Skip to main content

Debaryomyces hansenii supplementation in low fish meal diets promotes growth, modulates microbiota and enhances intestinal condition in juvenile marine fish

Abstract

Background

The development of a sustainable business model with social acceptance, makes necessary to develop new strategies to guarantee the growth, health, and well-being of farmed animals. Debaryomyces hansenii is a yeast species that can be used as a probiotic in aquaculture due to its capacity to i) promote cell proliferation and differentiation, ii) have immunostimulatory effects, iii) modulate gut microbiota, and/or iv) enhance the digestive function. To provide inside into the effects of D. hansenii on juveniles of gilthead seabream (Sparus aurata) condition, we integrated the evaluation of the main key performance indicators coupled with the integrative analysis of the intestine condition, through histological and microbiota state, and its transcriptomic profiling.

Results

After 70 days of a nutritional trial in which a diet with low levels of fishmeal (7%) was supplemented with 1.1% of D. hansenii (17.2 × 105 CFU), an increase of ca. 12% in somatic growth was observed together with an improvement in feed conversion in fish fed a yeast-supplemented diet. In terms of intestinal condition, this probiotic modulated gut microbiota without affecting the intestine cell organization, whereas an increase in the staining intensity of mucins rich in carboxylated and weakly sulphated glycoconjugates coupled with changes in the affinity for certain lectins were noted in goblet cells. Changes in microbiota were characterized by the reduction in abundance of several groups of Proteobacteria, especially those characterized as opportunistic groups. The microarrays-based transcriptomic analysis found 232 differential expressed genes in the anterior-mid intestine of S. aurata, that were mostly related to metabolic, antioxidant, immune, and symbiotic processes.

Conclusions

Dietary administration of D. hansenii enhanced somatic growth and improved feed efficiency parameters, results that were coupled to an improvement of intestinal condition as histochemical and transcriptomic tools indicated. This probiotic yeast stimulated host-microbiota interactions without altering the intestinal cell organization nor generating dysbiosis, which demonstrated its safety as a feed additive. At the transcriptomic level, D. hansenii promoted metabolic pathways, mainly protein-related, sphingolipid, and thymidylate pathways, in addition to enhance antioxidant-related intestinal mechanisms, and to regulate sentinel immune processes, potentiating the defensive capacity meanwhile maintaining the homeostatic status of the intestine.

Background

Yeasts are single-celled members of the fungi kingdom, widely distributed in nature, capable of living in extreme environments. Its biological relevance is determined by 1,500 species described so far, with a high impact on both plant and animal life [1], being also a pillar in the human food industry. Used for millennia due to their fermentative activity to produce highly demanded foods (bread, cheese, wine, beer, among others), for some years now yeasts have also been in the crosshair of the pharmaceutical industry [2]. Some authors have suggested their use as transport vehicles for bioactive compounds [3], drug delivery [4], or what is more widely considered, as natural probiotics for humans, livestock, and aquaculture species [5, 6]. Some of them have been isolated from the intestine of several species as part of their commensal microbiota, including fish [7]. In this sense, the reasons why some yeast species are highly interesting for their use as probiotics is because they may promote somatic growth, modulate the intestinal microbiota, as well as enhance host health and condition due to their contents in immunomodators like β-glucans among other bioactive compounds [8,9,10].

Several studies have recommended the inclusion of yeasts in livestock and aquaculture feeds, as an additive, due to their beneficial effects in key performance indicators related to growth and feed efficiency, and in terms of antioxidant performance, and immunity enhancement, among which is the resistance to infections [11,12,13]. On the other hand, yeasts and their metabolic products from food industry by-products could also be used to produce high quality alternative protein sources within a sustainable and circular economy approach [14,15,16]. It is known that yeasts are a rich source of nutrients and protein-related compounds with a wide range of bioactive activities. In this sense, yeasts cells contain different types of wall-related compounds, like glucans and polyamines (biologically active amines), which are essential for the maintenance of life [17]. Specifically, amines have been observed to be relevant to the normal cell development and are involved in several cellular processes, along with systemic benefits for the animals [18,19,20].

Among all yeast species, Saccharomyces cerevisiae might be the most studied yeast as a probiotic or alternative protein source. However, another yeast species that has received a lot of attention for its probiotic effect is Debaryomyces hansenii [21]. Contrary to other species, D. hansenii belongs to the normal microbiota of wild and cultivated animals [22], and it has been reported that it also promotes growth and development in various species at different stages, results that have been attributed to its glucans and polyamine content [21]. This probiotic has been tested at early life stages in aquatic species with promising results; for instance, larvae of Seriola rivoliana [23], Danio rerio [24], Dicentrarchus labrax [25] and Penaeus monodon [26] showed improved survival and development when fed a diet supplemented with D. hansenii. Similarly, other studies focused on juveniles and adults showed that a diet supplemented with this yeast had a positive effect on the host in terms of its physiological and immune responses, showing its capacity to counteract possible stress impacts as well as to enhance disease resistance [17, 27, 28], along with an improvement of the intestinal health and function [29]. However, little is known about the effects on fish performance, and especially about the modes of action on the intestinal mucosa, beyond a purely immunological aspect.

The aim of this study was to evaluate the effect of a feed supplemented with D. hansenii on gilthead seabream (Sparus aurata) on common key performance indicators (KPI) associated to somatic growth and feed efficiency coupled with the analysis of the transcriptomic profile and the histological organization of the anterior-mid intestine, and its autochthonous microbiota in order to provide inside into the effects of this yeast on host condition. The convergence offered in this study by the integration of these analytics in the intestinal mucosa in one of the most highly valued species in the Mediterranean [30], is highly relevant for the introduction and management of sustainable feeding strategies for the growing aquaculture industry.

Material and methods

Diet composition

To evaluate the effects of D. hansenii as a probiotic in aquafeeds, two diets were formulated with similar levels of protein content (48.4% crude protein), lipid content (17.2% crude fat), and energy levels (21.7 MJ/kg gross energy) (Table 1). Both diets, named Control and Yeast diets, were formulated with low levels of fishmeal (7%) where fishmeal was replaced by a blend of plant-based meals (wheat, corn, pea and soy) (Table 1). Diets only differed in the inclusion of D. hansenii (CBS 8339) at an inclusion rate of 1.1% (17.2 × 105 CFU). D. hansenii was provided by CIBNOR (La Paz, Mexico) and grown as described in Tovar-Ramírez et al. [25].

Table 1 Ingredients and proximal composition of the experimental diets

Both diets (2 mm pellet size) were manufactured by SPAROS Lda (Olhão, Portugal) in a temperature-controlled low-shear extruder as detailed in Gisbert et al. [31]. Samples of each diet were taken for proximate composition, and feeds were stored at 4 °C along the experimental feeding trial to prevent their oxidation.

Experimental model and design

A total of 500 S. aurata juveniles (body weight, BW = 14.5 ± 1.2 g; mean ± SD) were obtained from a commercial farm (PISCIMAR S.L.; AVRAMAR, Burriana, Spain). Fish were transported to IRTA-La Ràpita research facilities (La Ràpita, Spain), and acclimated for two weeks in two tanks of 2,000 L connected to a IRTAmar® recirculating system under natural photoperiod and constant temperature (23.0 ± 1.0 °C). During acclimatation, fish were fed with a commercial diet containing 48.5% protein, 18% lipid, and 18.5 MJ/kg digestible energy (Optibream, Skretting).

Before the trial, 200 fish (25 fish per tank; 4 tank replicates per diet) were gently anesthetized (50 mg/L tricaine methanesulfonate, MS-222, Sigma-Aldrich, Madrid, Spain) and individually measured in BW and standard length (SL) to the nearest 0.1 g and 1 mm, respectively. The trial lasted for 70 d, during which fish were fed at 3.5% of the stocked biomass by means of automatic feeders (ARVO-TEC T Drum 2000; Arvotec, Finland). The daily feed ratio was evenly distributed in 4 meals at 08:00, 11:00, 13:00 and 16:00 h (at each meal, the corresponding feed ration was distributed during 1 h). One hour after each meal, uneaten pellets were collected, dried overnight (120 °C), and weighted (g) for calculating daily feed ingesta values, while feed ration was adjusted to guarantee 10%–15% of uneaten pellets; thus, confirming that fish were fed at satiation. This trial was run in a recirculating system IRTAmar® under natural photoperiod conditions (14 h light/8 h dark), constant temperature (23.3 ± 1.3 °C), salinity (35–36 ppt), dissolved oxygen (5.7 ± 0.2 \(\mathrm{mg}/\mathrm L\)) (OXI330, Crison Instruments, Spain), and pH (8.2 ± 0.1) (pHmeter 507, Crison Instruments). Ammonia (0.13 ± 0.1 mg \({\mathrm{NH}}_{4}^{+}\)/L) and nitrite levels (0.18 ± 0.1 mg \({\mathrm{NO}}_{2}^{-}\)/L) (HACH DR 900 Colorimeter, Hach Company, Spain) were weekly controlled.

Growth and feed performance indicators were calculated using the following formulae:

  • \(\mathrm{Specific}\;\mathrm{growth}\;\mathrm{rate}\;\mathrm{in}\;\mathrm{body}\;\mathrm{weight},\;\mathrm{BW}\;\left(\mathrm{SGR},\;\%\;\mathrm{BW}/\mathrm d\right)\:=\:100\;\times\;\left[\left(\ln\;{\mathrm{BW}}_{\mathrm f}\;-\;\ln\;{\mathrm{BW}}_{\mathrm i})\;/\;\mathrm d\right)\right];\;\mathrm{where}\;{\mathrm{BW}}_{\mathrm f}\;\mathrm{and}\;{\mathrm{BW}}_{\mathrm i}\;\mathrm{are}\;\mathrm{the}\;\mathrm{final}\;\mathrm{and}\;\mathrm{initial}\;\mathrm{mean}\;\mathrm{BW}\;\mathrm{of}\;\mathrm{fish}.\)  

  • \(\mathrm{Survival}\;\mathrm{rate}\;(\mathrm{SR},\;\%)\:=\:{100\;\times\;(\mathrm{Final}\;\mathrm{number}\;\mathrm{of}\;\mathrm{fish}\;/\;\mathrm{Initial}\;\mathrm{number}\;\mathrm{of}\;\mathrm{fish}).}\) 

  • \(\mathrm{Feed}\;\mathrm{conversion}\;\mathrm{ratio}\;(\mathrm{FCR})\:=\:\mathrm{Feed}\;\mathrm{ingesta}\;(\mathrm g)\;/\;\mathrm{Increase}\;\mathrm{in}\;\mathrm{fish}\;\mathrm{biomass}\;(\mathrm g).\) 

At the end of the trial, all fish were individually captured, anaesthetised (100 mg/L MS222) and their BW and SL measured as previously described. In addition, fifteen fish per tank were euthanised (300 mg/L MS222) and their digestive tract removed for further transcriptomic and microbiome studies as described below. In this sense, the anterior-mid intestine was chosen for transcriptomic purposes because of its immunological relevance in comparison with other intestinal sections [32, 33].

Histological and histochemical analysis

Samples from the anterior-mid intestine (n = 12 randomly selected fish per experimental diet; 3 fish per tank) were embedded in paraffin and sagittally sectioned (5–6 μm). A total of 576 sections (2 per each sample × 24 samples × 12 histochemical techniques) were used for histological and histochemical purposes. Two sections per each sample were stained with haematoxylin–eosin for descriptive purposes; the rest were used for evaluating the histochemical properties of epithelial and mucous cells. In brief, Schiff, Periodic Acid Schiff (PAS), diastase-PAS and Alcian Blue (AB) pH 2.5, 1 and 0.5 (carboxylated and sulphated glycoconjugates/glycoproteins) techniques were used for studying carbohydrate distribution. Furthermore, several horseradish peroxidase (HRP) conjugated lectins (Sigma-Aldrich, Spain) were used for proper characterization of different glucidic residues bound to the glycoconjugates; in particular, Canavalia ensiformes/ConA (mannose and/or glucose), Ulex europeus/UEA-I (L-fucose), Triticum vulgaris/WGA (N-acetyl-D-glucosamine and/or N-acetyl-neuraminic acid, NeuNAc/sialic acid/NANA), Glycine max/SBA (a-N-acetyl-D-galactosamine) and Sambucus nigra/SNA (NeuNAc/sialic acid/NANA). Lectin concentrations ranged between 15 μg/L to 30 μg/mL. Regarding negative controls, omission of the respective lectin, substitution of lectin-HPR conjugates by TBS and treatments with different enzymes were performed according to Sarasquete et al. [34]. The peroxidase activity was visualized with 3,3-diaminobenzidine tetra hydrochloride/DAB and hydrogen peroxide (0.05%). All the techniques were performed according to Underwood [35] and following proper standardized techniques and protocols [36]. All reagents were purchased from Sigma-Aldrich (St Louis, MO, USA).

Histological images (600 dpi) were taken with a Leitz Wetzlar microscope with a built-in SPOT Insight Color camera (Ernst Leitz Wetzlar GmbH, Germany). Results were manually registered using a semi-quantitative assessment scoring based on color intensity scores (0, negative; 1, weak; 2, moderate; 3, intense; 4, very intense) from four independent observers, comparing the sections of the control with the experimental diet. The mucous cell count was determined in four different sites of each histological section, and the number of cells expressed per length unit of the basal lamina of the mucosal epithelium (1 mm) according to Yamamoto et al. [37].

Intestinal microbiota

DNA extraction and sequencing by Illumina MiSeq technology

The complete intestine of S. aurata (n = 8 per dietary condition) was taken for microbiome analyses. Samples were stored at −20 °C, thawed gradually on ice and the mucus intestinal contents (autochthonous microbiota) were collected for further analyses. A 1-mL aliquot per sample of intestinal contents were collected with 1 mL PBS. Total DNA was extracted from each sample according to protocol described in Tapia-Paniagua et al. [38], with minor modifications. DNA concentration and purity were quantified fluorometrically with Qubit™ dsDNA HS Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA) and by spectrophotometric and electrophoretic methods to study the degree of purity, quality, and DNA integrity. Isolated DNA was stored at –20 ºC until further processing and 30 ng used for subsequent analyses. 16S rRNA of samples was sequenced on Illumina MiSeq platform (Illumina, San Diego, CA, USA) with 2 × 300 bp paired-end sequencing in the Ultrasequencing Service of Malaga (Malaga, Spain). Sequencing was carried out using the primers 341F and 805R directed to the variable regions V3–V4 of the 16S rRNA gene [39]. Raw read sequences of the 16S rRNA gene from S. aurata intestinal microbiota are publicly available in the NCBI SRA depository (SRS16609731—SRS16609735) within BioProject PRJNA914904, with BioSample accession numbers SAMN32318396-SAMN32318408.

Bioinformatic analysis

Primers were removed and quality control were performed with cutadapt and FastQC software respectively. Barcoding sequences were processed using R DADA2 library. Briefly, forward and reverse reads were truncated with decreasing quality metrics while maintaining sequence overlap. Paired reads were assembled after error modelling and correction, creating amplicon sequence variants (ASVs). Chimeric ASVs were removed by reconstruction against more abundant parent ASVs. Taxonomy was assigned to representative sequence variants using SILVA release 138 database, clustered at 99% identity, and trimmed to the amplified region. Abundance of ASV of the intestinal microbiota was processed using phyloseq and vegan library in R statistical package. Shannon's alpha diversity indices, to evaluate taxonomic diversity and PCoA to beta diversity were performed. ASVs with abundance minor to 10 reads in at least of 10% of samples were filtered to taxonomy analyses. R library ggplot was used to represent the abundance of different taxonomy categories and differential abundance of taxa was carried out using the R package DESeq2 with a false discovery rate (FDR < 0.05).

Gut transcriptional analysis

RNA isolation and quality control

Total RNA from the anterior-mid intestine of nine randomly selected fish per dietary treatment (n = 4 fish per tank) was extracted using the RNeasy® Mini Kit (Qiagen, Germany). Total RNA was eluted in a final volume of 35 μL nuclease-free water and treated with DNAse (DNA-free™ DNA Removal Kit; Invitrogen, Lithuania). Total RNA concentration and purity were measured using Nanodrop-2000® spectrophotometer (Thermo Scientific, USA) and stored at −80 °C until analysis. Prior to hybridization with microarrays, RNA samples were diluted to 133.33 ng/μL concentration, checked for RNA integrity (Agilent 2100 Bioanalyzer; Agilent Technologies, Spain) and selected by the criteria of a RIN value > 8.5. Three different pools of samples per dietary treatment were established (n = 4 fish each pool); this choice of pooling individual samples resulted in the loss of information derived from individual sample variability.

Microarray hybridization and analysis

Anterior-mid intestine transcriptional analysis from both experimental groups was carried out using the Aquagenomics Sparus aurata Oligonucleotide Microarray v2.0 (4 × 44 K) (SAQ) platform. Detailed information and transcriptomic raw data are available at the Gene Expression Omnibus (GEO) public repository at the US National Center for Biotechnology Information (NCBI), accession numbers GPL13442, and GSE162504, respectively. Sample labelling, hybridization, washes, and scanning was performed as described in Firmino et al. [40]. Briefly, a one-color RNA labelling was used (Agilent One-Color RNA Spike-In kit; Agilent Technologies, USA). RNA from each sample pool (200 ng) was reverse-transcribed with spike-in. Then, total RNA was used as template for Cyanine-3 (Cy3) labelled cRNA synthesis and amplified with the Quick Amp Labelling kit (Agilent Technologies). cRNA samples were purified using the RNeasy® micro kit (Qiagen). Dye incorporation and cRNA yield were checked (NanoDrop ND-2000® spectrophotometer). Then, Cy3-labeled cRNA (1.5 mg) with specific activity > 6.0 pmol Cy3/mg cRNA was fragmented at 60 °C for 30 min and hybridized with the array in presence of hybridization buffer (Gene expression hybridization kit, Agilent Technologies) at 65 °C for 17 h. For washes, microarrays were incubated with Gene expression wash buffers, and stabilization and drying solution according to manufacturer instructions (Agilent Technologies). Microarray slides were then scanned (Agilent G2505B Microarray Scanner System), and spot intensities and other quality control features extracted (Agilent Feature Extraction software version 10.4.0.0). The extracted raw data were imported and analysed with GeneSpring (version 14.5 GX software, Agilent Technologies). To standardize the arrays intensity, 75% percentile normalization was used, and data were filtered by expression levels. The differential expressed genes (DEGs) were obtained from a gene-level differential expression analysis. Expression values with up-value < 0.05 were considered statistically significant. The DEGs were grouped according to their fold-change value (P-value < 0.05) and represented using the GraphPad softwarev7.0 for Windows. The 3D principal component analysis (PCA) was carried out using GeneSpring software (Agilent), four eigenvectors were calculated to describe the aggrupation of diets control and yeast in a 3D plot. The gene expression values (log-expression ratios) were represented by a hierarchical clustering heatmap analysis using GeneSpring software (Agilent).

The Search Tool for the Retrieval of Interacting Genes (STRING) public repository version 11.0 (https://string-db.org), and the application ClueGO (v2.5.9) and CluePedia (v1.5.9) using Cytoscape (v3.9.1) software, were used to generate the anterior-mid intestine transcripteractome and cluster aggrupation for the DEGs of fish fed the yeast-supplemented diet. A protein–protein interaction (PPI) Networks Functional Enrichment Analysis for all the differentially expressed genes (DEGs) was conducted with a medium-confidence interaction score (0.4) using Homo sapiens as model organism [40]. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of all the DEGs obtained were also assessed through STRING (P < 0.05). In order to confirm match of gene acronyms between both Homo sapiens and S. aurata species, human orthology identification based on gene/protein name was accessed through the Genecards (www.genecards.org) [41] and Uniprot (www.uniprot.org) databases. Additionally, protein–protein BLAST (BLASTp) was run (E < 10−7; query cover > 95%).

Statistical analysis

Data on growth, feed performance indicators and goblet cell density in the intestinal epithelium are expressed as mean ± standard deviation, and a probability value of P < 0.05 was considered as significant by means of a Student’s t-test. Regarding microbiota studies, F diversity indexes were compared between both dietary groups by means of a Student’s t-test (P < 0.05).

Results

Growth and feed performance indicators

At the end of the trial, fish fed low fish meal diet supplemented with D. hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) had significant differences compared with the fish fed the Control diet in terms of somatic growth. Data showed improved BWf, BWG and the SGR values in fish fed the Yeast diet compared with those fed the Control diet (Table 2; P < 0.05). Furthermore, FCR values were significantly lower in fish fed the Yeast diet when compared to the control ones (1.26 ± 0.07 vs. 1.06 ± 0.05, respectively). No significant differences in the FI and SR were found between both groups (Table 2; P > 0.05).

Table 2 Growth and feed performance indicators in gilthead sea bream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet)

Morphological organization of the anterior-mid intestine and histochemical properties of mucins produced by goblet cells

The mucosal layers from the anterior-mid intestine of fish fed both experimental diets were similar. In brief, both were lined by a simple columnar epithelium with basal nuclei, basophilic cytoplasm, and prominent brush border, with scattered goblet cells. No signals of histological alterations associated with inflammatory processes nor lymphocyte infiltrations in the anterior-mid intestine were observed in fish fed the Yeast diet compared to the control group (Fig. 1a and b). No significant differences in goblet cell density were detected when comparing both treatments (Fig. 2).

Fig. 1
figure 1

Histological organization of the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) (a) or devoid of the probiotic (Control diet) (b). Histochemical properties of mucins produced by intestinal goblet cells regarding their content on carboxylated and/or sulphated acidic groups from fish fed the Control diet (c) and the Yeast diet (d). Abbreviations: mc, mucous cells; bb, brush border. Staining: hematoxylin–eosin (a, b), Alcian Blue pH = 2.5 (c, d). Scale bar = 50 µm

Fig. 2
figure 2

Box-plot graph of goblet cell density in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). No statistically significant differences were detected among dietary treatments (Student’s t-test; P > 0.05). Mean line within the boxplot indicate the average value of the series; O and  = outlier points of the series

Regarding the histochemical properties of mucins produced by goblet cells from the anterior-mid intestine, results showed a variable richness of neutral glycoproteins (PAS and diastase-PAS positive) (Table 3). In addition, mucins showed a mixture of carboxylated (AB pH = 2.5) and sulphated acidic groups (weak and strongly ionized; AB pH = 1.0 and 0.5, respectively) (Table 3 and Fig. 1c). Regarding lectin histochemistry, a specific affinity for WGA, SNA, ConA and SBA lectins was detected in the mucinous content of goblet cells (Table 3 and Fig. 3). Moreover, no variations were detected in the distribution of mucosal cell glycoconjugates between the upper and the bottom areas of the intestinal folds. When comparing both dietary groups, the dietary administration of D. hansenii modified the composition of glycoproteins of mucins produced by goblet cells, with an increase in the staining intensity of mucins rich in carboxylated and weakly sulphated glycoconjugates (Table 3 and Fig. 1c and d). In addition, an increase in affinity for SBA lectin and a decrease in the affinity for the WGA lectin was found in the mucinous content of goblet cells, whereas no changes were detected regarding ConA, SNA and UEA-I lectins (Table 3 and Fig. 3).

Table 3 Histochemical results and affinity for different lectins in the anterior-mid intestinal mucosa of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet)
Fig. 3
figure 3

Histochemical localization of glycoconjugates containing sugar residues in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). Presence of glycoconjugates containing α/β-N-acetyl-D-galactosamine in mucous cells of S. aurata fed a Control (a) or a Yeast diet (b). Results denote a moderate increase in affinity for the SBA lectin in the mucous cells and in the intestinal epithelium from Yeast diet. Glycoconjugates containing N-acetyl-D-glucosamine and/or N-acetylneuraminic acid residues in mucous cells of S. aurata fed the Control or the Yeast diet (d). Results denote a moderate decrease in affinity for the WGA lectin in the mucous cells from the Yeast diet. Histochemical detection of glycoconjugates containing N-acetylneuraminic acid/sialic acid residues in the intestine from the Control (e) or Yeast (f) groups. Note the decrease in affinity for the SNA lectin in the anterior-mid intestinal epithelium of S. aurata fed the Yeast diet. Glycoconjugates containing a-mannose/a-glucose residues in anterior-mid intestine from S. aurata fed Control (g) or Yeast diets (h). Observe the increase in affinity for the ConA lectin in the intestinal epithelium of S. aurata fed the Diet Y. Abbreviations: e: epithelium; mc: mucous cells. Scale bar = 50 μm

Intestinal microbiota

Sequencing data analysis

After bioinformatic processing, an average of 117,038.2 reads per sample were obtained (minimum reading 71,378 and maximum 162,602 reading) that were classified into 1,594 ASVs.

The Shannon and Simpson alpha diversity indexes were calculated for each sample of experimental groups (Table 4). The alfa diversity values in the experimental groups did not show significant differences (P > 0.05). The PCoA analysis to check the beta diversity between the detected sequences did not show a clearly separated grouping of samples based on the diet (Additional file 1: Fig. S1).

Table 4 Alpha diversity indices of microbiota intestine population of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet)

Taxonomic composition of the intestinal microbiota

At phylum level, the predominant phyla detected in all fish fed both experimental diets were Proteobacteria, followed by Firmicutes (Fig. 4A), whereas the most abundant classes in all cases were alpha- and gamma-Proteobacteria (Fig. 4B). However, no significant differences were found among diets. A comparative analysis at genus level (abundance > 0.5%) showed a very similar abundance of sequences between specimens from both diets (Fig. 5), whereas Pseudoalteromonas, Acinetobacter, Pseudomonas, Shewanella and Vibrio were the most abundant genera.

Fig. 4
figure 4

Values of relative abundance at the phylum (A) and class (B) level, obtained for the anterior-mid intestine microbiota of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet)

Fig. 5
figure 5

Values of relative abundance at genus level, obtained for the anterior-mid intestine microbiota of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet)

DESeq2 analysis was applied to evaluate the significant differences (FDR < 0.05) between the abundance of the microbial taxa. Significant differences were detected regarding to the genera included in Protobacteria, and with abundance higher than 0.5% such as Acinetobacter, Comamonas and Pseudomonas, which showed decreased abundances in fish from the Yeast diet in comparison to those fed the Control diet (Fig. 6). Other significant differences were also detected within the Proteobacteria phylum, and particular in bacteria from the genus Anaerococcus, Ascidiaceihabitans, Hydrogenophaga and Variovorax that displayed lower abundances of ASVs in fish from the Yeast diet, whereas an increase of ASVs related to the genus Bacillus were found in the intestine of fish fed the Yeast diet (Fig. 6).

Fig. 6
figure 6

Logarithmic fold change of differentially abundant ASVs (DESeq2, FDR < 0.05) at the genus level in the anterior-mid intestine microbiota of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet)

Transcriptomics of the anterior-mid intestine

Transcriptomic profile

To evaluate the modulatory effects of the dietary administration of D. hansenii at 1.1% upon intestine transcriptome, we performed a microarray-based transcriptomic analysis. As shown in Fig. 7 and listed in Additional file 1: Table S1, a total of 232 differentially-expressed genes (DEGs) were found in the anterior-mid intestine when comparing samples from both diets (P < 0.05). Among these, 177 DEGs were up-regulated, and 55 DEGs were down-regulated showing both different levels of fold-change (FC) (Fig. 7A). The percentage expression pattern was similar in both up- and down-regulated genes, although in numerical terms most of the DEGs found were up-regulated with a FC > 2 (12 DEGs up-regulated vs. 1 DEG down-regulated), even some of these being expressed with a FC of 4, 6 or 10 (Fig. 7A and Additional file 1: Table S1). The 3D PCA analysis showed a clear grouping for both the Control and Yeast diets (Fig. 7B), meanwhile the hierarchical clustering heatmap confirmed the conservative response of the arrays, showing a clear differential expression profile of DEGs between both dietary groups (Fig. 7C).

Fig. 7
figure 7

Transcriptomic response of the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). A The green (up-regulated) and red (down-regulated) colour scheme indicate the gene modulation according to its magnitude interval (fold-change). B 3D principal component analysis (PCA). Control (red spheres) and D. hansenii groups (yellow spheres) are represented (each array included in the study is represented by one sphere). C Hierarchical clustering heatmap representing the 232 DEGs. The results of each microarray analysed for the control and yeast groups are shown. The green (up-regulation) and red (down-regulation) colour scheme indicates the gene normalized intensity values according to its magnitude interval

A biological network (transcripteractome) considering the complete DEGs annotated list was prepared in order to have an overview of the relationships between them (Fig. 8). The gene network resulted in 157 nodes/DEGs and 210 edges/interactions (with an expected number of 145 edges), excluding from the analysis the remaining DEGs (n = 75; 32% of the total DEGs), classified as unknown genes.

Fig. 8
figure 8

Biological network of the differentially expressed genes (DEGs) in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). Node fill with a continuous mapping colour from red (down-regulated DEGs) to green (up-regulated DEGs), represents the fold change (FC) intensity. Edge score was represented by doted lines with a continuous mapping from 0.35 (light Blue) to 1.00 (dark blue). The transcripteractome was obtained using Cytoscape (v3.9.1) platform with String (v1.7.1). For details, please refer to Additional file 1: Table S1

Functional enrichment analysis

The functional enrichment analysis of the transcripteractome helped us to identify several biological processes and pathways for the DEGs in the anterior-mid intestine of S. aurata fed the Yeast diet in comparison to their congeners from the control group (Control diet). Thus, 54 GO were identified (P < 0.05; Additional file 1: Table S2), whereas the top-10 representative processes per number of DEGs were as listed: “Nitrogen compound metabolic process” (60.5% of the total DEGs: 83 up-regulated vs. 12 down-regulated; GO:0006807), “Organic substance biosynthetic process” (30.6% of the total DEGs: 42 up-regulated vs. 6 down-regulated; GO:1901576), “Cellular response to chemical stimulus” (28.2% of the total DEGs: 35 up regulated vs. 9 down-regulated; GO:0070887), “Nucleic acid metabolic process” (24.8% of the total DEGs: 37 up-regulated vs. 2 down-regulated; GO:0090304), “Negative regulation of gene expression” (20.4% of the total DEGs: 27 up-regulated vs. 5 down-regulated; GO:0010629), “Cellular catabolic process” (18.5% of the total DEGs: 28 up-regulated vs. 1 down-regulated; GO:0044248), “Heterocycle biosynthetic process” (14.0% of the total DEGs: 21 up-regulated vs. 1 down-regulated; GO:0,018130), “Cellular response to oxygen-containing compound” (13.4% of the total DEGs: 16 up-regulated vs. 5 down-regulated; GO:1901701), “DNA metabolic process” (12.1% of the total DEGs: 17 up-regulated vs. 2 down-regulated; GO:0006259) and, “Symbiotic process” (11.5% of the total DEGs: 15 up-regulated vs. 3 down-regulated; GO:0044403) (Additional file 1: Table S3).

According to the results reported by the enrichment analysis, representative clusters of genes were classified as shown in Figs. 9, 10, 11, 12 and 13 and Additional file 1: Table S4. A significant number of genes were observed to be shared among the metabolic category: Protein Metabolism (31.2% of known genes; Fig. 9), Nucleotide Metabolism (8.3% of known genes; Fig. 10), and Lipid Metabolism (5.7% of known genes; Fig. 11). The other clustered categories contained genes related to the Regulation of Antioxidant Capacity (6.4% of known genes; Fig. 12), and to the Regulation of Immune Response (10.8% of known genes; Fig. 13). This clusters suggest a strong relationship between processes that favours metabolic and immune responses to the dietary inclusion of D. hansenii.

Fig. 9
figure 9

Protein metabolism cluster of the functional enrichment network for biological processes based on the related functions of the differentially expressed genes (DEGs) in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). The node size determinate the significance (Bonferroni step-down correction; small P < 0.05; medium P < 0.005; large P < 0.0005), the node colour determinate the principal GO process aggrupation, and the node numbers indicate different GO process classified in Additional file 1: Table S4

Fig. 10
figure 10

Nucleotide metabolism cluster of the functional enrichment network for biological processes based on the related functions of the differentially expressed genes (DEGs) in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). The node size determinate the significance (Bonferroni step-down correction; small P < 0.05; medium P < 0.005; large P < 0.0005), the node colour determinate the principal GO process aggrupation, and the node numbers indicate different GO process classified in Additional file 1: Table S4

Fig. 11
figure 11

Lipid metabolism cluster of the functional enrichment network for biological processes based on the related functions of the differentially expressed genes (DEGs) in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). The node size determinate the significance (Bonferroni step-down correction; small P < 0.05; medium P < 0.005; large P < 0.0005), the node colour determinate the principal GO process aggrupation, and the node numbers indicate different GO process classified in Additional file 1: Table S4

Fig. 12
figure 12

Regulation of Antioxidant Capacity cluster of the functional enrichment network for biological processes based on the related functions of the differentially expressed genes (DEGs) in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). The node size determinate the significance (Bonferroni step-down correction; small P < 0.05; medium P < 0.005; large P < 0.0005), the node colour determinate the principal GO process aggrupation, and the node numbers indicate different GO process classified in Additional file 1: Table S4

Fig. 13
figure 13

Regulation of Immune Response cluster of the functional enrichment network for biological processes based on the related functions of the differentially expressed genes (DEGs) in the anterior-mid intestine of gilthead seabream (Sparus aurata) fed experimental low fishmeal diets containing Debaryomyces hansenii (1.1%, 17.2 × 105 CFU; Yeast diet) or devoid of the probiotic (Control diet). The node size determinate the significance (Bonferroni step-down correction; small P < 0.05; medium P < 0.005; large P < 0.0005), the node colour determinate the principal GO process aggrupation, and the node numbers indicate different GO process classified in Additional file 1: Table S4

Three significative cellular components GO groups (80.2% of the total DEGs in GO:0043227; 73.9% in GO:0043231; 10.2% in GO:0005740) were associated with the single or double lipid bilayer membrane, which indicated that most of DEGs belong to the above-mentioned structures. On the other hand, it is also worth noting that some of these DEGs participate in mitochondrial processes (GO:0005739), proteasome complex (GO:0000502) and DNA replication (replication fork: GO:0005657; replisome: GO:0030894). For further details see Additional file 1: Table S5.

Discussion

Low fishmeal along with high plant-content meal diets are of common practices for the aquaculture feed industry. However, reducing the dependence on marine raw materials is still associated with variable side effects on fish nutrient metabolism, mucosal health, and disease resistance [42], especially due to the presence of antinutritional factors and the lack of essential amino acids and fatty acids in those alternative sources. Thus, it is necessary to develop complementary nutritional tools to reduce the negative-side effects associated with the substitution of traditional ingredients. In this sense, the use of functional feeds seems to be an appropriate strategy [43]. When considering new additives, yeasts are among those promising candidates showing a wide range of benefits on animal health and growth [21, 44]. In this sense, the present study aimed to determine the effects on growth and intestinal condition of a functional feed with a low fishmeal formula (7%) that was supplemented with D. hansenii in S. aurata, the main marine fish species farmed in the Mediterranean basin [30].

Effects of D. hansenii on fish performance

Under present experimental conditions, we found a significant body weight increase in fish fed the Yeast diet coupled to a lower feed conversion rate and similar feed intake values compared with those fish fed the Control diet. Considering that feeding costs represent up to 50%–70% of total production costs in intensive fish farms [45], and fishmeal being amongst the most expensive raw materials in aquafeeds [46]; the nutritional strategy tested in our study that consisted of combining a diet containing low fishmeal levels (7%) with the supplementation of a live probiotic seemed very promising when KPIs related to growth and feed utilization were considered. Furthermore, these results are of special relevance since this is the first study in fish juveniles showing the benefits of supplementing low fish meal diets with D. hansenii in terms of somatic growth and feed efficiency parameters. In this context, several previous studies had reported that the dietary inclusion of D. hansenii (intact or hydrolysed) in aquafeeds with high levels of fishmeal (39%–41%) did not result in significant differences in terms of fish growth performance in Atlantic salmon (Salmo salar) and tropical gar (Atractosteus tropicus) [27, 47]. Present findings when using low fishmeal diets may be due to the presence in D. hansenii of growth factors like biogenic amines, capable of promoting growth and intestinal functionality [21, 29, 48]. Biogenic amines or polyamines are essential metabolites found in almost all living organisms [49]. In particular, D. hansenii is especially rich in putrescine, spermidine, spermine and cadaverine [17, 50]. These polyamines are essential for many cell functions and play a key role in vertebrate development and cell growth [17, 51]. In particular, Tovar-Ramírez et al. [25] observed in European sea bass (Dicentrarchus labrax) an increase of larval survival and an enhancement of the digestive function, whereas Teles et al. [23] found that longfin yellowtail (Seriola rivoliana) larvae had higher growth and survival rates, and an earlier digestive tract maturation when fed with a diet supplemented with D. hansenii. These results found in fish larvae were similar to those found in the current study regardless of the developmental stage considered. Furthermore, although it is described that polyamines in high concentrations can generate adverse effects on animal health, their probiotic administration through D. hansenii in feed might be of great benefit to fish as reported in several studies [21, 25, 29, 48]. In addition, by participating in the protein metabolism, these polyamines could be of special interest in carnivorous species with high protein demand [51], since their dietary provision by D. hansenii may modulate the amino acid metabolism in fish, which could result in the promotion of fish growth by a better use of dietary proteins [52], as shown in our study.

Effect of D. hansenii diet on intestinal condition

The intestine is a complex organ composed of the digestive epithelium with its typical tissue organization in different mucosal layers, its associated lymphoid tissue, and the mucus layer covering the simple intestinal epithelium with its commensal microbiota. Furthermore, enterocytes form a selectively permeable barrier for water, electrolytes, and nutrients, while maintaining an effective defence against pathogens and tolerance towards dietary antigens [53]. Thus, studying the effect of the diet on tissue condition and its commensal microbiota is of importance when evaluating new nutritional strategies. In this context, the intestinal microbiota has been extensively studied in a multitude of fish species since microbial gut communities are essential for proper gastrointestinal development and are implicit in aiding digestive and immunological functions and disease resistance [54, 55]. However, how host-microbiota symbiosis and/or how feed and feed additives can modulate the normal microbiota in the host remains poorly understood, since gut microbial communities are strongly affected by dietary or environmental factors [56, 57]. Despite that, results in terms of phyla obtained in our study were in agreement within those from other authors [40, 58, 59]. In those studies, Proteobacteria has been reported as the most abundant phylum in the intestine of S. aurata, followed by Firmicutes, Actinobacteria, and Bacteroidetes. This conservative pattern at phyla level of microbial communities in the intestine was maintained in our study regardless of the diet evaluated.

At a lower taxonomic level, Pseudomonas spp. and Acinetobacter spp. were the dominant genera in the gut microbiome from both dietary groups. Pseudomonas spp. and Acinetobacter spp. are a part of the typical fish microbiota [60], whereas some strains have been reported as antagonistic against pathogenic microorganisms [61, 62]. However, under stressful conditions such as malnutrition and overcrowding their abundance and the presence of some opportunistic and pathogenic species may increase [63,64,65]. Under current experimental condition, the reduction in the relative abundance of these genera in fish fed the Yeast diet suggested that the dietary administration of D. hansenii may be a strategy for dealing with infections caused by opportunistic and antimicrobial-resistant bacteria, such as Pseudomonas spp. and Acinetobacter spp. [66,67,68,69,70]. Furthermore, D. hansenii supplementation also resulted in an increase of the relative abundances of ASVs related to the genus Bacillus in comparison to fish fed the Control diet. This Gram-positive endospore-forming genus has been widely tested as a probiotic on several aquatic cultured organisms, enhancing growth, modulating immune response, and promoting disease resistance [71, 72]. Similar to other species, D. hansenii are normally found as cohabitants in the gut of wild and farmed fish [21, 73], and seems to interact with the host, maintaining intestine microbiota homeostasis, and aiding to its recover in case of dysbiosis [74, 75]. Although in the current study, the low fishmeal diet did not result in any dietarily-induced inflammatory disorder of the intestinal mucosa or dysbiosis of the gut microbiome, these interactions in terms of DEGs related to membrane-linked organelles and symbiotic biological processes were identified by the transcriptomic functional analysis conducted, and further discussed in "Effect of D. hansenii diet on gut metabolism, homeostasis, and immunity".

The mucus layer that covers enterocytes is considered as the first line of innate host defence as well as the site where commensal microbiota attaches, preventing the colonisation of pathogenic organisms by occupying empty niches. The dynamic relationship between commensal bacteria and mucins secreted by goblet cells plays an important role in gut health and condition [76]. Commensal microbiota is adapted to the glycan rich environment of mucus, stablishing host-microbiota interactions, influencing on the quantitatively and qualitatively mucin glycosylation [77]. In the current study, the dietary administration of D. hansenii did not modify the histochemical properties of mucins produced by goblet cells in terms of neutral glycoproteins, but resulted in a slightly higher richness of carboxylated and weakly ionised sulphated glycoconjugates in fish fed the Yeast diet. These results are of special relevance since the pattern of O-glycosylation in mucin glycoproteins and their polypeptide backbone structure linked to some physical properties of the mucus layer as its viscosity and depth, are key elements in gut health and condition modulation [78]. Thus, the increase in carboxylated and weakly ionized acid sulphated grlycoproteins produced by goblet cells in fish fed the Yeast diet may be interpreted as beneficial due to an increase in mucus viscosity that would protect the host form opportunistic pathogenic bacterial [79, 80]. Furthermore, the inclusion of D. hansenii in the diet changed the glycosylation patterns of intestinal mucins, being of special relevance the increase in SBA lectin affinity rather than WGA, changes that may be partially associated to different rates of mucin glycosylation and goblet cell differentiation along the intestinal mucosa [34]. However, these results are of special relevance since lectins with affinity for SBA, which bind to GalNAc residues may contain sialic acid residues in the backbone region of the mucin [81], have been postulated to have an antiparasitic effect in turbot (Scophthalmus maximus) [82] and pathogenic organisms [78, 83]. Furthermore, the above-mentioned changes in lectin affinity may be supported by differential expression of b4galt3 and b3galt2 genes, which both participates in the elongation and branching biosynthetic steps of glycosylation pathways [84], and the up-expression of a Golgi mannosyl-oligosaccharide 1,2-α-mannosidases gene (man1a1), whose activity is a prerequisite for the formation of complex or hybrid glycans of mucins [85].

Overall, the results obtained showed the ability of D. hansenii to modulate the gut microbiota without affecting the intestine cell organization in S. aurata, mainly reducing several groups of Proteobacteria abundances, especially those opportunistic groups, and changing glycosylation patterns of intestinal mucins. It is suggested that these results, which can be attributed to the potential role of D. hansenii as a probiotic, involve some of the yeast cell wall components, such as β-glucan, polyamines and mannan molecules that have, apart from these attributes, the ability to modulate the response of the immune system [21, 86, 87]. However, the functional benefits of these findings need further attention, including the role of D. hansenii and the microbiota or their interactions, in terms of glycosylation patterns variation.

Effect of D. hansenii diet on gut metabolism, homeostasis, and immunity

In parallel with the improved performance and the modulation of the intestinal condition, several authors have suggested that the dietary administration of D. hansenii result in a concomitant modification of systemic physiological responses, and especially those related to the digestive function and immunity [17, 88]. In this sense, the dietary inclusion of D. hansenii modulated the expression of different genes in the anterior-mid intestine of S. aurata that were associated with biological processes related to gut metabolism, antioxidant defence, and immunity.

Compared with terrestrial animals, fish have higher protein requirements due to their biological lifestyle characteristics [89]. For this reason, it is not surprising that most of the DEGs with metabolic functions were mainly related to protein biosynthesis, turnover and degradation to cope with their increased growth rate. In our study, we found that the ubiquitin–proteasome system (UPS) was modulated by the regulation of some ubiquitin enzyme and ligases genes (uchl3, trim 25, trim39, ubr7 and ube2i) and proteasome subunits (psma4, psma6, psmb10, psmc2 and rad23a), which act as protein binding signalling and protein degradation, respectively. Both activities are suggested to be important mechanisms of aging, immune activity, cell quality control and protection, tissue regeneration, or DNA repair and gene transduction [90, 91]. On the other hand, we found several DEGs related to protein synthesis and modification, especially genes involved in ribosomal subunit components (rpl22, rpl38, mrps2, mrps9, and mrpl54), and amide-related biosynthetic processes. The regulation of several genes involved in amide biosynthesis may be attributed to the amine and polyamine content of D. hansenii included in the diet [17], which would support the above-mentioned benefits in terms of growth performance as described in several species [21].

Linked to the increased gut metabolic activity, we found other DEGs not directly related to protein metabolism that were associated with nucleotide metabolism and DNA elongation and repair. According to current results, dietary D. hansenii enhanced thymidylate nucleotide metabolism as indicated by the up-regulation of dtymk, tyms, dut, and dck genes. Thymidylate is necessary to synthetise deoxythymidine triphosphate (dTTP), an essential building block of DNA [92]. In particular, studies in larvae showed that TYMK, the protein codded by dtymk gene, and its deoxyribonucleotide triphosphate (dNTP) product, are vital in early embryogenesis and essential for neurodevelopment, and genome integrity in adult fish [93, 94]. These results would be in agreement with the reported beneficial effect of D. hansenii larval development and morphogenesis [23, 25].

The third main metabolic cluster identified in our study was related to lipid metabolism, mainly involving DEGs related to sphingolipid or ceramide metabolic processes (aldh3a2, elovl6, asah1, degs1, b3galt2 and b4galt3). Sphingolipids and ceramides are constituents of eucaryotic cell membranes with a wide variety of functions and properties. It is reported that sphingolipids have antimicrobial and immunomodulatory properties [95], along with bioactive signalling functions [96]. The DEG asah1 encodes for an acid ceramidase N-acylsphingosine amidohydrolase 1 (ASAH1), which catalyses the degradation of ceramide into sphingosine and free fatty acid and suggested to maintain ceramide homeostasis in the lysosome [97]. Furthermore, participates in sphingosine recycling pathway along with degs1 gene, which controls the step from dihydroceramides to ceramides, and suggested to be a promising biomarker of redox state [98].

An increase in the metabolism could lead to increased production of oxidative radicals, that in an unbalanced situation with the antioxidant defence capacity would produce oxidative damage [99]. In parallel to the above-mentioned enhancement of gut metabolic activity, we also found an increased expression of some genes from the electron chain and others also involved in mitochondrial respiration. Their activity produces chemical energy for the normal cell behaviour, but as a result the superoxide anion (\(\mathrm O_2^-\)), the precursor of most other reactive oxygen species (ROS), are also obtained [100]. To possibly counteract with this situation, we found the up-regulation of mgst3 and gsto1 genes in fish fed the Yeast diet, which both together are related with the glutathione cycle, in charge of detoxifying hydrogen peroxide (H2O2) [101], and selenot, which mediates oxidoreductase functions involved in redox homeostasis [102]. Magalhães et al. [103] reported in S. aurata different antioxidant levels depending on the organ analysed, being the enzymatic activity of catalase (CAT), superoxide dismutase (SOD) and glutathione reductase (GR) higher in the intestine, and glutathione peroxidase (GPx) in liver. The authors discussed that this pattern is expected due to the elevated cell turnover rate in the intestine that favours oxidative processes in a non-stressful situation, which is also proposed in our study by the supposedly increased gut cell metabolism linked to the increased somatic growth observed. As H2O2 plays important cellular functions, e.g., as a promoter for cell cycle progression, the activity of the glutathione cycle enzymes and cytochrome P450 (cyp1a1), also takes part in metabolic pathways like arachidonic, leukotriene, prostaglandin metabolism, or in estrogen metabolism [51, 104], as well as protecting lipids and proteins to be oxidised [105]. In that way, the up-regulation of malate/oxoglutarate carrier (slc25a11) that is related with the TCA cycle and also plays a key role maintaining GSH levels in the mitochondria [106], would reinforce this hypothesis. Thus, the up-regulation of these genes could be more related to metabolism than to a possible oxidative damage produced by the inclusion of yeast in the diet. This idea is also supported by some down-regulated genes related to oxidative stress like: serpinb1, which is suggested to protect the cell from proteases released into the cytoplasm during stress [107]; sin3a, whose expression is promoted under hypoxia stress [108]; and the anti-inflammatory cytokine IL-10, that is modulated under redox/oxidant perturbations [109]. Regarding antioxidant functions, dietary live yeast administration in D. labrax larvae, induced lower activity and expression levels of glutathione peroxidase and superoxide dismutase compared to fish fed control diet, assuming a possible involvement of superoxide anion retention in fish larvae, which could represent importance to the host to increase cell or tissue responsiveness to growth- and/or differentiation-enhancing factors [110].

As reviewed in Angulo et al. [21], D. hansenii dietary supplementation has been shown to promote gut protection in several aquatic and terrestrial species at immunological and gene expression levels. An exhaustive analysis of the DEGs promoted by the Yeast diet in our study showed different regulatory responses of the S. aurata gut immune system. Among them, we found regulatory viral genes up-regulated like the lsm14a, a sensor for both viral DNA and RNA which is important to initiate the IFN-β induction [111]; the trim25 gene, that encodes the TRIM25 E3 ubiquitin ligase protein, which mediates an essential step to initiate the antiviral responses in the cell through the pattern recognition receptor RIG-I [112]; and the phb2, a gene that encodes a prohibitin 2 protein related to the mitochondrial-mediated antiviral innate immunity by activating the RIG-I signalling pathway [113]. Yeast dietary inclusion also promoted the up-regulation of regulatory genes required for T and B cell development and proinflammatory mediators (nsd2, rabl3, inhbb, and parp1), a lymphocyte activator (slamf8), and genes involved in proteasomal generation of antigenic peptides (psma4 and psma6). All these genes might participate in the first steps of the immune response, promoting the regulation of sentinel immune responses, maintaining the immune cells quiescent to avoid autoimmunity, but prepared for a rapid activation [114]. Contrary to those up-regulated genes, we found down-regulated genes directly related with the activation of immune system processes: serpinb1, which is suggested to be activated upon an infection [115]; the tyrobp gene, which participates in the activation of immune cells involved in inflammatory reactions and as a component of NK cells with anti-viral functions [116]; and the cfh gene, which regulates the alternative pathway of the complement system [117]. We also found two cytokines down-regulated, a pro-inflammatory il15 gene, which promotes Th1 responses and T-cell maturation [118], and il10, an anti-inflammatory gene that is expressed under infection with stimulatory and inhibitory activities on several immune cells [119]. Altogether, it may be postulated that yeast supplementation in a low fish meal feed could promote the regulation of sentinel immune processes by increasing the potentiation of the innate immunity rather than activating a response of the immune markers. A good example of this hypothesis was shown by the down-regulation of psmb10, which is an IFN-inducible immunoproteasome, mainly activated during an immune response [120]. In that sense, it has been postulated that D. hansenii might modulate host’s immunity through its wall-related β-glucan and polyamine content, increasing functional and decreasing deleterious immune responses in fish [121]. How these components act or in which pathways are involved is still poorly studied. However, as β-glucans are considered pathogen-associated molecular patterns, there seems to be a consensus in its effects potentiating cellular responses and gene expression signalling pathways, activating the communication and activity of the adaptive immune system in several marine species [122,123,124], which could explain the vast response of the intestine at transcriptomic level.

Conclusions

The present multi-integrative study of the intestinal response to the probiotic D. hansenii when administered to S. aurata juveniles in diets with low fishmeal levels provided new insights into the use of this yeast and its metabolic and immunomodulatory components on farmed fish. Under current experimental conditions, we demonstrated that dietary administration of D. hansenii (1.1%, 17.2 × 105 CFU) enhanced somatic growth and improved feed efficiency parameters, results that were coupled to an improvement of intestinal condition as histochemical, microbiota and transcriptomic tools indicated. In particular, D. hansenii stimulated host-microbiota interactions without altering the intestinal cell organization nor generating dysbiosis, which demonstrated the safety use of the yeast as a probiotic. At the transcriptomic level, D. hansenii promoted metabolic pathways, mainly protein-related, sphingolipid, and thymidylate pathways, in addition to enhance antioxidant-related intestinal mechanisms, and to regulate sentinel immune processes, potentiating the defensive capacity meanwhile maintaining the homeostatic status of the intestine. These findings confirm the probiotic benefits of D. hansenii in the diet, and its use on cultured marine species beyond their larval stages.

Availability of data and materials

Detailed information and transcriptomic raw data are available at the Gene Expression Omnibus (GEO) public repository at the US National Center for Biotechnology Information (NCBI), accession numbers GPL13442, and GSE162504, respectively. Data on intestinal microbiota are publicly available in the NCBI SRA depository (SRS16609731—SRS16609735) within BioProject PRJNA914904, with BioSample accession numbers SAMN32318396 and SAMN32318408.

Abbreviations

AB:

Alcian blue

ASAH1:

N-acylsphingosine amidohydrolase 1

ASVs:

Amplicon sequence variants

BW:

Body weight

CAT:

Catalase

CFU:

Colony-forming unit

D.  hansenii :

Debaryomyces hansenii

DEGs:

Differential expressed genes

dNTP:

Deoxyribonucleotide triphosphate

dTTP:

Deoxythymidine triphosphate

FC:

Fold-change

FCR:

Feed conversion ratio

FDR:

False discovery rate

FI:

Feed intake

GEO:

Gene Expression Omnibus

GR:

Glutathione reductase

GO:

Gene Ontology

GPx:

Glutathione peroxidase

GSH:

Reduced glutathione

HRP:

Horseradish peroxidase

IFN-β:

Interferon beta

IL-10:

Interleukin 10

KEGG:

Kyoto Encyclopedia of Genes and Genomes

KPI:

Key performance indicators

NCBI:

National Center for Biotechnology Information

NK:

Natural killer

PAS:

Periodic Acid Schiff

PCA:

Principal component analysis

PCoA:

Principal coordinate analysis

PPI:

Protein–protein interaction

RIG-I:

Retinoic acid-inducible gene I

S.  aurata :

Sparus aurata

SBA:

Soybean agglutinin

SD:

Standard deviation

SGR:

Specific growth rate

SOD:

Superoxide dismutase

TCA:

Tricarboxylic acid cycle

TRIM25:

Tripartite motif containing 25

UPS:

Ubiquitin–proteasome system

WGA:

Wheat Germ Agglutinin (Triticum vulgaris lectin)

References

  1. Kurtzman CP, Piškur J. Comparative genomics: using fungi as models. Top Curr Genet. 2006;15:29–46. https://doi.org/10.1007/b95174.

    Article  CAS  Google Scholar 

  2. Walker GM. Yeasts. In: Schaechter M, editor. Desk encyclopedia of microbiology. 2nd ed. Academic Press; 2009. p. 1174–87.

  3. Dadkhodazade E, Khanniri E, Khorshidian N, Hosseini SM, Mortazavian AM, Kia EM. Yeast cells for encapsulation of bioactive compounds in food products: a review. Biotechnol Prog. 2021;37(4). https://doi.org/10.1002/btpr.3138

  4. Sabu C, Mufeedha P, Pramod K. Yeast-inspired drug delivery: biotechnology meets bioengineering and synthetic biology. Expert Opin Drug Deliv. 2018;16(1):27–41. https://doi.org/10.1080/17425247.2019.1551874.

    Article  CAS  PubMed  Google Scholar 

  5. Fernández-Pacheco P, Ramos Monge IM, Fernández-González M, Poveda Colado JM, Arévalo-Villena M. Safety evaluation of yeasts with probiotic potential. Front Nutr. 2021;8. https://doi.org/10.3389/fnut.2021.659328

  6. Rohani MF, Islam SM, Hossain MK, Ferdous Z, Siddik MA, Nuruzzaman M, et al. Probiotics, prebiotics and synbiotics improved the functionality of aquafeed: upgrading growth, reproduction, immunity and disease resistance in fish. Fish Shellfish Immunol. 2022;120:569–89. https://doi.org/10.1016/J.FSI.2021.12.037.

    Article  CAS  PubMed  Google Scholar 

  7. Valderrama B, Ruiz JJ, Gutiérrez MS, Alveal K, Caruffo M, Oliva M, et al. Cultivable yeast microbiota from the marine fish species Genypterus chilensis and Seriolella violacea. J Fungi (Basel). 2021;7(7). https://doi.org/10.3390/jof7070515

  8. Banu MR, Akter S, Islam MR, Mondol MN, Hossain MA. Probiotic yeast enhanced growth performance and disease resistance in freshwater catfish gulsa tengra, Mystus cavasius. Aquac Rep. 2020;16. https://doi.org/10.1016/J.AQREP.2019.100237

  9. Siddik MAB, Foysal J, Fotedar R, Francis DS, Gupta SK. Probiotic yeast Saccharomyces cerevisiae coupled with Lactobacillus casei modulates physiological performance and promotes gut microbiota in juvenile barramundi, Lates calcarifer. Aquaculture. 2021;546:737346. https://doi.org/10.1016/j.aquaculture.2021.737346.

  10. Islam SMM, Rohani MF, Shahjahan M. Probiotic yeast enhances growth performance of Nile tilapia (Oreochromis niloticus) through morphological modifications of intestine. Aquac Rep. 2021;21. https://doi.org/10.1016/J.AQREP.2021.100800

  11. Abdel-Tawwab M, Mounes HAM, Shady SHH, Ahmed KM. Effects of yucca, Yucca schidigera, extract and/or yeast, Saccharomyces cerevisiae, as water additives on growth, biochemical, and antioxidants/oxidant biomarkers of Nile tilapia. Oreochromis niloticus Aquaculture. 2021;533:736122. https://doi.org/10.1016/j.aquaculture.2020.736122.

    Article  CAS  Google Scholar 

  12. Yang X, Chi S, Tan B, Nie Q, Hu J, Dong X, et al. Yeast hydrolysate helping the complex plant proteins to improve the growth performance and feed utilization of Litopenaeus vannamei. Aquac Rep. 2020;17:100375. https://doi.org/10.1016/j.aqrep.2020.100375.

  13. Hao Q, Xia R, Zhang Q, Xie Y, Ran C, Yang Y, et al. Partially replacing dietary fish meal by Saccharomyces cerevisiae culture improve growth performance, immunity, disease resistance, composition and function of intestinal microbiota in channel catfish (Ictalurus punctatus). Fish Shellfish Immunol. 2022;125:220–9. https://doi.org/10.1016/j.fsi.2022.05.014.

    Article  CAS  PubMed  Google Scholar 

  14. Mo WY, Choi WM, Man KY, Wong MH. Food waste-based pellets for feeding grass carp (Ctenopharyngodon idellus): Adding baker’s yeast and enzymes to enhance growth and immunity. Sci Tot Env. 2020;707:134954. https://doi.org/10.1016/J.SCITOTENV.2019.134954.

    Article  CAS  Google Scholar 

  15. Estévez A, Padrell L, Iñarra B, Orive M, Martin DS. Brewery by-products (yeast and spent grain) as protein sources in gilthead seabream (Sparus aurata) feeds. Aquaculture. 2021;543:736921. https://doi.org/10.1016/j.aquaculture.2021.736921.

  16. Konstantinidis I, Firmino JP, Ruiz A, Iñarra B, San Martin D, Estévez A, et al. Brewer’s spent dry yeast modulates immunity in gilthead sea bream (Sparus aurata). Front Mar Sci. 2022;9:383. https://doi.org/10.3389/fmars.2022.857592.

  17. Reyes-Becerril M, Ascencio-Valle F, Tovar-Ramírez D, Meseguer J, Esteban MÁ. Effects of polyamines on cellular innate immune response and the expression of immune-relevant genes in gilthead seabream leucocytes. Fish Shellfish Immunol. 2011;30(1):248–54. https://doi.org/10.1016/j.fsi.2010.10.011.

    Article  CAS  PubMed  Google Scholar 

  18. Feddern V, Mazzuco H, Fonseca FN, de Lima GJMM, Feddern V, Mazzuco H, et al. A review on biogenic amines in food and feed: toxicological aspects, impact on health and control measures. Anim Prod Sci. 2019;59(4):608–18. https://doi.org/10.1071/AN18076.

    Article  Google Scholar 

  19. Gomez-Gomez HA, Minatel IO, Borges CV, Marques MOM, da Silva ET, Monteiro GC, et al. Phenolic compounds and polyamines in grape-derived beverages. J Agric Sci. 2018;10(12):65. https://doi.org/10.5539/jas.v10n12p65.

    Article  Google Scholar 

  20. Tsafack PB, Tsopmo A. Effects of bioactive molecules on the concentration of biogenic amines in foods and biological systems. Heliyon. 2022;8(9):e10456. https://doi.org/10.1016/j.heliyon.2022.e10456.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Angulo M, Reyes-Becerril M, Medina-Córdova N, Tovar-Ramírez D, Angulo C. Probiotic and nutritional effects of Debaryomyces hansenii on animals. Appl Microbiol Biotechnol. 2020;104(18):7689–99. https://doi.org/10.1007/S00253-020-10780-Z.

    Article  CAS  PubMed  Google Scholar 

  22. Raggi P, Lopez P, Diaz A, Carrasco D, Silva A, Velez A, et al. Debaryomyces hansenii and Rhodotorula mucilaginosa comprised the yeast core gut microbiota of wild and reared carnivorous salmonids, croaker and yellowtail. Environ Microbiol. 2014;16(9):2791–803. https://doi.org/10.1111/1462-2920.12397.

    Article  CAS  PubMed  Google Scholar 

  23. Teles A, Alvarez-González CA, Llera-Herrera R, Gisbert E, Salas-Leiva J, del Carmen Rodríguez-Jaramillo M, et al. Debaryomyces hansenii CBS 8339 promotes larval development in Seriola rivoliana. Aquaculture. 2022;560. https://doi.org/10.1016/j.aquaculture.2022.738587

  24. Vargas O, Gutiérrez MS, Caruffo M, Valderrama B, Medina DA, García K, et al. Probiotic yeasts and Vibrio anguillarum infection modify the microbiome of zebrafish larvae. Front Microbiol. 2021;12. https://doi.org/10.3389/fmicb.2021.647977

  25. Tovar-Ramírez D, Zambonino Infante J, Cahu C, Gatesoupe FJ, Vázquez-Juárez R. Influence of dietary live yeast on European sea bass (Dicentrarchus labrax) larval development. Aquaculture. 2004;234(1–4):415–27. https://doi.org/10.1016/j.aquaculture.2004.01.028.

    Article  Google Scholar 

  26. Sukumaran V, Lowman DW, Sajeevan TP, Philip R. Marine yeast glucans confer better protection than that of baker’s yeast in Penaeus monodon against white spot syndrome virus infection. Aquac Res. 2010;41(12):1799–805. https://doi.org/10.1111/j.1365-2109.2010.02520.x.

    Article  Google Scholar 

  27. Morales-Lange B, Djordjevic B, Gaudhaman A, Press CML, Olson J, Mydland LT, et al. Dietary inclusion of hydrolyzed Debaryomyces hansenii yeasts modulates physiological responses in plasma and immune organs of Atlantic salmon (Salmo salar) parr exposed to acute hypoxia stress. Front Physiol. 2022;13. https://doi.org/10.3389/fphys.2022.836810

  28. Angulo M, Reyes-Becerril M, Tovar-Ramírez D, Ascencio F, Angulo C. Debaryomyces hansenii CBS 8339 β-glucan enhances immune responses and down-stream gene signaling pathways in goat peripheral blood leukocytes. Dev Comp Immunol. 2018;88:173–82. https://doi.org/10.1016/j.dci.2018.07.017.

    Article  CAS  PubMed  Google Scholar 

  29. Reyes-Becerril M, Ascencio-Valle F, Meseguer J, Tapia-Paniagua ST, Moriñigo MA, Esteban MÁ. Debaryomyces hansenii L2-enriched diet enhances the immunity status, gene expression and intestine functionality in gilthead seabream (Sparus aurata L.). Aquac Res. 2012;43(8):1107–18. https://doi.org/10.1111/J.1365-2109.2011.02914.X.

    Article  CAS  Google Scholar 

  30. FAO. The State of World Fisheries and Aquaculture. Towards Blue Transformation. Rome. 2022;266. https://doi.org/10.4060/cc0461en

  31. Gisbert E, Skalli A, Campbell J, Solovyev MM, Rodríguez C, Dias J, et al. Spray-dried plasma promotes growth, modulates the activity of antioxidant defenses, and enhances the immune status of gilthead sea bream (Sparus aurata) fingerlings. J Anim Sci. 2015;93(1):278–86. https://doi.org/10.2527/JAS.2014-7491.

    Article  CAS  PubMed  Google Scholar 

  32. Oehlers SH, Flores MV, Chen T, Hall CJ, Crosier KE, Crosier PS. Topographical distribution of antimicrobial genes in the zebrafish intestine. Dev Comp Immunol. 2011;35:385–91. https://doi.org/10.1016/j.dci.2010.11.008.

    Article  CAS  PubMed  Google Scholar 

  33. Vallejos-Vidal E, Reyes-Cerpa S, Tort L, Polo J, Reyes-López FE, Gisbert E. Spray-dried porcine plasma promotes the association between metabolic and immunological processes at transcriptional level in gilthead sea bream (Sparus aurata) gut. Front Mar Sci. 2022;9. https://doi.org/10.3389/fmars.2022.814233

  34. Sarasquete C, Gisbert E, Ribeiro L, Vieira L, Dinis MT. Glyconjugates in epidermal, branchial and digestive mucous cells and gastric glands of gilthead sea bream, Sparus aurata, Senegal sole, Solea senegalensis and Siberian sturgeon Acipenser baeri development. Eur J Histochem. 2001;45(3):267–78. https://doi.org/10.4081/1637.

    Article  CAS  PubMed  Google Scholar 

  35. Underwood JCE. Histochemistry. Theoretical and applied. Vol. 2: Analytical technology. In: Pearse AGE, editor. Edinburgh: Churchill Livingstone; 1985. p. 234. https://doi.org/10.1002/path.1711470319.

  36. Sarasquete C, Cárdenas S, González de Canales ML, Pascual E. Oogenesis in the bluefin tuna, Thunnus thynnus L A histological and histochemical study. Histol Histopathol. 2002;17(3):775–88. https://doi.org/10.14670/HH-17.775.

    Article  CAS  PubMed  Google Scholar 

  37. Yamamoto T, Kawai K, Oshima S. Distribution of mucous cells on the body surface of Japanese flounder Paralichthys olivaceus. J Fish Biol. 2011;78(3):848–59. https://doi.org/10.1111/J.1095-8649.2010.02898.X.

    Article  CAS  PubMed  Google Scholar 

  38. Tapia-Paniagua ST, Chabrillón M, Díaz-Rosales P, de la Banda IG, Lobo C, Balebona MC, et al. Intestinal microbiota diversity of the flat fish Solea senegalensis (Kaup, 1858) following probiotic administration. Microb Ecol. 2010;60(2):310–9. https://doi.org/10.1007/s00248-010-9680-z.

    Article  PubMed  Google Scholar 

  39. Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013;41(1). https://doi.org/10.1093/nar/gks808

  40. Firmino JP, Vallejos-Vidal E, Balebona MC, Ramayo-Caldas Y, Cerezo IM, Salomón R, et al. Diet, immunity, and microbiota interactions: an integrative analysis of the intestine transcriptional response and microbiota modulation in gilthead seabream (Sparus aurata) fed an essential oils-based functional diet. Front Immunol. 2021;12. https://doi.org/10.3389/fimmu.2021.625297

  41. Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, et al. The GeneCards Suite: from gene data mining to disease genome sequence analyses. Curr Protoc Bioinformatics. 2016;54(1). https://doi.org/10.1002/CPBI.5

  42. Aragão C, Gonçalves AT, Costas B, Azeredo R, Xavier MJ, Engrola S. Alternative proteins for fish diets: implications beyond growth. Animals. 2022;12(9):1211. https://doi.org/10.3390/ANI12091211.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Martin SAM, Król E. Nutrigenomics and immune function in fish: new insights from omics technologies. Dev Comp Immunol. 2017;75:86–98. https://doi.org/10.1016/J.DCI.2017.02.024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Broadway PR, Carroll JA, Sanchez NCB. Live yeast and yeast cell wall supplements enhance immune function and performance in food-producing livestock: a Review. Microorganisms. 2015;3(3):417–27. https://doi.org/10.3390/MICROORGANISMS3030417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rana KJ, Siriwardena S, Hasan MR. Impact of rising feed ingredient prices on aquafeeds and aquaculture production, Vol. 541. FAO Fisheries and Aquaculture Techical Paper. Rome: Food and Agriculture Organization of the United Nations (FAO); 2009. p. 1–63.

  46. Tacon AGJ, Metian M. Global overview on the use of fish meal and fish oil in industrially compounded aquafeeds: Trends and future prospects. Aquaculture. 2008;285(1–4):146–58. https://doi.org/10.1016/j.aquaculture.2008.08.015.

    Article  CAS  Google Scholar 

  47. Hernández-López IA, Tovar-Ramírez D, de La Rosa-García S, Álvarez-Villagómez CS, Asencio-Alcudia GG, Martínez-Burguete T, et al. Dietary live yeast (Debaryomyces hansenii) provides no advantages in tropical gar, Atractosteus tropicus (Actinopterygii: Lepisosteiformes: Lepisosteidae), juvenile aquaculture. Acta Ichthyol Piscat. 2021;51(3):311–20. https://doi.org/10.3897/AIEP.51.67095.

    Article  Google Scholar 

  48. Agboola JO, Lapeña D, Øverland M, Arntzen MØ, Mydland LT, Hansen JØ. Yeast as a novel protein source - Effect of species and autolysis on protein and amino acid digestibility in Atlantic salmon (Salmo salar). Aquaculture. 2022;546:737312. https://doi.org/10.1016/J.AQUACULTURE.2021.737312.

  49. Baratella D, Bonaiuto E, Magro M, de Almeida RJ, Kanamori Y, Lima GPP, et al. Endogenous and food-derived polyamines: determination by electrochemical sensing. Amino Acids. 2018;50(9):1187–203. https://doi.org/10.1007/s00726-018-2617-4.

    Article  CAS  PubMed  Google Scholar 

  50. Helinck S, Perello MC, Deetae P, de Revel G, Spinnler HE. Debaryomyces hansenii, Proteus vulgaris, Psychrobacter sp. and Microbacterium foliorum are able to produce biogenic amines. Dairy Sci Technol. 2013;93(2):191–200. https://doi.org/10.1007/S13594-012-0102-5.

  51. Wang Q, Xu Z, Ai Q. Arginine metabolism and its functions in growth, nutrient utilization, and immunonutrition of fish. Anim Nutr. 2021;7(3):716–27. https://doi.org/10.1016/J.ANINU.2021.03.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Andersen SM, Holen E, Aksnes A, Ronnestad I, Zerrahn JE, Espe M. Dietary arginine affects energy metabolism through polyamine turnover in juvenile Atlantic salmon (Salmo salar). Br J Nutr. 2013;110(11):1968–77. https://doi.org/10.1017/S0007114513001402.

    Article  CAS  PubMed  Google Scholar 

  53. Celi P, Cowieson AJ, Fru-Nji F, Steinert RE, Kluenter AM, Verlhac V. Gastrointestinal functionality in animal nutrition and health: new opportunities for sustainable animal production. Anim Feed Sci Technol. 2017;234:88–100. https://doi.org/10.1016/j.anifeedsci.2017.09.012.

    Article  Google Scholar 

  54. Kim PS, Shin NR, Lee JB, Kim MS, Whon TW, Hyun DW, et al. Host habitat is the major determinant of the gut microbiome of fish. Microbiome. 2021;9(1):1–16. https://doi.org/10.1186/S40168-021-01113-X.

    Article  Google Scholar 

  55. Merrifield DL, Rodiles A. The fish microbiome and its interactions with mucosal tissues. In: Beck BH, Peatman E, editors. Mucosal Health Aquaculture. Academic Press; 2015. p. 273–95. https://doi.org/10.1016/B978-0-12-417186-2.00010-8.

  56. Hoseinifar SH, Dadar M, van Doan H, Harikrishnan R. Feed additives impacts on shellfish microbiota, health, and development. In: Derome N, editor. Microbial communities in aquaculture ecosystems. Cham: Springer; 2019. p. 143–63. https://doi.org/10.1007/978-3-030-16190-3_7.

  57. Xu L, Xiang P, Zhang B, Yang K, Liu F, Wang Z, et al. Host species influence the gut microbiota of endemic cold-water fish in upper Yangtze River. Front Microbiol. 2022;13. https://doi.org/10.3389/fmicb.2022.906299

  58. Solé-Jiménez P, Naya-Català F, Piazzon MC, Estensoro I, Calduch-Giner J, Sitjà-Bobadilla A, et al. Reshaping of gut microbiota in gilthead sea bream fed microbial and processed animal proteins as the main dietary protein source. Front Mar Sci. 2021;8. https://doi.org/10.3389/fmars.2021.705041

  59. Naya-Català F, do Vale Pereira G, Piazzon MC, Fernandes AM, Calduch-Giner JA, Sitjà-Bobadilla A, et al. Cross-talk between intestinal microbiota and host gene expression in gilthead sea bream (Sparus aurata) juveniles: insights in fish feeds for increased circularity and resource utilization. Front Physiol. 2021;12. https://doi.org/10.3389/fphys.2021.748265

  60. Egerton S, Culloty S, Whooley J, Stanton C, Ross RP. The gut microbiota of marine fish. Front Microbiol. 2018;9. https://doi.org/10.3389/fmicb.2018.00873

  61. Vijayan KK, Bright Singh IS, Jayaprakash NS, Alavandi Sv, Somnath Pai S, Preetha R, et al. A brackishwater isolate of Pseudomonas PS-102, a potential antagonistic bacterium against pathogenic vibrios in penaeid and non-penaeid rearing systems. Aquaculture. 2006;251(2–4):192–200. https://doi.org/10.1016/J.AQUACULTURE.2005.10.010.

    Article  CAS  Google Scholar 

  62. Hoque F, Jawahar Abraham T, Nagesh TS, Kamilya D. Pseudomonas aeruginosa FARP72 offers protection against Aeromonas hydrophila infection in Labeo rohita. Probiotics Antimicrob Proteins. 2019;11(3):973–80. https://doi.org/10.1007/s12602-018-9456-1.

    Article  PubMed  Google Scholar 

  63. Ciulli S, Volpe E, Sirri R, Passalacqua PL, Cesa Bianchi F, Serratore P, et al. Outbreak of mortality in Russian (Acipenser gueldenstaedtii) and Siberian (Acipenser baerii) sturgeons associated with sturgeon nucleo-cytoplasmatic large DNA virus. Vet Microbiol. 2016;191:27–34. https://doi.org/10.1016/J.VETMIC.2016.05.012.

    Article  CAS  PubMed  Google Scholar 

  64. Christon JH. The rasputin effect: when commensals and symbionts become parasitic. Adv Environ Microbiol. 2016;3. https://doi.org/10.1007/978-3-319-28170-4

  65. Wang YW, Zhu J, Ge XP, Sun SM, Su YL, Li B, et al. Effects of stocking density on the growth performance, digestive enzyme activities, antioxidant resistance, and intestinal microflora of blunt snout bream (Megalobrama amblycephala) juveniles. Aquac Res. 2019;50(1):236–46. https://doi.org/10.1111/ARE.13889.

  66. Baker-Austin C, Wright MS, Stepanauskas R, McArthur Jv. Co-selection of antibiotic and metal resistance. Trends Microbiol. 2006;14(4):176–82. https://doi.org/10.1016/J.TIM.2006.02.006.

    Article  CAS  PubMed  Google Scholar 

  67. Maniati M, Ikonomidis A, Mantzana P, Maniatis A, Pournaras S. O491 Pseudomonas aeruginosa with a novel blaVIM-4/blaP1b and a second class-1 integron, effiux pumps overexpression and repressed porin OprD. Int J Antimicrob Agents. 2007;29:S106. https://doi.org/10.1016/S0924-8579(07)70335-7.

    Article  Google Scholar 

  68. Moore JE, Huang J, Yu P, Ma C, Moore PJA, Millar BC, et al. High diversity of bacterial pathogens and antibiotic resistance in salmonid fish farm pond water as determined by molecular identification employing 16S rDNA PCR, gene sequencing and total antibiotic susceptibility techniques. Ecotoxicol Environ Saf. 2014;108:281–6. https://doi.org/10.1016/J.ECOENV.2014.05.022.

    Article  CAS  PubMed  Google Scholar 

  69. Xiong W, Sun Y, Zhang T, Ding X, Li Y, Wang M, et al. Antibiotics, antibiotic resistance genes, and bacterial community composition in fresh water aquaculture environment in China. Microb Ecol. 2015;70(2):425–32. https://doi.org/10.1007/S00248-015-0583-X.

    Article  CAS  PubMed  Google Scholar 

  70. Algammal AM, Mabrok M, Sivaramasamy E, Youssef FM, Atwa MH, El-kholy AW, et al. Emerging MDR-Pseudomonas aeruginosa in fish commonly harbor oprL and toxA virulence genes and blaTEM, blaCTX-M, and tetA antibiotic-resistance genes. Sci Rep. 2020;10:15961. https://doi.org/10.1038/s41598-020-72264-4.

  71. Olmos J, Acosta M, Mendoza G, Pitones V. Bacillus subtilis, an ideal probiotic bacterium to shrimp and fish aquaculture that increase feed digestibility, prevent microbial diseases, and avoid water pollution. Arch Microbiol. 2020;202(3):427–35. https://doi.org/10.1007/S00203-019-01757-2.

    Article  CAS  PubMed  Google Scholar 

  72. James G, Das BC, Jose S, Rejish RK. Bacillus as an aquaculture friendly microbe. Aquac Int. 2021;29(1):323–53. https://doi.org/10.1007/S10499-020-00630-0.

    Article  CAS  Google Scholar 

  73. Caruffo M, Navarrete N, Salgado O, Díaz A, López P, García K, et al. Potential probiotic yeasts isolated from the fish gut protect zebrafish (Danio rerio) from a Vibrio anguillarum challenge. Front Microbiol. 2015;6:1093. https://doi.org/10.3389/FMICB.2015.01093.

    Article  PubMed  PubMed Central  Google Scholar 

  74. He J, He Y, Pan D, Cao J, Sun Y, Zeng X. Associations of gut microbiota with heat stress-induced changes of growth, fat deposition, intestinal morphology, and antioxidant capacity in ducks. Front Microbiol. 2019;10:903. https://doi.org/10.3389/FMICB.2019.00903.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Shao X, Sun C, Tang X, Zhang X, Han D, Liang S, et al. Anti-inflammatory and intestinal microbiota modulation properties of Jinxiang garlic (Allium sativum L.) polysaccharides toward dextran sodium sulfate-induced colitis. J Agric Food Chem. 2020;68(44):12295–309. https://doi.org/10.1021/ACS.JAFC.0C04773.

  76. Cornick S, Tawiah A, Chadee K. Roles and regulation of the mucus barrier in the gut. Tissue Barriers. 2015;3(1–2). https://doi.org/10.4161/21688370.2014.982426

  77. Ouwerkerk JP, de Vos WM, Belzer C. Glycobiome: Bacteria and mucus at the epithelial interface. Best Pract Res Clin Gastroenterol. 2013;27(1):25–38. https://doi.org/10.1016/J.BPG.2013.03.001.

    Article  CAS  PubMed  Google Scholar 

  78. Salomón R, Reyes-López FE, Tort L, Firmino JP, Sarasquete C, Ortiz-Delgado JB, et al. Medicinal plant leaf extract from sage and lemon verbena promotes intestinal immunity and barrier function in gilthead seabream (Sparus aurata). Front Immunol. 2021;12. https://doi.org/10.3389/fimmu.2021.670279

  79. Čapkovičová A, Maková Z, Piešová E, Alves A, Faix Š, Faixová Z. Evaluation of the effects of salvia officinalis essential oil on plasma biochemistry, gut mucus and quantity of acidic and neutral mucins in the chicken gut. Acta Vet Brno. 2014;64(1):138–48. https://doi.org/10.2478/ACVE-2014-0014.

    Article  Google Scholar 

  80. Bosi A, Banfi D, Bistoletti M, Moretto P, Moro E, Crema F, et al. Hyaluronan: a neuroimmune modulator in the microbiota-gut axis. Cells. 2021;11(1):126. https://doi.org/10.3390/CELLS11010126.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Matrosovich M, Herrler G, Klenk HD. Sialic acid receptors of viruses. Top Curr Chem. 2015;367:1–28. https://doi.org/10.1007/128_2013_466.

    Article  CAS  PubMed  Google Scholar 

  82. Redondo MJ, Alvarez-Pellitero P. Carbohydrate patterns in the digestive tract of Sparus aurata L. and Psetta maxima (L.) (Teleostei) parasitized by Enteromyxum leei and E. scophthalmi (Myxozoa). Parasitol Int. 2010;59(3):445–53. https://doi.org/10.1016/J.PARINT.2010.06.005.

  83. Quintana-Hayashi MP, Padra M, Padra JT, Benktander J, Lindén SK. Mucus-pathogen interactions in the gastrointestinal tract of farmed animals. Microorganisms. 2018;6(2):55. https://doi.org/10.3390/MICROORGANISMS6020055.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Schjoldager KT, Narimatsu Y, Joshi HJ, Clausen H. Global view of human protein glycosylation pathways and functions. Nat Rev Mol Cell Biol. 2020;21(12):729–49. https://doi.org/10.1038/S41580-020-00294-X.

    Article  CAS  PubMed  Google Scholar 

  85. Legler K, Rosprim R, Karius T, Eylmann K, Rossberg M, Wirtz RM, et al. Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer. Br J Cancer. 2018;118(6):847–56. https://doi.org/10.1038/bjc.2017.472.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kumura H, Tanoue Y, Tsukahara M, Tanaka T, Shimazaki K. Screening of dairy yeast strains for probiotic applications. J Dairy Sci. 2004;87(12):4050–6. https://doi.org/10.3168/jds.S0022-0302(04)73546-8.

    Article  CAS  PubMed  Google Scholar 

  87. Liu Y, Liu X, Yang L, Qiu Y, Pang J, Hu X, et al. Adjuvanticity of β-glucan for vaccine against Trichinella spiralis. Front Cell Dev Biol. 2021;9:1803. https://doi.org/10.3389/FCELL.2021.701708.

    Article  Google Scholar 

  88. Gatesoupe FJ. Live yeasts in the gut: Natural occurrence, dietary introduction, and their effects on fish health and development. Aquaculture. 2007;267(1–4):20–30. https://doi.org/10.1016/J.AQUACULTURE.2007.01.005.

    Article  Google Scholar 

  89. Kaushik SJ, Seiliez I. Protein and amino acid nutrition and metabolism in fish: current knowledge and future needs. Aquac Res. 2010;41(3):322–32. https://doi.org/10.1111/J.1365-2109.2009.02174.X.

    Article  CAS  Google Scholar 

  90. Glickman MH, Ciechanover A. The ubiquitin-proteasome proteolytic pathway: Destruction for the sake of construction. Physiol Rev. 2002;82(2):373–428. https://doi.org/10.1152/physrev.00027.2001.

    Article  CAS  PubMed  Google Scholar 

  91. Kors S, Geijtenbeek K, Reits E, Schipper-Krom S. Regulation of proteasome activity by (post) transcriptional mechanisms. Front Mol Biosci. 2019;6:48. https://doi.org/10.3389/FMOLB.2019.00048.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Cohen EB, Gecki RC, Toker A. Metabolic pathway alterations in microvascular endothelial cells in response to hypoxia. PLoS One. 2020;15(7):e0232072. https://doi.org/10.1371/JOURNAL.PONE.0232072.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Frisk JH, Eriksson S, Pejler G, Wang L. Identification of a novel thymidylate kinase activity. Nucleosides Nucleotides Nucleic Acids. 2020;39(10–12):1359–68. https://doi.org/10.1080/15257770.2020.1755043.

    Article  CAS  PubMed  Google Scholar 

  94. Vanoevelen JM, Bierau J, Grashorn JC, Lambrichs E, Kamsteeg EJ, Bok LA, et al. DTYMK is essential for genome integrity and neuronal survival. Acta Neuropathol. 2022;143(2):245–62. https://doi.org/10.1007/S00401-021-02394-0.

    Article  PubMed  Google Scholar 

  95. Sepahi A, Cordero H, Goldfine H, Esteban MÁ, Salinas I. Symbiont-derived sphingolipids modulate mucosal homeostasis and B cells in teleost fish. Sci Rep. 2016;6:39054. https://doi.org/10.1038/srep39054.

  96. Kitatani K, Idkowiak-Baldys J, Hannun YA. The sphingolipid salvage pathway in ceramide metabolism and signaling. Cell Signal. 2008;20(6):1010–8. https://doi.org/10.1016/J.CELLSIG.2007.12.006.

    Article  CAS  PubMed  Google Scholar 

  97. Parveen F, Bender D, Law SH, Mishra VK, Chen CC, Ke LY. Role of ceramidases in sphingolipid metabolism and human diseases. Cells. 2019;8(12):1573. https://doi.org/10.3390/CELLS8121573.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Rodriguez-Cuenca S, Barbarroja N, Vidal-Puig A. Dihydroceramide desaturase 1, the gatekeeper of ceramide induced lipotoxicity. Biochim Biophys Acta. 2015;1851(1):40–50. https://doi.org/10.1016/J.BBALIP.2014.09.021.

    Article  CAS  PubMed  Google Scholar 

  99. Frisard M, Ravussin E. Energy metabolism and oxidative stress: impact on the metabolic syndrome and the aging process. Endocrine. 2006;29(1):27–32. https://doi.org/10.1385/ENDO:29:1:27.

    Article  CAS  PubMed  Google Scholar 

  100. Turrens JF. Mitochondrial formation of reactive oxygen species. J Physiol. 2003;552(2):335–44. https://doi.org/10.1113/JPHYSIOL.2003.049478.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Heo S, Kim S, Kang D. The role of hydrogen peroxide and peroxiredoxins throughout the cell cycle. Antioxidants. 2020;9(4):280. https://doi.org/10.3390/ANTIOX9040280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Mazza R, Gattuso A, Imbrogno S, Boukhzar L, Leo S, Mallouki BY, et al. Selenoprotein T as a new positive inotrope in the goldfish, Carassius auratus. J Exp Biol. 2019;222(11). https://doi.org/10.1242/jeb.201202

  103. Magalhães R, Guerreiro I, Santos RA, Coutinho F, Couto A, Serra CR, et al. Oxidative status and intestinal health of gilthead sea bream (Sparus aurata) juveniles fed diets with different ARA/EPA/DHA ratios. Sci Rep. 2020;10:13824. https://doi.org/10.1038/s41598-020-70716-5.

  104. Garner AP, Paine MJI, Rodriguez-Crespo I, Chinje EC, Ortiz P, Montellano D, et al. Nitric oxide synthases catalyze the activation of redox cycling and bioreductive anticancer agents 1. Cancer Res. 1999;59:1929–34.

  105. Sánchez-Nuño S, Sanahuja I, Fernández-Alacid L, Ordóñez-Grande B, Carbonell T, Ibarz A. Oxidative attack during temperature fluctuation challenge compromises liver protein homeostasis of a temperate fish model. Comp Biochem Physiol B Biochem Mol Biol. 2019;236. https://doi.org/10.1016/j.cbpb.2019.110311

  106. Marí M, de Gregorio E, de Dios C, Roca-Agujetas V, Cucarull B, Tutusaus A, et al. Mitochondrial glutathione: recent insights and role in disease. Antioxidants. 2020;9(10):909. https://doi.org/10.3390/ANTIOX9100909.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Torriglia A, Martin E, Jaadane I. The hidden side of SERPINB1/Leukocyte Elastase Inhibitor. Semin Cell Dev Biol. 2017;62:178–86. https://doi.org/10.1016/J.SEMCDB.2016.07.010.

    Article  CAS  PubMed  Google Scholar 

  108. Tiana M, Acosta-Iborra B, Puente-Santamaría L, Hernansanz-Agustin P, Worsley-Hunt R, Masson N, et al. The SIN3A histone deacetylase complex is required for a complete transcriptional response to hypoxia. Nucleic Acids Res. 2018;46(1):120–33. https://doi.org/10.1093/NAR/GKX951.

    Article  CAS  PubMed  Google Scholar 

  109. Haddad JJ, Fahlman CS. Redox- and oxidant-mediated regulation of interleukin-10: an anti-inflammatory, antioxidant cytokine? Biochem Biophys Res Commun. 2002;297(2):163–76. https://doi.org/10.1016/S0006-291X(02)02094-6.

    Article  CAS  PubMed  Google Scholar 

  110. Tovar-Ramírez D, Mazurais D, Gatesoupe JF, Quazuguel P, Cahu CL, Zambonino-Infante JL. Dietary probiotic live yeast modulates antioxidant enzyme activities and gene expression of sea bass (Dicentrarchus labrax) larvae. Aquaculture. 2010;300(1–4):142–7. https://doi.org/10.1016/J.AQUACULTURE.2009.12.015.

    Article  Google Scholar 

  111. Li Y, Chen R, Zhou Q, Xu Z, Li C, Wang S, et al. LSm14A is a processing body-associated sensor of viral nucleic acids that initiates cellular antiviral response in the early phase of viral infection. Proc Natl Acad Sci U S A. 2012;109(29):11770–5. https://doi.org/10.1073/pnas.120340510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Martín-Vicente M, Medrano LM, Resino S, García-Sastre A, Martínez I. TRIM25 in the regulation of the antiviral innate immunity. Front Immunol. 2017;8:1187. https://doi.org/10.3389/fimmu.2017.01187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Refolo G, Vescovo T, Piacentini M, Fimia GM, Ciccosanti F. Mitochondrial interactome: a focus on antiviral signaling pathways. Front Cell Dev Biol. 2020;8. https://doi.org/10.3389/fcell.2020.00008

  114. Devenish LP, Mhlanga MM, Negishi Y. Immune regulation in time and space: the role of local- and long-range genomic interactions in regulating immune responses. Front Immunol. 2021;12. https://doi.org/10.3389/fimmu.2021.662565

  115. Wang D, Gou M, Hou J, Pang Y, Li Q. The role of serpin protein on the natural immune defense against pathogen infection in Lampetra japonica. Fish Shellfish Immunol. 2019;92:196–208. https://doi.org/10.1016/J.FSI.2019.05.062.

    Article  CAS  PubMed  Google Scholar 

  116. Tomasello E, Vivier E. KARAP/DAP12/TYROBP: three names and a multiplicity of biological functions. Eur J Immunol. 2005;35(6):1670–7. https://doi.org/10.1002/EJI.200425932.

    Article  CAS  PubMed  Google Scholar 

  117. Wei X, Li J, Zhan X, Tu L, Huang H, Wang Y. A novel missense mutation in complement factor I predisposes patients to atypical hemolytic uremic syndrome: a case report. J Med Case Rep. 2022;16(1):1–6. https://doi.org/10.1186/s13256-022-03312-y.

    Article  Google Scholar 

  118. Zou J, Secombes CJ. The function of fish cytokines. Biology (Basel). 2016;5(2):23. https://doi.org/10.3390/BIOLOGY5020023.

    Article  PubMed  Google Scholar 

  119. Huo HJ, Chen SN, Li L, Nie P. Functional characterization of IL-10 and its receptor subunits in a perciform fish, the mandarin fish Siniperca chuatsi. Dev Comp Immunol. 2019;97:64–75. https://doi.org/10.1016/J.DCI.2019.03.017.

    Article  CAS  PubMed  Google Scholar 

  120. McConnell SC, Hernandez KM, Wcisel DJ, Kettleborough RN, Stemple DL, Yoder JA, et al. Alternative haplotypes of antigen processing genes in zebrafish diverged early in vertebrate evolution. Proc Natl Acad Sci USA. 2016;113(34):E5014–23. https://doi.org/10.1073/pnas.1607602113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Hadiuzzaman M, Moniruzzaman M, Shahjahan M, Bai SC, Min T, Hossain Z. β-glucan: mode of action and its uses in fish immunomodulation. Front Mar Sci. 2022;9. https://doi.org/10.3389/fmars.2022.905986

  122. Reyes-Becerril M, Angulo M, Sanchez V, Guluarte C, Angulo C. β-D-glucan from marine yeast Debaryomyces hansenii BCS004 enhanced intestinal health and glucan-expressed receptor genes in Pacific red snapper Lutjanus Peru. Microb Pathog. 2020;143:104141. https://doi.org/10.1016/J.MICPATH.2020.104141.

    Article  CAS  PubMed  Google Scholar 

  123. Machuca C, Méndez-Martínez Y, Reyes-Becerril M, Angulo C. Yeast β-glucans as fish immunomodulators: a review. Animals. 2022;12(16):2154. https://doi.org/10.3390/ANI12162154.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Vidya SRG, Vijaya Ch. Efficacy of β-glucan from Debaryomyces hansenii as an immunostimulant in Litopenaeus vannamei culture. Aquacult Int. 2021;29(4):1451–8. https://doi.org/10.1007/s10499-021-00678-6.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the technicians at the IRTA facilities for their valuable help during fish rearing.

Funding

This work has been financed through the DIETAplus project of JACUMAR (Junta de Cultivos Marinos, MAPAMA; Spanish government), which is co-funded with FEMP funds (EU). Furthermore, this research was funded by means of grants from the Spanish Government: PID2019-106878RB-I00 and IS was granted with a Postdoctoral fellowship (FJC2020-043933-I). EVV and FERL thank the support of Fondecyt iniciación (project number 11221308) and Fondecyt regular (project number 11221308) grants (Agencia Nacional de Investigacióny Desarrollo de Chile, Government of Chile), respectively. Collaboration between Ibero-American researchers has been done under the framework of the network LARVAplus “Strategies for the development and im-provement of fish larvae production in Ibero-America” (117RT0521) funded by the Ibero-American Program of Science and Technology for Development (CYTED, Spain).

Author information

Authors and Affiliations

Authors

Contributions

EG designed and carried out the experiments. Biological samplings were performed by EG, JPF, AR, and IS. The transcriptomic data analysis and interpretation were performed by IS, EV-V, and FER-L. IMC and MAM carried out the microbiota analyses. JBO-D and CS carried out the histological and histochemical analysis. DT-R reviewed and validated the methodology used in the study. The study was supervised by EG, IS, and LT. IS wrote the original draft. All the authors provided the critical feedback, read, and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Enric Gisbert.

Ethics declarations

Ethics approval and consent to participate

This study was conducted following the Guiding Principles for Biomedical Research Involving Animals (EU2010/63), the guidelines of the Spanish laws (law 32/2007 and RD 53/2013) and authorized by the Ethical Committee of the Institute for Research and Technology in Food and Agriculture (Spain), and Generalitat de Catalunya government for the use of laboratory animals (FUE-2020–01719167).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Supplementary Information

Additional file 1: Fig. S1:

Principal coordinate analysisof the anterior-mid intestine microbiota similarity. Table S1: Summary of the differentially expressed genesobtained from the microarray-based transcriptomic analysis for the gut of juvenile gilthead seabream fed the D. hansenii diet. Table S2: Functional enrichment analysis of the DEGs. Biological processidentified in the intestine of juvenile gilthead seabreamfed the D. hansenii diet. Table S3: Top-10 biological processof the DEGs identified in the intestine of juvenile gilthead seabreamfed the D. hansenii diet. Table S4: Representative cluster of the functional enrichment network for biological processes based on the related functions of the differentially expressed genesin the anterior-mid intestine. Table S5: Cellular componentand annotated keywordsof the DEGs identified in the intestine of juvenile gilthead seabreamfed the D. hansenii diet.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sanahuja, I., Ruiz, A., Firmino, J.P. et al. Debaryomyces hansenii supplementation in low fish meal diets promotes growth, modulates microbiota and enhances intestinal condition in juvenile marine fish. J Animal Sci Biotechnol 14, 90 (2023). https://doi.org/10.1186/s40104-023-00895-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-023-00895-4

Keywords