Next Article in Journal
Challenges and Approaches of Culturing the Unculturable Archaea
Next Article in Special Issue
Genetics and Genomics of Infectious Diseases in Key Aquaculture Species
Previous Article in Journal
Effects of Datura stramonium L. Invasion into Different Habitats on Native Plant Functional Traits and Soil Carbon, Nitrogen and Phosphorus Stoichiometric Characteristics
Previous Article in Special Issue
Expression and Immune Characterization of Major Histocompatibility Complex in Paralichthys olivaceus after Antigen Stimulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Synbiotic Agents and Their Active Components for Sustainable Aquaculture: Concepts, Action Mechanisms, and Applications

by
Vijayaram Srirengaraj
1,
Hary L. Razafindralambo
2,3,*,
Holy N. Rabetafika
2,
Huu-Thanh Nguyen
4 and
Yun-Zhang Sun
1,*
1
Fisheries College, Jimei University, Xiamen 361021, China
2
ProBioLab, 5004 Namur, Belgium
3
BioEcoAgro Joint Research Unit, TERRA Teaching and Research Centre, Sustainable Management of Bio-Agressors & Microbial Technologies, Gembloux Agro-Bio Tech—Université de Liège, 5030 Gembloux, Belgium
4
Department of Biotechnology, An Giang University, Long Xuyen City 90000, Vietnam
*
Authors to whom correspondence should be addressed.
Biology 2023, 12(12), 1498; https://doi.org/10.3390/biology12121498
Submission received: 31 October 2023 / Revised: 26 November 2023 / Accepted: 5 December 2023 / Published: 6 December 2023

Abstract

:

Simple Summary

Aquatic animals are consistently exposed to the threats of environmental deterioration and infection outbreaks because of the excessive use of antibiotics and synthetic drugs. This practice leads to the accumulation of residues in aquatic systems and the development of antimicrobial resistance among pathogens. Nature-based solutions, such as functional feeds containing synbiotics and their active components, such as probiotics, prebiotics, and postbiotics, play a crucial role in maintaining a healthy environment and promoting the well-being of animals in aquaculture. Drawing upon a thorough literature survey and experimental evidence, these agents have been shown beneficial to aquatic animals and their ecosystems. Consequently, these synbiotic agents and related components emerge as promising natural alternatives to traditional synthetic drugs and antibiotics in aquaculture.

Abstract

Aquaculture is a fast-emerging food-producing sector in which fishery production plays an imperative socio-economic role, providing ample resources and tremendous potential worldwide. However, aquatic animals are exposed to the deterioration of the ecological environment and infection outbreaks, which represent significant issues nowadays. One of the reasons for these threats is the excessive use of antibiotics and synthetic drugs that have harmful impacts on the aquatic atmosphere. It is not surprising that functional and nature-based feed ingredients such as probiotics, prebiotics, postbiotics, and synbiotics have been developed as natural alternatives to sustain a healthy microbial environment in aquaculture. These functional feed additives possess several beneficial characteristics, including gut microbiota modulation, immune response reinforcement, resistance to pathogenic organisms, improved growth performance, and enhanced feed utilization in aquatic animals. Nevertheless, their mechanisms in modulating the immune system and gut microbiota in aquatic animals are largely unclear. This review discusses basic and current research advancements to fill research gaps and promote effective and healthy aquaculture production.

1. Introduction

Aquaculture is an emerging sector that generates numerous employment opportunities and also addresses a fundamental need for essential nutrients in global food production [1]. However, it is presently faced with pressing challenges, especially the vulnerability of aquatic animals to ecological degradation and infectious outbreaks. A key contributing factor to these threats is the excessive use of antibiotics and synthetic drugs, which exert harmful effects on the aquatic environment [2,3].
The aquaculture production sector typically relies on traditional practices employing various antibiotics (e.g., chloramphenicol, fluoroquinolones, nitrofurans, quinolones, florfenicol, sufamerazine, chorionic gonadotropin, oxytetracycline dihydrate, and oxytetracycline hydrochloride) and synthetic chemicals (e.g., formalin, malachite green, potassium permanganate, and copper sulfate) to control related diseases [4]. However, some of these chemotherapeutic applications have been widely criticized given their negative impacts on marine debris gathering, drug resistance expansion, and immunosuppressant activity. For example, the use of formalin and potassium permanganate for pathogen control has resulted in adverse effects on fish like damage to gills (hyperplasia) and alteration in mucous cells [5,6]. The extensive application of antibiotics in aquaculture has led to their bioaccumulation in aquatic animals [7]. The intensive use of antibiotics and chemicals leads to the buildup of harmful residues, not only in aquatic animals but also in consumers, by causing side effects such as diarrhea, vomiting, and stomach problems. Moreover, the practice of these traditional methods has been reported to be ineffective in controlling diseases in large-scale aquaculture processes [8,9,10,11,12,13].
In fish, the gastrointestinal tract (GIT) microbiota plays several vital functions. These microbial consortia increase digestive action, enhance the immune system, protect against harmful microbes, and improve intestine development [14]. In recent years, some gnotobiotic (germ-free) animal models have been successfully used as wonderful tools for studying host–microbe interactions and investigating the role of gut microbiota in xenobiotic metabolism [15,16]. Through zebrafish (Danio rerio) models, researchers have observed that the presence of alkaline phosphatase in the brush border intestine plays a vital function in gut epithelium division, as well as in the modulation of gene expression in bacteria, which possesses various functional properties (e.g., epithelial maturation, hormone-secreting endocrine organs, and mucous secreting goblet cells) in the gastrointestinal tract in D. rerio larvae [17,18]. Recently, it was reported that TLR2/MyD88 signaling plays an essential role in innate immune recognition and activation during the colonization of two indigenous bacteria (Chryseobacterium ZOR0023 and Exiguobacterium ZWU0009) in zebrafish [19]. Indigenous probiotic strains have significant functions such as developing the immune system (nonspecific and specific immunity) and inducing different types of cytokines, namely, TNF-a, interleukins (IL-6, IL-10, IL-12), and IFN-c [20]. The indigenous probiotic Bacillus pumilus SE5 activates the expression of TLR2 signaling and antibacterial peptide genes in the intestine of groupers (Epinephelus coioides). Enhanced TLR2 signaling may result from the interaction of the host with the probiotic cell components [21,22]. In order to enhance the immune system in fish, the gut microbiota also provides important protection against pathogenic organisms [23,24].
Functional feed additives such as probiotics, prebiotics, and/or synbiotics in diets have been extensively recommended to maintain a healthy GIT microbial community, improve immunity, and consequently promote the health of cultured aquatic organisms [25,26,27]. These synbiotic- and component-based ingredients, consisting of live microorganisms, inert substrates, and a combination of both, possess a wide range of multiple functionalities. They represent alternative nature-based solutions for improving aquatic animal health and production [24,28,29]. This review provides insights into the basic and current developments in the utilization of probiotics, prebiotics, postbiotics, and synbiotics in aquaculture applications. It also presents a new way to develop a healthy and modern aquaculture industry.

2. Probiotics

2.1. Definition and Characteristic Features

The Food and Agriculture Organization (FAO) of the United Nations and the World Health Organization (WHO) define probiotics as “Live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [30]. Recently, the term probiotics has been associated with microbial feed additives that, when controlled in enough amounts, confer health and beneficial impacts on a host of aquatic animals [29].
Probiotics act as a defense system for the host against harmful microbes or foreign substances [31,32,33,34]. They also produce beneficial bioactive molecules such as enzymes, proteins, lipids, organic acids, and others. Some of these bioactive molecules improve binding to probiotics and reduce, therefore, the activity of pathogens in the gut region through the surface competition mechanism [35]. Probiotics play a significant role in strengthening the immune system of the host [36]. While earlier studies have noted the utilization of probiotics in pigs, poultry, cattle, and humans, their application in aquaculture is a relatively new idea [37,38]. Probiotics can be administered in two ways in aquaculture. They can be supplemented with feed to modulate gut microbes, or they can be directly added to the water, thereby inhibiting the growth of pathogens. These modes of administration are very critical in the utilization of probiotics in aquaculture [39,40]. Probiotics can have alive, dead, or microbial cell components and provide benefits to the host when added to feed or rearing water. This is achieved at least in part by improving the microbial balance of the host or ambient environment [40]. Figure 1 summarizes the different entryways of probiotics and their benefits in the aquaculture system.
Probiotics appear to be a new agent for the development of aquaculture systems, exerting several favorable effects on growth activity, immune systems, digestion, water quality, the inhibition of pathogens, and the regulation of the gut microbes of aquatic animals. The utilization of probiotics in aquaculture is a modern trend, although its effectiveness in the aquatic ecosystem has not been considered comprehensively. Probiotics are ubiquitous, commonly present in aquatic animals, and play an important protective role throughout the digestive system [41,42]. Mainly represented by Lactobacilli, these beneficial microorganisms are vital to preventing illnesses and improving aquatic animal GIT functions by excreting secondary metabolites such as lactic acid and other bioactive compounds [43,44]. These biomolecules, synthesized by probiotics, protect against inhibitory molecules from pathogens [45]. They can also be extracted from probiotics in terrestrial plants and marine life forms and then utilized to enhance disease resistance, develop the immune system, reduce environmental stress, and increase feed quality levels [46,47]. Advanced studies in this field have reported microbial by-product biomolecules such as enzymes, lipids, proteins, and immune toxins [48]. Nowadays, some probiotic products are commercially available and are already used in aquaculture as feed additives [49]. These microbial by-products are beneficial and are mainly helpful in enhancing the health status of aquatic animals.
Potential probiotic strains are assessed based on physiological, functional, and safety criteria such as stress resistance (e.g., acid and bile tolerance), gut epithelial adherence, survival rates, pathogen-inhibiting activities, large-scale cultivability, non-hemolytic activity, non-pathogenicity, the absence of plasmid-encoded antibiotic resistance genes, and beneficial effects on host animals. These include, for instance, their capacity as growth promoters and anti-inflammatory, antimutagenic, and immunostimulatory agents. Each new strain used for probiotic expansion mainly contains all the aforesaid features [28,50,51]. Current and potential probiotic species for use in aquaculture are listed in Table 1.

2.2. Possible Modes of Action of Probiotics in Aquaculture

The significant effects of probiotics, e.g., Bacillus spp. as feed supplements, include the improvement of growth performance, digestive enzyme activity, resistance to pathogens, and immune responses in aquatic animals [68,69]. Possible action modes of probiotics in aquaculture include the regulation of amino and fatty acid metabolisms, the excretion of digestive enzymes and vitamins or cofactors, the production of antagonistic compounds that inhibit bacteria, the enhancement of immune responses, the disruption of the quorum-sensing processes of pathogenic organisms, stress improvement, and heavy-metal detoxification.

2.2.1. Probiotics Act as Growth Enhancers in Aquaculture

Probiotics play a crucial role in digesting complex dietary macronutrients. Additionally, they contribute to the host’s nutrient and vitamin supply and provide essential digestive enzymes, thereby enhancing feed utilization and digestion.
One of the mechanisms that regulates the metabolism of amino and fatty acids is the capacity of various probiotic strains to produce vitamin B12, as revealed by a study on carp guts [70,71]. In addition, this is helpful for enhancing fish growth and eradicating vitamin B12 deficiency in fish [72]. Also, essential macronutrients are usually supplied through feed. Various micronutrients such as amino acids, vitamins, and fatty acids are very important for physiological functions as nutrients in aquatic animals [73,74,75]. For instance, diverse fish species such as carp (Cyprinus carpio), rainbow trout (Oncorhynchus mykiss), channel catfish (Ictalurus punctatus), and tilapia (Oreochromis niloticus) have been found to synthesize vitamin B12 [76,77,78]. The growth and survival rates of juvenile black tiger shrimp (Penaeus monodon) were enhanced when they were fed for 100 days with a combination of Lactobacillus spp., previously isolated from the GITs of chickens [79]. In fact, probiotics improve the digestive function of aquatic animals by producing or inducing the secretion of different kinds of extracellular enzymes such as proteases, amylases, and lipases.
The function of probiotics results in abridged feed cost, which accounts for 60–70% of the contribution cost of fish production [80,81]. Both the maximum growth performance and best feed conversion ratio were detected when O. niloticus was fed with the probiotic Micrococcus luteus [82,83]. Bacillus subtilis improved feed digestibility; enhanced weight gain and feed conversion; and significantly increased the survival rate of bullfrogs (Lithobates catesbeianus) fed different doses (2.5, 5.0, and 10.0 g/kg) [84,85]. Bacillus species aid in the digestion of aquatic animals by supplying exoenzymes (proteases, lipases, and amylases) that enhance digestive enzymes [86]. The addition of probiotics (a mixture of Streptococcus faecium, Lactobacillus acidophilus, and Saccharomyces cerevisiae) at a concentration of 0.1% to Nile tilapia fry diets was found to enhance animal growth and intestinal alkaline phosphatase activity [87].

2.2.2. Biocontrol of Bacterial Diseases in Aquaculture

In the past few decades, numerous studies have stated that probiotics synthesize different types of inhibitory substances responsible for antagonistic activity against pathogens. Two probiotic strains of LAB (Lactococcus lactis MM1 and Enterococcus faecium MM4) isolated from the intestine of the orange-spotted grouper (E. coioides) can secrete several inhibitory substances such as hydrogen peroxide and bacteriocin-like substances. These can be utilized to induce antimicrobial activity against different pathogens such as Staphylococcus aureus, V. harveyi, and V. metschnikovi, which affect groupers (E. coioides) [88,89]. The probiotic B. pumilus H2 has strong inhibitory activity against Vibrio spp. through its main mechanism of amicoumacin production, disrupting the cell membrane and cell lysis and thus showing anti-Vibrio activity [90,91]. The probiotic Bacillus velezensis cell-free supernatant contains different types of bioactive molecules that act against A. salmonicida infection [92]. The lipopeptide N3, synthesized by the probiotic Bacillus amyloliquefaciens M1, has strong antibacterial activity in the whole-cell membrane, which can exert significant effects from ion-conducting channels on the whole-cell membrane and membrane-active properties [93,94]. The probiotic species Clostridium butyricum, a culture supernatant, includes different types of inhibitory substances, mainly short-chain fatty acids (SCFAs); it can lower the pH of the intestine and thus decrease the growth of pathogens in fish intestinal epithelial cells [95]. The probiotic E. faecium was supplemented in the diets of Olive flounders and can enhance the antibacterial activity [96].

2.2.3. Biocontrol of Viral Diseases in Aquaculture

Microorganism strains with potential probiotic effects in aquaculture such as Pseudomonas spp., Vibrios spp., and Aeromonas spp. induce antiviral effects against hematopoietic necrosis virus (IHNV) infection [97,98]. Similarly, the potential probiotic strain Pseudoalteromonas undina VKM-124 has been used to improve Yellow Jack (Carangoides bartholomaei) larval survival and enhance antiviral effects against Neuro Necrosis Virus (SJNNV) infections [99,100].

2.2.4. Immunostimulant Agents in Aquaculture

Immunity development and modulation are among the various health benefits of probiotics in aquaculture. The majority of earlier studies have dealt with the health-boosting capabilities of probiotics in aquatic organisms. Currently, probiotics are significantly focused on the immunological development properties of the piscine immune system, including both innate and adaptive immunities [20]. Different types of probiotics improve various immunological properties, and notably, several fish use the efficiency of probiotics to vitalize teleost immunity in both in situ and ex situ conditions [101]. Although promising findings have been reported in previous studies, most immunostimulants do not progress to large-scale functions for fish. Since various immunostimulants in aquaculture produce similar effects, researchers have demonstrated the utilization of probiotics to enhance disease resistance and the immune system of carp fish species [102,103]. Several carp fish have shown an increase in the production of total serum protein, nitric oxide, lysozyme, albumin, and phagocytic activity via blood leucocytes; express IL-1b, superoxide anion, myeloperoxidase content, respiratory burst activity, and globulin levels; and complement C3, TNF-α, and lysozyme-C [102,104]. Current study reports indicate that probiotics (either single or mixed types) could enhance the immunological development of fish [105]. These reports have emphasized the immunomodulating properties of beneficial living cell organisms and the factors that facilitate the optimal induction of defense responses in the fish community. The probiotic strain B. pumilus SE5 has been isolated from the intestine of the fast-growing grouper, E. coioides [106,107], and subsequent studies have demonstrated that both viable and heat-inactivated B. pumilus SE5 could shape intestinal immunity and microbiota [108] and improve the growth performance and systemic immunity of E. coioides [109]. The dietary supplementation of the cell wall (CW), peptidoglycan (PG), and lipoteichoic acid (LTA) of the probiotic B. pumilus SE5 and its effect on intestinal immune-related gene expression and microbiota were evaluated in a 60-day feeding trial. The PG and LTA of the probiotic B. pumilus SE5 were more effective than the CW in shaping the intestinal immunity and microbiota of E. coioides [21], even though the mechanisms were largely unclear and needed further study.

2.2.5. Interference of Quorum Sensing in Aquaculture

Quorum sensing (QS) is a communication system among bacterial cells that is very useful in controlling different kinds of biological macromolecule expressions like virulence agents in cell-thickness-dependent comparative performance [110]. In this process, QS bacteria produce and generate tiny marker molecules called auto-inducers [111]. The disruption of the QS process in pathogenic organisms is a potential anti-infective strategy, and different types of methods have been used to investigate QS. These include the inhibition of signal molecule biosynthesis, the application of QS antagonists, the chemical inactivation of QS signals with oxidized halogen antimicrobials, signal molecule biodegradation with bacterial lactonases and bacterial and eukaryotic acylases, and the application of QS agonists in aquaculture [112,113]. N-acyl homoserine lactones (AHLs) are the most important family of QS auto-inducers utilized in Gram-negative bacteria, and their biodegradation is a potential way to interrupt QS [114]. Bacillus species were among the first bacteria documented to degrade AHLs through the production of lactonase enzymes. Probiotic Bacillus strains can effectively secrete quorum-quenching enzymes and could reduce the pathogenic activity of A. hydrophila YJ-1 and control gut microbiota [115,116]. The dietary supplementation of probiotics with quorum-quenching activity has been shown to increase the intestinal barrier function and enhance the immune system of crucian carp against A. hydrophila infection. The quorum-quenching bacteria increase the expression of the tight junction (TJ) proteins ZO-1 and Occludin, which control the permeability and absorption of the intestinal mucosal barrier of crucian carp [117]. Bacillus sp. QSI-1 has been reported to be a quorum quencher in virulence agent production and the biofilm arrangement of the zebrafish pathogen A. hydrophila. In experimental trials, fish fed with Bacillus sp. QSI-1 exhibited a relative survival percentage of 80.8% [118]. In another study, AHL-degrading Bacillus sp. was shown to protect shrimp (Penaeus monodon) against Vibrio harveyi infection [119]. Furthermore, Enterobacter sp. f003 and Staphylococcus sp. sw120, isolated from fish intestines and pond sediment, respectively, have demonstrated the ability to degrade acyl-homoserine lactones (AHLs) and protect against A. hydrophila infection in the cyprinid Carassius auratus gibelio [120]. In a biofilm system, bacteria are resistant to high temperatures, phagocytic cells, surfactants, antibiotics, and antibodies and can alter their vital transmissions via quorum-sensing signaling [121]. These findings suggest that bacteria capable of degrading AHLs should be considered an alternative to antibiotics in aquaculture for effectively controlling bacterial infections in fish.

2.2.6. Stress Improvement in the Aquaculture System

Stress in a fish’s life cycle disrupts all production. The cultured species may be weakened and averse to taking feed [122]. In this condition, probiotics in culture farms can decrease stress levels and help to enhance the innate immune system against pathogens and environmental stressors [123,124]. Probiotic treatments are very helpful in increasing the production of fish within the given time, and they also reduce the stress level in normal aquaculture practices.
Studies have concluded that the use of some probiotic strains increases chronic stress resistance in zebrafish (D. rerio) [125,126]. Supplementation with an experimental nutritional probiotic, Lactobacillus delbrueckii sp. Delbrueckii, in sea bass led to a decrease in cortisol levels from 25 to 59 days, which, in fish tissue, is a stress indicator since it is directly engaged with the host’s reaction to stress [127]. One more approach evaluated how fish treated with probiotics exhibited increased flexibility in stress tests when compared with a control group [81]. The antioxidative properties of the probiotic Lactobacillus fermentum induce protective action in the intestinal microbial ecosystem and help to overcome exo- and endogenous oxidative stress [128]. The probiotic strain Bacillus coagulans SCC-19 alleviates the nonspecific immune damage induced by cadmium in common carp while also relieving oxidative stress induced by cadmium in fish [129].

2.2.7. Reducing Heavy Metals in Aquaculture

Heavy metals such as lead (Pb), cadmium (Cd), silver (Ag), chromium (Cr), mercury (Hg), cobalt (Co), zinc (Zn), iron (Fe), and copper (Cu) are present in the soil, water, and atmosphere [130,131,132]. These metals can have toxic effects on all organisms and pose a huge risk to food quality, crops, and environmental quality. Heavy metals are mainly connected to anthropogenic action in the ecosystem [133]. Aqueous release from metal industries (steel, mining, and electroplating) contains elevated levels of heavy metals that end up in water bodies, and they are then also utilized for aquacultural action [134,135]. These heavy metals accumulate in fish tissue, and this is a matter of great concern with regard to humans consuming them via the food chain and breathing [133,135,136]. Their elimination is very helpful in reducing the toxic effects of the aquatic environment and outflow is, subsequently, imperative [137]. Among all the recommended methods of eliminating heavy metals is the process of utilizing microbes, which is cost-effective [138]. The action mechanisms of probiotics in detoxifying heavy metals can be classified into metabolically independent processes that do not require cellular energy, such as biosorption, and cellular-energy-dependent processes, namely, bioaccumulation and bioprecipitation [139].
Biosorption relies on a physicochemical process wherein cell-surface structures bind heavy metals through physical interactions. For example, Lactobacillus acidophilus and Bifidobacterium angulatum are effective in removing Cd, Pb, and As through electrostatic interactions between heavy-metal cations and the anionic functional groups of cell wall membranes [140]. Some probiotics release exopolysaccharides (EPSs), which can sequester heavy metals and reduce their bioavailability. The mechanisms underlying EPS-metal binding are mainly related to negatively charged acidic groups and steric structures on the surface of EPSs [141].
In bioaccumulation processes, probiotics accumulate heavy metals within their cells through energy-dependent processes. This can involve the synthesis and use of metal-binding proteins, such as metallothionein. For instance, Bacillus cereus can produce metallothionein in order to accumulate Pb [142].
Bioprecipitation involves the conversion of free metals into insoluble complexes, thereby reducing their bioavailability. Bacteria can catalyze oxidative and reductive processes to facilitate the precipitation of heavy metals. Micrococcus spp. have been demonstrated to be able to sequestrate heavy metals such as Zn, Cd, Pb, and Fe via calcite precipitation [143]
Generally, heavy metals activate the sporulation development of Bacillus species and thus decrease heavy metal absorption [134,144]. In addition, probiotic strains from aquatic farming sediments can be utilized as dietary supplements and help to remove heavy metals and metal-resistant microbes from the intestines of aquatic organisms, particularly fish, to control the progress of heavy metal accumulation [145].

2.3. Major Probiotic Genera as Biocontrol Agents in Aquaculture

The major probiotic genera used in aquaculture are Lactobacillus and Bacillus [146]. In most cases, Bacillus, Lactobacillus, Lactococcus, Leuconostoc, Pediococcus, and Weissella are isolated from fish and shellfish guts [147,148,149,150,151]. Supplementation in aquaculture feed is achieved using single-strain probiotics or associations of various bacteria as multi-strain probiotics (MSPs), which have been reported to have more beneficial effects on hosts owing to synergistic effects between various strains [152]. Table 2 lists some examples of probiotic-based functional feed additives for aquatic animals.

3. Prebiotics

Prebiotics are “non-digestible sugars, which helpfully influence the host by specifically enhancing the development of health-encouraging strains in the gut” [182,183]. Prebiotics improve the synbiotic association of the gut microbiota of the host [184] and are also known as immunosaccharides. There are various types of prebiotic compounds, including mannan oligosaccharide (MOS), fructooligosaccharide (FOS), and arabinooligosaccharide (AOS), all of which play a significant role in improving the natural immune system [185]. MOSs are most frequently used in animal diets. These prebiotics improve growth activity, feed utilization, survival rates, the development of immune reactions, and antagonistic activity against aquatic pathogens [186,187,188]. Oligosaccharide-type components have been connected to the development of immunity [189,190] and have been used extensively in diverse fish species such as Psetta maxima [13], Larimichthys crocea [191], Paralichthys olivaceus [192], Rutilus rutilus [193], Piaractus mesopotamicus [194], and Acipenser Persicus [195]. Previous study reports have examined the function of prebiotics in cultured finfish and shellfish, explaining that these compounds have significant effects on gut microbial composition, immune system, and infection resistance against pathogenic organisms in fish [196,197]. Previous studies have also verified the health-beneficial effects of prebiotics on growth and physiological status [198]. Prebiotics can improve the capability and feasibility of aquaculture production. The most frequently used prebiotics, including xylooligosaccharide (XOS), FOS, transgalactooligosaccharide (TGOS), glucooligosaccharide (GOS), soybean oligosaccharide (SBOS), polydextrose, inulin, and Lactosucrose, enhance aquaculture production [199]. Natural sources of prebiotics in vertebrates include onions, garlic, tomatoes, honey chicory, leeks, and so on [200].

3.1. Action in the Gastrointestinal Tracts of Aquatic Animals

Prebiotics exert possible effects on host biological responses, protecting fish species against harmful microbes and thus decreasing their mortality. However, an evaluation of the intestinal microbiota of important commercial fish like hybrid striped bass, channel catfish, salmonids, and tilapia is necessary to infer if there are any particular bacterial species that can be enhanced by the utilization of prebiotics. By increasing the production of volatile fatty acids (VFAs) in the GIT, the host’s advantage is the inhibition of potentially pathogenic organisms [201,202]. The synthesis of VFAs in the aquatic organism’s GIT indicates the presence of microbial communities [203]. Herbivorous fish were the first species (Kyphosus cornelii and K. sydneyanus) shown to contain VFAs synthesized by an intestinal bacterial community [204]. Another fish species, tilapia (Oreochromis mossambicus), was found to have VFAs produced by intestinal bacterial communities [205]. Prebiotics have numerous favorable effects on aquatic animals by enhancing disease resistance and improving nutrient accessibility [206]. Recently, our group evaluated the effects of FOS on the growth performance and predominant autochthonous intestinal microbiota of shrimp (L. vannamei) fed diets with fish meal partially replaced by soybean meal. The results showed that a dietary supplement of 2–4 g/kg of FOS could improve the growth performance and survival rate and exert a beneficial effect on the intestinal microbiota of shrimp. A dose adding 2–4 g/kg of FOS to shrimp diets with fish meal partially replaced by soybean meal was recommended [207,208].

3.2. Regulation in the Immune System of Aquatic Animals

In the past decades, prebiotics were used to regulate intestinal microbiota, modulate immunity, control pathogens, and increase the survival ability of aquatic animals, particularly fish such as sharks, rays, and bony fish [195]. Similar to all vertebrates, fish fully rely on their natural immunity against pathogens because of the restrictions on their adaptive immune functions [209]. There are various cellular and soluble components primarily concerned with immune responses, including phagocytes, leukocytes, and auxiliary cells, which are organized into tissues and organs, with leukocytes being the most functional. The impacts of prebiotics on immunity are indirect and involve the modification of gut microbes, thereby enhancing the immune system. Thus, these beneficial components assist in changing effectiveness, enhancing fish growth, and inducing inhibitory activity against pathogens by prohibiting linkage sites; natural organic acid (e.g., formic acid, lactic acid, acetic acid) syntheses; hydrogen peroxide; and numerous other compounds like bacteriocins, siderophores, lysozyme, and antibiotics. Through these action mechanisms, prebiotics can also cause changes in physiological and immunological responses in fish spleens, kidneys, and thymuses, which are major lymphoid organs [49,210]. The prebiotic components can act as growth promoters for commensal microbes by inhibiting the adhesion and assault of harmful microorganisms in epithelial cells. A beneficial effect of monosaccharide components arises, for instance, from enhancing immune functions, and it acts as a protection system for lymphoid organs.

3.2.1. Phagocytosis

Phagocytosis is the process by which immune cells like macrophages and neutrophils engulf and digest foreign cells or particles, such as bacteria, viruses, and cellular debris [211]. FOS (0.5%) is used to enhance the phagocytosis, respiratory burst, and phenoloxidase activity of sea cucumber coelomocytes and infection resistance against V. splendidus infection [212]. The phagocytic capability of inhabitant and obtained trout macrophages are related to the circumstances (i.e., in suspension versus attached and spread) of the cells at the time of particle treatment. Substrate binding and cell spreading may play a very important function in controlling the overall phagocytic capabilities of macrophages. Since the host’s resistance against infectious agents depends upon the phagocytic ability of the cells, the finding that obtained trout macrophages can surround a larger number of activity latex particles than inhabitant cells provides a better understanding of immune regulatory mechanisms in fish [213]. Dietary supplementation with FOS significantly improves lysozyme activity compared with control diet groups. However, the phagocytic percentage of the phagocytic index has no significant effects. In addition, a combination of FOS and MOS (5.0 g/kg) has shown a significant difference in the phagocytic activity of Japanese flounders [195].

3.2.2. Macrophage Activation

Macrophages play a very important role in the nonspecific and specific connections of immune function by synthesizing the highest level of immune reaction and eliminating harmful microbes. Macrophages are stimulated to produce diverse inflammatory cytokines like tumor necrosis factor (TNF), IL-1, IL-12, etc. [96]. The alterations to the physiology of macrophages as a result of environmental signals can benefit them with improved antimicrobial activity. Nevertheless, ecosystem signals do not always cause changes that improve macrophage immune activity. Both nonspecific and specific immune responses can result in macrophages that are more vulnerable to harmful infections and less prepared to generate cytokines that enhance immune system responses [214].

3.2.3. Respiratory Burst Activity

A respiratory burst is the fast release of reactive oxygen substances, namely, superoxide anions, hydrogen peroxide, and hydroxyl radicals. These reactive oxygen compounds are generally used to defend the ability of the host organism to counter harmful microbes. They are synthesized by activated phagocytes that are responsible for destroying microbes [215]. Respiratory burst analyses have been performed in naturally resistant cells and blood neutrophils using the NBT (nitro blue tetrazolium) and MPO (myeloperoxidase) methods. Inulin (5 g kg−1) has been utilized as a dietary nutrient supplement for Nile tilapia and has improved lysozyme and hematocrit NBT action. It can also significantly enhance the natural immune system and increase the survival rate against A. hydrophila infection [216,217].
Marine invertebrates contain enzymes such as tyrosinases, laccases, and catecholases, which can be modified to complement the system of prophenoloxidase. This enhancement improves antagonistic activity through processes like phagocytosis and respiratory burst via opsonization. In a study conducted on red swamp crayfish, the supplementation of a prebiotic nutrient diet with 8 and 10 g kg−1 of FOS over a 30-day trial period significantly enhanced phenoloxidase reactions, stimulated immune-related genes (lysozyme, crustin 1, SOD), and increased the survival rate and antibacterial activity against A. hydrophila infection [218].

3.2.4. Synthesis of Antibodies

B lymphocytes can produce special antibodies for recognizing specific microbial antigens, and these antibodies can neutralize antigens by surface binding and attaching to target cells. Prebiotics can stimulate the immune system like the production of antibodies. β-glucans, in particular, are known for their immunomodulatory effects because of their ability to bind to specific receptors on immune cells, such as macrophages, neutrophils, and natural killer cells, and enhance the release of signaling molecules such as cytokines. Such signaling molecules stimulate blood cells and enhance the secretion of antibodies that can recognize and bind to specific antigens (e.g., pathogens) [219,220]. The stimulation of antibody secretion (IgM) in crucian carp using glucans and astragalus polysaccharides as vaccine adjuvants has been demonstrated and has enhanced disease resistance against A. veronii [221]. The dietary supplementation of MOS and β-glucans was used to enhance the immune system of carp fry [222,223].

3.3. Major Prebiotics with Biocontrol Capabilities in Aquaculture

3.3.1. β-Glucan

There is much evidence available regarding the positive effects of prebiotics on immune responses, disease resistance, and growth performance upon oral delivery in a variety of farmed animals such as salmonids [224], sea bream [225], and shellfish [226]. The supplementation of β-glucan as a prebiotic enhances growth activity and higher resistance action against pathogens in P. vannamei [227]. The prebiotic administration of β-glucans in diets is used to increase disease resistance; its efficiency depends on its origin and structure [228]. The glucan substance extracted from the cell walls of yeast (S. cerevisiae) has the ability to enhance the nonspecific immune system and disease resistance in Atlantic salmon [229].

3.3.2. Oligosaccharide

Oligosaccharide components are crucial for the modulation of immune responses in fish species. The positive results of monosaccharide products have encouraged the development of various immunomodulating, environmentally friendly nutrient diet supplements for fish species [230]. Dietary supplementation with 1 to 1.5 g kg−1 of MOS is capable of improving the growth activity and the efficiency of common carp fingerlings, as well as their antibacterial resistance against A. hydrophila infections [231]. Nutrient feed additives (FOS) in beluga (Huso huso) juveniles have numerous beneficial effects such as gut microbiota modulation, immune response, digestive enzyme action, and growth performance [232]. Dietary supplementation with FOS at different concentrations (0%, 0.5%, and 1%) over 7 weeks in common carp has been proven to have significant effects on intestinal microbiota modulation and physiological response [233]. The dietary supplementation of MOS at 0.4% improves the growth performance and nonspecific immune responses of Asian catfish (Clarias batrachus) juveniles [234]. The prebiotic FOS, when used as a feed additive in juvenile large yellow croakers, has been found to improve growth action and digestive enzyme action [13,235].
Not all prebiotic substances have immunostimulant properties; only a few references are available regarding the effects of isomalto-oligosaccharide (IMO), which consists of a combination of isomaltotriose, isomaltose, panose, and isomaltotetraose, on aquatic animals. No clear statement has been recorded regarding immune responses [236].

3.3.3. Chitosan

Chitosan is a linear polysaccharide component of β-(1–4)-linked D-glucosamine and is synthesized through alkaline deacetylation. It is a major component of arthropod exoskeletons, like those of shrimps, crabs, insects, and lobsters. In aquaculture, chitosan induces immunostimulation effects in various species, namely, rainbow trout [237], olive flounder (Paralichthys olivaceus) [238], and salmonids [239]. The administration of chitosan in the nutrient feed of C. carpio koi for 75 days resulted in significant effects such as an enhanced immune response, improved lipid metabolism, enhanced growth performance, and modulated intestine microbiota, thereby protecting the fish from pathogen invasion [240].

3.3.4. Inulin

The prebiotic component inulin, a soluble plant fiber, is used in fish diets and plays a crucial role in enhancing the immune system in both mammals and fish. In aquaculture, inulin finds significant use by activating beneficial bacteria, inhibiting pathogens, and boosting immune system activity [241]. Inulin has the potential to mitigate inflammation induced by a high-carbohydrate diet, thereby enhancing pathogen resistance in fish. Additionally, supplementing with inulin leads to changes in gut microbiota composition and its metabolites. These alterations likely contribute to alleviating the metabolic syndromes induced by a high-carbohydrate diet in fish [242].
Figure 2 summarizes the main components of prebiotics from natural sources and their main action modes in improving host health. The functional feed additives of prebiotics in aquatic animals are summarized in Table 3.

4. Postbiotics

4.1. Concept, Definition, and Major Components of Postbiotics

The use of live microorganisms as probiotics may have potential issues associated with gene resistance acquisition and translocation and depends on their viability [245]. Likewise, it has been recognized that non-viable microorganisms, as well as their components and metabolites, can have positive effects on health, leading to the appearance of the postbiotic concept [246]. Postbiotics are defined by consensus panels as preparations of inactivated microorganisms and/or their components (cell fragments, cell walls, metabolites) that have beneficial health effects on hosts [247]. This definition does not include purified metabolites in the absence of cells or cell components. One definition defines postbiotics as dead microbes and/or cell structures or metabolites that are produced via bacterial lysis or secreted during the fermentation process [248].
Postbiotics include inactivated probiotics called paraprobiotics; metabolites like short-chain fatty acids (SCFAs), vitamins, and phenolic acids; secreted proteins and peptides; functional proteins and enzymes; cell wall components like LTAs and peptidoglycan (PG)-derived muropeptides; secreted and extracellular polysaccharides (EPSs); cell lysates; cellular components (glycans, enzymes); the microbial fraction; and surface molecules such as pili [249,250].
Figure 3 outlines the main postbiotic components.

4.2. Action Modes and Applications of Postbiotics in Aquaculture

The action mechanisms of postbiotics are still unclear, but it is generally assumed that they are similar to those of live probiotics [251]. Three main mechanisms are involved in postbiotic action modes.

4.2.1. Immunomodulation via Microbial Compounds

Postbiotics act on the immune system through two signaling pathways, namely, nuclear factor-kB (NF-kB) and mitogen-activated protein kinase (MAPK), which are involved in immune and inflammatory responses. Postbiotics stimulate the innate and adaptive immune systems via external Toll-like receptors (TLRs), which recognize associated pathogens and bind to specific patterns such as LTAs and PGs. They also interact with intracellular nucleotide-like receptors (NLRs) and nucleotide-binding and oligomerization domain (NOD)-like receptors, which can bind to molecules like lipopolysaccharide (LPS), PG, and flagellin, thereby activating innate immune signaling pathways [248,250]. The role of PG recognition proteins in innate immune responses against pathogens has been demonstrated in fish [252,253]. PG-derived muropeptides from bacterial cell walls have been shown to boost the immune systems of fish [254] and shrimp [255]. For instance, muropeptides isolated from Bifidobacterium thermophilum have been proven to enhance shrimp immunity by increasing phagocytic activity or activating immune genes [255,256].
Additionally, postbiotics can enhance epithelial barrier protection via cell surface molecules such as pili and secreted protein P40 [257]. For example, the role of Lactobacillus pentosus surface protein on immune responses has been demonstrated in shrimp (L. vannamei) infected with Vibrio parahaemolyticus [258].

4.2.2. Antagonizing Pathogens via Antimicrobial Activities

Postbiotics exhibit antimicrobial activities against various pathogens because of the presence of metabolites like peptides and organic acids [259]. Bacteriocin JFP2 isolated from B. amyloliquefaciens exhibits antimicrobial activity against the fish pathogen A. hydrophila [260]. The dietary addition of postbiotics containing LAB (Lactobacillus) has been reported to protect rainbow trout (O. mykiss) against the bacterial fish pathogen L. garvieae after 30 days of feeding [261].

4.2.3. Inhibition of Oxidation via Antioxidant Enzyme Systems and Metabolites

Various postbiotics obtained from LAB have been shown to exhibit antioxidant activity, mainly attributed to phenolic compounds [262]. L. plantarum postbiotics have been documented to enhance antioxidant activity in animals [263]. In aquaculture applications, the overall antioxidant status of shrimp fed with diets supplemented with C. butyricum postbiotics was improved regarding an increase in alkaline phosphatase, acid phosphatase, total nitric oxide synthase, lysozyme, peroxidase, superoxide dismutase activities, total antioxidant capacity, and phenoloxidase content in the serum [264].
In aquaculture, postbiotics have been used as growth promoters instead of antibiotics, for immune system stimulation, and as disease control [257,265,266]. Recently, the potential application of postbiotics in aquaculture water quality in order to modulate bacterioplankton communities and influence nutrient cycling and bacterial pathogen abundance was reported [267]. Figure 4 illustrates the potential applications of postbiotics in aquaculture. Table 4 shows some recent potential applications of postbiotics in aquaculture.

5. Synbiotics

Synbiotics refer to dietary additives that blend probiotics and prebiotics in a synergistic combination, thereby enhancing their beneficial effects. When either dietary additives or supplements are used, the resulting positive effects typically follow one of three patterns: ingredient effects, synergism, or potentiation. Supplementation outcomes occur when the combined effects of both additives used together approximate the sum of the effects of the individual supplements. In the case of synergism, the amalgamated result of the two products is significantly greater than the sum of the effects of each factor administered alone. The term potentiation is used differently; some pharmacologists interchange it with synergism to describe a result that is better than that of a supplement alone, while others use it to describe an outcome that is only present when both substances are used simultaneously [273,274].

5.1. Possible Modes of Action of Synbiotics in Aquaculture

5.1.1. Synbiotics Enhance Digestive Enzyme and Growth Performance

Dietary administration with synbiotics is helpful in enhancing the digestive enzymatic activities of fish, allowing the host to degrade more nutrients. This dietary method increases digestive action and likely enhances the weight gain rate and/or feed efficiency [275]. Nutrient diet supplementation with a mixture of probiotics and monosaccharides enhances feed efficiency and overall health in carp. However, limited data are available in aquaculture regarding the function of the nutrient diet supplementation of synbiotics in carp [24]. Nutrient diet administration with synbiotics enhances the lymphocytes and white blood cells in carp [276]. Synbiotics (IMBO), a combination of probiotics (E. faecium) and prebiotics (FOS), have been used to enhance the growth performance, survival rate, and digestive enzyme function of common carp fingerlings [277]. Dietary supplementation with FOS, MOS, and B. clausii can improve the growth performance and health benefits of the Japanese flounder more than a control diet [192]. Dietary supplementation with FOS and 1.35 × 107 CFU g−1 B. subtilis (single or mixed) increases the specific growth rate (SGR) and feed efficiency ratio (FER) compared with the groups without B. subtilis additives in juvenile large yellow croakers (Larimichthys crocea) [235]. Figure 5 illustrates the possible modes of action of synbiotics in aquaculture.

5.1.2. Synbiotics Improve Immune Response and Disease Resistance

An amalgamation of probiotic and prebiotic feed supplements is mainly helpful in enhancing the survival of beneficial organisms, as the presence of prebiotics protects well-organized fermentation. Finally, this rewards the host with a suitable approach [278]. The nutritional additives of probiotics and prebiotics (MOS, FOS, and inulin) enhance fish immune systems via the GIT [24,279,280]. A synbiotic composed of Pediococcus acidilactici and galactooligosaccharides improved immune parameters and antagonistic activity against S. iniae when administered to rainbow trout fingerlings for 8 weeks [281]. The combination of probiotic Bacillus spp. and 0.2% prebiotic isomaltooligosaccharide was used to improve immune functions in shrimp (Penaeus japonicas) against V. alginolyticus infection [282]. In addition, the blended use of Bacillus and molasses improved the microbial population and enhanced the development of the probiotic community and inhibitory activity against pathogens in Pacific white shrimp [283]. The effectiveness of a synbiotic treatment in conditions of defense against infectious factors can be evaluated with a confrontation examination given its regulatory power over harmful microbes and its capability to resist infections [284]. The functional feed additives of synbiotics in aquatic animals are summarized in Table 5.

6. Limitations of the Use of Synbiotic Agents in Aquaculture

The use of synbiotic agents in aquaculture instead of antibiotics has recently gained significant interest [290]. Probiotics have been shown to be effective in promoting growth, increasing immunity, and improving resistance to infections in aquatic animals [291]. The major limitation of their use comes from the problem of possible gene resistance acquisition and translocation, as well as the question of their viability and/or ability to colonize the fish gut [245]. The use of multi-strain probiotics increases the possibility of strain survival rates and, therefore, improves the beneficial effects on the growth, immunity, and infection resistance of aquatic animals [152]. Postbiotics present an advantage over probiotics because they do not have viability problems and are less susceptible to environmental conditions [245,292]. Additionally, they generally have a complex composition made up of several compounds that play multiple roles and have numerous beneficial effects on aquatic animals. However, their use in managing infectious diseases is still in its early stages [259].
Prebiotics, as inert biotic agents, are relatively safe and cost-effective alternatives to probiotics. Several studies on their immunostimulant properties and growth promotion in fish and shellfish have shown some evidence for their use in aquaculture [293]. Nevertheless, studies on the optimal dose should be carried out, as inadequate doses may lead to detrimental effects on aquatic animals [206,232]. Synbiotics improve the colonization of microorganisms in the intestines and are generally more effective than probiotics or prebiotics alone [292]. For example, Nile tilapia (O. niloticus) fed with synbiotics showed the highest increase in specific growth rate compared with a group fed with probiotics or prebiotics alone [276,294]. Extensive studies are still needed to specify the role of prebiotics, probiotics, postbiotics, and synbiotics in growth performance, intestinal health, and immune aspects with a focus on the mechanisms underlying the synbiotic diet in aquatic animals against various pathogens. The mode of administration and dose of the biotic agents are also important and certainly have an impact on their effectiveness [295].
The economic aspect of utilizing synbiotics and their components could be a limitation in aquaculture production. In the context of intensive aquaculture practices, the aspect of feeding comprises a substantial 60–80% of operational costs [296]. A Probiotic application in larval whiteleg shrimp (L. vannamei) resulted in a 6% increase in total production costs. However, the result of a higher survival rate contributed to a 44% reduction in unit production costs [297]. Studies on the feasibility of synbiotics in aquaculture have consistently shown improvement in economic efficiency compared with control diets, especially when aquatic animals have been under stress conditions such as high stocking density [298] or during the reproductive period [299].

7. Concluding Remarks and Future Perspectives

In conclusion, the aquaculture sector has experienced substantial growth in recent decades, confronting challenges related to environmental degradation and disease outbreaks, primarily because of the widespread prophylactic use of antibiotics and drugs. Synbiotic agents and their components, namely, probiotics, prebiotics, and postbiotics, emerge as natural and sustainable solutions considering their beneficial effects on growth performance, immunity, and overall health. These outcomes can be achieved by directly acting on aquatic animals through feeding or indirectly by improving the environment and water quality.
The direct-action mechanisms of these biotic family agents involve the modulation of the gut microbiota, leading to enhanced growth performance and feed utilization, as well as the reinforcement of the immune response, which helps aquatic animals resist pathogenic organisms. Indirectly, these natural solutions can assist in detoxifying the aquaculture system by removing heavy metals through biosorption, bioaccumulation, and bioprecipitation mechanisms, either through cellular-energy-dependent processes or not.
Moreover, these functional feed ingredients appear to be good alternatives to antibiotics and synthetic drugs given their multiple mechanisms of action in aquaculture, which help mitigate issues related to antibiotic resistance and the accumulation of harmful residues. While several study reports are available on probiotics, prebiotics, and synbiotics for the purpose of driving the development of aquaculture health and production, extensive studies are still needed at different levels for a deeper understanding of the mechanisms corresponding to the role of each component and combination in the growth performance, intestinal health, and immune aspects of aquatic animals. Furthermore, postbiotics, which are components or metabolites from dead probiotic microorganisms, such as functional amino acids, fatty acids, enzymes, exopolysaccharides, and organic acids, show promise as feed components because of their abilities to enhance the innate immune system, disease resistance, and growth and survival rates of aquatic animals.
Beyond the consideration of such biotic family agents and their combination with other functional ingredients such as herbs, it is also important to pay attention to combining biological solutions with other emerging technologies, such as nanoparticle-based delivery methods, in the future to improve efficiency in disease management, feeding formulation, and water quality.

Author Contributions

Conceptualization, V.S. and H.L.R.; writing—original draft preparation, V.S.; writing—review and editing, H.L.R., H.N.R. and H.-T.N.; supervision, H.L.R. and Y.-Z.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gephart, J.A.; Golden, C.D.; Asche, F.; Belton, B.; Brugere, C.; Froehlich, H.E.; Fry, J.P.; Halpern, B.S.; Hicks, C.C.; Jones, R.C. Scenarios for Global Aquaculture and Its Role in Human Nutrition. Rev. Fish. Sci. Aquac. 2020, 29, 122–138. [Google Scholar] [CrossRef]
  2. Pepi, M.; Focardi, S. Antibiotic-Resistant Bacteria in Aquaculture and Climate Change: A Challenge for Health in the Mediterranean Area. Int. J. Environ. Res. Public Health 2021, 18, 5723. [Google Scholar] [CrossRef] [PubMed]
  3. Tavares-Dias, M. Toxicity, Physiological, Histopathological and Antiparasitic Effects of the Formalin, a Chemotherapeutic of Fish Aquaculture. Aquac. Res. 2021, 52, 1803–1823. [Google Scholar] [CrossRef]
  4. Mohamed, S.; Nagaraj, G.; Chua, F.H.C.; Wang, Y.G. The Use of Chemicals in Aquaculture in Malaysia and Singapore. In Proceedings of the Use of Chemicals in Aquaculture in Asia, Iloilo, Philippines, 20–22 May 1996; Aquaculture Department, Southeast Asian Fisheries Development Center: Iloilo, Philippines, 2000; pp. 127–140. [Google Scholar]
  5. Ghelichpour, M.; Rajabiesterabadi, H.; Hoseini, S.M. Gill Histopathological Characteristics of Caspian Roach (Rutilus rutilus caspicus) Fingerlings Treated with Potassium Permanganate and Formalin. Aquac. Res. 2016, 47, 276–282. [Google Scholar] [CrossRef]
  6. Leal, J.F.; Neves, M.G.P.M.S.; Santos, E.B.H.; Esteves, V.I. Use of Formalin in Intensive Aquaculture: Properties, Application and Effects on Fish and Water Quality. Rev. Aquac. 2018, 10, 281–295. [Google Scholar] [CrossRef]
  7. Zhang, X.; Zhang, J.; Han, Q.; Wang, X.; Wang, S.; Yuan, X.; Zhang, B.; Zhao, S. Antibiotics in Mariculture Organisms of Different Growth Stages: Tissue-Specific Bioaccumulation and Influencing Factors. Environ. Pollut. 2021, 288, 117715. [Google Scholar] [CrossRef] [PubMed]
  8. Alfred, O.; Shaahu, A.; Orban, D.A.; Egwenomhe, M. An Overview on Understanding the Basic Concept of Fish Diseases in Aquaculture. IRE J. 2020, 4, 83–91. [Google Scholar]
  9. Essawi, T.; Srour, M. Screening of Some Palestinian Medicinal Plants for Antibacterial Activity. J. Ethnopharmacol. 2000, 70, 343–349. [Google Scholar] [CrossRef]
  10. Hoseinifar, S.H.; Zou, H.K.; Miandare, H.K.; Van Doan, H.; Romano, N.; Dadar, M. Enrichment of Common Carp (Cyprinus carpio) Diet with Medlar (Mespilus germanica) Leaf Extract: Effects on Skin Mucosal Immunity and Growth Performance. Fish Shellfish Immunol. 2017, 67, 346–352. [Google Scholar] [CrossRef]
  11. Musthafa, M.S.; Asgari, S.M.; Kurian, A.; Elumalai, P.; Ali, A.R.J.; Paray, B.A.; Al-Sadoon, M.K. Protective Efficacy of Mucuna pruriens (L.) Seed Meal Enriched Diet on Growth Performance, Innate Immunity, and Disease Resistance in Oreochromis Mossambicus against Aeromonas hydrophila. Fish Shellfish Immunol. 2018, 75, 374–380. [Google Scholar] [CrossRef]
  12. Safety, S. FDA Needs to Improve Oversight of Imported Seafood and Better Leverage Limited Resources; United States Government Accountability Office: Washington, DC, USA, 2011.
  13. Yilmaz, S.; Yilmaz, E.; Dawood, M.A.; Ringø, E.; Ahmadifar, E.; Abdel-Latif, H.M. Probiotics, Prebiotics, and Synbiotics Used to Control Vibriosis in Fish: A Review. Aquaculture 2022, 547, 737514. [Google Scholar] [CrossRef]
  14. Wang, A.R.; Ran, C.; Ringø, E.; Zhou, Z.G. Progress in Fish Gastrointestinal Microbiota Research. Rev. Aquac. 2018, 10, 626–640. [Google Scholar] [CrossRef]
  15. Luna, G.M.; Quero, G.M.; Kokou, F.; Kormas, K. Time to Integrate Biotechnological Approaches into Fish Gut Microbiome Research. Curr. Opin. Biotechnol. 2022, 73, 121–127. [Google Scholar] [CrossRef] [PubMed]
  16. Jia, P.-P.; Junaid, M.; Wen, P.-P.; Yang, Y.-F.; Li, W.-G.; Yang, X.-G.; Pei, D.-S. Role of Germ-Free Animal Models in Understanding Interactions of Gut Microbiota to Host and Environmental Health: A Special Reference to Zebrafish. Environ. Pollut. 2021, 279, 116925. [Google Scholar] [CrossRef]
  17. Bates, J.M.; Mittge, E.; Kuhlman, J.; Baden, K.N.; Cheesman, S.E.; Guillemin, K. Distinct Signals from the Microbiota Promote Different Aspects of Zebrafish Gut Differentiation. Dev. Biol. 2006, 297, 374–386. [Google Scholar] [CrossRef] [PubMed]
  18. Ye, L.; Rawls, J.F. Microbial Influences on Gut Development and Gut-Brain Communication. Development 2021, 148, dev194936. [Google Scholar] [CrossRef] [PubMed]
  19. Koch, B.E.; Yang, S.; Lamers, G.; Stougaard, J.; Spaink, H.P. Intestinal Microbiome Adjusts the Innate Immune Setpoint during Colonization through Negative Regulation of MyD88. Nat. Commun. 2018, 9, 4099. [Google Scholar] [CrossRef] [PubMed]
  20. Tran, N.T.; Yang, W.; Nguyen, X.T.; Zhang, M.; Ma, H.; Zheng, H.; Zhang, Y.; Chan, K.-G.; Li, S. Application of Heat-Killed Probiotics in Aquaculture. Aquaculture 2022, 548, 737700. [Google Scholar] [CrossRef]
  21. Yang, H.-L.; Sun, Y.-Z.; Hu, X.; Ye, J.; Lu, K.-L.; Hu, L.-H.; Zhang, J.-J. Bacillus Pumilus SE5 Originated PG and LTA Tuned the Intestinal TLRs/MyD88 Signaling and Microbiota in Grouper (Epinephelus coioides). Fish Shellfish Immunol. 2019, 88, 266–271. [Google Scholar] [CrossRef]
  22. Van Doan, H. Bacillus spp. in Aquaculture-Mechanisms and Applications: An Update View. In Probiotic Bacteria and Postbiotic Metabolites: Role in Animal and Human Health; Springer: Berlin/Heidelberg, Germany, 2021; pp. 1–59. [Google Scholar] [CrossRef]
  23. Kim, Y.-R.; Kim, E.-Y.; Choi, S.; Hossain, M.T.; Oh, R.-K.; Heo, W.-S.; Lee, J.-M.; Cho, Y.-C.; Kong, I.-S. Effect of a Probiotic Strain, Enterococcus Faecium, on the Immune Responses of Olive Flounder (Paralichthys olivaceus). J. Microbiol. Biotechnol. 2012, 22, 526–529. [Google Scholar] [CrossRef]
  24. Mugwanya, M.; Dawood, M.A.; Kimera, F.; Sewilam, H. Updating the Role of Probiotics, Prebiotics, and Synbiotics for Tilapia Aquaculture as Leading Candidates for Food Sustainability: A Review. Probiotics Antimicrob. Proteins 2021, 14, 130–157. [Google Scholar] [CrossRef] [PubMed]
  25. Hoseinifar, S.H.; Shakouri, M.; Yousefi, S.; Van Doan, H.; Shafiei, S.; Yousefi, M.; Mazandarani, M.; Mozanzadeh, M.T.; Tulino, M.G.; Faggio, C. Humoral and Skin Mucosal Immune Parameters, Intestinal Immune Related Genes Expression and Antioxidant Defense in Rainbow Trout (Oncorhynchus mykiss) Fed Olive (Olea europea L.) Waste. Fish Shellfish Immunol. 2020, 100, 171–178. [Google Scholar] [CrossRef] [PubMed]
  26. Da Silva Liebl, A.R.; Cáo, M.A.; dos Santos Nascimento, M.; da Castro, P.D.S.; Duncan, W.L.P.; Pantoja-Lima, J.; Aride, P.H.R.; Bussons, M.R.F.M.; Furuya, W.M.; Faggio, C. Dietary Lysine Requirements of Colossoma macropomum (Cuvier, 1818) Based on Growth Performance, Hepatic and Intestinal Morphohistology and Hematology. Vet. Res. Commun. 2022, 46, 9–25. [Google Scholar] [CrossRef] [PubMed]
  27. Mahboub, H.H.; Faggio, C.; Hendam, B.M.; Algharib, S.A.; Alkafafy, M.; Hashem, M.A.; Mahmoud, Y.K.; Khamis, T.; Abdel-Ghany, H.M.; Masoud, S.R. Immune-Antioxidant Trait, Aeromonas Veronii Resistance, Growth, Intestinal Architecture, and Splenic Cytokines Expression of Cyprinus carpio Fed Prunus armeniaca Kernel-Enriched Diets. Fish Shellfish Immunol. 2022, 124, 182–191. [Google Scholar] [CrossRef] [PubMed]
  28. Al-Shawi, S.G.; Dang, D.S.; Yousif, A.Y.; Al-Younis, Z.K.; Najm, T.A.; Matarneh, S.K. The Potential Use of Probiotics to Improve Animal Health, Efficiency, and Meat Quality: A Review. Agriculture 2020, 10, 452. [Google Scholar] [CrossRef]
  29. Adel, M.; Dawood, M.A. Probiotics Application: Implications for Sustainable Aquaculture. In Probiotic Bacteria and Postbiotic Metabolites: Role in Animal and Human Health; Springer: Berlin/Heidelberg, Germany, 2021; pp. 191–219. [Google Scholar]
  30. FAO/WHO Expert Consultation. Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Acid Bacteria; World Health Organization: Córdoba, Spain, 2001. [Google Scholar]
  31. Guardiola, F.A.; Porcino, C.; Cerezuela, R.; Cuesta, A.; Faggio, C.; Esteban, M.A. Impact of Date Palm Fruits Extracts and Probiotic Enriched Diet on Antioxidant Status, Innate Immune Response and Immune-Related Gene Expression of European Seabass (Dicentrarchus labrax). Fish Shellfish Immunol. 2016, 52, 298–308. [Google Scholar] [CrossRef] [PubMed]
  32. Van Doan, H.; Hoseinifar, S.H.; Ringø, E.; Ángeles Esteban, M.; Dadar, M.; Dawood, M.A.; Faggio, C. Host-Associated Probiotics: A Key Factor in Sustainable Aquaculture. Rev. Fish. Sci. Aquac. 2020, 28, 16–42. [Google Scholar] [CrossRef]
  33. Morshedi, V.; Bojarski, B.; Hamedi, S.; Torahi, H.; Hashemi, G.; Faggio, C. Effects of Dietary Bovine Lactoferrin on Growth Performance and Immuno-Physiological Responses of Asian Sea Bass (Lates calcarifer) Fingerlings. Probiotics Antimicrob. Proteins 2021, 13, 1790–1797. [Google Scholar] [CrossRef]
  34. Mirbakhsh, M.; Ghaednia, B.; Zorriehzahra, M.J.; Esmaeili, F.; Faggio, C. Dietary Mixed and Sprayed Probiotic Improves Growth Performance and Digestive Enzymes of Juvenile Whiteleg Shrimp (Litopenaeus vannamei, Boone, 1931). J. Appl. Aquac. 2022, 35, 823–836. [Google Scholar] [CrossRef]
  35. Misra, S.; Pandey, P.; Mishra, H.N. Novel Approaches for Co-Encapsulation of Probiotic Bacteria with Bioactive Compounds, Their Health Benefits and Functional Food Product Development: A Review. Trends Food Sci. Technol. 2021, 109, 340–351. [Google Scholar] [CrossRef]
  36. El-Kady, A.A.; Magouz, F.I.; Mahmoud, S.A.; Abdel-Rahim, M.M. The Effects of Some Commercial Probiotics as Water Additive on Water Quality, Fish Performance, Blood Biochemical Parameters, Expression of Growth and Immune-Related Genes, and Histology of Nile Tilapia (Oreochromis niloticus). Aquaculture 2022, 546, 737249. [Google Scholar] [CrossRef]
  37. Vijayaram, S.; Kannan, S. Probiotics: The Marvelous Factor and Health Benefits. Biomed. Biotechnol. Res. J. BBRJ 2018, 2, 1–8. [Google Scholar] [CrossRef]
  38. Cıl, G.I.; Bulut, G.; Budak, D.; Camkerten, G.; Camkerten, I. Probiotics and Functional Feed. In Probiotics, the Natural Microbiota in Living Organisms; CRC Press: Boca Raton, FL, USA, 2021; pp. 315–342. ISBN 1-351-02754-9. [Google Scholar]
  39. Foysal, M.J.; Alam, M.; Kawser, A.R.; Hasan, F.; Rahman, M.M.; Tay, C.-Y.; Prodhan, M.S.H.; Gupta, S.K. Meta-Omics Technologies Reveals Beneficiary Effects of Lactobacillus plantarum as Dietary Supplements on Gut Microbiota, Immune Response and Disease Resistance of Nile Tilapia (Oreochromis niloticus). Aquaculture 2020, 520, 734974. [Google Scholar] [CrossRef]
  40. Zaineldin, A.I.; Hegazi, S.; Koshio, S.; Ishikawa, M.; Dawood, M.A.; Dossou, S.; Yukun, Z.; Mzengereza, K. Singular Effects of Bacillus Subtilis C-3102 or Saccharomyces Cerevisiae Type 1 on the Growth, Gut Morphology, Immunity, and Stress Resistance of Red Sea Bream (Pagrus major); Mzuzu University: Mzuzu, Malawi, 2021. [Google Scholar] [CrossRef]
  41. Kong, Y.; Gao, C.; Du, X.; Zhao, J.; Li, M.; Shan, X.; Wang, G. Effects of Single or Conjoint Administration of Lactic Acid Bacteria as Potential Probiotics on Growth, Immune Response and Disease Resistance of Snakehead Fish (Channa argus). Fish Shellfish Immunol. 2020, 102, 412–421. [Google Scholar] [CrossRef] [PubMed]
  42. Kord, M.I.; Maulu, S.; Srour, T.M.; Omar, E.A.; Farag, A.A.; Nour, A.A.M.; Hasimuna, O.J.; Abdel-Tawwab, M.; Khalil, H.S. Impacts of Water Additives on Water Quality, Production Efficiency, Intestinal Morphology, Gut Microbiota, and Immunological Responses of Nile tilapia Fingerlings under a Zero-Water-Exchange System. Aquaculture 2022, 547, 737503. [Google Scholar] [CrossRef]
  43. Ayivi, R.D.; Gyawali, R.; Krastanov, A.; Aljaloud, S.O.; Worku, M.; Tahergorabi, R.; da Silva, R.C.; Ibrahim, S.A. Lactic Acid Bacteria: Food Safety and Human Health Applications. Dairy 2020, 1, 202–232. [Google Scholar] [CrossRef]
  44. Feng, T.; Wang, J. Oxidative Stress Tolerance and Antioxidant Capacity of Lactic Acid Bacteria as Probiotic: A Systematic Review. Gut Microbes 2020, 12, 1801944. [Google Scholar] [CrossRef]
  45. Tiwari, S.K.; Dicks, L.M.; Popov, I.V.; Karaseva, A.; Ermakov, A.M.; Suvorov, A.; Tagg, J.R.; Weeks, R.; Chikindas, M.L. Probiotics at War against Viruses: What Is Missing from the Picture? Front. Microbiol. 2020, 11, 1877. [Google Scholar] [CrossRef]
  46. Govindaraj, K.; Samayanpaulraj, V.; Narayanadoss, V.; Uthandakalaipandian, R. Isolation of Lactic Acid Bacteria from Intestine of Freshwater Fishes and Elucidation of Probiotic Potential for Aquaculture Application. Probiotics Antimicrob. Proteins 2021, 13, 1598–1610. [Google Scholar] [CrossRef]
  47. Khalid, F.; Khalid, A.; Fu, Y.; Hu, Q.; Zheng, Y.; Khan, S.; Wang, Z. Potential of Bacillus velezensis as a Probiotic in Animal Feed: A Review. J. Microbiol. 2021, 59, 627–633. [Google Scholar] [CrossRef]
  48. Vallesi, A.; Pucciarelli, S.; Buonanno, F.; Fontana, A.; Mangiagalli, M. Bioactive Molecules from Protists: Perspectives in Biotechnology. Eur. J. Protistol. 2020, 75, 125720. [Google Scholar] [CrossRef]
  49. Butt, U.D.; Lin, N.; Akhter, N.; Siddiqui, T.; Li, S.; Wu, B. Overview of the Latest Developments in the Role of Probiotics, Prebiotics and Synbiotics in Shrimp Aquaculture. Fish Shellfish Immunol. 2021, 114, 263–281. [Google Scholar] [CrossRef]
  50. Labba, I.-C.M.; Andlid, T.; Lindgren, Å.; Sandberg, A.-S.; Sjöberg, F. Isolation, Identification, and Selection of Strains as Candidate Probiotics and Starters for Fermentation of Swedish Legumes. Food Nutr. Res. 2020, 64. [Google Scholar] [CrossRef] [PubMed]
  51. Rajyalakshmi, K.; Babu, M.K.; Shabana, S.; Satya, A.K. Identification and Screening of Probiotics as a Biocontrol Agent against Pathogenic Vibriosis in Shrimp Aquaculture. Ann. Rom. Soc. Cell Biol. 2021, 25, 12292–12305. [Google Scholar]
  52. 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]
  53. Zhao, C.; Guo, G.; Li, Z.; Chen, J.; Ren, Y. Effects of Probiotics (Bacillus coagulans) Supplementation after Antibiotic Administration on Growth, Immunity, and Intestinal Microflora in Turbot Scophthalmus maximus. Aquac. Int. 2023. [Google Scholar] [CrossRef]
  54. Galagarza, O.A.; Smith, S.A.; Drahos, D.J.; Eifert, J.D.; Williams, R.C.; Kuhn, D.D. Modulation of Innate Immunity in Nile tilapia (Oreochromis niloticus) by Dietary Supplementation of Bacillus subtilis Endospores. Fish Shellfish Immunol. 2018, 83, 171–179. [Google Scholar] [CrossRef] [PubMed]
  55. Qin, L.U.; Xiang, J.; Xiong, F.; Wang, G.; Zou, H.; Li, W.; Li, M.; Wu, S. Effects of Bacillus Licheniformis on the Growth, Antioxidant Capacity, Intestinal Barrier and Disease Resistance of Grass Carp (Ctenopharyngodon idella). Fish Shellfish Immunol. 2020, 97, 344–350. [Google Scholar] [CrossRef]
  56. Das, S.; Mondal, K.; Sengupta, C. Evaluation of the Probiotic Potential of Streptomyces antibioticus and Bacillus cereus on Growth Performance of Freshwater Catfish Heteropneustes Fossilis. Aquac. Rep. 2021, 20, 100752. [Google Scholar] [CrossRef]
  57. Loghmani, H.; Khalili Hadad, B.; Kazempoor, R.; Sh, A.S. Investigation of the Effects of Bifidobacterium Bifidum as a Probiotic on Liver Function Enzymes Due to Exposure to E. Coli. O157H7 in Koi Fish (Cyprinus rubrofuscus). J. Surv. Fish. Sci. 2022, 5, 27-3. [Google Scholar] [CrossRef]
  58. Puvanendran, V.; Rud, I.; Breiland, M.S.W.; Arnesen, J.-A.; Axelsson, L. Probiotic Carnobacterium Divergens Increase Growth Parameters and Disease Resistance in Farmed Atlantic Cod (Gadus morhua) Larvae without Influencing the Microbiota. Aquaculture 2021, 532, 736072. [Google Scholar] [CrossRef]
  59. Tachibana, L.; Telli, G.S.; de Carla Dias, D.; Goncalves, G.S.; Ishikawa, C.M.; Cavalcante, R.B.; Natori, M.M.; Hamed, S.B.; Ranzani-Paiva, M.J.T. Effect of Feeding Strategy of Probiotic Enterococcus Faecium on Growth Performance, Hematologic, Biochemical Parameters and Non-Specific Immune Response of Nile Tilapia. Aquac. Rep. 2020, 16, 100277. [Google Scholar] [CrossRef]
  60. Tian, J.-X.; Kang, Y.-H.; Chu, G.-S.; Liu, H.-J.; Kong, Y.-D.; Zhao, L.-H.; Kong, Y.-X.; Shan, X.-F.; Wang, G.-Q. Oral Administration of Lactobacillus Casei Expressing Flagellin A Protein Confers Effective Protection against Aeromonas Veronii in Common Carp, Cyprinus carpio. Int. J. Mol. Sci. 2019, 21, 33. [Google Scholar] [CrossRef] [PubMed]
  61. Sagada, G.; Gray, N.; Wang, L.; Xu, B.; Zheng, L.; Zhong, Z.; Ullah, S.; Tegomo, A.F.; Shao, Q. Effect of Dietary Inactivated Lactobacillus plantarum on Growth Performance, Antioxidative Capacity, and Intestinal Integrity of Black Sea Bream (Acanthopagrus schlegelii) Fingerlings. Aquaculture 2021, 535, 736370. [Google Scholar] [CrossRef]
  62. Noshair, I.; Kanwal, Z.; Jabeen, G.; Arshad, M.; Yunus, F.-U.-N.; Hafeez, R.; Mairaj, R.; Haider, I.; Ahmad, N.; Alomar, S.Y. Assessment of Dietary Supplementation of Lactobacillus rhamnosus Probiotic on Growth Performance and Disease Resistance in Oreochromis niloticus. Microorganisms 2023, 11, 1423. [Google Scholar] [CrossRef] [PubMed]
  63. Zhu, C.-Z.; Li, D.; Chen, W.-J.; Ban, S.-N.; Liu, T.; Wen, H.; Jiang, M. Effects of Dietary Host-Associated Lactococcus lactis on Growth Performance, Disease Resistance, Intestinal Morphology and Intestinal Microbiota of Mandarin Fish (Siniperca chuatsi). Aquaculture 2021, 540, 736702. [Google Scholar] [CrossRef]
  64. Al-Hisnawi, A.; Rodiles, A.; Rawling, M.D.; Castex, M.; Waines, P.; Gioacchini, G.; Carnevali, O.; Merrifield, D.L. Dietary Probiotic Pediococcus acidilactici MA18/5M Modulates the Intestinal Microbiota and Stimulates Intestinal Immunity in Rainbow Trout (Oncorhynchus mykiss). J. World Aquac. Soc. 2019, 50, 1133–1151. [Google Scholar] [CrossRef]
  65. Liang, Q.; Liu, G.; Guo, Z.; Wang, Y.; Xu, Z.; Ren, Y.; Zhang, Q.; Cui, M.; Zhao, X.; Xu, D. Application of Potential Probiotic Strain Streptomyces sp. SH5 on Anti-Aeromonas Infection in Zebrafish Larvae. Fish Shellfish Immunol. 2022, 127, 375–385. [Google Scholar] [CrossRef]
  66. Boonanuntanasarn, S.; Ditthab, K.; Jangprai, A.; Nakharuthai, C. Effects of Microencapsulated Saccharomyces cerevisiae on Growth, Hematological Indices, Blood Chemical, and Immune Parameters and Intestinal Morphology in Striped Catfish, Pangasianodon hypophthalmus. Probiotics Antimicrob. Proteins 2019, 11, 427–437. [Google Scholar] [CrossRef]
  67. Zamini, A.; Tehranifard, A. Effects of Lactococcus lactis and Weissella Cibaria as Probiotic on Growth Performance, Intestinal Bacterial Flora, Digestive Enzymes and Intestinal Histology in Common Carp (Cyprinus Carpio). J. Aquac. Dev. 2021, 15, 13–29. [Google Scholar]
  68. Mingmongkolchai, S.; Panbangred, W. Bacillus Probiotics: An Alternative to Antibiotics for Livestock Production. J. Appl. Microbiol. 2018, 124, 1334–1346. [Google Scholar] [CrossRef]
  69. Ringø, E. Probiotics in Shellfish Aquaculture. Aquac. Fish. 2020, 5, 1–27. [Google Scholar] [CrossRef]
  70. Xia, Y.; Yu, E.; Lu, M.; Xie, J. Effects of Probiotic Supplementation on Gut Microbiota as Well as Metabolite Profiles within Nile tilapia, Oreochromis niloticus. Aquaculture 2020, 527, 735428. [Google Scholar] [CrossRef]
  71. Yukgehnaish, K.; Kumar, P.; Sivachandran, P.; Marimuthu, K.; Arshad, A.; Paray, B.A.; Arockiaraj, J. Gut Microbiota Metagenomics in Aquaculture: Factors Influencing Gut Microbiome and Its Physiological Role in Fish. Rev. Aquac. 2020, 12, 1903–1927. [Google Scholar] [CrossRef]
  72. Wu, G. Nutrition and Metabolism: Foundations for Animal Growth, Development, Reproduction, and Health. In Recent Advances in Animal Nutrition and Metabolism; Springer: Berlin/Heidelberg, Germany, 2022; pp. 1–24. [Google Scholar] [CrossRef]
  73. LeBlanc, J.G.; Chain, F.; Martín, R.; Bermúdez-Humarán, L.G.; Courau, S.; Langella, P. Beneficial Effects on Host Energy Metabolism of Short-Chain Fatty Acids and Vitamins Produced by Commensal and Probiotic Bacteria. Microb. Cell Factories 2017, 16, 79. [Google Scholar] [CrossRef] [PubMed]
  74. Morais, T.; Inácio, A.; Coutinho, T.; Ministro, M.; Cotas, J.; Pereira, L.; Bahcevandziev, K. Seaweed Potential in the Animal Feed: A Review. J. Mar. Sci. Eng. 2020, 8, 559. [Google Scholar] [CrossRef]
  75. Hardy, R.W.; Kaushik, S.J.; Mai, K.; Bai, S.C. Fish Nutrition—History and Perspectives. In Fish Nutrition; Elsevier: Amsterdam, The Netherlands, 2022; pp. 1–16. [Google Scholar]
  76. Uma, A.; Subash, P.; Abraham, T.J. Importance of Gut Microbiota in Fish–A Review. Indian J. Anim. Health 2020, 59, 181–194. [Google Scholar] [CrossRef]
  77. Singh, S.K.; Bhandari, M.P.; Shrestha, S.; Koirala, U.; Gurung, G.B. Supplementation of Commercial Probiotics in Feed for Growth and Survival of Rainbow Trout (Oncorhynchus mykiss). In Proceedings of the National Workshop on Livestock and Fisheries Research in Nepal, Kathmandu, Nepal, 3–4 March 2021; Volume 3, p. 275. [Google Scholar]
  78. Chen, J.; Sun, D.; Cui, H.; Rao, C.; Li, L.; Guo, S.; Yang, S.; Zhang, Y.; Cao, X. Toxic Effects of Carbon Quantum Dots on the Gut–Liver Axis and Gut Microbiota in the Common Carp Cyprinus carpio. Environ. Sci. Nano 2022, 9, 173–188. [Google Scholar] [CrossRef]
  79. Phianphak, W.; Rengpipat, S.; Piyatiratitivorakul, S.; Menasveta, P. Probiotic Use of Lactobacillus spp. for Black Tiger Shrimp, Penaeus monodon. J. Sci. Res. Chula Univ. 1999, 24, 41–58. [Google Scholar]
  80. Tuan, T.N.; Duc, P.M.; Hatai, K. Overview of the Use of Probiotics in Aquaculture. Int. J. Res. Fish. Aquac. 2013, 3, 89–97. [Google Scholar]
  81. El-Saadony, M.T.; Alagawany, M.; Patra, A.K.; Kar, I.; Tiwari, R.; Dawood, M.A.; Dhama, K.; Abdel-Latif, H.M. The Functionality of Probiotics in Aquaculture: An Overview. Fish Shellfish Immunol. 2021, 117, 36–52. [Google Scholar] [CrossRef]
  82. Yassir, R.Y.; Adel, M.E.; Azze, A. Use of Probiotic Bacteria as Growth Promoters, Antibacterial and the Effect on Physiological Parameters of Orechromis niloticus. J. Fish Dis. 2002, 22, 633–642. [Google Scholar]
  83. Swain, S.; Hauzoukim, S.K.G.; Das, S.K.; Roy, A. Application of Probiotics in Aquaculture. Pharma innov. 2021, 10, 146–149. Available online: https://www.thepharmajournal.com/archives/2021/vol10issue7S/PartC/S-10-6-136-245.pdf (accessed on 1 November 2023).
  84. Franca, F.M.; Danielle de Carla, D.; Teixeira, P.C.; Marcantonio, A.S.; de Stefani, M.V.; Antonucci, A.; Da Rocha, G.; Ranzani-PAIVA, M.J.T.; Ferreira, C.M. Efeito Do Probiótico Bacillus Subtilis No Crescimento, Sobrevivência e Fisiologia de Rãs-Touro (Rana catesbeiana). Bol. Inst. Pesca 2008, 34, 403–412. [Google Scholar]
  85. Wang, Z.; Yang, M.; Wang, L.; Lu, K.; Song, K.; Zhang, C. Bacillus Subtilis LCBS1 Supplementation and Replacement of Fish Meal with Fermented Soybean Meal in Bullfrog (Lithobates catesbeianus) Diets: Effects on Growth Performance, Feed Digestibility and Gut Health. Aquaculture 2021, 545, 737217. [Google Scholar] [CrossRef]
  86. Assan, D.; Kuebutornye, F.K.A.; Hlordzi, V.; Chen, H.; Mraz, J.; Mustapha, U.F.; Abarike, E.D. Effects of Probiotics on Digestive Enzymes of Fish (Finfish and Shellfish); Status and Prospects: A Mini Review. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2022, 257, 110653. [Google Scholar] [CrossRef] [PubMed]
  87. Lara-Flores, M.; Olivera-Castillo, L.; Olvera-Novoa, M.A. Effect of the Inclusion of a Bacterial Mix (Streptococcus faecium and Lactobacillus acidophilus), and the Yeast (Saccharomyces cerevisiae) on Growth, Feed Utilization and Intestinal Enzymatic Activity of Nile Tilapia (Oreochromis niloticus). Int. J. Fish. Aquac. 2010, 2, 93–101. [Google Scholar]
  88. Carnevali, O.; Sun, Y.-Z.; Merrifield, D.L.; Zhou, Z.; Picchietti, S. Probiotic Applications in Temperate and Warm Water Fish Species. In Aquaculture Nutrition: Gut Health, Probiotics and Prebiotics; Wiley: New York, NY, USA, 2014; pp. 253–289. [Google Scholar] [CrossRef]
  89. Loh, J.Y.; Chan, H.K.; Yam, H.C.; In, L.L.A.; Lim, C.S.Y. An Overview of the Immunomodulatory Effects Exerted by Probiotics and Prebiotics in Grouper Fish. Aquac. Int. 2020, 28, 729–750. [Google Scholar] [CrossRef]
  90. Gao, X.-Y.; Liu, Y.; Miao, L.-L.; Li, E.-W.; Hou, T.-T.; Liu, Z.-P. Mechanism of Anti-Vibrio Activity of Marine Probiotic Strain Bacillus Pumilus H2, and Characterization of the Active Substance. AMB Express 2017, 7, 23. [Google Scholar] [CrossRef]
  91. Kuebutornye, F.K.; Abarike, E.D.; Lu, Y.; Hlordzi, V.; Sakyi, M.E.; Afriyie, G.; Wang, Z.; Li, Y.; Xie, C.X. Mechanisms and the Role of Probiotic Bacillus in Mitigating Fish Pathogens in Aquaculture. Fish Physiol. Biochem. 2020, 46, 819–841. [Google Scholar] [CrossRef]
  92. Emam, A.M.; Dunlap, C.A. Genomic and Phenotypic Characterization of Bacillus velezensis AMB-Y1; a Potential Probiotic to Control Pathogens in Aquaculture. Antonie Leeuwenhoek 2020, 113, 2041–2052. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, H.-M.; Rong, Y.-J.; Zhao, M.-X.; Song, B.; Chi, Z.-M. Antibacterial Activity of the Lipopetides Produced by Bacillus Amyloliquefaciens M1 against Multidrug-Resistant Vibrio spp. Isolated from Diseased Marine Animals. Appl. Microbiol. Biotechnol. 2014, 98, 127–136. [Google Scholar] [CrossRef]
  94. Chau, K.M.; Van, T.T.H.; Quyen, D.V.; Le, H.D.; Phan, T.H.T.; Ngo, N.D.T.; Vo, T.D.T.; Dinh, T.T.; Le, H.T.; Khanh, H.H.N. Molecular Identification and Characterization of Probiotic Bacillus Species with the Ability to Control Vibrio spp. in Wild Fish Intestines and Sponges from the Vietnam Sea. Microorganisms 2021, 9, 1927. [Google Scholar] [CrossRef] [PubMed]
  95. Yin, Z.; Liu, Q.; Liu, Y.; Gao, S.; He, Y.; Yao, C.; Huang, W.; Gong, Y.; Mai, K.; Ai, Q. Early Life Intervention Using Probiotic Clostridium butyricum Improves Intestinal Development, Immune Response, and Gut Microbiota in Large Yellow Croaker (Larimichthys crocea) Larvae. Front. Immunol. 2021, 12, 640767. [Google Scholar] [CrossRef] [PubMed]
  96. Li, C.; Zhang, B.; Liu, C.; Zhou, H.; Wang, X.; Mai, K.; He, G. Effects of Dietary Raw or Enterococcus faecium Fermented Soybean Meal on Growth, Antioxidant Status, Intestinal Microbiota, Morphology, and Inflammatory Responses in Turbot (Scophthalmus maximus L.). Fish Shellfish Immunol. 2020, 100, 261–271. [Google Scholar] [CrossRef]
  97. Kamei, Y.; Yoshimizu, M.; Ezura, Y.; Kimura, T. Screening of Bacteria with Antiviral Activity from Fresh Water Salmonid Hatcheries. Microbiol. Immunol. 1988, 32, 67–73. [Google Scholar] [CrossRef]
  98. Hasan, K.N.; Banerjee, G. Recent Studies on Probiotics as Beneficial Mediator in Aquaculture: A Review. J. Basic Appl. Zool. 2020, 81, 53. [Google Scholar] [CrossRef]
  99. Maeda, M.; Nogami, K.; Kanematsu, M.; Hirayama, K. The Concept of Biological Control Methods in Aquaculture. Hydrobiologia 1997, 358, 285–290. [Google Scholar] [CrossRef]
  100. Mondal, H.; Chandrasekaran, N.; Mukherjee, A.; Thomas, J. Viral Infections in Cultured Fish and Shrimps: Current Status and Treatment Methods. Aquac. Int. 2022, 30, 227–262. [Google Scholar] [CrossRef]
  101. De Andrade Belo, M.A.; Charlie-Silva, I. Teleost Fish as an Experimental Model for Vaccine Development. In Vaccine Design: Methods and Protocols, Volume 2. Vaccines for Veterinary Diseases; Springer: Berlin/Heidelberg, Germany, 2022; pp. 175–194. [Google Scholar] [CrossRef]
  102. Dawood, M.A.; Koshio, S. Recent Advances in the Role of Probiotics and Prebiotics in Carp Aquaculture: A Review. Aquaculture 2016, 454, 243–251. [Google Scholar] [CrossRef]
  103. Akbari, H.; Shekrabi, S.P.H.; Soltani, M.; Mehrgan, M.S. Effects of Potential Probiotic Enterococcus Casseliflavus (EC-001) on Growth Performance, Immunity, and Resistance to Aeromonas Hydrophila Infection in Common Carp (Cyprinus carpio). Probiotics Antimicrob. Proteins 2021, 13, 1316–1325. [Google Scholar] [CrossRef]
  104. Rachmawati, R.A.; Mulyani, Y.; Rochima, E.; Grandiosa, R. The Effect of Induction of Bacteria Bacillus Subtilis in Feed on the Immune System of Carp (Cyprinus carpio Linnaeus, 1758). World Sci. News 2021, 160, 203–216. [Google Scholar]
  105. Shah, S.; Chesti, A.; Rather, M.; Manzoor, S.; Malik, R.; Khan, J. Effect of Probiotic (Bacillus Subtilis) on the Immune System of Fingerlings of Grass Carp, Ctenopharyngodon idella. Pharma Innov. J. 2021, 10, 769–772. [Google Scholar]
  106. Yan, Y.-Y.; Xia, H.-Q.; Yang, H.-L.; Hoseinifar, S.H.; Sun, Y.-Z. Effects of Dietary Live or Heat-inactivated Autochthonous Bacillus pumilus SE 5 on Growth Performance, Immune Responses and Immune Gene Expression in Grouper Epinephelus coioides. Aquac. Nutr. 2016, 22, 698–707. [Google Scholar] [CrossRef]
  107. Liu, Z.-Y.; Yang, H.-L.; Hu, L.-H.; Yang, W.; Ai, C.-X.; Sun, Y.-Z. Dose-Dependent Effects of Histamine on Growth, Immunity and Intestinal Health in Juvenile Grouper (Epinephelus coioides). Front. Mar. Sci. 2021, 8, 685720. [Google Scholar] [CrossRef]
  108. Yang, H.-L.; Xia, H.-Q.; Ye, Y.-D.; Zou, W.-C.; Sun, Y.-Z. Probiotic Bacillus Pumilus SE5 Shapes the Intestinal Microbiota and Mucosal Immunity in Grouper Epinephelus coioides. Dis. Aquat. Organ. 2014, 111, 119–127. [Google Scholar] [CrossRef] [PubMed]
  109. Yang, H.-L.; Hu, X.; Ye, J.-D.; Seerengaraj, V.; Yang, W.; Ai, C.-X.; Sun, Y.-Z. Cell Wall Components of Bacillus Pumilus SE5 Improved the Growth, Digestive and Immunity of Grouper (Epinephelus coioides). Curr. Chin. Sci. 2021, 1, 231–239. [Google Scholar] [CrossRef]
  110. Boo, A.; Amaro, R.L.; Stan, G.-B. Quorum Sensing in Synthetic Biology: A Review. Curr. Opin. Syst. Biol. 2021, 28, 100378. [Google Scholar] [CrossRef]
  111. Saeki, E.K.; Kobayashi, R.K.T.; Nakazato, G. Quorum Sensing System: Target to Control the Spread of Bacterial Infections. Microb. Pathog. 2020, 142, 104068. [Google Scholar] [CrossRef]
  112. Defoirdt, T.; Boon, N.; Bossier, P.; Verstraete, W. Disruption of Bacterial Quorum Sensing: An Unexplored Strategy to Fight Infections in Aquaculture. Aquaculture 2004, 240, 69–88. [Google Scholar] [CrossRef]
  113. Alexpandi, R.; Abirami, G.; Satish, L.; Swasthikka, R.P.; Krishnaveni, N.; Jayakumar, R.; Pandian, S.K.; Ravi, A.V. Tocopherol and Phytol Possess Anti-Quorum Sensing Mediated Anti-Infective Behavior against Vibrio campbellii in Aquaculture: An in Vitro and in Vivo Study. Microb. Pathog. 2021, 161, 105221. [Google Scholar] [CrossRef]
  114. Samrot, A.V.; Abubakar Mohamed, A.; Faradjeva, E.; Si Jie, L.; Hooi Sze, C.; Arif, A.; Chuan Sean, T.; Norbert Michael, E.; Yeok Mun, C.; Xiao Qi, N. Mechanisms and Impact of Biofilms and Targeting of Biofilms Using Bioactive Compounds—A Review. Medicina 2021, 57, 839. [Google Scholar] [CrossRef]
  115. Hoseinifar, S.H.; Sun, Y.-Z.; Wang, A.; Zhou, Z. Probiotics as Means of Diseases Control in Aquaculture, a Review of Current Knowledge and Future Perspectives. Front. Microbiol. 2018, 9, 2429. [Google Scholar] [CrossRef] [PubMed]
  116. 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] [PubMed]
  117. Jiang, Y.; Zhou, S.; Sarkodie, E.K.; Chu, W. The Effects of Bacillus cereus QSI-1 on Intestinal Barrier Function and Mucosal Gene Transcription in Crucian Carp (Carassius auratus gibelio). Aquac. Rep. 2020, 17, 100356. [Google Scholar] [CrossRef]
  118. Chu, W.; Zhou, S.; Zhu, W.; Zhuang, X. Quorum Quenching Bacteria Bacillus Sp. QSI-1 Protect Zebrafish (Danio rerio) from Aeromonas hydrophila Infection. Sci. Rep. 2014, 4, 5446. [Google Scholar] [CrossRef] [PubMed]
  119. Shaheer, P.; Sreejith, V.N.; Joseph, T.C.; Murugadas, V.; Lalitha, K.V. Quorum Quenching Bacillus spp.: An Alternative Biocontrol Agent for Vibrio harveyi Infection in Aquaculture. Dis. Aquat. Organ. 2021, 146, 117–128. [Google Scholar] [CrossRef] [PubMed]
  120. Li, M.; Xi, B.; Qin, T.; Chen, K.; Xie, J. Isolation and Characterization of AHL-Degrading Bacteria from Fish and Pond Sediment. J. Oceanol. Limnol. 2019, 37, 1460–1467. [Google Scholar] [CrossRef]
  121. Mamun, M.A.A.; Nasren, S.; Abhiman, P.B.; Rathore, S.S.; Sowndarya, N.S.; Ramesh, K.S.; Shankar, K.M. Investigation of Production, Formation and Characterization of Biofilm Cells of Aeromonas hydrophila for Oral Vaccination of Fish. J. Exp. Zool. India 2019, 22, 1115–1123. [Google Scholar]
  122. Assan, D.; Huang, Y.; Mustapha, U.F.; Addah, M.N.; Li, G.; Chen, H. Fish Feed Intake, Feeding Behavior, and the Physiological Response of Apelin to Fasting and Refeeding. Front. Endocrinol. 2021, 12, 798903. [Google Scholar] [CrossRef]
  123. Mohapatra, S.; Chakraborty, T.; Kumar, V.; DeBoeck, G.; Mohanta, K.N. Aquaculture and Stress Management: A Review of Probiotic Intervention. J. Anim. Physiol. Anim. Nutr. 2013, 97, 405–430. [Google Scholar] [CrossRef]
  124. Hoseinifar, S.H.; Yousefi, S.; Van Doan, H.; Ashouri, G.; Gioacchini, G.; Maradonna, F.; Carnevali, O. Oxidative Stress and Antioxidant Defense in Fish: The Implications of Probiotic, Prebiotic, and Synbiotics. Rev. Fish. Sci. Aquac. 2020, 29, 198–217. [Google Scholar] [CrossRef]
  125. Vianello, S.; Brazzoduro, L.; Dalla Valle, L.; Belvedere, P.; Colombo, L. Myostatin Expression during Development and Chronic Stress in Zebrafish (Danio rerio). J. Endocrinol. 2003, 176, 47–60. [Google Scholar] [CrossRef] [PubMed]
  126. Lutfi, E.; Basili, D.; Falcinelli, S.; Morillas, L.; Carnevali, O.; Capilla, E.; Navarro, I. The Probiotic Lactobacillus Rhamnosus Mimics the Dark-Driven Regulation of Appetite Markers and Melatonin Receptors’ Expression in Zebrafish (Danio Rerio) Larvae: Understanding the Role of the Gut Microbiome. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2021, 256, 110634. [Google Scholar] [CrossRef]
  127. Carnevali, O.; de Vivo, L.; Sulpizio, R.; Gioacchini, G.; Olivotto, I.; Silvi, S.; Cresci, A. Growth Improvement by Probiotic in European Sea Bass Juveniles (Dicentrarchus labrax, L.), with Particular Attention to IGF-1, Myostatin and Cortisol Gene Expression. Aquaculture 2006, 258, 430–438. [Google Scholar] [CrossRef]
  128. Castex, M.; Lemaire, P.; Wabete, N.; Chim, L. Effect of Dietary Probiotic Pediococcus Acidilactici on Antioxidant Defences and Oxidative Stress Status of Shrimp Litopenaeus Stylirostris. Aquaculture 2009, 294, 306–313. [Google Scholar] [CrossRef]
  129. Chang, X.; Chen, Y.; Feng, J.; Huang, M.; Zhang, J. Amelioration of Cd-Induced Bioaccumulation, Oxidative Stress and Immune Damage by Probiotic Bacillus coagulans in Common Carp (Cyprinus carpio L.). Aquac. Rep. 2021, 20, 100678. [Google Scholar] [CrossRef]
  130. Merola, C.; Bisegna, A.; Angelozzi, G.; Conte, A.; Abete, M.C.; Stella, C.; Pederiva, S.; Faggio, C.; Riganelli, N.; Perugini, M. Study of Heavy Metals Pollution and Vitellogenin Levels in Brown Trout (Salmo trutta trutta) Wild Fish Populations. Appl. Sci. 2021, 11, 4965. [Google Scholar] [CrossRef]
  131. Jyoti, D.; Sinha, R.; Faggio, C. Advances in Biological Methods for the Sequestration of Heavy Metals from Water Bodies: A Review. Environ. Toxicol. Pharmacol. 2022, 94, 103927. [Google Scholar] [CrossRef]
  132. Shahjahan, M.; Taslima, K.; Rahman, M.S.; Al-Emran, M.; Alam, S.I.; Faggio, C. Effects of Heavy Metals on Fish Physiology–A Review. Chemosphere 2022, 300, 134519. [Google Scholar] [CrossRef]
  133. Zaynab, M.; Al-Yahyai, R.; Ameen, A.; Sharif, Y.; Ali, L.; Fatima, M.; Khan, K.A.; Li, S. Health and Environmental Effects of Heavy Metals. J. King Saud Univ.-Sci. 2022, 34, 101653. [Google Scholar] [CrossRef]
  134. Stefanescu, I.A. Bioaccumulation of Heavy Metals by Bacillus megaterium from Phosphogypsum Waste. Sci. Study Res. Chem. Chem. Eng. Biotechnol. Food Ind. 2015, 16, 93. [Google Scholar]
  135. Sonone, S.S.; Jadhav, S.; Sankhla, M.S.; Kumar, R. Water Contamination by Heavy Metals and Their Toxic Effect on Aquaculture and Human Health through Food Chain. Lett. Appl. NanoBioSci. 2020, 10, 2148–2166. [Google Scholar] [CrossRef]
  136. Fatima, S.; Muzammal, M.; Rehman, A.; Rustam, S.A.; Shehzadi, Z.; Mehmood, A.; Waqar, M. Water Pollution on Heavy Metals and Its Effects on Fishes. Int. J. Fish Aquat. Stud. 2020, 8, 6–14. [Google Scholar]
  137. Moiseenko, T.I.; Gashkina, N.A. Distribution and Bioaccumulation of Heavy Metals (Hg, Cd and Pb) in Fish: Influence of the Aquatic Environment and Climate. Environ. Res. Lett. 2020, 15, 115013. [Google Scholar] [CrossRef]
  138. Rahman, Z.; Singh, V.P. Bioremediation of Toxic Heavy Metals (THMs) Contaminated Sites: Concepts, Applications and Challenges. Environ. Sci. Pollut. Res. 2020, 27, 27563–27581. [Google Scholar] [CrossRef] [PubMed]
  139. Wróbel, M.; Śliwakowski, W.; Kowalczyk, P.; Kramkowski, K.; Dobrzyński, J. Bioremediation of Heavy Metals by the Genus Bacillus. Int. J. Environ. Res. Public. Health 2023, 20, 4964. [Google Scholar] [CrossRef]
  140. Elsanhoty, R.M.; Al-Turki, I.A.; Ramadan, M.F. Application of Lactic Acid Bacteria in Removing Heavy Metals and Aflatoxin B1 from Contaminated Water. Water Sci. Technol. 2016, 74, 625–638. [Google Scholar] [CrossRef]
  141. Wang, Y.; Han, J.; Ren, Q.; Liu, Z.; Zhang, X.; Wu, Z. The Involvement of Lactic Acid Bacteria and Their Exopolysaccharides in the Biosorption and Detoxication of Heavy Metals in the Gut. Biol. Trace Elem. Res. 2023. [Google Scholar] [CrossRef]
  142. Murthy, S.; Bali, G.; Sarangi, S.K. Effect of Lead on Metallothionein Concentration in Leadresistant Bacteria Bacillus cereus Isolated from Industrial Effluent. Afr. J. Biotechnol. 2011, 10, 15966–15972. [Google Scholar] [CrossRef]
  143. Gomaa, E.Z. Biosequestration of Heavy Metals by Microbially Induced Calcite Precipitation of Ureolytic Bacteria. Rom. Biotechnol. Lett. 2018, 24, 147–153. [Google Scholar] [CrossRef]
  144. Fakhar, A.; Gul, B.; Gurmani, A.R.; Khan, S.M.; Ali, S.; Sultan, T.; Chaudhary, H.J.; Rafique, M.; Rizwan, M. Heavy Metal Remediation and Resistance Mechanism of Aeromonas, Bacillus, and Pseudomonas: A Review. Crit. Rev. Environ. Sci. Technol. 2022, 52, 1868–1914. [Google Scholar] [CrossRef]
  145. Rekadwad, B. Microbial Systematics; CRC Press: Boca Raton, FL, USA, 2021. [Google Scholar]
  146. Ringø, E.; Van Doan, H.; Lee, S.H.; Soltani, M.; Hoseinifar, S.H.; Harikrishnan, R.; Song, S.K. Probiotics, Lactic Acid Bacteria and Bacilli: Interesting Supplementation for Aquaculture. J. Appl. Microbiol. 2020, 129, 116–136. [Google Scholar] [CrossRef]
  147. Valipour, A.; Nedaei, S.; Noori, A.; Khanipour, A.A.; Hoseinifar, S.H. Dietary Lactobacillus Plantarum Affected on Some Immune Parameters, Air-Exposure Stress Response, Intestinal Microbiota, Digestive Enzyme Activity and Performance of Narrow Clawed Crayfish (Astacus leptodactylus, Eschscholtz). Aquaculture 2019, 504, 121–130. [Google Scholar] [CrossRef]
  148. Yi, Y.; Zhang, Z.; Zhao, F.; Liu, H.; Yu, L.; Zha, J.; Wang, G. Probiotic Potential of Bacillus velezensis JW: Antimicrobial Activity against Fish Pathogenic Bacteria and Immune Enhancement Effects on Carassius auratus. Fish Shellfish Immunol. 2018, 78, 322–330. [Google Scholar] [CrossRef] [PubMed]
  149. Huang, J.-B.; Wu, Y.-C.; Chi, S.-C. Dietary Supplementation of Pediococcus Pentosaceus Enhances Innate Immunity, Physiological Health and Resistance to Vibrio anguillarum in Orange-Spotted Grouper (Epinephelus coioides). Fish Shellfish Immunol. 2014, 39, 196–205. [Google Scholar] [CrossRef] [PubMed]
  150. Dejene, F.; Regasa Dadi, B.; Tadesse, D. In Vitro Antagonistic Effect of Lactic Acid Bacteria Isolated from Fermented Beverage and Finfish on Pathogenic and Foodborne Pathogenic Microorganism in Ethiopia. Int. J. Microbiol. 2021, 2021, 5370556. [Google Scholar] [CrossRef]
  151. Huy, N.D.; Ngoc, L.M.T.; Loc, N.H.; Lan, T.T.; Quang, H.T.; Dung, T. Isolation of Weissella Cibaria from Pacific White Shrimp (Litopenaeus vannamei) Gastrointestinal Tract and Evaluation of Its Pathogenic Bacterial Inhibition. Indian J. Sci. Technol. 2020, 13, 1200–1212. [Google Scholar] [CrossRef]
  152. Puvanasundram, P.; Chong, C.M.; Sabri, S.; Yusoff, M.S.; Karim, M. Multi-Strain Probiotics: Functions, Effectiveness and Formulations for Aquaculture Applications. Aquac. Rep. 2021, 21, 100905. [Google Scholar] [CrossRef]
  153. Midhun, S.J.; Neethu, S.; Arun, D.; Vysakh, A.; Divya, L.; Radhakrishnan, E.K.; Jyothis, M. Dietary Supplementation of Bacillus Licheniformis HGA8B Improves Growth Parameters, Enzymatic Profile and Gene Expression of Oreochromis niloticus. Aquaculture 2019, 505, 289–296. [Google Scholar] [CrossRef]
  154. Yang, G.; Tian, X.; Dong, S.; Peng, M.; Wang, D. Effects of Dietary Bacillus Cereus G19, B. Cereus BC-01, and Paracoccus marcusii DB11 Supplementation on the Growth, Immune Response, and Expression of Immune-Related Genes in Coelomocytes and Intestine of the Sea Cucumber (Apostichopus japonicus selenka). Fish Shellfish Immunol. 2015, 45, 800–807. [Google Scholar] [CrossRef] [PubMed]
  155. Zhao, Y.; Yuan, L.; Wan, J.; Sun, Z.; Wang, Y.; Sun, H. Effects of Potential Probiotic Bacillus Cereus EN25 on Growth, Immunity and Disease Resistance of Juvenile Sea Cucumber Apostichopus Japonicus. Fish Shellfish Immunol. 2016, 49, 237–242. [Google Scholar] [CrossRef]
  156. Zhang, J.-J.; Yang, H.-L.; Yan, Y.-Y.; Zhang, C.-X.; Ye, J.; Sun, Y.-Z. Effects of Fish Origin Probiotics on Growth Performance, Immune Response and Intestinal Health of Shrimp (Litopenaeus vannamei) Fed Diets with Fish Meal Partially Replaced by Soybean Meal. Aquac. Nutr. 2020, 26, 1255–1265. [Google Scholar] [CrossRef]
  157. Newaj-Fyzul, A.; Adesiyun, A.A.; Mutani, A.; Ramsubhag, A.; Brunt, J.; Austin, B. Bacillus Subtilis AB1 Controls Aeromonas Infection in Rainbow Trout (Oncorhynchus mykiss, Walbaum). J. Appl. Microbiol. 2007, 103, 1699–1706. [Google Scholar] [CrossRef] [PubMed]
  158. Sahandi, J.; Jafaryan, H.; Soltani, M.; Ebrahimi, P. The Use of Two Bifidobacterium Strains Enhanced Growth Performance and Nutrient Utilization of Rainbow Trout (Oncorhynchus mykiss) Fry. Probiotics Antimicrob. Proteins 2019, 11, 966–972. [Google Scholar] [CrossRef] [PubMed]
  159. Ringø, E.; Seppola, M.; Berg, A.; Olsen, R.E.; Schillinger, U.; Holzapfel, W. Characterization of Carnobacterium divergens Strain 6251 Isolated from Intestine of Arctic Charr (Salvelinus alpinus L.). Syst. Appl. Microbiol. 2002, 25, 120–129. [Google Scholar] [CrossRef]
  160. Ringø, E. The Ability of Carnobacteria Isolated from Fish Intestine to Inhibit Growth of Fish Pathogenic Bacteria: A Screening Study. Aquac. Res. 2008, 39, 171–180. [Google Scholar] [CrossRef]
  161. Vendrell, D.; Balcazar, J.L.; de Blas, I.; Ruiz-Zarzuela, I.; Gironés, O.; Muzquiz, J.L. Protection of Rainbow Trout (Oncorhynchus mykiss) from Lactococcosis by Probiotic Bacteria. Comp. Immunol. Microbiol. Infect. Dis. 2008, 31, 337–345. [Google Scholar] [CrossRef]
  162. Al-Dohail, M.A.; Hashim, R.; Aliyu-Paiko, M. Evaluating the Use of Lactobacillus acidophilus as a Biocontrol Agent against Common Pathogenic Bacteria and the Effects on the Haematology Parameters and Histopathology in African Catfish Clarias gariepinus Juveniles: L. Acidophilus as a Probiotic in African Catfish. Aquac. Res. 2011, 42, 196–209. [Google Scholar] [CrossRef]
  163. Zheng, C.N.; Wang, W. Effects of Lactobacillus Pentosus on the Growth Performance, Digestive Enzyme and Disease Resistance of White Shrimp, Litopenaeus vannamei (Boone, 1931). Aquac. Res. 2017, 48, 2767–2777. [Google Scholar] [CrossRef]
  164. Lee, S.H.; Beck, B.R.; Hwang, S.-H.; Song, S.K. Feeding Olive Flounder (Paralichthys olivaceus) with Lactococcus lactis BFE920 Expressing the Fusion Antigen of Vibrio OmpK and FlaB Provides Protection against Multiple Vibrio Pathogens: A Universal Vaccine Effect. Fish Shellfish Immunol. 2021, 114, 253–262. [Google Scholar] [CrossRef] [PubMed]
  165. Balcázar, J.L.; Vendrell, D.; De Blas, I.; Ruiz-Zarzuela, I.; Múzquiz, J.L. Effect of Lactococcus lactis CLFP 100 and Leuconostoc mesenteroides CLFP 196 on Aeromonas salmonicida Infection in Brown Trout (Salmo trutta). Microb. Physiol. 2009, 17, 153–157. [Google Scholar] [CrossRef] [PubMed]
  166. Thao, T.T.P.; Lan, T.T.P.; Phuong, T.V.; Truong, H.T.H.; Khoo, K.S.; Manickam, S.; Hoa, T.T.; Tram, N.D.Q.; Show, P.L.; Huy, N.D. Characterization Halotolerant Lactic Acid Bacteria Pediococcus pentosaceus HN10 and in Vivo Evaluation for Bacterial Pathogens Inhibition. Chem. Eng. Process.-Process Intensif. 2021, 168, 108576. [Google Scholar] [CrossRef]
  167. Tarkhani, R.; Imani, A.; Hoseinifar, S.H.; Moghanlou, K.S.; Manaffar, R. The Effects of Host-Associated Enterococcus faecium CGMCC1. 2136 on Serum Immune Parameters, Digestive Enzymes Activity and Growth Performance of the Caspian Roach (Rutilus rutilus caspicus) Fingerlings. Aquaculture 2020, 519, 734741. [Google Scholar] [CrossRef]
  168. Safari, R.; Adel, M.; Lazado, C.C.; Caipang, C.M.A.; Dadar, M. Host-Derived Probiotics Enterococcus casseliflavus Improves Resistance against Streptococcus Iniae Infection in Rainbow Trout (Oncorhynchus mykiss) via Immunomodulation. Fish Shellfish Immunol. 2016, 52, 198–205. [Google Scholar] [CrossRef]
  169. Ali, M.; Soltanian, S.; Mirghaed, A.T.; Akhlaghi, M.; Hoseinifar, S.H.; Esmailnejad, A. The Effect of Oral Administration of Lactic Acid Bacteria Isolated from Kefir on Intestinal Microbiota, Growth Performance and Survival in Juvenile Rainbow Trout, Oncorhynchus mykiss. Int. J. Aquat. Biol. 2020, 8, 35–49. [Google Scholar] [CrossRef]
  170. Sakai, M.; Yoshida, T.; Atsuta, S.; Kobayashi, M. Enhancement of Resistance to Vibriosis in Rainbow Trout, Oncorhynchus mykiss (Walbaum), by Oral Administration of Clostridium butyricum Bacterin. J. Fish Dis. 1995, 18, 187–190. [Google Scholar] [CrossRef]
  171. Meng, X.; Wu, S.; Hu, W.; Zhu, Z.; Yang, G.; Zhang, Y.; Qin, C.; Yang, L.; Nie, G. Clostridium butyricum Improves Immune Responses and Remodels the Intestinal Microbiota of Common Carp (Cyprinus carpio L.). Aquaculture 2021, 530, 735753. [Google Scholar] [CrossRef]
  172. Kahyani, F.; Pirali-Kheirabadi, E.; Shafiei, S.; Shenavar Masouleh, A. Effect of Dietary Supplementation of Potential Probiotic Weissella Confusa on Innate Immunity, Immune-related Genes Expression, Intestinal Microbiota and Growth Performance of Rainbow Trout (Oncorhynchus mykiss). Aquac. Nutr. 2021, 27, 1411–1420. [Google Scholar] [CrossRef]
  173. Rengpipat, S.; Rueangruklikhit, T.; Piyatiratitivorakul, S. Evaluations of Lactic Acid Bacteria as Probiotics for Juvenile Seabass Lates calcarifer. Aquac. Res. 2008, 39, 134–143. [Google Scholar] [CrossRef]
  174. Jinendiran, S.; Archana, R.; Sathishkumar, R.; Kannan, R.; Selvakumar, G.; Sivakumar, N. Dietary Administration of Probiotic Aeromonas veronii V03 on the Modulation of Innate Immunity, Expression of Immune-Related Genes and Disease Resistance against Aeromonas Hydrophila Infection in Common Carp (Cyprinus carpio). Probiotics Antimicrob. Proteins 2021, 13, 1709–1722. [Google Scholar] [CrossRef]
  175. Abd El-Rhman, A.M.; Khattab, Y.A.E.; Shalaby, A.M.E. Micrococcus Luteus and Pseudomonas Species as Probiotics for Promoting the Growth Performance and Health of Nile Tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2009, 27, 175–180. [Google Scholar] [CrossRef]
  176. ABdel-Tawwab, M.; Mousa, M.A.A.; Mohammed, M.A. Use of Live Baker’s Yeast, Saccharomyces Cerevisiae, in Practical Diet to Enhance the Growth Performance of Galilee Tilapia, Sarotherodon galilaeus (L.), and Its Resistance to Environmental Copper Toxicity. J. World Aquac. Soc. 2010, 41, 214–223. [Google Scholar] [CrossRef]
  177. Banu, M.R.; Akter, S.; Islam, M.R.; Mondol, M.N.; Hossain, M.A. Probiotic Yeast Enhanced Growth Performance and Disease Resistance in Freshwater Catfish Gulsa Tengra, Mystus Cavasius. Aquac. Rep. 2020, 16, 100237. [Google Scholar] [CrossRef]
  178. Reyes-Becerril, M.; Alamillo, E.; Angulo, C. Probiotic and Immunomodulatory Activity of Marine Yeast Yarrowia lipolytica Strains and Response against Vibrio parahaemolyticus in Fish. Probiotics Antimicrob. Proteins 2021, 13, 1292–1305. [Google Scholar] [CrossRef]
  179. Raida, M.K.; Larsen, J.L.; Nielsen, M.E.; Buchmann, K. Enhanced Resistance of Rainbow Trout, Oncorhynchus mykiss (Walbaum), against Yersinia Ruckeri Challenge Following Oral Administration of Bacillus subtilis and B. licheniformis (BioPlus2B). J. Fish Dis. 2003, 26, 495–498. [Google Scholar] [CrossRef] [PubMed]
  180. Sayed Hassani, M.H.; Jourdehi, A.Y.; Zelti, A.H.; Masouleh, A.S.; Lakani, F.B. Effects of Commercial Superzist Probiotic on Growth Performance and Hematological and Immune Indices in Fingerlings Acipenser baerii. Aquac. Int. 2020, 28, 377–387. [Google Scholar] [CrossRef]
  181. Zhang, M.; Pan, L.; Fan, D.; He, J.; Su, C.; Gao, S.; Zhang, M. Study of Fermented Feed by Mixed Strains and Their Effects on the Survival, Growth, Digestive Enzyme Activity and Intestinal Flora of Penaeus vannamei. Aquaculture 2021, 530, 735703. [Google Scholar] [CrossRef]
  182. Joseph, T.C.; Kumar, A.; Basha, K.A. Prophylactic Health Products in Aquaculture; ICAR-Central Institute of Fisheries Technology: Kerala, India, 2017. [Google Scholar]
  183. Lordan, C.; Thapa, D.; Ross, R.P.; Cotter, P.D. Potential for Enriching Next-Generation Health-Promoting Gut Bacteria through Prebiotics and Other Dietary Components. Gut Microbes 2020, 11, 1–20. [Google Scholar] [CrossRef]
  184. Peredo-Lovillo, A.; Romero-Luna, H.E.; Jiménez-Fernández, M. Health Promoting Microbial Metabolites Produced by Gut Microbiota after Prebiotics Metabolism. Food Res. Int. 2020, 136, 109473. [Google Scholar] [CrossRef]
  185. Wu, Y.; Chen, Y.; Lu, Y.; Hao, H.; Liu, J.; Huang, R. Structural Features, Interaction with the Gut Microbiota and Anti-Tumor Activity of Oligosaccharides. RSC Adv. 2020, 10, 16339–16348. [Google Scholar] [CrossRef] [PubMed]
  186. Elumalai, P.; Kurian, A.; Lakshmi, S.; Musthafa, M.S.; Ringo, E.; Faggio, C. Effect of Leucas Aspera against Aeromonas Hydrophila in Nile Tilapia (Oreochromis niloticus): Immunity and Gene Expression Evaluation. Turk. J. Fish. Aquat. Sci. 2021, 22, TRJFAS19802. [Google Scholar] [CrossRef]
  187. Kumar, J.; Priyadharshini, M.; Madhavi, M.; Begum, S.S.; Ali, A.J.; Musthafa, M.S.; Faggio, C. Impact of Hygrophila Auriculata Supplementary Diets on the Growth, Survival, Biochemical and Haematological Parameters in Fingerlings of Freshwater Fish Cirrhinus mrigala (Hamilton, 1822). Comp. Biochem. Physiol. A. Mol. Integr. Physiol. 2022, 263, 111097. [Google Scholar] [CrossRef] [PubMed]
  188. Aliab, S.S.R.; Ambasankar, K.; Praveena, P.E. Effect of Dietary Mannanoligosaccharide on Intestinal Microbiota and Immune Parameters of Asian Seabass (Lates calcarifer) Juveniles. J. Fish Res. 2021, 5, 16–21. [Google Scholar]
  189. Asaduzzaman, M.D.; Iehata, S.; Akter, S.; Kader, M.A.; Ghosh, S.K.; Khan, M.N.A.; Abol-Munafi, A.B. Effects of Host Gut-Derived Probiotic Bacteria on Gut Morphology, Microbiota Composition and Volatile Short Chain Fatty Acids Production of Malaysian Mahseer Tor Tambroides. Aquac. Rep. 2018, 9, 53–61. [Google Scholar] [CrossRef]
  190. Hu, J.; Zhang, J.; Wu, S. The Growth Performance and Non-Specific Immunity of Juvenile Grass Carp (Ctenopharyngodon idella) Affected by Dietary Alginate Oligosaccharide. 3 Biotech 2021, 11, 46. [Google Scholar] [CrossRef]
  191. Liu, Y.; Miao, Y.; Xu, N.; Ding, T.; Cui, K.; Chen, Q.; Zhang, J.; Fang, W.; Mai, K.; Ai, Q. Effects of Dietary Astragalus Polysaccharides (APS) on Survival, Growth Performance, Activities of Digestive Enzyme, Antioxidant Responses and Intestinal Development of Large Yellow Croaker (Larimichthys crocea) Larvae. Aquaculture 2020, 517, 734752. [Google Scholar] [CrossRef]
  192. Hamidoghli, A.; Won, S.; Lee, S.; Lee, S.; Farris, N.W.; Bai, S.C. Nutrition and Feeding of Olive Flounder Paralichthys olivaceus: A Review. Rev. Fish. Sci. Aquac. 2020, 28, 340–357. [Google Scholar] [CrossRef]
  193. Kazuń, B.; Małaczewska, J.; Kazuń, K.; Kamiński, R.; Adamek-Urbańska, D.; Żylińska-Urban, J. Dietary Administration of β-1, 3/1, 6-Glucan and Lactobacillus Plantarum Improves Innate Immune Response and Increases the Number of Intestine Immune Cells in Roach (Rutilus rutilus). BMC Vet. Res. 2020, 16, 216. [Google Scholar] [CrossRef]
  194. Deon, M.P.P.; Bicudo, Á.J.A.; Sado, R.Y. Performance, Hematology, and Immunology of Pacu in Response to Dietary Supplementation with Fructooligosaccharides. Pesqui. Agropecuária Bras. 2021, 56, e02460. [Google Scholar] [CrossRef]
  195. Yousefi, S.; Shokri, M.M.; Noveirian, H.A.; Hoseinifar, S.H. Effects of Dietary Yeast Cell Wall on Biochemical Indices, Serum and Skin Mucus Immune Responses, Oxidative Status and Resistance against Aeromonas hydrophila in Juvenile Persian Sturgeon (Acipenser persicus). Fish Shellfish Immunol. 2020, 106, 464–472. [Google Scholar] [CrossRef] [PubMed]
  196. Serradell, A.; Torrecillas, S.; Makol, A.; Valdenegro, V.; Fernández-Montero, A.; Acosta, F.; Izquierdo, M.S.; Montero, D. Prebiotics and Phytogenics Functional Additives in Low Fish Meal and Fish Oil Based Diets for European Sea Bass (Dicentrarchus labrax): Effects on Stress and Immune Responses. Fish Shellfish Immunol. 2020, 100, 219–229. [Google Scholar] [CrossRef] [PubMed]
  197. Priya, P.S.; Ashwitha, A.; Thamizharasan, K.; Harishkumar, M.; Dinesh, S.; Nithya, T.G.; Kamaraj, M. Synergistic Effect of Durian Fruit Rind Polysaccharide Gel Encapsulated Prebiotic and Probiotic Dietary Supplements on Growth Performance, Immune-Related Gene Expression, and Disease Resistance in Zebrafish (Danio rerio). Heliyon 2021, 7, e06669. [Google Scholar] [CrossRef] [PubMed]
  198. Mohammadian, T.; Ghanei-Motlagh, R.; Molayemraftar, T.; Mesbah, M.; Zarea, M.; Mohtashamipour, H.; Nejad, A.J. Modulation of Growth Performance, Gut Microflora, Non-Specific Immunity and Gene Expression of Proinflammatory Cytokines in Shabout (Tor grypus) upon Dietary Prebiotic Supplementation. Fish Shellfish Immunol. 2021, 112, 38–45. [Google Scholar] [CrossRef] [PubMed]
  199. Van Doan, H.; Hoseinifar, S.H.; Faggio, C.; Chitmanat, C.; Mai, N.T.; Jaturasitha, S.; Ringø, E. Effects of Corncob Derived Xylooligosaccharide on Innate Immune Response, Disease Resistance, and Growth Performance in Nile Tilapia (Oreochromis niloticus) Fingerlings. Aquaculture 2018, 495, 786–793. [Google Scholar] [CrossRef]
  200. Włodarczyk, M.; Śliżewska, K. Efficiency of Resistant Starch and Dextrins as Prebiotics: A Review of the Existing Evidence and Clinical Trials. Nutrients 2021, 13, 3808. [Google Scholar] [CrossRef] [PubMed]
  201. Vázquez, J.A.; González, M.; Murado, M.A. Effects of Lactic Acid Bacteria Cultures on Pathogenic Microbiota from Fish. Aquaculture 2005, 245, 149–161. [Google Scholar] [CrossRef]
  202. Poolsawat, L.; Li, X.; Xu, X.; Rahman, M.M.; Boonpeng, N.; Leng, X. Dietary Xylooligosaccharide Improved Growth, Nutrient Utilization, Gut Microbiota and Disease Resistance of Tilapia (Oreochromis niloticus x O. Aureus). Anim. Feed Sci. Technol. 2021, 275, 114872. [Google Scholar] [CrossRef]
  203. Maas, R.M.; Deng, Y.; Dersjant-Li, Y.; Petit, J.; Verdegem, M.C.; Schrama, J.W.; Kokou, F. Exogenous Enzymes and Probiotics Alter Digestion Kinetics, Volatile Fatty Acid Content and Microbial Interactions in the Gut of Nile Tilapia. Sci. Rep. 2021, 11, 8221. [Google Scholar] [CrossRef]
  204. Wang, M.; Zhou, J.; Selma-Royo, M.; Simal-Gandara, J.; Collado, M.C.; Barba, F.J. Potential Benefits of High-Added-Value Compounds from Aquaculture and Fish Side Streams on Human Gut Microbiota. Trends Food Sci. Technol. 2021, 112, 484–494. [Google Scholar] [CrossRef]
  205. Maas, R.M.; Verdegem, M.C.; Wiegertjes, G.F.; Schrama, J.W. Carbohydrate Utilisation by Tilapia: A Meta-analytical Approach. Rev. Aquac. 2020, 12, 1851–1866. [Google Scholar] [CrossRef]
  206. Wee, W.; Hamid, N.K.A.; 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. 2022, 9, 28–34. [Google Scholar] [CrossRef]
  207. Hu, X.; Yang, H.-L.; Yan, Y.-Y.; Zhang, C.-X.; Ye, J.; Lu, K.-L.; Hu, L.-H.; Zhang, J.-J.; Ruan, L.; Sun, Y.-Z. Effects of Fructooligosaccharide on Growth, Immunity and Intestinal Microbiota of Shrimp (Litopenaeus vannamei) Fed Diets with Fish Meal Partially Replaced by Soybean Meal. Aquac. Nutr. 2019, 25, 194–204. [Google Scholar] [CrossRef]
  208. Mustafa, A.; Buentello, A.; Gatlin, D., III; Lightner, D.; Hume, M.; Lawrence, A. Effects of Fructooligosaccharides (FOS) on Growth, Survival, Gut Microflora, Stress, and Immune Response in Pacific White Shrimp, Litopenaeus vannamei, Cultured in a Recirculating System. J. Immunoass. Immunochem. 2020, 41, 45–59. [Google Scholar] [CrossRef]
  209. Zhu, W.; Su, J. Immune Functions of Phagocytic Blood Cells in Teleost. Rev. Aquac. 2022, 14, 630–646. [Google Scholar] [CrossRef]
  210. Tran, N.T.; Li, S. Potential Role of Prebiotics and Probiotics in Conferring Health Benefits in Economically Important Crabs. Fish Shellfish Immunol. Rep. 2022, 3, 100041. [Google Scholar] [CrossRef] [PubMed]
  211. Uribe-Querol, E.; Rosales, C. Phagocytosis: Our Current Understanding of a Universal Biological Process. Front. Immunol. 2020, 11, 1066. [Google Scholar] [CrossRef]
  212. Zhao, Y.; Mai, K.; Xu, W.; Zhang, W.; Ai, Q.; Zhang, Y.; Wang, X.; Liufu, Z. Influence of Dietary Probiotic Bacillus TC22 and Prebiotic Fructooligosaccharide on Growth, Immune Responses and Disease Resistance against Vibrio Splendidus Infection in Sea Cucumber Apostichopus japonicus. J. Ocean Univ. China 2011, 10, 293–300. [Google Scholar] [CrossRef]
  213. Park, Y.; Zhang, Q.; Wiegertjes, G.F.; Fernandes, J.M.; Kiron, V. Adherent Intestinal Cells from Atlantic Salmon Show Phagocytic Ability and Express Macrophage-Specific Genes. Front. Cell Dev. Biol. 2020, 8, 580848. [Google Scholar] [CrossRef]
  214. Picchietti, S.; Miccoli, A.; Fausto, A.M. Gut Immunity in European Sea Bass (Dicentrarchus labrax): A Review. Fish Shellfish Immunol. 2021, 108, 94–108. [Google Scholar] [CrossRef]
  215. Maldonado, E.; Rojas, D.A.; Morales, S.; Miralles, V.; Solari, A. Dual and Opposite Roles of Reactive Oxygen Species (ROS) in Chagas Disease: Beneficial on the Pathogen and Harmful on the Host. Oxid. Med. Cell. Longev. 2020, 2020, 8867701. [Google Scholar] [CrossRef] [PubMed]
  216. Merrifield, D.L.; Ringo, E. Aquaculture Nutrition: Gut Health, Probiotics and Prebiotics; John Wiley & Sons: Hoboken, NJ, USA, 2014; ISBN 0-470-67271-4. [Google Scholar]
  217. Ghafarifarsani, H.; Rashidian, G.; Bagheri, T.; Hoseinifar, S.H.; Van Doan, H. Study on Growth Enhancement and the Protective Effects of Dietary Prebiotic Inulin on Immunity Responses of Rainbow Trout (Oncorhynchus mykiss) Fry Infected with Aeromonas Hydrophila. Ann. Anim. Sci. 2021, 21, 543–559. [Google Scholar] [CrossRef]
  218. Dong, C.; Wang, J. Immunostimulatory Effects of Dietary Fructooligosaccharides on Red Swamp Crayfish, Procambarus clarkii (Girard). Aquac. Res. 2013, 44, 1416–1424. [Google Scholar] [CrossRef]
  219. Khanjani, M.H.; Sharifinia, M.; Ghaedi, G. β-Glucan as a Promising Food Additive and Immunostimulant in Aquaculture Industry. Ann. Anim. Sci. 2022, 22, 817–827. [Google Scholar] [CrossRef]
  220. Raa, J. The Use of Immune-Stimulants in Fish and Shellfish Feeds. In Avances en Nutrición Acuícola V. Memorias del V Simposium Internacional de Nutrición Acuícola. 19–22 Noviembre, 2000; Cruz -Suárez, L.E., Ricque-Marie, D., Tapia-Salazar, M., Olvera-Novoa, M.A., y Civera-Cerecedo, R., Eds.; Mérida: Yucatán, Mexico, 2000. [Google Scholar]
  221. Song, H.; Zhang, S.; Yang, B.; Liu, Y.; Kang, Y.; Li, Y.; Qian, A.; Yuan, Z.; Cong, B.; Shan, X. Effects of Four Different Adjuvants Separately Combined with Aeromonas veronii Inactivated Vaccine on Haematoimmunological State, Enzymatic Activity, Inflammatory Response and Disease Resistance in Crucian Carp. Fish Shellfish Immunol. 2022, 120, 658–673. [Google Scholar] [CrossRef]
  222. Geraylou, Z.; Souffreau, C.; Rurangwa, E.; De Meester, L.; Courtin, C.M.; Delcour, J.A.; Buyse, J.; Ollevier, F. Effects of Dietary Arabinoxylan-Oligosaccharides (AXOS) and Endogenous Probiotics on the Growth Performance, Non-Specific Immunity and Gut Microbiota of Juvenile Siberian Sturgeon (Acipenser bábaerii). Fish Shellfish Immunol. 2013, 35, 766–775. [Google Scholar] [CrossRef]
  223. Khanjani, M.H.; Ghaedi, G.; Sharifinia, M. Effects of Diets Containing Β-glucan on Survival, Growth Performance, Haematological, Immunity and Biochemical Parameters of Rainbow Trout (Oncorhynchus mykiss) Fingerlings. Aquac. Res. 2022, 53, 1842–1850. [Google Scholar] [CrossRef]
  224. Marcos-López, M.; Rodger, H.D. Amoebic Gill Disease and Host Response in Atlantic Salmon (Salmo salar L.): A Review. Parasite Immunol. 2020, 42, e12766. [Google Scholar] [CrossRef]
  225. Reis, B.; Gonçalves, A.T.; Santos, P.; Sardinha, M.; Conceição, L.E.; Serradeiro, R.; Pérez-Sánchez, J.; Calduch-Giner, J.; Schmid-Staiger, U.; Frick, K. Immune Status and Hepatic Antioxidant Capacity of Gilthead Seabream Sparus aurata Juveniles Fed Yeast and Microalga Derived β-Glucans. Mar. Drugs 2021, 19, 653. [Google Scholar] [CrossRef]
  226. Pogue, R.; Murphy, E.J.; Fehrenbach, G.W.; Rezoagli, E.; Rowan, N.J. Exploiting Immunomodulatory Properties of β-Glucans Derived from Natural Products for Improving Health and Sustainability in Aquaculture-Farmed Organisms: Concise Review of Existing Knowledge, Innovation and Future Opportunities. Curr. Opin. Environ. Sci. Health 2021, 21, 100248. [Google Scholar] [CrossRef]
  227. Mameloco, E.J.; Traifalgar, R.F. Supplementation of Combined Mannan Oligosaccharide and β-Glucan Immunostimulants Improves Immunological Responses and Enhances Resistance of Pacific Whiteleg Shrimp, Penaeus vannamei, against Vibrio parahaemolyticus Infection. Int. Aquat. Res. 2020, 12, 291. [Google Scholar] [CrossRef]
  228. Cornet, V.; Khuyen, T.D.; Mandiki, S.N.; Betoulle, S.; Bossier, P.; Reyes-López, F.E.; Tort, L.; Kestemont, P. GAS1: A New β-Glucan Immunostimulant Candidate to Increase Rainbow Trout (Oncorhynchus mykiss) Resistance to Bacterial Infections with Aeromonas salmonicida achromogenes. Front. Immunol. 2021, 12, 693613. [Google Scholar] [CrossRef] [PubMed]
  229. Hansen, J.Ø.; Lagos, L.; Lei, P.; Reveco-Urzua, F.E.; Morales-Lange, B.; Hansen, L.D.; Schiavone, M.; Mydland, L.T.; Arntzen, M.Ø.; Mercado, L. Down-Stream Processing of Baker’s Yeast (Saccharomyces cerevisiae)–Effect on Nutrient Digestibility and Immune Response in Atlantic Salmon (Salmo salar). Aquaculture 2021, 530, 735707. [Google Scholar] [CrossRef]
  230. El-Nobi, G.; Hassanin, M.; Khalil, A.A.; Mohammed, A.Y.; Amer, S.A.; Montaser, M.M.; El-Sharnouby, M.E. Synbiotic Effects of Saccharomyces cerevisiae, Mannan oligosaccharides, and β-Glucan on Innate Immunity, Antioxidant Status, and Disease Resistance of Nile Tilapia, Oreochromis niloticus. Antibiotics 2021, 10, 567. [Google Scholar] [CrossRef] [PubMed]
  231. Ebrahimi, G.; Ouraji, H.; Khalesi, M.K.; Sudagar, M.; Barari, A.; Zarei Dangesaraki, M.; Jani Khalili, K.H. Effects of a Prebiotic, Immunogen®, on Feed Utilization, Body Composition, Immunity and Resistance to Aeromonas Hydrophila Infection in the Common Carp Cyprinus carpio (Linnaeus) Fingerlings. J. Anim. Physiol. Anim. Nutr. 2012, 96, 591–599. [Google Scholar] [CrossRef] [PubMed]
  232. Hoseinifar, S.H.; Mirvaghefi, A.; Mojazi Amiri, B.; Rostami, H.K.; Merrifield, D.L. The Effects of Oligofructose on Growth Performance, Survival and Autochthonous Intestinal Microbiota of Beluga (Huso huso) Juveniles. Aquac. Nutr. 2011, 17, 498–504. [Google Scholar] [CrossRef]
  233. Ziółkowska, E.; Bogucka, J.; Dankowiakowska, A.; Rawski, M.; Mazurkiewicz, J.; Stanek, M. Effects of a Trans-Galactooligosaccharide on Biochemical Blood Parameters and Intestine Morphometric Parameters of Common Carp (Cyprinus carpio L.). Animals 2020, 10, 723. [Google Scholar] [CrossRef]
  234. Akter, M.N.; Zahan, K.; Zafar, M.A.; Khatun, N.; Rana, M.S.; Mursalin, M.I. Effects of Dietary Mannan Oligosaccharide on Growth Performance, Feed Utilization, Body Composition and Haematological Parameters in Asian Catfish (Clarias batrachus) Juveniles. Turk. J. Fish. Aquat. Sci. 2021, 21, 559–567. [Google Scholar] [CrossRef]
  235. Ai, Q.; Xu, H.; Mai, K.; Xu, W.; Wang, J.; Zhang, W. Effects of Dietary Supplementation of Bacillus subtilis and Fructooligosaccharide on Growth Performance, Survival, Non-Specific Immune Response and Disease Resistance of Juvenile Large Yellow Croaker, Larimichthys Crocea. Aquaculture 2011, 317, 155–161. [Google Scholar] [CrossRef]
  236. Hu, Y.; Winter, V.; Gänzle, M. In Vitro Digestibility of Commercial and Experimental Isomalto-Oligosaccharides. Food Res. Int. 2020, 134, 109250. [Google Scholar] [CrossRef]
  237. Oushani, A.K.; Soltani, M.; Sheikhzadeh, N.; Mehrgan, M.S.; Islami, H.R. Effects of Dietary Chitosan and Nano-Chitosan Loaded Clinoptilolite on Growth and Immune Responses of Rainbow Trout (Oncorhynchus mykiss). Fish Shellfish Immunol. 2020, 98, 210–217. [Google Scholar] [CrossRef] [PubMed]
  238. Wu, Y.; Rashidpour, A.; Almajano, M.P.; Metón, I. Chitosan-Based Drug Delivery System: Applications in Fish Biotechnology. Polymers 2020, 12, 1177. [Google Scholar] [CrossRef] [PubMed]
  239. Andresen, A.M.S.; Gjøen, T. Chitosan Nanoparticle Formulation Attenuates Poly (I: C) Induced Innate Immune Responses against Inactivated Virus Vaccine in Atlantic Salmon (Salmo Salar). Comp. Biochem. Physiol. Part D Genom. Proteom. 2021, 40, 100915. [Google Scholar] [CrossRef]
  240. Kamilya, D.; Khan, M.I.R. Chitin and Chitosan as Promising Immunostimulant for Aquaculture. In Handbook of Chitin and Chitosan; Elsevier: Amsterdam, The Netherlands, 2020; pp. 761–771. [Google Scholar]
  241. De Campos, C.M.; Zanuzzo, F.S.; Gimbo, R.Y.; Favero, G.C.; Soares, M.P.; Pilarski, F.; Urbinati, E.C. Dietary Inulin Modulated the Cortisol Response and Increased the Protection against Pathogens in Juvenile Pacu (Piaractus mesopotamicus). Aquac. Res. 2022, 53, 860–869. [Google Scholar] [CrossRef]
  242. Wang, T.; Zhang, N.; Yu, X.-B.; Qiao, F.; Chen, L.-Q.; Du, Z.-Y.; Zhang, M.-L. Inulin Alleviates Adverse Metabolic Syndrome and Regulates Intestinal Microbiota Composition in Nile Tilapia (Oreochromis niloticus) Fed with High-Carbohydrate Diet. Br. J. Nutr. 2021, 126, 161–171. [Google Scholar] [CrossRef]
  243. Xue, S.; Xia, B.; Zhang, B.; Li, L.; Zou, Y.; Shen, Z.; Xiang, Y.; Han, Y.; Chen, W. Mannan Oligosaccharide (MOS) on Growth Performance, Immunity, Inflammatory and Antioxidant Responses of the Common Carp (Cyprinus carpio) under Ammonia Stress. Front. Mar. Sci. 2022, 9, 1062597. [Google Scholar] [CrossRef]
  244. Zhou, L.; Li, H.; Qin, J.G.; Wang, X.; Chen, L.; Xu, C.; Li, E. Dietary Prebiotic Inulin Benefits on Growth Performance, Antioxidant Capacity, Immune Response and Intestinal Microbiota in Pacific White Shrimp (Litopenaeus vannamei) at Low Salinity. Aquaculture 2020, 518, 734847. [Google Scholar] [CrossRef]
  245. Piqué, N.; Berlanga, M.; Miñana-Galbis, D. Health Benefits of Heat-Killed (Tyndallized) Probiotics: An Overview. Int. J. Mol. Sci. 2019, 20, 2534. [Google Scholar] [CrossRef]
  246. Aggarwal, S.; Sabharwal, V.; Kaushik, P.; Joshi, A.; Aayushi, A.; Suri, M. Postbiotics: From Emerging Concept to Application. Front. Sustain. Food Syst. 2022, 6, 887642. [Google Scholar] [CrossRef]
  247. 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. The International Scientific Association of Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef]
  248. Mantziari, A.; Salminen, S.; Szajewska, H. Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases. Microorganisms 2020, 8, 1510. [Google Scholar] [CrossRef] [PubMed]
  249. Nataraj, B.H.; Ali, S.A.; Behare, P.V.; Yadav, H. Postbiotics-Parabiotics: The New Horizons in Microbial Biotherapy and Functional Foods. Microb. Cell Factories 2020, 19, 168. [Google Scholar] [CrossRef] [PubMed]
  250. Jastrząb, R.; Graczyk, D.; Siedlecki, P. Molecular and Cellular Mechanisms Influenced by Postbiotics. Int. J. Mol. Sci. 2021, 22, 13475. [Google Scholar] [CrossRef]
  251. Vallejo-Cordoba, B.; Castro-López, C.; García, H.S.; González-Córdova, A.F.; Hernández-Mendoza, A. Postbiotics and Paraprobiotics: A Review of Current Evidence and Emerging Trends. Adv. Food Nutr. Res. 2020, 94, 1–34. [Google Scholar] [CrossRef] [PubMed]
  252. MacKenzie, S.A.; Roher, N.; Boltaña, S.; Goetz, F.W. Peptidoglycan, Not Endotoxin, Is the Key Mediator of Cytokine Gene Expression Induced in Rainbow Trout Macrophages by Crude LPS. Mol. Immunol. 2010, 47, 1450–1457. [Google Scholar] [CrossRef] [PubMed]
  253. Li, Q.; Cui, K.; Xu, D.; Wu, M.; Mai, K.; Ai, Q. Molecular Identification of Peptidoglycan Recognition Protein 5 and Its Functional Characterization in Innate Immunity of Large Yellow Croaker, Larimichthys crocea. Dev. Comp. Immunol. 2021, 124, 104130. [Google Scholar] [CrossRef] [PubMed]
  254. Casadei, E.; Bird, S.; Wadsworth, S.; Vecino, J.L.G.; Secombes, C.J. The Longevity of the Antimicrobial Response in Rainbow Trout (Oncorhynchus mykiss) Fed a Peptidoglycan (PG) Supplemented Diet. Fish Shellfish Immunol. 2015, 44, 316–320. [Google Scholar] [CrossRef]
  255. Song, X.; Zhang, Y.; Wei, S.; Huang, J. Effects of Different Enzymatic Hydrolysis Methods on the Bioactivity of Peptidoglycan in Litopenaeus vannamei. Chin. J. Oceanol. Limnol. 2013, 31, 374–383. [Google Scholar] [CrossRef]
  256. Itami, T.; Asano, M.; Tokushige, K.; Kubono, K.; Nakagawa, A.; Takeno, N.; Nishimura, H.; Maeda, M.; Kondo, M.; Takahashi, Y. Enhancement of Disease Resistance of Kuruma Shrimp, Penaeus Japonicus, after Oral Administration of Peptidoglycan Derived from Bifidobacterium thermophilum. Aquaculture 1998, 164, 277–288. [Google Scholar] [CrossRef]
  257. Teame, T.; Wang, A.; Xie, M.; Zhang, Z.; Yang, Y.; Ding, Q.; Gao, C.; Olsen, R.E.; Ran, C.; Zhou, Z. Paraprobiotics and Postbiotics of Probiotic lactobacilli, Their Positive Effects on the Host and Action Mechanisms: A Review. Front. Nutr. 2020, 7, 570344. [Google Scholar] [CrossRef]
  258. Du, Y.; Wang, B.; Jiang, K.; Wang, M.; Zhou, S.; Liu, M.; Wang, L. Exploring the Influence of the Surface Proteins on Probiotic Effects Performed by Lactobacillus Pentosus HC-2 Using Transcriptome Analysis in Litopenaeus vannamei Midgut. Fish Shellfish Immunol. 2019, 87, 853–870. [Google Scholar] [CrossRef] [PubMed]
  259. Sudhakaran, G.; Guru, A.; Haridevamuthu, B.; Murugan, R.; Arshad, A.; Arockiaraj, J. Molecular Properties of Postbiotics and Their Role in Controlling Aquaculture Diseases. Aquac. Res. 2022, 53, 3257–3273. [Google Scholar] [CrossRef]
  260. Kim, D.-H.; Subramanian, D.; Park, S.-H.; Jang, Y.-H.; Heo, M.-S. Assessment and Potential Application of the Probiotic Strain, Bacillus Amyloliquefaciens JFP2, Isolated from Fermented Seafood-Jeotgal in Flounder Paralichthys olivaceus Juveniles. Isr. J. Aquac.-Bamidgeh 2017, 69, 1–12. [Google Scholar] [CrossRef]
  261. Mora-Sánchez, B.; Balcázar, J.L.; Pérez-Sánchez, T. Effect of a Novel Postbiotic Containing Lactic Acid Bacteria on the Intestinal Microbiota and Disease Resistance of Rainbow Trout (Oncorhynchus mykiss). Biotechnol. Lett. 2020, 42, 1957–1962. [Google Scholar] [CrossRef] [PubMed]
  262. İncili, G.K.; Karatepe, P.; Akgöl, M.; Güngören, A.; Koluman, A.; İlhak, O.İ.; Kanmaz, H.; Kaya, B.; Hayaloğlu, A.A. Characterization of Lactic Acid Bacteria Postbiotics, Evaluation in-Vitro Antibacterial Effect, Microbial and Chemical Quality on Chicken Drumsticks. Food Microbiol. 2022, 104, 104001. [Google Scholar] [CrossRef]
  263. Chang, H.M.; Foo, H.L.; Loh, T.C.; Lim, E.T.C.; Abdul Mutalib, N.E. Comparative Studies of Inhibitory and Antioxidant Activities, and Organic Acids Compositions of Postbiotics Produced by Probiotic Lactiplantibacillus Plantarum Strains Isolated from Malaysian Foods. Front. Vet. Sci. 2021, 7, 602280. [Google Scholar] [CrossRef] [PubMed]
  264. Luo, K.; Tian, X.; Wang, B.; Wei, C.; Wang, L.; Zhang, S.; Liu, Y.; Li, T.; Dong, S. Evaluation of Paraprobiotic Applicability of Clostridium butyricum CBG01 in Improving the Growth Performance, Immune Responses and Disease Resistance in Pacific White Shrimp, Penaeus vannamei. Aquaculture 2021, 544, 737041. [Google Scholar] [CrossRef]
  265. Wu, X.; Teame, T.; Hao, Q.; Ding, Q.; Liu, H.; Ran, C.; Yang, Y.; Zhang, Y.; Zhou, Z.; Duan, M. Use of a Paraprobiotic and Postbiotic Feed Supplement (HWFTM) Improves the Growth Performance, Composition and Function of Gut Microbiota in Hybrid Sturgeon (Acipenser baerii x Acipenser Schrenckii). Fish Shellfish Immunol. 2020, 104, 36–45. [Google Scholar] [CrossRef]
  266. Del Valle, J.C.; Bonadero, M.C.; Gimenez, A.V.F. Saccharomyces cerevisiae as Probiotic, Prebiotic, Synbiotic, Postbiotics and Parabiotics in Aquaculture: An Overview. Aquaculture 2023, 569, 739342. [Google Scholar] [CrossRef]
  267. Sousa, J.M.G.; Louvado, A.; Coelho, F.; Oliveira, V.; Oliveira, H.; Cleary, D.F.R.; Gomes, N.C.M. In Vitro Study of the Modulatory Effects of Heat-Killed Bacterial Biomass on Aquaculture Bacterioplankton Communities. Sci. Rep. 2022, 12, 19699. [Google Scholar] [CrossRef]
  268. Feng, J.; Cai, Z.; Chen, Y.; Zhu, H.; Chang, X.; Wang, X.; Liu, Z.; Zhang, J.; Nie, G. Effects of an Exopolysaccharide from Lactococcus lactis Z-2 on Innate Immune Response, Antioxidant Activity, and Disease Resistance against Aeromonas hydrophila in Cyprinus Carpio L. Fish Shellfish Immunol. 2020, 98, 324–333. [Google Scholar] [CrossRef]
  269. Gao, Q.; Gao, Q.; Min, M.; Zhang, C.; Peng, S.; Shi, Z. Ability of Lactobacillus plantarum Lipoteichoic Acid to Inhibit Vibrio anguillarum-Induced Inflammation and Apoptosis in Silvery Pomfret (Pampus argenteus) Intestinal Epithelial Cells. Fish Shellfish Immunol. 2016, 54, 573–579. [Google Scholar] [CrossRef]
  270. Meng, D.; Hao, Q.; Zhang, Q.; Yu, Z.; Liu, S.; Yang, Y.; Ran, C.; Zhang, Z.; Zhou, Z. A Compound of Paraprobiotic and Postbiotic Derived from Autochthonous Microorganisms Improved Growth Performance, Epidermal Mucus, Liver and Gut Health and Gut Microbiota of Common Carp (Cyprinus carpio). Aquaculture 2023, 570, 739378. [Google Scholar] [CrossRef]
  271. Rimoldi, S.; Gini, E.; Koch, J.F.A.; Iannini, F.; Brambilla, F.; Terova, G. Effects of Hydrolyzed Fish Protein and Autolyzed Yeast as Substitutes of Fishmeal in the Gilthead Sea Bream (Sparus aurata) Diet, on Fish Intestinal Microbiome. BMC Vet. Res. 2020, 16, 118. [Google Scholar] [CrossRef]
  272. Van Nguyen, N.; Onoda, S.; Van Khanh, T.; Hai, P.D.; Trung, N.T.; Hoang, L.; Koshio, S. Evaluation of Dietary Heat-Killed Lactobacillus plantarum Strain L-137 Supplementation on Growth Performance, Immunity and Stress Resistance of Nile Tilapia (Oreochromis niloticus). Aquaculture 2019, 498, 371–379. [Google Scholar] [CrossRef]
  273. Dawood, M.A.; Abo-Al-Ela, H.G.; Hasan, M.T. Modulation of Transcriptomic Profile in Aquatic Animals: Probiotics, Prebiotics and Synbiotics Scenarios. Fish Shellfish Immunol. 2020, 97, 268–282. [Google Scholar] [CrossRef]
  274. Kuo, H.-W.; Chang, C.-C.; Cheng, W. Synbiotic Combination of Prebiotic, Cacao Pod Husk Pectin and Probiotic, Lactobacillus plantarum, Improve the Immunocompetence and Growth of Litopenaeus vannamei. Fish Shellfish Immunol. 2021, 118, 333–342. [Google Scholar] [CrossRef] [PubMed]
  275. Ghafarifarsani, H.; Hoseinifar, S.H.; Talebi, M.; Yousefi, M.; Van Doan, H.; Rufchaei, R.; Paolucci, M. Combined and Singular Effects of Ethanolic Extract of Persian Shallot (Allium Hirtifolium Boiss) and Synbiotic Biomin® IMBO on Growth Performance, Serum-and Mucus-Immune Parameters and Antioxidant Defense in Zebrafish (Danio rerio). Animals 2021, 11, 2995. [Google Scholar] [CrossRef] [PubMed]
  276. Mohammadi, G.; Hafezieh, M.; Karimi, A.A.; Azra, M.N.; Van Doan, H.; Tapingkae, W.; Abdelrahman, H.A.; Dawood, M.A. The Synergistic Effects of Plant Polysaccharide and Pediococcus Acidilactici as a Synbiotic Additive on Growth, Antioxidant Status, Immune Response, and Resistance of Nile Tilapia (Oreochromis niloticus) against Aeromonas Hydrophila. Fish Shellfish Immunol. 2022, 120, 304–313. [Google Scholar] [CrossRef] [PubMed]
  277. Dehaghani, P.G.; Baboli, M.J.; Moghadam, A.T.; Ziaei-Nejad, S.; Pourfarhadi, M. Effect of Synbiotic Dietary Supplementation on Survival, Growth Performance, and Digestive Enzyme Activities of Common Carp (Cyprinus carpio) Fingerlings. Czech J. Anim. Sci. 2015, 60, 224–232. [Google Scholar] [CrossRef]
  278. Kapoor, D.; Sharma, P.; Sharma, M.M.M.; Kumari, A.; Kumar, R. Microbes in Pharmaceutical Industry. In Microbial Diversity, Interventions and Scope; Springer: Berlin/Heidelberg, Germany, 2020; pp. 259–299. [Google Scholar] [CrossRef]
  279. Elabd, H.; Faggio, C.; Mahboub, H.H.; Emam, M.A.; Kamel, S.; El Kammar, R.; Abdelnaeim, N.S.; Shaheen, A.; Tresnakova, N.; Matter, A. Mucuna Pruriens Seeds Extract Boosts Growth, Immunity, Testicular Histology, and Expression of Immune-Related Genes of Mono-Sex Nile Tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2022, 127, 672–680. [Google Scholar] [CrossRef] [PubMed]
  280. Lumsangkul, C.; Linh, N.V.; Chaiwan, F.; Abdel-Tawwab, M.; Dawood, M.A.; Faggio, C.; Jaturasitha, S.; Van Doan, H. Dietary Treatment of Nile Tilapia (Oreochromis niloticus) with Aquatic Fern (Azolla caroliniana) Improves Growth Performance, Immunological Response, and Disease Resistance against Streptococcus agalactiae Cultured in Bio-Floc System. Aquac. Rep. 2022, 24, 101114. [Google Scholar] [CrossRef]
  281. Jamal, M.T.; Sumon, A.A.; Pugazhendi, A.; Al Harbi, M.; Hussain, A.; Haque, F. Use of Probiotics in Commercially Important Finfish Aquaculture. Int. J. Probiotics Prebiotics 2020, 15, 7–21. [Google Scholar] [CrossRef]
  282. Prabawati, E.; Hu, S.-Y.; Chiu, S.-T.; Balantyne, R.; Risjani, Y.; Liu, C.-H. A Synbiotic Containing Prebiotic Prepared from a By-Product of King Oyster Mushroom, Pleurotus eryngii and Probiotic, Lactobacillus plantarum Incorporated in Diet to Improve the Growth Performance and Health Status of White Shrimp, Litopenaeus Vannamei. Fish Shellfish Immunol. 2022, 120, 155–165. [Google Scholar] [CrossRef] [PubMed]
  283. Panigrahi, A.; Das, R.R.; Sivakumar, M.R.; Saravanan, A.; Saranya, C.; Sudheer, N.S.; Vasagam, K.K.; Mahalakshmi, P.; Kannappan, S.; Gopikrishna, G. Bio-Augmentation of Heterotrophic Bacteria in Biofloc System Improves Growth, Survival, and Immunity of Indian White Shrimp Penaeus Indicus. Fish Shellfish Immunol. 2020, 98, 477–487. [Google Scholar] [CrossRef]
  284. Rajeev, R.; Adithya, K.K.; Kiran, G.S.; Selvin, J. Healthy Microbiome: A Key to Successful and Sustainable Shrimp Aquaculture. Rev. Aquac. 2021, 13, 238–258. [Google Scholar] [CrossRef]
  285. Safari, O.; Sarkheil, M.; Shahsavani, D.; Paolucci, M. Effects of Single or Combined Administration of Dietary Synbiotic and Sodium Propionate on Humoral Immunity and Oxidative Defense, Digestive Enzymes and Growth Performances of African Cichlid (Labidochromis lividus) Challenged with Aeromonas hydrophila. Fishes 2021, 6, 63. [Google Scholar] [CrossRef]
  286. Ye, J.-D.; Wang, K.; Li, F.-D.; Sun, Y.-Z. Single or Combined Effects of Fructo- and Mannan Oligosaccharide Supplements and Bacillus clausii on the Growth, Feed Utilization, Body Composition, Digestive Enzyme Activity, Innate Immune Response and Lipid Metabolism of the Japanese Flounder Paralichth: Dietary Benefits of FOS, MOS and B. clausii in the Japanese Flounder. Aquac. Nutr. 2011, 17, e902–e911. [Google Scholar] [CrossRef]
  287. Hoseinifar, S.H.; Hoseini, S.M.; Bagheri, D. Effects of Galactooligosaccharide and Pediococcus Acidilactici on Antioxidant Defence and Disease Resistance of Rainbow Trout, Oncorhynchus Mykiss. Ann. Anim. Sci. 2017, 17, 217–227. [Google Scholar] [CrossRef]
  288. Wan, J.-J.; Pan, J.; Shen, M.-F.; Xue, H.; Sun, M.; Zhang, M.-Q.; Zhu, X.-H.; Ma, X. Changes in the Growth Performance, Antioxidant Enzymes and Stress Resistance Caused by Dietary Administration of Synbiotic (Fructooligosaccharide and Probiotics) in Juvenile Chinese Mitten Crab, Eriocheir Sinensis. Aquac. Int. 2022, 30, 467–481. [Google Scholar] [CrossRef]
  289. Maniat, M.; Salati, A.P.; Zanguee, N.; Mousavi, S.M.; Hoseinifar, S.H. Effects of Dietary Pediococcus acidilactici and Isomaltooligosaccharide on Growth Performance, Immunity, and Antioxidant Defense in Juvenile Common Carp. Aquac. Nutr. 2023, 2023, e1808640. [Google Scholar] [CrossRef] [PubMed]
  290. Rohani, M.F.; Islam, S.M.; Hossain, M.K.; Ferdous, Z.; Siddik, M.A.; Nuruzzaman, M.; Padeniya, U.; Brown, C.; Shahjahan, M. Probiotics, Prebiotics and Synbiotics Improved the Functionality of Aquafeed: Upgrading Growth, Reproduction, Immunity and Disease Resistance in Fish. Fish Shellfish Immunol. 2022, 120, 569–589. [Google Scholar] [CrossRef] [PubMed]
  291. Dawood, M.A.O.; Koshio, S.; Abdel-Daim, M.M.; Van Doan, H. Probiotic Application for Sustainable Aquaculture. Rev. Aquac. 2019, 11, 907–924. [Google Scholar] [CrossRef]
  292. Goh, J.X.H.; Tan, L.T.-H.; Law, J.W.-F.; Ser, H.-L.; Khaw, K.-Y.; Letchumanan, V.; Lee, L.-H.; Goh, B.-H. Harnessing the Potentialities of Probiotics, Prebiotics, Synbiotics, Paraprobiotics, and Postbiotics for Shrimp Farming. Rev. Aquac. 2022, 14, 1478–1557. [Google Scholar] [CrossRef]
  293. Carbone, D.; Faggio, C. Importance of Prebiotics in Aquaculture as Immunostimulants. Effects on Immune System of Sparus Aurata and Dicentrarchus Labrax. Fish Shellfish Immunol. 2016, 54, 172–178. [Google Scholar] [CrossRef] [PubMed]
  294. Ghaly, F.M.; Hussein, S.H.; Awad, S.M.; El-Makhzangy, A.A. Growth Promoter, Immune Response, and Histopathological Change of Prebiotic, Probiotic and Synbiotic Bacteria on Nile Tilapia. Saudi J. Biol. Sci. 2023, 30, 103539. [Google Scholar] [CrossRef]
  295. Jahangiri, L.; Esteban, M.Á. Administration of Probiotics in the Water in Finfish Aquaculture Systems: A Review. Fishes 2018, 3, 33. [Google Scholar] [CrossRef]
  296. Subedi, B.; Shrestha, A. A Review: Application of Probiotics in Aquaculture. Int. J. For. Anim. Fish. Res. 2020, 4, 5. [Google Scholar]
  297. Peñalosa-Martinell, D.; Araneda-Padilla, M.; Dumas, S.; Martinez-Díaz, S.; Vela-Magaña, M. The Use of Probiotics in Larval Whiteleg Shrimp ( Litopenaeus vannamei ) Production: A Marginal Analysis of Bioeconomic Feasibility. Aquac. Res. 2021, 52, 943–951. [Google Scholar] [CrossRef]
  298. De Azevedo, R.V.; Fosse Filho, J.C.; Cardoso, L.D.; da Mattos, D.C.; Vidal Júnior, M.V.; de Andrade, D.R. Economic Evaluation of Prebiotics, Probiotics and Symbiotics in Juvenile Nile Tilapia. Rev. Ciênc. Agronômica 2015, 46, 72–79. [Google Scholar] [CrossRef]
  299. Dias, D.d.C.; Furlaneto, F.d.P.B.; Sussel, F.R.; Tachibana, L.; Gonçalves, G.S.; Ishikawa, C.M.; Natori, M.M.; Ranzani-Paiva, M.J.T. Economic Feasibility of Probiotic Use in the Diet of Nile Tilapia, Oreochromis niloticus, during the Reproductive Period. Acta Sci. Anim. Sci. 2020, 42, e47960. [Google Scholar] [CrossRef]
Figure 1. Illustration of the use and impact of probiotics in aquaculture systems.
Figure 1. Illustration of the use and impact of probiotics in aquaculture systems.
Biology 12 01498 g001
Figure 2. Main chemical components of prebiotics from natural sources and their action modes in improving host health.
Figure 2. Main chemical components of prebiotics from natural sources and their action modes in improving host health.
Biology 12 01498 g002
Figure 3. Postbiotic main components and molecules.
Figure 3. Postbiotic main components and molecules.
Biology 12 01498 g003
Figure 4. Postbiotics in aquaculture.
Figure 4. Postbiotics in aquaculture.
Biology 12 01498 g004
Figure 5. Illustration of modes of action of synbiotics in aquaculture.
Figure 5. Illustration of modes of action of synbiotics in aquaculture.
Biology 12 01498 g005
Table 1. A list of current probiotic strains for use in aquaculture.
Table 1. A list of current probiotic strains for use in aquaculture.
GenusProbioticsExample of Target Fish SpeciesReferences
BacillusBacillus coagulansCommon carp (Cyprinus carpio), turbot (Scophthalmus Maximus)[52,53]
Bacillus subtilisNile tilapia (O. niloticus)[54]
Bacillus licheniformisGrass carp (Ctenopharyngodon idella)[55]
Bacillus cereusCatfish (Heteropneustes fossilis)[56]
BifidobacteriumBifidobacterim bifidusKoi fish (Cyprinus rubrofuscus)[57]
CarnobacteriumCarnobacterium divergensAtlantic cod (Gadus morhua)[58]
EnterococcusEnterococcus faeciumNile tilapia (O. niloticus)[59]
LactobacillusLactobacillus caseiCommon carp (Cyprinus carpio)[60]
L. plantarumBlack sea bream (Acanthopagrus schlegelii)[61]
L. rhamnosusNile tilapia (O. niloticus)[62]
LactococcusL. lactisMandarin fish (Siniperca chuatsi)[63]
PediococcusPediococcus acidilacticiRainbow trout (Oncorhynchus mykiss)[64]
StreptomycesStreptomyces sp.Zebrafish (Danio rerio)[65]
SaccharomycesSaccharomyces cerevisiaeStriped catfish (Pangasianodon hypophthalmus)[66]
WeissellaWeissella cibariaCommon carp (Cyprinus carpio)[67]
Table 2. Functional feed additives of major probiotics in aquatic animals.
Table 2. Functional feed additives of major probiotics in aquatic animals.
Probiotics OrganismsFunctions Aquatic OrganismsReferences
Bacillus
B. licheniformis HGA8B↑ growth performance and ↓ feed conversion ratio
Up-regulation of immune genes
O. niloticus[153]
B. cereus G19
B. cereus BC-01
↑ growth and immunityApostichopus japonicus[154]
B. cereus EN25Immunity and resistance against Vibrio splendidusA. japonicus[155]
B. pumilus SE5↑ growth and immunityL. vannamei[156]
B. subtilis AB1Bactericidal activity against Aeromonas infectionO. mykiss[157]
Bifidobacterium
Bifidobacterium animalis PTCC-1631 ↑ growth performance, digestion, and nutrient utilizationO. mykiss[158]
B. lactis PTCC-1736↑ growth, nutrient digestibility, and carcass compositionO. mykiss[158]
Carnobacterium
C. divergens
C. maltaromaticum
Antagonistic effects against V. anguillarum, V. viscosus, and A. salmonicida-[159,160]
Lactobacillus
L. plantarum CLFP↓ mortality against harmful strain L. garvieaeO. mykiss[161]
L. acidophilusSurvival against Staphylococcus xylosus, Aeromonas hydrophila gr.2, and Streptococcus agalactiae infectionClarias gariepinus[162]
L. pentosus↑ growth performance and feed conversion ratio
↑ survival against Vibrio species
L. vannamei[163]
Lactococcus
Lactococcus lactis BFE920 Activation of nonspecific immune system
Bactericidal activity against S. iniae
Paralichthys olivaceus[164]
Leuconostoc
Lc. Mesenteroides CLFP 196↑ survival against A. salmonicida infectionSalmo trutta[165]
Pediococcus
P. pentosaceus HN10↑ feed utilization, digestive enzyme activity, and anti-Vibrio activityL. vannamei[166]
Enterococcus
E. casseliflavus CGMCC1.2136 ↑ growth performance, immunity, and digestive enzyme activityRutilus rutilus caspicus[167]
E. casseliflavus↑ growth performance and disease resistance against S. iniaeO. mykiss[168]
E. durans↑ growth performance and survival rateO. mykiss[169]
Clostridium
C. butyricum↑ antibacterial activity against Vibriosis infectionO. mykiss[170]
C. butyricum↑ immunity; regulation of gut microbiota; antagonistic effects against Aeromonas sp., Vibrio sp., and Pseudomonas sp.C. carpio[171]
Weissella
W. confusa↑ growth performanceO. mykiss[172]
W. confusa↑ growth performance and antibacterial activity against A. hydrophilaLates calcarifer[173]
Other strains
A. veronii BA-1↑ immune system and antibacterial activityC. carpio[174]
Micrococcus luteus↑ growth performance and feed conversion ratioO. niloticus[175]
Pseudoalteromonas undina VKM-124↑ survival and antiviral activityCarangoides bartholomaei[99]
Yeast
S.cerevisiae↑ growth performance and resistance against waterborne Cu toxicitySarotherodon galilaeus[176]
S. cerevisiae↑ immunity and ↓ mortality against P. fluorescensMystus cavasius[177]
Yarrowia lipolytica↑ immune response, antioxidant status, and disease resistance against V. parahaemolyticus infection Lutjanus peru[178]
Multi-strain
B. subtilis and Bacillus licheniformis (BioPlus2B)↑ resistance against Y. ruckeri O. mykiss[179]
Lactobacillus delbrueckii
Lactobacillus rhamnosus
L. plantarum
B. bifidum
↑ growth performance and immunityAcipenser baerii[180]
Lactobacillus plantarum (STBL1), Saccharomyces cerevisiae (STBS1), and Bacillus safensis (SQVG18)↑ growth, antioxidant capacity, digestion, and gut microfloraP. vannamei[181]
decrease or reduction; ↑, increase or improvement.
Table 3. Functional feed additives of prebiotics in aquatic animals.
Table 3. Functional feed additives of prebiotics in aquatic animals.
PrebioticsFunctionsAquatic SpeciesReferences
FOS↑ growth, survival, and gut microbiota sectionL. vannamei[208]
β-glucan↑ growth, survival, and immune systemSparus aurata[225]
MOS↑ growth, immune system, antioxidant capacity, and intestinal healthCyprinus carpio[243]
Chitosan↑ growth, feed utilization, lipid metabolism, gut microbiota composition, and immune systemCyprinus carpio koi[240]
Inulin↑ growth, antioxidant capacity, immunity, and gut microbiota at low salinityL. vannamei[244]
↑, increase or improvement.
Table 4. Some recent potential applications of postbiotics in aquaculture.
Table 4. Some recent potential applications of postbiotics in aquaculture.
PostbioticsMicroorganism ProducerAquatic SpeciesApplicationsReferences
ExopolysaccharidesLactococcus lactis Z-2Common carp
(C. carpio)
Immunity enhancement
Resistance against A. hydrophila
[268]
Cell surface proteinsL. pentosusShrimp
(Litopenaeus vannamei)
Immune response improvement[258]
Cell wall components (PGs and LTA)B. pumilus SE5Grouper
(E. coioides)
Growth performance improvement
Innate and adaptive immunity amelioration
[109]
Lipoteichoic acidsL. plantarum LTASilvery pomfret
(Pampus argenteus)
Resistance against V. anguillarum-caused vibriosis [269]
Non-living microorganismsS. cerevisiae, B. velezensis and Cetobacterium someraeCommon carp
(C. carpio)
Gut microbiota improvement Enhancement of nonspecific immunity
Antioxidant status improvement
[270]
Dried autolyzed yeastGilthead sea bream (Sparus aurata)Intestinal microbiota improvement[271]
Rhodotorula minuta and Cetobacterium someraeHybrid sturgeon
(Acipenser baerii × Acipenser schrencki)
Growth performance improvement
Nonspecific immunity improvement
[265]
Heat-killed L. plantarum L-137Nile tilapia
(O. niloticus)
Growth performance stress resistance and immunity enhancement[272]
Table 5. Functional feed additives of synbiotics in aquatic animals.
Table 5. Functional feed additives of synbiotics in aquatic animals.
SynbioticsFunctionsAquatic OrganismsReferences
P.acidilactici + GOS↑ growth, survival, and digestive enzyme functionLabidochromis lividus[285]
B. clausii + FOS, MOS ↑ growth, survival, and digestive enzyme functionParalichthys olivaceus[286]
P.acidilactici + GOS↑ immunity and antagonistic activity against S. iniae infectionsOncorhynchus mykiss[287]
B. subtilis + L. acidophilus + S. cerevisiae + FOS↑ growth and feed efficiency ratioEriocheir sinensis[288]
P.acidilactici + IMO↑ growth, immune response, and antioxidant capacityC. carpio[289]
↑, increase or improvement.
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

Srirengaraj, V.; Razafindralambo, H.L.; Rabetafika, H.N.; Nguyen, H.-T.; Sun, Y.-Z. Synbiotic Agents and Their Active Components for Sustainable Aquaculture: Concepts, Action Mechanisms, and Applications. Biology 2023, 12, 1498. https://doi.org/10.3390/biology12121498

AMA Style

Srirengaraj V, Razafindralambo HL, Rabetafika HN, Nguyen H-T, Sun Y-Z. Synbiotic Agents and Their Active Components for Sustainable Aquaculture: Concepts, Action Mechanisms, and Applications. Biology. 2023; 12(12):1498. https://doi.org/10.3390/biology12121498

Chicago/Turabian Style

Srirengaraj, Vijayaram, Hary L. Razafindralambo, Holy N. Rabetafika, Huu-Thanh Nguyen, and Yun-Zhang Sun. 2023. "Synbiotic Agents and Their Active Components for Sustainable Aquaculture: Concepts, Action Mechanisms, and Applications" Biology 12, no. 12: 1498. https://doi.org/10.3390/biology12121498

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

Article Metrics

Back to TopTop