Hostname: page-component-7c8c6479df-27gpq Total loading time: 0 Render date: 2024-03-28T01:26:00.947Z Has data issue: false hasContentIssue false

Comprehensive biometric, biochemical and histopathological assessment of nutrient deficiencies in gilthead sea bream fed semi-purified diets

Published online by Cambridge University Press:  29 July 2015

Gabriel F. Ballester-Lozano
Affiliation:
Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal, IATS-CSIC, 12595 Castellón, Spain
Laura Benedito-Palos
Affiliation:
Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal, IATS-CSIC, 12595 Castellón, Spain
Itziar Estensoro
Affiliation:
Fish Pathology Group, Instituto de Acuicultura Torre de la Sal, IATS-CSIC, 12595 Castellón, Spain
Ariadna Sitjà-Bobadilla
Affiliation:
Fish Pathology Group, Instituto de Acuicultura Torre de la Sal, IATS-CSIC, 12595 Castellón, Spain
Sadasivam Kaushik
Affiliation:
INRA, UR1067 NuMeA Nutrition, Metabolism Aquaculture, F-64310 Saint Pée-sur Nivelle, France
Jaume Pérez-Sánchez*
Affiliation:
Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal, IATS-CSIC, 12595 Castellón, Spain
*
*Corresponding author: J. Pérez-Sánchez, fax +34 964319509, email jaime.perez.sanchez@csic.es
Rights & Permissions [Opens in a new window]

Abstract

Seven isoproteic and isolipidic semi-purified diets were formulated to assess specific nutrient deficiencies in sulphur amino acids (SAA), n-3 long-chain PUFA (n-3 LC-PUFA), phospholipids (PL), P, minerals (Min) and vitamins (Vit). The control diet (CTRL) contained these essential nutrients in adequate amounts. Each diet was allocated to triplicate groups of juvenile gilthead sea bream fed to satiety over an 11-week feeding trial period. Weight gain of n-3 LC-PUFA, P–Vit and PL–Min–SAA groups was 50, 60–75 and 80–85 % of the CTRL group, respectively. Fat retention was decreased by all nutrient deficiencies except by the Min diet. Strong effects on N retention were found in n-3 LC-PUFA and P fish. Combined anaemia and increased blood respiratory burst were observed in n-3 LC-PUFA fish. Hypoproteinaemia was found in SAA, n-3 LC-PUFA, PL and Vit fish. Derangements of lipid metabolism were also a common disorder, but the lipodystrophic phenotype of P fish was different from that of other groups. Changes in plasma levels of electrolytes (Ca, phosphate), metabolites (creatinine, choline) and enzyme activities (alkaline phosphatase) were related to specific nutrient deficiencies in PL, P, Min or Vit fish, whereas changes in circulating levels of growth hormone and insulin-like growth factor I primarily reflected the intensity of the nutritional stressor. Histopathological scoring of the liver and intestine segments showed specific nutrient-mediated changes in lipid cell vacuolisation, inflammation of intestinal submucosa, as well as the distribution and number of intestinal goblet and rodlet cells. These results contribute to define the normal range of variation for selected biometric, biochemical, haematological and histochemical markers.

Type
Full Papers
Copyright
Copyright © The Authors 2015 

Clinical haematology and basic blood biochemistry are common diagnostic tools to assess health and welfare in humans and most livestock production systems( Reference Knox, Reid and Irwin 1 , Reference Kerr 2 ). In fish, although there is experimental evidence that circulating electrolytes, metabolites and hormones highly reflect impaired growth performance, stress condition and disease outcome, the use of such analyses as diagnostic tools is poorly established in practice. This is due to the paucity of reliable information on reference values of haematology and blood biochemistry parameters in healthy and well-nourished fish. Some attempts have been made to compile available data in fish blood biochemistry and haematology( Reference Peres, Santos and Oliva-Teles 3 Reference Hrubec and Smith 6 ). However, for the majority of well-established farmed finfish, grown under different rearing conditions with diverse physiological status, validated data are lacking. Besides, important gaps on reliable clinical biomarkers are arising with the advent of new fish feeds with a maximised replacement of fishmeal (FM) and fish oil (FO) by alternative feedstuffs of terrestrial or marine origin. Similarly, for the histopathological scoring of relevant target tissues (liver, intestine), there is evidence of clinical signs of liver steatosis, accumulation of intestinal lipid droplets or intestine submucosa inflammation arising from lipid-related metabolic disorders( Reference Benedito-Palos, Navarro and Sitjà-Bobadilla 7 , Reference Torstensen and Tocher 8 ), but a direct link to a specific nutrient or a group of nutrients is lacking.

Therefore, there is an urgent need for reliable reference values, but also for the definition of blood and histopathological parameters that have specificity, sensitivity and diagnostic value for nutritional deficiencies. Thus, our experimental setup with a feeding trial of semi-purified diets formulated for a given nutritional deficiency in a typically marine fish such as the gilthead sea bream considered the following two major steps: (i) functional validation of a set of clinical data based on body composition, organosomatic indices, and blood haematology and biochemistry for the initial assessment of nutrient deficiencies in methionine (Met), essential fatty acids (EFA) such as the n-3 long-chain PUFA (n-3 LC PUFA), phospholipids (PL), P and micronutrients (minerals, vitamins); and (ii) histopathological scoring of liver and intestine sections as a complementary diagnostic tool. The studied nutrient deficiencies were chosen because they are constraining factors in practical marine fish feeds with a maximised FM/FO replacement. In parallel, current work is underway for the definition of the normal range of variation of selected biomarkers, integrating the data reported here with our own data in the framework of the ARRAINA (Advanced Research Initiatives for Nutrition and Aquaculture) EU project, where fish were fed through the production cycle with varying inclusion levels of FM and FO (from 40 % in the control (CTRL) diet to 7·5 % in the extreme low FM/FO diet).

Methods

Diets

Seven isonitrogenous (51–52 % of DM) and isolipidic (14·5–15·5 % of DM) diets were formulated. They were produced in a semi-industrial scale (Sparos LDA) (Table 1). All diets contained casein (20 %), casein hydrolysate (5 %), gelatin (5·8 %) and soya protein concentrate (34·5 %) as protein sources, and were supplemented with l-threonine (0·02 %). Taurine (0·3 %), betaine (0·3 %) and glucosamine (0·4 %) were added as attractants, and ethoxyquin (0·1 %) as the antioxidant in all diets. dl-Methionine was supplemented at 0·4 % in all diets, except in the diet designed to be deficient in sulphur amino acids (SAA diet). FO was added at 13·9 % in all diets, except in the fatty acid (FA)-deficient diet (n-3 LC-PUFA diet), in which FO was totally replaced by a blend of vegetable oils (VO) in order to reduce the EPA and DHA contents to trace levels (Table 2). Soya lecithin (2 %) was added as the unique source of PL in all diets, except in the PL-deficient diet. Calcium phosphate (2·2 %) was added in all diets, except in the P-deficient diet. Mineral premix based on available data on mineral requirements of fish( Reference Antony Jesu Prabhu, Schrama and Kaushik 9 ) was included at 2·2 % in all diets, except in the diet designed to be mineral deficient (Min diet). Vitamin premix based on NRC( 10 ) was added in all diets at an incorportaion level of 2 %, except in the diet designed to be vitamin deficient (Vit diet).

Table 1 Ingredients and chemical composition of the experimental diets

CTRL, control; SAA, sulphur amino acids; n-3 LC-PUFA, n-3 long-chain PUFA; PL, phospholipids; Min, minerals; Vit, vitamins.

* Supplied the following (g/kg mix): calcium hydrogen phosphate 500, calcium carbonate (40 % Ca) 215, sodium chloride 40, ferrous sulphate (21 % Fe) 20, manganese sulphate 3, zinc sulphate 4, copper sulphate 3, cobalt (II) chloride (25 % Co) 0·02, potassium iodine 0·04, sodium selenite 0·03, sodium fluoride 1, magnesium hydroxide (60 % Mg) 124 and potassium chloride 90.

Supplied the following (g/kg mix, except as noted): retinyl acetate 1, dl-cholecalciferol 2·5, dl-α tocopheryl acetate 5, menadione sodium bisulphite 1, ascorbic acid 20, thiamin 0·1, riboflavin 0·4, pyridoxine 0·3, vitamin B12 10 mg, nicotinic acid 1, pantothenic acid 2, folic acid 0·1, biotin 10 mg, choline chloride 200, inositol 30.

Table 2 Fatty acid composition of the experimental diets (% total fatty acid methyl esters) (Mean values of two determinations)

CTRL, control; SAA, sulphur amino acids; n-3 LC-PUFA, n-3 long-chain PUFA; PL, phospholipids; Min, minerals; Vit, vitamins; Tr, trace value<0·05.

* Fatty acids with at least twenty carbon atoms and more than three double bonds.

Feeding trial and fish sampling

Juvenile sea bream of Atlantic origin (Ferme Marine de Douhet, Ile d’Oléron, France) were acclimatised to laboratory conditions for one month before the start of a 12-week trial (May–July) in the indoor experimental facilities of the Institute of Aquaculture Torre de la Sal (IATS-CSIC). Following the acclimatisation period, fish of 15 g initial mean body weight were randomly distributed into 500 L tanks in triplicates of 35 fish each. Fish were fed to visual satiety one (12 h)/two times (9 h, 14 h) per day (6 d/week). The trial was conducted under natural photoperiod and temperature conditions at IATS latitude (40°5 N; 0°10E), and the water temperature was increased from 19°C in May to 24°C at the end of July. Water flow rate was 20 L/min, oxygen content of water effluents was always higher than 85 % saturation and unionised ammonia remained below toxic levels (<0·02 mg/l).

At the end of the trial and following overnight fasting (10–12 h in the morning), 18 fish per dietary treatment (six per tank) were randomly selected and decapitated under anaesthesia with 3-aminobenzoic acid ethyl ester (MS-222, 100 μg/ml). Blood was taken from the caudal vessels with heparinised syringes (less than 5 min for all the fish sampled from each tank). One aliquot was used for measurements of haematological parameters and respiratory burst (RB) activity of blood leucocytes. The remaining blood was centrifuged at 3000 g for 20 min at 4°C, and the plasma obtained was stored in separate aliquots at –20°C until further assays were performed. Viscera, liver and mesenteric fat were weighed and representative portions of the liver and intestine segments (anterior and posterior) were taken for histological processing. When blood and tissue collection was completed, additional fish (four fish per tank) were taken for whole body composition analyses.

The experimental protocol was approved by the Agencia Estatal Consejo Superior de Investigaciones Científicas, IATS Review Board, and all procedures were in accordance with the national and current EU legislations on the handling of experimental animals.

Chemical composition

Diets and fish for body composition analyses (a pooled sample of ten fish at the beginning and four fish per tank at the end of trial) were ground, and small aliquots were dried to determine moisture content. The remaining samples were freeze-dried and chemical analyses were carried out: DM by drying at 105°C for 24 h, protein (N×6·25) by the Kjeldahl method and fat after dichloromethane extraction by the Soxhlet method.

Total lipids for analyses of dietary FA acid composition were extracted by the method described by Folch et al.( Reference Folch, Lees and Sloane Stanley 11 ), with chloroform–methanol (2:1, v/v) containing 0·01 % butylated hydroxytoluene (BHT) as antioxidant. After the addition of nonadecanoic FA (19:0) as internal standard, total lipids were subjected to acid-catalysed transmethylation for 16 h at 50°C using 1 ml toluene and 2 ml of 1 % (v/v) sulphuric acid in methanol( Reference Christie 12 ). Fatty acid methyl esters (FAME) were extracted with hexane:diethyl ether (1:1) and purified by TLC (Silica gel G 60, 20×20 cm glass plates, Merck), using hexane:diethyl ether:acetic acid (85:15:1·5) as the solvent system. FAMEs were then analysed with Fisons Instruments GC 8000 Series (Fisons) Gas Chromatograph as described elsewhere( Reference Ballester-Lozano, Benedito-Palos and Navarro 13 ). Individual FAMEs were identified by comparison with well-characterised sardine oil (Marinol; Fishing Industry Research Institute) and the FAME 37 mix from Supelco. BHT and internal standards (19:0) were obtained from Sigma-Aldrich. All solvents used for lipid extraction and FA analyses were HPLC grade and were obtained from Merck.

Haematology

Hb concentration was determined using a HemoCue B-Haemoglobin Analyser® (AB Leo Diagnostic), which uses a modified azide methaemoglobin reaction for Hb quantification; haematocrit (Hct) was measured after centrifugation of the blood in heparinised capillary tubes at 13 000 g for 10 min. RBC counts were made in a Neubauer chamber using an isotonic solution (1 % NaCl). Erythrocyte osmotic fragility test was carried out in hypotonic NaCl solutions with haemolysis read at 540 nm. Median corpuscular fragility (MCF) was defined as the concentration of NaCl (g/l) causing 50 % lysis.

Blood biochemistry

Plasma glucose was measured by the glucose oxidase method (Thermo Fisher Scientific). Plasma TAG were determined using lipase/glycerol kinase/glycerol-3-phosphate oxidase reagent. Total plasma cholesterol was determined using cholesterol esterase/cholesterol dehydrogenase reagent (Thermo Fisher Scientific). HDL and LDL/VLDL-cholesterol were determined using a kit (EHDL-100) from BioAssay Systems, based on an improved polyethylene glycol precipitation method in which HDL and LDL/VLDL are separated and cholesterol concentrations are determined using cholesterol esterase/cholesterol dehydrogenase reagent. Total plasma proteins were measured with the Bio-Rad protein reagent (Bio-Rad) with bovine serum albumin as the standard.

Changes in plasma enzyme activities of alanine aminotransferase (ALAT, EC 2.6.1.2) (EALT-100), aspartate aminotransferase (ASAT, EC 2.6.1.1) (EASTR-100) and glutamate dehydrogenase (GLDH, EC 1.4.1.2) (DGLDH-100) were measured using colorimetric assay kits (BioAssay Systems). Plasma alkaline phosphatase (ALP, EC 3.1.3.1) activity was determined using a fluorimetric assay kit (QFAP-100, BioAssay Systems).

Plasma levels of creatinine (DICT-500), choline (ECHO-100), calcium (DICA-500), chloride (DICL-250), magnesium (DMG-250) and phosphate (DIPI-500) were measured using colorimetric assay kits (BioAssay Systems). Total antioxidant capacity was measured as Trolox activity using a microplate assay kit (709001) (Cayman Chemical). Plasma lysozyme activity was measured by a turbidimetric assay( Reference Ellis 14 ) adapted to microplates, as previously described( Reference Sitjà-Bobadilla, Peña-Llopis and Gómez-Requeni 15 ). Induction of RB activity in blood leucocytes was measured directly from heparinised blood, following the method described by Nikoskelainen et al.( Reference Nikoskelainen, Verho and Airas 16 ) with some modifications( Reference Saera-Vila, Calduch-Giner and Prunet 17 ).

Plasma growth hormone (GH) was determined by a homologous gilthead sea bream RIA as reported elsewhere( Reference Martínez-Barber, Pendón and Martí-Palanca 18 ). The sensitivity and midrange (ED50) of the assay were 0·15 and 1·8 ng/ml, respectively. Plasma insulin-like growth factors (IGF) were extracted by acid–ethanol cryoprecipitation( Reference Shimizu, Swanson and Fukada 19 ), and the concentration of IGF-I was measured by means of a generic fish IGF-I RIA validated for Mediterranean perciform fish( Reference Vega-Rubín de Celis, Gómez-Requeni and Pérez-Sánchez 20 ). The sensitivity and midrange of the assay were 0·05 and 0·7–0·8 ng/ml, respectively.

Histology

For histological examination, pieces of the liver, anterior (a piece immediately after the piloric caeca) and posterior (a piece immediately before the rectum) intestinal segments were fixed in 10 % buffered formalin, embedded in paraffin, 4-μm sectioned and stained with Giemsa and periodic acid–Schiff (PAS). Histochemical reactivity in tissues was evaluated by grading staining using the following scale: negative (–), slight (+), moderate (++) and marked (+++).

Statistical analysis

Data on growth performance and blood haematology and biochemistry were analysed by one-way ANOVA followed by the Student Newman–Keuls test (P<0·05). All the analyses were performed with SPSS 17.0 program (SPSS Inc.).

Results

Growth performance

Data on growth, somatic indices and body composition are shown in Table 3. As a general rule, nutrient-deficient diets reduced significantly feed intake, growth rates and feed efficiency in fish fed P and n-3 LC-PUFA diets. This resulted in a weight gain of 50 % (n-3 LC-PUFA), 60–75 % (P, Vit) and 80–85 % (PL, Min, SAA) of CTRL fish.

Table 3 Effect of nutrient deficiencies on growth performance of gilthead sea bream fed to visual satiety from May to July (13 weeks) (Mean values with their standard errors)

CTRL, control; SAA, sulphur amino acids; n-3 LC-PUFA, n-3 long-chain PUFA; PL, phospholipids; Min, minerals; Vit, vitamins.

Data on body weight, feed intake, growth indices and body composition are the mean values with their standard errors of the mean of triplicate tanks. Data on viscera, mesenteric fat and liver weight are the mean values with their standard errors of the mean of twenty fish.

a,b,c,d Mean values with unlike superscript letters in each row indicate significant differences among dietary treatments (Student Newman–Keuls test, P<0·05)

* Result values from one-way ANOVA.

Viscerosomatic index (VSI)=(100×viscera weight)/fish weight.

Mesenteric index (MSI)=(100×mesenteric fat weight)/fish weight.

§ Hepatosomatic index (HSI)=(100×liver weight)/fish weight.

|| Specific growth rate (SGR)=100×(ln final body weight–ln initial body weight)/d.

Feed efficiency (FE)=wet weight gain/dry feed intake.

Mesenteric fat index (MSI) was markedly reduced in Vit fish. The same was observed in PL and n-3 LC-PUFA groups, although there were no statistically significant differences. Conversely, MSI was significantly increased in P fish. Hepatosomatic index (HSI) was also altered by dietary treatments, and it was largely increased in n-3 LC-PUFA fish. The opposite was found in Min fish and in a lower extent in Met and Vit fish.

Dietary treatment also altered whole body composition with low protein and lipid content in fish fed P and Vit diets, respectively. This feature was related to a strong decrease in N retention in P and n-3 LC-PUFA fish. Lipid retention was significantly reduced by nutrient deficiencies in all experimental groups with the exception of Min fish.

Blood analyses

Data on blood analysis are shown in Table 4. Hb concentration, Hct and RBC counts were significantly lower in n-3 LC-PUFA fish than in CTRL fish. This feature was related to a greater osmotic fragility, evidenced by the significant increase of MCF values from 6·6 g/l in CTRL fish to 7·4 g/l in n-3 LC-PUFA fish. Other experimental groups did not show any statistically significant alterations in haematological parameters.

Table 4 Effect of nutrient deficiencies on basic plasma biochemistry of sea bream fed to visual satiety from May to July (13 weeks) (Mean values with their standard errors of ten fish)

CTRL, control; SAA, sulphur amino acids; n-3 LC-PUFA, n-3 long-chain PUFA; PL, phospholipids; Min, minerals; Vit, vitamins; MCF, median corpuscular fragility; ND, not determined; ALAT, alanine aminotransferase; ASAT, aspartate aminotransferase; GLDH, glutamate dehydrogenase; ALP, alkaline phosphatase; GH, growth hormone; IGF, insulin-like growth factors.

a,b,c Unlike superscript letters in each row indicate significant differences among dietary treatments (Student Newman–Keuls test, P<0·05)

* Result values from one-way ANOVA.

To convert Hb from g/dl to g/l, multiply by 10. To convert glucose from mg/dl to mmol/l, multiply by 0.0555. To convert cholesterol, HDL-cholesterol and VLDL/LDL-cholesterol from mg/dl to mmol/l, multiply by 0.0259. To convert creatinine from mg/dl to ��mol/l, multiply by 88.4. To convert Ca, chloride and phosphate from mg/dl to g/l, multiply by 10.

Blood biochemistry was altered in a nutrient-specific manner, and strong hypotriglyceridaemia, hypocholesterolaemia and hypoproteinaemia with decreased plasma levels of creatinine were found in n-3 LC-PUFA fish, but also in Vit fish. Hypoproteinaemia was a sign of SAA and PL deficiency, whereas hypertriglyceridaemia and hypercholesterolaemia were characteristic features of P fish. Low plasma choline levels were found in SAA, Min and Vit fish.

Plasma electrolytes were highly refractory to dietary treatment in our experimental conditions, with the exception of calcium and phosphate in fish fed diets not supplemented with the vitamin premix and inorganic P, respectively. Likewise, enzyme activities of ALAT, ASAT and GLDH were not modified by dietary treatment, whereas ALP activity was significantly decreased in PL fish but increased in P and Min fish groups. Lysozyme activity was not altered by any dietary treatment. In contrast, RB was triggered in a consistent manner in n-3 LC-PUFA fish. Plasma antioxidant capacity was also increased by nutrient deficiencies, although this feature was especially evident in fish fed P, Min and Vit diets.

Regarding growth factors, circulating levels of GH highly reflected the impairment of growth performance and the highest plasma concentration was observed in n-3 LC-PUFA fish, followed by fish fed P and Vit diets. The opposite was found for circulating levels of IGF-I, and the lowest IGF-I concentration was found in n-3 LC-PUFA and P fish groups.

Histopathological traits

The histological examination of the liver and intestine showed different features, which are summarised in Table 5. Representative differential microphotographs are also provided in Figs. 13. The highest level of fat accumulation either in the liver or in the anterior intestine was observed in fish fed n-3 LC-PUFA, but without reaching steatosis. Accumulation of glycogen in the liver (revealed by PAS staining) was observed in fish fed SAA, P and Vit diets, but it was not extreme. No fat accumulation was observed in the posterior intestine in any of the groups. Goblet cell content and number varied with the diet, and a clear decrease in the number of neutral mucins (stained with PAS) was observed in the anterior intestine of fish fed n-3 LC-PUFA, PL, P and Min diets. In all fish groups, the number of PAS+ goblet cells was lower in the posterior intestine compared with the anterior intestine, and only PL and Vit fish had slight staining. The number of Giemsa-stained goblet cells was also decreased in P, Min and Vit fish. The staining of the epithelial layer of the posterior intestine was biphasic in the samples from all the experimental diets, except SAA and PL, in which it was homogeneous as in CTRL fish. The number of granulocytes in the submucosae in the anterior intestine and posterior intestine was not outstanding, except in Vit fish in the anterior intestine. Vit was the only diet in which intra-epithelial lymphocytes were in higher number than in the CTRL diets in both intestinal segments. Another remarkable feature was the high number of rodlet cells in the posterior intestine epithelium of PL, P, Min and Vit fish, with the highest level in Min fish.

Fig. 1. Histochemical staining of liver sections from gilthead sea bream fed the control diet (a, b), the n-3 long-chain PUFA diet (c, d) or the sulphur amino acids diet (e, f). Stainings: Giemsa (a, c, e), periodic acid–Schiff (b, d, f). Scale bars=20 µm.

Fig. 2. Histochemical staining of anterior intestine sections from gilthead sea bream fed the control diet (a, b), the n-3 long-chain PUFA diet (c, d) or the vitamin diet (e, f). Stainings: Giemsa (a, c, e), periodic acid–Schiff (b, d, f). Scale bars=20 µm.

Fig. 3. Histochemical staining of posterior intestine sections from gilthead sea bream fed the control diet (a, b), the n-3 long-chain PUFA diet (c, d) or the mineral diet (e, f). Stainings: Giemsa (a, c, e), periodic acid–Schiff (b, d, f). Scale bars=20 µm. Inset in (c) shows detail of rodlet cells in the epithelium; inset in (e) shows the abundant granulocytes in the submucosa.

Table 5 Summary of the histological features observed in the liver and anterior (AI) and posterior (PI) intestine of fish fed control (CTRL) and nutrient-deficient diets*

SAA, sulphur amino acids; n-3 LC-PUFA, n-3 long-chain PUFA; PL, phospholipids; Min, minerals; Vit, vitamins; PAS, periodic acid–Schiff.

* The intensity of the features was graded from absence (−) to the highest observed level (+++).

Discussion

Comprehensive approaches have been used to address the total or partial FM/FO replacement in a wide range of finfish including rainbow trout( Reference Panserat, Ducasse-Cabanot and Plagnes-Juan 21 ) and typically marine fish such as European sea bass( Reference Kaushik, Covès and Dutto 22 ) and gilthead sea bream. In particular, for gilthead sea bream, the long-term consequences of feeding low FM/FO feeds on growth performance and endocrine status( Reference Benedito-Palos, Navarro and Sitjà-Bobadilla 7 , Reference Gómez-Requeni, Mingarro and Calduch-Giner 23 , Reference Benedito-Palos, Saera-Vila and Calduch-Giner 24 ), health and welfare( Reference Saera-Vila, Calduch-Giner and Prunet 17 , Reference Estensoro, Benedito-Palos and Palenzuela 25 Reference Pérez-Sánchez, Estensoro and Redondo 30 ), fish quality( Reference Ballester-Lozano, Benedito-Palos and Navarro 13 , Reference Ballester-Lozano, Benedito-Palos and Mingarro 31 , Reference Benedito-Palos, Bermejo-Nogales and Karampatos 32 ) and food safety( Reference Nácher-Mestre, Serrano and Benedito-Palos 33 , Reference Nácher-Mestre, Serrano and Benedito-Palos 34 ) have been considered in a highly integrated manner. However, knowledge on the specific effects and consequences of a given nutrient or a group of nutrients is mostly lacking. Thus, this is one of the first studies analysing at the same time the effects of dietary deletion or reduction of six different nutrients recognised as essential to fish( 10 ), which contributes to fill the gaps in the diagnosis of the nutritional fish condition under standardised rearing conditions. Of note, we did not have the same degree of deficiency for the six different nutrients tested. Indeed, EPA and DHA contents were reduced to trace levels in the n-3 LC-PUFA diet with the total replacement of FO by VO. Likewise, the main source of P in the CTRL diet was the added calcium phosphate, but it was more difficult to induce a severe Met deficiency while preserving the supply of other essential amino acids.

Adipose tissue (AT) is now recognised as an important target tissue for the diagnosis and treatment of most lipid metabolic disorders arising from an excessive lipid influx( Reference Unger, Clark and Scherer 35 ). Clinically, lipotoxicity not only appears with fattening, but also with hypoxia, blockage of glucocorticoid-sensitive pathways and the acquisition and maintenance of inflammatory phenotypes( Reference Nawrocki and Scherer 36 Reference Maury and Brichard 38 ). In humans as well as in other animal models, FO and n-3 LC-PUFA of FO are able to reverse these clinical symptoms, decreasing lipolysis and alleviating the inflammatory condition of AT, which in turn reduces the production of lipolytic cytokines, the release of free FA and thereby the risk of hepatic steatosis( Reference Puglisi, Hasty and Saraswathi 39 ). This liver syndrome is the result of a massive synthesis and/or deposition of TG in the form of lipid vacuoles, and it is commonly observed in many fish species challenged with xenobiotics and unbalanced diets( Reference Wolf and Wolfe 40 ). This metabolic derangement is often accompanied by the displacement of the nucleus of hepatocytes and even pyknosis. Relatively little is known about the ultimate mechanism, although the dietary protein source and protein/energy ratio have a major effect on the regulation of lipid metabolism in European sea bass( Reference Dias, Alvarez and Arzel 41 , Reference Dias, Alvarez and Diez 42 ). The replacement of FM and FO with plant protein and oil sources also has a number of effects on the regulation of intermediary metabolism in trout( Reference Panserat, Hortopan and Plagnes-Juan 43 ) and Atlantic salmon( Reference Leaver, Villeneuve and Obach 44 ). In gilthead sea bream, tissue FA uptake( Reference Saera-Vila, Calduch-Giner and Gómez-Requeni 45 , Reference Saera-Vila, Calduch-Giner and Navarro 46 ) and mitochondrial respiration uncoupling( Reference Bermejo-Nogales, Calduch-Giner and Pérez-Sánchez 47 ) are highly affected by FM and FO replacement with plant ingredients. In addition, increases in cell size and lipolytic rates are characteristic features of isolated adipocytes from fish fed either plant proteins or VO( Reference Cruz-García, Sánchez-Gurmaches and Bouraoui 48 , Reference Albalat, Gómez-Requeni and Rojas 49 ), and lipoid liver degeneration is frequently observed as a metabolic disturbance in gilthead sea bream fed high plant ingredient-based feeds( Reference Benedito-Palos, Navarro and Sitjà-Bobadilla 7 , Reference Gómez-Requeni, Mingarro and Calduch-Giner 23 , Reference Caballero, Izquierdo and Kjørsvik 50 ). Accordingly, the present study shows that the total replacement of FO by VO in FM-free diets (n-3 LC-PUFA diet) caused a slight reduction of MSI related to a lipodystrophic phenotype with clinical signs of hypolipidaemia and hepatomegaly (high HSI). The loss of mesenteric AT mass, together with low plasma levels of choline and calcium, was even higher in the fish fed diets with reduced vitamin supply. However, abnormal liver lipid deposition rates were not found by light microscopy in this group of fish. Therefore, the absence or modification of AT mass cannot be used as the only criterion for the risk assessment of liver steatosis.

The main signs of P deficiency in fish nutritional studies are poor bone mineralisation and bad growth performance( Reference Antony Jesu Prabhu, Schrama and Kaushik 9 , Reference Lall and Lewis-McCrea 51 ), which were correlated in the present study with low plasma phosphate concentrations and high plasma activities of ALP in the absence of apparent skeletal deformities. ALP activity is a well-known bone turnover biomarker( Reference Fjelldal, Lock and Hansen 52 ), but here we also observed an increased plasma ALP activity with the reduced dietary supply of trace minerals (iron, magnesium, zinc, copper and selenium) in Min fish. However, the increase in MSI in combination with hyperlipidaemia allows differentiating the effects of deficiency in P from those due to deficiencies in trace minerals. In this sense, it must be noted than an important role of P in the regulation of lipid metabolism has also been reported in other fish species( Reference Albrektsen, Hope and Aksnes 53 , Reference Vielma and Lall 54 ).

Hct and Hb values are general indicators of health, and these haematological parameters change in response to nutrient deficiencies, environmental conditions, growth status and anti-nutritional factors( Reference García-Garrido, Muñoz-Chapuli and Deandres 55 , Reference Lim and Lee 56 ). A high incidence of anaemia has been reported in yellowtail and parrot fish fed FM-free diets( Reference Takagi, Murata and Goto 57 , Reference Maita, Aoki and Yamagata 58 ). This has been attributed, at least in part, to taurine deficiency, and thus its supplementation seems to be required when these fish are fed low levels of taurine in FM- or plant protein-based diets( Reference Takagi, Murata and Goto 59 ). In our study, all the diets were supplemented with taurine (0·3 %), and signs of anaemia, such as low erythrocytes, Hct and Hb values, were found only in fish fed the n-3 LC-PUFA diet, which also showed an increased erythrocyte osmotic fragility, as reported in rats with deficiencies in LC-PUFA( Reference Ehrström, Harmsringdahl and Alling 60 ). Importantly, this clinical sign was not found in the PL fish group, and therefore contributes to better define the sometimes-overlapping signs of nutrient deficiencies in EFA and PL.

Previous studies in gilthead sea bream have already highlighted increased RB of blood leucocytes in fish fed diets with a high replacement of FO by VO( Reference Saera-Vila, Calduch-Giner and Prunet 17 , Reference Estensoro, Benedito-Palos and Palenzuela 25 ). This has been confirmed in our study, and importantly RB turned out to be a specific criterion for the diagnosis of n-3 LC-PUFA deficiency, as it was notably increased in this group and not with PL or other nutrient-induced deficiencies. Likewise, a close talk between PL and bone metabolism exists, and the decrease in plasma ALP activity is becoming an easy and highly valuable marker of PL deficiencies in gilthead sea bream. Of note, a high PL supply is required during early life stages in fish to improve survival rates and to decrease the incidence of skeletal deformities( Reference Tocher, Bendiksen and Campbell 61 ), but PL requirements in juvenile and adult fish are still controversial( Reference Niu, Liu and Tian 62 , Reference Daprà, Geurden and Corraze 63 ).

Plasma transaminases and GLDH are commonly used in clinical chemistry as markers of tissue damage( Reference Ozer, Ratner and Shaw 64 ), but the current results indicate that they are poorly informative of nutritionally mediated metabolic derangements in gilthead sea bream. In contrast, overall plasma total antioxidant activity was increased in parallel to the reduced growth performance, indicative of a reduced aerobic metabolism and, therefore, of a reduced production of reactive oxygen species. Strong support for this comes from inbreeding selection of rat strains, which demonstrates that most stressful and oxidative risk factors correlate with the low expression of genes required for mitochondrial biogenesis and oxidative phosphorylation( Reference Wisløff, Najjar and Ellingsen 65 ). Experimental evidence also indicates that plasma antioxidant capacity is increased by hypoxia exposure, probably due to the concurrent decrease of basal metabolism, mitochondrial respiration uncoupling and oxidative phosphorylation( Reference Bermejo-Nogales, Calduch-Giner and Pérez-Sánchez 66 ). However, the magnitude and even the direction of the change is poorly predictable when comparisons are made between this and previous feeding trials with practical diets containing FM( Reference Saera-Vila, Calduch-Giner and Prunet 17 ).

Most growth regulatory events in fish are mediated at the hormonal level by the GH/IGF axis, keeping pituitary GH secretion and hepatic/extra-hepatic IGF production under control( Reference Company, Astola and Pendón 67 Reference Reindl and Sheridan 69 ). Therefore, circulating GH and IGF-I are one of the most important endocrine determinants of growth in a vast array of stress and nutritional disorders arising from crowding and handling stress( Reference Saera-Vila, Benedito-Palos and Sitjà-Bobadilla 70 ), changes in ration size( Reference Pérez-Sánchez, Calduch-Giner and Mingarro 68 , Reference Pérez-Sánchez, Martí-Palanca and Kaushik 71 ), dietary protein/energy ratio( Reference Martí-Palanca, Martínez-Barbera and Pendón 72 , Reference Company, Calduch-Giner and Kaushik 73 ) and dietary protein and lipid sources( Reference Gómez-Requeni, Mingarro and Calduch-Giner 23 , Reference Benedito-Palos, Saera-Vila and Calduch-Giner 24 , Reference Gómez-Requeni, Mingarro and Kirchner 74 ). This notion was also found here, and importantly a close positive correlation between growth rates and circulating levels of IGF-I was evidenced, regardless of the nutrient deficiency. As expected, an opposite trend was found for GH and growth rates, which would reflect a lowered negative feedback inhibition of IGFs upon pituitary GH release as a result of a transcriptional defect in the signal transduction of GH receptors. This metabolic feature leads to liver GH resistance and reduced hepatic IGF production, in spite of increased plasma levels of GH, as it has been stated previously in a wide range of fish species, including gilthead sea bream( Reference Pérez-Sánchez, Martí-Palanca and Kaushik 71 , Reference Beckman, Shimizu and Gadberry 75 Reference Wilkinson, Porter and Woolcott 77 ).

Histological traits also gave interesting information on the possible pathological outcome of nutrient deficiencies. The highest accumulation of hepatic lipids was found in gilthead sea bream fed the n-3 LC-PUFA-deficient diet, followed by SAA, P and Min. However, lipid accumulation did not reach the highest score of steatosis observed by us with other dietary interventions( Reference Benedito-Palos, Navarro and Sitjà-Bobadilla 7 , Reference Sitjà-Bobadilla, Peña-Llopis and Gómez-Requeni 15 ), or by other authors using diets with an excess of dietary lipids( Reference Caballero, López-Calero and Socorro 78 ), EFA deficiencies( Reference Montero, Robaina and Socorro 79 ) and VO( Reference Caballero, Izquierdo and Kjørsvik 50 , Reference Wassef, Wahby and Sakr 80 , Reference Alexis 81 ). Glycogen accumulation was also high in the liver of SAA, P and Vit fish, although the observed glycogen deposition did not reach an extreme condition. Massive accumulation of supranuclear lipid droplets in the intestinal epithelial layer is also considered a sign of inadequate/unbalanced diets due to a reduced metabolisation of absorbed lipids, either because they are not needed or because they are absorbed in a higher amount than needed. This accumulation was not observed in the posterior intestine for any diet, but it was a clear feature in the anterior intestine after feeding n-3 LC-PUFA-, PL- and Vit-deficient diets. This epithelial accumulation stands as an early marker of deregulated lipid metabolism compared with lipid accumulation in the liver, as it was visible in PL and Vit fed fish, in which lipoid liver degeneration was not found.

Another histological feature with clear differences among fish was the number of goblet cells and their staining characteristics. In this sense, it is noteworthy that the n-3 LC-PUFA diet induced a strong reduction in the number of goblet cells, and the remaining goblet cells were not stained either by Giemsa or by PAS, indicating that mucin content was not neutral or acidic. Neutral mucins were also absent in the anterior intestine as a result feeding the different diets, except SAA and Vit, and in the posterior intestine, except PL and Vit. Previous gilthead sea bream studies have indicated that other models of nutritional- and parasite-induced enteritis also invoke modifications in goblet cell type and number( Reference Estensoro, Redondo and Salesa 26 , Reference Baeza-Ariño, Martínez-Llorens and Nogales-Mérida 82 ). In fact, the 66 % replacement of FO by VO in plant protein-based diets produced a significant decrease in GC with neutral and acidic mucins in the anterior intestine and medium intestine as well as in those with carboxylic mucins and sialic acid in the medium intestine, but no significant changes in the posterior intestine. In European sea bass fed mannan oligosaccharides, the number of goblet cells secreting acidic mucins was increased( Reference Torrecillas, Makol and Caballero 83 ). In yellow perch fed wheat–gluten–protein-based diets, even supplemented with free lysine, the number of goblet cells was also decreased( Reference Ostaszewska, Dabrowski and Kamaszewski 84 ). In contrast, Atlantic salmon( Reference Bakke-McKellep, Press and Baeverfjord 85 ), Atlantic cod( Reference Olsen, Hansen and Rosenlund 86 ) and carp( Reference Urán, Goncalves and Taverne-Thiele 87 ) fed high plant protein-based diets presented goblet cells hypertrophy and hyperplasia.

Rodlet cells are exclusive of teleost epithelial layers and represent a cell type whose function has not yet definitively been established, although considered to be closely linked to the immune system and osmoregulation( Reference Reite 88 ). Many studies consistently report an association between rodlet cells proliferation/hyperplasia and the presence of a variety of parasites, chemicals and environmental stressors( Reference Manera and Dezfuli 89 , Reference Shimada Borges, Salimbeni Vivai and Branco 90 ) and they even have been proposed as biomarkers( Reference Manera and Dezfuli 89 ). However, there is no previous report on the relationship with the diet. In the present study, the number of rodlet cells was increased only in the posterior intestine, notably in fish fed Min diet. The increased presence of rodlet cells could, therefore, be interpreted either as a sign of inflammation or as a sign of osmoregulatory imbalance and reinforce the idea that cell osmoregulation is dependent on trace minerals rather than P uptake. The higher presence of rodlet cells was coincident with other inflammatory markers (intra-epithelial lymphocytes and submucosal granulocytes) in fish fed Min, PL and Vit diets either in the anterior intestine or in the posterior intestine. In any case, the observed cellular inflammation was mild and by no means comparable with that caused by other dietary interventions in Atlantic salmon( Reference Baeverfjord and Krogdahl 91 ), common carp( Reference Urán, Goncalves and Taverne-Thiele 87 ) or gilthead sea bream( Reference Sitjà-Bobadilla, Peña-Llopis and Gómez-Requeni 15 ).

In summary, clinical blood biochemistry and tissue histopathology have been proved highly informative to assess the nutritional condition of farmed gilthead sea bream. The final diagnosis outcome might require confirmation by more specific assays, but the generated information is by itself useful for the overall assessment of fish performance and metabolic condition when the measured parameters are referred to a CTRL group or historical data for a given fish strain, life stage and/or rearing condition. All this information is summarised in online Supplementary Table S1 as a set of clinical signs for a given nutritional deficiency in gilthead sea bream. To establish the normal range of variation of these parameters as a function of season and growth performance, the data from this study were combined with those derived from other past and on-going feeding trials with practical diets. The rationale for this is to cover a wide range of variation for marine and plant ingredients without apparent detrimental effects on fish performance through the production cycle. The reference values for these studied biomarkers are shown in online Supplementary Table S2, and will be periodically updated on the basis of the data produced within the ARRAINA project.

Acknowledgements

The authors are grateful to M. A. González, P. Cabrera and I. Vicente for their excellent technical assistance in fish rearing and fish blood and tissue sampling.

This study was funded by the European Union (ARRAINA, FP7-KBBE-2011-5-288925, Advanced Research Initiatives for Nutrition and Aquaculture) projects. Additional funding was obtained from the Spanish MINECO (MI2-Fish, AGL2013-48560) and from Generalitat Valenciana (PROMETEO FASE II-2014/085). G. F. B.-L. was a recipient of a Spanish PhD fellowship from the Diputación Provincial de Castellón.

G. F. B.-L. and L. B.-P. performed blood analyses; I. E. and A. S.-B. performed and supervised the histopathological analyses and RBC counts; S. K. formulated the experimental diets; J. P.-S. designed and coordinated the work, wrote the manuscript and took primary responsibility for its final content. All the authors read and approved the final manuscript.

The authors declare that there are no conflicts of interest perceived to bias the study.

Supplementary Material

To view supplementary material for this article, please visit http://dx.doi.org/10.1017/S0007114515002354

References

1. Knox, KMG, Reid, SWJ, Irwin, T, et al. (1998) Objective interpretation of bovine clinical biochemistry data: application of Bayes law to a database model. Prev Vet Med 33, 147158.CrossRefGoogle ScholarPubMed
2. Kerr, MG (2008) Veterinary Laboratory Medicine: Clinical Biochemistry and Haematology, 2nd ed. London: Blackwell Science Ltd.Google Scholar
3. Peres, H, Santos, S & Oliva-Teles, A (2014) Blood chemistry profile as indicator of nutritional status in European seabass (Dicentrarchus labrax). Fish Physiol Biochem 40, 13391347.CrossRefGoogle ScholarPubMed
4. Peres, H, Santos, S & Oliva-Teles, A (2013) Selected plasma biochemistry parameters in gilthead seabream (Sparus aurata) juveniles. J Appl Ichthyol 29, 630636.CrossRefGoogle Scholar
5. Hille, S (1982) A literature review of the blood chemistry of rainbow trout, Salmo gairdneri Rich. J Fish Biol 20, 535569.Google Scholar
6. Hrubec, TC & Smith, SA (2010) Hematology of fishes. In Schalm’s Veterinary Hematology, 6th ed., pp. 9941003 [J Douglas, editor]. Singapore: Blackwell Publishing Ltd.Google Scholar
7. Benedito-Palos, L, Navarro, JC, Sitjà-Bobadilla, A, et al. (2008) High levels of vegetable oils in plant protein-rich diets fed to gilthead sea bream (Sparus aurata L.): growth performance, muscle fatty acid profiles and histological alterations of target tissues. Br J Nutr 100, 9921003.CrossRefGoogle ScholarPubMed
8. Torstensen, BE & Tocher, DR (2011) The effects of fish oil replacement on lipid metabolism of fish. In Fish Oil Replacement and Alternative Lipid Sources in Aquaculture Feeds , pp. 405437 [W-K Ng, GM Turchini and DR Tocher, editors]. Boca Raton, FL: CRC Press.Google Scholar
9. Antony Jesu Prabhu, P, Schrama, JW & Kaushik, SJ (2014) Mineral requirements of fish: a systematic review. Rev Aquacult 6, 148.Google Scholar
10. National Research Council (2011) Nutrient Requirements of Fish and Shrimp. Washington, DC: National Academy of Sciences.Google Scholar
11. Folch, J, Lees, M & Sloane Stanley, GH (1957) A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem 226, 497509.Google Scholar
12. Christie, WW (1982) Lipid Analysis. Isolation, Separation, Identification and Structural Analysis of Lipids. Oxford: Pergamon Press.Google Scholar
13. Ballester-Lozano, GF, Benedito-Palos, L, Navarro, JC, et al. (2011) Prediction of fillet fatty acid composition of market-size gilthead sea bream (Sparus aurata) using a regression modelling approach. Aquaculture 319, 8188.CrossRefGoogle Scholar
14. Ellis, AE (1990) Lysozyme assays. Fish Immunol Tech Commun 1, 101103.Google Scholar
15. Sitjà-Bobadilla, A, Peña-Llopis, S, Gómez-Requeni, P, et al. (2005) Effect of fish meal replacement by plant protein sources on non-specific defence mechanisms and oxidative stress in gilthead sea bream (Sparus aurata). Aquaculture 249, 387400.Google Scholar
16. Nikoskelainen, S, Verho, S, Airas, K, et al. (2005) Adhesion and ingestion activities of fish phagocytes induced by bacterium Aeromonas salmonicida can be distinguished and directly measured from highly diluted whole blood of fish. Dev Comp Immunol 29, 525537.CrossRefGoogle ScholarPubMed
17. Saera-Vila, A, Calduch-Giner, JA, Prunet, P, et al. (2009) Dynamics of liver GH/IGF axis and selected stress markers in juvenile gilthead sea bream (Sparus aurata) exposed to acute confinement: differential stress response of growth hormone receptors. Comp Biochem Physiol 154A, 197203.Google Scholar
18. Martínez-Barber, JP, Pendón, C, Martí-Palanca, H, et al. (1995) The use of recombinant gilthead sea bream (Sparus aurata) growth hormone for radioiodination and standard preparation in radioimmunoassay. Comp Biochem Physiol 110A, 335340.Google Scholar
19. Shimizu, M, Swanson, P, Fukada, H, et al. (2000) Comparison of extraction methods and assay validation for salmon insulin-like growth factor-I using commercially available components. Gen Comp Endocrinol 119, 2636.Google Scholar
20. Vega-Rubín de Celis, S, Gómez-Requeni, P & Pérez-Sánchez, J (2004) Production and characterization of recombinantly derived peptides and antibodies for accurate determinations of somatolactin, growth hormone and insulin-like growth factor-I in European sea bass (Dicentrarchus labrax). Gen Comp Endocrinol 139, 266277.Google Scholar
21. Panserat, S, Ducasse-Cabanot, S, Plagnes-Juan, E, et al. (2008) Dietary fat level modifies the expression of hepatic genes in juvenile rainbow trout (Oncorhynchus mykiss) as revealed by microarray analysis. Aquaculture 275, 235241.Google Scholar
22. Kaushik, SJ, Covès, D, Dutto, G, et al. (2004) Almost total replacement of fish meal by plant protein sources in the diet of a marine teleost, the European seabass, Dicentrarchus labrax . Aquaculture 230, 391404.Google Scholar
23. Gómez-Requeni, P, Mingarro, M, Calduch-Giner, JA, et al. (2004) Protein growth performance, amino acid utilisation and somatotropic axis responsiveness to fish meal replacement by plant protein sources in gilthead sea bream (Sparus aurata). Aquaculture 232, 493510.Google Scholar
24. Benedito-Palos, L, Saera-Vila, A, Calduch-Giner, JA, et al. (2007) Combined replacement of fish meal and oil in practical diets for fast growing juveniles of gilthead sea bream (Sparus aurata L.): networking of systemic and local components of GH/IGF axis. Aquaculture 267, 199212.Google Scholar
25. Estensoro, I, Benedito-Palos, L, Palenzuela, O, et al. (2011) The nutritional background of the host alters the disease course in a fish-myxosporean system. Vet Parasitol 175, 141150.Google Scholar
26. Estensoro, I, Redondo, MJ, Salesa, B, et al. (2012) Effect of nutrition and Enteromyxum leei infection on gilthead sea bream Sparus aurata intestinal carbohydrate distribution. Dis Aquat Organ 100, 2942.Google Scholar
27. Ganga, R, Montero, D, Bell, JG, et al. (2011) Stress response in sea bream (Sparus aurata) held under crowded conditions and fed diets containing linseed and/or soybean oil. Aquaculture 311, 215223.CrossRefGoogle Scholar
28. Calduch-Giner, J, Sitjà-Bobadilla, A, Davey, G, et al. (2012) Dietary vegetable oils do not alter the intestine transcriptome of gilthead sea bream (Sparus aurata), but modulate the transcriptomic response to infection with Enteromyxum leei . BMC Genomics 13, 470.Google Scholar
29. Pérez-Sánchez, J, Borrel, M, Bermejo-Nogales, A, et al. (2013) Dietary oils mediate cortisol kinetics and the hepatic mRNA expression profile of stress-responsive genes in gilthead sea bream (Sparus aurata) exposed to crowding stress. Implications on energy homeostasis and stress susceptibility. Comp Biochem Physiol 8D, 123130.Google Scholar
30. Pérez-Sánchez, J, Estensoro, I, Redondo, MJ, et al. (2013) Mucins as diagnostic and prognostic biomarkers in a fish-parasite model: transcriptional and functional analysis. PLOS ONE 8, e65457.CrossRefGoogle Scholar
31. Ballester-Lozano, GF, Benedito-Palos, L, Mingarro, M, et al. (2014) Up-scaling validation of a dummy regression approach for predictive modelling the fillet fatty acid composition of cultured European sea bass (Dicentrarchus labrax). Aquacult Res (epublication ahead of print version 26 August 2014).Google Scholar
32. Benedito-Palos, L, Bermejo-Nogales, A, Karampatos, AI, et al. (2011) Modelling the predictable effects of dietary lipid sources on the fillet fatty acid composition of one-year-old gilthead sea bream (Sparus aurata L.). Food Chem 124, 538544.Google Scholar
33. Nácher-Mestre, J, Serrano, R, Benedito-Palos, L, et al. (2010) Bioaccumulation of polycyclic aromatic hydrocarbons in gilthead sea bream (Sparus aurata L.) exposed to long term feeding trials with different experimental diets. Arch Environ Contam Toxicol 59, 137146.Google Scholar
34. Nácher-Mestre, J, Serrano, R, Benedito-Palos, L, et al. (2009) Effects of fish oil replacement and re-feeding on the bioaccumulation of organochlorine compounds in gilthead sea bream (Sparus aurata L.) of market size. Chemosphere 76, 811817.Google Scholar
35. Unger, RH, Clark, GO, Scherer, PE, et al. (2010) Lipid homeostasis, lipotoxicity and the metabolic syndrome. Biochim Biophys Acta 1801, 209214.Google Scholar
36. Nawrocki, AR & Scherer, PE (2005) The adipocyte as a drug discovery target. Drug Discov Today 10, 12191230.CrossRefGoogle ScholarPubMed
37. Bourlier, V & Bouloumie, A (2009) Role of macrophage tissue infiltration in obesity and insulin resistance. Diabetes Metab 35, 251260.Google Scholar
38. Maury, E & Brichard, SM (2010) Adipokine dysregulation, adipose tissue inflammation and metabolic syndrome. Mol Cell Endocrinol 314, 116.Google Scholar
39. Puglisi, MJ, Hasty, AH & Saraswathi, V (2011) The role of adipose tissue in mediating the beneficial effects of dietary fish oil. J Nutr Biochem 22, 101108.Google Scholar
40. Wolf, JC & Wolfe, MJ (2005) A brief overview of nonneoplastic hepatic toxicity in fish. Toxicol Pathol 33, 7585.Google Scholar
41. Dias, J, Alvarez, MJ, Arzel, J, et al. (2005) Dietary protein source affects lipid metabolism in the European seabass (Dicentrarchus labrax). Comp Biochem Physiol 142A, 1931.Google Scholar
42. Dias, J, Alvarez, MJ, Diez, A, et al. (1998) Regulation of hepatic lipogenesis by dietary protein/energy in juvenile European seabass (Dicentrarchus labrax). Aquaculture 161, 169186.Google Scholar
43. Panserat, S, Hortopan, GA, Plagnes-Juan, E, et al. (2009) Differential gene expression after total replacement of dietary fish meal and fish oil by plant products in rainbow trout (Oncorhynchus mykiss) liver. Aquaculture 294, 123131.Google Scholar
44. Leaver, MJ, Villeneuve, LA, Obach, A et al. (2008) Functional genomics reveals increases in cholesterol biosynthetic genes and highly unsaturated fatty acid biosynthesis after dietary substitution of fish oil with vegetable oils in Atlantic salmon (Salmo salar). BMC Genomics 9, 299.Google Scholar
45. Saera-Vila, A, Calduch-Giner, JA, Gómez-Requeni, P, et al. (2005) Molecular characterization of gilthead sea bream (Sparus aurata) lipoprotein lipase. Transcriptional regulation by season and nutritional condition in skeletal muscle and fat storage tissues. Comp Biochem Physiol 142B, 224232.Google Scholar
46. Saera-Vila, A, Calduch-Giner, JA, Navarro, I, et al. (2007) Tumour necrosis factor (TNF) as a regulator of fat tissue mass in the Mediterranean gilthead sea bream (Sparus aurata L.). Comp Biochem Physiol 146B, 338345.Google Scholar
47. Bermejo-Nogales, A, Calduch-Giner, J & Pérez-Sánchez, J (2010) Gene expression survey of mitochondrial uncoupling proteins (UCP1/UCP3) in gilthead sea bream (Sparus aurata L.). J Comp Physiol 180B, 685694.Google Scholar
48. Cruz-García, L, Sánchez-Gurmaches, J, Bouraoui, L, et al. (2011) Changes in adipocyte cell size, gene expression of lipid metabolism markers, and lipolytic responses induced by dietary fish oil replacement in gilthead sea bream (Sparus aurata L.). Comp Biochem Physiol 158A, 391399.Google Scholar
49. Albalat, A, Gómez-Requeni, P, Rojas, P, et al. (2005) Nutritional and hormonal control of lipolysis in isolated gilthead seabream (Sparus aurata) adipocytes. Am J Physiol Regul Integr Comp Physiol 289, R259R265.Google Scholar
50. Caballero, MJ, Izquierdo, MS, Kjørsvik, E, et al. (2004) Histological alterations in the liver of sea bream, Sparus aurata L., caused by short- or long-term feeding with vegetable oils. Recovery of normal morphology after feeding fish oil as the sole lipid source. J Fish Dis 27, 531541.Google Scholar
51. Lall, SP & Lewis-McCrea, LM (2007) Role of nutrients in skeletal metabolism and pathology in fish – an overview. Aquaculture 267, 319.Google Scholar
52. Fjelldal, PJ, Lock, E-J, Hansen, T, et al. (2012) Continuous light induces bone resorption and affects vertebral morphology in Atlantic salmon (Salmo salar L.) fed a phosphorous deficient diet. Aquacult Nutr 18, 610619.Google Scholar
53. Albrektsen, S, Hope, B & Aksnes, A (2009) Phosphorous (P) deficiency due to low P availability in fishmeal produced from blue whiting (Micromesistius poutassou) in feed for under-yearling Atlantic salmon (Salmo salar) smolt. Aquaculture 296, 318328.Google Scholar
54. Vielma, J & Lall, SP (1998) Control of phosphorus homeostasis of Atlantic salmon (Salmo salar) in fresh water. Fish Physiol Biochem 19, 8393.Google Scholar
55. García-Garrido, L, Muñoz-Chapuli, R & Deandres, AV (1990) Serum cholesterol and triglyceride levels in Scyliorhinus canicula during sexual maturation. J Fish Biol 36, 499509.Google Scholar
56. Lim, S-J & Lee, K-J (2009) Partial replacement of fish meal by cottonseed meal and soybean meal with iron and phytase supplementation for parrot fish Oplegnathus fasciatus . Aquaculture 290, 283289.Google Scholar
57. Takagi, S, Murata, H, Goto, T, et al. (2008) Taurine is an essential nutrient for yellowtail Seriola quinqueradiata fed non-fish meal diets based on soy protein concentrate. Aquaculture 280, 198205.Google Scholar
58. Maita, M, Aoki, H, Yamagata, Y, et al. (1998) Plasma biochemistry and disease resistance in yellowtail fed a non-fish meal diet. Fish Pathol 33, 5963.Google Scholar
59. Takagi, S, Murata, H, Goto, T, et al. (2010) Necessity of dietary taurine supplementation for preventing green liver symptom and improving growth performance in yearling red sea bream Pagrus major fed nonfishmeal diets based on soy protein concentrate. Fisheries Sci 76, 119130.Google Scholar
60. Ehrström, M, Harmsringdahl, M & Alling, C (1981) Osmotic fragility and fluidity of erythrocyte membranes from rats raised on an essential fatty acid deficient diet. A spin label study. Biochim Biophys Acta 644, 175182.Google Scholar
61. Tocher, DR, Bendiksen, E, Campbell, PJ, et al. (2008) The role of phospholipids in nutrition and metabolism of teleost fish. Aquaculture 280, 2134.Google Scholar
62. Niu, J, Liu, YJ, Tian, LX, et al. (2008) The effect of different levels of dietary phospholipid on growth, survival and nutrient composition of early juvenile cobia (Rachycentron canadum). Aquacult Nutr 14, 249256.Google Scholar
63. Daprà, F, Geurden, I, Corraze, G, et al. (2011) Physiological and molecular responses to dietary phospholipids vary between fry and early juvenile stages of rainbow trout (Oncorhynchus mykiss). Aquaculture 319, 377384.CrossRefGoogle Scholar
64. Ozer, J, Ratner, M, Shaw, M, et al. (2008) The current state of serum biomarkers of hepatotoxicity. Toxicology 245, 194205.Google Scholar
65. Wisløff, U, Najjar, SM, Ellingsen, Ø, et al. (2005) Cardiovascular risk factors emerge after artificial selection for low aerobic capacity. Science 307, 418420.Google Scholar
66. Bermejo-Nogales, A, Calduch-Giner, JA & Pérez-Sánchez, J (2014) Tissue-specific gene expression and functional regulation of uncoupling protein 2 (UCP2) by hypoxia and nutrient availability in gilthead sea bream (Sparus aurata): implications on the physiological significance of UCP1-3 variants. Fish Physiol Biochem 40, 751762.Google Scholar
67. Company, R, Astola, A, Pendón, C et al. (2001) Somatotropic regulation of fish growth and adiposity: growth hormone (GH) and somatolactin (SL) relationship. Comp Biochem Physiol 130C, 435445.Google Scholar
68. Pérez-Sánchez, J, Calduch-Giner, JA, Mingarro, M, et al. (2002) Overview of fish growth hormone family. New insights in genomic organization and heterogeneity of growth hormone receptors. Fish Physiol Biochem 27, 243258.CrossRefGoogle Scholar
69. Reindl, KM & Sheridan, MA (2012) Peripheral regulation of the growth hormone-insulin-like growth factor system in fish and other vertebrates. Comp Biochem Physiol 163A, 231245.Google Scholar
70. Saera-Vila, A, Benedito-Palos, L, Sitjà-Bobadilla, A, et al. (2009) Assessment of the health and antioxidant trade-off in gilthead sea bream (Sparus aurata L.) fed alternative diets with low levels of contaminants. Aquaculture 296, 8795.Google Scholar
71. Pérez-Sánchez, J, Martí-Palanca, H & Kaushik, SJ (1995) Ration size and protein intake affect circulating growth hormone concentration, hepatic growth hormone binding and plasma insulin-like growth factor-I immunoreactivity in a marine teleost, the gilthead bream (Sparus aurata). J Nutr 125, 546552.Google Scholar
72. Martí-Palanca, H, Martínez-Barbera, JP, Pendón, C, et al. (1996) Growth hormone as a function of age and dietary protein: energy ratio in a marine teleost, the gilthead sea bream (Sparus aurata). Growth Regul 6, 253259.Google Scholar
73. Company, R, Calduch-Giner, JA, Kaushik, S, et al. (1999) Growth performance and adiposity in gilthead sea bream (Sparus aurata): risks and benefits of high energy diets. Aquaculture 171, 279292.CrossRefGoogle Scholar
74. Gómez-Requeni, P, Mingarro, M, Kirchner, S, et al. (2003) Effects of dietary amino acid profile on growth performance, key metabolic enzymes and somatotropic axis responsiveness of gilthead sea bream (Sparus aurata). Aquaculture 220, 749767.Google Scholar
75. Beckman, BR, Shimizu, M, Gadberry, BA, et al. (2004) Response of the somatotropic axis of juvenile coho salmon to alterations in plane of nutrition with an analysis of the relationships among growth rate and circulating IGF-I and 41 kDa IGFBP. Gen Comp Endocrinol 135, 334344.Google Scholar
76. Pierce, AL, Shimizu, M, Beckman, BR, et al. (2005) Time course of the GH/IGF axis response to fasting and increased ration in chinook salmon (Oncorhynchus tshawytscha). Gen Comp Endocrinol 140, 192202.Google Scholar
77. Wilkinson, RJ, Porter, M, Woolcott, H, et al. (2006) Effects of aquaculture related stressors and nutritional restriction on circulating growth factors (GH, IGF-I and IGF-II) in Atlantic salmon and rainbow trout. Comp Biochem Physiol 145A, 214224.Google Scholar
78. Caballero, MJ, López-Calero, G, Socorro, J, et al. (1999) Combined effect of lipid level and fish meal quality on liver histology of gilthead seabream (Sparus aurata). Aquaculture 179, 277290.Google Scholar
79. Montero, D, Robaina, LE, Socorro, J, et al. (2001) Alteration of liver and muscle fatty acid composition in gilthead seabream (Sparus aurata) juveniles held at high stocking density and fed an essential fatty acid deficient diet. Fish Physiol Biochem 24, 6372.Google Scholar
80. Wassef, EA, Wahby, OM & Sakr, EM (2007) Effect of dietary vegetable oils on health and liver histology of gilthead seabream (Sparus aurata) growers. Aquacult Res 38, 852861.Google Scholar
81. Alexis, MN (1997) Fish meal and fish oil replacers in Mediterranean marine fish diets. Cahiers Opt Med 22, 183204.Google Scholar
82. Baeza-Ariño, R, Martínez-Llorens, S, Nogales-Mérida, S, et al. (2014) Study of liver and gut alterations in sea bream, Sparus aurata L., fed a mixture of vegetable protein concentrates. Aquacult Res (epublication ahead of print version 30 June 2014).Google Scholar
83. Torrecillas, S, Makol, A, Caballero, MJ, et al. (2011) Improved feed utilization, intestinal mucus production and immune parameters in sea bass (Dicentrarchus labrax) fed mannan oligosaccharides (MOS). Aquacult Nutr 17, 223233.Google Scholar
84. Ostaszewska, T, Dabrowski, K, Kamaszewski, M, et al. (2013) The effect of dipeptide, Lys-Gly, supplemented diets on digestive tract histology in juvenile yellow perch (Perca flavescens). Aquacult Nutr 19, 100109.Google Scholar
85. Bakke-McKellep, AM, Press, CM, Baeverfjord, G, et al. (2000) Changes in immune and enzyme histochemical phenotypes of cells in the intestinal mucosa of Atlantic salmon, Salmo salar L., with soybean meal-induced enteritis. J Fish Dis 23, 115127.Google Scholar
86. Olsen, RE, Hansen, AC, Rosenlund, G, et al. (2007) Total replacement of fish meal with plant proteins in diets for Atlantic cod (Gadus morhua L.) II – health aspects. Aquaculture 272, 612624.Google Scholar
87. Urán, PA, Goncalves, AA, Taverne-Thiele, JJ, et al. (2008) Soybean meal induces intestinal inflammation in common carp (Cyprinus carpio L.). Fish Shellfish Immunol 25, 751760.Google Scholar
88. Reite, OB (2005) The rodlet cells of teleostean fish: their potential role in host defence in relation to the role of mast cells/eosinophilic granule cells. Fish Shellfish Immunol 19, 253267.Google Scholar
89. Manera, M & Dezfuli, BS (2004) Rodlet cells in teleosts: a new insight into their nature and functions. J Fish Biol 65, 597619.Google Scholar
90. Shimada Borges, JC, Salimbeni Vivai, ABB, Branco, PC, et al. (2013) Effects of trophic levels (chlorophyll and phosphorous content) in three different water bodies (urban lake, reservoir and aquaculture facility) on gill morphology of Nile tilapia (Oreochromis niloticus). J Appl Ichthyol 29, 573578.Google Scholar
91. Baeverfjord, G & Krogdahl, A (1996) Development and regression of soybean meal induced enteritis in Atlantic salmon, Salmo salar L, distal intestine: a comparison with the intestines of fasted fish. J Fish Dis 19, 375387.Google Scholar
Figure 0

Table 1 Ingredients and chemical composition of the experimental diets

Figure 1

Table 2 Fatty acid composition of the experimental diets (% total fatty acid methyl esters) (Mean values of two determinations)

Figure 2

Table 3 Effect of nutrient deficiencies on growth performance of gilthead sea bream fed to visual satiety from May to July (13 weeks) (Mean values with their standard errors)

Figure 3

Table 4 Effect of nutrient deficiencies on basic plasma biochemistry of sea bream fed to visual satiety from May to July (13 weeks) (Mean values with their standard errors of ten fish)

Figure 4

Fig. 1. Histochemical staining of liver sections from gilthead sea bream fed the control diet (a, b), the n-3 long-chain PUFA diet (c, d) or the sulphur amino acids diet (e, f). Stainings: Giemsa (a, c, e), periodic acid–Schiff (b, d, f). Scale bars=20 µm.

Figure 5

Fig. 2. Histochemical staining of anterior intestine sections from gilthead sea bream fed the control diet (a, b), the n-3 long-chain PUFA diet (c, d) or the vitamin diet (e, f). Stainings: Giemsa (a, c, e), periodic acid–Schiff (b, d, f). Scale bars=20 µm.

Figure 6

Fig. 3. Histochemical staining of posterior intestine sections from gilthead sea bream fed the control diet (a, b), the n-3 long-chain PUFA diet (c, d) or the mineral diet (e, f). Stainings: Giemsa (a, c, e), periodic acid–Schiff (b, d, f). Scale bars=20 µm. Inset in (c) shows detail of rodlet cells in the epithelium; inset in (e) shows the abundant granulocytes in the submucosa.

Figure 7

Table 5 Summary of the histological features observed in the liver and anterior (AI) and posterior (PI) intestine of fish fed control (CTRL) and nutrient-deficient diets*

Supplementary material: File

Ballester-Lozano supplementary material

Table S1

Download Ballester-Lozano supplementary material(File)
File 24.4 KB
Supplementary material: File

Ballester-Lozano supplementary material

Table S2

Download Ballester-Lozano supplementary material(File)
File 28.9 KB