1
|
Estrada-Meza J, Videlo J, Bron C, Duchampt A, Saint-Béat C, Zergane M, Silva M, Rajas F, Bouret SG, Mithieux G, Gautier-Stein A. Intestinal gluconeogenesis controls the neonatal development of hypothalamic feeding circuits. Mol Metab 2024; 89:102036. [PMID: 39304064 PMCID: PMC11470480 DOI: 10.1016/j.molmet.2024.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE Intestinal gluconeogenesis (IGN) regulates adult energy homeostasis in part by controlling the same hypothalamic targets as leptin. In neonates, leptin exhibits a neonatal surge controlling axonal outgrowth between the different hypothalamic nuclei involved in feeding circuits and autonomic innervation of peripheral tissues involved in energy and glucose homeostasis. Interestingly, IGN is induced during this specific time-window. We hypothesized that the neonatal pic of IGN also regulates the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues. METHODS We genetically induced neonatal IGN by overexpressing G6pc1 the catalytic subunit of glucose-6-phosphatase (the mandatory enzyme of IGN) at birth or at twelve days after birth. The neonatal development of hypothalamic feeding circuits was studied by measuring Agouti-related protein (AgRP) and Pro-opiomelanocortin (POMC) fiber density in hypothalamic nuclei of 20-day-old pups. The effect of the neonatal induction of intestinal G6pc1 on sympathetic innervation of the adipose tissues was studied via tyrosine hydroxylase (TH) quantification. The metabolic consequences of the neonatal induction of intestinal G6pc1 were studied in adult mice challenged with a high-fat/high-sucrose (HFHS) diet for 2 months. RESULTS Induction of intestinal G6pc1 at birth caused a neonatal reorganization of AgRP and POMC fiber density in the paraventricular nucleus of the hypothalamus, increased brown adipose tissue tyrosine hydroxylase levels, and protected against high-fat feeding-induced metabolic disorders. In contrast, inducing intestinal G6pc1 12 days after birth did not impact AgRP/POMC fiber densities, adipose tissue innervation or adult metabolism. CONCLUSION These findings reveal that IGN at birth but not later during postnatal life controls the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues, promoting a better management of metabolism in adulthood.
Collapse
Affiliation(s)
| | - Jasmine Videlo
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Clara Bron
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Adeline Duchampt
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Mickael Zergane
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Marine Silva
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Fabienne Rajas
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Sebastien G Bouret
- University Lille, Inserm, CHU Lille, Laboratory of development and plasticity of the Neuroendocrine brain, Lille Neuroscience & Cognition, Inserm UMR-S1172, Lille, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | |
Collapse
|
2
|
Le Bourgot C, Lollier V, Richer Y, Thoulouze L, Svilar L, Le Gall S, Blat S, Le Huërou-Luron I. Maternal short chain fructo-oligosaccharides supplementation during late gestation and lactation influences milk components and offspring gut metabolome: a pilot study. Sci Rep 2024; 14:4236. [PMID: 38378944 PMCID: PMC10879084 DOI: 10.1038/s41598-024-54813-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Breast milk composition is influenced by maternal diet. This study aimed to evaluate if supplementation of maternal diet with a prebiotic fibre, through its potential effect on milk composition, can be a leverage to orientate the gut microbiota of infants in a way that would be beneficial for their health. Twelve sows received a diet supplemented with short chain fructo-oligosaccharides or maltodextrins during the last month of gestation and the lactation. Oligosaccharidic and lipidomic profiles of colostrum and mature milk (21 days), as well as faecal microbiota composition and metabolomic profile of 21 day-old piglets were evaluated. The total porcine milk oligosaccharide concentration tended to be lower in scFOS-supplemented sows, mainly due to the significant reduction of the neutral core oligosaccharides (in particular that of a tetrahexose). Maternal scFOS supplementation affected the concentration of 31 lipids (mainly long-chain triglycerides) in mature milk. Faecal short-chain fatty acid content and that of 16 bacterial metabolites were modified by scFOS supplementation. Interestingly, the integrative data analysis gave a novel insight into the relationships between (i) maternal milk lipids and PMOs and (ii) offspring faecal bacteria and metabolites. In conclusion, scFOS-enriched maternal diet affected the composition of mature milk, and this was associated with a change in the colonisation of the offspring intestinal microbiota.
Collapse
Affiliation(s)
- Cindy Le Bourgot
- Tereos, Scientific and Regulatory Affairs Department, Moussy-le-Vieux, France.
| | - Virginie Lollier
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Yoann Richer
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Loric Thoulouze
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Ljubica Svilar
- Cribiom, Centre de Recherche Cardiovasculaire et Nutrition C2VN, UMR INRAE 1260 INSERM 1263, University Aix-Marseille, Marseille, France
| | - Sophie Le Gall
- INRAE, UR1268 BIA, 44300, Nantes, France
- INRAE, PROBE Research Infrastructure, BIBS Facility, 44300, Nantes, France
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, University Rennes, 35590, Saint-Gilles, France
| | | |
Collapse
|
3
|
Kiernan DP, O’Doherty JV, Sweeney T. The Effect of Prebiotic Supplements on the Gastrointestinal Microbiota and Associated Health Parameters in Pigs. Animals (Basel) 2023; 13:3012. [PMID: 37835619 PMCID: PMC10572080 DOI: 10.3390/ani13193012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Establishing a balanced and diverse microbiota in the GIT of pigs is crucial for optimizing health and performance throughout the production cycle. The post-weaning period is a critical phase, as it is often associated with dysbiosis, intestinal dysfunction and poor performance. Traditionally, intestinal dysfunctions associated with weaning have been alleviated using antibiotics and/or antimicrobials. However, increasing concerns regarding the prevalence of antimicrobial-resistant bacteria has prompted an industry-wide drive towards identifying natural sustainable dietary alternatives. Modulating the microbiota through dietary intervention can improve animal health by increasing the production of health-promoting metabolites associated with the improved microbiota, while limiting the establishment and proliferation of pathogenic bacteria. Prebiotics are a class of bioactive compounds that resist digestion by gastrointestinal enzymes, but which can still be utilized by beneficial microbes within the GIT. Prebiotics are a substrate for these beneficial microbes and therefore enhance their proliferation and abundance, leading to the increased production of health-promoting metabolites and suppression of pathogenic proliferation in the GIT. There are a vast range of prebiotics, including carbohydrates such as non-digestible oligosaccharides, beta-glucans, resistant starch, and inulin. Furthermore, the definition of a prebiotic has recently expanded to include novel prebiotics such as peptides and amino acids. A novel class of -biotics, referred to as "stimbiotics", was recently suggested. This bioactive group has microbiota-modulating capabilities and promotes increases in short-chain fatty acid (SCFA) production in a disproportionally greater manner than if they were merely substrates for bacterial fermentation. The aim of this review is to characterize the different prebiotics, detail the current understating of stimbiotics, and outline how supplementation to pigs at different stages of development and production can potentially modulate the GIT microbiota and subsequently improve the health and performance of animals.
Collapse
Affiliation(s)
- Dillon P. Kiernan
- School of Veterinary Medicine, University College Dublin, Belfield, D04 W6F6 Dublin, Ireland;
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 W6F6 Dublin, Ireland;
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Belfield, D04 W6F6 Dublin, Ireland;
| |
Collapse
|
4
|
Lin A, Yan X, Xu R, Wang H, Su Y, Zhu W. Effects of lactic acid bacteria-fermented formula milk supplementation on colonic microbiota and mucosal transcriptome profile of weaned piglets. Animal 2023; 17:100959. [PMID: 37688970 DOI: 10.1016/j.animal.2023.100959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 09/11/2023] Open
Abstract
Supplemental probiotic fermented milk as a gut modulator can improve growth performance for weaned piglets by promoting the development of the small intestine in digestion and immune function. The effect on colon health might also play a considerable part in the favourable role of probiotic fermented milk in the growth performance improvement of weaned piglets; however, it has yet to be reported. This study aimed to investigate the effects of supplementation with lactic acid bacteria-fermented formula milk (LFM) on colonic morphology, microbiota composition, and mucosal transcriptome profile in weaned piglets. A total of 24 male weaned piglets were randomly divided into two groups: a control (CON) treatment or the LFM-supplemented treatment. Each group consisted of six replicates (cages) with two piglets per cage, and each piglet in the LFM group was supplemented with 80 mL LFM three times a day for 21 d, while the CON group was treated with the same amount of drinking water. Results showed that supplementation of LFM reduced the colonic histological damage scores and significantly increased the number of goblet cells per crypt. Furthermore, LFM consumption decreased the levels of pro-inflammation cytokines in the colonic mucosa. LFM downregulated the expression of inflammatory genes (CXCL9 and CXCL10) involving Toll-like receptor signalling pathway, immune response, and response to bacterium, and up-regulated two active genes (S100A8 and S100A9) involving the IL-17 signalling pathway and Toll-like receptor 4 binding. In addition, LFM could increase the potential probiotic genera containing Lachnospira and Anaerorhabdus furcosa group, which were positively related to short-chain fatty acid (SCFA) production. Correspondingly, LFM-fed piglets had higher total bacterial load and total SCFA concentration in the colonic digesta compared with the CON group. These novel findings support the benefits of LFM in enhancing intestinal homoeostasis and ameliorating weaning stress for weaned piglets, which is associated with the modulation of gut microbiota composition and immune-related genes.
Collapse
Affiliation(s)
- A Lin
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - X Yan
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - R Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - H Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Y Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China.
| | - W Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
5
|
Xu T, Guo Y, Zhang Y, Cao K, Zhou X, Qian M, Han X. Alleviative Effect of Probiotic Ferment on Lawsonia intracellularis Infection in Piglets. BIOLOGY 2023; 12:879. [PMID: 37372164 DOI: 10.3390/biology12060879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023]
Abstract
(1) Background: Lawsonia intracellularis (LI) is an obligate intracellular Gram-negative bacterium that causes porcine ileitis. Pigs infected with LI have severe ileal lesions and show symptoms of diarrhea, indigestion, and growth retardation. Previous studies found that probiotic ferment (FAM) improved the growth performance, gut barrier, and function in piglets. Therefore, we aimed to reveal the mechanism that FAM alleviates negative performance in LI-challenged piglets by characterizing the changes in intestinal integrity, function, and gut microbiota following FAM supplementation. (2) Methods: Twenty-four healthy piglets were randomly allotted to four treatments. Three groups were challenged with LI; both FAM addition and vaccination were performed to explore their positive effects on LI-infected piglets. (3) Results: Piglets infected with LI showed lower growth performance and typical pathological symptoms. Moreover, microscopic images showed that observed intestinal morphological damage could be repaired by FAM and vaccine. To explore the digestion of nutrients in piglets, both digestive enzyme activity and ileal transporter expression were performed to reveal the promoting effect of additives. Reduction of LI colonization intervention by FAM could also ameliorate abnormal differentiation and function of intestinal epithelial cells and alleviate severe inflammatory responses in piglets. Regarding the gut microbiota, both the structure and function of the ileal and colonic microbiota were altered following FAM supplementation. (4) Conclusions: In conclusion, probiotic ferment can reduce the colonization of LI in the ileum, improve intestinal damage, barrier function and microbiota structure, and enhance digestive enzyme activity and nutrient transport proteins expression, thereby improving piglet growth performance, which has the effect of preventing ileitis in pigs.
Collapse
Affiliation(s)
- Tingting Xu
- Hainan Institute, Zhejiang University, Yazhou Bay Science and Technology City, Sanya 572025, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yong Guo
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, College of Animal Science and Technology, Hangzhou 310022, China
| | - Yuanyuan Zhang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kai Cao
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinchen Zhou
- Hainan Institute, Zhejiang University, Yazhou Bay Science and Technology City, Sanya 572025, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mengqi Qian
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyan Han
- Hainan Institute, Zhejiang University, Yazhou Bay Science and Technology City, Sanya 572025, China
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Ferreres-Serafini L, Castillejos L, Martín M, Le Bourgot C, Martín-Orúe SM. Looking for Possible Benefits of Combining Short-Chain Fructo-Oligosaccharides (scFOS) with Saccharomyces cerevisiae Sc 47 on Weaned Pigs Orally Challenged with Escherichia coli F4 . Animals (Basel) 2023; 13:526. [PMID: 36766416 PMCID: PMC9913220 DOI: 10.3390/ani13030526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The objective of this work was to evaluate the effect of supplementing short-chain fructo-oligosaccharides (scFOS) combined or not with live yeast Saccharomyces cerevisiae Sc 47 on weanling pigs challenged with Escherichia coli F4+. We allocated ninety-six piglets to four experimental diets: control (CTR); supplemented with scFOS (5 g/kg Profeed® P95) (scFOS); S. cerevisiae Sc 47 (1 g/kg Actisaf® Sc 47 HR +) (YEA); or both (SYN). Parameters included: performance; E. coli F4+ detection; fermentation activity; inflammatory biomarkers; and ileal histomorphology. Our results showed that supplementing scFOS was able to reduce the incidence of diarrhea, and both supplements were able to lower counts of EHEC along the gut. Supplementing scFOS was mostly associated with changes in the gut ecosystem and increases in the lactobacilli population, while S. cerevisiae Sc 47 registered increases in the numbers of ileal intraepithelial lymphocytes. The synbiotic mixture showed the lowest diarrhea incidence and fecal scores, benefiting from complementary modes of action and possible synergistic effects due to a hypothesized yeast-LAB cross-feeding phenomenon in the foregut. In conclusion, our results evidence that supplementing scFOS or Saccharomyces cerevisiae Sc 47 is efficacious to fight post-weaning colibacillosis, and combining both could be beneficial in high-risk scenarios.
Collapse
Affiliation(s)
- Laia Ferreres-Serafini
- Animal Nutrition and Welfare Service (SNIBA), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Lorena Castillejos
- Animal Nutrition and Welfare Service (SNIBA), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Marga Martín
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | | | - Susana M. Martín-Orúe
- Animal Nutrition and Welfare Service (SNIBA), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| |
Collapse
|
7
|
Liu N, Shen H, Zhang F, Liu X, Xiao Q, Jiang Q, Tan B, Ma X. Applications and prospects of functional oligosaccharides in pig nutrition: A review. ANIMAL NUTRITION 2023. [DOI: 10.1016/j.aninu.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
8
|
Charton E, Bourgeois A, Bellanger A, Le-Gouar Y, Dahirel P, Romé V, Randuineau G, Cahu A, Moughan PJ, Montoya CA, Blat S, Dupont D, Deglaire A, Le Huërou-Luron I. Infant nutrition affects the microbiota-gut-brain axis: Comparison of human milk vs. infant formula feeding in the piglet model. Front Nutr 2022; 9:976042. [PMID: 36211510 PMCID: PMC9532976 DOI: 10.3389/fnut.2022.976042] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Early nutrition plays a dominant role in infant development and health. It is now understood that the infant diet impacts the gut microbiota and its relationship with gut function and brain development. However, its impact on the microbiota-gut-brain axis has not been studied in an integrative way. The objective here was to evaluate the effects of human milk (HM) or cow’s milk based infant formula (IF) on the relationships between gut microbiota and the collective host intestinal-brain axis. Eighteen 10-day-old Yucatan mini-piglets were fed with HM or IF. Intestinal and fecal microbiota composition, intestinal phenotypic parameters, and the expression of genes involved in several gut and brain functions were determined. Unidimensional analyses were performed, followed by multifactorial analyses to evaluate the relationships among all the variables across the microbiota-gut-brain axis. Compared to IF, HM decreased the α-diversity of colonic and fecal microbiota and modified their composition. Piglets fed HM had a significantly higher ileal and colonic paracellular permeability assessed by ex vivo analysis, a lower expression of genes encoding tight junction proteins, and a higher expression of genes encoding pro-inflammatory and anti-inflammatory immune activity. In addition, the expression of genes involved in endocrine function, tryptophan metabolism and nutrient transport was modified mostly in the colon. These diet-induced intestinal modifications were associated with changes in the brain tissue expression of genes encoding the blood-brain barrier, endocrine function and short chain fatty acid receptors, mostly in hypothalamic and striatal areas. The integrative approach underlined specific groups of bacteria (Veillonellaceae, Enterobacteriaceae, Lachnospiraceae, Rikenellaceae, and Prevotellaceae) associated with changes in the gut-brain axis. There is a clear influence of the infant diet, even over a short dietary intervention period, on establishment of the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Elise Charton
- STLO, INRAE, Institut Agro, Rennes, France
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | | | | | | - Patrice Dahirel
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | - Véronique Romé
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | | - Armelle Cahu
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | - Paul J. Moughan
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Carlos A. Montoya
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Smart Foods and Bioproducts Innovation Centre of Excellence, AgResearch Limited, Palmerston North, New Zealand
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | | | | - Isabelle Le Huërou-Luron
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- *Correspondence: Isabelle Le Huërou-Luron,
| |
Collapse
|
9
|
Maternal stevioside supplementation improves intestinal immune function of chicken offspring potentially via modulating gut microbiota and down-regulating the promoter methylation level of suppressor of cytokine signaling 1 (SOCS1). ANIMAL NUTRITION 2022; 10:329-346. [PMID: 35919247 PMCID: PMC9307571 DOI: 10.1016/j.aninu.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/18/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022]
Abstract
The intestinal immune function of chickens is limited during the early growing stage. Maternal nutritional intervention has been suggested to affect the innate immunity of offspring. The present study aimed to investigate the effects of maternal stevioside supplementation on the intestinal immune function of chicken offspring. A total of 120 Jinmao yellow-feathered breeder hens were fed a basal diet or a diet supplemented with 250 mg/kg stevioside for 5 weeks. During the last week, 200 breeding eggs from each group were collected for incubation. After hatching, 80 male offspring (40 chickens from each group) were randomly selected and fed the same basal diet for 28 d. In addition, 90 well-shaped fertile eggs of non-treated breeder hens were incubated for the in ovo injection experiment. Steviol dissolved in 20% glycerol was injected at 7 d of incubation. The results showed that maternal stevioside supplementation could improve embryonic development, jejunal integrity and proliferation in the jejunal crypt (P < 0.05). Maternal stevioside supplementation could also increase the innate transcription levels of cytokines and endotoxin tolerance-related factors in the jejunum of chicken offspring (P < 0.05). At 28 d of age, the offspring following maternal stevioside supplementation exhibited higher jejunal secretory immunoglobulin A and serum interferons levels (P < 0.05). A higher abundance of Lactobacillales induced by maternal stevioside supplementation was positively correlated with intestinal immune-related factors (P < 0.05). The in ovo injection with steviol did not alter either embryonic development or intestinal immune function of hatching chickens (P > 0.05). Furthermore, maternal stevioside supplementation could induce hypo-methylation on the promoter region of suppressor of cytokine signaling 1 (SOCS1). In conclusion, maternal stevioside supplementation could improve the intestinal immune function of chicken offspring potentially via modulating the gut microbiota and down-regulating the promoter methylation level of SOCS1.
Collapse
|
10
|
Rodríguez-Sorrento A, Castillejos L, López-Colom P, Cifuentes-Orjuela G, Moreno-Muñoz JA, Martín-Orúe SM. Assessment of the Effects of the Synbiotic Combination of Bifidobacterium longum subsp. infantis CECT 7210 and Oligofructose-Enriched Inulin Against Digestive Bacterial Infections in a Piglet Model. Front Microbiol 2022; 13:831737. [PMID: 35350617 PMCID: PMC8957890 DOI: 10.3389/fmicb.2022.831737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/11/2022] [Indexed: 11/24/2022] Open
Abstract
The use of bifidobacteria as probiotics has proven to be beneficial in gastroenteric infections. Furthermore, prebiotics such as inulin can enhance the survival and growth of these bacteria. Two trials were performed to evaluate the effects of the administration of Bifidobacterium longum subsp. infantis CECT 7210 and oligofructose-enriched inulin against Salmonella enterica serovar Typhimurium or enterotoxigenic Escherichia coli (ETEC) F4. A total of 72 (Salmonella trial) and 96 (ETEC F4 trial) weaned piglets were used in a 2 × 2 design (with or without synbiotic, inoculated or not with the pathogen). After adaptation, animals were orally inoculated. Performance and clinical signs were evaluated. On days 4 and 8 (Salmonella trial) and 3 and 7 (ETEC F4 trial) post-inoculation (PI), one animal per pen was euthanized. Blood, digestive content and tissue samples were collected and microbiological counts, fermentation products, serum inflammatory markers and ileum histomorphometry analysis were performed. Both challenges had an impact on faecal consistency (p < 0.001), including the faecal shedding of Salmonella and increased numbers of enterobacteria and coliforms. The synbiotic administration did not have any effect on pathogen loads but induced changes in the fermentation profile, such as increased valeric acid in both trials as well as decreased acetic acid, except for Salmonella-challenged animals. The effect on propionate varied among trials, increasing in challenged synbiotic-treated pigs and decreasing in non-challenged ones in the Salmonella trial (P interaction = 0.013), while the opposed occurred in the ETEC F4 trial (P interaction = 0.013). The administration of the synbiotic increased intraepithelial lymphocytes (IEL; p = 0.039) on day 8 PI in the Salmonella trial and a similar trend occurred in non-challenged pigs in the ETEC F4 trial (P interaction = 0.086). The results did not provide evidence of reduced pathogen load with the synbiotic, although a modulation in fermentative activity could be identified depending on the challenge. Consistent increases were found in IEL, suggesting that this synbiotic combination has some immunomodulatory properties.
Collapse
Affiliation(s)
- Agustina Rodríguez-Sorrento
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lorena Castillejos
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Paola López-Colom
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departamento de Producción Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Agraria del Ecuador (UAE), Guayaquil, Ecuador
| | | | | | - Susana M Martín-Orúe
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
11
|
Cai J, Wang N, Chen J, Wu A, Nepovimova E, Valis M, Long M, Wu W, Kuca K. Bacillus velezensis A2 Inhibited the Cecal Inflammation Induced by Zearalenone by Regulating Intestinal Flora and Short-Chain Fatty Acids. Front Nutr 2022; 9:806115. [PMID: 35360686 PMCID: PMC8963806 DOI: 10.3389/fnut.2022.806115] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/09/2022] [Indexed: 12/16/2022] Open
Abstract
Zearalenone (ZEA) as an estrogen-like mycotoxin can cause the inflammatory injury of the cecum. How to reduce the harm that ZEA causes to humans and animals is a current concern for researchers. In this study, we aimed to ascertain whether Bacillus velezensis A2 (A2) could alleviate injury caused by ZEA by regulating the intestinal flora and the content of short chain fatty acids in the cecum among mice. Our results showed that Bacillus velezensis A2 improved the fold height, myometrial thickness, and crypt depth of the cecum induced by ZEA. Enzyme-linked immunosorbent assay and Western blotting results showed that A2 could decrease the ZEA-induced increase in expression levels of IL-2, IL-6, IFN-γ, TNF-α, and FC. Studies also showed that A2 increased the content of SCFA in the cecum which was decreased by ZEA. The microbial communities in the cecum were changed when given ZEA or A2. A2 was found to greatly reduce the ZEN-induced increase in the relative abundance of p_Actinobacteria, p_Protebacteria, o_Coriobacteriales, g_Anaerotruncus, g_Pseudoflavonifractor, g_Lachnoclostridium, g_Enterorhabdus, and f_Oscillospiraceae, and increase the ZEN-induced decrease in the relative abundance of f_Coriobacteriales. Results indicated that Bacillus velezensis A2 can largely ameliorate the intestinal inflammatory injury induced by ZEA in mice by regulating the microflora and short chain fatty acids content.
Collapse
Affiliation(s)
- Jing Cai
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Nan Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jia Chen
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Aibo Wu
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czechia
| | - Martin Valis
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Miao Long
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
- *Correspondence: Miao Long,
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czechia
- Wenda Wu,
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czechia
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czechia
- Kamil Kuca, ;
| |
Collapse
|
12
|
Henriksen IW, Mejia JLC, Mentzel CMJ, Lindenberg F, Hansen AK. Oligosaccharide equine feed supplement, Immulix, has minor impact on vaccine responses in mice. Sci Rep 2022; 12:582. [PMID: 35022427 PMCID: PMC8755741 DOI: 10.1038/s41598-021-04132-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/19/2021] [Indexed: 12/26/2022] Open
Abstract
Several mammalian species are vaccinated in early life, but little is known about the effect of diet on vaccine response. Oligosaccharides are increasingly proposed as dietary supplement for young individuals due to their anti-inflammatory potential elicited through modulation of gut microbiota (GM). Also, diet, e.g. the size of the fat fraction, is known to modulate the GM. We tested if an oligosaccharide diet (Immulix) and/or increased dietary fat content affected antibody titers to a tetanus vaccine in 48 BALB/cJTac mice through GM modulation. Female mice had significantly higher IgG titers with higher variation compared to male mice. The effects of Immulix and/or increased fat content were minor. Immulix negatively affected IgG titers in male mice four weeks after secondary vaccination but upregulated Il1b gene expression in the spleen. Immulix had a downregulating effect on expression of Cd4 and Foxp3 in ileum only if the mice were fed the diet with increased fat. The diet with increased dietary fat increased Il1b but decreased Cd8a gene expression in the spleen. Immulix and diet affected GM composition significantly. Increased dietary fat content upregulated Lactobacillus animalis but downregulated an unclassified Prevotella spp. Immulix decreased Lactobacillales, Streptococcaceae and Prevotellaceae but increased Bacteroides. It is concluded that in spite of some minor influences on immune cell markers, cytokines and IgG titers Immulix feeding or increased dietary fat content did not have any biologically relevant effects on tetanus vaccine responses in this experiment in mice.
Collapse
Affiliation(s)
- Ida Wang Henriksen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
- Brogaarden Aps, Lynge, Denmark.
| | | | | | | | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| |
Collapse
|
13
|
Ali A, Hamzaid NH, Ismail NAS. The Interplay of Nutriepigenomics, Personalized Nutrition and Clinical Practice in Managing Food Allergy. Life (Basel) 2021; 11:1275. [PMID: 34833150 PMCID: PMC8623511 DOI: 10.3390/life11111275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Food allergy in children has been a common issue due to the challenges of prescribing personalized nutrition with a lack of nutriepigenomics data. This has indeed further influenced clinical practice for appropriate management. While allergen avoidance is still the main principle in food allergy management, we require more information to advance the science behind nutrition, genes, and the immune system. Many researchers have highlighted the importance of personalized nutrition but there is a lack of data on how the decision is made. Thus, this review highlights the relationship among these key players in identifying the solution to the clinical management of food allergy with current nutriepigenomics data. The discussion integrates various inputs, including clinical assessments, biomarkers, and epigenetic information pertaining to food allergy, to curate a holistic and personalized approach to food allergy management in particular.
Collapse
Affiliation(s)
- Adli Ali
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nur Hana Hamzaid
- Dietetic Program & Centre for Rehabilitation and Special Needs Studies (iCaRehab), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
14
|
Dietary supplementation of fructooligosaccharides alleviates enterotoxigenic E. coli-induced disruption of intestinal epithelium in a weaned piglet model. Br J Nutr 2021; 128:1526-1534. [PMID: 34763738 DOI: 10.1017/s0007114521004451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Diarrhea caused by pathogens such as enterotoxigenic E. coli (ETEC) is a serious threat to the health of young animals and human infants. Here, we investigated the protective effect of fructooligosaccharides (FOS) on the intestinal epithelium with ETEC-challenge in a weaned piglet model. Twenty-four weaned piglets were randomly divided into three groups: (1) non-ETEC-challenged control (CON), (2) ETEC-challenged control (ECON), and (3) ETEC challenge + 2.5 g/kg FOS (EFOS). On day 19, the CON pigs were orally infused with sterile culture, while the ECON and EFOS pigs were orally infused with active ETEC (2.5 × 109 colony-forming units). On day 21, pigs were slaughtered to collect venous blood and small intestine. Result showed that the pre-treatment of FOS improved the antioxidant capacity and the integrity of intestinal barrier in the ETEC-challenged pigs without affecting their growth performance. Specifically, comparing with ECON pigs, the level of GSH-Px (glutathione peroxidase) and CAT (catalase) in the plasma and intestinal mucosa of EFOS pigs was increased (P<0.05), and the intestinal barrier marked by ZO-1 and plasmatic DAO was also improved in EFOS pigs. A lower level (P<0.05) of inflammatory cytokines in the intestinal mucosa of EFOS pigs might be involved in the inhibition of TLR4/MYD88/NF-κB pathway. The apoptosis of jejunal cells in EFOS pigs was also lower than that in ECON pigs (P<0.05). Our findings provide convincing evidence of possible prebiotic and protective effect of FOS on the maintenance of intestinal epithelial function under the attack of pathogens.
Collapse
|
15
|
A pilot study about the development and characterization of a Roux en Y gastric bypass model in obese Yucatan minipigs. Sci Rep 2021; 11:20190. [PMID: 34642370 PMCID: PMC8511153 DOI: 10.1038/s41598-021-98575-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 07/21/2021] [Indexed: 11/30/2022] Open
Abstract
Performing the Roux-en-Y gastric bypass (RYGBP) in obese Yucatan minipigs provides an opportunity to explore the mechanisms behind the effects of this surgery in controlled environmental and nutritional conditions. We hypothesized that RYGBP in these minipigs would induce changes at multiple levels, as in obese humans. We sought to characterize RYGBP in a diet-induced obese minipig model, compared with a pair-fed sham group. After inducing obesity with an ad libitum high-fat/high-sugar diet, we performed RYGBP (n = 7) or sham surgery (n = 6). Oral glucose tolerance tests (OGTT) were performed before and after surgery. Histological analyses were conducted to compare the alimentary limb at sacrifice with tissue sampled during RYGBP surgery. One death occurred in the RYGBP group at postoperative day (POD) 3. Before sacrifice, weight loss was the same across groups. GLP-1 secretion (OGTT) was significantly higher at 15, 30 and 60 min at POD 7, and at 30 and 60 min at POD 30 in the RYGBP group. Incremental insulin area under the curve increased significantly after RYGBP (p = 0.02). RYGBP induced extensive remodeling of the alimentary limb. Results show that RYGBP can be safely performed in obese minipigs, and changes mimic those observed in humans.
Collapse
|
16
|
Chen J, Vitetta L. Modulation of Gut Microbiota for the Prevention and Treatment of COVID-19. J Clin Med 2021; 10:2903. [PMID: 34209870 PMCID: PMC8268324 DOI: 10.3390/jcm10132903] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 02/08/2023] Open
Abstract
The gut microbiota is well known to exert multiple benefits on human health including protection from disease causing pathobiont microbes. It has been recognized that healthy intestinal microbiota is of great importance in the pathogenesis of COVID-19. Gut dysbiosis caused by various reasons is associated with severe COVID-19. Therefore, the modulation of gut microbiota and supplementation of commensal bacterial metabolites could reduce the severity of COVID-19. Many approaches have been studied to improve gut microbiota in COVID-19 including probiotics, bacterial metabolites, and prebiotics, as well as nutraceuticals and trace elements. So far, 19 clinical trials for testing the efficacy of probiotics and synbiotics in COVID-19 prevention and treatment are ongoing. In this narrative review, we summarize the effects of various approaches on the prevention and treatment of COVID-19 and discuss associated mechanisms.
Collapse
Affiliation(s)
- Jiezhong Chen
- Medlab Clinical, Research Department, Sydney 2015, Australia;
| | - Luis Vitetta
- Medlab Clinical, Research Department, Sydney 2015, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
17
|
Gut health: The results of microbial and mucosal immune interactions in pigs. ACTA ACUST UNITED AC 2021; 7:282-294. [PMID: 34258416 PMCID: PMC8245825 DOI: 10.1016/j.aninu.2021.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/09/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
There are a large number of microorganisms in the porcine intestinal tract. These microorganisms and their metabolites contribute to intestinal mucosal immunity, which is of great importance to the health of the host. The host immune system can regulate the distribution and composition of intestinal microorganisms and regulate the homeostasis of intestinal flora by secreting a variety of immune effector factors, such as mucin, secretory immunoglobulin A (sIgA), regenerating islet-derived III (RegIII)γ, and defensin. Conversely, intestinal microorganisms can also promote the differentiation of immune cells including regulatory T cells (Treg) and Th17 cells through their specific components or metabolites. Studies have shown that imbalances in the intestinal flora can lead to bacterial translocation and compromised intestinal barrier function, affecting the health of the body. This review focuses on the composition of the pig intestinal flora and the characteristics of intestinal mucosal immunity, discusses the interaction mechanism between the flora and intestinal mucosal immunity, as well as the regulation through fecal microbiota transplantation (FMT), dietary nutritional composition, probiotics and prebiotics of pig intestinal microecology. Finally, this review provides insights into the relationship between intestinal microorganisms and the mucosal immune system.
Collapse
|
18
|
Azad MA, Gao J, Ma J, Li T, Tan B, Huang X, Yin J. Opportunities of prebiotics for the intestinal health of monogastric animals. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:379-388. [PMID: 33364453 PMCID: PMC7750794 DOI: 10.1016/j.aninu.2020.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
The goal of prebiotic applications from different sources is to improve the gut ecosystem where the host and microbiota can benefit from prebiotics. It has already been recognized that prebiotics have potential roles in the gut ecosystem because gut microbiota ferment complex dietary macronutrients and carry out a broad range of functions in the host body, such as the production of nutrients and vitamins, protection against pathogens, and maintenance of immune system balance. The gut ecosystem is very crucial and can be affected by numerous factors consisting of dietary constituents and commensal bacteria. This review focuses on recent scientific evidence, confirming a beneficial effect of prebiotics on animal health, particularly in terms of protection against pathogenic bacteria and increasing the number of beneficial bacteria that may improve epithelial cell barrier functions. It has also been reviewed that modification of the gut ecosystem through the utilization of prebiotics significantly affects the intestinal health of animals. However, the identification and characterization of novel potential prebiotics remain a topical issue and elucidation of the metagenomics relationship between gut microbiota alteration and prebiotic substances is necessary for future prebiotic studies.
Collapse
Affiliation(s)
- Md A.K. Azad
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Gao
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Ma
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Tiejun Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, 410125, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Xingguo Huang
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| |
Collapse
|
19
|
Csernus B, Czeglédi L. Physiological, antimicrobial, intestine morphological, and immunological effects of fructooligosaccharides in pigs. Arch Anim Breed 2020; 63:325-335. [PMID: 32964103 PMCID: PMC7500070 DOI: 10.5194/aab-63-325-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022] Open
Abstract
In pig nutrition, there are some periods when natural alternatives to
antibiotics are more required, such as during suckling and weaning.
Fructooligosaccharides (FOSs) are a group of prebiotics applied as feed
ingredients in animal nutrition since their positive effects on growth
performance, immunological parameters, intestinal microbiota, and gut
morphology are reported. Accordingly, FOS may be candidate molecules to
improve the mentioned properties in pigs. Previous studies defined FOS as inhibiting
the activity of pathogens and increasing the colonization of beneficial
bacteria in the gut, although metabolites of FOS decreased the intestinal pH
value. Beneficial effects on digestive-enzyme activities and on protein
digestion were determined in some studies. All of the three types of FOS
(inulin, oligomeric fructans, and short-chain FOSs) promoted the microbial
composition of the gut by increasing the colonizations of Lactobacillus, Bifidobacterium, and
Prevotella genus. FOS also affected the immune response directly and indirectly and
increased vaccine-specific IgA, serum IgG, and IgE levels. Moreover, FOS
enhanced the activation of T cells and altered the secretions of some
cytokines. Levels of vaccine-specific IgG could not be increased after FOS
supplements. In most cases, FOS modified intestinal morphological
parameters, such as longer villi, villus-height-to-crypt-depth ratio, and
thicker mucosa, which could suggest better absorptive functions. Results are
contradictory on growth performance, which might be influenced by the chemical
structure, the duration, and the dose of FOS, so further studies are
required. This review aims to gather information regarding immunological,
antimicrobial, intestine morphological, and growth performance properties of
fructooligosaccharides in pigs.
Collapse
Affiliation(s)
- Brigitta Csernus
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, 4032, Hungary.,Doctoral School of Animal Science, University of Debrecen, Debrecen, 4032, Hungary
| | - Levente Czeglédi
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, 4032, Hungary
| |
Collapse
|
20
|
Maternal Supplementation of Food Ingredient (Prebiotic) or Food Contaminant (Mycotoxin) Influences Mucosal Immune System in Piglets. Nutrients 2020; 12:nu12072115. [PMID: 32708852 PMCID: PMC7400953 DOI: 10.3390/nu12072115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022] Open
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.
Collapse
|
21
|
Maternal dietary resistant starch does not improve piglet's gut and liver metabolism when challenged with a high fat diet. BMC Genomics 2020; 21:439. [PMID: 32590936 PMCID: PMC7318506 DOI: 10.1186/s12864-020-06854-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Background In the past several years, the use of resistant starch (RS) as prebiotic has extensively been studied in pigs, and this mostly in the critical period around weaning. RS is believed to exert beneficial effects on the gastrointestinal tract mainly due to higher levels of short chain fatty acids (SCFAs) and an improved microbiota profile. In this study, sows were fed digestible starch (DS) or RS during late gestation and lactation and the possible maternal effect of RS on the overall health of the progeny was assessed. Since RS is also described to have a positive effect on metabolism, and to investigate a metabolic programming of the progeny, half of the piglets per maternal diet were assigned to a high fat diet from weaning on to 10 weeks after. Results No bodyweight differences were found between the four experimental piglet groups. The high fat diet did however impact back fat thickness and meat percentage whereas maternal diet did not influence these parameters. The impact of the high fat diet was also reflected in higher levels of serum cholesterol. No major differences in microbiota could be distinguished, although higher levels of SCFA were seen in the colon of piglets born from RS fed sows, and some differences in SCFA production were observed in the caecum, mainly due to piglet diet. RNA-sequencing on liver and colon scrapings revealed minor differences between the maternal diet groups. Merely a handful of genes was differentially expressed between piglets from DS and RS sows, and network analysis showed only one significant cluster of genes in the liver due to the maternal diet that did not point to meaningful biological pathways. However, the high fat diet resulted in liver gene clusters that were significantly correlated with piglet diet, of which one is annotated for lipid metabolic processes. These clusters were not correlated with maternal diet. Conclusions There is only a minor impact of maternal dietary RS on the progeny, reflected in SCFA changes. A high fat diet given to the progeny directly evokes metabolic changes in the liver, without any maternal programming by a RS diet.
Collapse
|
22
|
Ayuso M, Michiels J, Wuyts S, Yan H, Degroote J, Lebeer S, Le Bourgot C, Apper E, Majdeddin M, Van Noten N, Vanden Hole C, Van Cruchten S, Van Poucke M, Peelman L, Van Ginneken C. Short-chain fructo-oligosaccharides supplementation to suckling piglets: Assessment of pre- and post-weaning performance and gut health. PLoS One 2020; 15:e0233910. [PMID: 32502215 PMCID: PMC7274435 DOI: 10.1371/journal.pone.0233910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/14/2020] [Indexed: 01/04/2023] Open
Abstract
Farmers face difficulties in redeeming their investment in larger litter sizes since this comes with larger litter heterogenicity, lower litter resilience and risk of higher mortality. Dietary oligosaccharides, given to the sow, proved beneficial for the offspring’s performance. However, giving oligosaccharides to the suckling piglet is poorly explored. Therefore, this field trial studied the effect of dietary short-chain fructo-oligosaccharides (scFOS; 1g/day; drenched) supplementation to low (LBW, lower quartile), normal (NBW, two intermediate quartiles) and high (HBW, upper quartile) birth weight piglets from birth until 7 or 21 days of age. Performance parameters, gut microbiome and short-chain fatty acids profile of feces and digesta were assessed at birth (d 0), d 7, weaning (d 21.5) and 2 weeks post-weaning (d 36.5). Additional parameters reflecting gut health (intestinal integrity and morphology, mucosal immune system) were analysed at d 36.5. Most parameters changed with age or differed with the piglet’s birth weight. Drenching with scFOS increased body weight by 1 kg in NBW suckling piglets and reduced the post-weaning mortality rate by a 100%. No clear difference in the IgG level, the microbiota composition and fermentative activity between the treatment groups was observed. Additionnally, intestinal integrity, determined by measuring intestinal permeability and regenerative capacity, was similar between the treatment groups. Also, intestinal architecture (villus lenght, crypt depth) was not affected by scFOS supplementation. The density of intra-epithelial lymphocytes and the expression profiles (real-time qPCR) for immune system-related genes (IL-10, IL-1ß, IL-6, TNFα and IFNγ) were used to assess mucosal immunity. Only IFNγ expression, was upregulated in piglets that received scFOS for 7 days. The improved body weight and the reduced post-weaning mortality seen in piglets supplemented with scFOS support the view that scFOS positively impact piglet’s health and resilience. However, the modes of action for these effects are not yet fully elucidated and its potential to improve other performance parameters needs further investigation.
Collapse
Affiliation(s)
- Miriam Ayuso
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
- * E-mail:
| | - Joris Michiels
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sander Wuyts
- Department of Bioengineering, Faculty of Sciences, University of Antwerp, Wilrijk, Belgium
| | - Honglin Yan
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Jeroen Degroote
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sarah Lebeer
- Department of Bioengineering, Faculty of Sciences, University of Antwerp, Wilrijk, Belgium
| | | | | | - Maryam Majdeddin
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Noémie Van Noten
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Charlotte Vanden Hole
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Steven Van Cruchten
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Mario Van Poucke
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chris Van Ginneken
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
23
|
Short-chain fructo-oligosaccharides alleviates oxidized oil-induced intestinal dysfunction in piglets associated with the modulation of gut microbiota. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
24
|
Ramírez-Alarcón K, Sánchez-Agurto Á, Lamperti L, Martorell M. Epigenetics, Maternal Diet and Metabolic Programming. ACTA ACUST UNITED AC 2019. [DOI: 10.2174/1874196701907010045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background:
The maternal environment influences embryonic and fetal life. Nutritional deficits or excesses alter the trajectory of fetus/offspring’s development. The concept of “developmental programming” and “developmental origins of health and disease” consists of the idea that maternal diet may remodel the genome and lead to epigenetic changes. These changes are induced during early life, permanently altering the phenotype in the posterior adult stage, favoring the development of metabolic diseases such as obesity, dyslipidemia, hypertension, hyperinsulinemia, and metabolic syndrome. In this review, it is aimed to overview epigenetics, maternal diet and metabolic programming factors and determine which of these might affect future generations.
Scope and Approach:
Nutrients interfere with the epigenome by influencing the supply and use of methyl groups through DNA transmethylation and demethylation mechanisms. They also influence the remodeling of chromatin and arginine or lysine residues at the N-terminal tails of histone, thus altering miRNA expression. Fats, proteins, B vitamins and folates act as important cofactors in methylation processes. The metabolism of carbon in the methyl groups of choline, folic acid and methionine to S-Adenosyl Methionine (SAM), acts as methyl donors to methyl DNA, RNA, and proteins. B-complex vitamins are important since they act as coenzymes during this process.
Key Findings and Conclusion:
Nutrients, during pregnancy, potentially influence susceptibility to diseases in adulthood. Additionally, the deficit or excess of nutrients alter the epigenetic machinery, affecting genes and influencing the genome of the offspring and therefore, predisposing the development of chronic diseases in adults.
Collapse
|
25
|
Arnaud AP, Rome V, Richard M, Formal M, David-Le Gall S, Boudry G. Post-natal co-development of the microbiota and gut barrier function follows different paths in the small and large intestine in piglets. FASEB J 2019; 34:1430-1446. [PMID: 31914707 DOI: 10.1096/fj.201902514r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Gut microbiota and intestinal barrier co-develop after birth, establishing a homeostatic state whereby mucosal cells cohabit with commensal bacteria. We hypothesized that this post-natal co-development follows different timings depending on the intestinal site considered. Jejunal, ileal, and colonic luminal contents and mucosa were sampled in suckling piglets at post-natal day (PND) 0, 2, 7, 14, and 28. Jejunal, ileal, and colonic luminal microbiota (evaluated by 16S DNA sequencing followed by beta-diversity analysis) clustered at PND2 but colonic microbiota diverge afterwards (P < .05). Mucosal permeability, evaluated in Ussing chambers, increased with age in the jejunum and ileum (P < .05) but not the colon. Expression of pattern recognition receptor (PRR) exhibited different patterns (gradual or sharp increase, decrease, or no change with age, P < .05) depending on PRR and intestinal site considered. Principal component analysis of mucosa data revealed clear clustering of colonic samples, irrespective of the age and clustering of jejunal and ileal samples, with gradual changes with age. Correlation analysis highlighted three families correlating with mucosal parameters: Enterobacteriaceae in the jejunum, Peptostreptococcaceae in the ileum, and Micrococcaceae in the colon. In conclusion, small and large intestine display close microbiota composition early in life but distinct mucosal phenotype and follow very different post-natal development.
Collapse
Affiliation(s)
- Alexis Pierre Arnaud
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France.,Service de chirurgie pédiatrique, CHU rennes, University of Rennes 1, Rennes, France
| | - Véronique Rome
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | - Marion Richard
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | - Michèle Formal
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | | | - Gaëlle Boudry
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| |
Collapse
|
26
|
Val-Laillet D. Review: Impact of food, gut-brain signals and metabolic status on brain activity in the pig model: 10 years of nutrition research using in vivo brain imaging. Animal 2019; 13:2699-2713. [PMID: 31354119 DOI: 10.1017/s1751731119001745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The purpose of this review is to offer a panorama on 10 years of nutrition research using in vivo brain imaging in the pig model. First, we will review some work describing the brain responses to food signals, including basic tastants such as sweet and bitter at both oral and visceral levels, as well as conditioned preferred and aversive flavours. Second, we will have a look at the impact of weight gain and obesity on brain metabolism and functional responses, drawing the parallel with obese human patients. Third, we will evoke the concept of the developmental origins of health and diseases, and how the pig model can shed light on the importance of maternal nutrition during gestation and lactation for the development of the gut-brain axis and adaptation abilities of the progeny to nutritional environments. Finally, three examples of preventive or therapeutic strategies will be introduced: the use of sensory food ingredients or pre-, pro-, and postbiotics to improve metabolic and cognitive functions; the implementation of chronic vagus nerve stimulation to prevent weight gain and glucose metabolism alterations; and the development of bariatric surgery in the pig model for the understanding of its complex mechanisms at the gut-brain level. A critical conclusion will brush the limitations of neurocognitive studies in the pig model and put in perspective the rationale and ethical concerns underlying the use of pig experimentation in nutrition and neurosciences.
Collapse
Affiliation(s)
- D Val-Laillet
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, St Gilles, France
| |
Collapse
|
27
|
Opriessnig T, Karuppannan AK, Beckler D, Ait-Ali T, Cubas-Atienzar A, Halbur PG. Bacillus pumilus probiotic feed supplementation mitigates Lawsonia intracellularis shedding and lesions. Vet Res 2019; 50:85. [PMID: 31640784 PMCID: PMC6806562 DOI: 10.1186/s13567-019-0696-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/11/2019] [Indexed: 01/18/2023] Open
Abstract
The causative agent of ileitis, Lawsonia intracellularis, is commonly associated with diarrhea and reduced weight gain in growing pigs. The effect of in-feed probiotics on L. intracellularis infection dynamics was evaluated. In brief, 70 2.5-week-old-pigs were randomly divided into six groups with 10–20 pigs each. All pigs were fed an age appropriate base ration for the duration of the study, which was supplemented with one of three Bacillus strains including B. amyloliquefaciens (T01), B. licheniformis (T02) and B. pumilus (T03). Another group was orally vaccinated with a commercial live L. intracellularis vaccine (VAC) at 3 weeks of age. At 7 weeks of age, T01-LAW, T02-LAW, T03-LAW, VAC-LAW and the POS-CONTROL groups were challenged with L. intracellularis while the NEG-CONTROL pigs were not challenged. All pigs were necropsied 16 days later. By the time of inoculation, all VAC-LAW pigs had seroconverted and at necropsy 10–65% of the pigs in all other challenged groups were also seropositive. The results indicate a successful L. intracellularis challenge with highest bacterial DNA levels in POS-CONTROL pigs, VAC-LAW pigs and T01-LAW pigs. There was a delay in onset of shedding in T02-LAW and T03-LAW groups, which was reflected in less severe macroscopic and microscopic lesions, reduced intralesional L. intracellularis antigen levels and a lower area under the curve for bacterial shedding. Under the study conditions, two of the probiotics tested suppressed L. intracellularis infection. The obtained findings show the potential of probiotics in achieving antibiotic-free control of L. intracellularis.
Collapse
Affiliation(s)
- Tanja Opriessnig
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK. .,Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| | - Anbu K Karuppannan
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | | | - Tahar Ait-Ali
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Ana Cubas-Atienzar
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Patrick G Halbur
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| |
Collapse
|
28
|
Tanno H, Fujii T, Ose R, Hirano K, Tochio T, Endo A. Characterization of fructooligosaccharide-degrading enzymes in human commensal Bifidobacterium longum and Anaerostipes caccae. Biochem Biophys Res Commun 2019; 518:294-298. [PMID: 31420164 DOI: 10.1016/j.bbrc.2019.08.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/08/2019] [Indexed: 12/22/2022]
Abstract
Kestose and nystose are short chain fructooligosaccharides (scFOSs) with degrees of polymerization of 3 and 4, respectively. A previous study revealed that these scFOSs have different growth stimulation properties against two human commensals, i.e. Bifidobacterium longum subsp. longum and butyrogenic Anaerostipes caccae. The present study characterized genes involved in FOS metabolism in these organisms. A. caccae possesses a single gene cluster consisting of four genes, including a gene encoding the putative FOS degradation enzyme sucrose-6-phosphate hydrolase (S6PH). B. longum possesses two gene clusters consisting of three genes each, including genes encoding β-fructofuranosidase (CscA) and sucrose phosphorylase (ScrP). In A. caccae, the genes were highly transcribed in cells cultured with sucrose or kestose but poorly in cells cultured with glucose or nystose. Heterologously expressed S6PH degraded sucrose and kestose but not nystose. In B. longum, transcription of the genes was high in cells cultured with sucrose or kestose but was poor or not detected in cells cultured with glucose or nystose. Heterologously expressed CscA degraded sucrose, kestose and nystose, but ScrP degraded only sucrose. These data suggested that the different growth stimulation activities of kestose and nystose are due to different substrate specificities of FOS degradation enzymes in the organisms and/or induction activity of the genes in the two scFOSs. This is the first study characterizing the FOS metabolism at the transcriptional level and substrate-specificity of the degradation enzyme in butyrogenic human gut anaerobes.
Collapse
Affiliation(s)
- Hiroki Tanno
- Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, 099-2493, Hokkaido, Japan
| | - Tadashi Fujii
- B Food Science Co., Ltd., 24-12 Kitahama, Chita, 478-0046, Aichi, Japan
| | - Riichi Ose
- Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, 099-2493, Hokkaido, Japan
| | - Katsuaki Hirano
- B Food Science Co., Ltd., 24-12 Kitahama, Chita, 478-0046, Aichi, Japan
| | - Takumi Tochio
- B Food Science Co., Ltd., 24-12 Kitahama, Chita, 478-0046, Aichi, Japan
| | - Akihito Endo
- Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, 099-2493, Hokkaido, Japan.
| |
Collapse
|
29
|
San Andres JV, Mastromano GA, Li Y, Tran H, Bundy JW, Miller PS, Burkey TE. The effects of prebiotics on growth performance and in vitro immune biomarkers in weaned pigs. Transl Anim Sci 2019; 3:1315-1325. [PMID: 32704894 PMCID: PMC7200398 DOI: 10.1093/tas/txz129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/25/2019] [Indexed: 01/01/2023] Open
Abstract
The objective of the experiment was to investigate the effects of prebiotics in nursery pigs on growth performance and immune biomarkers. Sixty-four weaned pigs (31 ± 1 d; BW 8 ± 0.1 kg) of mixed gender were housed (4 pigs/pen) in an environmentally controlled nursery with ad libitum access to feed and water over a 35-d study. Pigs were randomly assigned to one of four treatments: control (53% corn, 32% SBM, 7% fishmeal, 8% others), control + 2.5% GroBiotic-S (GS), control + 0.05% chicory (CL), or control + 0.5% chicory (CH). Feeders and pigs were weighed weekly. On day 21, blood samples were obtained from three pigs/treatment for collection of peripheral blood mononuclear cells (PBMC). Isolated PBMC were cultured and subsequently challenged with lipopolysaccharide (LPS; 20 ng/mL). Cell culture supernatants were collected for quantification of the pro- and anti-inflammatory cytokines, interleukin (IL)-8 and IL-10, respectively. Dietary treatment had no effect on BW. At days 28 to 35, pigs fed GS (790 ± 15 g), CL (704 ± 15 g), or CH (692 ± 15 g) had greater (P < 0.05) ADG compared with control (643 ± 15 g) pigs. In addition, overall (days 0–35), pigs fed GS (823 ± 18 g), CL (783 ± 18 g), or CH (782 ± 18 g) had greater (P < 0.05) ADFI compared with control, and ADFI for GS-fed pigs was greater (P < 0.05) than either CL or CH. There was no difference in G:F among treatments. In vitro LPS challenge increased (P < 0.05) IL-8 secretion from PBMC isolated from CL (23,731 ± 3,221 pg/mL) pigs compared with control (10,061 ± 3,221 pg/mL) and CH (12,411 ± 3,221 pg/mL) pigs. Secretion of IL-10 from PBMC isolated from CL (63 ± 9 pg/mL) pigs was greater (P < 0.05) compared with control (22 ± 9 pg/mL) pigs and tended (P < 0.1) to be greater compared with CH (34 ± 9 pg/mL) pigs. Results indicate that inclusion of prebiotics in nursery pig diets has positive effects on growth performance and may have immunomodulatory effects (in vitro) on cells isolated from prebiotic-fed pigs.
Collapse
Affiliation(s)
- Joice V San Andres
- Department of Animal Science, University of Nebraska, Lincoln, NE.,Department of Animal Science, Central Luzon State University, Philippines
| | | | - Yanshuo Li
- Department of Animal Science, University of Nebraska, Lincoln, NE
| | - Huyen Tran
- Department of Animal Science, University of Nebraska, Lincoln, NE
| | - Justin W Bundy
- Department of Animal Science, University of Nebraska, Lincoln, NE
| | - Phillip S Miller
- Department of Animal Science, University of Nebraska, Lincoln, NE
| | - Thomas E Burkey
- Department of Animal Science, University of Nebraska, Lincoln, NE
| |
Collapse
|
30
|
Zhao W, Yuan M, Li P, Yan H, Zhang H, Liu J. Short-chain fructo-oligosaccharides enhances intestinal barrier function by attenuating mucosa inflammation and altering colonic microbiota composition of weaning piglets. ITALIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1080/1828051x.2019.1612286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Meng Yuan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Pengcheng Li
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Honglin Yan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
31
|
Jha R, Fouhse JM, Tiwari UP, Li L, Willing BP. Dietary Fiber and Intestinal Health of Monogastric Animals. Front Vet Sci 2019; 6:48. [PMID: 30886850 PMCID: PMC6409295 DOI: 10.3389/fvets.2019.00048] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/06/2019] [Indexed: 01/10/2023] Open
Abstract
Animal performance, feed efficiency, and overall health are heavily dependent on gut health. Changes in animal production systems and feed regulations away from the use of antibiotic growth promoters (AGP) have necessitated the identification of strategies to optimize gut health in novel and effective ways. Among alternatives to AGP, the inclusion of dietary fibers (DF) in monogastric diets has been attempted with some success. Alternative feedstuffs and coproducts are typically rich in fiber and can be used in the diets to reduce feed costs and optimize gut health. DF are naturally occurring compounds with a diverse composition and are present in all plant-based feedstuffs. DF stimulate the growth of health-promoting gut bacteria, are fermented in the distal small intestine and large intestine to short-chain fatty acids and have beneficial effects on the immune system. Maternal DF supplementation is one novel strategy suggested to have a beneficial programming effect on the microbial and immune development of their offspring. One mechanism by which DF improves gut health is through maintenance of an anaerobic intestinal environment that subsequently prevents facultative anaerobic pathogens from flourishing. Studies with pigs and poultry have shown that fermentation characteristics and their beneficial effects on gut health vary widely based on type, form, and the physico-chemical properties of the DF. Therefore, it is important to have information on the different types of DF and their role in optimizing gut health. This review will provide information and updates on different types of DF used in monogastric nutrition and its contribution to gut health including microbiology, fermentation characteristics, and innate and adaptive immune responses.
Collapse
Affiliation(s)
- Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Janelle M. Fouhse
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Utsav P. Tiwari
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Linge Li
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Benjamin P. Willing
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
32
|
Brugman S, Ikeda-Ohtsubo W, Braber S, Folkerts G, Pieterse CMJ, Bakker PAHM. A Comparative Review on Microbiota Manipulation: Lessons From Fish, Plants, Livestock, and Human Research. Front Nutr 2018; 5:80. [PMID: 30234124 PMCID: PMC6134018 DOI: 10.3389/fnut.2018.00080] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
During recent years the impact of microbial communities on the health of their host (being plants, fish, and terrestrial animals including humans) has received increasing attention. The microbiota provides the host with nutrients, induces host immune development and metabolism, and protects the host against invading pathogens (1-6). Through millions of years of co-evolution bacteria and hosts have developed intimate relationships. Microbial colonization shapes the host immune system that in turn can shape the microbial composition (7-9). However, with the large scale use of antibiotics in agriculture and human medicine over the last decades an increase of diseases associated with so-called dysbiosis has emerged. Dysbiosis refers to either a disturbed microbial composition (outgrowth of possible pathogenic species) or a disturbed interaction between bacteria and the host (10). Instead of using more antibiotics to treat dysbiosis there is a need to develop alternative strategies to combat disturbed microbial control. To this end, we can learn from nature itself. For example, the plant root (or "rhizosphere") microbiome of sugar beet contains several bacterial species that suppress the fungal root pathogen Rhizoctonia solani, an economically important fungal pathogen of this crop (11). Likewise, commensal bacteria present on healthy human skin produce antimicrobial molecules that selectively kill skin pathogen Staphylococcus aureus. Interestingly, patients with atopic dermatitis (inflammation of the skin) lacked antimicrobial peptide secreting commensal skin bacteria (12). In this review, we will give an overview of microbial manipulation in fish, plants, and terrestrial animals including humans to uncover conserved mechanisms and learn how we might restore microbial balance increasing the resilience of the host species.
Collapse
Affiliation(s)
- Sylvia Brugman
- Cell Biology and Immunology Group, Animal Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Corné M. J. Pieterse
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| | - Peter A. H. M. Bakker
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
33
|
Karuppannan AK, Opriessnig T. Lawsonia intracellularis: Revisiting the Disease Ecology and Control of This Fastidious Pathogen in Pigs. Front Vet Sci 2018; 5:181. [PMID: 30140680 PMCID: PMC6095029 DOI: 10.3389/fvets.2018.00181] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022] Open
Abstract
Lawsonia intracellularis is an anaerobic obligate intracellular bacterium infecting the small intestine and infrequently also the large intestine of pigs and other animals including hamsters and horses. The infection is characterized by proliferation, hemorrhage, necrosis, or any combination commonly referred to as "ileitis," affecting the health and production efficacy of farmed pigs. Despite decades of research on this pathogen, the pathogenesis and virulence factors of this organism are not clearly known. In pigs, prophylaxis against L. intracellularis infection is achieved by either administration of subtherapeutic levels of in-feed antibiotic growth promoters or vaccination. While the former approach is considered to be effective in L. intracellularis control, potential regulations on subtherapeutic antibiotics in many countries in the near future may necessitate alternative approaches. The potential of manipulating the gut microbiome of pigs with feed ingredients or supplements to control L. intracellularis disease burden is promising based on the current understanding of the porcine gut microbiome in general, as well as preliminary insights into the disease ecology of L. intracellularis infection accrued over the last 30 years.
Collapse
Affiliation(s)
- Anbu K. Karuppannan
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Tanja Opriessnig
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
34
|
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. FASEB J 2018; 33:301-313. [DOI: 10.1096/fj.201800108r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Cindy Le Bourgot
- Tereos Marckolsheim France
- INRAINSERMUniv RennesNutrition Metabolisms and CancerNuMeCan Rennes France
| | | | | | | | - Armelle Cahu
- INRAINSERMUniv RennesNutrition Metabolisms and CancerNuMeCan Rennes France
| | | | | | | | - Sophie Blat
- INRAINSERMUniv RennesNutrition Metabolisms and CancerNuMeCan Rennes France
| |
Collapse
|
35
|
Gautier Y, Luneau I, Coquery N, Meurice P, Malbert CH, Guerin S, Kemp B, Bolhuis JE, Clouard C, Le Huërou-Luron I, Blat S, Val-Laillet D. Maternal Western diet during gestation and lactation modifies adult offspring's cognitive and hedonic brain processes, behavior, and metabolism in Yucatan minipigs. FASEB J 2018; 32:fj201701541. [PMID: 29897815 DOI: 10.1096/fj.201701541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study explores the long-term effects of exposure to a maternal Western diet (WD) vs. standard diet (SD) in the Yucatan minipig, on the adult progeny at lean status ( n = 32), and then overweight status. We investigated eating behavior, cognitive abilities, brain basal glucose metabolism, dopamine transporter availability, microbiota activity, blood lipids, and glucose tolerance. Although both groups demonstrated similar cognitive abilities in a holeboard test, WD pigs expressed a higher stress level than did SD pigs (immobility, P < 0.05) and lower performance in an alley maze ( P = 0.06). WD pigs demonstrated lower dopamine transporter binding potential in the hippocampus and parahippocampal cortex ( P < 0.05 for both), as well as a trend in putamen ( P = 0.07), associated with lower basal brain activity in the prefrontal cortex and nucleus accumbens ( P < 0.05) compared with lean SD pigs. Lean WD pigs demonstrated a lower glucose tolerance than did SD animals (higher glucose peak, P < 0.05) and a tendency to a higher incremental area under the curve of insulin from 0 to 30 minutes after intravenous glucose injection ( P < 0.1). Both groups developed glucose intolerance with overweight, but WD animals were less impacted than SD animals. These results demonstrate that maternal diet shaped the offspring's brain functions and cognitive responses long term, even after being fed a balanced diet from weaning, but behavioral effects were only revealed in WD pigs under anxiogenic situation; however, WD animals seemed to cope better with the obesogenic diet from a metabolic standpoint.-Gautier, Y., Luneau, I., Coquery, N., Meurice, P., Malbert, C.-H., Guerin, S., Kemp, B., Bolhuis, J. E., Clouard, C., Le Huërou-Luron, I., Blat, S., Val-Laillet, D. Maternal Western diet during gestation and lactation modifies adult offspring's cognitive and hedonic brain processes, behavior, and metabolism in Yucatan minipigs.
Collapse
Affiliation(s)
- Yentl Gautier
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Isabelle Luneau
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Nicolas Coquery
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Paul Meurice
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | | | - Sylvie Guerin
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Bas Kemp
- Department of Animal Sciences, Wageningen University and Research, Adaptation Physiology Group, Wageningen, The Netherlands
| | - J Elizabeth Bolhuis
- Department of Animal Sciences, Wageningen University and Research, Adaptation Physiology Group, Wageningen, The Netherlands
| | - Caroline Clouard
- Department of Animal Sciences, Wageningen University and Research, Adaptation Physiology Group, Wageningen, The Netherlands
| | - Isabelle Le Huërou-Luron
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - Sophie Blat
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| | - David Val-Laillet
- INRA INSERM, Univ Rennes, Nutrition Metabolisms and Cancer (NuMeCan), Rennes Saint-Gilles, France
| |
Collapse
|
36
|
The role of gut microbiota in the effects of maternal obesity during pregnancy on offspring metabolism. Biosci Rep 2018; 38:BSR20171234. [PMID: 29208770 PMCID: PMC5897743 DOI: 10.1042/bsr20171234] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 02/07/2023] Open
Abstract
Obesity is considered a global epidemic. Specifically, obesity during pregnancy programs an increased risk of the offspring developing metabolic disorders in addition to the adverse effects on the mother per se Large numbers of human and animal studies have demonstrated that the gut microbiota plays a pivotal role in obesity and metabolic diseases. Similarly, maternal obesity during pregnancy is associated with alterations in the composition and diversity of the intestine microbial community. Recently, the microbiota in the placenta, amniotic fluid, and meconium in healthy gestations has been investigated, and the results supported the "in utero colonization hypothesis" and challenged the traditional "sterile womb" that has been acknowledged worldwide for more than a century. Thus, the offspring microbiota, which is crucial for the immune and metabolic function and further health in the offspring, might be established prior to birth. As a detrimental intrauterine environment, maternal obesity influences the microbial colonization and increases the risk of metabolic diseases in offspring. This review discusses the role of the microbiota in the impact of maternal obesity during pregnancy on offspring metabolism and further analyzes related probiotic or prebiotic interventions to prevent and treat obesity and metabolic diseases.
Collapse
|