Next Article in Journal
Effect of Chitosan on Ruminal Fermentation and Microbial Communities, Methane Emissions, and Productive Performance of Dairy Cattle
Next Article in Special Issue
Influence of Transportation on Stress Response and Cellular Oxidative Stress Markers in Juvenile Meagre (Argyrosomus regius)
Previous Article in Journal
Nociception in Chicken Embryos, Part III: Analysis of Movements before and after Application of a Noxious Stimulus
Previous Article in Special Issue
How Do Alternative Protein Resources Affect the Intestine Morphology and Microbiota of Atlantic Salmon?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Histomorphological Changes in Fish Gut in Response to Prebiotics and Probiotics Treatment to Improve Their Health Status: A Review

Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
*
Author to whom correspondence should be addressed.
Animals 2023, 13(18), 2860; https://doi.org/10.3390/ani13182860
Submission received: 9 August 2023 / Revised: 6 September 2023 / Accepted: 6 September 2023 / Published: 8 September 2023
(This article belongs to the Special Issue Morphological and Physiological Research on Fish)

Abstract

:

Simple Summary

Activities such as the digestion and absorption of feeds occur into the gastrointestinal tract (GIT), which also serves to excrete waste products of digestion. These processes occur thanks to the different species of microorganisms inhabiting the GIT, the microbiota, which contribute to the health status of fish by providing metabolic benefits and counteracting pathogen infection. The microbiota is affected by environmental conditions and by the dietary habits of fish species, and it may be modulated by the administration of feed additives based on prebiotics and probiotics. These represent a very useful tool to improve the health status of fish since they are able to enhance gut efficiency, nutrient uptake, defense against pathogens, and growth performance, as may also be assessed by histological endpoints. Overall, a better understanding of the appropriate administration of feed supplements for individual fish species is a promising strategy for improving of the quality and sustainability of the aquaculture sector.

Abstract

The gastrointestinal tract (GIT) promotes the digestion and absorption of feeds, in addition to the excretion of waste products of digestion. In fish, the GIT is divided into four regions, the headgut, foregut, midgut, and hindgut, to which glands and lymphoid tissues are associated to release digestive enzymes and molecules involved in the immune response and control of host-pathogens. The GIT is inhabited by different species of resident microorganisms, the microbiota, which have co-evolved with the host in a symbiotic relationship and are responsible for metabolic benefits and counteracting pathogen infection. There is a strict connection between a fish’s gut microbiota and its health status. This review focuses on the modulation of fish microbiota by feed additives based on prebiotics and probiotics as a feasible strategy to improve fish health status and gut efficiency, mitigate emerging diseases, and maximize rearing and growth performance. Furthermore, the use of histological assays as a valid tool for fish welfare assessment is also discussed, and insights on nutrient absorptive capacity and responsiveness to pathogens in fish by gut morphological endpoints are provided. Overall, the literature reviewed emphasizes the complex interactions between microorganisms and host fish, shedding light on the beneficial use of prebiotics and probiotics in the aquaculture sector, with the potential to provide directions for future research.

1. Introduction

In teleosts, the gastrointestinal tract (GIT) consists of a continuous hollow tube that opens to the outside at both ends, through the mouth at one end and through the anus at the other. Along this structure, it is possible to recognize areas that differ in histology and function since several activities occur, including digestion, absorption, and excretion of waste materials. As in mammals, a number of endogenous enzymes and key molecules produced by associated glands, such as the liver, pancreas, and cells of the intestinal wall, are released into the GIT to contribute to all of these actions [1]. The role of these enzymes is coupled with a variety of fermentation processes carried out by microorganisms (bacteria, fungi, and yeasts) colonizing the GIT that constitute the so-called “microbiota”, which has co-evolved with the host in a symbiotic relationship and is responsible for providing metabolic benefits and counteracting pathogen infection.
In fish, the number of microorganisms inhabiting the GIT has been estimated in the range of 107–108 per gram [2]. However, the GIT microbiota is highly variable and its normal variation in abundance and diversity of microorganisms is affected by several factors [3] including age, diet, host genetics, and the environment (freshwater, seawater). Feeding habits are surely one of the most relevant factors affecting the composition of the GIT microbiota among fish species. Indeed, it is well known how different dietary habits (herbivores, carnivores, omnivores, detritivores) come together with variations in both fish GIT morphology and microbial populations [3,4,5]. The strict correlation between the structures of the digestive apparatus and the feeding habit of fishes [6], as revealed in some morphometric parameters such as the intestinal length and area, highly influences the microbial populations detected along the alimentary canal [7]. On the other hand, microorganisms may also affect the digestive morphological structures and functions, both negatively [8], and positively, as in the case of probiotics [9].
In accordance with the currently adopted definition by the Food and Agricultural Organization of the United Nations (FAO) and the World Health Organization (WHO), probiotics are live microorganisms that, when administered in adequate amounts, confer health benefits to the host [10]. In fact, after administration, probiotics are able to colonize and multiply in the gut of the host and enact numerous beneficial effects by modulating various biological systems in the host [11,12], including immune status, growth performance, disease resistance, and feed conversion, with an overall improvement of the fish health status [13,14]. Probiotics, together with other additives such as prebiotics, which are referred to as the food or energy provider for good microorganisms [15], aim to optimize the host–microbiota ratio, which plays a key role in ensuring the proper functioning of the fish GIT [3,16]. Therefore, for all the above-mentioned reasons, the use of these feed additives has gained a key strategic role in the aquaculture sector [14,16].
Overall, this review focuses on the current knowledge regarding the use of a variety of probiotics and prebiotics in different fish species for the improvement of their health and gut efficiency. Furthermore, particular attention was paid to the mutual influences between the microbial populations and morphological features of the fish GIT areas, especially of the gut tract, which is usually investigated by histological assays to evaluate the performance of various feed additives in aquaculture.

2. The Gastrointestinal Tract (GIT) in Fish

As mentioned above, along the “tube”-termed GIT, it is possible to recognize different areas distinguishable from a functional and histological point of view. The main areas of the fish GIT can be summarized as follows:
  • Headgut: the area where it is possible to distinguish between a buccal and a pharyngeal cavity;
  • Foregut: the area that begins at the posterior edge of the gills and includes the oesophagus, the stomach, and the pylorus;
  • Midgut: the anterior intestine, which includes a variable number of pyloric caeca or appendages which are useful for increasing the surface area and optimizing the absorption of nutrients;
  • Hindgut: the area including the distal intestine and the anus.
The headgut, mainly characterized by the mouth cavity, plays the key role of ensuring feed acquisition; therefore, it is not unusual to observe differential features of this area among fish species in relation to their different feed habits. For instance, in lepidophagous fishes, the occurrence of a large sub-terminal mouth coupled to a unique arrangement of teeth on the jaws enables this species to perform its specialized feeding behaviour, consisting of scraping and eating the scales of other fish [7,8,9,10,11,12,13,14,15,16,17,18]. Additionally in the headgut, it is possible to find other distinctive anatomical structures such as the pharyngeal jaw apparatus that, combined with the oral jaws, allows some fish species to crush shells, mince feed, and separate edible material from unwanted debris, thus expanding their feeding capabilities [19].
The first part of the foregut is characterised by the oesophagus, with a squamous epithelium in the anterior part and a columnar one in the posterior part [20]. Noteworthy, the epithelium of the foregut has a different origin from that of the midgut, namely ectodermal for the foregut and endodermal for the midgut [21]. In addition, the foregut is characterized by the presence of goblet cells that, by producing mucous, facilitate the acquisition of food, preventing abrasion as well as invasion of pathogens [22]. These cells contribute to form the mucous layer, beneath which, a submucosal layer composed of a thick mass of loose connective tissue combined with a muscular layer is immediately arranged, which is crucial for the integrity of the oesophagus wall [23]. The occurrence of a muscular layer, which is also coupled with long branched folds in some species, suggests a certain elasticity and stretching capacity of the luminal surface area of the oesophagus, particularly in predatory fish [20,24]. Furthermore, in some fish species, the oesophagus is characterized by the presence, especially in the cranial portion, of several taste buds that are helpful for food selection [25].
Following the oesophagus, as in other vertebrates, it is quite common to recognise the stomach. Nevertheless, it is worthy to highlight that the stomach is lacking in 20% of fish species (i.e., Gobiidae, Blennidae, Cyprinadae), where it is partly replaced by other adaptations such as well-developed pharyngeal teeth, pharyngeal pockets, secretory glands present in the oesophagus, or a muscular gizzard [26,27]. In fish species where the stomach occurs, it may assume different conformations (straight, U-shaped, or Y-shaped), normally linked to the feeding habits of the specimens. For instance, the U-shaped stomach is typical of herbivores/omnivores, whereas the Y-shaped stomach is present in some predators since it allows the storage of large amounts of food [7,28]. Despite the different morphological structures, it is possible to recognize three distinct regions in fish stomach, defined as cardiac, fundic, and pyloric [17]. The primary roles of the fish stomach are the storage of food and the production of hydrochloric acid (HCl), which is necessary to promote the digestion process. The HCl secreted by the parietal cells of gastric glands generates the strongly acidic environment (pH < 2) of the gastric lumen, which is crucial for the correct activity of several digestive enzymes [21,22]. The gastric gland allocation appears to be greater in the anterior than in the posterior part of the stomach, the latter represented by the pyloric region that performs a key role in food storage rather than digestion. The occurrence of neutral mucins throughout the stomach, produced by the columnar epithelial cells, ensures a dual protective function against the presence of microorganisms and the high acidity of the gastric lumen [17,24,28]. Even though digestion into the stomach is primarily carried out by digestive enzymes, in some fish species (i.e., Clupeoidei, Channidae, Mugilidae, Acipenseridae, Coregoninae, and Chanidae), it is possible to notice a pyloric stomach able to grind food, acting as a sort of “gizzard” [5,21].
In the midgut, the forward part of the gut, the assimilation of digested food into the stomach begins. In order to fulfil this task, the pyloric caeca are located in this portion. The pyloric caeca are finger-like outgrowths whose shape permits an increase of the absorption surface area without promoting fermentation and storage, as demonstrated by [29]. Similar to the other portions of the GIT, differences in regard to size, state of branching, and connection to the gut are detectable in the various fish species. For instance, in the Mugilidae, the number of pyloric caeca is a crucial element for the identification of the species [30]. However, no clear correlation seems to exist between the pyloric caeca shape and feed habits of fish [31,32]. Interestingly, the midgut is the longest portion of the gut and its length is extremely variable and generally correlated with the feeding habits of the fish species. For instance, carnivorous species frequently possess intestines that are shorter than herbivorous fish, with large coils occurring in the former [33]. From a histological perspective, three layers, namely the mucosa, muscularis externa, and serosa, may be observed in the gut [26]. In the mucosa layer, the columnar epithelium plays the absorptive role, as testified by the presence of a distinctive brush border in the apical part. In the same layer, it is possible to observe other types of cells such as goblet-type mucous cells, lymphocytes, enteroendocrine cells, and rodlet cells [34], each one performing a different role. Indeed, the goblet cells are mucous cells [21], the enteroendocrine cells produce neuroendocrine substances [35], and the lymphocytes and rodlet cells have an immune function [36,37]. In contrast to mammals, in the fish gut, it is not possible to identify the villi organisation, but rather the existence of irregular random folds without crypts. Lieberkuhn crypt-like structures are encountered only in the Gadidae, such as cods [21,26].
The terminal part of the GIT is the hindgut. The hindgut of most fish is short and very difficult to distinguish from the midgut in terms of histomorphology, in particular with respect to changes in diameter or epithelial morphology. Additionally, in the hindgut, unlike the midgut, it is possible to observe an increased level of mucus production [5,32] and variations with regards to the mucosal fold and muscle thickness [21]. In the posterior intestine, unlike the midgut, a higher amount of goblet cells may be observed, resulting in greater mucous production. The rise in the presence of goblet cells from the anterior to posterior intestine, common in various fish species, provides epithelium protection and lubrification in order to facilitate food flow and defecation [17,22,38,39].

3. The Interaction between Fish Immune System and Microbiota

As previously described, a mucous layer is present, lining the entire lumen of the GIT. This mucous system allows for the development of an immune system, able to enhance innate or adaptive immune responses [40]. The immune system avoids the invasion of opportunistic and pathogenic microorganisms, whilst simultaneously preserving and enhancing the proliferation of commensal bacteria [41,42,43]. The innate immunity represents the main defence line for fish, despite the occurrence of an adaptive immune system, which has a restricted antibody repertoire and slower lymphocyte maturation compared to mammals [44]. The innate immunity is composed of physical barriers, humoral, and cellular components [40]. The physical barriers are characterized by a series of mucosal structures located in the skin, gills, and gut, which are able to prevent pathogen invasion. In the mucosal secretions, it is possible to observe several molecules such as:
  • Antimicrobial peptides (AMPs), which are small peptides (up to 50 amino acids residues). In fish species, it is possible to find a variety of major groups of AMPs such as the piscidins, cathelicidins, defensins, hepcidins, and high-density lipoproteins [44]. AMPs can guarantee general microbiota homeostasis thanks to their antimicrobial activity of immunity modulation [45].
  • Lysozymes, which are other useful components against pathogenic microorganisms. Indeed, these enzymes are able to cleave the glycosidic bond between the N-acetylmuramic acid and N-acetylglucosamine of bacterial cell wall peptidoglycans [46]. In fish, the role of lysozymes against pathogens has been demonstrated in several studies [47,48,49,50], which reported the modulation in their expression and activity versus several fish pathogen bacteria, such as Streptococcus iniae or Vibrio alginolyticus.
  • Complement system, which represents the element able to connect the innate and adaptive immunity. Approximately 30 inactive circulating proteins and membrane-bound receptors belong to this system [51]. The activation of these proteins is related to three different pathways: the classical pathway, where antigen-antibody complexes act as activators; the alternative pathway, where the activation is caused by the presence of several molecules on the surface of microorganisms without the antibodies attendance; and the lectin pathway, where the activation occurs after the mannose-binding lectin binds to the cell-surface carbohydrates of microorganisms [52,53]. Regardless of the three complement pathways, the C3 protein always plays the main role in the function of the complement system. Indeed, the C3 cleavage of C3 generates two protein subunits, namely C3a and C3b. C3b is fundamental because it activates the lytic pathway by attaching itself to the pathogen surface. In addition, C3b permits the sequential connection of C5b, C6, C7, C8, and C9 proteins to form the membrane-attack complex (MAC), able to provoke cell lysis [53].
  • Other useful compounds, such as transferrin, pentraxins, and lectins. These compounds represent further useful weapons to avoid the growth of dangerous bacteria and allow for beneficial microbiota homeostasis [43,44,51].
Among the physical barriers and antimicrobial compounds, several kinds of cells, such as the monocytes/macrophages, neutrophils, dendritic cells, and natural killer cells, represent the cellular component of the innate immune system in fish species. These cells are present in the Gut-Associated Lymphoid Tissue (GALT), where it is also possible to observe the lymphocytes, fundamental in the adaptive immune system. In contrast to mammals, in the fish gut, these cells are located between the epithelium and lamina propria, without a tissue organization [43]. Additionally, the epithelium is able to corroborate to the immune responses, producing specific receptors called Toll-like receptors (TLRs), which are important mediators of inflammatory pathways in the gut, playing a key role to recognize specific microbial patterns [54]. When this pattern is linked to the receptor, the cells start to produce several signal molecules (i.e., tumor necrosis factors (TNF), interleukins, and chemokines) able to guarantee the recruitment of innate immune cells. The innate immune cells (i.e., monocytes/macrophages, neutrophils, dendritic cells, and natural killer cells) use differential mechanisms of action to inhibit microbial proliferation. Indeed, in order to destroy pathogenic organisms, the innate immunity involves the activation of phagocytose (mediated by monocytes/macrophages) and the release of cytotoxic molecules (mediated by natural killer cells) or cytokines for the recruitment of adaptive immune cells [51]. For several years, researchers had supposed that adaptive immune cells did not have a specific role in the microbiota balance; however, more recent studies [55,56] suggested a possible implication in this context, mediated mainly by lymphocytes [57]. Although the role of the B and T lymphocytes seems to be not so different, some studies indicated that T and Z immunoglobulins (Ig) produced by the B lymphocyte are able to guarantee correct microbial colonization [43,58]. It has been reported that IgT is the primary immunoglobulin produced in response to the pathogenic microbiome in the fish gut and prevents any damaging effects of pathogens on the fish [3]. In general, the presence of specific bacteria is required for normal gut development in terms of the proliferation of epithelial cells and maturation of the immune system [43]. As a matter of fact, studies of the gnotobiotic zebrafish have pointed out the relevance of the microbiota in the proliferation of the intestinal epithelial cells by involving MyD88 [59] and in promoting the movement of immune cells to the intestine, such as the migration of neutrophils induced by commensal microorganisms activating serum amyloid A [60]. Additionally, the microbiota is crucial for regulating the neutrophil activity by enhancing the expression of several pro-inflammatory genes [61,62], thus improving infection resistance [43].

4. The Interaction between Fish GIT and Microbiota

The microorganisms that belong to fish microbiota can colonize several parts of the fish body such as the skin, gills, and obviously the gastrointestinal tract. Indeed, although the gut, with its morphology, helps the microbial colonization, other portions of the fish GIT can host different microorganisms, such as bacteria, fungi, and viruses [7,63]. Bacteria represent the most abundant component throughout the fish GIT, with an amount ranging from 107 to 1011 microorganisms per gram of intestinal content [2,12,64]. As a result, the majority of the research studies are primarily concerned with them. As in other animals, the microbiota colonization withstands the influence of different factors. Agents such as the environment (freshwater or saltwater), the feed habits (herbivores, carnivores, omnivores, or detritivores), and GIT shape are among the main ones [3]. The influence of the environment is confirmed by various studies [2,63] that highlighted how the genera Alteromonas, Flavobacterium, Pseudomonas, and Vibrio are more abundant in saltwater fish species, while in freshwater ones, the main microbial genera are represented by Aeromonas, Lactococcus, Pseudomonas, and Clostridium. A comparison of the main microorganisms found in the GIT of freshwater and saltwater fishes is reported in Table 1, together with data about the dominant bacteria stains typically detected in mammals.
As with other species, even among fish, it is possible to distinguish among different feed habits. The diet of a species is the result of specific adaptations that are also reflected in the digestive system, including the occurrence of digestive enzymes. Since a significant part of the digestive enzymes are synthesized by bacteria, the connection between the trophic level of a species and microbiota is not unusual [72]. Indeed, different studies [4,7,73] have documented the prevalence of some bacteria in fish species with similar feed habits [63], as also highlighted by [72], which reported the prevalence of cellulose-degrading bacteria such as Clostridium, Citrobacter, and Leptotrichia in the herbivores, while protease-producing bacteria, such as Cetobacterium and Halomonas, were reported in the carnivorous fish species. A more detailed list with the dominant microbial phyla found in the GIT of fish species of commercial relevance, grouped according to their feed habits, is reported in Table 2.
As described previously, in the fish GIT, is possible to recognize different regions, and each one is characterized by a different microbial density. Indeed, a certain amount of bacteria is detectable along the entire gastrointestinal tract, but some parts of the GIT are more colonized than others. In general, in fish, the bacterial colonization seems to follow an increasing trend from the stomach to the hindgut [63]. Nevertheless, this tendency is also associated with other different situations of constant microbial density along the fish GIT [95], or a reduction in the amount of microbiota from the mouth to the anus [96]. In addition to the previously reported quantitative differences, a wide range of microbial species can be identified in the various parts of the digestive tract in different fish species, as reported in Table 3.
Data reported in the previous published articles allow us to create a sort of “map” detailing one microbial species is more commonly present than another. The foregut, in particular the stomach, is not often subjected to microbiota analysis in comparison to the other regions of the fish GIT because it is thought that the data obtained can be easily affected by the fast movement of the food. Nevertheless, some research studies [7,97,98,99] have focused on the microbiota in the stomach, demonstrating the dominance of the phyla Firmicutes, Proteobacteria, and Actinobacteria in fish species such as Sparus aurata and Epinephelus awoara. These findings may suggest how these microbial phyla could likely adapt to both the low gastric pH and the activity of proteolytic enzymes [99].
In the other regions of the fish GIT, namely the midgut and hindgut, the physico-chemical conditions are more suitable for microbial proliferation than those observed in the foregut, particularly in stomach, and therefore, the bacteria present in the gut differ from those found in this area [105]. In certain cases, even the microbial population detected in the midgut and hindgut might differ, as different bacteria genera, such as Pseudomonas for the midgut and Vibrio for the hindgut, were, for instance, detected in Salmo salar and Gadus morhua [70,90]. Given the natural connection established between the host’s microbiota and its health status [43], the knowledge gained so far regarding the microbial communities offers useful information to optimize strategies of gut microbiota handling with the aim of enhancing the health status of fish, especially in the aquaculture sector [7].

5. Use of Feed Additives for Fish Health Improvement and Gut Efficiency

Over the last decades, the increased development of aquaculture activity has led to a growing attention on the wellness of fish species of commercial relevance. Nevertheless, the productivity growth of the aquaculture sector appears to be challenged by the frequent infections impacting the commercial fish species triggered by pathogens. Among the numerous forms of stressors, bacterial infections are one of the most widespread causes of harm in fish farming, with considerable economic implications [106]. To counteract this serious issue, as in other kinds of rearing, antibiotics are routinely used in the aquaculture sector. However, the extensive use of antibiotics to avoid the onset of diseases in aquaculture facilities may lead to the leakage of these drugs into wastewater, and consequently into rivers and seas [107,108]. Antibiotics, particularly those present in rivers, have the ability to permeate soil and enhance the selection and growth of antibiotic-resistant bacteria [109]. The variety of environmental implications notwithstanding, antibiotic usage can lead to a substantial loss of gut microbiota variability [110]. Generally, the conditions which fish are subjected to in fishery farms might be a cause of stress. Indeed, the continuous maintenance of different environmental factors such as temperature, photoperiod, and salinity in aquaculture plants is not always straightforward, and significant fluctuations in these parameters are a very common occurrence. As a result, the stress exposure in farmed fish may have a negative impact on a variety of physiological mechanisms, including the immune system [111]. Therefore, due to the huge impact that antibiotics have on the environment and health of farmed fish, it is crucial to establish alternative solutions capable of preserving the well being of farmed species and at the same time, reducing the ecological harm [112].
According to numerous recent studies [13,111,112,113,114], feed additives such as prebiotics and probiotics provide a tool capable of boosting aquaculture plant productivity by acting on a variety of ecological and biological aspects while minimizing the antibiotic consumption. Besides being identified as a very good alternative to the usage of antibiotics, the use of these feed additives in aquaculture represents a useful strategy for improving the overall performance of the aquaculture sector since it affects a variety of factors including rearing water quality [115,116], food absorption, and digestion, and has shown a positive effect on the growth performance of fish [117,118,119]. From an immunological perspective, the activity of these feed additives does not merely promote the proliferation of commensal bacteria at the detriment of pathogens. In actuality, it has been demonstrated that prebiotics and probiotics may enhance and improve the immune response of fish species by either increasing the leukocyte amount or the activity of key proteins such as lysozymes, as well as the complement system [120,121,122,123]. More in detail, data from the literature on the administration of specific prebiotics (Table 4) and probiotics (Table 5) acting as effective immunostimulants on aquaculture fish species from seawater and freshwater are listed in the following tables.
Ensuring the health of aquaculture species has an impact on parameters such as nutrient uptake and the growth performance of fish. Growth performance improvement is another relevant target for dietary supplements, since it can lead to a higher output, and hence to higher earnings for the fishery plant. The purpose of new feed formulations is, therefore, to assure an optimum feed absorption and performance of fish growth whilst reducing the rearing costs. In many cases, it seems fundamental to increase the digestive enzymes in order to maximize nutrient absorption. Indeed, numerous studies [117,156,219,220] have shown that the addition of prebiotics and probiotics enhances the nutrient absorption from feed, since they act on a variety of enzymes such as amylases and proteases [119]. However, in other cases, it was also observed that the new diet formulations could have a deleterious impact on the morphological structure of the fish gut, triggering some inflammatory responses [221]. This condition, especially if maintained for a prolonged period, might disrupt the architecture of the intestine, and therefore lead to the upset of the general health of the microbiota and the organism itself. To overcome this situation, the use of prebiotics and probiotics might be helpful. As a matter of fact, several studies [9,222,223] have demonstrated how the intake of prebiotics and probiotics may preserve and, in some cases, improve the morphology and the function of gut, often affected by the rearing conditions. More in detail, data from the literature on the administration of specific prebiotics (Table 6) and probiotics (Table 7) enhancing the gut efficiency and growth performance of aquaculture fish species from seawater and freshwater are listed in the following tables.

6. Use of Histomorphological Assays to Estimate the Quality of Feed Additives

As previously stated, the selection of new feed formulations plays an essential role in the well being of the specimens, and hence, the yield of the aquaculture activity [106]. Nevertheless, the formulation of optimal feeding diets for the wide variety of aquaculture fish species requires the implementation of different types of analyses to verify their impact on the health status of the specimens [251]. Among the different approaches employed [194,252], histomorphological assays represent a good biomarker for the assessment of the welfare of aquatic organisms [253,254,255,256,257] since it is able to promptly provide insights into the overall health status of individuals under examination. In particular, in the histological assays of fish guts, endpoints such as mucosal fold length (villi), muscle thickness (MT), and crypt depth (CD) might provide useful information regarding the efficiency of the gut in terms of nutrient absorption [223], while evaluation of the number of goblet cells and leukocytes may be useful for estimating the state of the immune system response, which is crucial for gut microbiota balance [222]. The evaluation of these endpoints may be performed by using both optical and electron microscopy, which offer useful information for an accurate assessment of the actions of prebiotics and probiotics at the fish-GIT-tissue level, as summarized in Table 8.
For instance, a number of histomorphological parameters were used to assess the impact of lactic acid bacteria and yeast on Oreochromis niloticus [222], including the haematoxylin and eosin (H/E) and periodic acid-Sciff (PAS) staining, which allowed for an observation of a relevant increase in the intestinal perimeter ratio and mucosal fold length (villi) after the probiotic treatment. These kinds of alterations in the fish gut structure may result in a facilitated and increased feed absorption and growth performance [264]. In parallel, the number of goblet cells and leukocytes in treated samples were found to be higher than those in control fish, highlighting the positive effect of the administrated microorganism on the immune system of fish. Similar results were obtained in O. niloticus challenged with the combined treatment of the probiotic P. acidilactici MA18/5M (Bactocell, Lallemand SAS, France) and the prebiotic short-chain fructo-oligosaccharides (FOS), usually termed as symbiotic [265], which enhanced the intestinal structure with increases in the mucosal fold (villi) height and the immune system with elevation of intraepithelial leukocytes, granulocytes, and goblet cells [258]. Notably, the combined administration of three probiotic strains (Bacillus coagulans, B. licheniformis and P. polymyxa) on the northern whiting Sillago sihama Forsskål induced positive effects in terms of gut wellness, resulting in higher uptake of intestinal nutrients and better growth performance, as demonstrated by an evaluation of some histological parameters (i.e., villus height and width, muscle thickness, and crypt depth) and growth and immune response endpoints [223].
The primary aim of these current research studies is to determine the optimal feed additives for each aquaculture species while also considering the dosage [266]. Indeed, the inappropriate usage of certain feed additives may have negative effects in certain circumstances, as reported in gilthead sea bream Sparus aurata, in which the combined intake of inulin and B. subtilis caused oedema and inflammations in the fish gut, as observed by transmission electron microscopy (TEM) [221]. Contrarily, Moroni et al. [9] documented in the same fish species that the probiotic Lactococcus lactis does not provoke any impairments in gut morphology nor inflammation. Therefore, these findings suggest the need to employ the most appropriate feed additive for each individual fish species. Interestingly, the supplementation of nucleotides in the diet of red drum Sciaenops ocellatus was capable of improving several histomorphological parameters in different gut tracts (distal intestine, mid-intestine, proximal intestine, pyloric caeca), enhancing the general health status of the fish and its performance in terms of nutrient absorption [230].
Feed additives can be therefore considered as a useful tool in the design of new food formulae for a more sustainable aquaculture. Indeed, the inclusion of the probiotics Lactobacillus brevis and L. buchneri in the diets of Seriola dumerili, in which fish oil was replaced by vegetable oils, reduced the thickness of submucosa layer of the posterior intestine, thus lowering inflammatory condition [259]. Lactobacillus spp., along with Bacillus spp., are probably among the most widely used probiotics in aquaculture [170,175,267,268]. Moreover, ref. [264] elucidated the ability of the probiotics B. licheniformis and B. subtilis coupled to the yeast S. cerevisiae to enhance several histological parameters such as the number of goblet and mucosal cells, and villus length and width. Interestingly, ref. [261] documented that although the individual use of the commercial probiotic Aquablend® had a significant effect on fold length in the proximal section of the GIT in Totoaba macdonaldi, its use in combination with the prebiotic GroBiotic®-A showed an increase in the values of these parameters, though not significantly, as a signal that the formulation used needs to be revised.
To accurately integrate and interpret the histological results, the use of a continuous scale scoring system may be helpful, as applied and reported in [262] for the evaluation of a number of histological parameters (i.e., intestinal folds, changes in enterocytes nucleous, supranuclear absorptive vacuolisation, connective tissue hyperplasia in the lamina propria and submucosa, infiltration of inflammatory intraepithelial leukocytes), that allowed for an understanding of the inefficiency of prebiotics (FOS and xylo-oligosaccharides) to counteract the negative impact provoked by diets including plant feedstuffs in the distal intestine of juveniles D. labrax.
Besides enhancing the general wellness of aquaculture fish species, feed additives (prebiotics and probiotics) play a pivotal role in challenging infections, providing a valid alternative to the use of antibiotics that may lead to antibiotic resistance after a prolonged period of time [175,256,257,269]. In [175], the efficiency of three Bacillus species in O. niloticus against A. hydrophila infection was assessed, resulting in improvements in some histological parameters of the fish gut (i.e., villus length and width, goblet cells count, intestinal epithelial muscle thickness). Noteworthy, the rise in the number of goblet cells is relevant to challenging infection, as these cells secrete mucus-containing bactericidal compounds which are useful against pathogens [115]. Therefore, this histological parameter, combined with other endpoints such as mortality rate, could provide relevant information on the immunostimulant and pathogen resistance effects of the tested feed additives. A further relevant histological parameter to estimate the pathogen resistance effects of feed additives is the level of intraepithelial leukocytes, as reported by [172,263] after using the commercial probiotic AquaStar® Growout (L. reuteri, B. subtilis, E. faecium and P. acidilactici) in tilapia O. niloticus. Finally, the evaluation of microvilli density by TEM, besides providing information on the absorptive surface area index (combined with numerical data of microvilli length and perimeter ratio), may also reveal tight junctions between enterocytes, and thus, the occurrence of a barrier against pathogens [263]. Therefore, these results point out the potential of the histological approach for obtaining a variety of precious information to delineate the gut health status of fish species treated with feed additives.

7. Conclusions

In this review, the close relationship between the microbiota and the GIT of fish is highlighted. The occurrence of different environmental conditions (freshwater and seawater) and dietary habits (herbivores, carnivores, omnivores, detritivores) are able to influence this ratio, resulting in changes both in terms of microbial colonization and histomorphological patterns, with repercussions in fish GIT efficiency. Therefore, the use of histological endpoints to assess the general health status of aquaculture fish species treated with feed additives (prebiotics and probiotics) represents a very useful strategy for understanding the capacity of these treatments to promote the interface between the microbiota and the gut, which is crucial for its proper performance in terms of nutrient uptake and defense against pathogens. Overall, an in-depth knowledge of the appropriate administration of feed supplements for individual fish species is a promising strategy to greatly enhance the aquaculture sector, both in terms of quality and sustainable production. Therefore, this review has the potential to provide directions for future research in this field.

Author Contributions

Conceptualization, T.C. and M.M.; data curation, G.D.M., T.C. and M.M.; writing—original draft preparation, G.D.M.; writing—review and editing, T.C. and M.M.; visualization, T.C. and G.D.M.; supervision, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

G.D.M. received a scholarship within the project “FISH PATH NET—Enhancement of Sicilian ichthyopathological centres”, funded by PO FEAMP 2014/2020.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ringø, E.; Zhou, Z.; Vecino, J.L.G.; Wadsworth, S.; Romero, J.; Krogdahl, Å.; Olsen, R.E.; Dimitroglou, A.; Foey, A.; Davies, S.; et al. Effect of dietary components on the gut microbiota of aquatic animals. A never-ending story? Aquac. Nutr. 2016, 22, 219–282. [Google Scholar] [CrossRef]
  2. Pérez, T.; Balcázar, J.L.; Ruiz-Zarzuela, I.; Halaihel, N.; Vendrell, D.; de Blas, I.; Múzquiz, J.L. Host–microbiota interactions within the fish intestinal ecosystem. Mucosal Immunol. 2010, 3, 355–360. [Google Scholar] [CrossRef] [PubMed]
  3. Diwan, A.D.; Harke, S.N.; Panche, A.N. Host-microbiome interaction in fish and shellfish: An overview. Fish Shellfish Immunol. Rep. 2023, 4, 100091. [Google Scholar] [CrossRef] [PubMed]
  4. Clements, K.D.; Angert, E.R.; Montgomery, W.L.; Choat, J.H. Intestinal microbiota in fishes: What’s known and what’s not. Mol. Ecol. 2014, 23, 1891–1898. [Google Scholar] [CrossRef]
  5. Ray, A.K.; Ringø, E. The Gastrointestinal Tract of Fish. In Aquaculture Nutrition: Gut Health, Probiotcs and Prebiotic; Merrifield, D., Ringø, E., Eds.; Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2014. [Google Scholar]
  6. Becker, A.G.; Gonçalves, J.F.; Oliveira Garcia, L.; Behr, E.R.; Graça, D.L.; Kurtz Filho, M.; Martins, T.; Baldisserotto, B. Morphometric parameters comparisons of the digestive tract of four teleosts with different feeding habits. Biol. Cienc. Rural 2010, 40, 862–866. [Google Scholar] [CrossRef]
  7. Egerton, S.; Culloty, S.; Whooley, J.; Stanton, C.; Ross, R.P. The gut microbiota of marine fish. Front. Microbiol. 2018, 9, 873. [Google Scholar] [CrossRef]
  8. Deng, M.; Yu, Z.; Geng, Y.; Wang, K.; Chen, D.; Huang, X.; Ou, Y.; Chen, Z.; Zhong, Z.; Lai, W. Outbreaks of Streptococcosis associated with Streptococcus iniae in Siberian sturgeon (Acipenser baerii) in China. Aquac. Res. 2017, 48, 909–919. [Google Scholar] [CrossRef]
  9. Moroni, F.; Naya-Català, F.; Piazzon, M.C.; Rimoldi, S.; Calduch-Giner, J.; Giardini, A.; Martínez, I.; Brambilla, F.; Pérez-Sánchez, J.; Terova, G. The effects of nisin-producing Lactococcus lactis strain used as probiotic on gilthead sea bream (Sparus aurata) growth, gut microbiota, and transcriptional response. Front. Mar. Sci. 2021, 8, 659519. [Google Scholar] [CrossRef]
  10. Food and Agricultural Organization of the United Nations; World Health Organization. Health and Nutrition Properties of Probiotics in Food including Powder Milk with Live Lactic Acid Bacteria. In Proceedings of the Report of a Joint FAO/WHO Expert Consultation on Evaluation of Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Acid Bacteria, Amerian Córdoba Park Hotel, Cordoba, Argentina, 1–4 October 2001. [Google Scholar]
  11. Nayak, S.K. Probiotics and immunity: A fish perspective. Fish Shellfish Immunol. 2010, 29, 2–14. [Google Scholar] [CrossRef]
  12. Nayak, S.K. Role of gastrointestinal microbiota in fish. Aquac. Res. 2010, 41, 1553–1573. [Google Scholar] [CrossRef]
  13. Akhter, N.; Wu, B.; Memon, A.M.; Mohsin, M. Probiotics and prebiotics associated with aquaculture: A review. Fish Shellfish Immunol. 2015, 45, 733–741. [Google Scholar] [CrossRef]
  14. Wee, W.; Hamid, N.K.A.; Mat, K.; Khalif, R.I.A.; Rusli, N.D.; Rahman, M.M.; Kabir, M.A.; Wei, L.S. The effects of mixed prebiotics in aquaculture: A review. Aquac. Fish. 2022; in press. [Google Scholar] [CrossRef]
  15. 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]
  16. Hancz, C. Application of probiotics for environmentally friendly and sustainable aquaculture: A Review. Sustainability 2022, 14, 15479. [Google Scholar] [CrossRef]
  17. Gosavi, S.M.; Verma, C.R.; Kharat, S.S.; Pise, M.; Kumkar, P. Structural adequacy of the digestive tract supports dual feeding habit in catfish Pachypterus khavalchor (Siluriformes: Horabagridae). Acta Histochem. 2019, 121, 437–449. [Google Scholar] [CrossRef]
  18. Takahashi, T.; Koblmüller, S. The adaptive radiation of cichlid fish in Lake Tanganyika: A morphological perspective. Int. J. Evol. Biol. 2011, 2011, 620754. [Google Scholar] [CrossRef] [PubMed]
  19. Wainwright, P.C. Functional morphology of the pharyngeal jaw apparatus. Fish Physiol. 2005, 23, 77–101. [Google Scholar]
  20. Kalhoro, H.; Tong, S.; Wang, L.; Hua, Y.; Volatiana, J.A.; Shao, Q. Morphological study of the gastrointestinal tract of Larimichthys crocea (Acanthopterygii: Perciformes). Zoologia 2018, 35, e25171. [Google Scholar] [CrossRef]
  21. Wilson, J.M.; Castro, L.F.C. Morphological Diversity of The Gastrointestinal Tract in Fishes. In The Multifunctional Gut of Fish; Grosell, M., Farrell, A.P., Brauner, C.J.B.T.-F.P., Eds.; Academic Press: Cambridge, MA, USA, 2010; pp. 1–55. [Google Scholar]
  22. Okuthe, G.E.; Bhomela, B. Morphology, histology and histochemistry of the digestive tract of the Banded tilapia, Tilapia sparrmanii (Perciformes: Cichlidae). Zoologia 2021, 37, e51043. [Google Scholar] [CrossRef]
  23. Abdulhadi, H.A. Some comparative histological studies on alimentary tract of Tilapia fish (Tilapia spilurus) and sea bream (Mylio cuvieri). Egypt. J. Aquat. Res. 2015, 31, 387–398. [Google Scholar]
  24. Alves, A.P.C.; Pereira, R.T.; Rosa, P.V. Morphology of the digestive system in carnivorous freshwater dourado Salminus brasiliensis. J. Fish Biol. 2021, 99, 1222–1235. [Google Scholar] [CrossRef]
  25. Mello, G.C.G.; Santos, M.L.; Arantes, F.P.; Pessali, T.C.; Brito, M.F.G.; Santos, J.E. Morphological characterisation of the digestive tract of the catfish Lophiosilurus alexandri Steindachner, 1876 (Siluriformes, Pseudopimelodidae). Acta Zool. 2019, 100, 14–23. [Google Scholar] [CrossRef]
  26. Flores, E.M.; Nguyen, A.T.; Odem, M.A.; Eisenhoffer, G.T.; Krachler, A.M. The zebrafish as a model for gastrointestinal tract–microbe interactions. Cell. Microbiol. 2020, 22, e13152. [Google Scholar] [CrossRef] [PubMed]
  27. Stevens, C.E.; Hume, I.D. Comparative Physiology of the Vertebrate Digestive System; Cambridge University Press: Cambridge, UK, 2004. [Google Scholar]
  28. Johnson, K.S.; Clements, K.D. Histology and ultrastructure of the gastrointestinal tract in four temperate marine herbivorous fishes. J. Morphol. 2022, 283, 16–34. [Google Scholar] [CrossRef] [PubMed]
  29. Buddington, R.K.; Diamond, J.M. Pyloric ceca of fish: A “new” absorptive organ. Am. J. Physiol. Liver Physiol. 1987, 252, G65–G76. [Google Scholar] [CrossRef]
  30. Farrag, M.G.; Azab, D.M.; Alabssawy, A.N. Comparative study on the histochemical structures of stomach, pyloric caeca and anterior intestine in the grey mullet, Mugil cephalus (Linnaeus, 1758). Egypt. J. Aquat. Biol. Fish. 2020, 24, 1055–1071. [Google Scholar] [CrossRef]
  31. Olsen, R.; Ringø, E. Lipid digestibility in fish: A review. Recent Res. Dev. Lipid Res. 1997, 1, 199–265. [Google Scholar]
  32. Ringø, E.; Olsen, R.E.; Mayhew, T.M.; Myklebust, R. Electron microscopy of the intestinal microflora of fish. Aquaculture 2003, 227, 395–415. [Google Scholar] [CrossRef]
  33. Banan Khojasteh, S.M. The morphology of the post-gastric alimentary canal in teleost fishes: A brief review. Int. J. Aquat. Sci. 2012, 3, 71–88. [Google Scholar]
  34. Kapoor, B.G.; Smit, H.; Verighina, I.A. The Alimentary Canal and Digestion in Teleosts; Russell, F.S., Yonge, M.B.T.-A., Eds.; Academic Press: Cambridge, MA, USA, 1976; pp. 109–239. [Google Scholar]
  35. Holmgren, S.; Olsson, C.B.T.-F.P. The neuronal and endocrine regulation of gut function. Fish Physiol. 2009, 28, 467–512. [Google Scholar]
  36. Manera, M.; Dezfuli, B.S. Rodlet cells in teleosts: A new insight into their nature and functions. J. Fish Biol. 2004, 65, 597–619. [Google Scholar] [CrossRef]
  37. Reite, O.B.; Evensen, O. Inflammatory cells of teleostean fish: A review focusing on mast cells/eosinophilic granule cells and rodlet cells. Fish Shellfish Immunol. 2006, 20, 192–208. [Google Scholar] [CrossRef] [PubMed]
  38. Cho, J.-H.; Park, J.W.; Ryu, Y.-W.; Kim, K.-W.; Hur, S.-W. Morphology, histology, and histochemistry of the digestive tract of the marbled flounder Pseudopleuronectes yokohamae. Animals 2023, 13, 936. [Google Scholar] [CrossRef] [PubMed]
  39. Alabssawy, A.N.; Khalaf-Allah, H.M.M.; Gafar, A.A. Anatomical and histological adaptations of digestive tract in relation to food and feeding habits of lizardfish, Synodus variegatus (Lacepède, 1803). Egypt. J. Aquat. Res. 2019, 45, 159–165. [Google Scholar] [CrossRef]
  40. Rauta, P.R.; Nayak, B.; Das, S. Immune system and immune responses in fish and their role in comparative immunity study: A model for higher organisms. Immunol. Lett. 2012, 148, 23–33. [Google Scholar] [CrossRef]
  41. Gomez, D.; Sunyer, J.O.; Salinas, I. The mucosal immune system of fish: The evolution of tolerating commensals while fighting pathogens. Fish Shellfish Immunol. 2013, 35, 1729–1739. [Google Scholar] [CrossRef]
  42. Kelly, C.; Salinas, I. Under pressure: Interactions between commensal microbiota and the teleost immune system. Front. Immunol. 2017, 8, 559. [Google Scholar] [CrossRef]
  43. Yu, Y.-Y.; Ding, L.-G.; Huang, Z.-Y.; Xu, H.-Y.; Xu, Z. Commensal bacteria-immunity crosstalk shapes mucosal homeostasis in teleost fish. Rev. Aquac. 2021, 13, 2322–2343. [Google Scholar] [CrossRef]
  44. Valero, Y.; Saraiva-Fraga, M.; Costas, B.; Guardiola, F.A. Antimicrobial peptides from fish: Beyond the fight against pathogens. Rev. Aquac. 2020, 12, 224–253. [Google Scholar] [CrossRef]
  45. Valero, Y.; Chaves-Pozo, E.; Meseguer, J.; Esteban, M.; Cuesta, A. Biologial Role of Fish Antimicrobial Peptides. In Antimicrobial Peptides: Properties, Functions and Role in Immune Response; Seong, M.D., Hak, Y.I., Eds.; Nova Science Publisher: Hauppauge, NY, USA, 2013; pp. 31–60. [Google Scholar]
  46. Vermassen, A.; Leroy, S.; Talon, R.; Provot, C.; Popowska, M.; Desvaux, M. Cell wall hydrolases in bacteria: Insight on the diversity of cell wall amidases, glycosidases and peptidases toward peptidoglycan. Front. Microbiol. 2019, 10, 331. [Google Scholar] [CrossRef]
  47. Gao, C.; Fu, Q.; Zhou, S.; Song, L.; Ren, Y.; Dong, X.; Su, B.; Li, C. The mucosal expression signatures of g-type lysozyme in turbot (Scophthalmus maximus) following bacterial challenge. Fish Shellfish Immunol. 2016, 54, 612–619. [Google Scholar] [CrossRef]
  48. Li, S.; Wang, D.; Liu, H.; Yin, J.; Lu, T. Expression and antimicrobial activity of c-type lysozyme in taimen (Hucho taimen, Pallas). Dev. Comp. Immunol. 2016, 63, 156–162. [Google Scholar] [CrossRef]
  49. Wei, S.; Huang, Y.; Huang, X.; Cai, J.; Wei, J.; Li, P.; Ouyang, Z.; Qin, Q. Molecular cloning and characterization of a new G-type lysozyme gene (Ec-lysG) in orange-spotted grouper, Epinephelus coioides. Dev. Comp. Immunol. 2014, 46, 401–412. [Google Scholar] [CrossRef]
  50. Yu, L.; Sun, B.; Li, J.; Sun, L. Characterization of a c-type lysozyme of Scophthalmus maximus: Expression, activity, and antibacterial effect. Fish Shellfish Immunol. 2013, 34, 46–54. [Google Scholar] [CrossRef] [PubMed]
  51. Smith, N.C.; Rise, M.L.; Christian, S.L. A comparison of the innate and adaptive immune systems in cartilaginous fish, ray-finned fish, and lobe-finned fish. Front. Immunol. 2019, 10, 2292. [Google Scholar] [CrossRef]
  52. Abram, Q.H.; Dixon, B.; Katzenback, B.A. Impacts of low temperature on the teleost immune system. Biology 2017, 6, 39. [Google Scholar] [CrossRef] [PubMed]
  53. Natnan, M.E.; Low, C.-F.; Chong, C.-M.; Bunawan, H.; Baharum, S.N. Integration of omics tools for understanding the fish immune response due to microbial challenge. Front. Mar. Sci. 2021, 8, 668771. [Google Scholar] [CrossRef]
  54. Sahoo, B.R. Structure of fish Toll-like receptors (TLR) and NOD-like receptors (NLR). Int. J. Biol. Macromol. 2020, 161, 1602–1617. [Google Scholar] [CrossRef] [PubMed]
  55. Nutsch, K.M.; Hsieh, C.-S. T cell tolerance and immunity to commensal bacteria. Curr. Opin. Immunol. 2012, 24, 385–391. [Google Scholar] [CrossRef] [PubMed]
  56. Yu, Y.-Y.; Kong, W.-G.; Xu, H.-Y.; Huang, Z.-Y.; Zhang, X.-T.; Ding, L.-G.; Dong, S.; Yin, G.-M.; Dong, F.; Yu, W.; et al. Convergent evolution of mucosal immune responses at the buccal cavity of teleost fish. iScience 2019, 19, 821–835. [Google Scholar] [CrossRef]
  57. Dawood, M.A.O. Nutritional immunity of fish intestines: Important insights for sustainable aquaculture. Rev. Aquac. 2021, 13, 642–663. [Google Scholar] [CrossRef]
  58. Bilal, S.; Etayo, A.; Hordvik, I. Immunoglobulins in teleosts. Immunogenetics 2021, 73, 65–77. [Google Scholar] [CrossRef] [PubMed]
  59. Cheesman, S.E.; Neal, J.T.; Mittge, E.; Seredick, B.M.; Guillemin, K. Epithelial cell proliferation in the developing zebrafish intestine is regulated by the Wnt pathway and microbial signaling via Myd88. Proc. Natl. Acad. Sci. USA 2011, 108, 4570–4577. [Google Scholar] [CrossRef] [PubMed]
  60. Kanther, M.; Tomkovich, S.; Xiaolun, S.; Grosser, M.R.; Koo, J.; Flynn, E.J., III; Jobin, C.; Rawls, J.F. Commensal microbiota stimulate systemic neutrophil migration through induction of serum amyloid A. Cell. Microbiol. 2014, 16, 1053–1067. [Google Scholar] [CrossRef] [PubMed]
  61. Galindo-Villegas, J.; García-Moreno, D.; De Oliveira, S.; Meseguer, J.; Mulero, V. Regulation of immunity and disease resistance by commensal microbes and chromatin modifications during zebrafish development. Proc. Natl. Acad. Sci. USA 2012, 109, E2605–E2614. [Google Scholar] [CrossRef] [PubMed]
  62. Rolig, A.S.; Parthasarathy, R.; Burns, A.R.; Bohannan, B.J.M.; Guillemin, K. Individual members of the microbiota disproportionately modulate host innate immune responses. Cell Host Microb. 2015, 18, 613–620. [Google Scholar] [CrossRef]
  63. 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]
  64. Rombout, J.H.W.M.; Abelli, L.; Picchietti, S.; Scapigliati, G.; Kiron, V. Teleost intestinal immunology. Fish Shellfish Immunol. 2011, 31, 616–626. [Google Scholar] [CrossRef]
  65. Fujimura, K.E.; Slusher, N.A.; Cabana, M.D.; Lynch, S.V. Role of the gut microbiota in defining human health. Expert Rev. Anti. Infect. Ther. 2010, 8, 435–454. [Google Scholar] [CrossRef]
  66. Hsiao, W.W.; Metz, C.; Singh, D.P.; Roth, J. The microbes of the intestine: An introduction to their metabolic and signaling capabilities. Endocrinol. Metab. Clin. N. Am. 2008, 37, 857–871. [Google Scholar] [CrossRef]
  67. Rajilic-Stojanovic, M.; Smidt, H.; de Vos, W.M. Diversity of the human gastrointestinal tract microbiota revisited. Environ. Microbiol. 2007, 9, 2125–2136. [Google Scholar] [CrossRef] [PubMed]
  68. Carda-Dieguez, M.; Mira, A.; Fouz, B. Pyrosequencing survey of intestinal microbiota diversity in cultured sea bass (Dicentrarchus labrax) fed functional diets. FEMS Microbiol. Ecol. 2014, 87, 451–459. [Google Scholar] [CrossRef] [PubMed]
  69. Desai, A.R.; Links, M.G.; Collins, S.A.; Mansfield, G.S.; Drew, M.D.; Van Kessel, A.G.; Hill, J.E. Effects of plant-based diets on the distal gut microbiome of rainbow trout (Oncorhynchus mykiss). Aquaculture 2012, 350, 134–142. [Google Scholar] [CrossRef]
  70. Ringø, E.; Sperstad, S.; Myklebust, R.; Refstie, S.; Krogdahl, Å. Characterisation of the microbiota associated with intestine of Atlantic cod (Gadus morhua L.): The effect of fish meal, standard soybean meal and a bioprocessed soybean meal. Aquaculture 2006, 261, 829–841. [Google Scholar] [CrossRef]
  71. Verner-Jeffreys, D.W.; Shields, R.J.; Bricknell, I.R.; Birkbeck, T.H. Changes in the gut-associated microflora during the development of Atlantic halibut (Hippoglossus hippoglossus L.) larvae in three British hatcheries. Aquaculture 2003, 219, 21–42. [Google Scholar] [CrossRef]
  72. Liu, H.; Guo, X.; Gooneratne, R.; Lai, R.; Zeng, C.; Zhan, F.; Wang, W. The gut microbiome and degradation enzyme activity of wild freshwater fishes influenced by their trophic levels. Sci. Rep. 2016, 6, 24340. [Google Scholar] [CrossRef] [PubMed]
  73. Parris, D.J.; Brooker, R.M.; Morgan, M.A.; Dixson, D.L.; Stewart, F.J. Whole gut microbiome composition of damselfish and cardinalfish before and after reef settlement. PeerJ 2016, 4, e2412. [Google Scholar] [CrossRef]
  74. Smriga, S.; Sandin, S.A.; Azam, F. Abundance, diversity, and activity of microbial assemblages associated with coral reef fish guts and feces. FEMS Microbiol. Ecol. 2010, 73, 31–42. [Google Scholar] [CrossRef]
  75. Miyake, S.; Ngugi, D.K.; Stingl, U. Diet strongly influences the gut microbiota of surgeonfishes. Molecul. Ecol. 2015, 24, 656–672. [Google Scholar] [CrossRef]
  76. Clements, K.D.; Pasch, I.B.; Moran, D.; Turner, S.J. Clostridia dominate 16S rRNA gene libraries prepared from the hindgut of temperate marine herbivorous fishes. Mar. Biol. 2007, 150, 1431–1440. [Google Scholar] [CrossRef]
  77. Fidopiastis, P.M.; Bezdek, D.J.; Horn, M.H.; Kandel, J.S. Characterizing the resident, fermentative microbial consortium in the hindgut of the temperate-zone herbivorous fish, Hermosilla azurea (Teleostei: Kyphosidae). Mar. Biol. 2006, 148, 631–642. [Google Scholar] [CrossRef]
  78. Sparagon, W.J.; Gentry, E.C.; Minich, J.J.; Vollbrecht, L.; Laurens, L.M.L.; Allen, E.E.; Sims, N.A.; Dorrestein, P.C.; Wegley, K.L.; Nelson, C.E. Fine scale transitions of the microbiota and metabolome along the gastrointestinal tract of herbivorous fishes. Anim. Microbiome 2022, 4, 33. [Google Scholar] [CrossRef] [PubMed]
  79. Li, J.; Ni, J.; Li, J.; Wang, C.; Li, X.; Wu, S.; Zhang, T.; Yu, Y.; Yan, Q. Comparative study on gastrointestinal microbiota of eight fish species with different feeding habits. J. Appl. Microbiol. 2014, 117, 1750–1760. [Google Scholar] [CrossRef]
  80. Geraylou, Z.; Souffreau, C.; Rurangwa, E.; Maes, G.E.; Spanier, K.I.; Courtin, C.M.; Delcour, J.A.; Buyse, J.; Ollevier, F. Prebiotic effects of arabinoxylan oligosaccharides on juvenile Siberian sturgeon (Acipenser baerii) with emphasis on the modulation of the gut microbiota using 454 pyrosequencing. FEMS Microbiol. Ecol. 2013, 86, 357–371. [Google Scholar] [CrossRef]
  81. Ward, N.L.; Steven, B.; Penn, K.; Methe, B.A.; Detrich, W.H. Characterization of the intestinal microbiota of two Antarctic notothenioid fish species. Extremophiles 2009, 13, 679–685. [Google Scholar] [CrossRef] [PubMed]
  82. Ransom, B.L. Intestinal Microbial Community Composition of Six Actinopterygii Fish Species in The Southeastern United States; University of Georgia: Athens, GA, USA, 2008. [Google Scholar]
  83. Sun, Y.; Yang, H.; Ling, Z.; Chang, J.; Ye, J. Gut microbiota of fast and slow growing grouper Epinephelus coioides. Afric. J. Microbiol. Res. 2009, 3, 637–640. [Google Scholar]
  84. Sugita, H.; Iwata, J.; Miyajima, C.; Kubo, T.; Noguchi, T.; Hashimoto, K.; Deguchi, Y. Changes in microflora of a puffer fish Fugu niphobles, with different water temperatures. Mar. Biol. 1989, 101, 299–304. [Google Scholar] [CrossRef]
  85. Star, B.; Haverkamp, T.H.; Jentoft, S.; Jakobsen, K.S. Next generation sequencing shows high variation of the intestinal microbial species composition in Atlantic cod caught at a single location. BMC Microbiol. 2013, 13, 248. [Google Scholar] [CrossRef]
  86. MacFariane, R.D.; McLaughlin, J.J.; Bullock, G.L. Quantitative and qualitative studies of gut flora in striped bass from estuarine and coastal marine environments. J. Wildl. Dis. 1986, 22, 344–348. [Google Scholar] [CrossRef]
  87. Newman Jr, J.T.; Cosenza, B.J.; Buck, J.D. Aerobic microflora of the bluefish (Pomatomus saltatrix) intestine. J. Fish. Res. Board Can. 1972, 29, 333–336. [Google Scholar] [CrossRef]
  88. Floris, R.; Manca, S.; Fois, N. Microbial ecology of intestinal tract of gilthead sea bream (Sparus aurata Linnaeus, 1758) from two coastal lagoons of Sardinia (Italy). Transitional Waters Bull. 2013, 7, 4–12. [Google Scholar]
  89. Skrodenytė-Arbačiauskienė, V.; Sruoga, A.; Butkauskas, D.; Skrupskelis, K. Phylogenetic analysis of intestinal bacteria of freshwater salmon Salmo salar and sea trout Salmo trutta trutta and diet. Fish. Sci. 2008, 74, 1307–1314. [Google Scholar] [CrossRef]
  90. Hovda, M.B.; Lunestad, B.T.; Fontanillas, R.; Rosnes, J.T. Molecular characterization of the intestinal microbiota of farmed Atlantic salmon (Salmo salar L.). Aquaculture 2007, 272, 581–588. [Google Scholar] [CrossRef]
  91. Bano, N.; DeRae Smith, A.; Bennett, W.; Vasquez, L.; Hollibaugh, J.T. Dominance of mycoplasma in the guts of the long-jawed mudsucker, Gillichthys mirabilis, from five California salt marshes. Environ. Microbiol. 2007, 9, 2636–2641. [Google Scholar] [CrossRef] [PubMed]
  92. Curson, A.R.; Sullivan, M.J.; Todd, J.D.M.; Johnston, A.W. Identification of genes for dimethyl sulfide production in bacteria in the gut of Atlantic Herring (Clupea harengus). ISME J. 2010, 4, 144–146. [Google Scholar] [CrossRef] [PubMed]
  93. Karthiayani, T.; Mahadeva Iyer, K. Quantitative and qualitative studies on the bacterial flora of fresh sardines. Fish. Technol. 1967, 4, 89–97. [Google Scholar]
  94. Svanevik, C.S.; Lunestad, B.T. Characterization of the microbiota of Atlantic mackerel (Scomber scombrus). Int. J. Food Microbiol. 2011, 151, 164–170. [Google Scholar] [CrossRef]
  95. Ringø, E.; Strøm, E. Microflora of Arctic charr, Salvelinus alpinus (L.): Gastrointestinal microflora of free-living fish and effect of diet and salinity on intestinal microflora. Aquac. Res. 1994, 25, 623–629. [Google Scholar] [CrossRef]
  96. Austin, B.; Al-Zahrani, A.M.J. The effect of antimicrobial compounds on the gastrointestinal microflora of rainbow trout, Salmo gairdneri Richardson. J. Fish Biol. 1988, 33, tb05444. [Google Scholar] [CrossRef]
  97. Silva, F.C.P.; Nicoli, J.R.; Zambonino-Infante, J.L.; Kaushik, S.; Gatesoupe, F.-J. Influence of the diet on the microbial diversity of faecal and gastrointestinal contents in gilthead sea bream (Sparus aurata) and intestinal contents in goldfish (Carassius auratus). FEMS Microbiol. Ecol. 2011, 78, 285–296. [Google Scholar] [CrossRef]
  98. Estruch, G.; Collado, M.C.; Peñaranda, D.S.; Tomás Vidal, A.; Jover Cerdá, M.; Pérez Martínez, G.; Martinez-Llorens, S. Impact of fishmeal replacement in diets for gilthead sea bream (Sparus aurata) on the gastrointestinal microbiota determined by pyrosequencing the 16S rRNA Gene. PLoS ONE 2015, 10, e0136389. [Google Scholar] [CrossRef]
  99. Zhou, Z.; Liu, Y.; Shi, P.; He, S.; Yao, B.; Ringø, E. Molecular characterization of the autochthonous microbiota in the gastrointestinal tract of adult yellow grouper (Epinephelus awoara) cultured in cages. Aquaculture 2009, 286, 184–189. [Google Scholar] [CrossRef]
  100. Xing, M.; Hou, Z.; Yuan, J.; Liu, Y.; Qu, Y.; Liu, B. Taxonomic and functional metagenomic profiling of gastrointestinal tract microbiome of the farmed adult turbot (Scophthalmus maximus). FEMS Microbiol. Ecol. 2013, 86, 432–443. [Google Scholar] [CrossRef]
  101. Parshukov, A.N.; Kashinskaya, E.N.; Simonov, E.P.; Hlunov, O.V.; Izvekova, G.I.; Andree, K.B.; Solovyev, M.M. The isolation of the intestinal gut microbiota of farmed rainbow trout, Oncorhynchus mykiss (Walbaum), depending on the infection status of the fish. Appl. Microbiol. 2019, 127, 379–395. [Google Scholar] [CrossRef] [PubMed]
  102. Wu, S.; Tian, J.; Wang, G.; Li, W.; Zou, H. Characterization of bacterial community in the stomach of yellow catfish (Pelteobagrus fulvidraco). World J. Microbiol. Biotechnol. 2012, 28, 2165–2174. [Google Scholar] [CrossRef] [PubMed]
  103. Zhao, N.; Guo, J.; Zhang, B.; Liu, K.; Liu, Y.; Shen, Y.; Li, J. Heterogeneity of the tissue-specific mucosal microbiome of normal grass carp (Ctenopharyngodon idella). Mar. Biotechnol. 2022, 24, 366–379. [Google Scholar] [CrossRef] [PubMed]
  104. Semova, I.; Carten, J.D.; Stombaugh, J.; Mackey, L.C.; Knight, R.; Farber, S.A.; Rawls, J.F. Microbiota regulate intestinal absorption and metabolism of fatty acids in the zebrafish. Cell Host Microbe 2012, 12, 277–288. [Google Scholar] [CrossRef]
  105. Zhou, Z.; Shi, P.; He, S.; Liu, Y.; Huang, G.; Yao, B.; Ringø, E. Identification of adherent microbiota in the stomach and intestine of emperor red snapper (Lutjanus sebae Cuvier) using 16S rDNA-DGGE. Aquac. Res. 2009, 40, 1213–1218. [Google Scholar] [CrossRef]
  106. Amenyogbe, E.; Chen, G.; Wang, Z.; Huang, J.; Huang, B.; Li, H. The exploitation of probiotics, prebiotics and synbiotics in aquaculture: Present study, limitations and future directions. A review. Aquac. Int. 2020, 28, 1017–1041. [Google Scholar] [CrossRef]
  107. Chen, J.; Xie, S. Overview of sulfonamide biodegradation and the relevant pathways and microorganisms. Sci. Total Environ. 2018, 640–641, 1465–1477. [Google Scholar] [CrossRef]
  108. Zhang, Q.-Q.; Ying, G.-G.; Pan, C.-G.; Liu, Y.-S.; Zhao, J.-L. Comprehensive evaluation of antibiotics emission and fate in the river basins of China: Source analysis, multimedia modeling, and linkage to bacterial resistance. Environ. Sci. Technol. 2015, 49, 6772–6782. [Google Scholar] [CrossRef] [PubMed]
  109. Thiele-Bruhn, S. Pharmaceutical antibiotic compounds in soils—A review. J. Plant Nutr. Soil Sci. 2003, 166, 145–167. [Google Scholar] [CrossRef]
  110. 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]
  111. Yukgehnaish, K.M.; 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]
  112. Van Doan, H.; Hoseinifar, S.H.; Ringø, E.; Ángeles Esteban, M.; Dadar, M.; Dawood, M.A.O.; Faggio, C. Host-associated probiotics: A key factor in sustainable aquaculture. Rev. Fish. Sci. Aquac. 2020, 28, 16–42. [Google Scholar] [CrossRef]
  113. 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]
  114. Chauhan, A.; Singh, R. Probiotics in aquaculture: A promising emerging alternative approach. Symbiosis 2019, 77, 99–113. [Google Scholar] [CrossRef]
  115. Elsabagh, M.; Mohamed, R.; Moustafa, E.M.; Hamza, A.; Farrag, F.; Decamp, O.; Dawood, M.A.O.; Eltholth, M. Assessing the impact of Bacillus strains mixture probiotic on water quality, growth performance, blood profile and intestinal morphology of Nile tilapia, Oreochromis niloticus. Aquac. Nutr. 2018, 24, 1613–1622. [Google Scholar] [CrossRef]
  116. Wang, Y.-B.; Tian, Z.-Q.; Yao, J.-T.; Li, W. Effect of probiotics, Enteroccus faecium, on tilapia (Oreochromis niloticus) growth performance and immune response. Aquaculture 2008, 277, 203–207. [Google Scholar] [CrossRef]
  117. Makled, S.O.; Hamdan, A.M.; El-Sayed, A.-F.M.; Hafez, E.E. Evaluation of marine psychrophile, Psychrobacter namhaensis SO89, as a probiotic in Nile tilapia (Oreochromis niloticus) diets. Fish Shellfish Immunol. 2017, 61, 194–200. [Google Scholar] [CrossRef]
  118. Soleimani, N.; Hoseinifar, S.H.; Merrifield, D.L.; Barati, M.; Abadi, Z.H. Dietary supplementation of fructooligosaccharide (FOS) improves the innate immune response, stress resistance, digestive enzyme activities and growth performance of Caspian roach (Rutilus rutilus) fry. Fish Shellfish Immunol. 2012, 32, 316–321. [Google Scholar] [CrossRef]
  119. Sumon, M.S.; Ahmmed, F.; Khushi, S.S.; Ahmmed, M.K.; Rouf, M.A.; Chisty, M.A.H.; Sarower, M.G. Growth performance, digestive enzyme activity and immune response of Macrobrachium rosenbergii fed with probiotic Clostridium butyricum incorporated diets. J. King Saud Univ. Sci. 2018, 30, 21–28. [Google Scholar] [CrossRef]
  120. Ibrahem, M.D.; Fathi, M.; Mesalhy, S.; Abd El-Aty, A.M. Effect of dietary supplementation of inulin and vitamin C on the growth, hematology, innate immunity, and resistance of Nile tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2010, 29, 241–246. [Google Scholar] [CrossRef]
  121. 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]
  122. Ramesh, D.; Vinothkanna, A.; Rai, A.K.; Vignesh, V.S. Isolation of potential probiotic Bacillus spp. and assessment of their subcellular components to induce immune responses in Labeo rohita against Aeromonas hydrophila. Fish Shellfish Immunol. 2015, 45, 268–276. [Google Scholar] [CrossRef] [PubMed]
  123. Torrecillas, S.; Makol, A.; Benítez-Santana, T.; Caballero, M.J.; Montero, D.; Sweetman, J.; Izquierdo, M. Reduced gut bacterial translocation in European sea bass (Dicentrarchus labrax) fed mannan oligosaccharides (MOS). Fish Shellfish Immunol. 2011, 30, 674–681. [Google Scholar] [CrossRef] [PubMed]
  124. Geraylou, Z.; Souffreau, C.; Rurangwa, E.; D’Hondt, S.; Callewaert, L.; Courtin, C.M.; Delcour, J.A.; Buyse, J.; Ollevier, F. Effects of arabinoxylan-oligosaccharides (AXOS) on juvenile Siberian sturgeon (Acipenser baerii) performance, immune responses and gastrointestinal microbial community. Fish Shellfish Immunol. 2012, 33, 718–724. [Google Scholar] [CrossRef] [PubMed]
  125. Chang, C.S.; Huang, S.L.; Chen, S.; Chen, S.N. Innate immune responses and efficacy of using mushroom beta-glucan mixture (MBG) on orange-spotted grouper, Epinephelus coioides, aquaculture. Fish Shellfish Immunol. 2013, 35, 115–125. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Lin, S.; Mao, S.; Guan, Y.; Luo, L.; Pan, Y. Effects of dietary chitosan oligosaccharides and Bacillus coagulans on the growth, innate immunity, and resistance of koi (Cyprinus carpio koi). Aquaculture 2012, 342, 36–41. [Google Scholar] [CrossRef]
  128. Cui, L.; Xu, W.; Ai, Q.; Wang, D.; Mai, K. Effects of dietary chitosan oligosaccharide complex with rare earth on growth performance and innate immune response of turbot, Scophthalmus maximus L. Aquac. Res. 2013, 44, 683–690. [Google Scholar] [CrossRef]
  129. Gu, M.; Pan, S.; Li, Q.; Qi, Z.; Deng, W.; Bai, N. Chitosan and chitooligosaccharides attenuate soyabean meal-induced intestinal inflammation of turbot (Scophthalmus maximus): Possible involvement of NF-кB, activator protein-1 and mitogen-activated protein kinases pathways. Brit. J. Nutr. 2021, 126, 1651–1662. [Google Scholar] [CrossRef]
  130. Pérez-Jiménez, G.M.; Peña-Marín, E.S.; Maytorena-Verdugo, C.I.; Sepúlveda-Quiroz, C.A.; Jiménez-Martínez, L.D.; De la Rosa-García, S.; Asencio-Alcudia, G.G.; Martinez, R.; Tovar-Ramirez, D.; Galaviz, M.; et al. Incorporation of fructooligosaccharides in diets influence growth performance, digestive enzyme activity, and expression of intestinal barrier function genes in tropical gar (Atractosteus tropicus) larvae. Fishes 2022, 7, 137. [Google Scholar] [CrossRef]
  131. Zhang, C.N.; Li, X.F.; Xu, W.N.; Jiang, G.Z.; Lu, K.L.; Wang, L.N.; Liu, W.B. Combined effects of dietary fructooligosaccharide and Bacillus licheniformis on innate immunity, antioxidant capability and disease resistance of triangular bream (Megalobrama terminalis). Fish Shellfish Immunol. 2013, 35, 13801386. [Google Scholar] [CrossRef] [PubMed]
  132. Poolsawat, L.; Li, X.; Yang, H.; Yang, P.; Kabir Chowdhury, M.A.; Yusuf, A.; Leng, X. The potentials of fructooligosaccharide on growth, feed utilization, immune and antioxidant parameters, microbial community and disease resistance of tilapia (Oreochromis niloticus × O. aureus). Aquac. Res. 2020, 51, 4430–4442. [Google Scholar] [CrossRef]
  133. Grisdale-Helland, B.; Helland, S.J.; Gatlin, D.M. III. The effects of dietary supplementation with mannanoligosaccharide, fructooligosaccharide, or galactooligosaccharide on the growth and feed utilization of Atlantic salmon (Salmo salar). Aquaculture 2008, 283, 163–167. [Google Scholar] [CrossRef]
  134. 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 Japanese flounder Paralichthys olivaceus. Aquac. Nutr. 2011, 17, e902e11. [Google Scholar]
  135. Akrami, R.; Iri, Y.; Khoshbavar Rostami, H.; Razeghi Mansour, M. Effect of dietary supplementation of fructooligosaccharide (FOS) on growth performance, survival, lactobacillus bacterial population and hemato-immunological parameters of stellate sturgeon (Acipenser stellatus) juvenile. Fish Shellfish Immunol. 2013, 35, 1235–1239. [Google Scholar] [CrossRef]
  136. Zhang, Q.; Yu, H.; Tong, T.; Tong, W.; Dong, L.; Xu, M.; Wang, Z. Dietary supplementation of Bacillus subtilis and fructooligosaccharide enhance the growth, non-specific immunity of juvenile ovate pompano, Trachinotus ovatus and its disease resistance against Vibrio vulnificus. Fish Shellfish Immunol. 2014, 38, 7–14. [Google Scholar] [CrossRef]
  137. Yousefi, S.; Hoseinifar, S.H.; Paknejad, H.; Hajimoradloo, A. The effects of dietary supplement of galactooligosaccharide on innate immunity, immune related genes expression and growth performance in zebrafish (Danio rerio). Fish Shellfish Immunol. 2018, 73, 192–196. [Google Scholar] [CrossRef]
  138. Ajdari, A.; Ghafarifarsani, H.; Hoseinifar, S.H.; Javahery, S.; Narimanizad, F.; Gatphayak, K.; Van Doan, H. Effects of dietary supplementation of PrimaLac, inulin, and Biomin Imbo on growth performance, antioxidant, and innate immune responses of common carp (Cyprinus carpio). Aquac. Nutr. 2022, 2022, 8297479. [Google Scholar] [CrossRef]
  139. Ahmdifar, E.; Akrami, R.; Ghelichi, A.; Mohammadi Zarejabad, A. Effects of different dietary prebiotic inulin levels on blood serum enzymes, hematologic, and biochemical parameters of great sturgeon (Huso huso) juveniles. Comp. Clin. Pathol. 2011, 20, 447–451. [Google Scholar] [CrossRef]
  140. Cerezuela, R.; Cuesta, A.; Meseguer, J.; Esteban, M. Effects of dietary inulin and heat-inactivated Bacillus subtilis on gilthead seabream (Sparus aurata L.) innate immune parameters. Benef. Microb. 2012, 3, 77–81. [Google Scholar] [CrossRef]
  141. Syed Raffic Ali, S.; Ambasankar, K.; Praveena, P.E.; Nandakumar, S.; Saiyad Musthafa, M. Effect of dietary prebiotic inulin on histology, immuno-haematological and biochemical parameters of Asian seabass (Lates calcarifer). Aquac. Res. 2018, 49, 2732–2740. [Google Scholar] [CrossRef]
  142. Rairakhwada, D.; Pal, A.K.; Bhathena, Z.P.; Sahu, N.P.; Jha, A.; Mukherjee, S.C. Dietary microbial levan enhances cellular non-specific immunity and survival of common carp (Cyprinus carpio) juveniles. Fish Shellfish Immunol. 2007, 22, 477–486. [Google Scholar] [CrossRef] [PubMed]
  143. Gupta, S.K. Ameliorative and protective effects of prebiotic, microbial levan in common carp (Cyprinus carpio) fry under experimental exposure to fipronil. Int. J. Aquat. Biol. 2021, 9, 134–147. [Google Scholar]
  144. Gupta, S.K.; Pal, A.K.; Sahu, N.P.; Dalvi, R.; Kumar, V.; Mukherjee, S.C. Microbial levan in the diet of Labeo rohita Hamilton juveniles: Effect on non-specific immunity and histopathological changes after challenge with Aeromonas hydrophila. J. Fish Dis. 2008, 31, 649–657. [Google Scholar] [CrossRef]
  145. Huang, M.Y.; Chang, C.I.; Chang, C.C.; Tseng, L.W.; Pan, C.L. Effects of dietary levan on growth performance, nonspecific immunity, pathogen resistance, and body composition of orange-spotted grouper (Epinephelus coioides H.). Aquac. Res. 2014, 46, 2752–2762. [Google Scholar] [CrossRef]
  146. Staykov, Y.; Spring, P.; Denev, S.; Sweetman, J. Effect of a mannan oligosaccharide on the growth performance and immune status of rainbow trout (Oncorhynchus mykiss). Aquac. Int. 2007, 15, 153–161. [Google Scholar] [CrossRef]
  147. Rodriguez-Estrada, U.; Satoh, S.; Haga, Y.; Fushimi, H.; Sweetman, J. Effects of single and combined supplementation of Enterococcus faecalis, mannan oligosaccharide and polyhydrobutyrate acid on growth performance and immune response of rainbow trout (Oncorhynchus mykiss). Aquac. Sci. 2009, 57, 609–617. [Google Scholar]
  148. Rodríguez-Estrada, U.; Satoh, S.; Haga, Y.; Fushimi, H.; Sweetman, J. Effects of inactivated Enterococcus faecalis and mannan oligosaccharide and their combination on growth, immunity, and disease protection in rainbow trout. N. Am. J. Aquacult. 2013, 75, 416–428. [Google Scholar] [CrossRef]
  149. Yoshida, T.; Kruger, R.; Inglis, V. Augmentation of nonspecific protection in African catfish, Clarias gariepinus (Burchell), by the long-term oral administration of immunostimulants. J. Fish Dis. 1995, 8, 195–198. [Google Scholar] [CrossRef]
  150. Talpur, A.D.; Munir, M.B.; Mary, A.; Hashim, R. Dietary probiotics and prebiotics improved food acceptability, growth performance, haematology and immunological parameters, and disease resistance against Aeromonas hydrophila in snakehead (Channa striata) fingerlings. Aquaculture 2014, 426, 14–20. [Google Scholar] [CrossRef]
  151. Akter, M.; Hashim, R.; Sutriana, A.; Nor, S.A.M. Influence of mannan oligosaccharide supplementation on haematological and immunological responses and disease resistance of striped catfish (Pangasianodon hypophthalmus Sauvage, 1878) juveniles. Aquac. Int. 2019, 27, 1535–1551. [Google Scholar] [CrossRef]
  152. Zhou, Q.-C.; Buentello, J.A.; Gatlin, D.M. Effects of dietary prebiotics on growth performance, immune response and intestinal morphology of red drum (Sciaenops ocellatus). Aquaculture 2010, 309, 253–257. [Google Scholar] [CrossRef]
  153. Lokesh, J.; Fernandes, J.M.O.; Korsnes, K.; Bergh, Ø.; Brinchmann, M.F.; Kiron, V. Transcriptional regulation of cytokines in the intestine of Atlantic cod fed yeast derived mannan oligosaccharide or b-Glucan and challenged with Vibrio anguillarum. Fish Shellfish Immunol. 2012, 33, 626–631. [Google Scholar] [CrossRef]
  154. El-Nokrashy, A.M.; El-Banna, R.A.; Edrise, B.M.; Abdel-Rahim, M.M.; Jover-Cerdá, M.; Tomás-Vidal, A.; Prince, A.; Davies, S.; El-Haroun, E.; Goda, A.S. Impact of nucleotide enriched diets on the production of gilthead seabream, Sparus aurata fingerlings by modulation of liver mitochondrial enzyme activitity, antioxidant status, immune gene expression, and gut microbial ecology. Aquaculture 2021, 535, 736398. [Google Scholar] [CrossRef]
  155. Lee, P.P.; Lin, Y.H.; Chen, M.C.; Cheng, W. Dietary administration of sodium alginate ameliorated stress and promoted immune resistance of grouper Epinephelus coioides under cold stress. Fish Shellfish Immunol. 2017, 65, 127–135. [Google Scholar] [CrossRef]
  156. Yang, P.; Hu, H.; Liu, Y.; Li, Y.; Ai, Q.; Xu, W.; Zhang, W.; Zhang, Y.; Zhang, Y.; Mai, K. Dietary stachyose altered the intestinal microbiota profile and improved the intestinal mucosal barrier function of juvenile turbot, Scophthalmus maximus L. Aquaculture 2018, 486, 98–106. [Google Scholar] [CrossRef]
  157. Pérez-Sánchez, J.; Benedito-Palos, L.; Estensoro, I.; Petropoulos, Y.; Calduch-Giner, J.A.; Browdy, C.L.; Sitjà-Bobadilla, A. Effects of dietary NEXT ENHANCE® 150 on growth performance and expression of immune and intestinal integrity related genes in gilthead sea bream (Sparus aurata L.). Fish Shellfish Immunol. 2015, 44, 117–128. [Google Scholar] [CrossRef]
  158. Firmino, J.P.; Fernández-Alacid, L.; Vallejos-Vidal, E.; Salomón, R.; Sanahuja, I.; Tort, L.; Ibarz, A.; Reyes-Lopez, F.E.; Gisbert, E. Carvacrol, thymol, and garlic essential oil promote skin innate immunity in gilthead seabream (Sparus aurata) through the multifactorial modulation of the secretory pathway and enhancement of mucus protective capacity. Front. Immunol. 2021, 12, 633621. [Google Scholar] [CrossRef] [PubMed]
  159. Shukry, M.; El-Kader, M.F.A.; Hendam, B.M.; Dawood, M.A.O.; Farrag, F.A.; Aboelenin, S.M.; Soliman, M.M.; Abdel-Latif, H.M. Dietary Aspergillus oryzae modulates serum biochemical indices, immune responses, oxidative stress, and transcription of HSP70 and cytokine genes in Nile tilapia exposed to salinity stress. Animals 2021, 11, 1621. [Google Scholar] [CrossRef] [PubMed]
  160. Ramesh, D.; Souissi, S.; Ahamed, T.S. Effects of the potential probiotics Bacillus aerophilus KADR3 in inducing immunity and disease resistance in Labeo rohita. Fish Shellfish Immunol. 2017, 70, 408–415. [Google Scholar] [CrossRef] [PubMed]
  161. Nandi, A.; Banerjee, G.; Dan, S.K.; Ghosh, K.; Ray, A.K. Evaluation of in vivo probiotic efficiency of Bacillus amyloliquefaciens in Labeo rohita challenged by pathogenic strain of Aeromonas hydrophila MTCC 1739. Probiotics Antimicrob. Proteins 2018, 10, 391–398. [Google Scholar] [CrossRef] [PubMed]
  162. Truong Thy, H.T.; Tri, N.N.; Quy, O.M.; Fotedar, R.; Kannika, K.; Unajak, S.; Areechon, N. Effects of the dietary supplementation of mixed probiotic spores of Bacillus amyloliquefaciens 54A, and Bacillus pumilus 47B on growth, innate immunity and stress responses of striped catfish (Pangasianodon hypophthalmus). Fish Shellfish Immunol. 2017, 60, 391–399. [Google Scholar] [CrossRef] [PubMed]
  163. Zhang, J.; Huang, M.; Feng, J.; Chen, Y.; Li, M.; Chang, X. Effects of dietary Bacillus licheniformis on growth performance, intestinal morphology, intestinal microbiome, and disease resistance in common carp (Cyprinus carpio L.). Aquac. Int. 2021, 29, 1343–1358. [Google Scholar] [CrossRef]
  164. Gobi, N.; Vaseeharan, B.; Chen, J.-C.; Rekha, R.; Vijayakumar, S.; Anjugam, M.; Iswarya, A. Dietary supplementation of probiotic Bacillus licheniformis Dahb1 improves growth performance, mucus and serum immune parameters, antioxidant enzyme activity as well as resistance against Aeromonas hydrophila in tilapia Oreochromis mossambicus. Fish Shellfish Immunol. 2018, 74, 501–508. [Google Scholar] [CrossRef]
  165. Tarnecki, A.M.; Wafapoor, M.; Phillips, R.N.; Rhody, N.R. Benefits of a Bacillus probiotic to larval fish survival and transport stress resistance. Sci. Rep. 2019, 9, 4892. [Google Scholar] [CrossRef]
  166. Weifen, L.; Xiaoping, Z.; Wenhui, S.; Bin, D.; Quan, L.; Luoqin, F.; Jiajia, Z.; Yue, W.; Dongyou, Y. Effects of Bacillus preparations on immunity and antioxidant activities in grass carp (Ctenopharyngodon idellus). Fish Physiol. Biochem. 2012, 38, 1585–1592. [Google Scholar] [CrossRef]
  167. Gobi, N.; Malaikozhundan, B.; Sekar, V.; Shanthi, S.; Vaseeharan, B.; Jayakumar, R.; Nazar, A.K. GFP tagged Vibrio parahaemolyticus Dahv2 infection and the protective effects of probiotic Bacillus licheniformis Dahb1 on the growth, immune and antioxidant responses in Pangasius hypophthalmus. Fish Shellfish Immunol. 2016, 52, 230–238. [Google Scholar] [CrossRef]
  168. Mukherjee, A.; Chandra, G.; Ghosh, K. Single or conjoint application of autochthonous Bacillus strains as potential probiotics: Effects on growth, feed utilization, immunity and disease resistance in rohu, Labeo rohita (Hamilton). Aquaculture 2019, 512, 734302. [Google Scholar] [CrossRef]
  169. Srisapoome, P.; Areechon, N. Efficacy of viable Bacillus pumilus isolated from farmed fish on immune responses and increased disease resistance in Nile tilapia (Oreochromis niloticus): Laboratory and on-farm trials. Fish Shellfish Immunol. 2017, 67, 199–218. [Google Scholar] [CrossRef] [PubMed]
  170. Liu, S.; Wang, S.; Cai, Y.; Li, E.; Ren, Z.; Wu, Y.; Guo, W.; Sun, Y.; Zhou, Y. Beneficial effects of a host gut-derived probiotic, Bacillus pumilus, on the growth, non-specific immune response and disease resistance of juvenile golden pompano, Trachinotus ovatus. Aquaculture 2020, 514, 734446. [Google Scholar] [CrossRef]
  171. Sun, Y.-Z.; Yang, H.-L.; Huang, K.-P.; Ye, J.-D.; Zhang, C.-X. Application of autochthonous Bacillus bioencapsulated in copepod to grouper Epinephelus coioides larvae. Aquaculture 2013, 392, 44–50. [Google Scholar] [CrossRef]
  172. Standen, B.T.; Peggs, D.L.; Rawling, M.D.; Foey, A.; Davies, S.J.; Santos, G.A.; Merrifield, D.L. Dietary administration of a commercial mixed-species probiotic improves growth performance and modulates the intestinal immunity of tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2016, 49, 427–435. [Google Scholar] [CrossRef] [PubMed]
  173. Lee, S.; Katya, K.; Park, Y.; Won, S.; Seong, M.; Bai, S.C. Comparative evaluation of dietary probiotics Bacillus subtilis WB60 and Lactobacillus plantarum KCTC3928 on the growth performance, immunological parameters, gut morphology and disease resistance in Japanese eel, Anguilla japonica. Fish Shellfish Immunol. 2017, 61, 201–210. [Google Scholar] [CrossRef] [PubMed]
  174. Ghanei-Motlagh, R.; Mohammadian, T.; Gharibi, D.; Khosravi, M.; Mahmoudi, E.; Zarea, M.; El-Matbouli, M.; Menanteau-Ledouble, S. Quorum quenching probiotics modulated digestive enzymes activity, growth performance, gut microflora, haemato-biochemical parameters and resistance against Vibrio harveyi in Asian seabass (Lates calcarifer). Aquaculture 2021, 531, 735874. [Google Scholar] [CrossRef]
  175. Kuebutornye, F.K.A.; Wang, Z.; Lu, Y.; Abarike, E.D.; Sakyi, M.E.; Li, Y.; Xie, C.X.; Hlordzi, V. Effects of three host-associated Bacillus species on mucosal immunity and gut health of Nile tilapia, Oreochromis niloticus and its resistance against Aeromonas hydrophila infection. Fish Shellfish Immunol. 2020, 97, 83–95. [Google Scholar] [CrossRef]
  176. Saputra, F.; Shiu, Y.L.; Chen, Y.C.; Puspitasari, A.W.; Danata, R.H.; Liu, C.H.; Hu, S.Y. Dietary supplementation with xylanase-expressing B. áamyloliquefaciens R8 improves growth performance and enhances immunity against Aeromonas hydrophila in Nile tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2016, 58, 397–405. [Google Scholar] [CrossRef]
  177. Xia, Y.; Wang, M.; Gao, F.; Lu, M.; Chen, G. Effects of dietary probiotic supplementation on the growth, gut health and disease resistance of juvenile Nile tilapia (Oreochromis niloticus). Anim. Nutr. 2020, 6, 69–79. [Google Scholar] [CrossRef]
  178. Bandyopadhyay, P.; Das Mohapatra, P.K. Effect of a probiotic bacterium Bacillus circulans PB7 in the formulated diets: On growth, nutritional quality and immunity of Catla catla (Ham.). Fish Physiol. Biochem. 2009, 35, 467–478. [Google Scholar] [CrossRef] [PubMed]
  179. Niu, K.-M.; Khosravi, S.; Kothari, D.; Lee, W.-D.; Lim, J.-M.; Lee, B.-J.; Kim, K.-W.; Lim, S.-G.; Lee, S.-M.; Kim, S.-K. Effects of dietary multi-strain probiotics supplementation in a low fishmeal diet on growth performance, nutrient utilization, proximate composition, immune parameters, and gut microbiota of juvenile olive flounder (Paralichthys olivaceus). Fish Shellfish Immunol. 2019, 93, 258–268. [Google Scholar] [CrossRef] [PubMed]
  180. Mohammadi, G.; Adorian, T.J.; Rafiee, G. Beneficial effects of Bacillus subtilis on water quality, growth, immune responses, endotoxemia and protection against lipopolysaccharide-induced damages in Oreochromis niloticus under biofloc technology system. Aquac. Nutr. 2020, 26, 1476–1492. [Google Scholar] [CrossRef]
  181. Hassaan, M.S.; Soltan, M.A.; Jarmołowicz, S.; Abdo, H.S. Combined effects of dietary malic acid and Bacillus subtilis on growth, gut microbiota and blood parameters of Nile tilapia (Oreochromis niloticus). Aquac. Nutr. 2018, 24, 83–93. [Google Scholar] [CrossRef]
  182. Meidong, R.; Khotchanalekha, K.; Doolgindachbaporn, S.; Nagasawa, T.; Nakao, M.; Sakai, K.; Tongpim, S. Evaluation of probiotic Bacillus aerius B81e isolated from healthy hybrid catfish on growth, disease resistance and innate immunity of Plamong Pangasius bocourti. Fish Shellfish Immunol. 2018, 73, 1–10. [Google Scholar] [CrossRef]
  183. Reyes-Becerril, M.; Angulo, C.; Angulo, M.; Esteban, M.Á. Probiotic properties of Debaryomyces hansenii BCS004 and their immunostimulatory effect in supplemented diets for gilthead seabream (Sparus aurata). Aquac. Res. 2021, 52, 2715–2726. [Google Scholar] [CrossRef]
  184. 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]
  185. Baños, A.; Ariza, J.J.; Nuñez, C.; Gil-Martínez, L.; García-López, J.D.; Martínez-Bueno, M.; Valdivia, E. Effects of Enterococcus faecalis UGRA10 and the enterocin AS-48 against the fish pathogen Lactococcus garvieae. Studies in vitro and in vivo. Food Microbiol. 2019, 77, 69–77. [Google Scholar] [CrossRef]
  186. Tarkhani, R.; Imani, A.; Hoseinifar, S.H.; Moghanlou, K.S.; Manaffar, R. The effects of host-associated Enterococcus faecium CGMCC1.2136 on serum immnune parameters, digestive enzymes activity and growth performance of the Caspian roach (Rutilus caspicus) fingerlings. Aquaculture 2020, 519, 734741. [Google Scholar] [CrossRef]
  187. Picchietti, S.; Fausto, A.M.; Randelli, E.; Carnevali, O.; Taddei, A.R.; Buonocore, F.; Scaigliati, G.; Abelli, L. Early treatment with Lactobacillus delbrueckii strain induces an increase in intestinal T-cells and granulocytes and modulates immune-related genes of larval Dicentrarchus labrax (L.). Fish Shellfish Immunol. 2009, 26, 368–376. [Google Scholar] [CrossRef]
  188. Mohammadi, G.; Rafiee, G.; Tavabe, K.R.; Abdel-Latif, H.M.; Dawood, M.A. The enrichment of diet with beneficial bacteria (single-or multi-strain) in biofloc system enhanced the water quality, growth performance, immune responses, and disease resistance of Nile tilapia (Oreochromis niloticus). Aquaculture 2021, 539, 736640. [Google Scholar] [CrossRef]
  189. Hooshyar, Y.; Abedian Kenari, A.; Paknejad, H.; Gandomi, H. Effects of Lactobacillus rhamnosus ATCC 7469 on different parameters related to health status of rainbow trout (Oncorhynchus mykiss) and the protection against Yersinia ruckeri. Probiotics Antimicrob. Proteins 2020, 12, 1370–1384. [Google Scholar] [CrossRef] [PubMed]
  190. Hoseinifar, S.H.; Roosta, Z.; Hajimoradloo, A.; Vakili, F. The effects of Lactobacillus acidophilus as feed supplement on skin mucosal immune parameters, intestinal microbiota, stress resistance and growth performance of black swordtail (Xiphophorus helleri). Fish Shellfish Immunol. 2015, 42, 533–538. [Google Scholar] [CrossRef] [PubMed]
  191. Al-Dohail, M.A.; Hashim, R.; Aliyu-Paiko, M. Effects of the probiotic, Lactobacillus acidophilus, on the growth performance, haematology parameters and immunoglobulin concentration in African Catfish (Clarias gariepinus, Burchell 1822) fingerling. Aquac. Res. 2009, 40, 1642–1652. [Google Scholar] [CrossRef]
  192. Adeshina, I.; Abubakar, M.I.O.; Ajala, B.E. Dietary supplementation with Lactobacillus acidophilus enhanced the growth, gut morphometry, antioxidant capacity, and the immune response in juveniles of the common carp, Cyprinus carpio. Fish Physiol. Biochem. 2020, 46, 1375–1385. [Google Scholar] [CrossRef]
  193. Zhang, C.-N.; Zhang, J.-L.; Guan, W.-C.; Zhang, X.-F.; Guan, S.-H.; Zeng, Q.-H.; Cheng, G.-F.; Cui, W. Effects of Lactobacillus delbrueckii on immune response, disease resistance against Aeromonas hydrophila, antioxidant capability and growth performance of Cyprinus carpio Huanghe var. Fish Shellfish Immunol. 2017, 68, 84–91. [Google Scholar] [CrossRef]
  194. Ahmadifar, E.; Moghadam, M.S.; Dawood, M.A.O.; Hoseinifar, S.H. Lactobacillus fermentum and/or ferulic acid improved the immune responses, antioxidative defence and resistance against Aeromonas hydrophila in common carp (Cyprinus carpio) fingerlings. Fish Shellfish Immunol. 2019, 94, 916–923. [Google Scholar] [CrossRef]
  195. Hamdan, A.M.; El-Sayed, A.F.M.; Mahmoud, M.M. Effects of a novel marine probiotic, Lactobacillus plantarum AH 78, on growth performance and immune response of Nile tilapia (Oreochromis niloticus). J. Appl. Microbiol. 2016, 120, 1061–1073. [Google Scholar] [CrossRef]
  196. Van Doan, H.; Kurian, A.; Hoseinifar, S.H.; Sel-audom, M.; Jaturasitha, S.; Tongsiri, S.; Ringø, E. Dietary inclusion of orange peels derived pectin and Lactobacillus plantarum for Nile tilapia (Oreochromis niloticus) cultured under indoor biofloc systems. Aquaculture 2019, 508, 98–105. [Google Scholar] [CrossRef]
  197. Van Doan, H.; Hoseinifar, S.H.; Khanongnuch, C.; Kanpiengjai, A.; Unban, K.; Van Kim, V.; Srichaiyo, S. Host-associated probiotics boosted mucosal and serum immunity, disease resistance and growth performance of Nile tilapia (Oreochromis niloticus). Aquaculture 2018, 491, 94–100. [Google Scholar] [CrossRef]
  198. Xia, Y.; Lu, M.; Chen, G.; Cao, J.; Gao, F.; Wang, M.; Liu, Z.; Zhang, D.; Zhu, H.; Yi, M. Effects of dietary Lactobacillus rhamnosus JCM1136 and Lactococcus lactis subsp. Lactis JCM5805 on the growth, intestinal microbiota, morphology, immune response and disease resistance of juvenile Nile tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2018, 76, 368–379. [Google Scholar] [CrossRef] [PubMed]
  199. 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]
  200. Abdelfatah, E.N.; Mahboub, H.H.H. Studies on the effect of Lactococcus garvieae of dairy origin on both cheese and Nile tilapia (O. niloticus). Int. J. Vet. Sci. Med. 2018, 6, 201–207. [Google Scholar] [CrossRef] [PubMed]
  201. Nguyen, T.L.; Park, C.-I.; Kim, D.-H. Improved growth rate and disease resistance in olive flounder, Paralichthys olivaceus, by probiotic Lactococcus lactis WFLU12 isolated from wild marine fish. Aquaculture 2017, 471, 113–120. [Google Scholar] [CrossRef]
  202. Feng, J.; Chang, X.; Zhang, Y.; Zhang, X.Y.; Nie, G. Effects of Lactococcus lactis from Cyprinus carpio L. as probiotics on growth performance, innate immune response and disease resistance against Aeromonas hydrophila. Fish Shellfish Immunol. 2019, 93, 73–81. [Google Scholar] [CrossRef]
  203. Sewaka, M.; Trullas, C.; Chotiko, A.; Rodkhum, C.; Chansue, N.; Boonanuntanasam, S.; Pirarat, N. Efficacy of synbiotic Jerusalem artichoke and Lactobacillus rhamnosus GG-supplemented diets on growth performance, serum biochemical parameters, intestinal morphology, immune parameters and protection against Aeromonas veronii in juvenile red tilapia (Oreochromis spp.). Fish Shellfish Immunol. 2019, 86, 260–268. [Google Scholar]
  204. Feng, J.; Li, D.; Liu, L.; Tang, Y.; Du, R. Characterization and comparison of the adherence and immune modulation of two gut Lactobacillus strains isolated from Paralichthys olivaceus. Aquaculture 2018, 499, 381–388. [Google Scholar] [CrossRef]
  205. Dowidar, M.F.; Abd ElAzeem, S.; Khater, A.M.; Awad Somayah, M.; Metwally, S.A. Improvement of growth performance, immunity and disease resistance in Nile tilapia, Oreochromis niloticus, by using dietary probiotics supplementation. J. Anim. Sci. Vet. Med. 2018, 3, 35–46. [Google Scholar] [CrossRef]
  206. Chen, S.W.; Liu, C.H.; Hu, S.Y. Dietary administration of probiotic Paenibacillus ehimensis NPUST1 with bacteriocin-like activity improves growth performance and immunity against Aeromonas hydrophila and Streptococcus iniae in Nile tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2019, 84, 695–703. [Google Scholar] [CrossRef]
  207. Gupta, A.; Gupta, P.; Dhawan, A. Paenibacillus polymyxa as a water additive improved immune response of Cyprinus carpio and disease resistance against Aeromonas hydrophila. Aquac. Rep. 2016, 4, 86–92. [Google Scholar] [CrossRef]
  208. Ferguson, R.M.W.; Merrifield, D.L.; Harper, G.M.; Rawling, M.D.; Mustafa, S.; Picchietti, S.; Balcazar, J.L.; Davies, S.J. The effect of Pediococcus acidilactici on the gut microbiota and immune status of on-growing red tilapia (Oreochromis niloticus). J. Appl. Microbiol. 2010, 109, 851–862. [Google Scholar] [CrossRef]
  209. Standen, B.T.; Rawling, M.D.; Davies, S.J.; Castex, M.; Foey, A.; Gioacchini, G.; Carnevali, O.; Merrifield, D.L. Probiotic Pediococcus acidilactici modulates both localised intestinal- and peripheral-immunity in tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2013, 35, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  210. Hoseinifar, S.H.; Hossein, M.; Paknejad, H.; Safari, R.; Jafar, A.; Yousefi, M.; Doan, H.V.; Mozanzadeh, M.T. Enhanced mucosal immune responses, immune related genes and growth performance in common carp (Cyprinus carpio) juveniles fed dietary Pediococcus acidilactici MA18/5M and raffinose. Dev. Comp. Immunol. 2019, 94, 59–65. [Google Scholar] [CrossRef]
  211. Gong, L.; He, H.; Li, D.; Cao, L.; Khan, T.A.; Li, Y.; Pan, L.; Yan, L.; Ding, X.; Sun, Y.; et al. A new isolate of Pediococcus pentosaceus (SL001) with antibacterial activity against fish pathogens and potency in facilitating the immunity and growth performance of grass carps. Front. Microbiol. 2019, 10, 1384. [Google Scholar] [CrossRef] [PubMed]
  212. Eissa, N.; Abou-ElGheit, E. Dietary supplementation impacts of potential non-pathogenic isolates on growth performance, hematological parameters and disease resistance in Nile tilapia (Oreochromis niloticus). J. Vet. Adv. 2014, 4, 712–719. [Google Scholar] [CrossRef]
  213. 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] [PubMed]
  214. Abdel-Tawwab, M.; Abdel-Rahman, A.M.; Ismael, N.E. Evaluation of commercial live bakers’ yeast, Saccharomyces cerevisiae as a growth and immunity promoter for Fry Nile tilapia, Oreochromis niloticus (L.) challenged in situ with Aeromonas hydrophila. Aquaculture 2008, 280, 185–189. [Google Scholar] [CrossRef]
  215. Cordero, H.; Morcillo, P.; Cuesta, A.; Brinchmann, M.F.; Esteban, M.A. Differential proteome profile of skin mucus of gilthead seabream (Sparus aurata) after probiotic intake and/or overcrowding stress. J. Proteom. 2016, 132, 41–50. [Google Scholar] [CrossRef]
  216. Wu, Z.-Q.; Jiang, C.; Ling, F.; Wang, G.-X. Effects of dietary supplementation of intestinal autochthonous bacteria on the innate immunity and disease resistance of grass carp (Ctenopharyngodon idellus). Aquaculture 2015, 438, 105–114. [Google Scholar] [CrossRef]
  217. Schaeck, M.; Reyes-López, F.E.; Vallejos-Vidal, E.; Van Cleemput, J.; Duchateau, L.; Van den Broeck, W.; Tort, L.; Decostere, A. Cellular and transcriptomic response to treatment with the probiotic candidate Vibrio lentus in gnotobiotic sea bass (Dicentrarchus labrax) larvae. Fish Shellfish Immunol. 2017, 63, 147–156. [Google Scholar] [CrossRef]
  218. 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]
  219. De Souza, E.M.; de Souza, R.C.; Melo, J.F.B.; da Costa, M.M.; de Souza, A.M.; Copatti, C.E. Evaluation of the effects of Ocimum basilicum essential oil in Nile tilapia diet: Growth, biochemical, intestinal enzymes, haematology, lysozyme and antimicrobial challenges. Aquaculture 2019, 504, 7–12. [Google Scholar] [CrossRef]
  220. Sun, Y.-Z.; Yang, H.-L.; Ma, R.-L.; Song, K.; Li, J.-S. Effect of Lactococcus lactis and Enterococcus faecium on growth performance, digestive enzymes and immune response of grouper Epinephelus coioides. Aquac. Nutr. 2012, 18, 281–289. [Google Scholar] [CrossRef]
  221. Cerezuela, R.; Fumanal, M.; Tapia-Paniagua, S.T.; Meseguer, J.; Moriñigo, M.Á.; Esteban, M.Á. Changes in intestinal morphology and microbiota caused by dietary administration of inulin and Bacillus subtilis in gilthead sea bream (Sparus aurata L.) specimens. Fish Shellfish Immunol. 2013, 34, 1063–1070. [Google Scholar] [CrossRef] [PubMed]
  222. Abdel-Aziz, M.; Bessat, M.; Fadel, A.; Elblehi, S. Responses of dietary supplementation of probiotic effective microorganisms (EMs) in Oreochromis niloticus on growth, hematological, intestinal histopathological, and antiparasitic activities. Aquac. Int. 2020, 28, 947–963. [Google Scholar] [CrossRef]
  223. Amoah, K.; Dong, X.; Tan, B.; Zhang, S.; Chi, S.; Yang, Q.; Liu, H.; Yang, Y.; Zhang, H. Effects of three probiotic strains (Bacillus coagulans, B. licheniformis and Paenibacillus polymyxa) on growth, immune response, gut morphology and microbiota, and resistance against Vibrio harveyi of northern whitings, Sillago sihama Forsskál (1775). Anim. Feed Sci. Technol. 2021, 277, 114958. [Google Scholar] [CrossRef]
  224. Selim, K.M.; Reda, R.M. Beta-glucans and mannan oligosaccharides enhance growth and immunity in Nile tilapia. N. Am. J. Aquac. 2015, 77, 22–30. [Google Scholar] [CrossRef]
  225. Munir, M.B.; Hashim, R.; Chai, Y.H.; Marsh, T.L.; Nor, S.A.M. Dietary prebiotics and probiotics influence growth performance, nutrient digestibility and the expression of immune regulatory genes in snakehead (Channa striata) fingerlings. Aquaculture 2016, 460, 59–68. [Google Scholar] [CrossRef]
  226. 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]
  227. Sutriana, A.; Hashim, R.; Akter, M.N.; Mohd Nor, S.A. Galactooligosaccharide and a combination of yeast and β-glucan supplements enhance growth and improve intestinal condition in striped catfish Pangasianodon hypophthalmus fed soybean meal diets. Fish Sci. 2018, 84, 523–533. [Google Scholar] [CrossRef]
  228. Hossain, M.S.; Koshio, S.; Ishikawa, M.; Yokoyama, S.; Sony, N.M.; Ono, S.; Fujieda, T. Comparison of the effects of inosine and inosine monophosphate on growth, immune response, stress resistance and gut morphology of juvenile red sea bream, Pagrus major. Aquaculture 2016, 458, 64–74. [Google Scholar] [CrossRef]
  229. Lu, Z.-Y.; Feng, L.; Jiang, W.-D.; Wu, P.; Liu, Y.; Kuang, S.-Y.; Tang, L.; Zhou, X.-Q. Mannan oligosaccharides improved growth performance and antioxidant capacity in the intestine of on-growing grass carp (Ctenopharyngodon idella). Aquac. Rep. 2020, 17, 100313. [Google Scholar] [CrossRef]
  230. Cheng, Z.; Buentello, A.; Gatlin, D.M. Dietary nucleotides influence immune responses and intestinal morphology of red drum Sciaenops ocellatus. Fish Shellfish Immunol. 2011, 30, 143–147. [Google Scholar] [CrossRef] [PubMed]
  231. Bunnoy, A.; Na-Nakorn, U.; Srisapoome, P. Probiotic effects of a novel strain, Acinetobacter KU011TH, on the growth performance, immune responses, and resistance against Aeromonas hydrophila of bighead catfish (Clarias macrocephalus Günther, 1864). Microorganisms 2019, 7, 613. [Google Scholar] [CrossRef] [PubMed]
  232. Giannenas, I.; Karamaligas, I.; Margaroni, M.; Pappas, I.; Mayer, E.; Encarnacao, P.; Karagouni, E. Effect of dietary incorporation of a multi-strain probiotic on growth performance and health status in rainbow trout (Oncorhynchus mykiss). Fish Physiol. Biochem. 2015, 41, 119–128. [Google Scholar] [CrossRef] [PubMed]
  233. Maas, R.M.; Verdegem, M.C.; Debnath, S.; Marchal, L.; Schrama, J.W. Effect of enzymes (phytase and xylanase), probiotics (B. amyloliquefaciens) and their combination on growth performance and nutrient utilisation in Nile tilapia. Aquaculture 2021, 533, 736226. [Google Scholar] [CrossRef]
  234. Wang, Y. Use of probiotics Bacillus coagulans, Rhodopseudomonas palustris and Lactobacillus acidophilus as growth promoters in grass carp (Ctenopharyngodon idella) fingerlings. Aquac. Nutr. 2011, 17, e372–e378. [Google Scholar] [CrossRef]
  235. Yanbo, W.; Zirong, X. Effect of probiotics for common carp (Cyprinus carpio) based on growth performance and digestive enzyme activities. Anim. Feed Sci. Technol. 2006, 127, 283–292. [Google Scholar] [CrossRef]
  236. Asaduzzaman, M.; Sofia, E.; Shakil, A.; Haque, N.F.; Khan, M.N.A.; Ikeda, D.; Kinoshita, S.; Abol-Munafi, A.B. Host gut derived probiotic bacteria promote hypertrophic muscle progression and upregulate growth-related gene expression of slow-growing Malaysian mahseer Tor tambroides. Aquac. Rep. 2018, 9, 37–45. [Google Scholar] [CrossRef]
  237. Jang, W.J.; Lee, J.M.; Hasan, M.T.; Lee, B.-J.; Lim, S.G.; Kong, I.-S. Effects of probiotic supplementation of a plant-based protein diet on intestinal microbial diversitydigestive enzyme activity, intestinal structure, and immunity in olive flounder (Paralichthys olivaceus). Fish Shellfish Immunol. 2019, 92, 719–727. [Google Scholar] [CrossRef]
  238. Munirasu, S.; Ramasubramanian, V.; Arunkumar, P. Effect of probiotics diet on growth and biochemical performance of freshwater fish Labeo rohita fingerlings. J. Entomol. Zool. Stud. 2017, 5, 1374–1379. [Google Scholar]
  239. Adeoye, A.A.; Yomla, R.; Jaramillo-Torres, A.; Rodiles, A.; Merrifield, D.L.; Davies, S.J. Combined effects of exogenous enzymes and probiotic on Nile tilapia (Oreochromis niloticus) growth, intestinal morphology and microbiome. Aquaculture 2016, 463, 61–70. [Google Scholar] [CrossRef]
  240. Haque, M.M.; Hasan, N.A.; Eltholth, M.M.; Saha, P.; Mely, S.S.; Rahman, T.; Murray, F.J. Assessing the impacts of in-feed probiotic on the growth performance and health condition of pangasius (Pangasianodon hypophthalmus) in a farm trial. Aquac. Rep. 2021, 20, 100699. [Google Scholar] [CrossRef] [PubMed]
  241. Silva, V.V.; Salomao, R.A.S.; Mareco, E.A.; Dal Pai, M.; Santos, V.B. Probiotic additive affects muscle growth of Nile tilapia (Oreochromis niloticus). Aquac. Res. 2021, 52, 2061–2069. [Google Scholar] [CrossRef]
  242. Mohammadian, T.; Nasirpour, M.; Tabandeh, M.R.; Mesbah, M. Synbiotic effects of b-glucan, mannan oligosaccharide and Lactobacillus casei on growth performance, intestine enzymes activities, immunehematological parameters and immune-related gene expression in common carp, Cyprinus carpio: An experimental infection with Aeromonas hydrophila. Aquaculture 2019, 511, 634197. [Google Scholar]
  243. Muchlisin, Z.A.; Murda, T.; Yulvizar, C.; Dewiyanti, I.; Fadli, N.; Afrido, F.; Muhammadar, A.A. Growth performance and feed utilization of keureling fish Tor tambra (Cyprinidae) fed formulated diet supplemented with enhanced probiotic. F1000Research 2017, 6, 137. [Google Scholar] [CrossRef]
  244. Selvaraj, R.; Bogar, T. Effect of probiotic supplemented feed on growth performance of molly fish (Poecilia sphenops) in Recirculating aquaculture system. Pharma Innov. J. 2019, 8, 533–537. [Google Scholar]
  245. Abd El-Rhman, A.M.; Khattab, Y.A.; Shalaby, A.M. 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]
  246. Sankar, H.; Philip, B.; Philip, R.; Singh, I.S.B. Effect of probiotics on digestive enzyme activities and growth of cichlids, Etroplus suratensis (Pearl spot) and Oreochromis mossambicus (Tilapia). Aquac. Nutr. 2017, 23, 852–864. [Google Scholar] [CrossRef]
  247. Islam, S.M.; Rohani, M.F.; Shahjahan, M. Probiotic yeast enhances growth performance of Nile tilapia (Oreochromis niloticus) through morphological modifications of intestine. Aquac. Rep. 2021, 21, 100800. [Google Scholar] [CrossRef]
  248. Jahan, N.; Islam, S.M.; Rohani, M.F.; Hossain, M.T.; Shahjahan, M. Probiotic yeast enhances growth performance of rohu (Labeo rohita) through upgrading hematology, and intestinal microbiota and morphology. Aquaculture 2021, 545, 737243. [Google Scholar] [CrossRef]
  249. 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]
  250. Dharmaraj, S.; Dhevendaran, K. Evaluation of Streptomyces as a probiotic feed for the growth of ornamental fish Xiphophorus helleri. Food Technol. Biotechnol. 2010, 48, 497–504. [Google Scholar]
  251. Rohani, M.F.; Islam, S.M.M.; Hossain, M.K.; Ferdous, Z.; Siddik, M.A.B.; 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]
  252. Mohapatra, S.; Chakraborty, T.; Prusty, A.K.; PaniPrasad, K.; Mohanta, K.N. Beneficial effects of dietary probiotics mixture on hemato-immunology and cell apoptosis of Labeo rohita fingerlings reared at higher water temperatures. PLoS ONE 2014, 9, e100929. [Google Scholar] [CrossRef]
  253. Afsa, S.; De Marco, G.; Giannetto, A.; Parrino, V.; Cappello, T.; ben Mansour, H.; Maisano, M. Histological endpoints and oxidative stress transcriptional responses in the Mediterranean mussel Mytilus galloprovincialis exposed to realistic doses of salicylic acid. Environ. Toxicol. Pharmacol. 2022, 92, 103855. [Google Scholar] [CrossRef] [PubMed]
  254. Afsa, S.; De Marco, G.; Cristaldi, A.; Giannetto, A.; Galati, M.; Billè, B.; Oliveri Conti, G.; ben Mansour, H.; Ferrante, M.; Cappello, T. Single and combined effects of caffeine and salicylic acid on mussel Mytilus galloprovincialis: Changes at histomorphological, molecular and biochemical levels. Environ. Toxicol. Pharmacol. 2023, 101, 104167. [Google Scholar] [CrossRef]
  255. De Marco, G.; Conti, G.O.; Giannetto, A.; Cappello, T.; Galati, M.; Iaria, C.; Pulvirenti, E.; Capparucci, F.; Mauceri, A.; Ferrante, M.; et al. Embryotoxicity of polystyrene microplastics in zebrafish Danio rerio. Environ. Res. 2022, 208, 112552. [Google Scholar] [CrossRef]
  256. Noureen, A.; De Marco, G.; Rehman, N.; Jabeen, F.; Cappello, T. Ameliorative hematological and histomorphological effects of dietary Trigonella foenum-graecum seeds in common carp (Cyprinus carpio) exposed to copper oxide nanoparticles. Int. J. Environ. Res. Public Health 2022, 19, 13462. [Google Scholar] [CrossRef]
  257. Noureen, A.; Jabeen, F.; Wajid, A.; Kazim, M.Z.; Safdar, N.; Cappello, T. Natural bioactive phytocompounds to reduce toxicity in common carp Cyprinus carpio: A challenge to environmental risk assessment of nanomaterials. Water 2023, 15, 1152. [Google Scholar] [CrossRef]
  258. Abid, A.; Davies, S.J.; Waines, P.; Emery, M.; Castex, M.; Gioacchini, G.; Carnevali, O.; Bickerdike, R.; Romero, J.; Merrifield, D.L. Dietary synbiotic application modulates Atlantic salmon (Salmo salar) intestinal microbial communities and intestinal immunity. Fish Shellfish Immunol. 2013, 35, 1948–1956. [Google Scholar] [CrossRef] [PubMed]
  259. Milián-Sorribes, M.C.; Martínez-Llorens, S.; Cruz-Castellón, C.; Jover-Cerdá, M.; Tomás-Vidal, A. Effect of fish oil replacement and probiotic addition on growth, body composition and histological parameters of yellowtail (Seriola dumerili ). Aquac. Nutr. 2021, 27, 3–16. [Google Scholar] [CrossRef]
  260. Darafsh, F.; Soltani, M.; Abdolhay, H.A.; Shamsaei Mehrejan, M. Improvement of growth performance, digestive enzymes and body composition of Persian sturgeon (Acipenser persicus) following feeding on probiotics: Bacillus licheniformis, Bacillus subtilis and Saccharomyces cerevisiae. Aquac. Res. 2020, 51, 957–964. [Google Scholar] [CrossRef]
  261. González-Félix, M.L.; Gatlin, D.M.; Urquidez-Bejarano, P.; de la Reé-Rodríguez, C.; Duarte-Rodríguez, L.; Sánchez, F.; Casas-Reyes, A.; Yamamoto, F.Y.; Ochoa-Leyva, A.; Perez-Velazquez, M. Effects of commercial dietary prebiotic and probiotic supplements on growth, innate immune responses, and intestinal microbiota and histology of Totoaba macdonaldi. Aquaculture 2018, 491, 239–251. [Google Scholar] [CrossRef]
  262. Guerreiro, I.; Couto, A.; Pérez-Jiménez, A.; Oliva-Teles, A.; Enes, P. Gut morphology and hepatic oxidative status of European sea bass (Dicentrarchus labrax) juveniles fed plant feedstuffs or fishmeal-based diets supplemented with short-chain fructo-oligosaccharides and xylo-oligosaccharides. Br. J. Nutr. 2015, 114, 1975–1984. [Google Scholar] [CrossRef] [PubMed]
  263. Standen, B.T.; Rodiles, A.; Peggs, D.L.; Davies, S.J.; Santos, G.A.; Merrifield, D.L. Modulation of the intestinal microbiota and morphology of tilapia, Oreochromis niloticus, following the application of a multi-species probiotic. Appl. Microbiol. Biotechnol. 2015, 99, 8403–8417. [Google Scholar] [CrossRef]
  264. Pirarat, N.; Pinpimai, K.; Endo, M.; Katagiri, T.; Ponpornpisit, A.; Chansue, N.; Maita, M. Modulation of intestinal morphology and immunity in nile tilapia (Oreochromis niloticus) by Lactobacillus rhamnosus GG. Res. Vet. Sci. 2011, 91, e92–e97. [Google Scholar] [CrossRef]
  265. Yilmaz, S.; Yilmaz, E.; Dawood, M.A.O.; Ringø, E.; Ahmadifar, E.; Abdel-Latif, H.M.R. Probiotics, prebiotics, and synbiotics used to control vibriosis in fish: A review. Aquaculture 2022, 547, 737514. [Google Scholar] [CrossRef]
  266. Mugwanya, M.; Dawood, M.A.O.; Kimera, F.; Sewilam, H. Updating the role of probiotics, prebiotics, andsSynbiotics for Tilapia aquaculture as leading candidates for food sustainability: A Review. Probiotics Antimicrob. Proteins 2022, 14, 130–157. [Google Scholar] [CrossRef]
  267. Jiang, H.; Bian, Q.; Zeng, W.; Ren, P.; Sun, H.; Lin, Z.; Tang, Z.; Zhou, X.; Wang, Q.; Wang, Y.; et al. Oral delivery of Bacillus subtilis spores expressing grass carp reovirus VP4 protein produces protection against grass carp reovirus infection. Fish Shellfish Immunol. 2019, 84, 768–780. [Google Scholar] [CrossRef]
  268. Van Doan, H.; Hoseinifar, S.H.; Tapingkae, W.; Seel-audom, M.; Jaturasitha, S.; Dawood, M.A.O.; Wongmaneeprateep, S.; Thu, T.T.N.; Esteban, M.Á. Boosted growth performance, mucosal and serum immunity, and disease resistance Nile Tilapia (Oreochromis niloticus) fingerlings using corncob-derived xylooligosaccharide and Lactobacillus plantarum CR1T5. Probiotics Antimicrob. Proteins 2020, 12, 400–411. [Google Scholar] [CrossRef] [PubMed]
  269. Pérez-Sánchez, T.; Ruiz-Zarzuela, I.; de Blas, I.; Balcázar, J.L. Probiotics in aquaculture: A current assessment. Rev. Aquac. 2014, 6, 133–146. [Google Scholar] [CrossRef]
Table 1. Comparison of the main microorganisms found in the gastrointestinal tract (GIT) of mammals, freshwater, and saltwater fishes.
Table 1. Comparison of the main microorganisms found in the gastrointestinal tract (GIT) of mammals, freshwater, and saltwater fishes.
Amount of Bacteria in GIT Dominant Microbial Phyla Less Abundant Microbial Phyla References
Mammals108 microbial cells for gram of intestinal contentBacteroidetes,
Firmicutes
Proteobacteria,
Actinobacteria, Fusobacteria, Cyanobacteria
[65,66,67]
Freshwater fishes107 to 1011 microbial cells for gram of intestinal contentProteobacteria,
Fusobacteria, Firmicutes, Bacteroidetes, Actinobacteria
Aeromonas, Pseudomonas, Bacteroides type A
Verrucomicrobia,
Enterobacteriaceae, Micrococcus, Acinetobacter, Clostridium, Bacteroides type B, Fusarium, Plesiomonas
[11,12,68,69,70,71]
Saltwater fishes107 to 1011 microbial cells for gram of intestinal contentProteobacteria,
Fusobacteria, Firmicutes, Bacteroidetes, Actinobacteria
Vibrio, Pseudomonas, Acinetobacter, Corynebacterium, Alteromonas, Flavobacterium, Micrococcus
Verrucomicrobia[11,12,68,69,70]
Table 2. Comparison of the main microorganisms found in the gastrointestinal tract of fish species according to their feed habits. (SW—seawater; FW—freshwater; AE—anadromous euryhaline).
Table 2. Comparison of the main microorganisms found in the gastrointestinal tract of fish species according to their feed habits. (SW—seawater; FW—freshwater; AE—anadromous euryhaline).
Feed HabitsFish SpeciesDominant Microbial PhylaReferences
HerbivoresAcanthurus nigricans
SW
Bacteroidetes, Proteobacteria, Firmicutes[74]
Acanthurus sp.
SW
Epulopiscium[75]
Aplodactylus arctidens
SW
Clostridium, Eubacterium desmolans, Papillibacter cinnaminovorans[76]
Chlorurus sordidus
SW
Vibrio, Photobacterium[74]
Hermosilla azurea
SW
Enterovibrio, Bacteroides, Faecalibacterium,
Desulfovibrio
[77]
Kyphosus spp.
SW
Gamma-proteobacteria, Clostridia, Bacteroidia, Erysipelotrichia (Vibrionaceae, Pasteurellaceae, Erysipelotrichaceae)[78]
Odax pullus
SW
Clostridium[76]
Ctenopharyngodon idellus
FW
Clostridium, Citrobacter, Leptotrichia[72]
Megalobrama amblycephala
FW
Brevundimonas, Massilia, Curvibacter, Acinetobacter, Clostridium, Citrobacter, Leptotrichia[72,79]
CarnivoresAcipenser baerii
SW
Cetobacterium[80]
Chaenocephalus aceratus
SW
Photobacterium[81]
Cynoscion nebulosus
SW
Escherichia[82]
Epinephelus coioides
SW
Bacillus, Vibrio, Delftia, Psychroacter, Acinetobacter, Pseudomonas[83]
Fugu niphobles
SW
Vibrio, Pseudomonas, Flavobacterium[84]
Gadus morhua
SW
Clostridium, Vibrio[85]
Hippoglossus hippoglossus
SW
Vibrionaceae (in larval and juvenile stages), Photobacterium phosphoreum (in adults)[71]
Lutjanusn bohar
SW
Vibrio, Photobacterium[74]
Morone saxatilis
SW
Aeromonas, Pseudomonas, Vibrio[86]
Notothenia coriiceps
SW
Photobacterium, Vibrio[81]
Paralichthys lethostigma
SW
Clostridium, Photobacterium,[82]
Pomatomus saltatrix
SW
Vibrio, Pseudomonas, Enterobacteraceae[87]
Sciaenops ocellatus
SW
Mycoplasmataceae, Photobacterium, Cetobacterium, Clostridiaceae, Vibrio[82]
Sparus aurata
SW
Pseudomonas[88]
Salmo trutta trutta
SW
Aeromonas, Pseudomonas[89]
Salmo salar
AE
Acinetobacter junii, Mycoplasma, Lactobacillus, Photobacterium phosphoreum, Lactococcus, Bacillus[90]
Culter alburnus
FW
Cetobacterium, Halomonas[72]
Pelteobagrus fulvidraco
FW
Clostridium, Yersinia, Aeromonas,
Clostridiales
[79]
Siniperca chuatsi
FW
Cetobacterium, Halomonas[72]
OnnivoresGillichthys mirabilis
SW
Mycoplasma[91]
Lagodon rhomboides
SW
Clostridium, Mycoplasma, Photobacterium, Propionibacterium, Staphylococcus, Pseudomonas, Corynebacterium[82]
Cariassus auratus
FW
Brevundimonas, Massilia, Curvibacter, Delftia
Clostridium, Cetobacterium Halomonas
[72,79]
Cyprinus carpio
FW
Brevundimonas, Massilia, Curvibacter, Sphingobacteriales, Cetobacterium, Aeromonas, Chitinophaga, Halomonas, Clostridium[72,79]
ZooplanktivoresApogonidae
SW
Vibrionaceae, Pasteurellaceae, Vibrio harveyi, Shewanella sp., Endozoicomonas sp.[73]
Clupea harengis
SW
Pseudomonas, Alteromonas, Psychrobacter[92]
Pomacentridae
SW
Vibrionaceae, Pasteurellaceae, Vibrio harveyi, Shewanella sp., Endozoicomonas sp.[73]
Sardinella longiceps
SW
Achromobacter, Vibrio, Pseudomonas[93]
Scomber scombrus
SW
Psychrobacter, Vibrio, Shewanella[94]
Syngnathus scovelli
SW
Proteobacteria[82]
Table 3. Distribution of microorganisms along the gastrointestinal tract (GIT) of different fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline).
Table 3. Distribution of microorganisms along the gastrointestinal tract (GIT) of different fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline).
GITFish SpeciesDominant PhylaReferences
StomachSparus aurata
SW
Firmicutes, Proteobacteria, Bacteroidetes (Bacillales, Flavobacteriaceae)[97]
Sparus aurata
SW
Firmicutes, Proteobacteria, Actinobacteria[98]
Epinephelus awoara
SW
Firmicutes, Proteobacteria, Actinobacteria,
Deinococcus-Thermus, Planctomycete
[99]
Scophthalmus maximus
SW
Proteobacteria, Firmicutes, Bacteroidetes, Tenericutes, Actinobacteria, Cyanobacteria[100]
Oncorhynchus mykiss
FW
Proteobacteria, Firmicutes, Fusobacteria (Cetobacterium dominanti)[101]
Foregut
(oesophagus, stomach, pylorus)
Salmo salar
AE
Gadus morhua
SW
Proteobacteria
(Janthinobacterium, Pseudomonas, Acinetobacter, Vibrio)
[70,90]
Scophthalmus maximus
SW
Proteobacteria, Firmicutes, Tenericutes, Actinobacteria, Cyanobacteria[100]
MidgutSalmo salar
AE
Gadus morhua
SW
Proteobacteria
(Photobacterium phosphoreum, Pseudomonas)
[70,90]
Pelteobagrus
fulvidraco
FW
Firmicutes, Proteobacteria, Bacteriodetes, Fusobacteria[102]
Oncorhynchus mykiss
FW
Proteobacteria, Firmicutes, Fusobacteria, Bacteroidetes (Bacillus)[101]
Ctenopharyngodon idella
FW
Proteobacteria, Firmicutes, Actinobacteria (Anoxybacillus, Leuconostoc, Clostridium, Actinomyces, Citrobacter)[103]
HindgutSalmo salar
AE
Gadus morhua
SW
Vibrio, P. phosphoreum[70,90]
Scophthalmus maximus
SW
Proteobacteria, Firmicutes, Tenericutes[100]
Acipenser baerii
AE
Fusobacteria/Firmicutes, Chlamydiae, Bacteriodetes, Actinobacteria[80]
Danio rerio
FW
Firmicutes, Proteobatceria,
Bacteriodetes
[104]
Table 4. Administration of prebiotics acting as effective immunostimulants in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline; MOS—mannan-oligosaccharides; FOS—fructo-oligosaccharides; COS—chito-oligosaccharides; GOS—galacto-oligosaccharides).
Table 4. Administration of prebiotics acting as effective immunostimulants in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline; MOS—mannan-oligosaccharides; FOS—fructo-oligosaccharides; COS—chito-oligosaccharides; GOS—galacto-oligosaccharides).
PrebioticsFish SpeciesDosage and Timing of AdministrationInitial Weight
(g)
EffectsReferences
Arabinoxylan-oligosaccharidesAcipenser baerii
AE
2% for 28 days and 12 weeks20.00Increase of phagocytic activity of leukocytes[124]
β-glucanEpinephelus coioides
SW
1, 2 g/kg for 30 days6.40 ± 0.65Enhancing of innate immune responses[125]
β-glucan + MOSCyprinus carpio
FW
Immunogen®
at 0.5, 1, 1.5, 2.5 g/kg for 8 weeks
11.12 ± 0.55Increase of leukocyte count and resistance to Aereomonas hydrophila[126]
COSCyprinus carpio
FW
0.2% for 8 weeks24.90 ± 0.52Increase of lysozyme level[127]
COSScophthalmus maximus
SW
75, 150, 300, 600, 1200 mg/kg for 56 days2.10 ± 0.10Increase of phagocytic activity at doses > 300 mg/kg[128]
COSScophthalmus maximus
SW
2 g/kg for 56 days11.00Reduction of gene expression of pro-inflammatory cytokines[129]
FOSAtractosteus tropicus
SW
(Larvae)
7.5 g/kg for 15 days0.03 ± 0.006No significant increase in gene expression of pro-inflammatory cytokines[130]
FOSMegalobrama terminalis
FW
0.3, 0.6% for 8 weeks30.50 ± 0.50Increase of various factors of immune system (IgM and complement proteins)[131]
FOSOreochromis niloticus
FW
0.5, 1, 2, 4 g/kg for 8 weeks5.00 ± 0.02 gEnhancing of immune and antioxidant responses, and resistance to A. hydrophila
(optimum at 1 g/kg)
[132]
FOSSalmo salar
AE
1% for 4 months200.20 ± 0.60No changes on immune system parameters [133]
FOSRutilus rutilus
FW
1%, 2%, 3% for 7 weeks0.67 ± 0.03Increase of immunoglobulins and lysozyme activity [118]
FOSParalichthys olivaceus
SW
0.5% for 56 days21.00Increase of lysozyme activity[134]
FOSAcipenser stellatus
AE
1%, 2% for 11 weeks30.16 ± 0.14Increase of lysozyme activity at 1% dose[135]
FOSTrachinotus ovatus
SW
2, 4 g/kg for 56 days10.32Increase of lysozyme activity and immunoglobulins[136]
GOS Danio rerio
FW
0.5%, 1%, 2% for 56 days0.045 ± 0.001 Enhancement of immune system parameters[137]
InulinCyprinus carpio
FW
1, 2 g/kg for 60 days25.37 ± 0.22Enhancement of immunoglobulins of epithelial mucosa
(optimum at 2 g/kg)
[138]
InulinHuso huso
AE
(juveniles)
1%, 2%, 3% for 8 weeks16.14 ± 0.38Increase of leukocyte count[139]
InulinSparus orata
SW
10 g/kg for 2 and 4 weeks50.00Increase of immunglobulins IgM and immune responses, resistance to P. damselae[140]
InulinOreochromis niloticus
FW
5 g/kg for 1 and 2 months11.00Increase of di haematocrit level, lysozyme activity and resistance to A. hydrophila[120]
InulinLates calcarifer
FW
15, 20 g/kg for 60 days7.14 ± 0.05Enhancement of immune and blood parameters [141]
LevanCyprinus carpio
FW
0.1%, 0.2%, 0.5% for 75 days9.00 ± 0.50Increase of lysozyme level
(optimum at 0.5% dose)
[142]
LevanCyprinus carpio
FW
0.75% for 60 days3.31 ± 0.52Increase of myeloperoxidases and immunoglobulins [143]
LevanLabeo rohita
rohu
FW
0.1%, 0.25%, 0.50%, 0.75%, 1%, 1.25%
for 60 days
4.50 ± 0.14Increase of lysozyme level
(optimum at 1.25% dose)
[144]
LevanEpinephelus coioides
SW
5, 10, 25, 50 g/kg for 12 weeks6.00Increase of lysozyme level
(optimum at 25 g/kg)
[145]
MOSSalmo salar
AE
10 g/kg for 4 months200.20 ± 0.60Reduction of lysozyme activity[133]
MOSDicentrarchus labrax
SW
2%, 4% for 8 weeks116.00Increase of leukocyte activity and resistance to Vibrio anguillarum
at 4% dose
[123]
MOSOncorhynchus mykiss
FW
2 g/kg for 42 days30.00Increase of antibody titers and lysozyme[146]
MOSOncorhynchus mykiss
FW
0.4% for 12 weeks13.20Increase of phagocytic activity, haematocrit and resistance to Vibrio anguillarum[147]
MOSOncorhynchus mykiss
FW
0.25%, 0.5% for 12 weeks36.27 ± 0.42Increase of phagocytic activity, haematocrit and resistance to Aereomonas salmonicida
at 0.5% dose
[148]
MOSClarias gariepinus
FW
10 g/kg for 45 days35.00Increase of lysozyme activity[149]
MOSChanna striata
FW
2 g/Kg for 12 weeks10.00Increase of lysozyme activity[150]
MOSPangasianodon hypophthalmus
FW
0.2%, 0.4%, 0.6%, 0.8%
for 12 weeks
20.41 ± 1.64Enhancement of immune parameters (immunoglobulins, lysozyme, leukocyte count) and resistance to A. hydrophila[151]
MOSSciaenops ocellatus
FW
10 g/kg for 6–8 weeks10.00Increase of lysozyme activity[152]
MOSGadus morhua
SW
1 g/kg for 5 weeks90.00Increase of expression of interleukins and resistance to V. anguillarum[153]
Nucleotides
(NucleoforceFish™)
Sparus aurata
SW
250,
500 mg/kg
for 150 days
0.36 ± 0.002Increase of gene expression of interleukins, hepcidin and B receptors of T cells[154]
Sodium alginateEpinephelus coioides
SW
10 g/kg for 12 days19.50 ± 0.50Increase of immune responses (complement system, phagocytic activity, lysozyme)[155]
StachyoseScophthalmus maximus
SW
1.25%, 5% for 12 weeks4.63 ± 0.01Inhibition of microbial pathogen growth and increase of beneficial ones
(optimum at 5% dose)
[156]
Thymol and carvacrolSparus aurata
SW
0.01% for 9 weeks26.00 ± 1.00Increase of interleukins, cytokines and inflammatory responses [157]
Thymol, carvacrol and essential oils from Allium spp.Sparus aurata
SW
0.5%40.30 ± 0.1Increase of immune innate responses at epithelial level and cortisol-mediated response, and lower microbial growth [158]
Table 5. Administration of probiotics acting as effective immunostimulants in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline; CFU—colony-forming unit).
Table 5. Administration of probiotics acting as effective immunostimulants in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline; CFU—colony-forming unit).
ProbioticsFish SpeciesDosage and Timing of AdministrationInitial Weight
(g)
EffectsReferences
Aspergillus oryzaeOreochromis niloticus
FW
1 × 1010 CFU/g19.50 ± 0.50Increase of non-specific immune and antioxidant responses[159]
Bacillus aerophilusLabeo rohita
FW
1 × 106, 1 × 107, 1 × 108, 1 × 109 CFU/g for 6 weeks35.00–40.00Increase of lysozyme and phagocytic activity, and resistance to A. hydrophila
(at 1 × 108 CFU/g)
[160]
Bacillus amyloliquefaciensLabeo rohita
FW
1 × 105, 1 × 107, 1 × 109 CFU/g for 70 days20.23Increase of antibody titers [161]
Bacillus amyloliquefacien, Bacillus pumilusPangasianodon Hypophthalmus
FW
1 × 108, 3 × 108, 5 × 108 CFU/g for 90 days15.30 ± 1.20Increase of lysozyme and phagocytic activity, and resistance to E. ictaluri with reduced death rate
(optimum at 5 × 108 CFU/g)
[162]
Bacillus licheniformisCyprinus carpio
FW
1 × 106, 1 × 107, 1 × 108 CFU/g for 60 days38.58 ± 0.42Increase of pro-and anti-inflammatory citokines and resistance to A. hydrophila
(optimum at 1 × 108 CFU/g)
[163]
Bacillus licheniformisOreochromis mossambicus FW1 × 105, 1 × 107 CFU/g for 4 weeks24.00 ± 2.50Increase of lysozyme activity and resistance to a A. hydrophila[164]
Bacillus licheniformis, Bacillus amyloliquefaciensCentropomus undecimalis SW
(Larvae)
1 × 1013 CFU/g for 28 days Reduction of lysozyme activity and not significant increase of SOD[165]
Bacillus licheniformis, Bacillus pumilusLabeo rohita
FW
1 × 108 CFU/g for 14 days50.00–60.00Increase of immune response and resistance to A. hydrophila[122]
Bacillus licheniformi, Bacillus subtilisCtenopharyngodon idella FW1 × 108 CFU/g for 4 weeks45.00General enhancement of immune parameters[166]
Bacillus licheniformis Dahb1Pangasius hypophthalmus
FW
1 × 105, 1 × 107 CFU/mL for 24 days15.00 ± 2.50Increase of lysozyme activity and myeloperoxidase
(optimum at 1 × 105 CFU/mL)
[167]
Bacillus methylotrophicus, Bacillus amyloliquefaciens, Bacillus licheniformisLabeo rohita
FW
1 × 107 cells/g for 60 days~50.00 Increase of immune and blood parameters, and resistance to A. hydrophila[168]
Bacillus pumilusOreochromis niloticus
FW
1 × 106, 1 × 107, 1 × 108, 1 × 109 CFU/kg for 30 days and 4 months50.00Increase of phagocytic activity and resistance to Streptococcus agalactiae
(at 1 × 108, 1 × 109 CFU/kg)
[169]
Bacillus pumilus A97Trachinotus ovatus
SW
1 × 108 CFU/g for 56 days5.95 ± 1.69 Increase of expression of genes of non-specific immune responses (Toll-like receptor) and resistance to Vibrio ponticus[170]
Bacillus pumilus, Bacillus clausiiEpinephelus coioides
SW
(Larvae)
1 × 106 bacteria/mL for 28 days Increased resistance to Aliivibrio fischeri, Vibrio
scophthalmi and Vibrio sp.
[171]
Bacillus spp., Enterococcus spp., Lactobacillus spp. (AquaStar®) Oreochromis niloticus
FW
3 g/kg for 6 weeks29.02 ± 0.33Increase of expression of genes of immune responses (interleukins and cytokines), intraepithelial leukocytes and intestinal mucous cells[172]
Bacillus subtilisOncorhynchus mykiss
FW
1 × 104, 1 × 105, 1 × 106, 1 × 107, 1 × 108, 1 × 109 CFU/g for 14 days30.00Increase of leukocytes, lysozyme activity and resistance to Aeromonas sp.
(optimum at 1 × 107 CFU/g)
[121]
Bacillus subtilisAnguilla japonica
FW
1 × 106, 1 × 107, 1 × 108 CFU/g for 8 weeks8.29 ± 0.06Increase of lysozyme activity and IgM
(optimum at 1 × 107 CFU/g)
[173]
Bacillus thuringiensis, Bacillus cereusLates calcarifer
FW
1 × 1012 CFU/kg for 35 days75.00 ± 0.60Enhancement of antioxidant response resistance to pathogens (V. harvey)[174]
Bacillus velezensis TPS3N, Bacillus subtilis TPS, Bacillus amyloliquefaciens TPS1Oreochromis niloticus
FW
1 × 108 CFU/mL for 4 weeks46.24 ± 0.48Increase of lysozyme activity, IgM at epithelial mucosa and intestine, survival rate to A. hydrophila[175]
Bacillus amyloliquefaciensOreochromis niloticus
FW
2 × 106 CFU/g for 60 days1.49 ± 0.15Enhancement of immune parameters and resistance to A. hydrophila[176]
Bacillus cereus, Bacillus subtilisOreochromis niloticus
FW
0.5 × 108, 1 × 108 CFU/g for 90 days0.20 ± 0.05Enhancement of immune status and resistance to diseases
(optimum at 1 × 108 CFU/g)
[177]
Bacillus circulansCatla catla
SW
2 × 104, 2 × 105, 2 × 106 CFU/100 g for 60 days6.48 ± 0.04Enhancement of non-specific immune parameters
(optimum at 2 × 105 CFU/100 g)
[178]
Bacillus pumilus, Bacillus clausiiEpinephelus coioides
SW
(Larvae)
2 × 106 bacetria/mL for 28 days Increased resistance to Aliivibrio fischeri, Vibrio scophthalmi, and Vibrio sp.[171]
Bacillus spp. + Lactobacillus spp. + S. cerevisiae Paralichthys olivaceus
SW
1 × 108, 1 × 109 CFU/kg for 12 weeks13.50 ± 0.01Enhancement of immune parameters[179]
Bacillus subtilisOreochromis niloticus
FW
0, 1, 2, 3, 4 g (1.19 × 108 CFU/g)/kg for 50 days14.82 ± 0.42Enhancement of immune responses (optimum at 3 g/kg)[180]
Bacillus subtilisOreochromis niloticus
FW
1.1 × 105 CFU/g for 84 days5.26 ± 0.06Enhancement of blood parameters (haematocrit and leukocytes)[181]
Bacillus aeriusPangasius bocourti
FW
1 × 107 CFU/g for 60 days69.00Increase of phagocytic activity, lysozyme, complement, and resistance to A. hydrophila[182]
Debaryomyces hansenii BCS004Sparus aurata
SW
106 CFUg−1 for 4 weeks80.00 ± 5.00Enhancement of immune parameters (phagocytic activity, increase of IgM)[183]
Enterococcus casseliflavus
EC-001
Cyprinus carpio
FW
1 × 1010, 1 × 1011, 1 × 1012 CFU/kg for 56 days12.00 ± 0.50Increase of immune response and resistance to infections
(optimum at 1 × 1012 CFU/kg)
[184]
Enterococcus faecalisOncorhynchus mykiss
FW
5 × 108 CFU/g for 30 days50.00Increase of resistance to L. garvieae CECT 527[185]
Enterococcus faeciumRutilus rutilus caspicus
AE
1 × 107, 1 × 108 CFU/g12.00Increase of immunoglobulins and lysozyme activity [186]
Enterococcus faeciumOreochromis niloticus
FW
1 × 107 CFU/mL for 40 days6.83 ± 0.18Enhancement of immune parameters[116]
L. delbrueckii sp. delbrueckii AS13BDicentrarchus labrax
SW
(Larvae)
1 × 105 bacteria/cm3 for 74 days Increase of T cells and granulocytes of intestinal mucosa [187]
Lactobacillus plantarum (alone);
Bacillus subtilis,
L. plantarum,
L. rhamnosus,
L. acidophilus,
L. delbrueckii (in mixture)
Oreochromis niloticus
FW
1 × 108 CFU/g for 112 days (by biofloc system)8.63 ± 3.35Increase of immune and antioxidant response, and resistance to pathogens (A. hydrophila)[188]
Lactobacillus rhamnosus ATCC 7469Oncorhynchus mykiss
FW
1 × 109 CFU/kg for 60 days18.41 ± 0.32Enhancement of non-specific immune parameters, gene expression of IL-1 and TNF, antioxidant activity and resistance to pathogens (Yersinia ruckeri)[189]
Lactobacillus acidophilusXiphophorus helleri
FW
1.5 × 108, 3 × 108, 6 × 108 CFU/g for 70 days0.03 ± 0.001Increase of gene expression of immune responses
(optimum at 6 × 108 CFU/g)
[190]
Lactobacillus acidophilusClarias gariepinus
FW
3 × 107 CFU/g for 21 days5.31 ± 0.10Enhancement of blood parameters[191]
Lactobacillus acidophilusCyprinus carpio
FW
1 × 102, 1 × 104, 1 × 106 CFU/kg for 56 days21.34 ± 1.85Increase of innate immune and antioxidant responses
(optimum at 1 × 106 CFU/kg)
[192]
Lactobacillus delbrueckiiCyprinus Carpio
FW
1 × 106, 1 × 107 CFU/g for 8 weeks1.05 ± 0.03Enhancement of intestinal immune parameters and resistance to A. hydrophila[193]
Lactobacillus fermentumCyprinus carpio
FW
1 × 108 CFU/g for 56 days3.90 ± 0.20 Enhancement of immune responses and resistance to pathologies[194]
Lactobacillus plantarumOreochromis niloticus
FW
3.4 × 108, 6.8 × 108, 1.3 × 109 CFU/g for 40 days24.50Increase of gene expression of interleukins and cytokines
(optimum at 6.8 × 108 CFU/g)
[195]
Lactobacillus plantarumOreochromis niloticus
FW
1 × 108 CFU/g for 12 weeks4.90 ± 0.04Increase of lysozyme level in serum and epithelial mucosa, phagocytic activity and resistance to Streptococcus agalctiae[112]
Lactobacillus plantarumOreochromis niloticus
FW
1 × 108 CFU/g for 84 days5.92 ± 0.08 Increase of immune response in serum and epithelial mucosa[196]
Lactobacillus plantarum, Bacillus velezensis H3.1Oreochromis niloticus
FW
1 × 107 CFU/g Bacillus velezensis;
1 × 108 CFU/g Lactobacillus plantarum for 30 days
21.80 ± 0.03Increase of lysozyme and phagocytic activities, complement proteins and resistance to Streptococcus agalactiae[197]
Lactobacillus rhamnosus JCM1136, Lactococcus lactis subsp. lactis JCM5805Oreochromis niloticus
FW
(Juveniles)
1 × 108 CFU/g for 6 weeks0.20 ± 0.05Increase of gene expression of immune response (interleukins, interferons) and survival rate[198]
Lactobacillus spp., Bacillus subtilis, Bifidobacterium bifidumAcipenser baerii
AE
(Fingerlings)
1 × 106, 2 × 106, 3 × 106 CFU/g for 56 days10.50 ± 0.14Increase of lysozyme and IgM levels
(optimum at 3 × 106 CFU/g)
[199]
Lactococcus garvieaeOreochromis niloticus
FW
1 × 107 CFU/g for 2 weeks50.00 ± 5.00 Increase of IgM and lysozyme activity[200]
Lactococcus lactisParalichthys olivaceus
SW
1 × 109 CFU/g for 8 weeks80.84 ± 9.37Enhancement of innate immune response and resistance to Streptococcus[201]
Lactococcus lactisCyprinus carpio
FW
5 × 108 CFU/g for 8 weeks33.07 ± 0.55Increase of pro-inflammatory cytokines[202]
Lactococcus lactisSparus aurata
SW
2 × 109, 5 × 109 CFU/kg for 12 weeks70.00–90.00Increase of gene expression of immune responses[9]
Lactobacillus rhamnosus GGOreochromis niloticus
FW
1 × 108 CFU/g for 2 weeks14.05 ± 0.42Increase of lysozyme activity, mucous cells at intestine and resistance to Aeromonas veroni[203]
Lactobacillus sakei PO11, Lb. plantarum PO23Paralichthys olivaceus
SW
1 × 1011 CFU/g for 27 days35.00 ± 5.00Increase of gene expression of immune responses
(interleukins and immunoglobulins)
[204]
Bacillus subtilis and Saccharomyces cerevisiaeOreochromis niloticus
FW
6 × 107 CFU/g for 56 days24.01 ± 0.02Enhancement of innate immune responses and resistance to pathologies[205]
Paenibacillus ehimensisOreochromis niloticus
FW
1 × 106, 1 × 107 CFU/g for 60 days5.53 ± 0.45 Enhancement of immune response and resistance to pathologies[206]
Paenibacillus polymyxaCyprinus carpio
FW
1 × 103, 1 × 104, 1 × 105 CFU/mL for 8 weeks23.17Increase of lysozyme activity, myeloperoxidase, and resistance to
A. hydrophila
(optimum at 1 × 103 CFU/mL)
[207]
Pediococcus acidilacticiOreochromis niloticus
FW
1 × 1010 CFU/kg for 32 days175.00Increase of lysozyme activity and leukocytes[208]
Pediococcus acidilacticiOreochromis niloticus
FW
2.81 × 106 CFU/g for 6 weeks9.19 ± 0.04Increase of leukocytes and intestinal mucous cells [209]
Pediococcus acidilacticiCyprinus carpio
FW
6 × 108 CFU/g for 60 days10.00 ± 2.50Increase of Ig and proteases of epithelial mucosa, gene expression of lysozyme and TNF-α[210]
Pediococcus pentosaceusCtenopharyngodon idella FW1 × 109 CFU/g for 30 days32.10 ± 9.00Increase of gene expression of immune response and resistance to A. hydrophila[211]
Pseudomonas fluorescens biovar I, II and IIIOreochromis niloticus
FW
1 × 1011 CFU/kg for 45 days2.93 ± 0.22Enhancement of non-specific immune and blood parameters [212]
Psychrobacter namhaensis SO89Oreochromis niloticus
FW
2.8 × 107, 5.6 × 107 for 50 days4.58 ± 0.14Increase of haematocrit, leukocytes and other immune parameters [117]
Saccharomyces cerevisiaePangasianodon hypophthalmus
FW
1 × 109, 1 × 1011 CFU/kg for 120 days55.00–65.00Enhancement of immune parameters (immunglobulines, lysozyme activity)[213]
Saccharomyces cerevisiaeOreochromis niloticus
FW
0.25, 0.50, 1, 2, 5 g/kg for 12 weeks0.33Enhancement of immune parameters and resistance to pathologies
(optimum at 1 g/kg)
[214]
Shewanella putrefaciens Pdp11Sparus orata
SW
1 × 108 CFU/g for 15–30 days104.20 ± 7.40Increase of lysozyme activity and gene expression of immune responses[215]
Shewanella xiamenensis, Aeromonas veroniiCtenopharyngodon idella FW1 × 108 cell/g for 28 days35.00 ± 5.00Enhancement of immune parameters, and resistance to A. hydrophila[216]
Vibrio lentusDicentrarchus labrax
SW
(Larvae)
1 × 106 CFU/mL for 10 days post-hatching Increase of transcription of genes of immune responses[217]
Weissella confusaOncorhynchus mykiss
FW
1.5 × 1010, 3 × 1010, 4.5 × 1010 CFU/kg for 60 days115.00 ± 2.60Increase of lysozyme activity and expression of INF-γ, TNF-α and IL-8 at 3 × 1010 CFU/kg[218]
Table 6. Administration of prebiotics enhancing the gut efficiency and growth performance in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline; MOS—mannan-oligosaccharides; GOS—galacto-oligosaccharides; COS—chito-oligosaccarides; FOS—fructo-oligosaccharides).
Table 6. Administration of prebiotics enhancing the gut efficiency and growth performance in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline; MOS—mannan-oligosaccharides; GOS—galacto-oligosaccharides; COS—chito-oligosaccarides; FOS—fructo-oligosaccharides).
PrebioticsFish SpeciesDosage and Timing of AdministrationInitial Weight
(g)
EffectsReferences
β-glucan + MOSOreochromis niloticus
FW
(Fingerlings)
1.5, 3 g/kg for 60 days8.70 ± 0.40Enhancement of growth performance and histological parameters of the intestine[224]
β-glucan, GOS, MOSChanna striata
FW
2 g/kg (β-glucan), 5 g/kg (GOS), 5 g/kg (MOS) for 16 weeks 22.40Enhancement of growth performance and protein digestibility [225]
β-glucan + MOSTor grypus
FW
1.5% (MOS), 1.5% (β-glucan) for 90 days35.00 ± 1.20Enhancement of growth performance and increase of protein content in fish[226]
COSScophthalmus maximus
SW
2 g/kg for 56 days11.00Enhancement of growth performance and positive effects on intestine structure [129]
FOSAtractosteus tropicus
SW
(Larvae)
7.5 g/kg for 15 days0.03Enhancement of growth performance and digestive capacity[130]
FOS Sciaenops ocellatus
FW
1% for 8 weeks7.00Increase in height of intestinal microvilli [152]
FOS Oreochromis niloticus
FW
0.5, 1, 2, 4 g/kg for 8 weeks5.00 ± 0.02Increase of amylase enzyme and height of anterior intestinal microvilli with enhancement of growth parameters
(optimum at 1 g/kg)
[132]
GOS Sciaenops ocellatus
FW
1% for 8 weeks7.00Increase in height of intestinal microvilli[152]
GOS Pangasius hypophthalmus FW10 g/kg for 12 weeks16.45 ± 0.07Increase of digestive enzymes activity and length of posterior intestinal microvilli [227]
Inosine,
monophosphate inosine
Pagrus major
SW
0.2%, 0.4%, 0.6%, 0.8% for 10 weeks6.60Increase in height of enterocytes, intestinal folds and microvilli
(optimum at 0.4%)
[228]
InulinSciaenops ocellatus
FW
1% for 8 weeks7.00Increase in height of intestinal microvilli[152]
InulinLates calcarifer
FW
15, 20 g/kg for 60 days7.14 ± 0.05Increase of intestinal adsorbent surface [141]
MOS Sciaenops ocellatus
FW
1% for 8 weeks7.00Increase in height of intestinal microvilli[152]
MOS Ctenopharyngodon idella
FW
200, 400, 600, 800 and 1000 mg/kg for 60 days215.85 ± 0.30Enhancement of the intestinal health status in fish with enteritis by Aeromonas hydrophila
(optimum at 400 mg/kg)
[229]
NucleotidesSciaenops ocellatus
FW
1% for 6 weeks7.10Increase in height of intestinal folds and microvilli[230]
Nucleotides
(NucleoforceFish™)
Sparus aurata
SW
250,
500 mg/kg
for 150 days
0.36 ± 0.002Enhancement of growth parameters [154]
Essential oils of Ocimum basilicum (EOOB)Oreochromis niloticus
FW
0.25, 0.5, 1, 2 mL of EOOB/kg for 45 days12.13 ± 0.11 Enhancement of digestive enzymes activity (amylases)
(optimum at 1 mL/kg)
[219]
StachyoseScophthalmus maximus L.
SW
1.25%, 5%
for 12 weeks
4.63 ± 0.01Increase of gene expression of occludins and ZO-1, and intestinal health
(optimum at 5%)
[156]
Thymol and carvacrolSparus aurata
SW
From 0.005 to 0.03% for 9 weeks26.00–27.00Increase in number of enterocytes e mucous cells in intestinal epithelium, and enhancement of feed gain ratio at 0.01%[157]
Table 7. Administration of probiotics enhancing the gut efficiency and growth performance in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline).
Table 7. Administration of probiotics enhancing the gut efficiency and growth performance in different aquaculture fish species. (SW—seawater; FW—freshwater; AE—anadromous euryhaline).
ProbioticsFish SpeciesDosage and Timing of AdministrationInitial Weight
(g)
EffectsReferences
Acinetobacter KU011THClarias macrocephalus
FW
1 × 105, 1 × 107, 1 × 109 CFU/kg and 1 × 103, 1 × 104, 1 × 105 CFU/mL 150.00 Enhancement of growth performance at 1 × 109 CFU/kg and 1 × 103 CFU/mL[231]
Acinetobacter, Vibrio, Bacillus, Alcaligens, Photobacterium, FlavobacteriumOncorhynchus mykiss
FW
3.9 × 109 CFU/g for 56 days113.00 ± 10.40Enhancement of growth performance[232]
Bacillus amyloliquefaciensOreochromis niloticus
FW
60 mg/kg for 42 days39.00 Enhancement of growth performance[233]
Bacillus coagulans, Rhodopseudomonas palustris, Lactobacillus acidophilusCtenopharyngodon idella
FW
1 × 106 CFU/g for 60 days2.10 ± 0.09Increase in weight and digestive enzymes activity [234]
Bacillus licheniformisCyprinus carpio
FW
1 × 106, 1 × 107, 1 × 108 CFU/g for 60 days38.58 ± 0.42Enhancement of growth performance and increase in height of intestinal microvilli
(optimum at 1 × 108 CFU/g)
[163]
Bacillus thuringiensis, Bacillus cereusLates calcarifer
FW
1 × 1012 CFU/kg for 42 days50.00 ± 0.50 Enhancement of growth performance[174]
Bacillus velezensis
TPS3N,
Bacillus subtilis TPS4, Bacillus amyloliquefaciens
Oreochromis niloticus
FW
1 × 108 CFU/mL for 4 weeks46.24 ± 0.48Enhancement of digestive enzymes activity and increase in thickness and length of intestinal microvilli, and number of mucous cells [175]
Bacillus sp.Cyprinus carpio
FW
~1 × 1011 CFU/g for 60 days5.90–7.10Enhancement of digestive enzymes activity and food conversion rate [235]
Bacillus sp., Alcaligenes sp.Tor tambroides
FW
1 × 108 CFU/g for 90 days1.39 ± 0.06Enhancement of growth rate and digestive enzymes activity, and positive effects on intestinal morphology [236]
Bacillus sp. SJ-10, Lactobacillus plantarumParalichthys olivaceus
SW
1 × 108 CFU/g for 8 weeks14.92 ± 0.21Not significant enhancement of some histo-morphological intestinal parameters and increase in some digestive enzymes activity [237]
Bacillus subtilisOreochromis niloticus
FW
1.1 × 105 CFU/g for 84 days5.26 ± 0.06Enhancement of growth performance [181]
Bacillus subtilis and Lactobacillus rhamnosusLabeo rohita
FW
(Fingerlings)
1 × 107 CFU/g for 60 days0.38 ± 0.015 Enhancement of growth performance and nutrients usage [238]
Bacillus subtilis,
Bacillus licheniformis,
Bacillus pumilus
Oreochromis niloticus
FW
20 mg/kg for 49 days34.56 ± 0.05 Increase in number of microvilli and lysozyme activity, no changes in growth performance and blood and immune parameters[239]
Bacillus subtilis,
Bacillus licheniformis,
Bacillus pumilus
Panganodon hypophthalmus
FW
1 × 1010 CFU/g for 110 days0.03Enhancement of growth performance and survival rate[240]
Bacillus, Bifidobacterium, Enterococcus, Lactobacillus, Pediococcus sp.Oreochromis niloticus
FW
(Fingerlings)
1, 2 g/kg for 90 days5.00Increase in final weight and muscle mass [241]
Enterococcus casseliflavus
(EC-001)
Cyprinus carpio
FW
1 × 1010, 1 × 1011, 1 × 1012 CFU/kg for 56 days12.00 ± 0.50Increase in weight at
1 × 1011 and 1 × 1012 CFU/kg
[184]
Enterococcus faeciumOreochromis niloticus
FW
1 × 107 CFU/mL for 40 days6.83 ± 0.18Increase in final weight[116]
Lactobacillus. acidophilusCyprinus carpio FW1 × 102, 1 × 104, 1 × 106 CFU/kg for 56 days21.34 ± 1.85 Enhancement of length and width of microvilli of growth performance (optimum at 1 × 106 CFU/kg)[192]
Lactobacillus rhamnosus ATCC 7469Oncorhynchus mykiss
FW
1 × 109 CFU/kg for 60 days18.41 ± 0.32Enhancement of growth parameters[189]
Lactobacillus acidophilusXiphophorus helleri
FW
1.5 × 108, 3 × 108, 6 × 108 CFU/g for 70 days3.90 ± 0.20 Enhancement of growth performance
(optimum at 6 × 108 CFU/g)
[190]
Lactobacillus caseiCyprinus carpio
FW
1 × 108 CFU/g for 30 days68.40 ± 5.90Enhancement of intestinal enzyme parameters[242]
Lactobacillus casei
(Yakult®)
Tor tambra
FW
(Larvae)
0, 5, 10, 15 mL/kg for 80 days0.03 Enhancement of growth performance
and nutrients usage
(optimum at 10 mL/kg)
[243]
Lactobacillus delbrueckiiPoecilia sphenops
FW
0.8 g/kg for 30 days0.90 ± 0.02 Enhancement of growth performance and survival rate[244]
Lactobacillus plantarumOreochromis niloticus
FW
1 × 108 CFU/g for 84 days5.92 ± 0.08 Enhancement of growth performance[196]
Lactobacillus. rhamnosus GGOreochromis niloticus
FW
1 × 108 CFU/g for 2 weeks14.05 ± 0.42Enhancement of histo-morphological intestinal parameters [203]
Lactobacillus rhamnosus JCM1136, Lactobacillus lactis subsp. lactis JCM1136Oreochromis niloticus
FW
1 × 108 CFU/g for 6 weeks0.20 ± 0.05Increase in length and density of intestinal microvilli [198]
Micrococcus luteusOreochromis niloticus
FW
1 × 107 cells/g for 90 days2.35 ± 0.10Enhancement of growth performance[245]
Micrococcus MCCB 104, Bacillus MCCB 101Oreochromis mossambicus
FW
1 × 103 CFU per individual for 28 days Increase of intestinal and hepato-pancreatic enzymes activity[246]
Pediococcus acidilacticiOreochromis niloticus
FW
1 × 1010 CFU/kg for 32 days175.00No significant change in growth parameters [208]
Pediococcus pentosaceus SL001Ctenopharyngodon idella
FW
1 × 109 CFU/g for 30 days32.10 ± 9.00Increase in number of goblet cells and length of intestinal villi [211]
Pseudomonas fluorescens biovar I, II and IIIOreochromis niloticus
FW
1 × 1011 CFU/kg for 45 days2.93 ± 0.22Enhancement of growth performance and blood and non-specific immune parameters [212]
Saccharomyces cerevisiaePangasianodon hypophthalmus
FW
1 × 109, 1 × 1011 CFU/kg for 120 days55.00–65.00Enhancement of growth rate, food conversion rate and immune parameters [213]
Saccharomyces cerevisiae
(Angel Yeast Co. Ltd., China)
Oreochromis niloticus
FW
(Fingerlings)
0, 1, 2, 4 g/kg for 60 days 7.55 ± 1.25Increase of growth performance and change of histo-morphological intestinal parameters
(optimum at 4 g/kg)
[247]
Saccharomyces cerevisiaeLabeo rohita
FW
(Fingerlings)
0, 1, 2, 4 g/kg for 90 days5.69 ± 0.02Enhancement of growth rate, histological parameters and nutrients usage
(optimum at 4 g/Kg)
[248]
Saccharomyces cerevisiaeMystus cavasius
FW
0, 0.5, 1, 1.5 g/kg for 75 days0.50 ± 0.20 Enhancement of growth performance
(optimum at 1 g/kg)
[249]
Streptomyces spp.Xiphophorus helleri
FW
10 g/kg for 50 days0.60Enhancement of growth performance and nutrients usage[250]
Table 8. The impact of some prebiotics/probiotics on the gut of aquaculture fish species assessed by histological endpoints. (↑: increase; ↓: decrease; =: no change).
Table 8. The impact of some prebiotics/probiotics on the gut of aquaculture fish species assessed by histological endpoints. (↑: increase; ↓: decrease; =: no change).
Prebiotics/ProbioticsFish SpeciesHistological EndpointsReferences
Lactic acid bacteria SaccharomycesOreochromis niloticus↑ intestinal perimeter ratio, mucosal fold length, goblet cells and leukocytes[222]
P. acidilactici MA18/5M + short-chain fructo-oligosaccharidesSalmo salar↑ mucosal fold height, intraepithelial leukocytes, granulocytes, and goblet cells[258]
Bacillus coagulans, B. licheniformis and Paenibacillus polymyxaSillago sihama↑ villus height, villus width, muscle thickness, and crypt depth[223]
Bacillus subtilis + inulinSparus aurata L.↑ enterocyte vacuolisation, microvilli disruption[221]
Lactococcus lactisSparus aurata= mucosal folds, connective tissue, lamina propria of simple folds, supranuclear vacuoles[9]
NucleotidesSciaenops ocellatus↑ microvillus height, enterocyte height, = fold height[230]
Lactobacillus brevis and L. buchneriSeriola dumerili↓ thickness of submucosa layer[259]
Bacillus licheniformis, Bacillus subtilis + Saccharomyces cerevisiaeAcipenser persicus↑ goblet and mucosal cell number ↑ villus length and width[260]
Aquablend® + GroBiotic®Totoaba macdonaldi↑ fold length, = fold width, = enterocyte height, = microvilli height[261]
Short-chain fructo-oligosaccharides and xylo-oligosaccharidesDicentrarchus labrax↓ mucosal folds, = enterocytes nucleous, ↑ supranuclear absorptive vacuolisation, = hyperplasia in the lamina propria and submucosa, ↑ intraepithelial leucocytes[262]
Bacillus velezensis TPS3N, Bacillus subtilisTPS4, Bacillus amyloliquefaciens TPS17 (as singular and combined suspension)Oreochromis niloticus↑ villus length and width, goblet cells count, intestinal epithelial muscle thickness[175]
AquaStar® Growout (Lactobacillus reuteri, Bacillus subtilis, Enterococcus faecium and Pediococcus acidilactici)Oreochromis niloticus↑ intraepithelial leucocytes, microvilli density[172,209,263]
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

De Marco, G.; Cappello, T.; Maisano, M. Histomorphological Changes in Fish Gut in Response to Prebiotics and Probiotics Treatment to Improve Their Health Status: A Review. Animals 2023, 13, 2860. https://doi.org/10.3390/ani13182860

AMA Style

De Marco G, Cappello T, Maisano M. Histomorphological Changes in Fish Gut in Response to Prebiotics and Probiotics Treatment to Improve Their Health Status: A Review. Animals. 2023; 13(18):2860. https://doi.org/10.3390/ani13182860

Chicago/Turabian Style

De Marco, Giuseppe, Tiziana Cappello, and Maria Maisano. 2023. "Histomorphological Changes in Fish Gut in Response to Prebiotics and Probiotics Treatment to Improve Their Health Status: A Review" Animals 13, no. 18: 2860. https://doi.org/10.3390/ani13182860

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