1
|
Schumacher SM, Doyle WJ, Hill K, Ochoa-Repáraz J. Gut microbiota in multiple sclerosis and animal models. FEBS J 2025; 292:1330-1356. [PMID: 38817090 PMCID: PMC11607183 DOI: 10.1111/febs.17161] [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: 10/17/2023] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
Multiple sclerosis (MS) is a chronic central nervous system (CNS) neurodegenerative and neuroinflammatory disease marked by a host immune reaction that targets and destroys the neuronal myelin sheath. MS and correlating animal disease models show comorbidities, including intestinal barrier disruption and alterations of the commensal microbiome. It is accepted that diet plays a crucial role in shaping the microbiota composition and overall gastrointestinal (GI) tract health, suggesting an interplay between nutrition and neuroinflammation via the gut-brain axis. Unfortunately, poor host health and diet lead to microbiota modifications that could lead to significant responses in the host, including inflammation and neurobehavioral changes. Beneficial microbial metabolites are essential for host homeostasis and inflammation control. This review will highlight the importance of the gut microbiota in the context of host inflammatory responses in MS and MS animal models. Additionally, microbial community restoration and how it affects MS and GI barrier integrity will be discussed.
Collapse
Affiliation(s)
| | | | - Kristina Hill
- Department of Biological Sciences, Boise State University, Boise, ID 83725
| | | |
Collapse
|
2
|
Xiong L, Zhang Z, Dong S, Lin T, Yue X, Chen F, Guan W, Zhang S. Maternal consumption of glycerol monolaurate optimizes milk fatty acid profile and enhances piglet gut health in association with G protein-coupled receptor 84 (GPR84) activation. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:387-403. [PMID: 40034459 PMCID: PMC11872655 DOI: 10.1016/j.aninu.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025]
Abstract
This study evaluated the effect of maternal glycerol monolaurate (GML) supplementation during late gestation and lactation on sow reproductive performance, transfer of immunity and redox status, milk fat and fatty acid profile, and fecal microbiota. Eighty multiparous sows (Landrace × Large white) were randomly allocated to two treatment groups (with or without 1000 mg/kg GML) with 40 replicates per treatment. The feeding experiment lasted from d 85 of gestation (G85) to d 23 of lactation (L23). The samples were collected on d 1 (L1) and 21 (L21) of lactation. Our results showed that maternal GML supplementation significantly increased litter weight (P = 0.002), average daily gain of piglets (P = 0.048), and sow average daily feed intake (P = 0.032). Compared with CON group, the concentrations of lauric acid (C12:0; P = 0.022), C16:0 (P = 0.001), and total saturated fatty acids (P = 0.006) in colostrum as well as C12:0 in L21 milk (P = 0.001) were higher in GML group. Besides, the concentrations of immunoglobulin A (IgA) and IgG in colostrum as well as sow and piglet plasma, the total antioxidant capacity and superoxide dismutase activity in sow colostrum were also significantly higher in the GML group (P < 0.05). Microbiome results showed that GML addition increased fecal microbial alpha diversity as well as the relative abundances of short chain fatty acids producing bacteria Ruminococcaceae and Parabacteroides; and decreased the harmful Proteobacteria of sows (P < 0.05). The Spearman analysis showed that the microbial biomarkers Prevotellaceae, Ruminococcaceae, and Parabacteroides were positively correlated with IgA and IgG of sow plasma and milk (P < 0.05). Besides, maternal GML addition up-regulated the relative protein expressions of proliferating cell nuclear antigen, cyclin D1, G protein-coupled receptor 84 (GPR84) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway in the duodenum and jejunum of piglets. Collectively, current findings suggested that maternal GML supplementation enhanced piglet growth during lactation, which might be associated with improving milk fat and lauric acid contents, microbiota derived immunoglobulins transfer, and gut health through potential involvement of GPR84 and PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Liang Xiong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhijin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shiqi Dong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tongbin Lin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xianhuai Yue
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
3
|
Fang X, Wang Z, Chen Q, Du Y, Sun H, Liu H, Feng Y, Li Z, Teng T, Shi B. Protective effect of the branched short-chain fatty acid isobutyrate on intestinal damage in weaned piglets through intestinal microbiota remodeling. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:1556-1568. [PMID: 39412364 DOI: 10.1002/jsfa.13930] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 01/14/2025]
Abstract
BACKGROUND Postweaning intestinal damage in piglets is a challenging issue in the livestock industry. Short-chain fatty acids (SCFAs) are important metabolic products of the gut microbiota and are widely recognized for their role in maintaining normal colonic function and regulating the intestinal immune system. However, the effects of branched short-chain fatty acid (BSCFA) isobutyrate on intestinal health remain largely unknown. This study aims to explore the potential of isobutyrate for alleviating postweaning intestinal damage. RESULTS This study indicates that isobutyrate can alleviate diarrhea in weaned piglets, enhance their growth performance, and optimize the gut microbiota. This is mainly achieved through increasing the relative abundance of probiotic bacteria such as Lactobacillus, Megasphaera, and Prevotellaceae_UCG-003, while concurrently reducing the relative abundance of potentially harmful bacteria such as Clostridium_sensu_stricto-1 and Escherichia-Shigella. It promotes the production of SCFAs, including acetate, isobutyrate, and butyrate. Furthermore, it activates G-protein-coupled receptors (GPR43/109A), inhibits the TLR4/MyD88 signaling pathway, strengthens the intestinal barrier function, and regulates the expression of related cytokines. CONCLUSION In summary, exogenous isobutyrate can be considered a promising feed additive for improving the intestinal microbiota and regulating intestinal health in piglets. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiuyu Fang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhengyi Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Qinrui Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yongqing Du
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Haowen Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Haiyang Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Ye Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Teng Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
4
|
Park S, Sun S, Wongchanla S, Chen Y, Li X, Liu Y. Dietary supplementation of blend of organic acids and monoglycerides alleviated diarrhea and systemic inflammation of weaned pigs experimentally infected with enterotoxigenic Escherichia coli F18. J Anim Sci Biotechnol 2025; 16:11. [PMID: 39838409 PMCID: PMC11752944 DOI: 10.1186/s40104-024-01148-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The emergence of antibiotic resistant microorganisms associated with conventional swine production practices has increased interest in acid-based compounds having antimicrobial properties and other biological functions as nutritional interventions. Despite the interest in organic acids and monoglycerides, few studies have examined the effects of the combination of these acid-based additives in weaned pigs under disease challenge conditions. Therefore, this study aimed to investigate the effects of dietary supplementation with blend of organic acids and/or medium-chain fatty acid monoglycerides on intestinal health and systemic immunity of weaned pigs experimentally infected with an enterotoxigenic Escherichia coli (ETEC) F18 at 4-week of age. RESULTS Dietary supplementation of organic acids, monoglycerides, or both organic acids and monoglycerides (combination) reduced (P < 0.05) the diarrhea frequency of ETEC F18-infected pigs throughout the experimental period (d -7 to 21 post-inoculation). This is consistent with the reduced (P < 0.05) proportion of β-hemolytic coliforms in feces observed for the organic acid and combination treatments on d 10 post-inoculation. Supplementation of organic acids, monoglycerides, or combination also reduced (P < 0.05) bacterial translocation in mesenteric lymph nodes on d 21 post-inoculation. Pigs fed with monoglycerides or combination had lower (P < 0.05) white blood cells on d 5 post-inoculation, and pigs fed the combination also had lower (P < 0.05) lymphocytes than pigs in control group. Monoglyceride supplementation increased (P < 0.05) white blood cells and neutrophils compared with control group on d 14 post-inoculation. However, supplementation with organic acid blend, monoglyceride blend, or combination did not affect growth performance in this experiment. CONCLUSIONS Supplementation with monoglycerides or organic acids alone or in combination improves the detrimental effects of ETEC F18 infection in weaned pigs, as indicated by reduced diarrhea, fecal shedding of β-hemolytic coliforms, and bacterial translocation, and thus enhancing disease resistance. Monoglycerides reduced the inflammatory response during peak infection, but their immunomodulatory and possible synergistic effects with organic acids need to be further investigated.
Collapse
Affiliation(s)
- Sangwoo Park
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Shuhan Sun
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | | | - Ying Chen
- Animal Nutrition, Eastman Chemical Company, Kingsport, TN, USA
| | - Xunde Li
- School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
5
|
Zhao C, Li Y, Wang H, Solomon AI, Wang S, Dong X, Song B, Ren Z. Dietary supplementation with compound microecological preparations: effects on the production performance and gut microbiota of lactating female rabbits and their litters. Microbiol Spectr 2025; 13:e0006724. [PMID: 39611688 PMCID: PMC11705915 DOI: 10.1128/spectrum.00067-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 08/04/2024] [Indexed: 11/30/2024] Open
Abstract
Early weaning is frequently accompanied by a significant increase in diarrhea and mortality rates, which reduces rabbits' performance. Although antibiotics can reduce pathogenic bacteria, they also harm beneficial microorganisms and disrupt the normal intestinal microbiota balance. In order to find non-residue and non-toxic alternatives to antibiotics to ensure the safety of animal products, we conducted a study on the effect of compound microecological preparations supplementation on lactating female rabbits and their offspring. A total of 60 female rabbits were randomly assigned to four groups: CON, supplemented with probiotics at 3, 6, and 9 g/female rabbit/day from day 24 of gestation until weaning. We observed that probiotics supplementation significantly enhanced production performance (P < 0.05), immune and antioxidant function (P < 0.05), as well as intestinal flora composition in lactating rabbits and their offspring. Notably, compared with the control group, the experimental group exhibited a 19.23%, 44.22%, and 24.57% increase in milk yield (P = 0.002). Regarding rabbit growth performance, the average body weight of young rabbits in the experimental group showed a significant increase of 3.59%, 10.22%, and 6.74% at day 35 (P = 0.022), whereas the average daily gain (ADG) of rabbits aged between 21 and 35 days was significantly elevated by 4.94%, 17.06%, and 6.28% in the experimental group (P < 0.001). In conclusion, probiotics supplementation can significantly enhance lactation performance, promote growth and disease resistance in rabbits, as well as improve intestinal health when administered at a dosage of 6 g/day. Moreover, the limited sample size in this study may hinder the detection of subtle effects, and augmenting the sample size will bolster the reliability of the study findings. IMPORTANCE The intestinal environment of rabbits is fragile and susceptible to environmental influences, leading to inflammatory intestinal diseases. Adding antibiotics to rabbit feed can achieve the effect of preventing and treating inflammation, which can also lead to the imbalance of the gut microbiota and residual antibiotics in agricultural products. Composite probiotics are live microbial feed additives composed of various ratios of probiotics and have become the most promising alternative to antibiotics due to their residue-free and non-toxic properties. The aim of this study was to investigate the impact of compound probiotics on lactating female rabbits and their offspring. Our findings highlight the potential of compound microecological preparations as an effective strategy for enhancing lactation performance, immune function, and antioxidant capacity in rabbits. The supplementation of probiotics through rabbit milk offers a promising approach to optimize the growth and health outcomes of newborn rabbits.
Collapse
Affiliation(s)
- Chengcheng Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Youhao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ahamba Ifeanyi Solomon
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuhui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xianggui Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Bing Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhanjun Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
6
|
Dong S, Zhang N, Wang J, Cao Y, Johnston LJ, Ma Y. Effects of Medium- and Short-Chain Fatty Acids on Growth Performance, Nutrient Digestibility, Gut Microbiota and Immune Function in Weaned Piglets. Animals (Basel) 2024; 15:37. [PMID: 39794980 PMCID: PMC11718992 DOI: 10.3390/ani15010037] [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: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
The aim of this study was to investigate the combination effects of α-glycerol monolaurate (GML) and glyceryl tributyrate (TB) on growth performance, nutrient digestibility, gut microbiota, and immune function in weaned piglets. A total of 120 weaned piglets with an average body weight (BW) of 6.88 kg were randomly allocated to one of the three dietary treatments: (1) CON: a basal diet; (2) 0.1%: a basal diet with 0.1% MSCFA (GML/TB = 1:1); (3) 0.2%: a basal diet with 0.2% MSCFA (GML/TB = 1:1). The experiment lasted 28 days. There were no differences on average daily growth (ADG), average daily feed intake (ADFI), and feed conversion ratio (FCR). Supplementation with 0.1% MSCFA increased apparent total tract digestibility (ATTD) of crude protein (CP) and gross energy (GE, p < 0.05) on d 14 and increased GE (p < 0.05) on d 28 compared with the CON group. The ATTD of dry matter (DM), organic matter (OM) and crude protein (CP) of piglets supplemented with 0.1% MSCFA was higher (p < 0.05). Compared with the CON group, supplementation with 0.1% MSCFA increased immunoglobulin M (IgM) concentration, decreased interleukin-6 (IL-6) content (p < 0.05) on d 14 and decreased malonaldehyde (MDA), interleukin-1beta (IL-1β), IL-6 concentrations (p < 0.05) on d 28. Supplementation with 0.1% MSCFA increased total antioxidant capacity (T-AOC) concentration (p < 0.05), decreased GSH-Px, MDA content (p < 0.05) in jejunum compared with the CON group. Moreover, supplementation with MSCFA increased the activity of duodenal lipase (p < 0.05) and the abundance of firmicutes and decreased the abundance of proteobacteria compared with the CON group. Overall, supplementation with MSCFA can improve nutrient digestibility, enhance immunity and antioxidant capacity, and improve the intestinal health of piglets. The combined use of MSCFA is a nutrition regulation strategy worthy of further exploration in modern animal husbandry.
Collapse
Affiliation(s)
- Shuang Dong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (N.Z.)
| | - Nan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (N.Z.)
| | - Jihua Wang
- Galido Biotechnology Co., Ltd., Wuhan 430074, China; (J.W.); (Y.C.)
| | - Yu Cao
- Galido Biotechnology Co., Ltd., Wuhan 430074, China; (J.W.); (Y.C.)
| | - Lee J. Johnston
- Department of Animal Science, West Central Research and Outreach Center, University of Minnesota, Morris, MN 56267, USA;
| | - Yongxi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (N.Z.)
| |
Collapse
|
7
|
Huangfu W, Ma J, Zhang Y, Liu M, Liu B, Zhao J, Wang Z, Shi Y. Dietary Fiber-Derived Butyrate Alleviates Piglet Weaning Stress by Modulating the TLR4/MyD88/NF-κB Pathway. Nutrients 2024; 16:1714. [PMID: 38892647 PMCID: PMC11174469 DOI: 10.3390/nu16111714] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
During weaning, piglets are susceptible to intestinal inflammation and impairment in barrier function. Dietary fiber (DF) plays an active role in alleviating weaning stress in piglets. However, the effects of different sources of dietary fiber on the performance of weaned piglets are inconsistent, and the mechanisms through which they affect intestinal health need to be explored. Therefore, in this study, sixty weaned piglets were randomly divided into three treatment groups: basal diet (control, CON), beet pulp (BP), and alfalfa meal (AM) according to the feed formulation for a 28-day trial. The results showed that both AM and BP groups significantly reduced diarrhea rate and serum inflammatory factors (IL-1β and TNF-α) and increased antioxidant markers (T-AOC and SOD), in addition to decreasing serum MDA and ROS concentrations in the AM group. At the same time, piglets in the AM group showed a significant reduction in serum intestinal permeability indices (LPS and DAO) and a substantial increase in serum immunoglobulin levels (IgA, IgG, and IgM) and expression of intestinal barrier-associated genes (Claudin1, Occludin, ZO-1, and MUC1), which resulted in an improved growth performance. Interestingly, the effect of DF on intestinal inflammation and barrier function can be attributed to its modulation of gut microbes. Fiber-degrading bacteria enriched in the AM group (Christensenellaceae_R-7_group, Pediococcus and Weissella) inhibited the production of TLR4- through the promotion of SCFAs (especially butyrate). MyD88-NF-κB signaling pathway activation reduces intestinal inflammation and repairs intestinal barrier function. In conclusion, it may provide some theoretical support and rationale for AM to alleviate weaning stress and improve early intestinal dysfunction, which may have implications for human infants.
Collapse
Affiliation(s)
- Weikang Huangfu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
| | - Jixiang Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
| | - Yan Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
| | - Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (W.H.); (J.M.); (Y.Z.); (M.L.); (B.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| |
Collapse
|
8
|
Yang M, Zhang J, Yan H, Pan Y, Zhou J, Zhong H, Wang J, Cai H, Feng F, Zhao M. A comprehensive review of medium chain monoglycerides on metabolic pathways, nutritional and functional properties, nanotechnology formulations and applications in food system. Crit Rev Food Sci Nutr 2024; 65:2943-2964. [PMID: 38779723 DOI: 10.1080/10408398.2024.2353403] [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] [Indexed: 05/25/2024]
Abstract
A large and growing body of literature has investigated the broad antibacterial spectrum and strong synergistic antimicrobial activity of medium chain monoglycerides (MCMs) have been widely investigated. Recently, more and more researches have focused on the regulation of MCMs on metabolic health and gut microbiota both in vivo and in vitro. The current review summarizes the digestion, absorption and metabolism of MCMs. Subsequently, it focuses on the functional and nutritional properties of MCMs, including the antibacterial and antiviral characteristics, the modulation of metabolic balance, the regulation of gut microbiota, and the improvement in intestinal health. Additionally, we discuss the most recent developments and application of MCMs using nanotechnologies in food industry, poultry and pharmaceutical industry. Additionally, we analyze recent application examples of MCMs and their nanotechnology formation used in food. The development of nanotechnology platforms facilitating molecular encapsulation and functional presentation contribute to the application of hydrophobic fatty acids and monoglycerides in food preservation and their antibacterial effectiveness. This study emphasizes the metabolic mechanisms and biological activity of MCMs by summarizing the prevailing state of knowledge on this topic, as well as providing insights into prospective techniques for developing the beneficial applications of MCMs to realize the industrialized production.
Collapse
Affiliation(s)
- Mengyu Yang
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| | - Junhui Zhang
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| | - Heng Yan
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| | - Ya Pan
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| | - Jie Zhou
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Jing Wang
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
- Ningbo Innovation Center, Zhejiang University, Ningbo, China
- Guangdong Qingyunshan Pharmaceutical Co., Ltd, Shaoguan, China
| | - Haiying Cai
- School of Biological & Chemical Engineering, Zhejiang Key Lab for Chem & Bio Processing Technology of Farm Product, Zhejiang University of Science and Technology, Hangzhou, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Galeeva JS, Fedorov DE, Starikova EV, Manolov AI, Pavlenko AV, Selezneva OV, Klimina KM, Veselovsky VA, Morozov MD, Yanushevich OO, Krikheli NI, Levchenko OV, Andreev DN, Sokolov FS, Fomenko AK, Devkota MK, Andreev NG, Zaborovskiy AV, Bely PA, Tsaregorodtsev SV, Evdokimov VV, Maev IV, Govorun VM, Ilina EN. Microbial Signatures in COVID-19: Distinguishing Mild and Severe Disease via Gut Microbiota. Biomedicines 2024; 12:996. [PMID: 38790958 PMCID: PMC11118803 DOI: 10.3390/biomedicines12050996] [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: 04/14/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has significantly impacted global healthcare, underscoring the importance of exploring the virus's effects on infected individuals beyond treatments and vaccines. Notably, recent findings suggest that SARS-CoV-2 can infect the gut, thereby altering the gut microbiota. This study aimed to analyze the gut microbiota composition differences between COVID-19 patients experiencing mild and severe symptoms. We conducted 16S rRNA metagenomic sequencing on fecal samples from 49 mild and 43 severe COVID-19 cases upon hospital admission. Our analysis identified a differential abundance of specific bacterial species associated with the severity of the disease. Severely affected patients showed an association with Enterococcus faecium, Akkermansia muciniphila, and others, while milder cases were linked to Faecalibacterium prausnitzii, Alistipes putredinis, Blautia faecis, and additional species. Furthermore, a network analysis using SPIEC-EASI indicated keystone taxa and highlighted structural differences in bacterial connectivity, with a notable disruption in the severe group. Our study highlights the diverse impacts of SARS-CoV-2 on the gut microbiome among both mild and severe COVID-19 patients, showcasing a spectrum of microbial responses to the virus. Importantly, these findings align, to some extent, with observations from other studies on COVID-19 gut microbiomes, despite variations in methodologies. The findings from this study, based on retrospective data, establish a foundation for future prospective research to confirm the role of the gut microbiome as a predictive biomarker for the severity of COVID-19.
Collapse
Affiliation(s)
- Julia S. Galeeva
- Research Institute for Systems Biology and Medicine, Department of Mathematical Biology and Bioinformatics, Moscow 117246, Russia; (D.E.F.); (E.V.S.); (A.I.M.); (A.V.P.)
| | - Dmitry E. Fedorov
- Research Institute for Systems Biology and Medicine, Department of Mathematical Biology and Bioinformatics, Moscow 117246, Russia; (D.E.F.); (E.V.S.); (A.I.M.); (A.V.P.)
| | - Elizaveta V. Starikova
- Research Institute for Systems Biology and Medicine, Department of Mathematical Biology and Bioinformatics, Moscow 117246, Russia; (D.E.F.); (E.V.S.); (A.I.M.); (A.V.P.)
| | - Alexander I. Manolov
- Research Institute for Systems Biology and Medicine, Department of Mathematical Biology and Bioinformatics, Moscow 117246, Russia; (D.E.F.); (E.V.S.); (A.I.M.); (A.V.P.)
| | - Alexander V. Pavlenko
- Research Institute for Systems Biology and Medicine, Department of Mathematical Biology and Bioinformatics, Moscow 117246, Russia; (D.E.F.); (E.V.S.); (A.I.M.); (A.V.P.)
| | - Oksana V. Selezneva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (O.V.S.); (K.M.K.); (V.A.V.); (M.D.M.)
| | - Ksenia M. Klimina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (O.V.S.); (K.M.K.); (V.A.V.); (M.D.M.)
| | - Vladimir A. Veselovsky
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (O.V.S.); (K.M.K.); (V.A.V.); (M.D.M.)
| | - Maxim D. Morozov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (O.V.S.); (K.M.K.); (V.A.V.); (M.D.M.)
| | - Oleg O. Yanushevich
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Natella I. Krikheli
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Oleg V. Levchenko
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Dmitry N. Andreev
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Filipp S. Sokolov
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Aleksey K. Fomenko
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Mikhail K. Devkota
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Nikolai G. Andreev
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Andrey V. Zaborovskiy
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Petr A. Bely
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Sergei V. Tsaregorodtsev
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Vladimir V. Evdokimov
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Igor V. Maev
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Vadim M. Govorun
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| | - Elena N. Ilina
- Department of Clinical Dentistry, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia; (O.O.Y.); (N.I.K.); (O.V.L.); (D.N.A.); (F.S.S.); (A.K.F.); (M.K.D.); (N.G.A.); (A.V.Z.); (P.A.B.); (S.V.T.); (V.V.E.); (I.V.M.); (V.M.G.)
| |
Collapse
|
10
|
Lin Y, Zhai JL, Wang YT, Guo PT, Zhang J, Wang CK, Jin L, Gao YY. Potassium diformate alleviated inflammation of IPEC-J2 cells infected with EHEC. Vet Microbiol 2024; 291:110013. [PMID: 38364468 DOI: 10.1016/j.vetmic.2024.110013] [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: 10/12/2023] [Revised: 01/16/2024] [Accepted: 01/28/2024] [Indexed: 02/18/2024]
Abstract
Potassium diformate (KDF) is a kind of formate, which possesses the advantages of antimicrobial activity, growth promotion and preventing diarrhea in weaned piglets. However, the researches of KDF in animal production mostly focused on apparent indexes such as growth performance and the mechanisms of KDF on intestinal health have not been reported. Thus, porcine small intestinal epithelial cells (IPEC-J2) infected with Enterohemorrhagic Escherichia coli (EHEC) was used to investigate the role of KDF on alleviating intestinal inflammation in this study. The 0.125 mg/mL KDF treated IPEC-J2 cells for 6 h and IPEC-J2 cells challenged with 5 × 107 CFU/mL EHEC for 4 h were confirmed as the optimum concentration and time for the following experiment. The subsequent experiment was divided into four groups: control group (CON), EHEC group, KDF group, KDF+EHEC group. The results showed that KDF increased the cell viability and the gene expression levels of SGLT3 and TGF-β, while decreased the content of IL-1β compared with the CON group. The cell viability and the gene expressions of SGLT1, SGLT3, GLUT2, Claudin-1, Occludin and TGF-β, and the protein expression of ZO-1 in EHEC group were lower than those in CON group, whereas the gene expressions of IL-1β, TNF, IL-8 and TLR4, and the level of phosphorylation NF-кB protein were increased. Pretreatment with KDF reduced the content of IgM and IL-1β, the gene expressions of IL-1β, TNF, IL-8 and TLR4 and the level of phosphorylation NF-кB protein, and increased the gene expression of TGF-β and the protein expression of Occludin in IPEC-J2 cells infected EHEC. In conclusion, 0.125 mg/mL KDF on IPEC-J2 cells for 6 h had the beneficial effects on ameliorating the intestinal inflammation because of reduced pro-inflammatory cytokines and enhanced anti-inflammatory cytokines through regulating NF-кB signaling pathway under the EHEC challenge.
Collapse
Affiliation(s)
- Ying Lin
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jun-Lei Zhai
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ya-Ting Wang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ping-Ting Guo
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jing Zhang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chang-Kang Wang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ling Jin
- China National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Yu-Yun Gao
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
11
|
Zhang F, Chen M, Liu X, Ji X, Li S, Jin E. New insights into the unfolded protein response (UPR)-anterior gradient 2 (AGR2) pathway in the regulation of intestinal barrier function in weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:225-232. [PMID: 38033605 PMCID: PMC10685161 DOI: 10.1016/j.aninu.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/05/2023] [Accepted: 08/11/2023] [Indexed: 12/02/2023]
Abstract
Sustained dysfunction of the intestinal barrier caused by early weaning is a major factor that induces postweaning diarrhea in weaned piglets. In both healthy and diseased states, the intestinal barrier is regulated by goblet cells. Alterations in the characteristics of goblet cells are linked to intestinal barrier dysfunction and inflammatory conditions during pathogenic infections. In this review, we summarize the current understanding of the mechanisms of the unfolded protein response (UPR) and anterior gradient 2 (AGR2) in maintaining intestinal barrier function and how modifications to these systems affect mucus barrier characteristics and goblet cell dysregulation. We highlight a novel mechanism underlying the UPR-AGR2 pathway, which affects goblet cell differentiation and maturation and the synthesis and secretion of mucin by regulating epidermal growth factor receptor and mucin 2. This study provides a theoretical basis and new insights into the regulation of intestinal health in weaned piglets.
Collapse
Affiliation(s)
- Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Mengxian Chen
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Shenghe Li
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| |
Collapse
|
12
|
Kong L, Cai Y, Pan X, Xiao C, Song Z. Glycerol monolaurate improves intestinal morphology and antioxidant status by suppressing inflammatory responses and nuclear factor kappa-B signaling in lipopolysaccharide-exposed chicken embryos. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:297-306. [PMID: 38033609 PMCID: PMC10684993 DOI: 10.1016/j.aninu.2023.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/15/2023] [Accepted: 06/29/2023] [Indexed: 12/02/2023]
Abstract
Medium-chain fatty acids and their derivatives are natural ingredients that support immunological functions in animals. The effects of glycerol monolaurate (GML) on intestinal innate immunity and associated molecular mechanisms were investigated using a chicken embryo model. Sixty-four Arbor Acres broiler embryos were randomly allocated into four groups. On embryonic day 17.5, the broiler embryos were administered with 9 mg of GML, which was followed by a 12-h incubation period and a 12-h challenge with 32 μg of lipopolysaccharide (LPS). On embryonic day 18.5, the jejunum and ileum were harvested. Results indicated that GML reversed the LPS-induced decline in villus height and upregulated the expression of mucin 2 (P < 0.05). GML decreased LPS-induced malondialdehyde production and boosted antioxidant enzyme activity (P < 0.05). GML alleviated LPS-stimulated intestinal secretion of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) (P < 0.05). GML also normalized LPS-induced changes in the gene expression of Toll-like receptor 4, nuclear factor kappa-B p65 (NF-κB p65), cyclooxygenase-2, NOD-like receptor protein 3, IL-18, zonula occludens 1, and occludin (P < 0.05). GML enhanced as well the expression of AMP-activated protein kinase α1 and claudin 1 (P < 0.05). In conclusion, GML improved intestinal morphology and antioxidant status by alleviating inflammatory responses and modulating NF-κB signaling in LPS-challenged broiler embryos.
Collapse
Affiliation(s)
- Linglian Kong
- Key Laboratory of Efficient Utilization of Nongrain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Yuanli Cai
- College of Life Science, Qilu Normal University, Jinan, Shandong 250200, China
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Nongrain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Chuanpi Xiao
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Nongrain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong 271018, China
| |
Collapse
|
13
|
Qi Y, Zheng T, Yang S, Zhang Q, Li B, Zeng X, Zhong Y, Chen F, Guan W, Zhang S. Maternal sodium acetate supplementation promotes lactation performance of sows and their offspring growth performance. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:213-224. [PMID: 37484994 PMCID: PMC10362078 DOI: 10.1016/j.aninu.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 07/25/2023]
Abstract
Milk yield and composition are critical determining factors for the early growth and development of neonates. The objective of this experiment was to comprehensively evaluate the effects of dietary sodium acetate (SA) supplementation on the milk yield and composition of sows and the growth performance of their offspring. A total of 80 sows (Landrace × Yorkshire, 3 to 6 parity) were randomly assigned to 2 groups (with or without 0.1% SA) from d 85 of gestation to d 21 of lactation. The result shows that maternal 0.1% SA supplementation significantly increased sows milk yield, milk fat, immunoglobulin A (IgA) and IgG content in milk (P < 0.05), with the up-regulation of short-chain fatty acids receptors (GPR41 and GPR43) expression and the activation of mammalian target of rapamycin complex C1 (mTORC1) signaling pathway. Consistently, in our in vitro experiment, SA also activated mTORC1 signaling in porcine mammary epithelial cells (P < 0.05). Furthermore, the improvement of milk quality and quantity caused by maternal SA supplementation led to the increase in body weight (BW) and average daily weight gain (ADG) of weaning piglets, with the improvement of gut health and colonization of the beneficial bacteria (P < 0.05). In conclusion, maternal supplementation of 0.1% SA improved the lactation performance (milk yield and milk fat) of sows, possibly with the activation of GPR41/GPR43-mTORC1 signaling. Furthermore, enhanced milk quality improved growth performance, gut health and the colonization of beneficial microbial flora of their piglets.
Collapse
Affiliation(s)
- Yingao Qi
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
| | - Yongxing Zhong
- Chia Tai Conti Agri-Husbandry Group Co., Ltd, Shenzhen, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
14
|
Chen F, Wang Y, Wang K, Chen J, Jin K, Peng K, Chen X, Liu Z, Ouyang J, Wang Y, Zhang X, Zou H, Zhou J, He B, Lin Q. Effects of Litsea cubeba essential oil on growth performance, blood antioxidation, immune function, apparent digestibility of nutrients, and fecal microflora of pigs. Front Pharmacol 2023; 14:1166022. [PMID: 37465523 PMCID: PMC10350539 DOI: 10.3389/fphar.2023.1166022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
The purpose of this experiment was to investigate the effects of Litsea cubeba essential oil (LCO) on the growth performance, blood antioxidation, immune function, apparent digestibility of nutrients, and fecal microflora in fattening pigs. A total of 120 pigs were randomly assigned to five groups, with six replicate pens per treatment and four pigs per pen, and they were fed basal diet, chlortetracycline (CTC), and low-, medium-, and high-concentration LCO. The results of the study showed that compared with the control treatment and CTC addition treatment of the basic diet, the catalase level in the serum of the pigs treated with 500 mg/kg LCO in the diet of finishing pigs was significantly increased (p < 0.05). The apparent digestibility of crude protein, crude ash, and calcium in pigs with different levels of LCO was significantly increased compared with the control treatments fed the basal diet (p < 0.05). In addition, compared with the control treatment fed the basal diet and the treatment with CTC, the apparent digestibility of ether extract in pigs treated with medium-dose LCO was significantly increased (p < 0.05), and the apparent digestibility of pigs was significantly increased after the addition of low-dose LCO (p < 0.05). Among the genera, the percentage abundance of SMB53 (p < 0.05) was decreased in the feces of the CTC group when compared to that in the medium-LCO group. At the same time, the relative abundance of L7A_E11 was markedly decreased in the feces of the control and medium- and high-concentration LCO group than that in the CTC group (p < 0.05). In conclusion, adding the level of 250 mg/kg LCO in the diet of pig could improve the growth performance and blood physiological and biochemical indicators of pigs, improve the antioxidant level of body and the efficiency of digestion and absorption of nutrients, and show the potential to replace CTC.
Collapse
Affiliation(s)
- Fengming Chen
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Yushi Wang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Kaijun Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Jiayi Chen
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Ke Jin
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Kaiqiang Peng
- Hunan Nuoz Biological Technology Co., Ltd., Yiyang, Hunan, China
| | - Xu Chen
- Hunan Nuoz Biological Technology Co., Ltd., Yiyang, Hunan, China
| | - Zhimou Liu
- Hunan Nuoz Biological Technology Co., Ltd., Yiyang, Hunan, China
| | - Jiang Ouyang
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Yong Wang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Xiaoya Zhang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Haowei Zou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Jun Zhou
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Binsheng He
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Qian Lin
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| |
Collapse
|
15
|
Zhao H, Tian M, Xiong L, Lin T, Zhang S, Yue X, Liu X, Chen F, Zhang S, Guan W. Maternal supplementation with glycerol monolaurate improves the intestinal health of suckling piglets by inhibiting the NF-κB/MAPK pathways and improving oxidative stability. Food Funct 2023; 14:3290-3303. [PMID: 36938595 DOI: 10.1039/d3fo00068k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
Glycerol monolaurate (GML) is a food safe emulsifier and a kind of MCFA monoglyceride that has been proven to confer positive benefits in improving animal health, production and feed digestibility as a feed additive. This study aims to evaluate whether supplementation of a sow diet with GML could affect the intestinal barrier function and antioxidant status of newborn piglets and to explore its regulatory mechanism. A total of 80 multiparous sows were divided into two groups, which were fed a basal diet or a basal diet supplemented with 0.1% GML. The results indicated that maternal supplementation with GML significantly increased fat, lactose and protein in sow colostrum, as well as fat and protein in sow 14-day milk (P < 0.05). The results showed that GML significantly reduced the concentrations of IL-12 in the duodenum, TNF-α, IL-1β and IL-12 in the jejunum, and IL-1β in the ileum of piglets (P < 0.05). Higher concentrations of T-AOC, T-SOD, GSH and GSH-Px and lower MDA in the intestine were observed in the GML group than in the control group. Correspondingly, the villi height, crypt depth and the ratio of villi height to crypt depth (V/C) in the jejunum and the V/C in the ileum in the GML group were significantly higher than those in the control group (P < 0.05). Moreover, the GML group displayed significantly increased protein abundance of zonula occludens (ZO)-1, occludin, and claudin-1 in the small intestine (P < 0.05), mRNA expression of mucins (MUCs) in the small intestine (MUC-1, MUC-3 and MUC-4), and mRNA expression of porcine beta defensins (pBDs) in the duodenum (pBD1 and pBD2), jejunum (pBD1, pBD2 and pBD129) (P < 0.05), and ileum (pBD2, pBD3 and pBD114) (P < 0.05). Further research showed that GML significantly reduced the phosphorylation of the NF-κB/MAPK pathways in the small intestine (P < 0.05). In addition, the results of 16S rDNA sequencing showed that maternal supplementation with GML altered the colonic microbiotic structure of piglets, and reduced the relative abundance of Escherichia shigella. In summary, a sow diet supplemented with GML enhanced the offspring's intestinal oxidative stability and barrier function and attenuated the offspring's intestinal inflammatory response, possibly by suppressing the activation of the NF-κB/MAPK pathways.
Collapse
Affiliation(s)
- Hao Zhao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Min Tian
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Liang Xiong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Tongbin Lin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Shuchang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xianhuai Yue
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xinghong Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| |
Collapse
|
16
|
Su B, Wang Y, Jian S, Tang H, Deng H, Zhu L, Zhao X, Liu J, Cheng H, Zhang L, Hu Y, Xu Z. In vitro and in vivo antiviral activity of monolaurin against Seneca Valley virus. Front Vet Sci 2023; 10:980187. [PMID: 36777661 PMCID: PMC9911909 DOI: 10.3389/fvets.2023.980187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Introduction Surveillance of the Seneca Valley virus (SVV) shows a disproportionately higher incidence on Chinese pig farms. Currently, there are no vaccines or drugs to treat SVV infection effectively and effective treatment options are urgently needed. Methods In this study, we evaluated the antiviral activity of the following medium-chain fatty acids (MCFAs) or triglycerides (MCTs) against SVV: caprylic acid, caprylic monoglyceride, capric monoglyceride, and monolaurin. Results In vitro experiments showed that monolaurin inhibited viral replication by up to 80%, while in vivo studies showed that monolaurin reduced clinical manifestations, viral load, and organ damage in SVV-infected piglets. Monolaurin significantly reduced the release of inflammatory cytokines and promoted the release of interferon-γ, which enhanced the viral clearance activity of this type of MCFA. Discussion Therefore, monolaurin is a potentially effective candidate for the treatment of SVV infection in pigs.
Collapse
Affiliation(s)
- Bo Su
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Yingjie Wang
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Shanqiu Jian
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China
| | - Xiaonan Zhao
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China
| | - Jian Liu
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China
| | - Huangzuo Cheng
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China
| | - Lina Zhang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Youjun Hu
- Innovation Center of Guangdong Nuacid Biotechnology Co., Ltd., Qingyuan, China,*Correspondence: Youjun Hu ✉
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichaun Agricultural University, Chengdu, China,Zhiwen Xu ✉
| |
Collapse
|
17
|
Yi H, Wang Z, Yang B, Yang X, Gao K, Xiong Y, Wu Q, Qiu Y, Hu S, Wang L, Jiang Z. Effects of zinc oxide and condensed tannins on the growth performance and intestinal health of weaned piglets in ETEC-challenged environment. Front Microbiol 2023; 14:1181519. [PMID: 37180229 PMCID: PMC10172512 DOI: 10.3389/fmicb.2023.1181519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
This experiment was conducted to evaluate effects of zine oxide (ZnO) and condensed tannins (CT), independently or in combination, on the growth performance and intestinal health of weaned piglets in enterotoxigenic Escherichia coli (ETEC-K88)-challenged environment. Randomly divided 72 weaned piglets into 4 groups. Dietary treatments included the following: basic diet group (CON), 1,500 mg/kg zinc oxide group (ZnO), 1,000 mg/kg condensed tannins group (CT), and 1,500 mg/kg zinc oxide +1,000 mg/kg condensed tannins group (ZnO + CT). Dietary ZnO supplementation decreased diarrhea rate from 0 to 14 days, 15 to 28 days, and 0 to 28 days (p < 0.05) and no significant on growth performance. The effect of CT on reducing diarrhea rate and diarrhea index was similar to the results of ZnO. Compared with the CON group, ZnO increased the ileum villus height and improved intestinal barrier function by increasing the content of mucin 2 (MUC-2) in jejunum and ileum mucosa and the mRNA expression of zonula occludens-1 (ZO-1) in jejunum (p < 0.05) and the expression of Occludin in duodenum and ileum (p < 0.05). The effects of CT on intestinal barrier function genes were similar to that of ZnO. Moreover, the mRNA expression of cystic fibrosis transmembrane conductance regulator (CFTR) in jejunum and ileum was reduced in ZnO group (p < 0.05). And CT was also capable of alleviating diarrhea by decreasing CFTR expression and promote water reabsorption by increasing AQP3 expression (p < 0.05). In addition, pigs receiving ZnO diet had higher abundance of phylum Bacteroidetes, and genera Prevotella, and lower phylum Firmicutes and genera Lactobacillus in colonic contents. These results indicated that ZnO and CT can alleviate diarrhea and improve intestinal barrier function of weaned pigs in ETEC-challenged environment. In addition, the application of ZnO combined with CT did not show synergistic effects on piglet intestinal health and overall performance. This study provides a theoretical basis for the application of ZnO in weaning piglet production practices, we also explored effects of CT on the growth performance and intestinal health of weaned piglets in ETEC-challenged environment.
Collapse
|
18
|
Chen J, Xia Y, Hu Y, Zhao X, You J, Zou T. A blend of formic acid, benzoic acid, and tributyrin alleviates ETEC K88-induced intestinal barrier dysfunction by regulating intestinal inflammation and gut microbiota in a murine model. Int Immunopharmacol 2023; 114:109538. [PMID: 36502593 DOI: 10.1016/j.intimp.2022.109538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
This study aimed to investigate the effects of an organic acid (OA) blend on intestinal barrier function, intestinal inflammation, and gut microbiota in mice challenged with enterotoxigenic Escherichia coli K88 (ETEC K88). Ninety female Kunming mice (7 weeks old) were randomly allotted to five treatments with six replicates per treatment and three mice per replicate. The five treatments were composed of the non-ETEC K88 challenge group and ETEC K88 challenge + OA blend groups (0, 0.6 %, 1.2 %, and 2.4 % OA blend). The OA blend consisted of 47.5 % formic acid, 47.5 % benzoic acid, and 5 % tributyrin. The feeding trial lasted for 15 days, and mice were intraperitoneally injected with PBS or ETEC K88 solution on day 15. At 24 h post-challenge, one mouse per replicate was selected for sample collection. The results showed that a dosage of 0.6 % OA blend alleviated the ETEC K88-induced intestinal barrier dysfunction, as indicated by the elevated villus height and the ratio of villus height to crypt depth of jejunum, and the reduced serum diamine oxidase (DAO) and D-lactate levels, as well as the up-regulated mRNA levels of ZO-1, Claudin-1, and Occludin in jejunum mucosa of mice. Furthermore, dietary addition with 0.6 % OA blend decreased ETEC K88-induced inflammation response, as suggested by the decreased TNF-α and IL-6 levels, and the increased IgA level in the serum, as well as the down-regulated mRNA level of TNF-α, IL-6, IL-1β, TLR-4, MyD88, and MCP-1 in jejunum mucosa of mice. Regarding gut microbiota, the beta-diversity analysis revealed a remarkable clustering between the 0.6 % OA blend group and the ETEC K88 challenge group. Supplementation of 0.6 % OA blend decreased the relative abundance of Firmicutes, and increased the relative abundance of Bacteroidota, Desulfobacterota, and Verrucomicrobiota of colonic digesta in mice. Also, the butyric acid content in the colonic digesta of mice was increased by dietary 0.6 % OA blend supplementation. Collectively, a dosage of 0.6 % OA blend could alleviate the ETEC K88-induced intestinal barrier dysfunction by regulating intestinal inflammation and gut microbiota of mice.
Collapse
Affiliation(s)
- Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China; Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Yingying Xia
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China; Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Youjun Hu
- Guangdong Nuacid Biotech Co., Ltd, Qingyuan 511500, China
| | - Xiaolan Zhao
- Guangdong Nuacid Biotech Co., Ltd, Qingyuan 511500, China
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China; Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Tiande Zou
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China; Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
19
|
Zinc Laurate Protects against Intestinal Barrier Dysfunction and Inflammation Induced by ETEC in a Mice Model. Nutrients 2022; 15:nu15010054. [PMID: 36615713 PMCID: PMC9824434 DOI: 10.3390/nu15010054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infection is one of the most common bacterial causes of diarrhea in children and young farm animals. Medium-chain fatty acids (MCFAs) have been widely used for their antibacterial and immune functions. However, there is limited information regarding the role of MCFAs chelated with Zn in diarrhea induced by ETEC infection. Here, zinc laurate (ZnLa) was used to evaluate its protective effect in a mice diarrhea model induced by ETEC. A total of 45 ICR-weaned female mice were randomly assigned to marginal zinc deficiency (dZn), dZn, and ETEC infection groups (dZn+ETEC); ETEC infection was co-treated with a low, middle, or high dose of ZnLa (ZnLa LOW+ETEC, ZnLa MID+ETEC, and ZnLa HIGH+ETEC), respectively, to explore the effect and its mechanism of ZnLa on diarrhea and intestinal health of mice challenged with ETEC. To further compare the antibacterial efficiency of ZnLa and ZnSO4 in mice with ETEC infection, a total of 36 ICR-weaned female mice were randomly divided into ZnLa, ZnLa+ETEC, ZnSO4, and ZnSO4 and ETEC infection groups (ZnSO4+ETEC); moreover, the growth curve of ETEC also compared ZnLa and ZnSO4 in vitro. Mice pretreated with ZnLa were effectively guarded against body weight losses and increases in diarrhea scores induced by ETEC. ZnLa pretreatment also prevented intestinal barrier damage and ion transport in mice challenged with ETEC, as evidenced by the fact that the intestinal villus height and the ratio of villus height and crypt depth, tight junction protein, and Na+ absorption were higher, whereas intestinal permeability and anion secretion were lower in mice pretreated with ZnLa. In addition, ZnLa conferred effective protection against ETEC-induced intestinal inflammatory responses, as the increases in protein and mRNAs of proinflammatory cytokines were prevented in serum and jejunum, which was likely associated with the TLR4/MYD88/NF-κB signaling pathway. The increase in ETEC shedding and virulence-related gene expression was prevented in mice with ZnLa pretreatment. Finally, the growth of ETEC and virulence-related gene expression were lower in the ZnLa group than in ZnSO4 with an equal concentration of zinc. These findings suggest that ZnLa is a promising prevention strategy to remedy ETEC infection.
Collapse
|
20
|
Cui C, Wu C, Wang J, Ma Z, Zheng X, Zhu P, Wang N, Zhu Y, Guan W, Chen F. Restored intestinal integrity, nutrients transporters, energy metabolism, antioxidative capacity and decreased harmful microbiota were associated with IUGR piglet's catch-up growth before weanling. J Anim Sci Biotechnol 2022; 13:129. [PMID: 36229888 PMCID: PMC9564052 DOI: 10.1186/s40104-022-00770-8] [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: 04/12/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is a major inducer of higher morbidity and mortality in the pig industry and catch-up growth (CUG) before weanling could significantly restore this negative influence. But there was limited knowledge about the underlying mechanism of CUG occurrence. METHODS Eighty litters of newborn piglets were divided into normal birth weight (NBW) and IUGR groups according to birth weight. At 26 d, those piglets with IUGR but over average body weight of eighty litters of weaned piglets were considered as CUG, and the piglets with IUGR still below average body weight were considered as NCUG. This study was conducted to systemically compare the intestinal difference among NBW, CUG and NCUG weaned piglets considering the crucial role of the intestine for piglet growth. RESULTS The results indicated that the mRNA expression of nutrients (amino acids, glucose, and fatty acids) transporters, and mitochondrial electron transport chain (ETC) I were upregulated in CUG piglets' gut with improved morphology compared with those NCUG, as well as the ratio of P-AMPK/AMPK protein expression which is the indicator of energy metabolism. Meanwhile, CUG piglet's gut showed higher antioxidative capacity with increased SOD and GSH-Px activity, decreased MDA levels, as well as higher mRNA expressions of Nrf2, Keap1, SOD, and GSH-Px. Furthermore, inflammatory parameters including TNF-α, IL-1β, IL-6, and IL-12 factors, and the activation of MAPK and NF-κB signaling pathways were significantly elevated in the NCUG intestine, while the protein expression of ZO-1, Occludin and Claudin-1 was reduced. The alpha diversity of fecal microbiota was higher in CUG piglets in contrast with NCUG piglets, and the increased beneficial bacteria and decreased pathogenic bacteria was also observed in CUG piglets. CONCLUSIONS CUG piglet's intestine showed comprehensive restoration including higher nutrients transport, energy metabolism, antioxidant capacity, and intestinal physical barrier, while lower oxidative stress, inflammatory response, and pathogenic microbiota.
Collapse
Affiliation(s)
- Chang Cui
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Caichi Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jun Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ziwei Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Pengwei Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Nuan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuhua Zhu
- Shenzhen Kingsino Technology CO., LTD, Shenzhen, 518107, China.,Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, 430070, China.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518116, China
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China. .,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China. .,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
21
|
Dahmer PL, Harrison OL, Jones CK. Effects of formic acid and glycerol monolaurate on weanling pig growth performance, fecal consistency, fecal microbiota, and serum immunity. Transl Anim Sci 2022; 6:txac145. [PMID: 36425847 PMCID: PMC9682209 DOI: 10.1093/tas/txac145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/21/2022] [Indexed: 03/05/2024] Open
Abstract
A total of 350 weanling pigs (DNA 400 × 200; initially, 5.67 ± 0.06 kg BW) were used in a 42-day study with 5 pigs per pen and 14 replicate pens per treatment. At weaning, pigs were allotted to pens in a completely randomized design and pens of pigs were randomly assigned to one of five dietary treatments: 1) negative control (CON; standard nursery diet containing only 150 ppm Zn from trace mineral premix and no acidifier); 2) control diet with 3,000 ppm added zinc from ZnO included in phase 1 and 2,000 ppm added zinc from ZnO included in phase 2 (ZnO); 3) control diet with 0.70% formic acid (FA; Amasil NA; BASF, Florham, NJ); 4) control diet with 0.18% glycerol monolaurate (GML; Natural Biologics GML, Natural Biologics, Newfield, NY); and 5) control diet with a 1.0% blend of formic acid and glycerol monolaurate (FORMI; FORMI 3G, ADDCON GmbH, Bitterfeld-Wolfen, Germany). Pigs were fed treatment diets from d 0 to d 28 and were then fed a common diet from d 28 to d 42. From days 0 to 7, pigs fed ZnO or FORMI had increased (P = 0.03) ADG compared to pigs fed CON, with no difference in feed intake (P > 0.05). Overall, pigs fed GML had reduced (P < 0.0001) ADG compared with those fed the CON, ZnO, or FORMI diets. Fecal DM was evaluated from days 7 to 28 and there was a treatment × day interaction (P = 0.04). Pigs fed GML had a lower fecal DM % on day 7, but a higher fecal DM % on days 14 and 21; however, no differences in fecal DM were observed on day 28. Fresh fecal samples were collected from the same randomly selected pig on days 0 and 14 (70 pigs total;14 pigs per treatment) for analysis of fecal microbial populations using 16S rDNA sequencing. Dietary treatment did not significantly impact fecal microbiota at the phyla level, but pigs fed ZnO had an increased relative abundance (P < 0.01) of the family Clostridiaceae. A blood sample was also collected from one pig per pen on days 0 and 14 for analysis of serum IgA, IgG, and TNF-α. There was no evidence that dietary treatment effected IgA, IgG, or TNF-α concentrations. The effect of sampling day was significant (P < 0.05), where circulating IgA and TNF-α was increased and IgG was decreased from days 0 to 14. In summary, there is potential for a blend of formic acid and GML to improve growth performance immediately post-weaning without negatively impacting fecal consistency. Formic acid and GML alone or in combination did not impact fecal microbial populations or serum immune parameters.
Collapse
Affiliation(s)
- Payton L Dahmer
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, USA
| | - Olivia L Harrison
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, USA
| | - Cassandra K Jones
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
22
|
He KJ, Dong JH, Ouyang XM, Huo YN, Cheng XS, Lin Y, Li Y, Gong G, Liu J, Ren JL, Guleng B. Glycerol monolaurate ameliorates DSS-induced acute colitis by inhibiting infiltration of Th17, neutrophils, macrophages and altering the gut microbiota. Front Nutr 2022; 9:911315. [PMID: 36034889 PMCID: PMC9413164 DOI: 10.3389/fnut.2022.911315] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background and aims Inflammatory bowel disease (IBD) places a heavy medical burden on countries and families due to repeated and prolonged attacks, and the incidence and prevalence of IBD are increasing worldwide. Therefore, finding an effective treatment is a matter of great urgency. Glycerol monolaurate (GML), which has a twelve-carbon chain, is a compound naturally found in human breast milk. Some studies have shown that GML has antibacterial and anti-inflammatory effects. However, the specific mechanism of action remains unclear. Methods Acute colitis was established in mice using 3% DSS, and glycerol monolaurate (500 mg·kg-1) was administered for two weeks. QPCR and western blotting were performed to examine the inflammatory status. Mice described were subjected to flow cytometry analysis for immune cell activation. Results GML treated alleviated macroscopic symptoms such as shortened colons, increased spleen weight, and caused weight loss in mice with DSS-induced colitis. In addition, GML decreased the expression of pro-inflammatory factors (NF-α, IL-1β and IL-1α) and increased the expression of anti-inflammatory factors (IL-10 and TGF-β). GML inhibited the activation of the MAPK and NF-κB signalling pathways, improved tissue damage, and increased the expression of intestinal tight junction proteins. In addition, LPMCs extracted from intestinal tissue via flow cytometry showed that GML treatment led to a decrease of Th17 cells, Neutrophils and Macrophages. 16S rDNA sequencing showed that GML increased the abundance of commensal bacterium such as Akkermansia and Lactobacillus murinus. Conclusions We showed that oral administration of GML ameliorated DSS-induced colitis by inhibiting infiltration of Th17 cells, Neutrophils, and Macrophages, protecting the intestinal mucosal barrier and altered the abundance of commensal bacterium. This study provides new insights into the biological function and therapeutic potential of GML in the treatment of IBD.
Collapse
Affiliation(s)
- Ke-Jie He
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,Binhai County People's Hospital, Yancheng, China
| | - Jia-Hui Dong
- Binhai County People's Hospital, Yancheng, China
| | - Xiao-Mei Ouyang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ya-Ni Huo
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao-Shen Cheng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ying Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yue Li
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guoyu Gong
- Cancer Research Center and Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Jingjing Liu
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,Cancer Research Center and Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
23
|
Tian M, Li L, Tian Z, Zhao H, Chen F, Guan W, Zhang S. Glyceryl butyrate attenuates enterotoxigenic Escherichia coli-induced intestinal inflammation in piglets by inhibiting the NF-κB/MAPK pathways and modulating the gut microbiota. Food Funct 2022; 13:6282-6292. [PMID: 35607985 DOI: 10.1039/d2fo01056a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aims of this study were to evaluate whether a diet supplemented with glyceryl butyrate could attenuate the immune-inflammatory response in piglets challenged with enterotoxigenic Escherichia coli (ETEC), and to explore the mechanisms of its regulation. Eighteen weaning piglets were assigned to three diets: basal diet (CON), antibiotics diet (ATB), and 0.5% glyceryl butyrate diet (GB group). Significantly lower concentrations of IL-1β, IL-6 and TNF-α in the jejunum and IL-6 in the ileum were observed in the GB group than that in the CON group (P < 0.05). Moreover, a decreasing trend of IL-1β (P = 0.075) and TNF-α (P = 0.070) was observed in the ileum in the GB group. Correspondingly, the GB group had significantly increased mRNA expression of porcine beta defensins (pBDs) in the jejunum (pBD1, pBD2, pBD114 and pBD129) and ileum (pBD2, pBD3, pBD114 and pBD129) (P < 0.05), and protein abundance of Claudin 1, Occludin, and ZO-1 in the jejunum and ileum (P < 0.05). Further research results showed that the improvement of beta defensins and tight junctions in the GB group was related to the decreased phosphorylation of the NFκB/MAPK pathway. In addition, the results of 16S rDNA sequencing showed that glycerol butyrate supplementation altered the ileal microbiota composition of piglets, increasing the relative abundance of Lactobacillus reuteri, Lactobacillus salivarius, and Lactobacillus agrilis. In summary, glyceryl butyrate attenuated the immune-inflammatory response in piglets challenged with ETEC by inhibiting the NF-κB/MAPK pathways and modulating the gut microbiota, and thus improved piglet intestinal health.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Lilang Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhezhe Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Hao Zhao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
24
|
Ge L, Liu D, Mao X, Liu S, Guo J, Hou L, Chen X, Huang K. Low Dose of Deoxynivalenol Aggravates Intestinal Inflammation and Barrier Dysfunction Induced by Enterotoxigenic Escherichia coli Infection through Activating Macroautophagy/NLRP3 Inflammasomes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3009-3022. [PMID: 35201764 DOI: 10.1021/acs.jafc.1c07834] [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: 06/14/2023]
Abstract
The toxicity of deoxynivalenol (DON) in healthy humans and animals has been extensively studied. However, whether the natural-low-dose DON is scatheless under unhealthy conditions, especially intestinal injury, is unknown. Infection of enterotoxigenic Escherichia coli (ETEC) is a classical intestinal injury model. In this study, we explored the effects of low-dose DON on intestinal injury induced by the ETEC infection and the underlying mechanism in piglets, mice, and IPEC-J2 monolayer cells. Results showed that significant growth slowdown, severe diarrhea, and intestinal damage, bacterial multiplication, and translocation were observed in the experimental group (low-dose DON, 0.75 mg/kg in feed for piglets, and 1 mg/kg body weight for mice, combined with the ETEC infection). Meanwhile, more aggressive intestinal inflammation and barrier dysfunction were observed in animals and IPEC-J2 monolayer cells. Higher expression levels of NLRP3 inflammasome and LC3B were observed in jejunum and IPEC-J2 in the experimental group. After treatment with NLRP3 or caspase1 inhibitors, excessive intestinal inflammation rather than barrier dysfunction in the experimental group was limited. CRISPR-Cas9-mediated knockout of LC3B alleviated intestinal inflammation and barrier dysfunction and also inhibited NLRP3 inflammasome. In conclusion, a low dose of DON aggravates intestinal inflammation and barrier dysfunction induced by the ETEC infection by activating macroautophagy and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xinru Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Junyan Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Lili Hou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| |
Collapse
|
25
|
Zhang Q, Yi D, Ji C, Wu T, Wang M, Guo S, Wang L, Zhao D, Hou Y. Monolaurin Confers a Protective Effect Against Porcine Epidemic Diarrhea Virus Infection in Piglets by Regulating the Interferon Pathway. Front Immunol 2022; 12:797476. [PMID: 35095875 PMCID: PMC8793282 DOI: 10.3389/fimmu.2021.797476] [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: 10/18/2021] [Accepted: 12/27/2021] [Indexed: 01/14/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) has reemerged as the main pathogen of piglets due to its high mutation feature. Monolaurin (ML) is a natural compound with a wide range of antibacterial and antiviral activities. However, the role of ML in PEDV infection is still unknown. This study aimed to evaluate the effect of ML on the growth performance, intestinal function, virus replication and cytokine response in piglets infected with PEDV, and to reveal the mechanism through proteomics analysis. Piglets were orally administrated with ML at a dose of 100 mg/kg·BW for 7 days before PEDV infection. Results showed that although there was no significant effect on the growth performance of piglets, ML administration alleviated the diarrhea caused by PEDV infection. ML administration promoted the recovery of intestinal villi, thereby improving intestinal function. Meanwhile, PEDV replication was significantly inhibited, and PEDV-induced expression of IL-6 and IL-8 were decreased with ML administration. Proteomics analyses showed that 38 proteins were differentially expressed between PEDV and ML+PEDV groups and were significantly enriched in the interferon-related pathways. This suggests ML could promote the restoration of homeostasis by regulating the interferon pathway. Overall, the present study demonstrated ML could confer a protective effect against PEDV infection in piglets and may be developed as a drug or feed additive to prevent and control PEDV disease.
Collapse
Affiliation(s)
- Qian Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Dan Yi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Changzheng Ji
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Tao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Manli Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Shuangshuang Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Lei Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Di Zhao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
26
|
Li L, Wang H, Zhang N, Zhang T, Ma Y. Effects of α-glycerol monolaurate on intestinal morphology, nutrient digestibility, serum profiles, and gut microbiota in weaned piglets. J Anim Sci 2022; 100:6528997. [PMID: 35167667 PMCID: PMC8956132 DOI: 10.1093/jas/skac046] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/12/2022] [Indexed: 12/18/2022] Open
Abstract
This experiment was conducted to investigate the effects of dietary supplementation of α-glycerol monolaurate (α-GML) on the growth performance, nutrient digestibility, serum profiles, intestinal morphology, and gut microbiota of weaned piglets. A total of 96 healthy 28-d-old (Duroc × Landrace × Yorkshire) weaned piglets with body weight of 8.34 ± 0.05 kg were randomly divided into 2 treatment groups with 6 replicate pens and 8 piglets per pen. The control group was fed a basal diet and the experimental group was fed the basal diet supplemented with 1,000 mg/kg α-GML. The experiment lasted for 28 d. Dietary supplementation with α-GML had no effect on average daily gain, average daily feed intake, or gain to feed ratio in piglets (P > 0.05); however, it reduced (P < 0.05) diarrhea rate of piglets on days 15 to 28. The apparent total tract digestibility of dry matter (DM), crude protein (CP), ether extract (EE), and gross energy (GE) on day 14, and DM, organic matter, CP, EE, and GE on day 28 increased (P < 0.05) with α-GML supplementation. Moreover, higher (P < 0.05) glutathione peroxidase activity and interleukin-10 (IL-10) concentration, and lower (P < 0.05) malondialdehyde and tumor necrosis factor-α concentrations were observed in piglets supplemented with α-GML compared with the control group on day 14. Compared with the control group, the villus height/crypt depth in the duodenum and villus height in the jejunum and ileum were significantly greater (P < 0.05) in the α-GML group. Dietary α-GML supplementation significantly increased (P < 0.05) the relative abundance of Firmicutes, while decreasing (P < 0.05) Bacteroidota and Campilobacterota in the cecal contents; significantly increased (P < 0.05) the relative proportion of Lactobacillus and Blautia species, reduced (P < 0.05) Eubacterium_rectale_ATCC_33656, Campylobacter, and uncultured_bacterium_Alloprevotella species. Thus, dietary α-GML supplementation at 1,000 mg/kg reduces diarrhea rate, improves intestinal morphology, nutrient digestibility, antioxidant capacity, and immune status, and ameliorates gut microbiota in weaned piglets.
Collapse
Affiliation(s)
- Longxian Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Huakai Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Nan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Tuan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yongxi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China,Corresponding author:
| |
Collapse
|
27
|
Peng SS, Li Y, Chen Q, Hu Q, He Y, Che L, Jiang PP. Intestinal and Mucosal Microbiome Response to Oral Challenge of Enterotoxigenic Escherichia coli in Weaned Pigs. Pathogens 2022; 11:pathogens11020160. [PMID: 35215105 PMCID: PMC8879466 DOI: 10.3390/pathogens11020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/10/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is closely associated with diarrhoea in children in resource-limited countries. This study aims to investigate the change of the mucosal microbiome and protein expression in the ileum induced by E. coli K88 (ETEC) using pigs as a model. Seven weaned male pigs were orally given ETEC (1 × 109 CFU, n = 7), and the other seven received saline (CON, n = 7). Ileal tissues were obtained 48 hours after the ETEC challenge for both proteomic and mucosal microbiome analyses. Nine proteins were found with altered abundance between the two groups, including a decrease in FABP1 and FABP6, involved in bile acid circulation. The TLR-9 mediated pathway was also affected showing increased transcription of genes SIGIRR and MyD88. Correlations between the ileal proteins and mucosal bacterial taxa found included a positive correlation between Lactobacilllus and PPP3CA (r = 0.9, p < 0.001) and a negative correlation between Prevotella with CTNND1 (r = −0.7, p < 0.01). In conclusion, ETEC infection caused inflammation and impaired the circulation of bile acids and the mucosal microbiome may affect the expression of intestinal proteins. Further studies are needed to explain the exact roles of these affected processes in the pathogenesis of ETEC-triggered diarrhoea.
Collapse
Affiliation(s)
- Shan-Shan Peng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China;
| | - Yingjie Li
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
| | - Qiuhong Chen
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
| | - Qi Hu
- The Neomics Institute, Shenzhen 518122, China;
| | - Ying He
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
| | - Lianqiang Che
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
- Correspondence: (L.C.); (P.-P.J.)
| | - Ping-Ping Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China;
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
- Correspondence: (L.C.); (P.-P.J.)
| |
Collapse
|
28
|
Kong L, Wang Z, Xiao C, Zhu Q, Song Z. Glycerol Monolaurate Ameliorated Intestinal Barrier and Immunity in Broilers by Regulating Intestinal Inflammation, Antioxidant Balance, and Intestinal Microbiota. Front Immunol 2021; 12:713485. [PMID: 34630388 PMCID: PMC8496679 DOI: 10.3389/fimmu.2021.713485] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022] Open
Abstract
This study was conducted to investigate the impact of glycerol monolaurate (GML) on performance, immunity, intestinal barrier, and cecal microbiota in broiler chicks. A total of 360 one-day-old broilers (Arbor Acres) with an average weight of 45.7 g were randomly allocated to five dietary groups as follows: basal diet and basal diets complemented with 300, 600, 900, or 1200 mg/kg GML. Samples were collected at 7 and 14 days of age. Results revealed that feed intake increased (P < 0.05) after 900 and 1200 mg/kg GML were administered during the entire 14-day experiment period. Dietary GML decreased (P < 0.05) crypt depth and increased the villus height-to-crypt depth ratio of the jejunum. In the serum and jejunum, supplementation with more than 600 mg/kg GML reduced (P < 0.05) interleukin-1β, tumor necrosis factor-α, and malondialdehyde levels and increased (P < 0.05) the levels of immunoglobulin G, jejunal mucin 2, total antioxidant capacity, and total superoxide dismutase. GML down-regulate (P < 0.05) jejunal interleukin-1β and interferon-γ expression and increased (P < 0.05) the mRNA level of zonula occludens 1 and occludin. A reduced (P < 0.05) expression of toll-like receptor 4 and nuclear factor kappa-B was shown in GML-treated groups. In addition, GML modulated the composition of the cecal microbiota of the broilers, improved (P < 0.05) microbial diversity, and increased (P < 0.05) the abundance of butyrate-producing bacteria. Spearman’s correlation analysis revealed that the genera Barnesiella, Coprobacter, Lachnospiraceae, Faecalibacterium, Bacteroides, Odoriacter, and Parabacteroides were related to inflammation and intestinal integrity. In conclusion, GML ameliorated intestinal morphology and barrier function in broiler chicks probably by regulating intestinal immune and antioxidant balance, as well as intestinal microbiota.
Collapse
Affiliation(s)
- Linglian Kong
- Department of Animal Science and Technology, Shandong Agricultural University, Taian, China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Ageing, College of Life Sciences, Yantai University, Yantai, China
| | - Chuanpi Xiao
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Qidong Zhu
- Department of Animal Science and Technology, Shandong Agricultural University, Taian, China
| | - Zhigang Song
- Department of Animal Science and Technology, Shandong Agricultural University, Taian, China
| |
Collapse
|
29
|
Ma J, Piao X, Shang Q, Long S, Liu S, Mahfuz S. Mixed organic acids as an alternative to antibiotics improve serum biochemical parameters and intestinal health of weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:737-749. [PMID: 34466678 PMCID: PMC8379140 DOI: 10.1016/j.aninu.2020.11.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/09/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022]
Abstract
The primary aim of this experiment was to critically explore the relationship between the different levels of mixed organic acids (MOA) and growth performance, serum antioxidant status and intestinal health of weaned piglets, as well as to investigate the potential possibility of MOA alternative to antibiotics growth promoters (AGP). A total of 180 healthy piglets (Duroc × [Landrace × Yorkshire]; weighing 7.81 ± 1.51 kg each, weaned at d 28) were randomly divided into 5 treatments: 1) basal diet (CON); 2) CON + chlorinomycin (75 mg/kg) + virginiamycin (15 mg/kg) + guitaromycin (50 mg/kg) (AGP); 3) CON + MOA (3,000 mg/kg) (OA1); 4) CON + MOA (5,000 mg/kg) (OA2); 5) CON + MOA (7,000 mg/kg) (OA3). This study design included 6 replicates per treatment with 6 piglets per pen (barrow:gilt = 1:1) and the experiment was separated into phase 1 (d 1 to 14) and phase 2 (d 15 to 28). In phases 1, 2 and overall, compared with the CON, the feed conversion ratio (FCR) was reduced (P < 0.01) and the average daily gain (ADG) was increased (P < 0.05) in piglets supplemented with AGP, OA1 and OA2. The concentration of serum immunoglobulins G (IgG) was improved (P < 0.05) in piglets supplemented with OA2 in phase 2. In the jejunum and ileum, the villus height:crypt depth ratio was significantly increased (P < 0.01) in piglets fed AGP and OA1. The mRNA expression level of claudin-1 and zonula occludens-1 (ZO-1) (P < 0.01) was up-regulated in piglets supplemented with OA1 and OA2. The piglets fed AGP, OA1 and OA2 showed an increase (P < 0.05) in the content of acetate acid and total volatile fatty acids (TVFA) in the cecum, and butyric acid and TVFA in the colon compared with CON. Also, OA1 lowered (P < 0.05) the content of Lachnospiraceae in piglets. These results demonstrated that MOA at 3,000 or 5,000 mg/kg could be an alternative to antibiotics due to the positive effects on performance, immune parameters, and intestinal health of weaned piglets. However, from the results of the quadratic fitting curve, it is inferred that MOA at a dose of 4,000 mg/kg may produce a better effect.
Collapse
Affiliation(s)
- Jiayu Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiangshu Piao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qinghui Shang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shenfei Long
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Sujie Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shad Mahfuz
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
30
|
Barberis E, Amede E, Tavecchia M, Marengo E, Cittone MG, Rizzi E, Pedrinelli AR, Tonello S, Minisini R, Pirisi M, Manfredi M, Sainaghi PP. Understanding protection from SARS-CoV-2 using metabolomics. Sci Rep 2021; 11:13796. [PMID: 34226622 PMCID: PMC8257707 DOI: 10.1038/s41598-021-93260-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
The COVID-19 pandemic is still raging in most countries. Although the recent mass vaccination campaign has opened a new chapter in the battle against SARS-CoV-2, the world is still far from herd immunity. There is an urgent need to identify healthy people at high risk of contracting COVID-19, as well as supplements and nutraceuticals that can reduce the risk of infection or mitigate symptoms. In the present study, a metabolic phenotype that could protect individuals from SARS-CoV-2 infection or predispose them to developing COVID-19 was investigated. Untargeted metabolomics was performed on serum samples collected from 51 healthcare workers who were in good health at the beginning of the COVID-19 outbreak in Italy, and who were later exposed to the same risk of developing COVID-19. Half of them developed COVID-19 within three weeks of the blood collection. Our results demonstrate the presence of a specific signature associated with protection from SARS-CoV-2. Circulating monolaurin, which has well-known antiviral and antibacterial properties, was higher in protected subjects, suggesting a potential defensive role against SARS-CoV-2 infection; thus, dietary supplements could boost the immune system against this infection. In addition, our data demonstrate that people with higher levels of cholesterol are at higher risk of developing COVID-19. The present study demonstrates that metabolomics can be of great help for developing personalized medicine and for supporting public healthcare strategies. Studies with larger cohorts of subjects are necessary to confirm our findings.
Collapse
Affiliation(s)
- Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Elia Amede
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Matteo Tavecchia
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Micol G Cittone
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Azienda Ospedaliero-Universitaria "Maggiore Della Carità", Novara, Italy
| | - Eleonora Rizzi
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Azienda Ospedaliero-Universitaria "Maggiore Della Carità", Novara, Italy
| | - Anita R Pedrinelli
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Azienda Ospedaliero-Universitaria "Maggiore Della Carità", Novara, Italy
| | - Stelvio Tonello
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Azienda Ospedaliero-Universitaria "Maggiore Della Carità", Novara, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Azienda Ospedaliero-Universitaria "Maggiore Della Carità", Novara, Italy
| |
Collapse
|
31
|
Cox LM, Maghzi AH, Liu S, Tankou SK, Dhang FH, Willocq V, Song A, Wasén C, Tauhid S, Chu R, Anderson MC, De Jager PL, Polgar-Turcsanyi M, Healy BC, Glanz BI, Bakshi R, Chitnis T, Weiner HL. Gut Microbiome in Progressive Multiple Sclerosis. Ann Neurol 2021; 89:1195-1211. [PMID: 33876477 DOI: 10.1002/ana.26084] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE This study was undertaken to investigate the gut microbiome in progressive multiple sclerosis (MS) and how it relates to clinical disease. METHODS We sequenced the microbiota from healthy controls and relapsing-remitting MS (RRMS) and progressive MS patients and correlated the levels of bacteria with clinical features of disease, including Expanded Disability Status Scale (EDSS), quality of life, and brain magnetic resonance imaging lesions/atrophy. We colonized mice with MS-derived Akkermansia and induced experimental autoimmune encephalomyelitis (EAE). RESULTS Microbiota β-diversity differed between MS patients and controls but did not differ between RRMS and progressive MS or differ based on disease-modifying therapies. Disease status had the greatest effect on the microbiome β-diversity, followed by body mass index, race, and sex. In both progressive MS and RRMS, we found increased Clostridium bolteae, Ruthenibacterium lactatiformans, and Akkermansia and decreased Blautia wexlerae, Dorea formicigenerans, and Erysipelotrichaceae CCMM. Unique to progressive MS, we found elevated Enterobacteriaceae and Clostridium g24 FCEY and decreased Blautia and Agathobaculum. Several Clostridium species were associated with higher EDSS and fatigue scores. Contrary to the view that elevated Akkermansia in MS has a detrimental role, we found that Akkermansia was linked to lower disability, suggesting a beneficial role. Consistent with this, we found that Akkermansia isolated from MS patients ameliorated EAE, which was linked to a reduction in RORγt+ and IL-17-producing γδ T cells. INTERPRETATION Whereas some microbiota alterations are shared in relapsing and progressive MS, we identified unique bacteria associated with progressive MS and clinical measures of disease. Furthermore, elevated Akkermansia in MS may be a compensatory beneficial response in the MS microbiome. ANN NEUROL 2021;89:1195-1211.
Collapse
Affiliation(s)
- Laura M Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Amir Hadi Maghzi
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Shirong Liu
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | | | - Fyonn H Dhang
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Valerie Willocq
- Department of Neurology, Harvard Medical School, Harvard University Wyss Institute for Biologically Inspired Engineering, Boston, MA
| | - Anya Song
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Caroline Wasén
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Shahamat Tauhid
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Renxin Chu
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Mark C Anderson
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Philip L De Jager
- Department of Neurology, Columbia University Medical Center, New York, NY
| | - Mariann Polgar-Turcsanyi
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Brian C Healy
- Department of Neurology, Biostatistics Center, Massachusetts General Hospital, Brigham and Women's Hospital, Boston, MA
| | - Bonnie I Glanz
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Rohit Bakshi
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Tanuja Chitnis
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
32
|
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: 4.3] [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
|
33
|
Pastor N, Collado MC, Manzoni P. Phytonutrient and Nutraceutical Action against COVID-19: Current Review of Characteristics and Benefits. Nutrients 2021; 13:464. [PMID: 33573173 PMCID: PMC7910826 DOI: 10.3390/nu13020464] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
The trend toward using phytonutrients and/or nutraceuticals (P/Ns) with the aim of impacting immune health has increased in recent years. The main reason is that properties of P/Ns are associated with possible immunomodulating effects in the prevention and complementary treatment of viral diseases, including COVID-19 and other respiratory infections. In the present review, we assess the scientific plausibility of specific P/Ns for this purpose of preventative and therapeutic interventions against COVID-19, with an emphasis on safety, validity, and evidence of efficacy against other viruses. Five potential candidates have been identified after reviewing available studies (in silico, in vitro, and in vivo) in which certain flavonoids have demonstrated a potential for use as adjuvant therapeutic agents against viral infections, including COVID-19. As these are often better tolerated than pharmacological treatments, their use could be more widely considered if additional detailed studies can validate the existing evidence.
Collapse
Affiliation(s)
- Nitida Pastor
- Department of Medical Affairs, Clinical Research, Mead Johnson Nutrition, Evansville, IN 47721, USA
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Agustin Escardino 7, 46980 Paterna, Valencia, Spain;
| | - Paolo Manzoni
- Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, 13875 Biella, Italy;
- Neonatology and NICU, Sant’Anna Hospital, AOU Città della Salute e della Scienza, 10136 Torino, Italy
| |
Collapse
|
34
|
Li J, Huang L, Yu LT, Tao G, Wang ZY, Hao WZ, Huang JQ. Feruloylated Oligosaccharides Alleviate Central Nervous Inflammation in Mice Following Spinal Cord Contusion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:15490-15500. [PMID: 33170671 DOI: 10.1021/acs.jafc.0c05553] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
As one of the empirical models of the chronic central inflammatory response, a spinal cord injury (SCI) deteriorates the neuronal survival and results in irreversible motor and sensory dysfunction below the injury area. Our previous studies have reported that maize bran feruloylated oligosaccharides (FOs) exert significant anti-inflammatory activities both in diabetes and colitis. However, no direct evidence of FOs alleviating central nervous inflammation was stated. This study aimed to investigate the therapeutic effect of FOs on SCI and its potential mechanism. Our results indicated that 4 weeks of FO administration effectively mitigated the inflammatory response via decreasing the number of microglia (labelled with Iba1), result in the expression of IL-1α, IL-2, IL-6, IL-18 and TNF-α downregulating, but the level of IL-10 and BDNF increases in the injured spinal cord. Moreover, FOs enhanced neuronal survival, ameliorated the scar cavities, and improved behaviors, including Basso mouse scale (BMS) scores and the gait of mice after SCI. Together, these results demonstrated that administration of FOs showed superior functional recovery effects in a SCI model. Also, FOs may modulate inflammatory activities by regulating the expression of proinflammatory factors, decreasing the production of inflammatory cells, and promoting functional recovery through the MAPK pathway following SCI.
Collapse
Affiliation(s)
- Jing Li
- Integrated Chinese and Western Medicine Postdoctoral research station, Jinan University, Guangzhou, Guangdong 510632, China
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
| | - Ling-Tai Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
| | - Gabriel Tao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston 77204, United States
| | - Zi-Ying Wang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, Guangdong 510632, China
| | - Wen-Zhi Hao
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jun-Qing Huang
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| |
Collapse
|
35
|
Tian M, Chen J, Wu Z, Song H, Yang F, Cui C, Chen F, Zhang S, Guan W. Fat Encapsulation Reduces Diarrhea in Piglets Partially by Repairing the Intestinal Barrier and Improving Fatty Acid Transport. Animals (Basel) 2020; 11:ani11010028. [PMID: 33375218 PMCID: PMC7824132 DOI: 10.3390/ani11010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
(1) Background: Nutritional strategies to enhance gut function and reduce the piglet diarrhea rate are critical to increase the growth performance of piglets. The purpose of this study was to investigate whether dietary fat types and/or fat microencapsulation techniques are involved in regulating the fatty acid transport system and the mechanical and immunological barriers of the small intestine. (2) Methods: Three hundred twenty-four weaning piglets were randomly divided into three groups fed a soybean oil diet (SBO, control group, 6.0% soybean oil), palm oil diet (PO, 6.0% palm oil), or encapsulated palm oil diet (EPO, 7.5% encapsulated palm oil). (3) Results: A significantly lower mRNA expression of the claudin was observed in the duodenum and jejunum of the PO group than in the SBO group (p < 0.05). However, the mRNA expression and protein abundance of claudin and ZO-1 in the jejunum of the EPO group were higher (p < 0.05) than in the PO group. Porcine β-defensin (pBD) secretion was not significantly different between the SBO and PO groups (p > 0.05), while the pBD-2 levels were significantly different (p < 0.05). Compared with the PO group, the EPO group exhibited a significantly increased secretion of pBD-2 and pBD-129 in the small intestine (p < 0.05) and pBD-1 in the jejunum and ileum (p < 0.05). The protein abundances of apolipoprotein AIV (Apo AIV) and intestinal fatty acid binding protein (I-FABP) were significantly lower in the PO group than in the SBO group (p < 0.05). Simultaneously, the protein abundances of fatty acid transport protein 4 (FATP4), fatty acid translocase (CD36), and I-FABP were higher in the EPO group than in the PO group. Furthermore, the low digestibility of palm oil (PO group) might negatively regulate intestinal tight junctions, fatty acid transporters, lipoproteins, and β-defensin through the activation of the AMPK/mTORC1 and AMPK/Sirt1/NF-κB pathways. (4) Conclusions: In summary, microencapsulation techniques might alleviate the negative effects of palm oil and help to improve the intestinal fatty acid transport system and barrier function.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Jiaming Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Zhihui Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Hanqing Song
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Fei Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Chang Cui
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (S.Z.); (W.G.); Tel./Fax: +86-20-85284837 (S.Z.); +86-20-85284837 (W.G.)
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (S.Z.); (W.G.); Tel./Fax: +86-20-85284837 (S.Z.); +86-20-85284837 (W.G.)
| |
Collapse
|
36
|
Yang Y, Zhang T, Zhou G, Jiang X, Tao M, Zhang J, Zeng X, Wu Z, Pan D, Guo Y. Prevention of Necrotizing Enterocolitis through Milk Polar Lipids Reducing Intestinal Epithelial Apoptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:7014-7023. [PMID: 32515192 DOI: 10.1021/acs.jafc.0c02629] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Neonatal necrotizing enterocolitis (NEC) is a common and devastating disease. The objective of this research was to investigate the protective mechanisms of milk polar lipids (MPLs) on the attenuation of lipopolysaccharides (LPS)-induced intestinal inflammation and apoptosis. MPLs were extracted from buttermilk and analyzed using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). A neonatal NEC rat model was used to investigate the effects of MPLs on NEC and its underlying mechanisms. Hematoxylin-eosin (H&E) staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL) assay were used to observe intestinal morphological changes and intestinal epithelial cell apoptosis, which showed that MPLs could reduce NEC symptoms and intestinal apoptosis. The expressions of IL-6, IL-8, and TNF-α in the MPL group was significantly downregulated (P < 0.05), and the expression levels of IL-10 were significantly upregulated (P < 0.05). At the same time, MPLs also significantly reduced (P < 0.05) activation of the LPS-induced TLR4/NF-κB signaling pathway. Furthermore, MPLs inhibit apoptosis by reducing the expressions of Bax, caspase-9, and caspase-3 and by increasing the expression of Bcl-2. In conclusion, MPLs could reduce NEC symptoms in mice by inhibiting cell inflammation and protecting against intestinal apoptosis.
Collapse
Affiliation(s)
- Yujie Yang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| | - Tao Zhang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| | - Guangyu Zhou
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| | - Xiaoxiao Jiang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| | - Mingxuan Tao
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| | - Jiaxin Zhang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| | - Xiaoqun Zeng
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Zhen Wu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Daodong Pan
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Yuxing Guo
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu, P. R. China
| |
Collapse
|
37
|
Boyko OO, Brygadyrenko VV. The impact of certain flavourings and preservatives on the survivability of eggs of Ascaris suum and Trichuris suis. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The article describes a laboratory study of nematocidal properties of flavourings with antibacterial effect against Ascaris suum (Goeze, 1782) and Trichuris suis Schrank, 1788. In the experiments, eight concentrations of food additives with antibacterial properties were used: cinnamaldehyde, benzoic acid, formic acid, linalool, citral, β-ionone. Minimum LC50 value for eggs of A. suum was observed while using cinnamaldehyde and benzoic acid – 1.62 ± 0.37% and 1.69 ± 0.14%, and for eggs of T. suis – 0.57 ± 0.03% and 1.80 ± 0.11% respectively. The lowest influence on the development of eggs of nematodes of pigs’ A. suum and T. suis was exerted by formic acid, linalool, citral and β-ionone. In eggs of A. suum and T. suis, larvae formed in 21 and 50 days even during exposure to 3% emulsions of these substances. The strongest negative impact on the eggs of parasitic nematodes was displayed by cinnamaldehyde flavouring. Further study on nematocidal properties of flavourings, as well as their mixtures, would contribute to the development of preparations which would have a strong effect on eggs and larvae of nematodes of animals and humans.
Collapse
|
38
|
Luise D, Correa F, Bosi P, Trevisi P. A Review of the Effect of Formic Acid and Its Salts on the Gastrointestinal Microbiota and Performance of Pigs. Animals (Basel) 2020; 10:E887. [PMID: 32438743 PMCID: PMC7278376 DOI: 10.3390/ani10050887] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 12/20/2022] Open
Abstract
Out of the alternatives to antibiotics and zinc oxide, organic acids, or simply acidifiers, play significant roles, especially in ensuring gut health and the growth performance of pigs. Regarding acidifiers, formic acid and its salts have shown very promising results in weaning, growing and finishing pigs. Although it is known that the main mechanisms by which acidifiers can improve livestock performance and health are related to the regulation of gastrointestinal pH, an improvement in intestinal digestibility and mineral utilization, and their antimicrobial properties against specific pathogens has been observed, while poor consensus remains in relation to the effect of acidifers on bacteria and the complex microbiome. Therefore, the aim of the present review was to critically evaluate the effects of formic acid and its salts on the performance and the gastrointestinal microbiota balance of pigs.
Collapse
Affiliation(s)
- Diana Luise
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 40127 Bologna, Italy; (F.C.); (P.B.); (P.T.)
| | | | | | | |
Collapse
|
39
|
Teng T, Gao F, He W, Fu H, Guo J, Bai G, Shi B. An Early Fecal Microbiota Transfer Improves the Intestinal Conditions on Microflora and Immunoglobulin and Antimicrobial Peptides in Piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:4830-4843. [PMID: 32252520 DOI: 10.1021/acs.jafc.0c00545] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The goal of this study was to investigate the effects of early fecal microbial transfer (FMT) on the microflora of recipient piglets, where Yorkshire newborn piglets and Min sows (an indigenous pig breed in China) were used as the fecal recipients and donors, respectively, to reveal the changes in immunity and development-related functions of the intestinal mucosa driven by FMT. The recipient group was inoculated with fecal microbial fluids from days 1 to 10. On day 21, the relative abundance of the Proteobacteria was reduced; the concentrations of immunoglobulin M (IgM) and immunoglobulin G (IgG) in the jejunal mucosa, and that of IgG in the ileal mucosa of the recipient group, were increased (P < 0.05). On day 40, the relative abundance of the Firmicutes in the recipient group was increased, while that of Bacteroides was decreased. The concentrations of IgG and IgM in the ileal mucosa of the recipient group were increased. FMT protected the intestine by modulating the antimicrobial peptides of the intestinal mucosa (P < 0.05). The results of this study revealed that early FMT can improve the gut microbiota, intestinal mucosal immunity, and intestinal development-related functions of Yorkshire piglets.
Collapse
Affiliation(s)
- Teng Teng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| | - Feng Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| | - Wei He
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| | - Huiyang Fu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| | - Jing Guo
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| | - Guangdong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|