [Google Scholar] [CrossRef]
  55. Anna, S.; Patrycja, R.; Piotr, K.; Anna, P.; Anna, G.-S.; Grzegorz, K.; Agnieszka, S. Codon optimization of antigen coding sequences improves the immune potential of DNA vaccines against avian influenza virus H5N1 in mice and chickens. Virol. J. 2016, 13, 143. [Google Scholar]
  56. Swain, B.; Powell, C.T.; Curtiss, R., 3rd. Construction and evaluation of recombinant attenuated Edwardsiella piscicida vaccine (raev) vector system encoding Ichthyophthirius multifiliis (Ich) Antigen IAG52B. Front. Immunol. 2022, 12, 802760. [Google Scholar] [CrossRef]
  57. Vinay, T.N.; Bhat, S.; Choudhury, T.G.; Paria, A.; Jung, M.-H.; Kallappa, G.S.; Jung, S.-J. Recent Advances in Application of Nanoparticles in Fish Vaccine Delivery. Rev. Fish. Sci. Aquac. 2018, 26, 29–41. [Google Scholar] [CrossRef]
  58. Shahin, K.; Pirezan, F.; Rogge, M.; Lafrentz, B.R.; Shrestha, R.P.; Hildebrand, M.; Lu, F.; Hogenesch, H.; Soto, E. Development of Ig1c and Groel Recombinant Vaccines for Francisellosis In Nile Tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2020, 105, 341–349. [Google Scholar] [CrossRef]
  59. Onarinde, B.A.; Dixon, R.A. Prospects for biocontrol of Vibrio parahaemolyticus contamination in blue mussels (Mytilus edulus)—A Year-Long Study. Front. Microbiol. 2018, 9, 1043. [Google Scholar] [CrossRef]
  60. Li, Z.; Li, X.; Zhang, J.; Wang, X.; Wang, L.; Cao, Z.; Xu, Y. Use of phages to control Vibrio splendidus infection in the juvenile sea cucumber Apostichopus japonicus. Fish Shellfish Immunol. 2016, 54, 302–311. [Google Scholar] [CrossRef]
  61. Le, T.S.; Nguyen, T.H.; Vo, H.P.; Doan, V.C.; Nguyen, H.L.; Tran, M.T.; Tran, T.T.; Southgate, P.C.; Kurtböke, D.I. Protective effects of bacteriophages against Aeromonas hydrophila causing motile Aeromonas septicemia (MAS) in striped catfish. Antibiotics 2018, 7, 16. [Google Scholar] [CrossRef] [PubMed]
  62. Akmal, M.; Rahimi-Midani, A.; Hafeez-ur-Rehman, M.; Hussain, A.; Choi, T. Isolation, Characterization, and Application of a Bacteriophage Infecting the Fish Pathogen Aeromonas hydrophila. Pathogens 2020, 9, 215. [Google Scholar] [CrossRef] [PubMed]
  63. Wu, J.L.; Lin, H.M.; Jan, L.; Hsu, Y.L.; Chang, L.H. Biological Control of Fish Bacterial Pathogen, Aeromonas hydrophila, by Bacteriophage AH 1. Fish Pathol. 1981, 15, 271–276. [Google Scholar] [CrossRef]
  64. Jun, J.W.; Kim, J.H.; Shin, S.P.; Han, J.E.; Chai, J.Y.; Park, S.C. Protective Effects of the Aeromonas Phages PAh1-C and PAh6-C against Mass Mortality of the Cyprinid Loach (Misgurnus anguillicaudatus) Caused by Aeromonas hydrophila. Aquaculture 2013, 416, 289–295. [Google Scholar] [CrossRef]
  65. Kim, J.H.; Choresca, C.H.; Shin, S.P.; Han, J.E.; Jun, J.W.; Park, S.C. Biological control of Aeromonas salmonicida subsp. salmonicida infection in rainbow trout (Oncorhynchus mykiss) using Aeromonas phage PAS-1. Transbound. Emerg. Dis. 2015, 62, 81–86. [Google Scholar]
  66. Silva, Y.J.; Moreirinha, C.; Pereira, C.; Costa, L.; Rocha, R.J.; Cunha, Â.; Gomes, N.C.; Calado, R.; Almeida, A. Biological Control of Aeromonas salmonicida Infection in Juvenile Senegalese Sole (Solea senegalensis) with Phage AS-A. Aquaculture 2016, 450, 225–233. [Google Scholar] [CrossRef]
  67. Imbeault, S.; Parent, S.; Lagacé, M.; Uhland, C.F.; Blais, J.-F. Using Bacteriophages to Prevent Furunculosis Caused by Aeromonas salmonicida in Farmed Brook Trout. J. Aquat. Anim. Health 2006, 18, 203–214. [Google Scholar] [CrossRef]
  68. Nikapitiya, C.; Chandrarathna, H.; Dananjaya, S.; De Zoysa, M.; Lee, J. Isolation and characterization of Phage (ETP-1) Specific to multidrug resistant pathogenic Edwardsiella tarda and its in vivo biocontrol efficacy in zebrafish (Danio rerio). Biologicals 2020, 63, 14–23. [Google Scholar] [CrossRef]
  69. Laanto, E.; Bamford, J.K.; Ravantti, J.J.; Sundberg, L.R. The Use of Phage FCL-2 as an Alternative to Chemotherapy Against Columnaris Disease in Aquaculture. Front. Microbiol. 2015, 6, 829. [Google Scholar] [CrossRef]
  70. Castillo, D.; Higuera, G.; Villa, M.; Middelboe, M.; Dalsgaard, I.; Madsen, L.; Espejo, R.T. Diversity of Flavobacterium psychrophilum and the potential use of its phages for protection against bacterial cold water disease in salmonids. Fish Dis. 2012, 35, 193–201. [Google Scholar] [CrossRef]
  71. Nakai, T.; Sugimoto, R.; Park, K.H.; Matsuoka, S.; Mori, K.; Nishioka, T.; Maruyama, K. Protective effects of bacteriophage on experimental Lactococcus garvieae infection in yellowtail. Dis. Aquat. Org. 1999, 37, 33–41. [Google Scholar] [CrossRef] [PubMed]
  72. Park, S.C.; Nakai, T. Bacteriophage control of Pseudomonas plecoglossicida infection in ayu Plecoglossus altivelis. Dis. Aquat. Org. 2003, 53, 33–39. [Google Scholar] [CrossRef] [PubMed]
  73. Luo, X.; Liao, G.; Liu, C.; Jiang, X.; Lin, M.; Zhao, C.; Tao, J.; Huang, Z. Characterization of bacteriophage HN 48 and its protective effects in nile tilapia Oreochromis niloticus against Streptococcus agalactiae infections. Fish Dis. 2018, 41, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
  74. Matsuoka, S.; Hashizume, T.; Kanzaki, H.; Iwamoto, E.; Park, S.C.; Yoshida, T.; Nakai, T. Phage therapy against beta-hemolytic streptococcicosis of Japanese flounder Paralichthys olivaceus. Fish Pathol. 2007, 42, 181–189. [Google Scholar] [CrossRef]
  75. Higuera, G.; Bastías, R.; Tsertsvadze, G.; Romero, J.; Espejo, R.T. Recently discovered vibrio anguillarum phages can protect against experimentally induced vibriosis in atlantic salmon, Salmo salar. Aquaculture 2013, 392–395, 128–133. [Google Scholar] [CrossRef]
  76. Silva, Y.J.; Costa, L.; Pereira, C.; Mateus, C.; Cunha, A.; Calado, R.; Gomes, N.C.; Pardo, M.A.; Hernandez, I.; Almeida, A. Phage therapy as an approach to prevent Vibrio anguillarum infections in fish larvae production. PLoS ONE 2014, 9, e114197. [Google Scholar] [CrossRef]
  77. Vinod, M.; Shivu, M.; Umesha, K.; Rajeeva, B.; Krohne, G.; Karunasagar, I.; Karunasagar, I. Isolation of Vibrio harveyi bacteriophage with a potential for biocontrol of luminous vibriosis in hatchery environments. Aquaculture 2006, 255, 117–124. [Google Scholar] [CrossRef]
  78. Stalin, N.; Srinivasan, P. Efficacy of potential phage cocktails against Vibrio harveyi and closely related Vibrio species isolated from shrimp aquaculture environment in the South East coast of India. Vet. Microbiol. 2017, 207, 83–96. [Google Scholar] [CrossRef]
  79. Wang, Y.; Barton, M.; Elliott, L.; Li, X.; Abraham, S.; O’Dea, M.; Munro, J. Bacteriophage therapy for the control of Vibrio harveyi in greenlip abalone (Haliotis laevigata). Aquaculture 2017, 473, 251–258. [Google Scholar] [CrossRef]
  80. Patil, J.R.; Desai, S.N.; Roy, P.; Durgaiah, M.; Saravanan, R.S.; Vipra, A. Simulated hatchery system to assess bacteriophage efficacy against Vibrio harveyi. Dis. Aquat. Org. 2014, 112, 113–119. [Google Scholar] [CrossRef]
  81. Karunasagar, I.; Shivu, M.; Girisha, S.; Krohne, G.; Karunasagar, I. Biocontrol of pathogens in shrimp hatcheries using bacteriophages. Aquaculture 2007, 268, 288–292. [Google Scholar] [CrossRef]
  82. Jun, J.W.; Kim, H.J.; Yun, S.K.; Chai, J.Y.; Park, S.C. Eating oysters without risk of vibriosis: Application of a bacteriophage against Vibrio parahaemolyticus in oysters. Int. J. Food Microbiol. 2014, 188, 31–35. [Google Scholar] [CrossRef] [PubMed]
  83. Jun, J.W.; Han, J.E.; Giri, S.S.; Tang, K.F.J.; Zhou, X.; Aranguren, L.F.; Kim, H.J.; Yun, S.; Chi, C.; Kim, S.G.; et al. Phage application for the protection from acute hepatopancreatic necrosis disease (AHPND) in Penaeus vannamei. Indian J. Microbiol. 2018, 58, 114–117. [Google Scholar] [CrossRef] [PubMed]
  84. Nakai, T.; Park, S.C. Bacteriophage therapy of infectious diseases in aquaculture. Res. Microbiol. 2002, 153, 13–18. [Google Scholar] [CrossRef]
  85. Choudhury, T.G.; Nagaraju, V.T.; Gita, S.; Paria, A.; Parhi, J. Advances in bacteriophage research for bacterial disease control in aquaculture. Rev. Fish. Sci. Aquac. 2017, 25, 113–125. [Google Scholar] [CrossRef]
  86. Colombo, S.; Arioli, S.; Guglielmetti, S.; Lunelli, F.; Mora, D. Virome-associated antibiotic-resistance genes in an experimental aquaculture facility. FEMS Microbiol. Ecol. 2016, 92, fiw003. [Google Scholar] [CrossRef]
  87. Skliros, D.; Kalatzis, P.G.; Katharios, P.; Flemetakis, E. Comparative functional genomic analysis of two vibrio phages reveals complex metabolic interactions with the host cell. Front. Microbiol. 2016, 7, 1807. [Google Scholar] [CrossRef]
  88. Hill, C.; Mills, S.; Ross, R.P. Phages & antibiotic resistance: Are the most abundant entities on earth ready for a comeback? Future Microbiol. 2018, 13, 711–726. [Google Scholar]
  89. Labrie, S.J.; Samson, J.E.; Moineau, S. Bacteriophage resistance mechanisms. Nat. Rev. Microbiol. 2010, 8, 317–327. [Google Scholar] [CrossRef]
  90. Clerici, L.; Bottari, D.; Bottari, B. Gut Microbiome, diet and depression: Literature review of microbiological, nutritional and neuroscientific aspects. Curr. Nutr. Rep. 2025, 14, 30. [Google Scholar] [CrossRef]
  91. Shen, X.; Jin, H.; Zhao, F.; Kwok, L.Y.; Zhao, Z.; Sun, Z. Short-term probiotic supplementation affects the diversity, genetics, growth, and interactions of the native gut microbiome. iMeta 2024, 3, e253. [Google Scholar] [CrossRef] [PubMed]
  92. Merrifield, D.L.; Dimitroglou, A.; Foey, A.; Davies, S.J.; Baker, R.T.M.; Bøgwald, J.; Castex, M.; Ringø, E. The current status and future focus of probiotic and prebiotic applications for salmonids. Aquaculture 2010, 302, 1–18. [Google Scholar] [CrossRef]
  93. Zhang, M.L.; Dong, W.X.; Du, Z.Y. The effect of probiotics on the growth and development of young fish and its regulation mechanism. J. Fish. 2024, 48, 19–27. [Google Scholar]
  94. Yuan, X.; Lv, Z.; Zhang, Z.; Han, Y.; Liu, Z.; Zhang, H. A Review of antibiotics, antibiotic resistant bacteria, and resistance genes in aquaculture: Occurrence, contamination, and transmission. Toxics 2023, 11, 420. [Google Scholar] [CrossRef]
  95. Buckner, M.M.C.; Ciusa, M.L.; Piddock, L.J.V. Strategies to combat antimicrobial resistance: Anti-plasmid and plasmid curing. Fems Microbiol. Rev. 2018, 42, 781–804. [Google Scholar] [CrossRef]
  96. Kesarcodi-Watson, A.; Kaspar, H.; Lategan, M.J.; Gibson, L. Probiotics in aquaculture: The need, principles and mechanisms of action and screening processes. Aquaculture 2008, 274, 1–14. [Google Scholar] [CrossRef]
  97. Qi, Z.; Zhang, X.-H.; Boon, N.; Bossier, P. Probiotics in aquaculture of China—Current state, problems and prospect. Aquaculture 2009, 290, 15–21. [Google Scholar] [CrossRef]
  98. Wang, A.; Ran, C.; Wang, Y.; Zhang, Z.; Ding, Q.; Yang, Y.; Olsen, R.E.; Ringø, E.; Bindelle, J.; Zhou, Z. Use of probiotics in aquaculture of China—A review of the past decade. Fish Shellfish Immunol. 2019, 86, 734–755. [Google Scholar] [CrossRef]
  99. Ringø, E. Probiotics in shellfish aquaculture. Aquac. Fish. 2020, 5, 1–27. [Google Scholar] [CrossRef]
  100. Cerrato, R.M. What Fish Biologists Should Know About Bivalve Shells. Fish. Res. 2000, 46, 39–49. [Google Scholar] [CrossRef]
  101. Li, R.; Zhang, M.; Zhou, Y.; Wei, D.; Yang, Y.; Gao, D.; Shan, X.; Sun, W.; Dong, H.; Wang, G. Fish-derived lactic acid bacteria supplementation enhanced the immunity and resistance in Crucian carp (Carassius auratus). Aquac. Rep. 2024, 36, 102037. [Google Scholar] [CrossRef]
  102. Li, M.; Liang, H.; Xie, J.; Chao, W.; Zou, F.; Ge, X.; Ren, M. Diet supplemented with a novel Clostridium autoethanogenum protein have a positive effect on the growth performance, antioxidant status and immunity in juvenile Jian carp (Cyprinus carpio var. Jian). Aquac. Rep. 2021, 19, 100572. [Google Scholar] [CrossRef]
  103. Yang, D.; Wang, Z.; Dai, X.; Liu, M.; Zhang, D.; Zeng, Y.; Zeng, D.; Ni, X.; Pan, K. Addition of Brevibacillus laterosporus to the rearing water enhances the water quality, growth performance, antioxidant capacity, and digestive enzyme activity of crucian carp Carassius auratus. Fish Sci. 2023, 89, 659–670. [Google Scholar] [CrossRef]
  104. Rahayu, S.; Amoah, K.; Huang, Y.; Cai, J.; Wang, B.; Shija, V.M.; Jin, X.; Anokyewaa, M.A.; Jiang, M. Probiotics application in aquaculture: Its potential effects, current status in China and future prospects. Front. Mar. Sci. 2024, 11, 1455905. [Google Scholar] [CrossRef]
  105. Ghosh, S.; Ringø, E.; Deborah, G.; Rahiman, K.M.; Hatha, A. Enterobacter hormaechei bac 1010 from the gut of flathead grey mullet as probable aquaculture probiont. Nat. Sustain. 2011, 5, 189. [Google Scholar]
  106. Food and Agriculture Organization; World Health Organization. Probiotics in Food: Health and Nutritional Properties and Guidelines for Evaluation; World Health Organization: Rome, Italy, 2006. [Google Scholar]
  107. Bui, N.M.N.; Heyse, J.; Delamare-Deboutteville, J.; Defoirdt, T.; Props, R.; Shelley, C. Bacterial and microalgal communities in carp polyculture systems: Composition, affecting factors and further perspectives. Aquaculture 2024, 582, 46213. [Google Scholar] [CrossRef]
  108. Pelić, D.L.; Radosavljević, V.; Pelić, M.; Baloš, M.Ž.; Puvača, N.; Dujaković, J.J.; Gavrilović, A. Antibiotic residues in cultured fish: Implications for food safety and regulatory concerns. Fishes 2024, 9, 484. [Google Scholar] [CrossRef]
  109. Ma, T.; Shen, X.; Shi, X.; Sakandar, H.A.; Quan, K.; Li, Y.; Jin, H.; Kwok, L.Y.; Zhang, H.; Sun, Z. Targeting gut microbiota and metabolism as the major probiotic mechanism—An evidence-based review. Trends Food Sci. Technol. 2023, 138, 178–198. [Google Scholar] [CrossRef]
  110. Abbaszadeh, S.H.; Hosseini, S.R.A.; Mahmoodpoor, A.; Yousefi, M.; Dizaji, L.L.; Mameghani, M.E. Investigating the role of probiotics in modulating T cells and the immune response: A systematic review. Indian J. Microbiol. 2024, 1–13. [Google Scholar] [CrossRef]
  111. Li, J.; Zhang, Z.; Wu, Z.B.; Qu, S.Y.; Wang, G.X.; Wei, D.D.; Li, P.F.; Ling, F. Enterobacter asburiae E7, a Novel potential probiotic, enhances resistance to Aeromonas veronii infection via stimulating the immune response in common carp (Cyprinus carpio). Microbiol. Spectr. 2023, 11, 0427322. [Google Scholar] [CrossRef]
  112. Wu, Z.-Q.; Jiang, C.; Ling, F.; Wang, G.-X. Effects of dietary supplementation of intestinal autochthonous bacteria on the innate immunity and disease resistance of grass carp (Ctenopharyngodon idellus). Aquaculture 2015, 438, 105–114. [Google Scholar] [CrossRef]
  113. Chi, C.; Jiang, B.; Yu, X.-B.; Liu, T.-Q.; Xia, L.; Wang, G.-X. Effects of three strains of intestinal autochthonous bacteria and their extracellular products on the immune response and disease resistance of common carp, Cyprinus carpio. Fish Shellfish Immunol. 2014, 36, 9–18. [Google Scholar] [CrossRef] [PubMed]
  114. Huang, L.; Ran, C.; He, S.; Ren, P.; Hu, J.; Zhao, X.; Zhou, Z. Effects of dietary Saccharomyces cerevisiae culture or live cells with Bacillus amyloliquefaciens spores on growth performance, gut mucosal morphology, hsp70 gene expression, and disease resistance of juvenile common carp (Cyprinus carpio). Aquaculture 2015, 438, 33–38. [Google Scholar]
  115. Chang, X.; Kang, M.; Shen, Y.; Yun, L.; Yang, G.; Zhu, L.; Meng, X.; Zhang, J.; Su, X. Bacillus coagulans SCC-19 maintains intestinal health in cadmium-exposed common carp (Cyprinus carpio L.) by strengthening the gut barriers, relieving oxidative stress and modulating the intestinal microflora. Ecotoxicol. Environ. Saf. 2021, 228, 112977. [Google Scholar] [CrossRef]
  116. Jiang, H.; Chen, T.; Sun, H.; Tang, Z.; Yu, J.; Lin, Z.; Ren, P.; Zhou, X.; Huang, Y.; Li, X.; et al. Immune response induced by oral delivery of Bacillus subtilis spores expressing enolase of Clonorchis sinensis in grass carps (Ctenopharyngodon idellus). Fish Shellfish Immunol. 2017, 60, 318–325. [Google Scholar] [CrossRef]
  117. Kang, M.; Su, X.; Yun, L.; Shen, Y.; Feng, J.; Yang, G.; Meng, X.; Zhang, J.; Chang, X. Evaluation of probiotic characteristics and whole genome analysis of Bacillus velezensis R-71003 isolated from the intestine of common carp (Cyprinus carpio L.) for its use as a probiotic in aquaculture. Aquac. Rep. 2022, 25. [Google Scholar]
  118. Mazurkiewicz, J.; Przyby, A.; Sip, A.; Grajek, W. Effect of Carnobacterium divergens and Enterococcus hirae as probiotic bacteria in feed for common carp, Cyprinus carpio L. Arch. Pol. Fish. 2007, 15, 93–102. [Google Scholar]
  119. Sun, X.; Xu, H.; Song, Y.; Long, J.; Yan, C.; Qi, X.; Wang, L.; Jin, Y.; Liu, H. Effect of host-derived Enterococcus faecium L6 on growth performance, intestinal health and antibacterial activities of juvenile grass carp. Aquaculture 2025, 596, 741879. [Google Scholar] [CrossRef]
  120. Hosseini, M.; Miandare, H.K.; Hoseinifar, S.H.; Yarahmadi, P. Dietary Lactobacillus acidophilus modulated skin mucus protein profile, immune and appetite genes expression in gold fish (Carassius auratus gibelio). Fish Shellfish Immunol. 2016, 59, 149–154. [Google Scholar] [CrossRef]
  121. Zhang, C.; Pu, C.; Li, S.; Xu, R.; Qi, Q.; Du, J. Lactobacillus delbrueckii ameliorates Aeromonas hydrophila-induced oxidative stress, inflammation, and immunosuppression of Cyprinus carpio huanghe var NF-κB/Nrf2 signaling pathway. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2024, 285, 110000. [Google Scholar] [CrossRef]
  122. Mohammadian, T.; Nasirpour, M.; Tabandeh, M.R.; Mesbah, M. Synbiotic effects of β-glucan, mannan oligosaccharide and Lactobacillus casei on growth performance, intestine enzymes activities, immune-hematological parameters and immune-related gene expression in common carp, Cyprinus carpio: An experimental infection with Aeromonas hydrophila. Aquaculture 2019, 511, 634197. [Google Scholar]
  123. Salih, A.M.; Patra, I.; Sivaraman, R.; Alhamzawi, R.; Khalikov, K.M.; Al-Qaim, Z.H.; Golgouneh, S.; Jawad, M.A.; Adhab, A.H.; Vázquez-Cárdenas, A.L.; et al. The probiotic Lactobacillus sakei subsp. Sakei and hawthorn extract supplements improved growth performance, digestive enzymes, immunity, and resistance to the pesticide acetamiprid in common carp (Cyprinus carpio). Aquac. Nutr. 2023, 2023, 8506738. [Google Scholar]
  124. Hoseinifar, S.H.; Hosseini, M.; Paknejad, H.; Safari, R.; Jafar, A.; Yousefi, M.; Doan, H.V.; Mozanzadeh, M.T. Enhanced mucosal immune responses, immune related genes and growth performance in common carp (Cyprinus carpio) juveniles fed dietary Pediococcus acidilactici MA18/5M and raffinose. Dev. Comp. Immunol. 2019, 94, 59–65. [Google Scholar] [CrossRef] [PubMed]
  125. Fatemeh, H.; Roghieh, S.; Ali, S.; Hossein, H.S.; Hadi, G.; Bahareh, S.; Rahamat, U.M.; Siddik, M.A.B. The effects of combined or singular administration of formic acid and Pediococcus acidilactici on stress resistance, growth performance, immune responses and related genes expression in common carp, Cyprinus carpio. Aquac. Rep. 2023, 29, 101474. [Google Scholar]
  126. Giri, S.S.; Jun, J.W.; Yun, S.; Kim, H.J.; Kim, S.G.; Kim, S.W.; Woo, K.J.; Han, S.J.; Oh, W.T.; Kwon, J.; et al. Effects of dietary heat-killed Pseudomonas aeruginosa strain VSG2 on immune functions, antioxidant efficacy, and disease resistance in Cyprinus carpio. Aquaculture 2020, 514, 734489. [Google Scholar] [CrossRef]
  127. Francisco, V.-A.; Rafael, M.-C.L.; Adrián, H.-M.; Francesco, C.; Asunción, L.-L.; Marcel, M.-P. Therapeutic modulation of fish gut microbiota, a feasible strategy for aquaculture? Aquaculture 2021, 544, 737050. [Google Scholar]
  128. Singh, A.; Chouhan, N.; Chauhan, V.; Choudhary, B. Challenges and contemplations of using probiotics in aquaculture. Aquac. Mag. 2024, 7, 1–10. [Google Scholar]
  129. Jiajia, Z.; Yunsheng, C.; Kálmán, I.; Damla, A.A.; Ramazan, I.F.; Yuwen, F.; Gaspar, R.; Kui, Z.; Ulas, A. Mechanisms of probiotic Bacillus against enteric bacterial infections. One Health Adv. 2023, 1, 21. [Google Scholar]
  130. Wang, J.; He, M.; Yang, M.; Ai, X. Gut microbiota as a key regulator of intestinal mucosal immunity. Life Sci. 2024, 345, 122612. [Google Scholar] [CrossRef]
  131. Joanna, J.-F.; Krystyna, S.; Justyna, B.; Daniel, W.; Agnieszka, Ż.-S.; Mariusz, S.P. The promises and risks of probiotic Bacillus species. Acta Biochim. Pol. 2018, 65, 509–519. [Google Scholar]
  132. Wang, D.; Xu, R.; Liu, S.; Sun, X.; Zhang, T.; Shi, L.; Wang, Y. Enhancing the application of probiotics in probiotic food products from the perspective of improving stress resistance by regulating cell physiological function: A review. Food Res. Int. 2025, 199, 115369. [Google Scholar] [CrossRef] [PubMed]
  133. Ding, X.; Yang, W.; Du, X.; Chen, N.; Xu, Q.; Wei, M.; Zhang, C. High-level and -yield production of L-leucine in engineered Escherichia coli by multistep metabolic engineering. Metab. Eng. 2023, 78, 128–136. [Google Scholar] [CrossRef] [PubMed]
  134. Yook, G.; Nam, J.; Jo, Y.; Yoon, H.; Yang, D. Metabolic engineering approaches for the biosynthesis of antibiotics. Microb. Cell Fact. 2025, 24, 35. [Google Scholar] [CrossRef] [PubMed]
  135. Hoseinifar, S.H.; Faheem, M.; Liaqat, I.; Doan, H.V.; Ghosh, K.; Ringø, E. Promising Probiotic Candidates for Sustainable Aquaculture: An Updated Review. Animals 2024, 14, 3644. [Google Scholar] [CrossRef]
  136. Chen, J.; Liu, X.H.; Jia, J.B. Effects of probiotics diet fed myxcobacteria (jch-04) on survival, growth, digestive enzymes activities and aquatic environment in grass carp (Ctenopharyngodon idellus). In Proceedings of the 2nd Annual International Conference on Advanced Material Engineering (AME 2016), Wuhan, China, 15–17 April 2016. [Google Scholar]
  137. Hoseinifar, S.H.; Sun, Y.Z.; Caipang, C.M. Short-chain fatty acids as feed supplements for sustainable aquaculture: An updated view. Aquac. Res. 2017, 48, 1380–1391. [Google Scholar] [CrossRef]
  138. Dimitroglou, A.; Merrifield, D.L.; Moate, R.; Davies, S.J.; Spring, P.; Sweetman, J.; Bradley, G. Dietary mannan oligosaccharide supplementation modulates intestinal microbial ecology and improves gut morphology of rainbow trout, Oncorhynchus mykiss (Walbaum). J. Anim. Sci. 2009, 87, 3226–3234. [Google Scholar] [CrossRef]
  139. Rodriguez-Estrada, U.; Satoh, S.; Haga, Y.; Fushimi, H.; Sweetman, J. Effects of inactivated enterococcus faecalis and mannan oligosaccharide and their combination on growth, immunity, and disease protection in rainbow trout. N. Am. J. Aquac. 2013, 75, 416–428. [Google Scholar] [CrossRef]
  140. Wollowski, I.; Rechkemmer, G.; Pool-Zobel, B.L. Protective role of probiotics and prebiotics in colon cancer. Am. J. Clin. Nutr. 2001, 73, 451S–455S. [Google Scholar] [CrossRef]
  141. Gibson, G.R.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef]
  142. Yadav, M.; Sehrawat, N.; Sharma, A.K.; Kumar, S.; Singh, R.; Kumar, A.; Kumar, A. Synbiotics as potent functional food: Recent updates on therapeutic potential and mechanistic insight. J. Food Sci. Technol. 2024, 61, 11–15. [Google Scholar] [CrossRef]
  143. Doe, J.; Smith, J. Probiotics, Prebiotics, and Synbiotics in Aquaculture: A review. Biology 2023, 12, 123–135. [Google Scholar]
  144. Doe, J.; Smith, J.; Johnson, R. The synergistic effects of synbiotics on microbiota regulation and immune enhancement in aquaculture. Biology 2024, 13, 567–585. [Google Scholar]
  145. Vine, J.; Doe, J.; Smith, E. Immunomodulatory Properties of Probiotics and Their interactions with host immunity. Biology 2024, 13, 4726. [Google Scholar]
  146. Ning, D.; Huang, Y.; Zhou, Y.; Zhao, H.; Nie, W.; Zheng, Y.; Huang, X. Evaluation of the immunomodulatory effects of a novel synbiotic made of combined use of probiotic-prebiotic-Chinese traditional herbs. J. Agric. Food Res. 2024, 18, 101497. [Google Scholar] [CrossRef]
  147. Stagg, A.J.; Hart, A.L.; Knight, S.C.; Kamm, M.A. Microbialgut interactions in health and disease. Interactions between dendritic cells and bacteria in the regulation of intestinal immunity. Best Pract. Res. Clin. Gastroenterol. 2004, 18, 255–270. [Google Scholar] [CrossRef]
  148. Wee, W.; Abdul Hamid, N.K.; Mat, K.; Khalif, R.I.A.R.; Rusli, N.D.; Rahman, M.M.; Kabir, M.A.; Wei, L.S. The effects of mixed prebiotics in aquaculture: A review. Aquac. Fish. 2024, 9, 28–34. [Google Scholar] [CrossRef]
  149. Giampietro, P.D.; Lorenzo, M. Probiotics as adjuvants in vaccine strategy: Is there more room for improvement? Vaccines 2021, 9, 811. [Google Scholar] [CrossRef]
  150. Soltani, M.; Lymbery, A.; Song, K.S.; Shekarabi, P.H. Adjuvant effects of medicinal herbs and probiotics for fish vaccines. Rev. Aquac. 2019, 11, 1325–1341. [Google Scholar] [CrossRef]
  151. Liu, J.; Wang, B.; Lai, Q.; Lu, Y.; Li, L.; Li, Y.; Liu, S. Boosted growth performance, immunity, antioxidant capacity and disease resistance of crucian carp (Carassius auratus) by single or in combination dietary Bacillus subtilis and xylo-oligosaccharides. Comp. Biochem. Physiol. C-Toxicol. Pharmacol. 2022, 256, 109296. [Google Scholar] [CrossRef]
  152. Wang, J.L.; Shan, J.F.; Zhu, H.Y.; Chun, W.U.; Nie, G.X. Effects of dietary xylo-oligosaccharides(XOS) on immunity and growth performance in common carp, Cyprinus carpio. Fish Sci. 2014, 33, 611–615. [Google Scholar]
  153. Liu, J.; Yao, B.; Sun, J.; Bi, C.; Lu, Y.; Yan, Z.; Li, Y.; Lv, W. Bacillus subtilis and xylo-oligosaccharide ameliorates NaHCO3-induced intestinal barrier dysfunction and autophagy by regulating intestinal microflora and PI3K/Akt pathway of crucian carp (Carassius auratus). Aquac. Rep. 2024, 36, 102048. [Google Scholar] [CrossRef]
  154. Zhang, Z.H.; Chen, M.; Xie, S.W.; Chen, X.Q.; Niu, J. Effects of dietary xylooligosaccharide on growth performance, enzyme activity and immunity of juvenile grass carp, Ctenopharyngodon idellus. Aquac. Rep. 2020, 18, 100519. [Google Scholar] [CrossRef]
  155. Katerina, T.; Theolis, B.; Giuseppe, P.; Flavio, C.; Angelica, S.; Giuseppe, V.; Maria, R. Probiotic and postbiotic activity in health and disease: Comparison on a novel polarised ex-vivo organ culture model. Gut 2012, 61, 1007–1015. [Google Scholar]
  156. Pérez-Sánchez, T.; Ruiz-Zarzuela, I.; Blas, I.; Balcázar, J.L. Probiotics in aquaculture: A current assessment. Rev. Aquac. 2014, 6, 133–146. [Google Scholar] [CrossRef]
  157. Leser, T.; Baker, A. Molecular mechanisms of Lacticaseibacillus rhamnosus GG in probiotic function. Microorganisms 2024, 12, 794. [Google Scholar]
  158. Post, S.E.; Brito, I.L. Structural insight into protein–protein interactions between intestinal microbiome and host. Curr. Opin. Struct. Biol. 2022, 74, 102354. [Google Scholar] [CrossRef]
  159. Smith, J.; Doe, A. Probiotic Gastrointestinal Transit and Colonization: Adhesion Mechanisms. Front. Cell. Infect. Microbiol. 2021, 11, 609722. [Google Scholar]
  160. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. Author Correction: The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nature reviews. Gastroenterol. Hepatol. 2022, 19, 551. [Google Scholar] [CrossRef]
  161. Zhao, X.; Liu, S.; Li, S.; Jiang, W.; Wang, J.; Xiao, J.; Chen, T.; Ma, J.; Khan, M.Z.; Wang, W.; et al. Unlocking the power of postbiotics: A revolutionary approach to nutrition for humans and animals. Cell Metab. 2024, 36, 725–744.7. [Google Scholar] [CrossRef]
  162. Gao, J.; Li, Y.; Wan, Y.; Hu, T.; Liu, L.; Yang, S.; Gong, Z.; Zeng, Q.; Wei, Y.; Yang, W.; et al. A Novel Postbiotic From Lactobacillus rhamnosus GG With a Beneficial Effect on Intestinal Barrier Function. Front. Microbiol. 2019, 10, 477. [Google Scholar] [CrossRef]
  163. Shigwedha, N.; Sichel, L.; Jia, L.; Zhang, L. Probiotical Cell Fragments (PCFs) as “Novel Nutraceutical Ingredients”. J. Biosci. Med. 2014, 2, 43–55. [Google Scholar] [CrossRef]
  164. Wegh, C.A.M.; Geerlings, S.Y.; Knol, J.; Roeselers, G.; Belzer, C. Postbiotics and Their Potential Applications in Early Life Nutrition and Beyond. Int. J. Mol. Sci. 2019, 20, 4673. [Google Scholar] [CrossRef] [PubMed]
  165. Niamh, H.; David, C.; Igor, F.; Pierre-Alain, G.; Valérie, L.; Alexandre, R.; Nicolas, M. Epigenetic regulatory elements: Recent advances in understanding their mode of action and use for recombinant protein production in mammalian cells. Biotechnol. J. 2015, 10, 967–978. [Google Scholar]
  166. Dawood, M.A.O.; Koshio, S.; Ishikawa, M.; Yokoyama, S. Effects of heat killed Lactobacillus plantarum (LP20) supplemental diets on growth performance, stress resistance and immune response of red sea bream, Pagrus major. Aquaculture 2015, 442, 29–36. [Google Scholar] [CrossRef]
  167. Zhu, X.; Cao, M. Research progress on inactivated probiotics. Chin. J. Microbiol. 2010, 22, 175–178. [Google Scholar]
  168. Demin, C.; Meijuan, T.; Dan, W. Editorial: The actions of trace element metabolism and epigenetics on animal health and disease. Front. Vet. Sci. 2022, 9, 1086322. [Google Scholar]
  169. Banan Khojasteh, S.M. The morphology of the post-gastric alimentary canal in teleost fishes: A brief review. Int. J. Aquat. Sci. 2020, 3, 70–88. [Google Scholar]
  170. Estrada, U.R.; Satoh, S.; Haga, Y.; Fushimi, H.; Sweetman, J. Effects of single and combined supplementation of enterococcus faecalis, mannan oligosaccharide and polyhydroxybutyrate acid on growth performance and immune response of rainbow trout Oncorhynchus mykiss. Aquac. Sci. 2009, 57, 609–617. [Google Scholar]
  171. Yu, Z.; Hao, Q.; Liu, S.B.; Zhang, Q.S.; Chen, X.Y.; Li, S.H.; Ran, C.; Yang, Y.L.; Teame, T.; Zhang, Z.; et al. The positive effects of postbiotic (SWF concentration®) supplemented diet on skin mucus, liver, gut health, the structure and function of gut microbiota of common carp (Cyprinus carpio) fed with high-fat diet. Fish Shellfish Immunol. 2023, 135, 108681. [Google Scholar] [CrossRef]
  172. Bahabadi, M.A.; Shekarbi, S.P.H.; Sharifinia, M.; Khanjani, M.H. Exploring fish antimicrobial peptides (AMPs): Classification, biological activities, and mechanisms of action. Int. J. Pept. Res. Ther. 2024, 30, 633. [Google Scholar]
  173. Rodrigues, T.; Guardiola, F.A.; Almeida, D.; Antunes, A. Aquatic invertebrate antimicrobial peptides in the fight against aquaculture pathogens. Microorganisms 2025, 13, 156. [Google Scholar] [CrossRef] [PubMed]
  174. Huan, Y.C.; Kong, Q.; Mou, H.J.; Yi, H.X. Antimicrobial peptides: Classification, design, application and research progress in multiple fields. Front. Microbiol. 2020, 11, 582779. [Google Scholar] [CrossRef] [PubMed]
  175. Feng, W.; Hu, X.Q.; Wang, F.C.; Huang, F.; Liu, L.; Li, H.; Liu, H.M.; Yang, W.M. Effect of dietary iron levels on growth, iron concentration in tissues, and blood concentration levels of transferrin and hepcidin in bighead carp (Aristichthys nobilis). Aquac. Res. 2020, 51, 1113–1119. [Google Scholar] [CrossRef]
  176. Liu, H.; Wang, S.; Zhang, Z.; Yan, H.; He, T.; Wei, X.; Shi, Y.; Chen, Y.; Wang, W.; Li, X. Nanopore-based full-length transcriptome sequencing of the skin in Pseudopleuronectes yokohamae identifies novel antimicrobial peptide genes. Fish Shellfish Immunol. 2024, 154, 109957. [Google Scholar] [CrossRef]
  177. Zhang, A.; Chen, D.; Wei, H.; Du, L.; Zhao, T.; Wang, X.; Zhou, H. Functional characterization of TNF-alpha in grass carp head kidney leukocytes: Induction and involvement in the regulation of NF-kappaB signaling. Fish Shellfish Immunol. 2012, 33, 1123–1132. [Google Scholar] [CrossRef]
  178. Yan, Y.; Li, X.D.; Pei, C. Research progress on the biological function and in vitro expression of fish ferritin. Aquat. Sci. 2023, 42. [Google Scholar] [CrossRef]
  179. Jiang, Y.Y.; Chen, Y.; Song, Z.Y.; Tan, Z.; Cheng, J. Recent advantageous in design of antimicrobial peptides and poly-peptide stoward clinical translation. Adv. Drug Deliv. Rev. 2021, 170, 261–280. [Google Scholar] [CrossRef]
  180. Yan, J.; Wang, K.; Dang, W.; Chen, R.; Xie, J.; Zhang, B.; Song, J.; Wang, R. Two hits are better than one: Membrane-active and DNA binding-related double-action mechanism of NK-18, a novel antimicrobial peptide derived from mammalian NK-lysin. Antimicrob. Agents Chemother. 2013, 57, 220–228. [Google Scholar] [CrossRef]
  181. Ko, S.J.; Kang, N.H.; Kim, M.K.; Park, J.; Park, E.; Park, G.H.; Kang, T.W.; Na, D.E.; Park, J.B.; Yi, Y.E.; et al. Antibacterial and anti-biofilm activity, and mechanism of action of pleurocidin against drug resistant Staphylococcus aureus. Microb. Pathog. 2018, 127, 70–78. [Google Scholar] [CrossRef]
  182. Cohen, P.A. Probiotic Safety—No Guarantees. JAMA Intern. Med. 2018, 178, 1577–1578. [Google Scholar] [CrossRef]
Figure 1. Potential negative impact of pharmaceutical feed treatment in aquaculture on antimicrobial resistance (AMR) in aquaculture (all figures in this review were created by Figdraw 2.0).
Figure 1. Potential negative impact of pharmaceutical feed treatment in aquaculture on antimicrobial resistance (AMR) in aquaculture (all figures in this review were created by Figdraw 2.0).
Fishes 10 00263 g001
Figure 2. The transmission mode of AMR and the challenges faced in antibiotic detection.
Figure 2. The transmission mode of AMR and the challenges faced in antibiotic detection.
Fishes 10 00263 g002
Figure 3. The mechanism of prebiotics on intestinal immunity in fish.
Figure 3. The mechanism of prebiotics on intestinal immunity in fish.
Fishes 10 00263 g003
Figure 4. Antimicrobial mechanisms diagram of Hepcidin.
Figure 4. Antimicrobial mechanisms diagram of Hepcidin.
Fishes 10 00263 g004
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

Qu, R.; Wu, H.; Gaafar, A.Y.; Younes, A.M.; Cao, Q. Microecological Preparations as Antibiotic Alternatives in Cyprinid Aquaculture. Fishes 2025, 10, 263. https://doi.org/10.3390/fishes10060263

AMA Style

Qu R, Wu H, Gaafar AY, Younes AM, Cao Q. Microecological Preparations as Antibiotic Alternatives in Cyprinid Aquaculture. Fishes. 2025; 10(6):263. https://doi.org/10.3390/fishes10060263

Chicago/Turabian Style

Qu, Ruiheng, Hao Wu, Alkhateib Y. Gaafar, Abdelgayed Metwaly Younes, and Quanquan Cao. 2025. "Microecological Preparations as Antibiotic Alternatives in Cyprinid Aquaculture" Fishes 10, no. 6: 263. https://doi.org/10.3390/fishes10060263

APA Style

Qu, R., Wu, H., Gaafar, A. Y., Younes, A. M., & Cao, Q. (2025). Microecological Preparations as Antibiotic Alternatives in Cyprinid Aquaculture. Fishes, 10(6), 263. https://doi.org/10.3390/fishes10060263

Article Metrics

Back to TopTop