Histomorphological Changes in Fish Gut after Prebiotics/Probiotics Treatment: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

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.

  • fish gastrointestinal tract (GIT)
  • feed additives
  • microbial communities
  • fish welfare assessment
  • histological assays

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].

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].
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 [19]. 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 [20]. 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 [21]. 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 [22]. 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 [19][23]
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 [24][25]. 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. 
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 [26]. 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 [27]. However, no clear correlation seems to exist between the pyloric caeca shape and feed habits of fish [28][29].
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][29] and variations with regards to the mucosal fold and muscle thickness [20]. 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][21][30][31].

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 [32]. The immune system avoids the invasion of opportunistic and pathogenic microorganisms, whilst simultaneously preserving and enhancing the proliferation of commensal bacteria [33][34][35]. 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 [36]. The innate immunity is composed of physical barriers, humoral, and cellular components [32]. 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 [36]. AMPs can guarantee general microbiota homeostasis thanks to their antimicrobial activity of immunity modulation [37].
  • 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 [38]. In fish, the role of lysozymes against pathogens has been demonstrated in several studies [39][40][41][42], 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 [43]. 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 [44][45]. 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 [45].
  • 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 [35][36][43].
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 [35].

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][46]. 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][47]. 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][46] 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.
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 [48]
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 [46]
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][49][50][51] 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
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 [52]. 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 [53][54]

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 [55]. 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 [56][57]. Antibiotics, particularly those present in rivers, have the ability to permeate soil and enhance the selection and growth of antibiotic-resistant bacteria [58]. The variety of environmental implications notwithstanding, antibiotic usage can lead to a substantial loss of gut microbiota variability [59]. Generally, the conditions which fish are subjected to in fishery farms might be a cause of stress.
According to numerous recent studies [13][60][61][62][63], 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 [64][65], food absorption, and digestion, and has shown a positive effect on the growth performance of fish [66][67][68]. From an immunological perspective, the activity of these feed additives does not merely promote the proliferation of commensal bacteria at the detriment of pathogens. 
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 [66][69][70][71] 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 [68]. 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 [72]. 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.

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 [55]. 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 [73]. Among the different approaches employed [74][75], histomorphological assays represent a good biomarker for the assessment of the welfare of aquatic organisms [76][77][78][79][80] 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 [81], 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 [82]. 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.
For instance, a number of histomorphological parameters were used to assess the impact of lactic acid bacteria and yeast on Oreochromis niloticus [82], 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 [83]. 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 [84], 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 [85].
The primary aim of these current research studies is to determine the optimal feed additives for each aquaculture species while also considering the dosage [86]. 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) [72]
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 [87]. Lactobacillus spp., along with Bacillus spp., are probably among the most widely used probiotics in aquaculture [88][89][90][91]
To accurately integrate and interpret the histological results, the use of a continuous scale scoring system may be helpful, as applied and reported in [92] 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 [79][80][89][93]. In [89], 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 [64]

This entry is adapted from the peer-reviewed paper 10.3390/ani13182860

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.
  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.
  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.
  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.
  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.
  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.
  7. Egerton, S.; Culloty, S.; Whooley, J.; Stanton, C.; Ross, R.P. The gut microbiota of marine fish. Front. Microbiol. 2018, 9, 873.
  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.
  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.
  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.
  11. Nayak, S.K. Probiotics and immunity: A fish perspective. Fish Shellfish Immunol. 2010, 29, 2–14.
  12. Nayak, S.K. Role of gastrointestinal microbiota in fish. Aquac. Res. 2010, 41, 1553–1573.
  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.
  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.
  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.
  16. Hancz, C. Application of probiotics for environmentally friendly and sustainable aquaculture: A Review. Sustainability 2022, 14, 15479.
  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.
  18. Takahashi, T.; Koblmüller, S. The adaptive radiation of cichlid fish in Lake Tanganyika: A morphological perspective. Int. J. Evol. Biol. 2011, 2011, 620754.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. Stevens, C.E.; Hume, I.D. Comparative Physiology of the Vertebrate Digestive System; Cambridge University Press: Cambridge, UK, 2004.
  26. Buddington, R.K.; Diamond, J.M. Pyloric ceca of fish: A “new” absorptive organ. Am. J. Physiol. Liver Physiol. 1987, 252, G65–G76.
  27. 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.
  28. Olsen, R.; Ringø, E. Lipid digestibility in fish: A review. Recent Res. Dev. Lipid Res. 1997, 1, 199–265.
  29. Ringø, E.; Olsen, R.E.; Mayhew, T.M.; Myklebust, R. Electron microscopy of the intestinal microflora of fish. Aquaculture 2003, 227, 395–415.
  30. 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.
  31. 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.
  32. 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.
  33. 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.
  34. Kelly, C.; Salinas, I. Under pressure: Interactions between commensal microbiota and the teleost immune system. Front. Immunol. 2017, 8, 559.
  35. 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.
  36. 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.
  37. 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.
  38. 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. Abram, Q.H.; Dixon, B.; Katzenback, B.A. Impacts of low temperature on the teleost immune system. Biology 2017, 6, 39.
  45. 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.
  46. Wang, A.R.; Ran, C.; Ringø, E.; Zhou, Z.G. Progress in fish gastrointestinal microbiota research. Rev. Aquac. 2018, 10, 626–640.
  47. Rombout, J.H.W.M.; Abelli, L.; Picchietti, S.; Scapigliati, G.; Kiron, V. Teleost intestinal immunology. Fish Shellfish Immunol. 2011, 31, 616–626.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. Chen, J.; Xie, S. Overview of sulfonamide biodegradation and the relevant pathways and microorganisms. Sci. Total Environ. 2018, 640–641, 1465–1477.
  57. 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.
  58. Thiele-Bruhn, S. Pharmaceutical antibiotic compounds in soils—A review. J. Plant Nutr. Soil Sci. 2003, 166, 145–167.
  59. 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.
  60. 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.
  61. 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.
  62. Dawood, M.A.O.; Koshio, S.; Abdel-Daim, M.M.; Van Doan, H. Probiotic application for sustainable aquaculture. Rev. Aquac. 2019, 11, 907–924.
  63. Chauhan, A.; Singh, R. Probiotics in aquaculture: A promising emerging alternative approach. Symbiosis 2019, 77, 99–113.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. Pérez-Sánchez, T.; Ruiz-Zarzuela, I.; de Blas, I.; Balcázar, J.L. Probiotics in aquaculture: A current assessment. Rev. Aquac. 2014, 6, 133–146.
More
This entry is offline, you can click here to edit this entry!
Video Production Service