1
|
Wang L, Lv Z, Ning X, Yue Z, Wang P, Liu C, Jin S, Li X, Yin Q, Zhu Q, Chang J. The effects of compound probiotics on production performance, rumen fermentation and microbiota of Hu sheep. Front Vet Sci 2024; 11:1440432. [PMID: 39545259 PMCID: PMC11560882 DOI: 10.3389/fvets.2024.1440432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 10/03/2024] [Indexed: 11/17/2024] Open
Abstract
Fungal probiotics have the potential as feed additives, but less has been explored in ruminant feed up to date. This study aimed to determine the effect of compound probiotics (CPs) with Aspergillus oryzae 1, Aspergillus oryzae 2 and Candida utilis on Hu sheep's growth performance, rumen fermentation and microbiota. A total of 120 male Hu sheep, aged 2 months and with the body weight of 16.95 ± 0.65 kg were divided into 4 groups. Each group consisted of 5 replicates, with 6 sheep per replicate. Group A was the control group fed with the basal diet. Group B, C and D was supplemented with the basal diet by adding 400, 800 and 1,200 grams per ton (g/t) CPs, respectively. The feeding trial lasted for 60 days after a 10-day adaptation period. The results showed that the average daily gain (ADG) of sheep in the CPs groups were significantly higher, the feed/gain were significantly lower than those in group A in the later stage and the overall period. The addition of CPs increased the economic benefit. The levels of CD4+ and the CD4+/CD8+ ratio in the CPs groups were higher than those in Group A. The levels of GSH, IgG, IL-2, IL-6, and IFN-γ in group C were significantly elevated compared with group A. Group B showed a significant increase in rumen NH3-N and cellulase activity. There was no difference in VFAs content between group A and group B, however, with the increasing addition of CPs, the butyric acid and isobutyric acid content tended to decrease. The rumen microbiota analysis indicated that the CPs addition increased the Firmicutes and Proteobacteria abundances, decreased the Bacteroidetes abundance. The correlation analysis showed that Prevotella was negatively correlated with ADG, and the addition of 400 CPs in group B reduced Prevotella's relative abundance, indicating CPs increased sheep growth by decreasing Prevotella abundance. The CPs addition reduced caspase-3, NF-κB and TNF-α expression in liver, jejunum and rumen tissues. In conclusion, the addition of CPs increased the sheep production performance, reduced inflammation, improved rumen and intestinal health. Considering the above points and economic benefits, the optimal addition of CPs as an additive for Hu sheep is 800 g/t.
Collapse
Affiliation(s)
- Lijun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhanqi Lv
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | | | - Zhiguang Yue
- Henan Anjin Biotechnology Co., Ltd., Xinxiang, China
| | - Ping Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Chaoqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Sanjun Jin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Xinxin Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Qingqiang Yin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Qun Zhu
- Henan Delin Biological Product Co., Ltd., Xinxiang, China
| | - Juan Chang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
2
|
Iatcu OC, Hamamah S, Covasa M. Harnessing Prebiotics to Improve Type 2 Diabetes Outcomes. Nutrients 2024; 16:3447. [PMID: 39458444 PMCID: PMC11510484 DOI: 10.3390/nu16203447] [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: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiota, a complex ecosystem of microorganisms in the human gastrointestinal tract (GI), plays a crucial role in maintaining metabolic health and influencing disease susceptibility. Dysbiosis, or an imbalance in gut microbiota, has been linked to the development of type 2 diabetes mellitus (T2DM) through mechanisms such as reduced glucose tolerance and increased insulin resistance. A balanced gut microbiota, or eubiosis, is associated with improved glucose metabolism and insulin sensitivity, potentially reducing the risk of diabetes-related complications. Various strategies, including the use of prebiotics like inulin, fructooligosaccharides, galactooligosaccharides, resistant starch, pectic oligosaccharides, polyphenols, β-glucan, and Dendrobium officinale have been shown to improve gut microbial composition and support glycemic control in T2DM patients. These prebiotics can directly impact blood sugar levels while promoting the growth of beneficial bacteria, thus enhancing glycemic control. Studies have shown that T2DM patients often exhibit a decrease in beneficial butyrate-producing bacteria, like Roseburia and Faecalibacterium, and an increase in harmful bacteria, such as Escherichia and Prevotella. This review aims to explore the effects of different prebiotics on T2DM, their impact on gut microbiota composition, and the potential for personalized dietary interventions to optimize diabetes management and improve overall health outcomes.
Collapse
Affiliation(s)
- Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
3
|
Yoshikawa S, Itaya K, Hoshina R, Tashiro Y, Suda W, Cho Y, Matsuura M, Shindo C, Ito T, Hattori M, Miyamoto H, Kodama H. Thermophile-fermented feed modulates the gut microbiota related to lactate metabolism in pigs. J Appl Microbiol 2024; 135:lxae254. [PMID: 39333026 DOI: 10.1093/jambio/lxae254] [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: 04/16/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
AIMS Extracts of fermented feed obtained via fermentation of marine animal resources with thermophilic Bacillaceae bacteria increase the fecundity of livestock. The intestinal bacterial profiles in response to long-term administration of this extract to pigs were investigated. METHODS AND RESULTS Half of a swine farm was supplied with potable water containing an extract of fermented feed for more than 2 years, whereas the other half was supplied with potable water without the extract. Feces from 6-month-old pigs rearing in these two areas were collected. 16S rRNA gene sequencing and isolation of lactic acid bacteria revealed an increase in the D/L-lactate-producing bacterium, Lactobacillus amylovorus, and a decrease in several members of Clostridiales following administration of fermented feed. A lactate-utilizing bacterium, Megasphaera elsdenii, was more abundant in the feces of pigs in the fermented feed group. All representative isolates of M. elsdenii showed rapid utilization of D-lactate relative to L-lactate, and butyrate and valerate were the main products. CONCLUSION The probiotic effect of fermented feed is associated with the modulation of lactate metabolism in the digestive organs of pigs.
Collapse
Affiliation(s)
- Shota Yoshikawa
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| | - Kaede Itaya
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| | - Ryo Hoshina
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| | - Yukihiro Tashiro
- Institute of Advanced Study, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
- Laboratory of Functional Food Design, Department of Functional Metabolic Design, Bio-Architecture Center, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Yuichiro Cho
- Department of Anatomy and Physiological Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Makiko Matsuura
- Sermas Co., Ltd., 4-3-5 Onitaka, Ichikawa City, Chiba 272-0015, Japan
| | - Chie Shindo
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Toshiyuki Ito
- Keiyo Gas Energy Solution Co. Ltd., 4-3-5 Onitaka, Ichikawa City, Chiba 272-0015, Japan
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
- School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Hirokuni Miyamoto
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
- Sermas Co., Ltd., 4-3-5 Onitaka, Ichikawa City, Chiba 272-0015, Japan
- Japan Eco-science (Nikkan Kagaku) Co. Ltd., 11-1-211 Shiomigaokacho, Chiba City, Chiba 260-0034, Japan
| | - Hiroaki Kodama
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| |
Collapse
|
4
|
Ferreres-Serafini L, Martín-Orúe SM, Sadurní M, Jiménez J, Moreno-Muñoz JA, Castillejos L. Supplementing infant milk formula with a multi-strain synbiotic and osteopontin enhances colonic microbial colonization and modifies jejunal gene expression in lactating piglets. Food Funct 2024; 15:6536-6552. [PMID: 38807503 DOI: 10.1039/d4fo00489b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
A total of ninety-six weaned piglets were assigned to four dietary treatments in a 2 × 2 design. The treatments included: a standard milk formula (CTR); CTR + probiotics (6.4 × 108 cfu L-1Bifidobacterium longum subsp. infantis CECT 7210 and 1.1 × 108 cfu L-1Lactobacillus rhamnosus NH001) + prebiotics (galacto-oligosaccharides 4.36 g L-1 and human-milk-oligosaccharide 0.54 g L-1) (SYN); CTR + osteopontin (0.43 g L-1) (OPN); and CTR + SYN + OPN (CON). Daily records including feed intake, body weight, and clinical signs, were maintained throughout the 15-day trial. At the end of the study samples from blood, digestive content, and gut tissues were collected to determine serum TNF-α, intestinal fermentative activity (SCFA and ammonia), colonic microbiota (16S rRNA Illumina-MiSeq), histomorphology, and jejunal gene expression (Open-Array). No statistical differences were found in weight gain; however, the animals supplemented with osteopontin exhibited higher feed intake. In terms of clinical signs, synbiotic supplementation led to a shorter duration of diarrhoea episodes. Regarding gut health, the sequenced faecal microbiota revealed better control of potentially dysbiotic bacteria with the CON diet at day 15. In the colon compartment, a significant increase in SCFA concentration, a decrease in ammonia concentration, and a significant decrease in intraepithelial lymphocyte counts were particularly observed in CON animals. The supplemented diets were also associated with modified jejunal gene expression. The synbiotic combination was characterized by the upregulation of genes related to intestinal maturation (ALPI, SI) and nutrient transport (SLC13A1, SLC15A1, SLC5A1, SLC7A8), and the downregulation of genes related to the response to pathogens (GBP1, IDO, TLR4) or the inflammatory response (IDO, IL-1β, TGF-β1). Osteopontin promoted the upregulation of a digestive function gene (GCG). Correlational analysis between the microbiota population and various intestinal environmental factors (SCFA concentration, histology, and gene expression) proposes mechanisms of communication between the gut microbiota and the host. In summary, these results suggest an improvement in the colonic colonization process and a better modulation of the immune response when milk formula is supplemented with the tested synbiotic combined with osteopontin, benefiting from a synergistic effect.
Collapse
Affiliation(s)
- Laia Ferreres-Serafini
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Susana Mª Martín-Orúe
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Meritxell Sadurní
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Jesús Jiménez
- Laboratorios Ordesa S.L., Parc Científic de Barcelona, C/Baldiri Reixac 15-21, 08028 Barcelona, Spain.
| | - José Antonio Moreno-Muñoz
- Laboratorios Ordesa S.L., Parc Científic de Barcelona, C/Baldiri Reixac 15-21, 08028 Barcelona, Spain.
| | - Lorena Castillejos
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| |
Collapse
|
5
|
Zhao XQ, Wang L, Zhu CL, Xue XH, Xia XJ, Wu XL, Wu YD, Liu SQ, Zhang GP, Bai YY, Fotina H, Hu JH. Oral Administration of the Antimicrobial Peptide Mastoparan X Alleviates Enterohemorrhagic Escherichia coli-Induced Intestinal Inflammation and Regulates the Gut Microbiota. Probiotics Antimicrob Proteins 2024; 16:138-151. [PMID: 36515889 DOI: 10.1007/s12602-022-10013-x] [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] [Accepted: 11/18/2022] [Indexed: 12/15/2022]
Abstract
The gut microbiota plays an important role in intestinal immune system development and in driving inflammation. Antibiotic administration for therapeutic purposes causes an imbalance in the gut microbiota. Antimicrobial peptides can regulate the gut microbiota and maintain intestinal homeostasis. The aim of this study was to investigate the anti-inflammatory effects and regulation of the gut microbiota by the orally administered antimicrobial peptide mastoparan X (MPX). In this study, Escherichia coli was used to induce intestinal inflammation, and the results showed that MPX+ E. coli alleviated weight loss and intestinal pathological changes in necropsy specimens of E. coli-infected mice. MPX+ E. coli reduced the serum levels of the inflammation-related proteins interleukin-2, interleukin-6, tumour necrosis factor-α, myeloperoxidase, and lactate dehydrogenase on days 7 and 28. Furthermore, MPX+ E. coli increased the length of villi and reduced the infiltration of inflammatory cells into the jejunum and colon post infection. Scanning electron microscopy and transmission electron microscopy results showed that MPX could improve the morphology of jejunum villi and microvilli and increase tight junction protein levels. 16S rRNA sequencing analysis of caecal content samples showed that the species diversity and richness were lower in the E. coli-infected group. At the genus level, MPX+ E. coli significantly reduced the abundance of Bacteroidales and Alistipes and enhanced the relative abundance of Muribaculaceae. Alpha-diversity analyses (Shannon index) showed that MPX significantly increased the microbial diversity of mice. Overall, this study is the first to investigate the effects of oral administration of MPX on intestinal inflammation and the gut microbiota, providing a new perspective regarding the prevention of enteritis and maintenance of intestinal homeostasis.
Collapse
Affiliation(s)
- Xue Qin Zhao
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.
- Divisions of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Chun Ling Zhu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Xiang Hong Xue
- Divisions of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiao Jing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Xi Long Wu
- State Key Laboratory of Marine Resource Utilization in South China Sea, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Yun Di Wu
- State Key Laboratory of Marine Resource Utilization in South China Sea, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Shan Qin Liu
- School of Chemistry and Chemical Engineering, Henan Institute of Science and Technology, Xinxiang, China
| | - Gai Ping Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Yue Yu Bai
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hanna Fotina
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine.
| | - Jian He Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.
| |
Collapse
|
6
|
Liao SF, Ji F, Fan P, Denryter K. Swine Gastrointestinal Microbiota and the Effects of Dietary Amino Acids on Its Composition and Metabolism. Int J Mol Sci 2024; 25:1237. [PMID: 38279233 PMCID: PMC10816286 DOI: 10.3390/ijms25021237] [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: 11/05/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/28/2024] Open
Abstract
Many researchers consider gut microbiota (trillions of microorganisms) an endogenous organ of its animal host, which confers a vast genetic diversity in providing the host with essential biological functions. Particularly, the gut microbiota regulates not only gut tissue structure but also gut health and gut functionality. This paper first summarized those common bacterial species (dominated by the Firmicutes, Bacteroidota, and Proteobacteria phyla) in swine gut and then briefly discussed their roles in swine nutrition and health, which include roles in nutrient metabolism, pathogen exclusion, and immunity modulation. Secondly, the current knowledge on how dietary nutrients and feed additives affect the gut bacterial composition and nutrient metabolism in pigs was discussed. Finally, how dietary amino acids affect the relative abundances and metabolism of bacteria in the swine gut was reviewed. Tryptophan supplementation promotes the growth of beneficial bacteria and suppresses pathogens, while arginine metabolism affects nitrogen recycling, impacting gut immune response and health. Glutamate and glutamine supplementations elevate the levels of beneficial bacteria and mitigate pathogenic ones. It was concluded that nutritional strategies to manipulate gut microbial ecosystems are useful measures to optimize gut health and gut functions. For example, providing pigs with nutrients that promote the growth of Lactobacillus and Bifidobacterium can lead to better gut health and growth performance, especially when dietary protein is limited. Further research to establish the mechanistic cause-and-effect relationships between amino acids and the dynamics of gut microbiota will allow swine producers to reap the greatest return on their feed investment.
Collapse
Affiliation(s)
- Shengfa F. Liao
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA; (P.F.)
| | - Feng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China;
| | - Peixin Fan
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA; (P.F.)
| | - Kristin Denryter
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA; (P.F.)
| |
Collapse
|
7
|
Tian S, Wang J, Gao R, Zhao F, Wang J, Zhu W. Galacto-Oligosaccharides Alleviate LPS-Induced Immune Imbalance in Small Intestine through Regulating Gut Microbe Composition and Bile Acid Pool. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17615-17626. [PMID: 37947505 DOI: 10.1021/acs.jafc.3c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Recent evidence suggests that the protective effect of gut microbiota on intestinal inflammation can be achieved through a microbe-bile acids (BAs) mechanism. Galacto-oligosaccharides (GOS) are a kind of prebiotic that alter gut microbiota composition. To verify whether GOS has a protective effect on intestinal inflammation through a microbe-BAs mechanism, this research was performed in a lipopolysaccharide (LPS) porcine model with the presence or absence of GOS. GOS prevented LPS-induced production of pro-inflammatory cytokines, the decrease of bacterial bile salt hydrolase-containing bacteria abundance, and the decrease of chendoxycholic acid (CDCA) level in piglets. Additionally, CDCA decreased LPS-induced production of pro-inflammatory cytokines, induced the expression of the takeda G-protein receptor 5 (TGR5), and its downstream cyclic adenosine monophosphate (cAMP) production in lamina propria-derived CD11b+ cells. The cAMP inhibitor eliminated the protective effect of CDCA on lamina propria-derived CD11b+ cells. These results suggested that GOS reduced the production of pro-inflammatory cytokines and inhibited NF-κB activation via microbe-BA-dependent TGR5-cAMP signaling in LPS-challenged piglets.
Collapse
Affiliation(s)
- Shiyi Tian
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, P. R. China
| | - Jue Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Ren Gao
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Fangzhou Zhao
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Jing Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| |
Collapse
|
8
|
Payen C, Kerouanton A, Novoa J, Pazos F, Benito C, Denis M, Guyard M, Moreno FJ, Chemaly M. Effects of Major Families of Modulators on Performances and Gastrointestinal Microbiota of Poultry, Pigs and Ruminants: A Systematic Approach. Microorganisms 2023; 11:1464. [PMID: 37374967 DOI: 10.3390/microorganisms11061464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Considering the ban on the use of antibiotics as growth stimulators in the livestock industry, the use of microbiota modulators appears to be an alternative solution to improve animal performance. This review aims to describe the effect of different families of modulators on the gastrointestinal microbiota of poultry, pigs and ruminants and their consequences on host physiology. To this end, 65, 32 and 4 controlled trials or systematic reviews were selected from PubMed for poultry, pigs and ruminants, respectively. Microorganisms and their derivatives were the most studied modulator family in poultry, while in pigs, the micronutrient family was the most investigated. With only four controlled trials selected for ruminants, it was difficult to conclude on the modulators of interest for this species. For some modulators, most studies showed a beneficial effect on both the phenotype and the microbiota. This was the case for probiotics and plants in poultry and minerals and probiotics in pigs. These modulators seem to be a good way for improving animal performance.
Collapse
Affiliation(s)
- Cyrielle Payen
- French Agency for Food, Environmental and Occupational Health and Safety, ANSES, Hygiene and Quality of Poultry, Pig Products Unit, 22440 Ploufragan, France
| | - Annaëlle Kerouanton
- French Agency for Food, Environmental and Occupational Health and Safety, ANSES, Hygiene and Quality of Poultry, Pig Products Unit, 22440 Ploufragan, France
| | - Jorge Novoa
- Computational Systems Biology Group, National Centre for Biotechnology (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Florencio Pazos
- Computational Systems Biology Group, National Centre for Biotechnology (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Carlos Benito
- Instituto de Gestión de la Innovación y del Conocimiento, INGENIO (CSIC and U. Politécnica de Valencia), Edificio 8E, Cam. de Vera, 46022 Valencia, Spain
| | - Martine Denis
- French Agency for Food, Environmental and Occupational Health and Safety, ANSES, Hygiene and Quality of Poultry, Pig Products Unit, 22440 Ploufragan, France
| | - Muriel Guyard
- French Agency for Food, Environmental and Occupational Health and Safety, ANSES, Hygiene and Quality of Poultry, Pig Products Unit, 22440 Ploufragan, France
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación (CIAL), CSIC-UAM, CEI (UAM + CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Marianne Chemaly
- French Agency for Food, Environmental and Occupational Health and Safety, ANSES, Hygiene and Quality of Poultry, Pig Products Unit, 22440 Ploufragan, France
| |
Collapse
|
9
|
Lin A, Yan X, Wang H, Su Y, Zhu W. Effects of lactic acid bacteria-fermented formula milk supplementation on ileal microbiota, transcriptomic profile, and mucosal immunity in weaned piglets. J Anim Sci Biotechnol 2022; 13:113. [PMID: 36199127 PMCID: PMC9536082 DOI: 10.1186/s40104-022-00762-8] [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: 03/22/2022] [Accepted: 07/31/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lactic acid bacteria (LAB) participating in milk fermentation naturally release and enrich the fermented dairy product with a broad range of bioactive metabolites, which has numerous roles in the intestinal health-promoting of the consumer. However, information is lacking regarding the application prospect of LAB fermented milk in the animal industry. This study investigated the effects of lactic acid bacteria-fermented formula milk (LFM) on the growth performance, intestinal immunity, microbiota composition, and transcriptomic responses in weaned piglets. A total of 24 male weaned piglets were randomly divided into the control (CON) and LFM groups. Each group consisted of 6 replicates (cages) with 2 piglets per cage. Each piglet in the LFM group were supplemented with 80 mL LFM three times a day, while the CON group was treated with the same amount of drinking water. RESULTS LFM significantly increased the average daily gain of piglets over the entire 14 d (P < 0.01) and the average daily feed intake from 7 to 14 d (P < 0.05). Compared to the CON group, ileal goblet cell count, villus-crypt ratio, sIgA, and lactate concentrations in the LFM group were significantly increased (P < 0.05). Transcriptomic analysis of ileal mucosa identified 487 differentially expressed genes (DEGs) between two groups. Especially, DEGs involved in the intestinal immune network for IgA production pathways, such as polymeric immunoglobulin receptor (PIGR), were significantly up-regulated (P < 0.01) by LFM supplementation. Moreover, trefoil factor 2 (TFF2) in the LFM group, one of the DEGs involved in the secretory function of goblet cells, was also significantly up-regulated (P < 0.01). Sequencing of the 16S rRNA gene of microbiota demonstrated that LFM led to selective enrichment of lactate-producing and short-chain fatty acid (SCFA)-producing bacteria in the ileum, such as an increase in the relative abundance of Enterococcus (P = 0.09) and Acetitomaculum (P < 0.05). CONCLUSIONS LFM can improve intestinal health and immune tolerance, thus enhancing the growth performance of weaned piglets. The changes in microbiota and metabolites induced by LFM might mediate the regulation of the secretory function of goblet cells.
Collapse
Affiliation(s)
- Ailian Lin
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.,National Center for International Research On Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoxi Yan
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.,National Center for International Research On Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hongyu Wang
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.,National Center for International Research On Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yong Su
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China. .,National Center for International Research On Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Weiyun Zhu
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.,National Center for International Research On Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
10
|
Mei Z, Yuan J, Li D. Biological activity of galacto-oligosaccharides: A review. Front Microbiol 2022; 13:993052. [PMID: 36147858 PMCID: PMC9485631 DOI: 10.3389/fmicb.2022.993052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Galacto-oligosaccharides (GOS) are oligosaccharides formed by β-galactosidase transgalactosylation. GOS is an indigestible food component that can pass through the upper gastrointestinal tract relatively intact and ferment in the colon to produce short-chain fatty acids (SCFAs) that further regulate the body’s intestinal flora. GOS and other prebiotics are increasingly recognized as useful food tools for regulating the balance of colonic microbiota-human health. GOS performed well compared to other oligosaccharides in regulating gut microbiota, body immunity, and food function. This review summarizes the sources, classification, preparation methods, and biological activities of GOS, focusing on the introduction and summary of the effects of GOS on ulcerative colitis (UC), to gain a comprehensive understanding of the application of GOS.
Collapse
Affiliation(s)
- Zhaojun Mei
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People’s Hospital, Luzhou, China
| | - Jiaqin Yuan
- Department of Orthopedics, The Second People’s Hospital of Yibin, Yibin, China
| | - Dandan Li
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Dandan Li,
| |
Collapse
|
11
|
Xu Z, Hou Y, Sun J, Zhu L, Zhang Q, Yao W, Fan X, Zhang K, Piao JG, Wei Y. Deoxycholic acid-chitosan coated liposomes combined with in situ colonic gel enhances renal fibrosis therapy of emodin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154110. [PMID: 35487039 DOI: 10.1016/j.phymed.2022.154110] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/30/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Renal fibrosis is the final common pathological feature of various chronic kidney diseases (CKD). Despite recent advances, development of new treatments strategy is needed. Emodin (EMO), an important ingredient of Chinese medicine, rhubarb (Polygonaceae Rheum palmatum l.), has been reported to inhibit the development of renal fibrosis effectively. However, the poor oral bioavailability of EMO and the insufficient monotherapy therapy compromise its efficacy. PURPOSE In order to enhance renal fibrosis therapy of emodin, an innovative combination therapy based on deoxycholic acid-chitosan coated liposomes (DCS-Lips) and in situ colonic gel (IGE) was developed. METHODS For one, the DCS-Lips were prepared via electrostatic interaction by mixing anionic conventional Lips with cationic DCS, deoxycholic acid conjugated on the backbone of chitosan. The cellular uptake of FITC-labeled DCS-Lips in Caco-2 cell monolayer was evaluated by CLSM and flow cytometry, respectively. Permeability study was carried out using Caco-2 cell monolayer. For another, EMO-loaded in situ colonic gel (EMO-IGE) was prepared by mixing EMO nanosuspensions and plain in situ gel, which was obtained by the cold method. The EMO-IGE was assessed for morphology, gelation temperature, viscosity and in vitro drug release. Finally, the therapeutic efficacy of the combination strategy, oral DCS-Lips formulations and in situ colonic gel, was evaluated in unilateral ureteral obstruction (UUO) rat model. Additionally, 16S rDNA sequencing was performed on rats faces to investigate whether the combination strategy improves the microbial dysbiosis in UUO rats. RESULTS The prepared DCS-Lips produced small, uniformly sized nanoparticles, and significantly enhanced the cellular uptake and in vitro permeability of EMO compared to non-coated liposomes. Moreover, the EMO-IGE was characterized by short gelation time, optimal gelling temperature, and excellent viscosity. In UUO model, the combination of DCS-Lips (gavage) and IGE (enema) attenuated renal fibrosis effectively. The results of 16S rDNA sequencing illustrated that IGE could restore the gut microbial dysbiosis of UUO rats. CONCLUSION Overall, the combination of DCS-Lips and EMO-IGE alleviated renal fibrosis effectively, resulting from the improved oral bioavailability of EMO by DCS-Lips and the restoration of gut microbiota by EMO-IGE, thus, presenting an innovative and promising potential for renal fibrosis treatment.
Collapse
Affiliation(s)
- Zhishi Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Yu Hou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jiang Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qibin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Wenjie Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xudong Fan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Ke Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
12
|
Michel C, Blottière HM. Neonatal Programming of Microbiota Composition: A Plausible Idea That Is Not Supported by the Evidence. Front Microbiol 2022; 13:825942. [PMID: 35783422 PMCID: PMC9247513 DOI: 10.3389/fmicb.2022.825942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Underpinning the theory "developmental origins of health and disease" (DOHaD), evidence is accumulating to suggest that the risks of adult disease are in part programmed by exposure to environmental factors during the highly plastic "first 1,000 days of life" period. An elucidation of the mechanisms involved in this programming is challenging as it would help developing new strategies to promote adult health. The intestinal microbiome is proposed as a long-lasting memory of the neonatal environment. This proposal is supported by indisputable findings such as the concomitance of microbiota assembly and the first 1,000-day period, the influence of perinatal conditions on microbiota composition, and the impact of microbiota composition on host physiology, and is based on the widely held but unconfirmed view that the microbiota is long-lastingly shaped early in life. In this review, we examine the plausibility of the gut microbiota being programmed by the neonatal environment and evaluate the evidence for its validity. We highlight that the capacity of the pioneer bacteria to control the implantation of subsequent bacteria is supported by both theoretical principles and statistical associations, but remains to be demonstrated experimentally. In addition, our critical review of the literature on the long-term repercussions of selected neonatal modulations of the gut microbiota indicates that sustained programming of the microbiota composition by neonatal events is unlikely. This does not exclude the microbiota having a role in DOHaD due to a possible interaction with tissue and organ development during the critical windows of neonatal life.
Collapse
Affiliation(s)
| | - Hervé M. Blottière
- Nantes Université, INRAE, UMR 1280, PhAN, Nantes, France
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
| |
Collapse
|
13
|
The Role of Gut Microbiota in the Skeletal Muscle Development and Fat Deposition in Pigs. Antibiotics (Basel) 2022; 11:antibiotics11060793. [PMID: 35740199 PMCID: PMC9220283 DOI: 10.3390/antibiotics11060793] [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: 05/17/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/02/2022] Open
Abstract
Pork quality is a factor increasingly considered in consumer preferences for pork. The formation mechanisms determining meat quality are complicated, including endogenous and exogenous factors. Despite a lot of research on meat quality, unexpected variation in meat quality is still a major problem in the meat industry. Currently, gut microbiota and their metabolites have attracted increased attention in the animal breeding industry, and recent research demonstrated their significance in muscle fiber development and fat deposition. The purpose of this paper is to summarize the research on the effects of gut microbiota on pig muscle and fat deposition. The factors affecting gut microbiota composition will also be discussed, including host genetics, dietary composition, antibiotics, prebiotics, and probiotics. We provide an overall understanding of the relationship between gut microbiota and meat quality in pigs, and how manipulation of gut microbiota may contribute to increasing pork quality for human consumption.
Collapse
|
14
|
Tian S, Wang J, Gao R, Wang J, Zhu W. Early-life galacto-oligosaccharides supplementation alleviates the small intestinal oxidative stress and dysfunction of lipopolysaccharide-challenged suckling piglets. J Anim Sci Biotechnol 2022; 13:70. [PMID: 35655292 PMCID: PMC9164537 DOI: 10.1186/s40104-022-00711-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/01/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Galacto-oligosaccharides (GOS) are non-digestible food ingredients that promote the growth of beneficial bacteria in the gut. This study investigated the protective effect of the early-life GOS supplement on the piglets' gut function against the oxidative stress induced by lipopolysaccharide (LPS)-challenge. METHODS Eighteen neonatal piglets were assigned to three groups including CON, LPS and LPS + GOS groups. The piglets in CON group and LPS group received physiological saline, while those in LPS + GOS group received GOS solution for 13 d after birth. On d 14, the piglets in LPS group and LPS + GOS group were injected with LPS solutions, while the piglets in CON group were injected with the same volume of physiological saline. RESULTS The results showed that the early-life GOS supplement blocked the LPS-induced reactive oxygen species (ROS) secretion, malondialdehyde (MDA) production and the increase of pro-apoptotic factor expression. Meanwhile, the early-life GOS supplement improved the activities of antioxidant enzymes, disaccharidase enzymes activities, and digestive enzymes activities, and increased the mRNA abundance of the gene related to nutrient digestion and absorption and the relative protein expression of tight junction. The study also showed that the early-life GOS supplement improved the expression of Hemeoxygenase-1 (HO-1) and NAD(P)H/quinone acceptor oxidoreductase-1 (NQO-1), and activated the AMP-activated protein kinase (AMPK). CONCLUSIONS These results suggested that GOS enhanced the gut function, reduced the ROS production and pro-apoptotic factors gene expression, and activated the AMPK signaling pathway in LPS-challenged piglets.
Collapse
Affiliation(s)
- Shiyi Tian
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jue Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ren Gao
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jing Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
15
|
Tian S, Wang J, Gao R, Wang J, Zhu W. Galacto-oligosaccharides directly attenuate lipopolysaccharides-induced inflammatory response, oxidative stress and barrier impairment in intestinal epithelium. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
16
|
Differential effects of early-life and post-weaning galactooligosaccharides intervention on colonic bacterial composition and function in weaning piglets. Appl Environ Microbiol 2021; 88:e0131821. [PMID: 34705551 DOI: 10.1128/aem.01318-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recently, we have proved that the early-life galactooligosaccharides (GOS) intervention could improve the colonic function by altering the bacterial composition in the suckling piglets. However, whether the early-life GOS (ELG) intervention could have a long influence of the colonic microbiota, and the ELG and post-weaning GOS (PWG) combined intervention would have an interaction effect on maintaining colonic health in weaning piglets remain to be explored. Thus in this study, we illustrated the differential effect of ELG and PWG intervention on colonic microbiota and colonic function of weaning piglets. Our results showed that both the ELG and PWG intervention decreased the diarrhea frequency of weaning piglets, while the PWG intervention increased colonic indexes. After 16S rRNA MiSeq sequencing of gut bacteria belonged to colonic niches (mucosa and digesta), the PWG increased the α-diversity of colonic mucosal bacteria was revealed. In addition, we found both the ELG and PWG intervention enriched the abundance of short chain fatty acids (SCFAs) producer in different colonic niches and increased total SCFAs concentrations in colonic digesta. These changes selectively modulated the mRNA expression of pattern recognition receptors and barrier proteins in the colonic mucosa. Of note, the combined effect of ELG and PWG effectively enhanced colonic SCFAs producer enrichment and up-regulated the butyrate concentration. Meanwhile, the gene expression of MyD88-NFκB signaling and the pro-inflammatory cytokines contents were markedly reduced under the combined effect of ELG and PWG. Importance Reducing the disorders of gut ecosystem is an effective way to relieve weaning stresses of piglets and save economic losses in the modern swine industry. To this end, prebiotics were often added in diet during the weaning transition. In present study, we demonstrated that the ELG and PWG intervention had shown different effects on the bacterial composition of different colonic niches and colonic function in the weaning piglets. Especially under the combined effect of ELG and PWG intervention, the gene expression of MyD88-NFκB signaling and the contents of pro-inflammation cytokines decreased with the increasing concentration of butyrate, which is one of the important microbial metabolites in the colon of weaning piglets. These findings further provided new insights into nutritional interventions to alleviate intestinal ecosystem dysbiosis and gut dysfunction in the piglets during the weaning transition.
Collapse
|
17
|
Xu Y, Xie L, Tang J, He X, Zhang Z, Chen Y, Zhou J, Gan B, Peng W. Morchella importuna Flavones Improve Intestinal Integrity in Dextran Sulfate Sodium-Challenged Mice. Front Microbiol 2021; 12:742033. [PMID: 34552579 PMCID: PMC8451270 DOI: 10.3389/fmicb.2021.742033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/02/2021] [Indexed: 12/16/2022] Open
Abstract
Morchella importuna, as an edible fungus, has various health benefits. However, the effects of M. importuna on intestinal health are rarely investigated. Hence, this study aims to ascertain the influences of flavones from the fruiting bodies of M. importuna (hereinafter abbreviated as MIF) on dextran sulfate sodium (DSS)-induced damage to intestinal epithelial barrier in C57BL/6J mice. In this (14-day) study, 144 C57BL/6J mice were divided into four groups: (1) Control; (2) DSS treatment; (3) DSS treatment + 100 mg/kg MIF (LMIF); (4) DSS treatment + 200 mg/kg MIF (HMIF). On days 8-14, mice in the challenged groups were challenged with 3.5% DSS, while the control group received an equal volume of normal saline. Then, serum and intestinal samples were obtained from all mice. The results showed that MIF ingestion enhanced intestinal integrity in DSS-challenged mice, as evinced by the elevated (p < 0.05) abundances of occludin, claudin-1, and zonula occludens-1 proteins. Meanwhile, MIF ingestion reduced (p < 0.05) the colonic interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) concentrations and increased the superoxide dismutase and catalase activities and Shannon and Simpson indices in DSS-challenged mice. Moreover, MIF ingestion reduced (p < 0.05) the abundance of phospho-nuclear factor (NF)-κB and increased the abundance of phospho-Nrf2 in DSS-challenged mice. Taken together, MIF protects against intestinal barrier injury in C57BL/6J mice via a mechanism that involves inhibiting NF-κB activation and promoting Nrf2 activation, as well as regulating intestinal microbiota.
Collapse
Affiliation(s)
- Yingyin Xu
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Liyuan Xie
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Jie Tang
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Xiaolan He
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Zhiyuan Zhang
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Ying Chen
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Jie Zhou
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Bingcheng Gan
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Weihong Peng
- National-Local Joint Engineering Laboratory of Breeding and Cultivation of Edible and Medicinal Fungi, Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
18
|
Schroyen M, Li B, Arévalo Sureda E, Zhang Y, Leblois J, Deforce D, Van Nieuwerburgh F, Wavreille J, Everaert N. Pre-Weaning Inulin Supplementation Alters the Ileal Transcriptome in Pigs Regarding Lipid Metabolism. Vet Sci 2021; 8:vetsci8100207. [PMID: 34679037 PMCID: PMC8539436 DOI: 10.3390/vetsci8100207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022] Open
Abstract
Prebiotics, such as inulin, are non-digestible compounds that stimulate the growth of beneficial microbiota, which results in improved gut and overall health. In this study, we were interested to see if, and how, the ileal transcriptome altered after inulin administration in the pre-weaning period in pigs. Seventy-two Piétrain–Landrace newborn piglets were divided into three groups: (a) a control (CON) group (n = 24), (b) an inulin (IN)-0.5 group (n = 24), and (c) an IN-0.75 group (n = 24). Inulin was provided as a solution and administered twice a day. At week 4, eight piglets per group, those closest to the average in body weight, were sacrificed, and ileal scrapings were collected and analyzed using 3′ mRNA massively parallel sequencing. Only minor differences were found, and three genes were differentially expressed between the CON and IN-0.5 group, at an FDR of 10%. All three genes were downregulated in the IN-0.5 group. When comparing the CON group with the IN-0.75 group, five genes were downregulated in the IN-0.75 group, including the three genes seen earlier as differentially expressed between CON and IN-0.5. No genes were found to be differential expressed between IN-0.5 and IN-0.75. Validation of a selection of these genes was done using qRT-PCR. Among the downregulated genes were Angiopoietin-like protein 4 (ANGPTL4), Aquaporin 7 (AQP7), and Apolipoprotein A1 (APOA1). Thus, although only a few genes were found to be differentially expressed, several of them were involved in lipid metabolism, belonging to the peroxisome proliferator-activated receptor (PPAR) signaling pathway and known to promote lipolysis. We, therefore, conclude that these lipid metabolism genes expressed in the ileum may play an important role when supplementing piglets with inulin early in life, before weaning.
Collapse
Affiliation(s)
- Martine Schroyen
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, B-5030 Gembloux, Belgium; (M.S.); (B.L.); (E.A.S.); (Y.Z.)
| | - Bing Li
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, B-5030 Gembloux, Belgium; (M.S.); (B.L.); (E.A.S.); (Y.Z.)
| | - Ester Arévalo Sureda
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, B-5030 Gembloux, Belgium; (M.S.); (B.L.); (E.A.S.); (Y.Z.)
| | - Yuping Zhang
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, B-5030 Gembloux, Belgium; (M.S.); (B.L.); (E.A.S.); (Y.Z.)
| | - Julie Leblois
- Association Wallonne de l’Élevage asbl (AWÉ), B-5590 Ciney, Belgium;
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium; (D.D.); (F.V.N.)
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium; (D.D.); (F.V.N.)
| | - José Wavreille
- Walloon Agricultural Research Center, Department of Production and Sectors, B-5030 Gembloux, Belgium;
| | - Nadia Everaert
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, B-5030 Gembloux, Belgium; (M.S.); (B.L.); (E.A.S.); (Y.Z.)
- Correspondence: ; Tel.: +32-81-62-24-48
| |
Collapse
|
19
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Effects of a Postbiotic and Prebiotic Mixture on Suckling Rats' Microbiota and Immunity. Nutrients 2021; 13:2975. [PMID: 34578853 PMCID: PMC8469903 DOI: 10.3390/nu13092975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023] Open
Abstract
Human milk serves as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as prebiotics, probiotics and postbiotics. The aim of this study was to examine the effects of the supplementation with a postbiotic (LactofidusTM) and its combination with the prebiotics short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) in a preclinical model of healthy suckling rats. Pups were supplemented daily with LactofidusTM (POST group) and/or scGOS/lcFOS (P+P and PRE groups, respectively). Body weight and fecal consistency were analyzed. At the end of the study, immunoglobulin (Ig) profile, intestinal gene expression, microbiota composition and short chain fatty acid (SCFA) proportion were quantified. The supplementation with all nutritional interventions modulated the Ig profile, but the prebiotic mixture and the postbiotic induced differential effects: whereas scGOS/lcFOS induced softer feces and modulated microbiota composition and SCFA profile, Lactofidus™ upregulated Toll-like receptors gene expression. The use of the combination of scGOS/lcFOS and Lactofidus™ showed the effects observed for the oligosaccharides separately, as well as showing a synergistic impact on animal growth. Thus, the combined use of both products seems to be a good strategy to modulate immune and microbial features in early life.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
20
|
He N, Wang Y, Zhou Z, Liu N, Jung S, Lee MS, Li S. Preventive and Prebiotic Effect of α-Galacto-Oligosaccharide against Dextran Sodium Sulfate-Induced Colitis and Gut Microbiota Dysbiosis in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:9597-9607. [PMID: 34378931 DOI: 10.1021/acs.jafc.1c03792] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
β-Galacto-oligosaccharide (β-GOS) showed great potential in ulcerative colitis (UC) adjuvant therapy. Herein, the preventive and prebiotic effect of enzymatic-synthesized α-linked galacto-oligosaccharide (α-GOS) was investigated in dextran sodium sulfate-induced colitis and gut microbiota dysbiosis mice. Compared with β-GOS, the α-GOS supplement was more effective in improving preventive efficacy, promoting colonic epithelial barrier integrity, and alleviating inflammation cytokines. Moreover, the activation of the NOD-like receptor (NLR) family member NLRP3 inflammasome-mediated inflammation was significantly inhibited by both α-GOS and β-GOS. Gut microbiota analysis showed that α-GOS treatment reshaped the dysfunctional gut microbiota. The subsequent Spearman's correlation coefficient analysis indicated that these gut microbiota changes were significantly correlated with the inflammatory parameters. These results suggested that the enzymatic-synthesized α-GOS is a promising therapeutic agent in UC prevention and adjuvant treatment by maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China
| | - Yueyuan Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China
| | - Zihan Zhou
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China
| | - Nian Liu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China
| | - Samil Jung
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul 140-742, Korea
| | - Myeong-Sok Lee
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul 140-742, Korea
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul 140-742, Korea
| |
Collapse
|
21
|
Wu Y, Zhang X, Pi Y, Han D, Feng C, Zhao J, Chen L, Che D, Bao H, Xie Z, Wang J. Maternal galactooligosaccharides supplementation programmed immune defense, microbial colonization and intestinal development in piglets. Food Funct 2021; 12:7260-7270. [PMID: 34165467 DOI: 10.1039/d1fo00084e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The benefits of galactooligosaccharides (GOS) in neonates have been confirmed. However, the effects of nutritional programming by maternal GOS intervention on microbial colonization and intestinal development in the offspring remain unclear. In the present study, late gestational sows were fed with GOS (10 g d-1 added into the diet) or not until parturition, and the performances, immune status, microbiota composition and intestinal barriers in their piglets on day 21 were compared. GOS supplementation in pregnant sows improved their litter characteristics and the growth performance of their piglets during the neonatal stage (day 21), and elevated the plasma IgA levels in both sows and their piglets (P < 0.05). GOS intervention enriched fecal Alloprevotella and Ruminoclostridium_1 in gestational sows and vertically increased fecal Alloprevotella and Ruminococcaceae in their piglets (P < 0.05). Moreover, maternal GOS intervention increased fecal acetate (P < 0.05) and improved the intestinal barriers of their piglets by upregulating intestinal tight junctions (Occludin, Claudin-1, ZO-1), the goblet cell number and Mucin-2 (P < 0.05), which correlated positively with the colonized microbiota (P < 0.05). In summary, GOS supplementation for sows during late gestation nutritionally programmed maternal specific microbes and IgA of their offspring. This neonatal programming showed positive potential in promoting the intestinal barriers, immune defense, and growth performance of the piglets. Our findings provide evidence for maternal nutritional programming in neonates and insights for future application of GOS in maternal-neonatal nutrition.
Collapse
Affiliation(s)
- Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang H, Xia P, Lu Z, Su Y, Zhu W. Metabolome-Microbiome Responses of Growing Pigs Induced by Time-Restricted Feeding. Front Vet Sci 2021; 8:681202. [PMID: 34239912 PMCID: PMC8258120 DOI: 10.3389/fvets.2021.681202] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/20/2021] [Indexed: 01/25/2023] Open
Abstract
Time-restricted feeding (TRF) mode is a potential strategy in improving the health and production of farm animals. However, the effect of TRF on microbiota and their metabolism in the large intestine of the host remains unclear. Therefore, the present study aimed to investigate the responses of microbiome and metabolome induced by TRF based on a growing-pig model. Twelve crossbred growing barrows were randomly allotted into two groups with six replicates (1 pig/pen), namely, the free-access feeding group (FA) and TRF group. Pigs in the FA group were fed free access while the TRF group were fed free access within a regular time three times per day at 07:00–08:00, 12:00–13:00, and 18:00–19:00, respectively. Results showed that the concentrations of NH4-N, putrescine, cadaverine, spermidine, spermine, total biogenic amines, isobutyrate, butyrate, isovalerate, total SCFA, and lactate were increased while the pH value in the colonic digesta and the concentration of acetate was decreased in the TRF group. The Shannon index was significantly increased in the TRF group; however, no significant effects were found in the Fisher index, Simpson index, ACE index, Chao1 index, and observed species between the two groups. In the TRF group, the relative abundances of Prevotella 1 and Eubacterium ruminantium group were significantly increased while the relative abundances of Clostridium sensu sticto 1, Lactobacillus, and Eubacterium coprostanoligenes group were decreased compared with the FA group. PLS-DA analysis revealed an obvious and regular variation between the FA and TRF groups, further pathway enrichment analysis showed that these differential features were mainly enriched in pyrimidine metabolism, nicotinate and nicotinamide metabolism, glycerolipid metabolism, and fructose and mannose metabolism. In addition, Pearson's correlation analysis indicated that the changes in the microbial genera were correlated with the colonic metabolites. In conclusion, these results together indicated that although the overall microbial composition in the colon was not changed, TRF induced the gradient changes of the nutrients and metabolites which were correlated with certain microbial genera including Lactobacillus, Eubacterium_ruminantium group, Eubacterium coprostanoligenes group, Prevotella 1, and Clostridium sensu sticto 1. However, more studies are needed to understand the impacts of TRF on the health and metabolism of growing pigs.
Collapse
Affiliation(s)
- Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Pengke Xia
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Zhiyang Lu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
23
|
Rodríguez-Sorrento A, Castillejos L, López-Colom P, Cifuentes-Orjuela G, Rodríguez-Palmero M, Moreno-Muñoz JA, Luise D, Trevisi P, Martín-Orúe SM. Effects of the Administration of Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001 and Their Synbiotic Combination With Galacto-Oligosaccharides Against Enterotoxigenic Escherichia coli F4 in an Early Weaned Piglet Model. Front Microbiol 2021; 12:642549. [PMID: 33935999 PMCID: PMC8086512 DOI: 10.3389/fmicb.2021.642549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/23/2021] [Indexed: 01/09/2023] Open
Abstract
We evaluated the potential of multi-strain probiotic (Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001) with or without galacto-oligosaccharides against enterotoxigenic Escherichia coli (ETEC) F4 infection in post-weaning pigs. Ninety-six piglets were distributed into 32 pens assigned to five treatments: one non-challenged (CTR+) and four challenged: control diet (CTR-), with probiotics (>3 × 1010 CFU/kg body weight each, PRO), prebiotic (5%, PRE), or their combination (SYN). After 1 week, animals were orally inoculated with ETEC F4. Feed intake, weight, and clinical signs were recorded. On days 4 and 8 post-inoculation (PI), one animal per pen was euthanized and samples from blood, digesta, and tissues collected. Microbiological counts, ETEC F4 real-time PCR (qPCR) quantification, fermentation products, serum biomarkers, ileal histomorphometry, and genotype for mucin 4 (MUC4) polymorphism were determined. Animals in the PRO group had similar enterobacteria and coliform numbers to the CTR+ group, and the ETEC F4 prevalence, the number of mitotic cells at day 4 PI, and villus height at day 8 PI were between that observed in the CTR+ and CTR- groups. The PRO group exhibited reduced pig major acute-phase protein (Pig-MAP) levels on day 4 PI. The PRE diet group presented similar reductions in ETEC F4 and Pig-MAP, but there was no effect on microbial groups. The SYN group showed reduced fecal enterobacteria and coliform counts after the adaptation week but, after the inoculation, the SYN group showed lower performance and more animals with high ETEC F4 counts at day 8 PI. SYN treatment modified the colonic fermentation differently depending on the MUC4 polymorphism. These results confirm the potential of the probiotic strains and the prebiotic to fight ETEC F4, but do not show any synergy when administered together, at least in this animal model.
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
| | | | | | | | - Diana Luise
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Susana María Martín-Orúe
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
24
|
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: 25] [Impact Index Per Article: 8.3] [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
|
25
|
Du L, Sun Y, Wang Q, Wang L, Zhang Y, Li S, Jin H, Yan S, Xiao X. Integrated metabolomics and 16S rDNA sequencing to investigate the mechanism of immune-enhancing effect of health Tonic oral liquid. Food Res Int 2021; 144:110323. [PMID: 34053528 DOI: 10.1016/j.foodres.2021.110323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/08/2021] [Accepted: 03/14/2021] [Indexed: 10/21/2022]
Abstract
Health Tonic oral liquid (HT) is a popular functional food in China and is used to enhance host immune response. However, its mechanisms of action are still poorly understood. In this work, we combined ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF MS) serum metabolomics with 16S rDNA sequencing to evaluate the effects of HT on metabolomics profiling and microbial community signatures. Short-chain fatty acids (SCFAs) contents in fecal were quantified through gas chromatography-mass spectrometry (GC-MS). Results indicated that HT use leads to a significant increase in IgG, IgM and IgA. Thirty-four metabolites were identified and quantified using metabolomics, most were aromatic amino acids and metabolites involved in glucose metabolism. HT intervention significantly increased the abundance of Alloprevotella, which may contribute to intestinal barrier integrity and inflammatory response inhabitation. Most SCFAs were highly expressed following HT intake. In summary, HT use maintains glucose and lipid metabolism balance, promotes high expressions of beneficial bacteria, and exerts promising immunomodulatory effects.
Collapse
Affiliation(s)
- Lijing Du
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yuanfang Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Qian Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| | - Leqi Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| | - Yiping Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Shasha Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huizi Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shikai Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China.
| | - Xue Xiao
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
26
|
The effect of natural antimicrobials against Campylobacter spp. and its similarities to Salmonella spp, Listeria spp., Escherichia coli, Vibrio spp., Clostridium spp. and Staphylococcus spp. Food Control 2021. [DOI: 10.1016/j.foodcont.2020.107745] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
27
|
Trevisi P, Luise D, Correa F, Bosi P. Timely Control of Gastrointestinal Eubiosis: A Strategic Pillar of Pig Health. Microorganisms 2021; 9:313. [PMID: 33546450 PMCID: PMC7913656 DOI: 10.3390/microorganisms9020313] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
The pig gastrointestinal tract (GIT) is an open ecosystem in which microorganisms and their host are mutually involved and continually adapt to different factors and problems which may or may not be host dependent or due to the production system. The aim of the present review is to highlight the factors affecting the GIT microbial balance in young pigs, focusing on the pre- and post-weaning phases, to define a road map for improving pig health and the production efficiency of the food chain. Birth and weaning body weight, physiological maturation, colostrum and milk (composition and intake), genetic background, environmental stressors and management practices, antibiotic use and diet composition are considered. Overall, there is a lack of knowledge regarding the effect that some factors, including weaning age, the use of creep feed, the composition of the colostrum and milk and the use of antibiotics, may have on the gut microbiome of piglets. Furthermore, the information on the gut microbiome of piglets is mainly based on the taxonomy description, while there is a lack of knowledge regarding the functional modification of the microbiota, essential for the exploitation of microbiota potential for modulating pig physiology.
Collapse
Affiliation(s)
- Paolo Trevisi
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 40127 Bologna, Italy; (D.L.); (F.C.); (P.B.)
| | | | | | | |
Collapse
|
28
|
Wang H, Xu R, Zhang H, Su Y, Zhu W. Swine gut microbiota and its interaction with host nutrient metabolism. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:410-420. [PMID: 33364457 PMCID: PMC7750828 DOI: 10.1016/j.aninu.2020.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/09/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Gut microbiota is generally recognized to play a crucial role in maintaining host health and metabolism. The correlation among gut microbiota, glycolipid metabolism, and metabolic diseases has been well reviewed in humans. However, the interplay between gut microbiota and host metabolism in swine remains incompletely understood. Given the limitation in conducting human experiments and the high similarity between swine and humans in terms of anatomy, physiology, polyphagy, habits, and metabolism and in terms of the composition of gut microbiota, there is a pressing need to summarize the knowledge gained regarding swine gut microbiota, its interplay with host metabolism, and the underlying mechanisms. This review aimed to outline the bidirectional regulation between gut microbiota and nutrient metabolism in swine and to emphasize the action mechanisms underlying the complex microbiome-host crosstalk via the gut microbiota-gut-brain axis. Moreover, it highlights the new advances in knowledge of the diurnal rhythmicity of gut microbiota. A better understanding of these aspects can not only shed light on healthy and efficient pork production but also promote our knowledge on the associations between gut microbiota and the microbiome-host crosstalk mechanism. More importantly, knowledge on microbiota, host health and metabolism facilitates the development of a precise intervention therapy targeting the gut microbiota.
Collapse
Affiliation(s)
- Hongyu 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
| | - Rongying 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
| | - He Zhang
- 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
| | - Yong 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
| | - Weiyun 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
|
29
|
Lee SH, You HS, Kang HG, Kang SS, Hyun SH. Association between Altered Blood Parameters and Gut Microbiota after Synbiotic Intake in Healthy, Elderly Korean Women. Nutrients 2020; 12:nu12103112. [PMID: 33053824 PMCID: PMC7650560 DOI: 10.3390/nu12103112] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/25/2020] [Accepted: 10/09/2020] [Indexed: 12/20/2022] Open
Abstract
Synbiotics intake can alter the composition of intestinal microbes beneficially. We aimed to detect the changes in the intestinal microbiomes of 37 healthy elderly Korean women after the intake of a synbiotic drink. This was a longitudinal study controlled with a temporal series, including a control period of 3 weeks before intake, synbiotic intake for 3 weeks, and a washout period of 3 weeks. Fecal microbiota composition was analyzed by sequencing the V3-V4 hypervariable regions of 16S rRNA. Physical fecal activity increased with improvement in fecal shape. Thirty intestinal bacterial taxa were observed to change only after the intake period. In particular, Ellagibacter appeared only after ingestion. In addition, the abundance of Terrisporobacter showed a positive correlation with C-reactive protein, triglyceride. Lachnospiraceae_uc, Eubacterium_g5, and Blautia had a positive correlation with creatinine, whereas PAC001100_g had a negative correlation with creatinine. Short-term (3 weeks) intake of symbiotic organisms changes the composition of the gut microbiota in healthy elderly Korean women.
Collapse
Affiliation(s)
- Song Hee Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea; (S.H.L.); (H.S.Y.)
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea;
| | - Hee Sang You
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea; (S.H.L.); (H.S.Y.)
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea;
| | - Hee-Gyoo Kang
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea;
- Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Sang Sun Kang
- Department of Biology Education, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Korea;
| | - Sung Hee Hyun
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea; (S.H.L.); (H.S.Y.)
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, 77 Gyeryong-ro, 771 Beon-gil, Jung-gu, Daejeon 34824, Korea;
- Correspondence: ; Tel.: +82-10-9412-8853
| |
Collapse
|
30
|
Wu Y, Zhang X, Han D, Ye H, Tao S, Pi Y, Zhao J, Chen L, Wang J. Short Administration of Combined Prebiotics Improved Microbial Colonization, Gut Barrier, and Growth Performance of Neonatal Piglets. ACS OMEGA 2020; 5:20506-20516. [PMID: 32832803 PMCID: PMC7439367 DOI: 10.1021/acsomega.0c02667] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/23/2020] [Indexed: 05/04/2023]
Abstract
This study was conducted to investigate the effects of short administration with the combination (GMF) of galactooligosaccharides (GOS), milk fat globule membrane (MFGM), and fructooligosaccharides (FOS) on microbiota, intestinal barriers, and growth performance of neonatal piglets. Sixteen newborn piglets were divided into two groups: GMF group and CON group; GMF solution (5 mL) and saline (5 mL) were, respectively, administered to piglets in the GMF group and CON group once a day during the first week after birth. The results showed that GMF administration improved the growth performance of neonatal piglets on day 8 and day 21, coupled with the enriched genus Lactobacillus on day 8 and the increased genera norank_f__Muribaculaceae, Christensenellaceae_R-7_group, Enterococcus, and Romboutsia on day 21. Additionally, GMF administration increased luminal acetate and propionate levels, upregulated the gene expressions of intestinal tight junctions (Occludin, Claudins, and ZO-1), mucins (Mucin-1, Mucin-2, Mucin-4, and Mucin-20), and cytokines (TNF-α, IL-1β, and IL-22) while decreased the plasma diamine oxidase (DAO) level on day 21. The correlation analysis showed a positive relationship between the colonized beneficial microbiota and the modified intestinal barrier genes. In conclusion, the first week administration of GMF facilitated the colonization of beneficial bacteria, promoted intestinal development by enhancing microbiota-associated intestinal barrier functions, and improved the growth performance of the piglets during the whole neonatal period. Our findings provide guidelines for combined prebiotics application in modulating the microbial colonization and intestinal development of the neonates.
Collapse
Affiliation(s)
- Yujun Wu
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Xiangyu Zhang
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Han
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Hao Ye
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Shiyu Tao
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Yu Pi
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Junying Zhao
- National
Engineering Center of Dairy for Early Life Health, Beijing Sanyuan Foods Co. Ltd, Beijing 100163, China
| | - Lijun Chen
- National
Engineering Center of Dairy for Early Life Health, Beijing Sanyuan Foods Co. Ltd, Beijing 100163, China
| | - Junjun Wang
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
31
|
Wan J, Zhang J, Chen D, Yu B, Huang Z, Mao X, Zheng P, Yu J, He J. Alterations in intestinal microbiota by alginate oligosaccharide improve intestinal barrier integrity in weaned pigs. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
32
|
Lafontaine GMF, Fish NM, Connerton IF. In Vitro Evaluation of the Effects of Commercial Prebiotic GOS and FOS Products on Human Colonic Caco-2 Cells. Nutrients 2020; 12:nu12051281. [PMID: 32366023 PMCID: PMC7282019 DOI: 10.3390/nu12051281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 01/04/2023] Open
Abstract
Prebiotic oligosaccharides are widely used as human and animal feed additives for their beneficial effects on the gut microbiota. However, there are limited data to assess the direct effect of such functional foods on the transcriptome of intestinal epithelial cells. The purpose of this study is to describe the differential transcriptomes and cellular pathways of colonic cells directly exposed to galacto-oligosaccharides (GOS) and fructo-oligosaccharides (FOS). We have examined the differential gene expression of polarized Caco–2 cells treated with GOS or FOS products and their respective mock-treated cells using mRNA sequencing (RNA-seq). A total of 89 significant differentially expressed genes were identified between GOS and mock-treated groups. For FOS treatment, a reduced number of 12 significant genes were observed to be differentially expressed relative to the control group. KEGG and gene ontology functional analysis revealed that genes up-regulated in the presence of GOS were involved in digestion and absorption processes, fatty acids and steroids metabolism, potential antimicrobial proteins, energy-dependent and -independent transmembrane trafficking of solutes and amino acids. Using our data, we have established complementary non-prebiotic modes of action for these frequently used dietary fibers.
Collapse
Affiliation(s)
- Geraldine M. Flaujac Lafontaine
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK;
| | - Neville M. Fish
- Saputo Dairy UK, Innovation Centre, Harper Adams University, Newport TF10 8NB, UK;
| | - Ian F. Connerton
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK;
- Correspondence:
| |
Collapse
|
33
|
Flaujac Lafontaine GM, Richards PJ, Connerton PL, O’Kane PM, Ghaffar NM, Cummings NJ, Fish NM, Connerton IF. Prebiotic Driven Increases in IL-17A Do Not Prevent Campylobacter jejuni Colonization of Chickens. Front Microbiol 2020; 10:3030. [PMID: 32010094 PMCID: PMC6972505 DOI: 10.3389/fmicb.2019.03030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/17/2019] [Indexed: 01/13/2023] Open
Abstract
Worldwide Campylobacter jejuni is a leading cause of foodborne disease. Contamination of chicken meat with digesta from C. jejuni-positive birds during slaughter and processing is a key route of transmission to humans through the food chain. Colonization of chickens with C. jejuni elicits host innate immune responses that may be modulated by dietary additives to provide a reduction in the number of campylobacters colonizing the gastrointestinal tract and thereby reduce the likelihood of human exposure to an infectious dose. Here we report the effects of prebiotic galacto-oligosaccharide (GOS) on broiler chickens colonized with C. jejuni when challenged at either an early stage in development at 6 days of age or 20 days old when campylobacters are frequently detected in commercial flocks. GOS-fed birds had increased growth performance, but the levels of C. jejuni colonizing the cecal pouches were unchanged irrespective of the age of challenge. Dietary GOS modulated the immune response to C. jejuni by increasing cytokine IL-17A expression at colonization. Correspondingly, reduced diversity of the cecal microbiota was associated with Campylobacter colonization in GOS-fed birds. In birds challenged at 6 days-old the reduction in microbial diversity was accompanied by an increase in the relative abundance of Escherichia spp. Whilst immuno-modulation of the Th17 pro-inflammatory response did not prevent C. jejuni colonization of the intestinal tract of broiler chickens, the study highlights the potential for combinations of prebiotics, and specific competitors (synbiotics) to engage with the host innate immunity to reduce pathogen burdens.
Collapse
Affiliation(s)
- Geraldine M. Flaujac Lafontaine
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Philip J. Richards
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Phillippa L. Connerton
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Peter M. O’Kane
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Nacheervan M. Ghaffar
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Nicola J. Cummings
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Neville M. Fish
- Saputo Dairy UK, Dairy Crest Innovation Centre, Harper Adams University, Newport, United Kingdom
| | - Ian F. Connerton
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| |
Collapse
|
34
|
Wang J, Tian S, Wang J, Zhu W. Early galactooligosaccharide intervention alters the metabolic profile, improves the antioxidant capacity of mitochondria and activates the AMPK/Nrf2 signaling pathway in suckling piglet liver. Food Funct 2020; 11:7280-7292. [DOI: 10.1039/d0fo01486a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The early GOS intervention altered the composition of the hepatic metabolic profile by promoting lipid catabolism and regulating amino acid metabolism in the suckling piglets.
Collapse
Affiliation(s)
- Jue Wang
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Shiyi Tian
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| |
Collapse
|
35
|
Tian S, Shi Q, Zhu Y, Yang H, Wang J, Zhu W. Significant changes in caecal microbial composition and metabolites of weaned piglets after protein restriction and succedent realimentation. J Anim Physiol Anim Nutr (Berl) 2019; 104:1126-1133. [PMID: 31840859 DOI: 10.1111/jpn.13268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
The present study aimed to investigate the effects of protein restriction and subsequent realimentation on caecal barrier function, caecal microbial composition and metabolites in weaned piglets. Thirty-six 28-day-old weaned piglets were randomly assigned to a control group and a treatment group. The piglets were fed diets containing 18.83% (normal) or 13.05% (low) of crude protein from the 1st to 14th day, after which all piglets were fed diets containing 18.83% of crude protein from the 15th to 28th day. The results showed that protein restriction increased caecal bacterial diversity and richness as well as the abundance of Ruminococcus 2, Faecalibacterium and Lachnospiraceae_uncultured, but reduced the abundance and the gene copies of Lactobacillus in the treatment group compared with the control group on day 14. Protein restriction also decreased the concentrations of isovaleric acid and total branched-chain fatty acids. During the succedent protein realimentation stage, the abundance of Ruminococcaceae UCG-014 and the concentrations of lactic acid, acetic acid, butyric acid and total short-chain fatty acids were increased in the treatment group on day 28. Furthermore, the ammonia concentration was reduced, while the gene mRNA levels of caecal barrier function were increased in the treatment group both on days 14 and 28. In conclusion, dietary protein restriction and realimentation could change caecal microbial composition and metabolites, and eventually influence caecal barrier function. The present study may provide a new insight into protein restriction and realimentation in weaned piglets.
Collapse
Affiliation(s)
- Shiyi Tian
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qing Shi
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yizhi Zhu
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Huairong Yang
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jing Wang
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, Joint International Research Laboratory of Animal Health and Food Safety, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|