#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Effects of the solubility of yeast cell wall preparations on their potential prebiotic properties in dogs


Authors: Stephanie de Souza Theodoro aff001;  Thaila Cristina Putarov aff001;  Caroline Tiemi aff001;  Lara Mantovani Volpe aff001;  Carlos Alberto Ferreira de Oliveira aff002;  Maria Beatriz de Abreu Glória aff003;  Aulus Cavalieri Carciofi aff001
Authors place of work: Veterinary Medicine and Surgery Department, College of Agrarian and Veterinarian Sciences (FCAV), São Paulo State University–UNESP, Jaboticabal, São Paulo, Brazil aff001;  Biorigin Brasil, Lençois Paulistas, São Paulo, Brazil aff002;  Pharmacy Faculty, Minas Gerais Federal University (UFMG), Belo Horizonte, Minas Gerais, Brazil aff003
Published in the journal: PLoS ONE 14(11)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0225659

Summary

Derivatives of yeast cell wall (YCW) have been studied for their potential prebiotic effects. Recently, new purified and soluble preparations have been developed in an attempt to increase their biological actions. Two YCW preparations, one conventional and another with higher solubility of the mannan oligosaccharide fraction, were evaluated on dogs. One food formulation was used, divided into the following treatments: CON–control, without yeast cell wall addition; YCW–addition of 0.3% of a conventional yeas cell wall extract; YCWs–addition of 0.3% of a yeast cell wall extract with high mannan oligosaccharide solubility. Twenty-four beagle dogs were used, eight per food, distributed on a block design. Blocks lasted 32 days, and TNF-a, IL-6, IL-10, ex vivo production of hydrogen peroxide and nitric oxide by peripheral neutrophils and monocytes, phagocytic index, and fecal IgA were evaluated at the beginning and end of each period. Additionally, nutrient digestibility, feces production and quality, and fermentation products were quantified. The results were evaluated by analysis of variance and compared using the Tukey test (P<0.05), using the basal immunological parameters as a covariate. The inclusion of YCWs reduced fat digestibility (P<0.05), increased the concentration of butyrate and putrescine, and reduced lactate in feces (P<0.05), showing that mannan oligosaccharide solubilization resulted in higher fermentation of this compound and altered the metabolism of the gut microbiota. Lower IL-6 on serum was verified for dogs fed the YCWs diet (P<0.05), suggesting a reduction in the inflammatory activity of dogs. Higher phagocytic index was verified for peripheral monocytes after the intake of the YCW food, suggesting better innate immunity. In conclusion, the solubilization of the mannooligosaccharide fraction alters its interaction with gut microbiota and biological actions in animals, although both yeast cell wall preparations exhibited prebiotic effects on dogs.

Keywords:

Yeast – Dogs – Diet – Cell walls – Gastrointestinal tract – Monocytes – Neutrophils – Fermentation

Introduction

The health of the gut is dependent on a dynamic interrelationship between the gut microbiota and gut nutrition [1,2], reflecting directly on the immunological status and general health of dogs [3,4]. It is postulated that the intestinal microbiota performs at least three main functions: protection, nutrition and metabolic control [5]. The microbiota acts as a barrier with important protective effect against pathogens; performs the fermentation of dietary nondigestible residues and endogenous substances, allowing the production of important nutrients for gut mucosa such as short-chain fatty acids; controls the proliferation and differentiation of intestinal epithelial cells; and contributes to immune system development and homeostasis [5].

Because intestinal microbes subsist on products resulting from the interaction between the host and its diet, food composition is one of the most important factors for gut microbiota maintenance, structure and function [1,6,7]. In this regard, yeast cell wall (YCW) may be an important energy source for intestinal microorganisms [8] and has been studied as a prebiotic candidate for dogs [9,10]. Mainly composed of carbohydrates and proteins, their main chemical constituents are mannose, glucose and N-acetylglucosamine (chitin) [11,12]. The YCW apparently meets the three essential criteria of a prebiotic [13], it is resistant to gastric acidity and hydrolysis by mammalian enzymes and to gastrointestinal absorption, is fermented by intestinal microbiota, and selectively stimulates the growth and/or activity of intestinal bacteria associated with health and wellbeing [1,14,15].

Among the possible mechanisms implicated for host health, prebiotics such as the YCW may promote short chain fatty acid (SCFA) production, colon pH regulation, and competition against pathogens for cell mucosa receptors [16]. Experimental data on animal studies have shown that the gut-associated lymphoid tissue (GALT) may be the primary target of the immunomodulatory effect of prebiotics [17,18], and the enterocytes are key intermediates that transmit signals from the intestinal lumen to the GALT [18]. Increase in serum lymphocyte concentration and decline in plasma neutrophils was reported in dogs fed YCW, indicative of an improvement in immunological status [19]. However, most publications on dogs only evaluated digestibility and fermentation products, and few evaluated the effects of the YCW on immunity. The SCFA generated after microbial fermentation of the YCW components may also modulate inflammation, since butyric acid may inhibit the production of the proinflammatory cytokines IL-2 and IFN-γ, and acetic and propionic acids may increase the production of the immunoregulatory cytokine IL-10 [20,21].

In recent years, specific strains of Saccharomyces cerevisiae and special techniques to separate and purify specific components of the cell wall structure have been developed. More purified than conventional YCW derivates, which consist of simple dried cell walls after the cellular content removal, these preparations have higher concentrations of soluble mannan oligosaccharides, smaller particle size and higher solubility in water, which are characteristics that may influence YCW exposure to gut microbiota and the host mucosa, potentially inducing different biological responses [10,22]. Based on these developments, the present study evaluated the effects of the incorporation in extruded diets of two preparations of Saccharomyces cerevisiae cell wall, differing in solubility in water of mannan oligosaccharides, on nutrient digestibility, microbial fermentation products in feces, and certain immunological parameters of adult dogs.

Experimental methods

The study was conducted in the Laboratory of Research in Nutrition and Nutritional Diseases of Dogs and Cats, College of Agrarian and Veterinarian Sciences, Sao Paulo State University (UNESP), Jaboticabal, SP, Brazil. All procedures with animals followed the ethical principles adopted by the Brazilian College of Animal Experimentation and were previously approved by the Ethics Committee on the Use of Animals (protocol number: 011937/17).

Test products

Two yeast derivates were used, obtained by the industrial purification of Saccharomyces cerevisiae cell wall (Biorigin, Lençóis Paulista, Sao Paulo, Brazil). After industrial purification, the yeasts culture was submitted to autolysis where intracellular enzymes are activated by appropriate processing conditions resulting in a partial degradation of the cell wall structures, followed by centrifugation and separation of the yeast extract from the yeast cell wall [23]. By this processing the standard purified Yeast Cell Wall (YCW) product was obtained with a water solubility index of approximately 20% (Table 1). Further, the purified yeast cell wall was submitted to a processing of chemical hydrolysis by acids [24,25], in order to partially solubilize the mannan-protein outer layer to obtain the soluble Yeast Cell Wall (YCWs) product, which presented 40% of water solubility index. Mainly mannan oligosaccharides were solubilized during the preparation of the extract, resulting in 2.1% soluble mannan oligosaccharides on the YCW and 22.2% soluble mannan oligosaccharides on the YCWs. The water solubility index was determined as previously described [26].

Tab. 1. Characteristics of the yeast cell wall derivates used on the study.
Characteristics of the yeast cell wall derivates used on the study.

Animals

Twenty-four adult Beagle dogs, males and females, with 3.5±0.91 years of age and weighing 11.95±1.12 kg were used. All animals belong to the kennel of the Laboratório de Pesquisa em Nutrição e Doenças Nutricionais de Cães e Gatos, FCAV/UNESP, Jaboticabal, Brazil. The mean body condition score of the dogs was 6.0±1.2, on a scale from 1 to 9 [27]. Prior to the study, dogs were submitted to physical, hematological, and serum biochemical evaluations by a veterinarian, and all were considered healthy.

Experimental design

The study included three experimental diets and was conducted in a randomized block design with two blocks of 12 dogs each and four dogs per diet in each block, totaling eight animals (repetitions) per diet (treatment). The blocking factor was time, due to available structure for research. Each block lasted 32 days and included testing the following: phagocytic activity of peripheral monocyte and neutrophils were evaluated on days 0, 15 and 30; cytokines in peripheral blood and the in vitro production of hydrogen peroxide and nitric oxide in cell culture were evaluated on days 0 and 30; IgA content in feces was evaluated prior to study (feces collected from days -2 to 0) and after 30 days of diet intake (days 30 to 32); total feces collection for digestibility measurement was performed from days 16 to 20; fresh feces collection to analyze fermentation products, pH and biogenic amines was conducted on days 23 to 25.

The amount of food offered was initially calculated considering the food metabolizable energy content, estimated by its chemical composition, and the individual energy requirement of laboratory dogs [28]. The daily amount was provided once a day (at 10 am). Offered and refused food was weighed, and the intake was recorded. Dogs were then weighed weekly, and the amount of food provided adjusted such that animals maintained a constant body weight throughout the study. Water was provided ad libitum. During the study dogs were housed in kennels measuring 1.5 m x 3.5 m with a solarium, and released daily in a collective playground for exercise and socialization.

Experimental diets

A single formulation based on corn grain, poultry byproduct meal, poultry fat and sugarcane fiber was used (Table 2), balanced for adult dogs according to the nutritional recommendations of the European Pet Food Industry Federation [29]. Sugarcane fiber was used due to its low fermentation [30], reducing interference with formation of fermentation products. The experimental diets were obtained by the addition of the different yeast cell wall extracts, added in replacement of corn (on an as-fed basis): CON—control diet, without inclusion of yeast cell wall extract; YCW—inclusion of 0.30% of YCW; YCWs—inclusion of 0.30% of YCWs.

Tab. 2. Ingredient and chemical composition of the food used on the study.
Ingredient and chemical composition of the food used on the study.

Dietary formulations were processed at the Extrusion Laboratory of the College of Agrarian and Veterinarian Sciences, Sao Paulo State University (UNESP), Jaboticabal, SP, Brazil. A single lot of raw materials was used for the three experimental diets. Ingredients were weighed and mixed before being ground in a hammer mill (Tigre, Moinhos Tigre, São Paulo, SP), fitted with a 0.8 mm sieve screen size, and extruded in a single-screw extruder (Model Mex-250, Manzoni Industria Ltda, Campinas, SP), with an average extrusion capacity of 250 kg/h. The temperature of the extruder preconditioner was kept higher than 85°C by direct steam injection. After extrusion, the kibbles were dried in a forced air dryer at 105°C for approximately 20 min and coated with poultry fat and liquid palatant.

Digestibility protocol, feces production and characteristics

This evaluation followed recommendations and procedures previously described [29]. Dogs were individually housed for 5 days in stainless steel metabolic cages, and each contained an apparatus to collect feces and urine separately. Food consumption was recorded daily, measuring the offered and refused amounts. Feces were quantitative collected at least twice a day for 120 h, weighed, and stored frozen at -15°C until analysis. After the end of the collection period, feces were thawed to room temperature and homogenized, compounding a single sample per dog, and then they were dried in a forced-air oven (320-SE, FANEM, São Paulo, Brazil) at 55°C for 72 hours. Predried feces and diets were ground in a knife type mill (MOD 340, ART LAB, São Paulo, Brazil) with a 1 mm sieve for laboratory analysis.

The gross energy (GE) content of diets and fecal samples was determined using a bomb calorimeter (IKA C2000 Basic, IKA-Werke GmbH & Co. KG, Staufen, Germany). Dry matter (DM) was determined by oven-drying the sample (method 934.01), ash was measured by muffle furnace incineration (method 942.05), crude protein was estimated using a LECO nitrogen/protein determination (FP-528, LECO Corporation, Saint Joseph, USA; method 990.03), total fat was assessed using the acid-hydrolyzed fat assay (method 954.02), and organic matter (OM) was calculated as DM minus ash. All samples were analyzed in duplicate, and the analyses were repeated when the variation between replicates was greater than 5%.

Fecal score was determined using the following system [31]: 0 = watery liquid that can be poured; 1 = soft, unformed; 2 = soft, malformed stool that assumes the shape of its container; 3 = soft, formed, and moist stool that retains its shape; 4 = well-formed and consistent stool that does not adhere to the floor; and 5 = hard, dry pellets, which are small and hard masses.

Fecal pH and fermentation products

For this evaluation, from days 23 to 25 for each block fecal samples were collected immediately after elimination for three consecutive days. Fecal pH was determined for 2 g of fresh feces mixed with 6 mL of ultrapure water, using a pH meter (model DM20; Digicrom Analitica LTDA, São Paulo, Brazil). Approximately 10 g of fresh feces was homogenized and mixed with 30 mL of a 16% (vol/vol) formic acid solution and precipitated to determine the volatile fatty acids (VFA). Next, the mixture was centrifuged (5810R; Eppendorf, Hamburg, Germany) 3 times at 4,500 x g for 15 min at 4°C. The supernatant was retained, and the pellet was discarded. The short-chain fatty acids (SCFA) and branched-chain fatty acids (BCFA) of the supernatant were determined by gas chromatography (model 9001; Finnigan Corporation, San Jose, CA) as previously described [32]. Lactic acid was measured by mixing 3 g of feces with 9 mL of distilled water. This mixture was centrifuged 3 times at 4,500 x g at 4°C for 15 min. The supernatant was obtained, and the pellet was discarded. The analysis of lactic acid was performed by spectrophotometry (Spectrophotometer Quick-Lab; Drake Eletronica e Comércio, São José do Rio Preto, São Paulo, Brazil) [33]; samples were quantified by comparing them with a standard curve for lactic acid. The concentration of ammonia was assessed in the same extracts prepared for the VFA. The extracts were thawed at room temperature, and 2 mL of each extract was diluted into 13 mL of distilled water and submitted to distillation in a nitrogen system (Tecnal TE-036/1; Tecnal Equipamentos Científicos, Piracicaba, São Paulo, Brazil).

To determine concentrations of biogenic amines in feces, five grams of fresh feces was homogenized and added to 7 mL of a 5% trichloroacetic acid solution and then mixed for 3 min by vortex and centrifuged at 10,000 x g for 20 min at 4°C (5810R; Eppendorf, Hamburg, Germany) [34]. The supernatant was filtered with qualitative filter paper, and the residue was extracted twice using 7 and 6 mL of a 5% trichloroacetic acid solution, separately. Then, the supernatants were filtered and pooled. The final volume obtained was recorded and frozen. Biogenic amine concentrations were determined in the supernatant by HPLC (HPLC model LC-10AD; Shimadzu Corporation, Kyoto, Japan).

Fecal Ig A

Fresh feces (immediately after elimination) was collected for three consecutive days before and 30 days after the intake of the experimental diets. For each period, fecal samples were pooled by dog, and fecal IgA was extracted using a saline solution [35]. Approximately 1 gram of feces was weighed and diluted in 10 mL of extraction buffer composed of 0.01 M phosphate-buffered saline (PBS) (pH 7.4), 0.5% Tween (Sigma-Aldrich, St Louis, MO, USA), and 0.05% sodium azide. After homogenization, the fecal suspensions were centrifuged at 1,500 x g for 20 min at 5°C. Then, 1 mL of the supernatant was transferred to a sterile microtube containing 20 μL of a protease-inhibitor cocktail (Sigma-Aldrich, St Louis, MO, USA). To remove the residues, samples were centrifuged at 15,000 x g for 15 min at 5°C, and the supernatants were kept in microtubes at -20°C until analysis.

The quantification of IgA was performed by an ELISA kit for canine IgA determination (Bethyl Laboratories, Montgomery, TX, USA). Optical density (OD) was read at 450 nm with a Microplate Reader (MRX TC Plus, Dynex Technology, Chantilly, Virginia, EUA). To calculate the IgA concentration, the OD of the samples was compared to the OD of a standard with a known concentration of IgA. The standard canine IgA sample was provided in the kit, and seven dilutions of the standard were made in order to develop a regression curve between OD and IgA amount. Samples were analyzed in duplicate, and the analysis was repeated when the variation between replicates was greater than 10%.

TNF-α, IL-6 and IL-10 on blood serum

For analyses of tumor necrosis factor alpha (TNF-α) and interleukins 6 (IL-6) and 10 (IL-10), on days zero and 30 blood samples (3 mL) were collected via jugular puncture and placed in tubes without anticoagulant. Afterwards, the samples were centrifuged at 3,500 x g for 10 min (5810R; Eppendorf, Hamburg, Germany), and the serum was stored frozen at -80°C until analysis. The dosage was estimated using a Luminex kit specific to dogs, according to the manufacturer's recommendations (MILLIPLEX MAP ELISA Canine Cytokine / Chemokine Magnetic Bead Panel—Immunology Multiplex Assay—Merck Millipore, St Charles-Missouri-USA).

Phagocytic activity

Phagocytic activity was measured on days 0, 15 and 30 using a commercial kit (pHrodo E. coli BioParticles, Molecular Probes Inc., Oregon, USA). Blood samples were collected by jugular puncture and placed in heparinized tubes. Then, 100 μL of each sample was incubated with 20 μL of pHrodo E. coli BioParticles, a reagent provided by the commercial kit. For each blood sample two tubes were prepared with the bioparticles; one was placed on ice, and the other kept in a water bath at 37 ºC for 15 min. Next, the incubated samples were lysed, followed by centrifugation and washing using the proper reagents as recommended by the manufacturer. Two negative control samples were run together on each collection day, both tubes with no bioparticles, but one placed on ice and the other kept at 37 ºC. Samples were analyzed using a flow cytometer (FACSCanto, Becton Dickinson Immunocytometry System, Mountain View, CA, USA), and the results were expressed as the percentage of fluorescence signal inside the desired population of neutrophils and monocytes. The target cell population was gated according to its volume and complexity [36].

Determination of hydrogen peroxide (H2O2) and nitric oxide (NO) production

Blood samples (6 mL) were collected with heparin via jugular puncture and added to 4.5 mL of Histopaque 1119 and 3 mL of Histopaque 1077 (Sigma Aldrich, St Louis, MO, EUA) in 15-mL conical centrifuge tubes. Tubes were centrifuged at 700 x g for 30 min at room temperature. After centrifugation, two distinct opaque layers separated, the mononuclear and granulocyte cells. Each layer was collected separately, transferred to a 50-mL conical centrifuge tube and washed at least twice with isotonic phosphate buffered saline by centrifugation at 360 x g for 10 min at room temperature. Erythrocyte lysis was conducted when necessary using 2 mL of ACK (Ammonium-Chloride-Potassium) solution (0.15 M ammonium chloride; 10 mM potassium bicarbonate; 0.1 mM EDTA) for a maximum of 2 min.

Cells were suspended in complete medium (RPMI 1640, Merck KGaA, Darmstadt, German), added to 40 mg/mL gentamicin and 10% fetal bovine serum, and the concentration was adjusted to 2 x 105 neutrophils or monocytes/mL. Then, suspensions were placed in 96-well flat plates (100 μL/well). Mononuclear cells were kept at 37°C in a humidified 5% CO2 incubator (Thermo Fisher Scientific, Waltham, MA, USA) for one hour for monocytes to adhere to the well surfaces, then supernatants were carefully discarded with a pipette and complete medium was added to each well. For monocytes, one plate was incubated for H2O2 production analysis and one for NO production analysis. For neutrophils, the supernatant from H2O2 production was used to conduct the NO analysis.

For H2O2 production, a total of 24 wells received 100 μL of sample; 12 of them were maintained as suspensions of nonstimulated cells, and the others 12 wells were stimulated with LPS (1 μg/well—E. coli Lipopolysaccharide, Sigma Aldrich, St. Louis, USA). Plates were maintained at 37°C in a humidified 5% CO2 incubator for 36 hours. The H2O2 production was measured as previously described [37,38]. Buffer solution (100 μL/well) consisting of 7.8 mL distilled water (dH2O), 0.8 mL of solution A (800 mL dH2O, 80 g NaCl, 2 g KCl, 2 g KH2PO4, 11.5 g Na2HPO4), 0.1 mL of solution B (100 mL dH2O, 1 g CaCl2), 0.1 mL of solution C (100 mL dH2O, 1 g MgCl2), 0.1 mL of phenol red (100 mL dH2O, 1 g phenol red), 0.1 mL of peroxidase (10 mg horseradish peroxidase, 2 mL PBS) and 1 mL of glucose (100 mL dH2O, 1 g glucose), was added to each well. Phorbol myristate acetate (PMA, 10 μL/well) was added to half of the nonstimulated wells and to half of LPS-stimulated wells and kept at 37°C in a humidified 5% CO2 incubator for one hour. Consequently, there were six replications for each cell condition: six wells for nonstimulated cells, six wells for nonstimulated + PMA, six wells for LPS-stimulated cells, and six wells for LPS-stimulated cells + PMA. After one-hour incubation, the reaction was stopped with 10 μL of 1 N NaOH. The absorbance was read in a microplate reader (iMarkMicroplate Absorbance Reader 168–1135, Bio-Rad, Hercules, California, USA) at 595 nm. The results were expressed in μM amounts of H2O2/2x105 cells. A hydrogen peroxide standard curve was constructed for each plate with a range of 0.25 to 16.00 nM of H2O2.

The NO production was assessed by the colorimetric method of the GRIESS reaction [39]. The analyses were conducted in six repetitions for nonstimulated cells and six repetitions for LPS-stimulated cells (1 μg/well E. coli Lipopolysaccharide, Sigma Aldrich, St. Louis, USA), totaling 12 wells per sample. One-hundred μL of GRIESS reagents diluted 1:1 (n-(1-naftil)-etil-enediamin diluted 0.1% in dH2O, 1% sulfonamide diluted in 5% H2PO4, Sigma Aldrich, St Louis, MO, USA) were added to the supernatant. The absorbance was read in a microplate reader (iMarkMicroplate Absorbance Reader 168–1135, Bio-Rad, Hercules, California, USA) at 540 nm. The results were expressed as μM amounts of NO/2x105 cells. An NO standard curve was constructed for each plate with a range of 0.78 to 100 μM of NO.

Statistical analysis

All variables were previously tested for normality or errors using the Cramer-von Misses test and for homoscedasticity using the Levene test. When necessary, logarithmic transformation (log x + 1) or lambda transformation was applied. For the immunological parameters, data were submitted to analysis of variance considering the effects of block, animal and diet. Differences among groups was detected at baseline, and for this reason the time 0 (baseline) was used as a covariate. When results of the F-test were significant, multiple comparisons of the means were made using Tukey’s test. Data obtained for nutrient digestibility, fecal parameters and fermentation products were submitted to analysis of variance and, when significant, compared by Tukey's test (P<0.05). Values of P<0.05 were considered significant, and P<0.10 as a trend. The analysis was conducted using the computer program R (version 3.3.3).

Results

Dogs showed proper food intake and maintained a constant body weight throughout the experimental period, with no episodes of food rejection, vomiting, or diarrhea. The food intake did not differ among diets (P>0.05). For the digestibility evaluation, DM intake was similar, resulting in similar nutrient intake by the animals (Table 3). The total tract apparent digestibility of nutrients was similar, except that fat digestibility was lower for dogs fed the YCWs food (P<0.05) than for CON. Feces production, DM, score and pH were also similar among treatments (P>0.05), as shown in (Table 4).

Tab. 3. Body weight (kg), nutrient intake (g/dog/day) and coefficients of total tract apparent digestibility (%) of nutrients of diets for dogs with the additions of different yeast cell wall preparations (mean and standard error of the mean).
Body weight (kg), nutrient intake (g/dog/day) and coefficients of total tract apparent digestibility (%) of nutrients of diets for dogs with the additions of different yeast cell wall preparations (mean and standard error of the mean).
Tab. 4. Feces production and characteristics of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Feces production and characteristics of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).

For fermentation products, higher concentrations of butyrate (approximately 25% more) and lower lactate (approximately 64% less) were verified in the feces of dogs fed the YCWs than in the other two foods (P<0.05), and there were no other detectable differences (Table 5).

Tab. 5. Volatile fatty acids (mMol/g of dry matter), ammonia and lactic acid concentration (mMol/kg of dry matter) on the feces of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Volatile fatty acids (mMol/g of dry matter), ammonia and lactic acid concentration (mMol/kg of dry matter) on the feces of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).

Only the biogenic amines putrescine, cadaverine, spermidine and phenylethylamine were detected in significant amounts in the dog feces (Table 6). Among them, putrescine was approximately 70% higher for dogs fed the YCWs than for those fed CON (P<0.05). Additionally, the feces of dogs fed the YCWs diet tentend to present higher spermidine concentration than CON (P = 0.096). The fecal concentration of IgA was similar between dogs fed the experimental foods, as presented in (Table 7).

Tab. 6. Biogenic amines concentration (mg/100 g of dry matter) on the feces of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Biogenic amines concentration (mg/100 g of dry matter) on the feces of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Tab. 7. Immunoglobulin A concentration (mg/g of dry matter) on the feces of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Immunoglobulin A concentration (mg/g of dry matter) on the feces of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).

Among the evaluated cytokines (Table 8), dogs fed the YCWs diet exhibited lower IL-6 serum concentration than did animals fed the CON diet (P<0.05), but similar values in comparison with dogs fed the YCW food. Dogs fed the YCW diet tended to present lower IL-6 and TNF-α values than animals fed the control food (P<0.10). No differences were detected for the other cytokines evaluated. No differences between diets were verified for H2O2 or NO production for monocytes or neutrophils, as shown in (Table 9).

Tab. 8. Serum cytokines concentrations (pg/mL) of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Serum cytokines concentrations (pg/mL) of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Tab. 9. Hydrogen peroxide (H2O2; μM of H2O2/2x105 cells) and nitrogen oxide (NO; μM of NO/2x105 cells) production in cell cultures of monocytes and neutrophils from the peripheral blood of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>; μM of H<sub>2</sub>O<sub>2</sub>/2x10<sup>5</sup> cells) and nitrogen oxide (NO; μM of NO/2x10<sup>5</sup> cells) production in cell cultures of monocytes and neutrophils from the peripheral blood of dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).

On day 30, the blood monocyte phagocytic index was 37% higher for dogs fed the YCW than the control diet (P<0.05), while dogs fed the YCWs food showed intermediate results. The neutrophil phagocytic activity of dogs did not differ among diets, having been elevated since the beginning of the study (Table 10).

Tab. 10. Monocyte and neutrophils phagocytic index (% of positive cells) from dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).
Monocyte and neutrophils phagocytic index (% of positive cells) from dogs fed diets with the addition of different yeast cell wall preparations (mean and standard error of the mean).

Discussion

Consistent with previous studies [40,41,42], the use of yeast cell wall was shown to be safe, as no changes in fecal quality or clinical condition of the animals was observed during the experimental period. The reduction of fat apparent digestibility after the consumption of the YCWs treatment can be attributed to the elevated solubility in water of yeast cell wall, perhaps behaving in the intestinal tract as a soluble and fermentable fiber that may interfere with fat absorption in dogs, as demonstrated by previous studies [43,44]. This effect on fat digestibility was not observed for the conventional YCW preparation, in agreement with previous publications on dogs [40,41]. The implications of the observed reduction in fat digestibility should be explored in future studies, including its use in low energy foods, but the magnitude of the fat digestibility reduction was low and its relevance to canine nutrition uncertain.

The experimental diets were formulated with sugarcane fiber, composed of approximately 45.8% cellulose, 28.1% hemicellulose and 9.3% lignina [45], an insoluble fiber source with very low fermentability [8,30] that was selected to not interfere with SCFA production. Under this condition, the intake of the more soluble yeast cell wall preparation, higher in soluble mannan oligosaccharides, changed the metabolism and fermentation products generated by the gut microbiota, as evidenced by higher fecal butyrate and putrescine and lower fecal lactate than in the other treatments. A higher production of SCFA, and especially of butyrate, is one of the outcomes expected from an effective prebiotic [13,46,47], suggesting an advantage for the YCWs. It is interesting that the traditional YCW, which is more insoluble, did not interfere with fermentation end-products formation; these data indicate that the solubility of the carbohydrate fractions of the yeast cell wall may be a key factor for product interaction with the gut microbiota.

In addition to its role as a source of energy for colonocytes, butyrate has been explored for its ability to directly affect cell growth and differentiation and to reduce cell inflammation [1,48,49]. In different cellular and animal models, butyrate reduced inflammation and improved the barrier function of the gut, reducing the production of proinflammatory cytokines [50,51,52]. Therefore, increased butyrate concentration is generally associated with improved health [53,54] and is one of the main objectives of prebiotic supplementations of diets.

Lactate is also produced as a result of carbohydrate fermentation by colon microbiota [55]; however, it does not exhibit a cumulative effect, as it is a substrate for several bacteria that utilize it, producing propionate and butyrate [56]. Thus, lactate concentrations may be interpreted considering the rates of production and consumption [56], which can explain the lower lactate and higher butyrate concentrations for dogs fed the YCWs diet. This altered the butyrate-to-lactate ratio, also exemplifying the impact of the YCWs on gut microbiota metabolism.

Amines are mainly formed through the decarboxylation of amino acids by the microorganisms of the gastrointestinal tract [57]. The fecal concentrations of amines observed in the present study are comparable to those previously reported in dogs [19,40,58,59], although the interpretation of amine concentrations in dog feces is difficult, due to the very limited information available regarding normal or desired levels [41]. In the present study, as the protein source (a possible source of amines) in diets was the same, the increased putrescine concentration (and the tendency of increased spermidine) may be explained by higher intestinal formation after the intake of the YCWs. Putrescine is produced by the decarboxylation of ornithine and arginine and, in turn, is progressively metabolized to spermidine, justifying the concomitant increase of both amines [60]. A previous study in our laboratory did not find an effect of yeast cell wall on fecal amine concentrations [41], reinforcing the lack of effect of the YCW diet in the present study, and suggesting that the soluble mannan oligosaccharides fraction of the YCWs in fact altered the fermentation profile of the gut microbiota.

Several favorable and harmful physiological processes involve the action of amines, especially the polyamines [61]. They are present in all living cells and are necessary for the normal development and repair of intestinal mucosa cells [62,63]. However, their activity has also been associated with the incidence of colorectal cancers [64], and high concentrations are related to inflammation, oxidative stress and genotoxicity [65]. Therefore, a significant reduction of polyamine concentrations in the intestinal lumen is not interesting, since the polyamine depletion (intracellular) directly affects the apoptosis of epithelial cells [40]; however, high amounts may also be undesirable. In a study with dogs in different age groups, higher putrescine, cadaverine, and spermine were observed in feces of older dogs compared to adult dogs, and higher spermidine was found in feces of dogs fed a diet based on soybean meal, which was linked by the authors to a higher IgA content in feces and better intestinal health [3].

IgA was evaluated in the present study, as it is an important marker of the mucosal immunity status [42,66], representing an essential factor in the protection against infectious agents, allergens and foreign proteins [3,67,68,69]. The main function of secretory IgA is to prevent bacteria and viruses from attaching and invading enterocytes [70,71]. The evaluation of this immunoglobulin is also of interest to clinicians to assess specific responses to antigens or in the diagnosis of IgA deficiency [66]. Studies with newborn animals have shown an effect of YCW on IgA secretion [72], differing from the present results. Perhaps the use of animals with mature immunity and the lack of immunological challenge in the present study may have interfered with the evaluation of the possible effect of the yeast cell wall preparations on the secretion of IgA, as also observed when the prebiotic resistant starch was evaluated in healthy adult dogs [73].

Cytokines was only evaluated after 30 days of diet intake, and due this was not possible to evaluate the kinetic of these compounds. The reduced IL-6 in serum of dogs fed the YCWs diet may also result, at least partially, from the higher butyrate formation in the intestine. Butyrate appears to be more potent than acetate or propionate in inducing immunomodulatory effects, as it affects the activity of histone deacetylases, which are responsible for decreasing the secretion of IL-12 and IL-6 cytokines by dendritic cells and allow dendritic cells to enhance mucosal regulatory T-cells [74]. However, a tendency for lower IL-6 and TNF-α in serum was also verified for dogs fed the YCW diet, which did not alter SCFA fecal concentration. These data corroborate findings of other researchers [75], which evaluated the action of a yeast cell wall fraction called "mannoprotein", added to a liquid diet for rats with Salmonella infection. The authors also found lower expression of TNF-α and IL-6 mRNA in the jejunum, ileum and colon tissues in the treatment groups and concluded that yeast cell wall derivates may lower the inflammatory response, protecting the intestinal tissue. Therefore, one may speculate that YCW may have a direct action on intestinal cells, reducing proinflammatory cytokines, in a mechanism independent of SCFA formation.

The increase in peripheral monocyte phagocytic activity in dogs fed the YCW diet was relevant, as this phenomenon is an important criterion for evaluating innate immunity [76,77]. Phagocytic cells act as the first line of defense against microorganisms [78], with monocytes being the key mediators of the early inflammatory response to infection. Considering the lack of changes in fermentation products in feces, it is possible to attribute this effect to a direct interaction of the mannan oligosaccharide or the b-1.3/1.6 glucan fractions of the YCW with the dendritic cells of the intestinal mucosa [79,80] which could demonstrate the ability of the YCW to modulate the immune system directly [49]. The ability of b-glucans to increase monocyte and neutrophil phagocytic percentages is well demonstrated for several species, including dogs [77,81,82]. However, a direct interaction of the mannan oligosaccharide fraction and the immune system has also been described [83,84] and cannot be excluded.

Peripheral blood mononuclear and polymorphonuclear (neutrophils) cells are traditionally used to evaluate in vitro responses of blood-derived immune cells to various antigens [85]. Although in the present study cells were stimulated by lipopolysaccharide and phorbol myristate acetate and substantially increased H2O2 and NO production, no diet effect was verified. Several studies regarding dietary intervention also found no effect on H2O2 or NO production [65,85]. The procedure is laborious, expensive and requires large volumes of blood to obtain the appropriate number of cells. In addition, cell sorting can stimulate cells and lead to loss of specific populations, leading to results that may not reflect the condition in vivo [85].

Some limitations of the present study may need to be considered. Only healthy dogs were used, and the period of dietary intake was not previously studied to determine if it was sufficient to express the complete effects of both yeast cell wall preparations. Consequently, possible differences between groups in the immunological system and microbiota metabolism could not be observed, and the long-term effects of the products are not known.

Conclusion

Under the conditions of the present research, positive immunomodulatory effects were verified for both yeast cell wall preparations. The addition of YCWs to an extruded diet changed intestinal microbiota metabolism, as verified by increased butyrate and putrescine and reduced lactate. YCWs in the diet also reduced inflammatory markers, which was verified by a reduction of serum IL-6 in dogs. The conventional YCW also tended to reduce IL-6 and TNF-α, and stimulated innate immunity, verified by an increase in peripheral monocyte phagocytic activity.

Supporting information

S1 Table [xlsx]
Full data set.


Zdroje

1. Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. 2017; 8(2): 172–184. doi: 10.1080/19490976.2017.1290756 28165863

2. Bibi S, Navarre DA, Sun X, Du M, Rasco B, Zhu M. Beneficial effect of potato consumption on gut microbiota and intestinal epithelial health. Am. J. Potato Res. 2019; https://doi.org/10.1007/s12230-018-09706-3.

3. Maria APJ, Ayane L, Putarov TC, Loureiro BA, Neto BP, Casagrande MF, et al. The effect of age and carbohydrate and protein sources on digestibility, fecal microbiota, fermentation products, fecal IgA, and immunological blood parameters in dogs. J Anim Sci. 2017; 95(6): 2452–2466. doi: 10.2527/jas.2016.1302 28727033

4. Coman MM, Verdenelli MC, Cecchini C, Belà B, Gramenzi A, Orpianesi C, et al. Probiotic characterization of Lactobacillus isolates from canine faeces. J Appl Microbiol. 2019; 126(4): 1245–1256. doi: 10.1111/jam.14197 30614169

5. Ballesteros-Pomar MD, Arnaiz EG. Papel de los prebióticos y los probióticos en la funcionalidad de la microbiota del paciente con nutrición enteral. Nutr Hosp. 2018; 35(2): 18–26. http://dx.doi.org/10.20960/nh.1956.

6. Delzenne N, Neyrinck A, Cani P. Gut microbiota and metabolic disorders: How prebiotic can work? British Journal of Nutrition. 2013; 109 (2): S81–S85. doi: 10.1017/S0007114512004047 23360884

7. Townsend GE, Han W, Schwalm ND, Raghavan V, Barry NA, Goodman AL, et al. Dietary sugar silences a colonization factor in a mammalian gut symbiont. PNAS. 2019; 116 (1): 233–238. doi: 10.1073/pnas.1813780115 30559205

8. Calabrò S, Carciofi AC, Musco N, Tudisco R, Gomes MOS, Cutrignelli MI. Fermentation characteristics of several carbohydrate sources for dog diets using the in vitro gas production technique. Italian Journal of Animal Science. 2013; 12(1): 21–27.

9. Gouveia EMMF, Silva IS, Nakazato G, Onselem VJV, Corrêa RAC, Araujo FR, et al. Action of phosphorylated mannanoligosaccharides on immune and hematological responses and fecal consistency of dogs experimentally infected with enteropathogenic Escherichia coli strains. Brazilian Journal of Microbiology. 2013; 44 (2): 499–504. doi: 10.1590/S1517-83822013000200027 24294246

10. Spring P, WenK C, Connolly A, Kiers A. A review of 733 published trials on Bio-Mos®, a mannan oligosaccharide, and Actigen®, a second generation mannose rich fraction, on farm and companion animals. Journal of Applied Animal Nutrition. 2015; 3 (7): 1–11. doi: 10.1017/jan.2015.6

11. Northcote DH; Horne RW. The chemical composition and structure of the yeast cell wall. Biochem J. 1952; 51(2): 232–236.2. doi: 10.1042/bj0510232 14944578

12. Aguilar-Uscanga B, François JM. A study of the yeast cell wall composition and structure in response to growth conditions and mode of cultivation. Letters in Applied Microbiology. 2003; 37: 268–274. doi: 10.1046/j.1472-765x.2003.01394.x 12904232

13. Gibson GR, Probert HM, Van Loo J, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev. 2004; 17: 259–75; doi: 10.1079/NRR200479 19079930. http://dx.doi.org/ 10.1079/NRR200479.

14. Fowler J, Kakani R, Haq A, Byrd JA, Bailey CA. Growth promoting effects of prebiotic yeast cell wall products in starter broilers under an immune stress and Clostridium perfringens challenge. The Journal of Applied Poultry Research. 2015; 24: 66–72. https://doi.org/10.3382/japr/pfv010.

15. Park SH, Lee SI, Ricke SC. Microbial Populations in Naked Neck Chicken Ceca Raised on Pasture Flock Fed with Commercial Yeast Cell Wall Prebiotics via an Illumina MiSeq Platform. PLOS ONE. 2016; 11(3): e0151944. doi: 10.1371/journal.pone.0151944 26992104

16. Park SH, Lee SI, Kim SA, Christensen K, Ricke SC. Comparison of antibiotic supplementation versus a yeast-based prebiotic on the cecal microbiome of commercial broilers. PLOS ONE. 2017; 12(8): e0182805. https://doi.org/10.1371/ journal.pone.0182805.

17. Schley PD, Field CJ. The immune-enhancing effects of dietary fibres and prebiotics. British Journal of Nutrition. 2002; 87 (2): S221–S230. doi: 10.1079/BJN/2002541

18. Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall R, Rowland I, et al. Prebiotic effects: metabolic and health benefits. British Journal of Nutrition. 2010; 104(S2): S1–S63. https://doi.org/10.1017/S0007114510003363.

19. Swanson KS, Grieshop CM, Flickinger EA, Healy H-P, Dawson KA, Merchen NR, et al. Effects of supplemental fructooligosaccharides plus mannan oligosaccharides on immune function and ileal and fecal microbial population in adult dogs. Archives of Animal Nutrition. 2002; 56: 309–318. doi: 10.1080/00039420214344 12462915

20. Cavaglieri CR, Nishiyama A, Fernandes LC, Curi R, Miles EA, Calder PC. Differential effects of short-chain fatty acids on proliferation and production of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sciences. 2003; 73: 1683–1690. doi: 10.1016/s0024-3205(03)00490-9 12875900

21. Kaisar MMM, Pelgrom LR, van der Ham AJ, Yazdanbakhsh M and Everts B. Butyrate Conditions Human Dendritic Cells to Prime Type 1 Regulatory T Cells via both Histone Deacetylase Inhibition and G Protein-Coupled Receptor 109A Signaling. Front. Immunol. 2017; 8:1429. doi: 10.3389/fimmu.2017.01429 29163504

22. Vetvicka V, Carlos Oliveira C. β-(1–3)(1–6)-D-glucans Modulate Immune Status and Blood Glucose Levels in Dogs. British Journal of Pharmaceutical Research. 2014; 4(8): 981–991.

23. Podpora B, Świderski F, Sadowska A, Rakowska R, Wasiak-Zys G. Spent brewer’s yeast extracts as a new component of functional food. Czech J. Food Sci., 2016; 34: 554–563.

24. Peat S, Whelan WJ, Edwards TE. Polysaccharides of baker’s yeast. Part IV. Mannan. Journal of the Chemical Society (Resumed). 1961; 29–34.

25. Ogawa K, Nishikori J, Ino T, Matsuda K. Chemical Structures of Oligosaccharides Obtained from Partial Acid Hydrolysates of Saccharomyces cerevisiae Mannan. Biosci. Biotech. Biochem., 1994; 58 (3): 560–562.

26. Anderson RA. Water absorption and solubility and amylograph characteristics of roll-cooked small grain products. Cereal Chern. 1981; 59(4): 265–269.

27. Laflamme DP. Development and validation of body condition score system for dogs. Canine Practices. 1997; 22: 10–15.


28. NRC—National Research Council. Nutrient requirements of dogs and cats. Washington, DC: National Academy Press, 2006.

29. FEDIAF—Fédération Européenne de L’industrie des Aliments pour Animaux Familiers. The European Pet Food Industry Federation. Nutritional Guidelines. 2017.

30. Loureiro BA, Sakomura NK, Vasconcellos RS, Sembenelli G, Gomes MOS. Insoluble fibres, satiety and food intake in cats fed kibble diets. J. Anim. Physiol. Anim. Nutr. (In press.) 2017; 101(5): 824–834. doi: 10.1111/jpn.12468 27080580

31. Carciofi AC, Takakura FS, De-Oliveira LD, Teshima E, Jeremias JT, Brunetto MA, et al. Effects of six carbohydrate sources on dog diet digestibility and postprandial glucose and insulin response. Journal of Animal Physiology and Animal Nutrition. 2008; 98: 326–336.

32. Erwin ES, Marco GJ, Emery EM. Volatile fatty acid analyses of blood and rumen fluid by gas chromatography. Journal of Dairy Science. 1961; 44: 1768–1771.

33. Pryce JD. A modification of the Barker-Summerson method for the determination of latic acid. The Analist. 1969; 94: 1121–1151.

34. Vale SR, Gloria MB. Determination of biogenic amines in cheese. J. AOAC Int. 1997; 80(5): 1006–1012. 9325578

35. Peters IR, Calvert EL, Hall EJ, Day MJ. Measurement of immunoglobulin concentrations in the feces of healthy dogs. Clin. Diagn. Lab. Immunol. 2004; 11(5): 841–848. doi: 10.1128/CDLI.11.5.841-848.2004 15358641

36. Neaga A, Lefor J, Lich KE, Liparoto SF, Xiao YQ. Development and validation of a flow cytometric method to evaluate phagocytosis of pHrodo™ BioParticles® by granulocytes in multiple species. Journal of Immunological Methods. 2013; 390(1–2): 9–17. doi: 10.1016/j.jim.2011.06.027 21767540

37. Pick E, Keisari Y. A simple colorimetric method for the measurement of hydrogen peroxide produced by cells in culture. Journal of Immunological Methods. 1980; 38: 161–170. doi: 10.1016/0022-1759(80)90340-3 6778929

38. Pick E, Mizel D. Rapid microassays for measurement of superoxide and hydrogen peroxide production by macrophages in culture using an automatic enzyme immunoassay reader. Journal of Immunological Methods. 1981; 46: 211–226. doi: 10.1016/0022-1759(81)90138-1 6273471

39. Grenn LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite, and [15N] nitrate in biological fluids. Analytical Biochemistry. 1982; 126: 131–138. doi: 10.1016/0003-2697(82)90118-x 7181105

40. Middelbos IS, Fastinger ND, Fahey GC Jr. Evaluation of fermentable oligosaccharides in diets fed to dogs in comparison to fiber standards. J. Anim. Sci. 2007; 85: 3033–3044. doi: 10.2527/jas.2007-0080 17686893

41. Gomes MOS, Beraldo MC, Putarov TC, Brunetto MA, Zaine L, Gloria MBA, et al. Old beagle dogs have lower faecal concentrations of some fermentation products and lower peripheral lymphocyte counts than young adult beagles. British Journal of Nutrition. 2011; 106: S187–S190. doi: 10.1017/S0007114511002960 22005424

42. Lin CY, Alexander C, Steelman AJ, Warzecha CM, Godoy MRC, Swanson KS. Effects of a Saccharomyces cerevisiae fermentation product on fecal characteristics, nutrient digestibility, fecal fermentative end-products, fecal microbial populations, immune function, and diet palatability in adult dogs. Journal of Animal Science. 2019. https://doi.org/10.1093/jas/skz064.

43. Diez M, Hornick JL, Baldwin P, Eenaeme CV, Istasse L. The influence of sugar-beet fiber, guar gum and inulin on nutrient digestibility, water consumption and plasma metabolites in healthy Beagle dogs. Res. Vet. Sci. 1998; 64: 91–96. doi: 10.1016/s0034-5288(98)90001-7 9625462

44. de-Oliveira LD, Takakura FS, Kienzle E, Brunetto MA, Teshima E, Pereira GT, et al. Fibre analysis and fibre digestibility in pet foods–a comparison of total dietary fibre, neutral and acid detergent fibre and crude fibre. Animal Physiology and Animal Nutrition. 2012; 96: 895–906. https://doi.org/10.1111/j.1439-0396.2011.01203.x

45. Monti M, Gibson M, Loureiro BA, Sá FC, Putarov TC, Villaverde C, et al. Influence of dietary fiber on macrostructure and processing traits of extruded dog foods. Anim. Feed Sci. Technol. 2016, 220: 93–102. doi: 10.1016/j.anifeedsci.2016.07.009

46. Teng P-Y, Kim WK. Review: Roles of Prebiotics in Intestinal Ecosystem of Broilers. Front. Vet. Sci. 2018; 5: 245. doi: 10.3389/fvets.2018.00245 30425993

47. Santos JPF, Aquino AAA, Glória MBA, Avila-CampoS MJ, Oba PM, Santos KM, et al. Effects of dietary yeast cell wall on faecal bacteria and fermentation products in adult cats. J Anim Physiol Anim Nutr. 2018; 102(4): 1–11. doi: 10.1111/jpn.12918 29761557

48. Rivière A, Selak M, Lantin D, Leroy F, De Vuyst L. Bifidobacteria and butyrate-producing colon bacteria: Importance and strategies for their stimulation in the human gut. Front. Microbiol. 2016; 7: 979. doi: 10.3389/fmicb.2016.00979 27446020

49. Chung WSF, Meijerink M, Zeuner B, Holck J, Louis P, Meyer A, et al. Prebiotic potential of pectin and pectic oligosaccharides to promote anti-inflammatory commensal bacteria in the human colon. FEMS Microbiology Ecology. 2017; 93(11): 1–9. https://doi.org/10.1093/femsec/fix127.

50. Inan MS, Rasoulpour RJ, Yin L, Hubbard K, Rosenberg DW, Giardina C. The luminal short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell line. Gastroenterology, 2000; 118(4): 724–734. doi: 10.1016/s0016-5085(00)70142-9 10734024

51. Yin L, Laevsky G, Giardina C. Butyrate Suppression of Colonocyte NF-κB Activation and Cellular Proteasome Activity. Journal of Biological Chemistry. 2001; 276(48): 44641–44646. doi: 10.1074/jbc.M105170200 11572859

52. Jiminez JA, Uwiera TC, Abbott DW, Uwiera RRE, Inglis GD. Butyrate Supplementation at High Concentrations Alters Enteric Bacterial Communities and Reduces Intestinal Inflammation in Mice Infected with Citrobacter rodentium. mSphere. 2017. 2(4): 1–21. https://doi.org/10.1128/mSphere.00243-17.

53. Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. 2014. Chapter Three–the role of short-chain fatty acids in health and disease. Adv. Immunol. 121: 91–119. doi: 10.1016/B978-0-12-800100-4.00003-9 24388214

54. Ríos-Covían D, Ruas-Madiedo P, Margolles A, Gueimonde M, Reyes-Gavilán CG, Salazar N. Intestinal short chain fatty acids and their link with diet and human health. Front. Microbiol. 2016; 7: 185. doi: 10.3389/fmicb.2016.00185 26925050

55. Fischer MM, Kessler AM, de Sá LR, Vasconcellos RS, Filho FO, Nogueira SP, et al. Fiber fermentability effects on energy and macronutrient digestibility, fecal traits, postprandial metabolite responses, and colon histology of overweight cats. J. Anim. Sci. 2012; 90(7): 2233–45. doi: 10.2527/jas.2011-4334 Epub 2012 Jan 13. 22247109

56. Moens F, Abbeele PV, Basit AW, Dodo C, Chatterjee R, Smith B, et al. A four-strain probiotic exerts positive immunomodulatory effects by enhancing colonic butyrate production in vitro. International Journal of Pharmaceutics. 2019. 555: 1–10. doi: 10.1016/j.ijpharm.2018.11.020 30445175

57. Hussein HS; Flickinger EA; Fahey GC Jr. Petfood Applications of Inulin and Oligofructose. Journal of Nutrition. 1999; 129:1454–1456.

58. Flickinger EA, Schreijen EMWC, Patil AR, Houssein HS, Grieshop CM, Merchen NR, et al. Nutrient digestibilities, microbial populations, and protein catabolites as affected by fructan supplementation of dog diets. Journal of Animal Science. 2003; 80: 2008–2018.

59. Propst EL, Flickinger EA, Bauer LL, Merchen NR, Fahey C Jr. A dose-response experiment evaluating the effects of oligofructose and inulin on nutrient digestibility, stool quality, and fecal protein catabolites in healthy adult dogs. J. Anim. Sci. 2003; 81: 3057–3066. doi: 10.2527/2003.81123057x 14677862

60. Hanfrey CC, Pearson BM, Hazeldine S, Lee J, Gaskin DJ, Woster PM, et al. Alternative spermidine biosynthetic route is critical for growth of Campylobac- ter jejuni and is the dominant polyamine pathway in human gut microbiota, J. Biol. Chem. 2011; 286: 43301–43312. doi: 10.1074/jbc.M111.307835 22025614

61. Kalac P. Health effects and occurrence of dietary polyamines: A review for the period 2005–mid 2013. Food Chemistry. 2014; 161: 27–39. doi: 10.1016/j.foodchem.2014.03.102 24837918

62. Wang JY, Johnson LR. Luminal polyamines stimulate repair of gastric mucosal stress ulcers. Gastrointestinal and Liver Physiology. 1990; 259(4): G584–G592. https://doi.org/10.1152/ajpgi.1990.259.4.G584

63. Loser C, Eisel A, Harms D, Folsch UR. Dietary polyamines are essential luminal growth factors for small intestinal and colonic mucosal growth and development. Gut. 1999; 44:12–16. doi: 10.1136/gut.44.1.12 9862820

64. Milovic V, Turchanowa L. Polyamines and colon cancer. Biochem. Soc. Trans. 2003; 31: 381–383. doi: 10.1042/bst0310381 12653643

65. Hoyles L, Swann J. Influence of the Human Gut Microbiome on the Metabolic Phenotype. The Handbook of Metabolic Phenotyping. Elsevier Inc.; 2019.

66. Norris CR, Gershwin LJ. Evaluation of systemic and secretory IgA concentrations and immunohistochemical stains for IgA-containing B cells in mucosal tissues of an Irish setter with selective IgA deficiency. J Am Anim Hosp Assoc. 2003; 39: 247–250. doi: 10.5326/0390247 12755197

67. Lycke N, Erlandsson L, Ekman L, Schon K, Leandreson T. Lack of J chain inhibits the transport of gut IgA and abrogates the development of intestinal antitoxic protection. J Immunol. 1999; 163: 913–919. 10395687

68. Wijburg OLC, Uren TK, Simpfendorfer K, Johansen F-E, Brandtzaeg P, Stugnell RA. Innate secretory antibodies protect against natural Salmonella typhimurium infection. J Exp Med. 2006; 203: 21–26. doi: 10.1084/jem.20052093 16390940

69. Zaine L, Ferreira C, Gomes MOS, Monti M, Tortola L, Vasconcellos RS, et al. Faecal IgA concentration is influenced by age in dogs. British Journal of Nutrition. 2011; 106: S183–S186. doi: 10.1017/S0007114511000559 22005423

70. Mayer L. Mucosal immunity and gastrointestinal antigen processing. J. Pediatr. Gastroenterol. Nutr. 2000; 30: S4–S12. doi: 10.1097/00005176-200001001-00002 10634293

71. Woof JM, Kerr MA. The function of immunoglobulin A in immunity. Journal of Pathology. 2006; 208: 270–282. doi: 10.1002/path.1877 16362985

72. Heinrichs AJ, Heinrichs BS, Jones CM. Fecal and saliva IgA secretion when feeding a concentrated mannan oligosaccharide to neonatal dairy calves. The Professional Animal Scientist. 2013; 29: 457–462.

73. Peixoto MC, Ribeira EM, Maria APJ, Loureiro BA, di Santo LG, Putarov TC, et al. Effect of resistant starch on the intestinal health of old dogs: fermentation products and histological features of the intestinal mucosa. J Anim Physiol Anim Nutr. 2018; 102: 111–121.

74. Frei R, Akdis M, O’Mahony L. Prebiotics, probiotics, synbiotics, and the immune system: experimental data and clinical evidence. Curr. Opin. Gastroenterol. 2015; 31: 153–158. doi: 10.1097/MOG.0000000000000151 25594887

75. Posadas SJ, Caz V, Caballero I, Cendejas E, Quilez I, Largo C, et al. Effects of mannoprotein E1 in liquid diet on inflammatory response and TLR5 expression in the gut of rats infected by Salmonella typhimurium. BMC Gastroenterology. 2010; 10: 58. doi: 10.1186/1471-230X-10-58 20529359

76. Song SK, Beck BR, Kim D, Park J, Jungjoon K, Kim HD, et al. Prebiotics as immunostimulants in aquaculture: A review. Fish & Shellfish Immunology. 2014; 40: 40–48.

77. Oliveira CAF, Vetvicka V, Zanuzzo FS. β-Glucan successfully stimulated the immune system in different jawed vertebrate species. Comparative Immunology, Microbiology and Infectious Diseases. 2019; 62: 1–6. doi: 10.1016/j.cimid.2018.11.006 30711038

78. Gourbeyre P, Denery S, Bodinier M. Probiotics, prebiotics, and synbiotics: impact on the gut immune system and allergic reactions. Journal of Leukocyte Biology. 2011; 89: 685–695. doi: 10.1189/jlb.1109753 21233408

79. Esteban MA, Rodriguez A, Mesguer J. Glucan receptor but not mannose receptor is involved in the phagocytosis of Saccharomyces cerevisiae by seabream (Sparus auratus L.) blood leucocytes. Fish Shellfish Immunol 2004; 16: 447–51. doi: 10.1016/j.fsi.2003.07.004 15123311

80. Abu-Elala N, Mohamed M, Mohamed M. Use of different Saccharomyces cerevisiae biotic forms as immune-modulator and growth promoter for Oreochromis niloticus challenged with some fish pathogens. International Journal of Veterinary Science and Medicine. 2013; 1: 21–29. https://doi.org/10.1016/j.ijvsm.2013.05.001

81. Vetvicka V, Vashishta A, Saraswat-ohri S, Vetvickova J. Immunological effects of yeast- and mushroom-derived beta-glucans. Journal of medicinal food. 2008; 11(4): 615–622. doi: 10.1089/jmf.2007.0588 19053851

82. Rodriguez-Estrada U, Satoh S, Haga Y, Fushimi H, Sweetman J. Effects of inactivated Enterococcus faecalis and mannan oligosaccharide and their combination on growth, immunity, and disease protection in rainbow trout. N. Am. J. Aquacult. 2013; 75: 416–428. doi: 10.1080/15222055.2013.799620

83. Torrecillas S, Makol A, Caballero M, Montero D, Ginés R, Sweetman J, et al. Improved feed utilization, intestinal mucus production and immune parameters in sea bass (Dicentrarchus labrax) fed mannan oligosaccharides (MOS). Aquaculture Nutrition. 2011; 17: 223–233. doi: 10.1111/j.1365-2095.2009.00730.x

84. Devi G, Harikrishnan R, Paray BA, Al-Sadoon MK, Hoseinifar SH, Balasundaram C. Comparative immunostimulatory effect of probiotics and prebiotics in Channa punctatus against Aphanomyces invadans. Fish & Shellfish Immunology. 2019; 86: 965–973, https://doi.org/10.1016/j.fsi.2018.12.051.

85. Schmitz S, Henrich M, Neiger R, Werling D, Allenspach K. Comparison of TNFα responses induced by Toll-like receptor ligands and probiotic Enterococcus faecium in whole blood and peripheral blood mononuclear cells of healthy dogs. Veterinary Immunology and Immunopathology. 2013; 153: 170–174. doi: 10.1016/j.vetimm.2013.02.014 23507437


Článok vyšiel v časopise

PLOS One


2019 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#