Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(10): e23149, 2023 10.
Article in English | MEDLINE | ID: mdl-37671857

ABSTRACT

The gut microbiota plays a key role in the postnatal development of the intestinal epithelium. However, the bacterial members of the primocolonizing microbiota driving these effects are not fully identified and the mechanisms underlying their long-term influence on epithelial homeostasis remain poorly described. Here, we used a model of newborn piglets treated during the first week of life with the antibiotic colistin in order to deplete specific gram-negative bacteria that are transiently dominant in the neonatal gut microbiota. Colistin depleted Proteobacteria and Fusobacteriota from the neonatal colon microbiota, reduced the bacterial predicted capacity to synthetize lipopolysaccharide (LPS), and increased the concentration of succinate in the colon. The colistin-induced disruption of the primocolonizing microbiota was associated with altered gene expression in the colon epithelium including a reduction of toll-like receptor 4 (TLR4) and lysozyme (LYZ). Our data obtained in porcine colonic organoid cell monolayers suggested that these effects were not driven by the variation of succinate or LPS levels nor by a direct effect of colistin on epithelial cells. The disruption of the primocolonizing microbiota imprinted colon epithelial stem cells since the expression of TLR4 and LYZ remained lower in organoids derived from colistin-treated piglet colonic crypts after several passages when compared to control piglets. Finally, the stable imprinting of LYZ in colon organoids was independent of the H3K4me3 level in its transcription start site. Altogether, our results show that disruption of the primocolonizing gut microbiota alters epithelial innate immunity in the colon and imprints stem cells, which could have long-term consequences for gut health.


Subject(s)
Microbiota , Animals , Swine , Toll-Like Receptor 4 , Colistin , Lipopolysaccharides , Stem Cells , Succinates , Succinic Acid , Colon , Homeostasis
2.
Nutrients ; 12(7)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708852

ABSTRACT

The early life period is crucial for the maturation of the intestinal barrier, its immune system, and a life-long beneficial host-microbiota interaction. The study aims to assess the impact of a beneficial dietary (short-chain fructooligosaccharides, scFOS) supplementation vs. a detrimental dietary environment (such as mycotoxin deoxynivalenol, DON) on offspring intestinal immune system developmental profiles. Sows were given scFOS-supplemented or DON-contaminated diets during the last 4 weeks of gestation, whereas force-feeding piglets with DON was performed during the first week of offspring life. Intestinal antigen-presenting cell (APC) subset frequency was analyzed by flow cytometry in the Peyer's patches and in lamina propria and the responsiveness of intestinal explants to toll-like receptor (TLR) ligands was performed using ELISA and qRT-PCR from post-natal day (PND) 10 until PND90. Perinatal exposure with scFOS did not affect the ontogenesis of APC. While it early induced inflammatory responses in piglets, scFOS further promoted the T regulatory response after TLR activation. Sow and piglet DON contamination decreased CD16+ MHCII+ APC at PND10 in lamina propria associated with IFNγ inflammation and impairment of Treg response. Our study demonstrated that maternal prebiotic supplementation and mycotoxin contamination can modulate the mucosal immune system responsiveness of offspring through different pathways.


Subject(s)
Food Contamination/analysis , Immune System/metabolism , Mucous Membrane/metabolism , Mycotoxins/toxicity , Prebiotics/administration & dosage , Animal Feed/analysis , Animal Feed/toxicity , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Cytokines/metabolism , Diet/veterinary , Dietary Supplements , Female , Interferon-gamma/metabolism , Maternal Nutritional Physiological Phenomena/drug effects , Mycotoxins/administration & dosage , Oligosaccharides/administration & dosage , Pregnancy , Pregnancy, Animal/drug effects , Pregnancy, Animal/immunology , Receptors, IgG/metabolism , Swine , Trichothecenes/administration & dosage , Trichothecenes/toxicity
3.
Front Behav Neurosci ; 13: 161, 2019.
Article in English | MEDLINE | ID: mdl-31379533

ABSTRACT

Psychological chronic stress is an important risk factor for major depressive disorder, of which consequences have been widely studied in rodent models. This work aimed at describing a pig model of chronic stress based on social isolation, environmental impoverishment and unpredictability. Three groups of animals of both sexes were constituted. Two were exposed to the psychosocial stressors while receiving (SF, n = 12) or not (SC, n = 22) the antidepressant fluoxetine, and a third group (NSC, n = 22) remained unstressed. Animals were observed in home pens and during dedicated tests to assess resignation and anxiety-like behaviors. Brain structure and function were evaluated via proton MRS and fMRI. Hippocampal molecular biology and immunodetection of cellular proliferation (Ki67+) and neuron maturation (DCX+) in the dentate gyrus were also performed. Salivary cortisol, fecal short-chain fatty acids (SCFAs), and various plasmatic and intestinal biomarkers were analyzed. Compared to NSC, SC animals showed more resignation (p = 0.019) and had a higher level of salivary cortisol (p = 0.020). SC brain responses to stimulation by a novel odor were lower, similarly to their hippocampal neuronal density (p = 0.015), cellular proliferation (p = 0.030), and hippocampal levels of BDNF and 5-HT1AR (p = 0.056 and p = 0.007, respectively). However, the number of DCX+ cells was higher in the ventral dentate gyrus in this group (p = 0.025). In addition, HOMA-IR was also higher (p < 0.001) and microbiota fermentation activity was lower (SCFAs, SC/NSC: p < 0.01) in SC animals. Fluoxetine partially or totally reversed several of these effects. Exposure to psychosocial stressors in the pig model induced effects consistent with the human and rodent literature, including resignation behavior and alterations of the HPA axis and hippocampus. This model opens the way to innovative translational research exploring the mechanisms of chronic stress and testing intervention strategies with good face validity related to human.

4.
FASEB J ; 33(1): 301-313, 2019 01.
Article in English | MEDLINE | ID: mdl-29975568

ABSTRACT

Perinatal nutrition programs physiologic and metabolic functions, with consequences on the susceptibility to develop metabolic diseases in adulthood. The microbiota represents a key factor of such programming. We investigated whether perinatal prebiotic [short-chain fructooligosaccharides (scFOS)] supplementation improved adult metabolic health in association with microbiota changes in pigs used as human model. Sows were supplemented with scFOS or not during the end of gestation and the entire lactation, and offspring received scFOS accordingly during 1 mo after weaning. Pigs were then fed a standard diet for 5 mo, followed by a high-fat diet for 3 mo once adults. Perinatal scFOS supplementation induced a persistent modulation of the composition of the fecal microbiota in adulthood, notably by increasing the Prevotella genus. Meanwhile, scFOS animals displayed improved capacity to secrete glucagon-like peptide-1 and improved pancreas sensitivity to glucose without any changes in peripheral insulin sensitivity. Perinatal scFOS supplementation also increased ileal secretory IgA secretion and alkaline phosphatase activity and decreased TNF-α expression in adipose tissue. In conclusion, perinatal scFOS supplementation induced long-lasting modulation of intestinal microbiota and had beneficial consequences on the host physiology in adulthood. Our results highlight the key role of perinatal nutrition on later microbiota and host metabolic adaptation to an unbalanced diet.-Le Bourgot, C., Ferret-Bernard, S., Apper, E., Taminiau, B., Cahu, A., Le Normand, L., Respondek, F., Le Huërou-Luron, I., Blat, S. Perinatal short-chain fructooligosaccharides program intestinal microbiota and improve enteroinsular axis function and inflammatory status in high-fat diet-fed adult pigs.


Subject(s)
Animal Feed/analysis , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Inflammation/veterinary , Intestinal Diseases/veterinary , Oligosaccharides/administration & dosage , Swine Diseases/prevention & control , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Dietary Supplements , Feces/microbiology , Female , Glucose/metabolism , Glucose Tolerance Test , Inflammation/drug therapy , Inflammation/etiology , Insulin/metabolism , Intestinal Diseases/drug therapy , Intestinal Diseases/etiology , Pregnancy , Swine , Swine Diseases/etiology
5.
Front Microbiol ; 9: 2584, 2018.
Article in English | MEDLINE | ID: mdl-30420848

ABSTRACT

Propionibacterium freudenreichii is a beneficial bacterium, used as a cheese starter, which presents versatile probiotic properties. These properties are strain-dependent. We hypothesized they may also be delivery vehicle-dependent. In this study, we thus explored in healthy piglets how the cheese matrix affects the immunomodulatory properties of P. freudenreichii. During 2 weeks, three groups of weaned piglets consumed, respectively, P. freudenreichii as a liquid culture (PF-culture), P. freudenreichii under the form of a cheese (PF-cheese), or a control sterile cheese matrix (Cheese-matrix). The in vivo metabolic activity of P. freudenreichii was assessed by determining short chain fatty acids (SCFA) concentration and bifidobacteria population in feces. Whatever the delivery vehicle, P. freudenreichii was metabolically active in piglets' colon and enhanced both bifidobacteria and SCFA in feces. P. freudenreichii consumption decreased the secretion of TNFα and of IL-10 by peripheral blood mononuclear cells (PBMC). It did not alter IL-10, IFNγ, IL-17, and TNFα secretion in mesenteric lymph node immune cells (MLNC). PF-cheese enhanced significantly Treg phenotype, while PF-culture decreased significantly Th17 phenotype in PBMC and MLNC. Remarkably, only PF-cheese induced an increase of Th2 phenotype in PBMC and MLNC. Ex vivo stimulation of PBMC and MLNC by Lipopolysaccharides and Concanavalin A emphasized the difference in the immunomodulatory responses between PF-culture and PF-cheese group, as well as between PBMC and MLNC. This study shows the importance to consider the delivery vehicle for probiotic administration. It confirms the anti-inflammatory potential of P. freudenreichii. It opens new perspectives for the use propionibacteria-fermented products as preventive agents for inflammatory bowel diseases and intestinal infectious diseases.

6.
Sci Rep ; 8(1): 11656, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30076313

ABSTRACT

Clinical and animal studies have demonstrated beneficial effects of early consumption of dairy lipids and a probiotic, Lactobacillus fermentum (Lf), on infant gut physiology. The objective of this study was to investigate their long-term effects on gut microbiota and host entero-insular axis and metabolism. Piglets were suckled with a milk formula containing only plant lipids (PL), a half-half mixture of plant lipids and dairy lipids (DL), or this mixture supplemented with Lf (DL + Lf). They were weaned on a standard diet and challenged with a high-energy diet until postnatal day 140. DL and DL + Lf modulated gut microbiota composition and metabolism, increasing abundance of several Clostridia genera. Moreover, DL + Lf specifically decreased the faecal content of 2-oxoglutarate and lysine compared to PL and 5-aminovalerate compared to PL and DL. It also increased short-chain fatty acid concentrations like propionate compared to DL. Furthermore, DL + Lf had a beneficial effect on the endocrine function, enhancing caecal GLP-1 and GLP-1 meal-stimulated secretion. Correlations highlighted the consistent relationship between microbiota and gut physiology. Together, our results evidence a beneficial programming effect of DL + Lf in infant formula composition on faecal microbiota and entero-insular axis function.


Subject(s)
Gastrointestinal Microbiome/drug effects , Infant Formula/chemistry , Lipids/administration & dosage , Probiotics/administration & dosage , Animals , Dietary Supplements , Feces/microbiology , Humans , Infant , Limosilactobacillus fermentum/chemistry , Lipids/chemistry , Milk/chemistry , Probiotics/chemistry , Swine , Swine, Miniature
7.
Eur J Nutr ; 57(2): 463-476, 2018 Mar.
Article in English | MEDLINE | ID: mdl-27744547

ABSTRACT

PURPOSE: Although composition of infant formula has been significantly improved during the last decade, major differences with the composition and structure of breast milk still remain and might affect nutrient digestion and gut biology. We hypothesized that the incorporation of dairy fat in infant formulas could modify their physiological impacts by making their composition closer to that of human milk. The effect of milk fat and milk fat globule membrane (MFGM) fragments in infant formulas on gut digestion, mucosal immunity and microbiota composition was evaluated. METHODS: Three formulas containing either (1) vegetable lipids stabilized only by proteins (V-P), (2) vegetable lipids stabilized by a mixture of proteins and MFGM fragments (V-M) and (3) a mixture of milk and vegetable lipids stabilized by a mixture of proteins and MFGM fragments (M-M) were automatically distributed to 42 newborn piglets until slaughter at postnatal day (PND) 7 or 28, and compared to a fourth group of sow's suckling piglets (SM) used as a breast-fed reference. RESULTS: At both PND, casein and ß-lactoglobulin digestion was reduced in M-M proximal jejunum and ileum contents compared to V-P and V-M ones leading to more numerous ß-Cn peptides in M-M contents. The IFNγ cytokine secretion of ConA-stimulated MLN cells from M-M piglets tended to be higher than in V-P ones at PND 7 and PND 28 and was closer to that of SM piglets. No dietary treatment effect was observed on IL-10 MLN cell secretion. Changes in faecal microbiota in M-M piglets resulted in an increase in Proteobacteria and Bacteroidetes and a decrease in Firmicutes phyla compared to V-P ones. M-M piglets showed higher abundances of Parabacteroides, Escherichia/Shigella and Klebsiella genus. CONCLUSIONS: The incorporation of both milk fat and MFGM fragments in infant formula modifies protein digestion, the dynamic of the immune system maturation and the faecal microbiota composition.


Subject(s)
Animal Nutritional Physiological Phenomena , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Immunomodulation , Milk/chemistry , Models, Immunological , Plant Oils/administration & dosage , Animals , Animals, Newborn , Caseins/administration & dosage , Caseins/metabolism , Cytokines/metabolism , Digestion , Feces/microbiology , Gastrointestinal Contents/chemistry , Gastrointestinal Contents/microbiology , Glycolipids/administration & dosage , Glycolipids/metabolism , Glycoproteins/administration & dosage , Glycoproteins/metabolism , Humans , Infant Formula , Infant Nutritional Physiological Phenomena , Infant, Newborn , Lactoglobulins/administration & dosage , Lactoglobulins/metabolism , Lipid Droplets , Lymph Nodes/growth & development , Lymph Nodes/immunology , Lymph Nodes/metabolism , Milk/metabolism , Plant Oils/metabolism , Plant Proteins, Dietary/administration & dosage , Plant Proteins, Dietary/metabolism , Sus scrofa/growth & development
8.
FASEB J ; 32(4): 2160-2171, 2018 04.
Article in English | MEDLINE | ID: mdl-29242276

ABSTRACT

Butyrate can improve gut functions, whereas histone deacetylase inhibitors might alleviate neurocognitive alterations. Our aim was to assess whether oral butyrate could modulate brain metabolism and plasticity and if this would relate to gut function. Sixteen pigs were subjected to sodium butyrate (SB) supplementation via beverage water or water only [control (C)]. All pigs had blood sampled after 2 and 3 wk of treatment, and were subjected to a brain positron emission tomography after 3 wk. Animals were euthanized after 4 wk to sample pancreas, intestine, and brain for gut physiology and anatomy measurements, as well as hippocampal histology, Ki67, and doublecortin (DCX) immunohistochemistry. SB compared with C treatment triggered basal brain glucose metabolism changes in the nucleus accumbens and hippocampus ( P = 0.003), increased hippocampal granular cell layer volume ( P = 0.006), and neurogenesis (Ki67: P = 0.026; DCX: P = 0.029). After 2 wk of treatment, plasma levels of glucose, insulin, lactate, glucagon-like peptide 1, and peptide tyrosine tyrosine remained unchanged. After 3 wk, plasma levels of lactate were lower in SB compared with C animals ( P = 0.028), with no difference for glucose and insulin. Butyrate intake impacted very little gut anatomy and function. These results demonstrate that oral SB impacted brain functions with little effects on the gut.-Val-Laillet, D., Guérin, S., Coquery, N., Nogret, I., Formal, M., Romé, V., Le Normand, L., Meurice, P., Randuineau, G., Guilloteau, P., Malbert, C.-H., Parnet, P., Lallès, J.-P., Segain, J.-P. Oral sodium butyrate impacts brain metabolism and hippocampal neurogenesis, with limited effects on gut anatomy and function in pigs.


Subject(s)
Butyric Acid/pharmacology , Hippocampus/drug effects , Histamine Antagonists/pharmacology , Intestines/drug effects , Neurogenesis , Administration, Oral , Animals , Blood Glucose/metabolism , Butyric Acid/administration & dosage , Butyric Acid/adverse effects , Female , Hippocampus/growth & development , Hippocampus/metabolism , Histamine Antagonists/administration & dosage , Histamine Antagonists/adverse effects , Insulin/blood , Intestines/physiology , Lactic Acid/blood , Swine
9.
Br J Nutr ; 117(1): 83-92, 2017 01.
Article in English | MEDLINE | ID: mdl-28115029

ABSTRACT

Prebiotic supplementation modulates immune system development and function. However, less is known about the effects of maternal prebiotic consumption on offspring intestinal defences and immune system responsiveness. We investigated the effects of maternal short-chain fructo-oligosaccharide (scFOS) supplementation on mucin-secreting cells, ileal secretory IgA and cytokine secretion of weaned offspring and their humoral response to an oral vaccine against obligate intracellular Lawsonia intracellularis. Sows were fed a control diet (CTRL) or scFOS-supplemented diet during the last third of gestation and throughout lactation. At weaning, each litter was divided into two groups receiving a post-weaning CTRL or scFOS diet for a month. Pigs from the four groups were either non-vaccinated (n 16) or vaccinated (n 117) at day 33. Biomarkers related to intestinal defences and immune parameters were analysed 3 weeks later. SCFA production was assessed over time in suckling and weaned pigs. Maternal scFOS supplementation improved ileal cytokine secretions (interferon (IFN)-γ, P<0·05; IL-4, P=0·07) and tended to increase caecal goblet cell number (P=0·06). It increased IgA vaccine response in the serum (P<0·01) and ileal mucosa (P=0·08). Higher bacterial fermentative activity was observed during lactation (total faecal SCFA, P<0·001) and after weaning (colonic butyrate, P=0·10) in pigs from scFOS-supplemented mothers. No synergistic effect between maternal and post-weaning scFOS supplementation was observed. Therefore, maternal scFOS supplementation has long-lasting consequences by strengthening gut defences and immune response to a vaccine against an intestinal obligate intracellular pathogen. Prebiotic consumption by gestating and lactating mothers is decisive in modulating offspring intestinal immunity.


Subject(s)
Bacterial Vaccines/immunology , Butyrates/blood , Cytokines/metabolism , Goblet Cells/physiology , Lawsonia Bacteria , Oligosaccharides/administration & dosage , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Cytokines/genetics , Desulfovibrionaceae Infections/microbiology , Desulfovibrionaceae Infections/veterinary , Diet/veterinary , Dietary Supplements , Female , Maternal Nutritional Physiological Phenomena , Oligosaccharides/chemistry , Prebiotics , Swine , Swine Diseases/prevention & control
10.
PLoS One ; 9(9): e107508, 2014.
Article in English | MEDLINE | ID: mdl-25238157

ABSTRACT

Peripartum nutrition is crucial for developing the immune system of neonates. We hypothesized that maternal short-chain fructooligosaccharide (scFOS) supplementation could accelerate the development of intestinal immunity in offspring. Thirty-four sows received a standard or a scFOS supplemented diet (10 g scFOS/d) for the last 4 weeks of gestation and the 4 weeks of lactation. Colostrum and milk immunoglobulins (Ig) and TGFß1 concentrations were evaluated on the day of delivery and at d 6 and d 21 postpartum. Piglet intestinal structure, the immunologic features of jejunal and ileal Peyer's patches, and mesenteric lymph node cells were analysed at postnatal d 21. Short-chain fatty acid concentrations were measured over time in the intestinal contents of suckling and weaned piglets. Colostral IgA (P<0.05) significantly increased because of scFOS and TGFß1 concentrations tended to improve (P<0.1). IFNγ secretion by stimulated Peyer's patch and mesenteric lymph node cells, and secretory IgA production by unstimulated Peyer's patch cells were increased (P<0.05) in postnatal d 21 scFOS piglets. These differences were associated with a higher proportion of activated CD25+CD4α+ T cells among the CD4+ helper T lymphocytes (P<0.05) as assessed by flow cytometry. IFNγ secretion was positively correlated with the population of activated T lymphocytes (P<0.05). Total short-chain fatty acids were unchanged between groups during lactation but were higher in caecal contents of d 90 scFOS piglets (P<0.05); specifically propionate, butyrate and valerate. In conclusion, we demonstrated that maternal scFOS supplementation modified the intestinal immune functions in piglets in association with increased colostral immunity. Such results underline the key role of maternal nutrition in supporting the postnatal development of mucosal immunity.


Subject(s)
Colostrum/immunology , Dietary Supplements , Intestines/immunology , Oligosaccharides/pharmacology , Swine/immunology , Animals , Female , Intestines/growth & development , Pregnancy , Prenatal Nutritional Physiological Phenomena , Swine/metabolism
11.
J Nutr Biochem ; 25(10): 1090-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25087993

ABSTRACT

We recently observed that maternal 18:3n-3 increases piglet jejunal permeability. We hypothesized that this would favor intestinal lipopolysaccharide (LPS) passage and alter gut immune system education toward this bacterial ligand. Sows were fed 18:3n-3 or 18:2n-6 diets throughout gestation and lactation. In each litter, two piglets were given oral Gram-negative spectrum antibiotic from post-natal day (PND) 14 to 28. All piglets were weaned on a regular diet at PND28. 18:3n-3 piglets exhibited greater jejunal permeability to FITC-LPS at PND28. Levels of 18:3n-3 but neither 20:5n-3 nor 20:4n-6 were greater in mesenteric lymph nodes (MLN) of 18:3n-3 piglets. Jejunal explant or MLN cell cytokine responses to LPS were not influenced by the maternal diet. Antibiotic increased jejunal permeability to FITC-LPS and lowered the level of 20:5n-3 in MLN, irrespective of the maternal diet. At PND52, no long-lasting effect of the maternal diet or antibiotic treatment on jejunal permeability was noticed. 18:3n-3 and 20:4n-6 levels were greater and lower, respectively, in MLN of 18:3n-3 compared to 18:2n-6 piglets. IL-10 production by MLN cells in response to LPS was greater in the 18:3n-3 group, irrespective of the neonatal antibiotic treatment. IL-8 secretion by jejunal explants in response to LPS was lower in antibiotic-treated 18:3n-3 compared to 18:2n-6 piglets. Finally, proportion of MHC class II(+) antigen-presenting cells was greater in 18:3n-3 than 18:2n-6 MLN cells. In conclusion, maternal 18:3n-3 directs the intestinal immune response to LPS toward an anti-inflammatory profile beyond the breastfeeding period; microbiota involvement seems dependent of the immune cells considered.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Jejunum/drug effects , Lipopolysaccharides/adverse effects , Animals , Animals, Newborn , Cells, Cultured , Cytokines/metabolism , DNA, Bacterial/genetics , Diet/veterinary , Fatty Acids, Omega-6/pharmacology , Female , Inflammation/pathology , Jejunum/immunology , Jejunum/microbiology , Maternal Nutritional Physiological Phenomena , Mesenteric Veins/drug effects , Mesenteric Veins/metabolism , Microbiota , Permeability , Pregnancy , Pregnancy Outcome , Swine , Weaning
12.
BMC Vet Res ; 8: 70, 2012 May 28.
Article in English | MEDLINE | ID: mdl-22640469

ABSTRACT

BACKGROUND: Physiology of the exocrine pancreas has been well studied in domestic and in laboratory animals as well as in humans. However, it remains quite unknown in wildlife mammals. Roe deer and cattle (including calf) belong to different families but have a common ancestor. This work aimed to evaluate in the Roe deer, the adaptation to diet of the exocrine pancreatic functions and regulations related to animal evolution and domestication. RESULTS: Forty bovine were distributed into 2 groups of animals either fed exclusively with a milk formula (monogastric) or fed a dry feed which allowed for rumen function to develop, they were slaughtered at 150 days of age. The 35 Roe deer were wild animals living in the temperate broadleaf and mixed forests, shot during the hunting season and classified in two groups adult and young. Immediately after death, the pancreas was removed for tissue sample collection and then analyzed. When expressed in relation to body weight, pancreas, pancreatic protein weights and enzyme activities measured were higher in Roe deer than in calf. The 1st original feature is that in Roe deer, the very high content in pancreatic enzymes seems to be related to specific digestive products observed (proline-rich proteins largely secreted in saliva) which bind tannins, reducing their deleterious effects on protein digestion. The high chymotrypsin and elastase II quantities could allow recycling of proline-rich proteins. In contrast, domestication and rearing cattle resulted in simplified diet with well digestible components. The 2nd feature is that in wild animal, both receptor subtypes of the CCK/gastrin family peptides were present in the pancreas as in calf, although CCK-2 receptor subtype was previously identified in higher mammals. CONCLUSIONS: Bovine species could have lost some digestive capabilities (no ingestion of great amounts of tannin-rich plants, capabilities to secrete high amounts of proline-rich proteins) compared with Roe deer species. CCK and gastrin could play an important role in the regulation of pancreatic secretion in Roe deer as in calf. This work, to the best of our knowledge is the first study which compared the Roe deer adaptation to diet with a domesticated animal largely studied.


Subject(s)
Adaptation, Physiological/physiology , Cattle/physiology , Deer/physiology , Pancreas/physiology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Cattle/genetics , Deer/genetics , Diet/veterinary , Digestion/physiology , Male , Organ Size , Pancreas/anatomy & histology , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...