1
|
Jiang Z, Yang M, Su W, Mei L, Li Y, Guo Y, Li Y, Liang W, Yang B, Huang Z, Wang Y. Probiotics in piglet: from gut health to pathogen defense mechanisms. Front Immunol 2024; 15:1468873. [PMID: 39559358 PMCID: PMC11570287 DOI: 10.3389/fimmu.2024.1468873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Various problems and obstacles are encountered during pig farming, especially the weaning phase when switching from liquid to solid feed. Infection by pathogenic bacteria causes damage to the intestinal barrier function of piglets, disrupts the balance of the intestinal microbiota, and destroys the chemical, mechanical, and immune barriers of the intestinal tract, which is one of the main causes of gut inflammation or gut diseases in piglets. The traditional method is to add antibiotics to piglet diets to prevent bacterial infections. However, long-term overuse of antibiotics leads to bacterial resistance and residues in animal products, threatening human health and causing gut microbiota dysbiosis. In this context, finding alternatives to antibiotics to maintain pre- and post-weaning gut health in piglets and prevent pathogenic bacterial infections becomes a real emergency. The utilization of probiotics in piglet nutrition has emerged as a pivotal strategy to promote gut health and defend against pathogenic infections, offering a sustainable alternative to traditional antibiotic usage. This review introduces recent findings that underscore the multifaceted roles of probiotics in enhancing piglet welfare, from fortifying the gut barrier to mitigating the impacts of common bacterial pathogens. Meanwhile, this study introduces the functions of probiotics from different perspectives: positive effects of probiotics on piglet gut health, protecting piglets against pathogen infection, and the mechanisms of probiotics in preventing pathogenic bacteria.
Collapse
Affiliation(s)
- Zipeng Jiang
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
- South China University of Technology, School of Biology and Biological Engineering, Guangzhou, China
| | - Mingzhi Yang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weifa Su
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liang Mei
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
| | - Yuqi Li
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
| | - Yuguang Guo
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
| | - Yangyuan Li
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
| | - Weifan Liang
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
| | - Bo Yang
- South China University of Technology, School of Biology and Biological Engineering, Guangzhou, China
| | - Zhiyi Huang
- Guangdong VTR Bio-tech Co., Ltd, R&D Center, Zhuhai, China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Yu X, Ma F, Dai H, Liu J, Hashem NM, Sun P. Effects of Different Galacto-Oligosaccharide Supplementation on Growth Performance, Immune Function, Serum Nutrients, and Appetite-Related Hormones in Holstein Calves. Animals (Basel) 2023; 13:3366. [PMID: 37958121 PMCID: PMC10649109 DOI: 10.3390/ani13213366] [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: 09/07/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Our previous study showed that early supplementation with 10 g/(d·head) of galacto-oligosaccharides (GOS) in newborn Holstein dairy calves reduced the incidence of diarrhea and improved growth performance and mineral absorption. Since the dose of 10 g/(d·head) was the lowest by dose screening in our previous study, the present study was designed to investigate whether a lower amount of GOS has similar effects on growth performance, immune function, serum nutrients in newborn Holstein heifer calves, and to further investigate its effect on appetite-related hormones. Twenty-eight healthy newborn (1 day of age) Holstein heifers with similar average body weight (41.18 ± 1.90 kg) were randomly divided into four groups (n = 7): the control group (CON group), which received heated raw milk, and three experimental groups, which received heated raw milk supplemented with 2.5 (GOS2.5 group), 5 (GOS5 group), and 10 g/(d·head) (GOS10 group) GOS. All heifer calves were fed the same starter for 28 d. Supplementation with GOS linearly increased the final body weight, average daily gain, and feed efficiency in heifer calves (p < 0.01). Compared with the control group, the average daily gain and feed efficiency of heifer calves were significantly higher in the GOS5 and GOS10 groups than in the control group (p < 0.05). Furthermore, supplementation with GOS quadratically enhanced the starter and total average daily feed intake of the heifers (p < 0.01), especially in the GOS2.5 and GOS5 groups, (p < 0.05 vs. CON). The serum concentration of immunoglobulin A was linearly increased by GOS supplementation (p < 0.05), and the levels in the GOS5 and GOS10 groups were significantly higher than those in the CON group. Meanwhile, GOS linearly decreased serum interleukin-1β and interleukin-6 concentrations (p < 0.05). The serum concentration of triglycerides was also linearly decreased (p < 0.05), whereas total protein and blood urea nitrogen were linearly increased (p < 0.05). Supplementation with GOS linearly decreased the serum concentration of leptin (p < 0.05) but increased cholecystokinin and glucagon-like peptide-1 (p < 0.05). Increasing doses of GOS linearly improved serum calcium and copper concentrations (p < 0.01) and quadratically enhanced the concentration of magnesium, which peaked in the GOS5 group (p < 0.05). In conclusion, GOS supplementation reduced the incidence of diarrhea and improved the growth performance and immune function of Holstein heifer calves.
Collapse
Affiliation(s)
- Xin Yu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fengtao Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Haonan Dai
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junhao Liu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nesrein M. Hashem
- Department of Animal and Fish Production, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt;
| | - Peng Sun
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
3
|
Chen Z, Yang H, Fu H, Wu L, Liu M, Jiang H, Liu Q, Wang Y, Xiong S, Zhou M, Sun X, Chen C, Huang L. Gut bacterial species in late trimester of pregnant sows influence the occurrence of stillborn piglet through pro-inflammation response. Front Immunol 2023; 13:1101130. [PMID: 36741405 PMCID: PMC9890068 DOI: 10.3389/fimmu.2022.1101130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Maternal gut microbiota is an important regulator for the metabolism and immunity of the fetus during pregnancy. Recent studies have indicated that maternal intestinal microbiota is closely linked to the development of fetus and infant health. Some bacterial metabolites are considered to be directly involved in immunoregulation of fetus during pregnancy. However, the detailed mechanisms are largely unknown. In this study, we exploited the potential correlation between the gut microbiota of pregnant sows and the occurrence of stillborn piglets by combining the 16S rRNA gene and metagenomic sequencing data, and fecal metabolome in different cohorts. The results showed that several bacterial species from Bacteroides, potential pathogens, and LPS-producing bacteria exhibited significantly higher abundances in the gut of sows giving birth to stillborn piglets. Especially, Bacteroides fragilis stood out as the key driver in both tested cohorts and showed the most significant association with the occurrence of stillborn piglets in the DN1 cohort. However, several species producing short-chain fatty acids (SCFAs), such as Prevotella copri, Clostridium butyricum and Faecalibacterium prausnitzii were enriched in the gut of normal sows. Functional capacity analysis of gut microbiome revealed that the pathways associated with infectious diseases and immune diseases were enriched in sows giving birth to stillborn piglets. However, energy metabolism had higher abundance in normal sows. Fecal metabolome profiling analysis found that Lysophosphatidylethanolamine and phosphatidylethanolamine which are the main components of cell membrane of Gram-negative bacteria showed significantly higher concentration in stillbirth sows, while SCFAs had higher concentration in normal sows. These metabolites were significantly associated with the stillborn-associated bacterial species including Bacteroides fragilis. Lipopolysaccharide (LPS), IL-1β, IL-6, FABP2, and zonulin had higher concentration in the serum of stillbirth sows, indicating increased intestinal permeability and pro-inflammatory response. The results from this study suggested that certain sow gut bacterial species in late trimester of pregnancy, e.g., an excess abundance of Bacteroides fragilis, produced high concentration of LPS which induced sow pro-inflammatory response and might cause the death of the relatively weak piglets in a farrow. This study provided novel evidences about the effect of maternal gut microbiota on the fetus development and health.
Collapse
Affiliation(s)
| | - Hui Yang
- *Correspondence: Lusheng Huang, ; Congying Chen, ; Hui Yang,
| | | | | | | | | | | | | | | | | | | | - Congying Chen
- *Correspondence: Lusheng Huang, ; Congying Chen, ; Hui Yang,
| | - Lusheng Huang
- *Correspondence: Lusheng Huang, ; Congying Chen, ; Hui Yang,
| |
Collapse
|
4
|
Rodríguez-Sorrento A, Castillejos L, López-Colom P, Cifuentes-Orjuela G, Rodríguez-Palmero M, Moreno-Muñoz JA, Luise D, Trevisi P, Martín-Orúe SM. Effects of the Administration of Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001 and Their Synbiotic Combination With Galacto-Oligosaccharides Against Enterotoxigenic Escherichia coli F4 in an Early Weaned Piglet Model. Front Microbiol 2021; 12:642549. [PMID: 33935999 PMCID: PMC8086512 DOI: 10.3389/fmicb.2021.642549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/23/2021] [Indexed: 01/09/2023] Open
Abstract
We evaluated the potential of multi-strain probiotic (Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001) with or without galacto-oligosaccharides against enterotoxigenic Escherichia coli (ETEC) F4 infection in post-weaning pigs. Ninety-six piglets were distributed into 32 pens assigned to five treatments: one non-challenged (CTR+) and four challenged: control diet (CTR-), with probiotics (>3 × 1010 CFU/kg body weight each, PRO), prebiotic (5%, PRE), or their combination (SYN). After 1 week, animals were orally inoculated with ETEC F4. Feed intake, weight, and clinical signs were recorded. On days 4 and 8 post-inoculation (PI), one animal per pen was euthanized and samples from blood, digesta, and tissues collected. Microbiological counts, ETEC F4 real-time PCR (qPCR) quantification, fermentation products, serum biomarkers, ileal histomorphometry, and genotype for mucin 4 (MUC4) polymorphism were determined. Animals in the PRO group had similar enterobacteria and coliform numbers to the CTR+ group, and the ETEC F4 prevalence, the number of mitotic cells at day 4 PI, and villus height at day 8 PI were between that observed in the CTR+ and CTR- groups. The PRO group exhibited reduced pig major acute-phase protein (Pig-MAP) levels on day 4 PI. The PRE diet group presented similar reductions in ETEC F4 and Pig-MAP, but there was no effect on microbial groups. The SYN group showed reduced fecal enterobacteria and coliform counts after the adaptation week but, after the inoculation, the SYN group showed lower performance and more animals with high ETEC F4 counts at day 8 PI. SYN treatment modified the colonic fermentation differently depending on the MUC4 polymorphism. These results confirm the potential of the probiotic strains and the prebiotic to fight ETEC F4, but do not show any synergy when administered together, at least in this animal model.
Collapse
Affiliation(s)
- Agustina Rodríguez-Sorrento
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lorena Castillejos
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Paola López-Colom
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | - Diana Luise
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Susana María Martín-Orúe
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
5
|
Scorletti E, Afolabi PR, Miles EA, Smith DE, Almehmadi A, Alshathry A, Childs CE, Fabbro SD, Beavis J, Moyses HE, Clough GF, Sethi JK, Patel J, Wright M, Breen DJ, Peebles C, Darekar A, Aspinall R, Fowell AJ, Dowman JK, Nobili V, Targher G, Delzenne NM, Bindels LB, Calder PC, Byrne CD. Synbiotics Alter Fecal Microbiomes, But Not Liver Fat or Fibrosis, in a Randomized Trial of Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology 2020; 158:1597-1610.e7. [PMID: 31987796 PMCID: PMC7613160 DOI: 10.1053/j.gastro.2020.01.031] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/05/2019] [Accepted: 01/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Dysbiosis of the intestinal microbiota has been associated with nonalcoholic fatty liver disease (NAFLD). We investigated whether administration of a synbiotic combination of probiotic and prebiotic agents affected liver fat content, biomarkers of liver fibrosis, and the composition of the fecal microbiome in patients with NAFLD. METHODS We performed a double-blind phase 2 trial of 104 patients with NAFLD in the United Kingdom. Participants (mean age, 50.8 ± 12.6 years; 65% men; 37% with diabetes) were randomly assigned to groups given the synbiotic agents (fructo-oligosaccharides, 4 g twice per day, plus Bifidobacterium animalis subspecies lactis BB-12; n = 55) or placebo (n = 49) for 10-14 months. Liver fat content was measured at the start and end of the study by magnetic resonance spectroscopy, and liver fibrosis was determined from a validated biomarker scoring system and vibration-controlled transient elastography. Fecal samples were collected at the start and end of the study, the fecal microbiome were analyzed by 16S ribosomal DNA sequencing. RESULTS Mean baseline and end-of-study magnetic resonance spectroscopy liver fat percentage values were 32.3% ± 24.8% and 28.5% ± 20.1% in the synbiotic group and 31.3% ± 22% and 25.2% ± 17.2% in the placebo group. In the unadjusted intention-to-treat analysis, we found no significant difference in liver fat reduction between groups (β = 2.8; 95% confidence interval, -2.2 to 7.8; P = .30). In a fully adjusted regression model (adjusted for baseline measurement of the outcome plus age, sex, weight difference, and baseline weight), only weight loss was associated with a significant decrease in liver fat (β = 2; 95% confidence interval, 1.5-2.6; P = .03). Fecal samples from patients who received the synbiotic had higher proportions of Bifidobacterium and Faecalibacterium species, and reductions in Oscillibacter and Alistipes species, compared with baseline; these changes were not observed in the placebo group. Changes in the composition of fecal microbiota were not associated with liver fat or markers of fibrosis. CONCLUSIONS In a randomized trial of patients with NAFLD, 1 year of administration of a synbiotic combination (probiotic and prebiotic) altered the fecal microbiome but did not reduce liver fat content or markers of liver fibrosis. (ClinicalTrials.gov, Number: NCT01680640).
Collapse
Affiliation(s)
- Eleonora Scorletti
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom; Department of Gastroenterology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania.
| | - Paul R. Afolabi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| | - Elizabeth A. Miles
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Debbie E. Smith
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| | - Amal Almehmadi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Albandri Alshathry
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Caroline E. Childs
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Stefania Del Fabbro
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Josh Beavis
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Helen E. Moyses
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| | - Geraldine F. Clough
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jaswinder K. Sethi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Janisha Patel
- Hepatology, Department of Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mark Wright
- Hepatology, Department of Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - David J. Breen
- Department of Radiology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Charles Peebles
- Department of Radiology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Angela Darekar
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Richard Aspinall
- Department of Hepatology, Portsmouth Hospitals NHS Trust, Queen Alexandra Hospital, Portsmouth, UK
| | - Andrew J. Fowell
- Department of Hepatology, Portsmouth Hospitals NHS Trust, Queen Alexandra Hospital, Portsmouth, UK
| | - Joanna K. Dowman
- Department of Hepatology, Portsmouth Hospitals NHS Trust, Queen Alexandra Hospital, Portsmouth, UK
| | - Valerio Nobili
- Hepatology, Gastroenterology and Nutrition Unit, IRCCS "Bambino Gesù" Children's Hospital, Rome, Italy,Department of Pediatric, University "La Sapienza", Rome, Italy
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Philip C. Calder
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK,Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christopher D. Byrne
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| |
Collapse
|
6
|
Zhong S, Zhou Z, Liang Y, Cheng X, Li Y, Teng W, Zhao M, Liu C, Guan M, Zhao C. Targeting strategies for chemotherapy-induced peripheral neuropathy: does gut microbiota play a role? Crit Rev Microbiol 2019; 45:369-393. [PMID: 31106639 DOI: 10.1080/1040841x.2019.1608905] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a progressive, often irreversible condition that produces severe neurological deficits. Emerging data suggest that chemotherapy also exerts detrimental effects on gut microbiota composition and intestinal permeability, contributing to dysbiosis and inflammation. Compared with other complications associated with chemotherapy, such as diarrhoea and mucositis, CIPN is of particular concern because it is the most common reason for terminating or suspending treatment. However, specific and effective curative treatment strategies are lacking. In this review, we provide an update on current preclinical and clinical understandings about the role of gut microbiota in CIPN. The gut microbiota serves as an intersection between the microbiome-gut-brain and the neuroimmune-endocrine axis, forming a complex network that can directly or indirectly affect key components involved in the manifestations of CIPN. Herein, we discuss several potential mechanisms within the context of the networks and summarize alterations in gut microbiome induced by chemotherapeutic drugs, providing great potential for researchers to target pathways associated with the gut microbiome and overcome CIPN.
Collapse
Affiliation(s)
- Shanshan Zhong
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Zhike Zhou
- Department of Geriatrics, The First Hospital of China Medical University , Shenyang , PR China
| | - Yifan Liang
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Xi Cheng
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University , Shenyang , PR China
| | - Weiyu Teng
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Mei Zhao
- Department of Cardiology, Shengjing Hospital of China Medical University , Shenyang , PR China
| | - Chang Liu
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Meiting Guan
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Chuansheng Zhao
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| |
Collapse
|
7
|
Barba-Vidal E, Martín-Orúe SM, Castillejos L. Practical aspects of the use of probiotics in pig production: A review. Livest Sci 2019. [DOI: 10.1016/j.livsci.2019.02.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
8
|
Brugman S, Ikeda-Ohtsubo W, Braber S, Folkerts G, Pieterse CMJ, Bakker PAHM. A Comparative Review on Microbiota Manipulation: Lessons From Fish, Plants, Livestock, and Human Research. Front Nutr 2018; 5:80. [PMID: 30234124 PMCID: PMC6134018 DOI: 10.3389/fnut.2018.00080] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
During recent years the impact of microbial communities on the health of their host (being plants, fish, and terrestrial animals including humans) has received increasing attention. The microbiota provides the host with nutrients, induces host immune development and metabolism, and protects the host against invading pathogens (1-6). Through millions of years of co-evolution bacteria and hosts have developed intimate relationships. Microbial colonization shapes the host immune system that in turn can shape the microbial composition (7-9). However, with the large scale use of antibiotics in agriculture and human medicine over the last decades an increase of diseases associated with so-called dysbiosis has emerged. Dysbiosis refers to either a disturbed microbial composition (outgrowth of possible pathogenic species) or a disturbed interaction between bacteria and the host (10). Instead of using more antibiotics to treat dysbiosis there is a need to develop alternative strategies to combat disturbed microbial control. To this end, we can learn from nature itself. For example, the plant root (or "rhizosphere") microbiome of sugar beet contains several bacterial species that suppress the fungal root pathogen Rhizoctonia solani, an economically important fungal pathogen of this crop (11). Likewise, commensal bacteria present on healthy human skin produce antimicrobial molecules that selectively kill skin pathogen Staphylococcus aureus. Interestingly, patients with atopic dermatitis (inflammation of the skin) lacked antimicrobial peptide secreting commensal skin bacteria (12). In this review, we will give an overview of microbial manipulation in fish, plants, and terrestrial animals including humans to uncover conserved mechanisms and learn how we might restore microbial balance increasing the resilience of the host species.
Collapse
Affiliation(s)
- Sylvia Brugman
- Cell Biology and Immunology Group, Animal Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Corné M. J. Pieterse
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| | - Peter A. H. M. Bakker
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
9
|
Fomenky BE, Chiquette J, Lessard M, Bissonnette N, Talbot G, Chouinard YP, Ibeagha-Awemu EM. Saccharomyces cerevisiae var. boulardii CNCM I-1079 and Lactobacillus acidophilus BT1386 influence innate immune response and serum levels of acute-phase proteins during weaning in Holstein calves. CANADIAN JOURNAL OF ANIMAL SCIENCE 2018. [DOI: 10.1139/cjas-2017-0120] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The aims of this study were to investigate the effect of Saccharomyces cerevisiae var. boulardii CNCM I-1079 (SCB) or Lactobacillus acidophilus BT1386 (LA) on (1) innate immune response, (2) markers of acute-phase reaction, and (3) immune gene expression of rumen and ileum tissues of Holstein calves. Forty eight calves (∼5 d old) were randomly allocated to four treatments as follows: (1) control (CTRL) fed milk replacer followed by starter feed, (2) CTRL supplemented with SCB in milk and feed, (3) CTRL supplemented with LA in milk and feed, and (4) CTRL supplemented with antibiotics (ATB; chlortetracycline and neomycin in milk, and chlortetracycline in feed). Tumor necrosis factor α (TNF-α) decreased (P < 0.05) on day 66 (post-weaning) for the ATB-treated calves. There were no treatment effects on production of interferon γ (IFN-γ) and interleukin 6 (IL-6) proteins and on expression of TLR4, TLR6, TLR9, TLR10, CLDN3, MUC1, and MUC20 genes. Calves fed SCB or LA had a greater (P < 0.05) oxidative burst at weaning (day 53) compared with CTRL. Oxidative burst was also greater (P < 0.05) after weaning (day 59 and day 87) for SCB-fed calves. Calves fed SCB and ATB had higher (P < 0.05) phagocytosis activity during weaning (day 47) compared with CTRL. The concentration of serum amyloid A2 (SAA2) increased (P < 0.05) in SCB- and LA-fed calves (day 53), whereas the concentration of C-reactive protein (CRP) increased (P < 0.05) in SCB-fed calves during weaning as compared with CTRL. Our results suggest that SCB could improve innate immune response (oxidative burst and phagocytosis) and markers of acute-phase reaction (CRP and SAA2), especially during critical periods like weaning.
Collapse
Affiliation(s)
- Bridget E. Fomenky
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC J1M 0C8, Canada
- Département des Sciences Animales, Université Laval, Québec, QC G1V 0A6, Canada
| | - Johanne Chiquette
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC J1M 0C8, Canada
| | - Martin Lessard
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC J1M 0C8, Canada
| | - Nathalie Bissonnette
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC J1M 0C8, Canada
| | - Guylaine Talbot
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC J1M 0C8, Canada
| | - Yvan P. Chouinard
- Département des Sciences Animales, Université Laval, Québec, QC G1V 0A6, Canada
| | - Eveline M. Ibeagha-Awemu
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC J1M 0C8, Canada
| |
Collapse
|
10
|
Arenas‐Padilla M, Duarte‐Gutiérrez J, Mata‐Haro V. Bifidobacterium animalis ssp. lactis Bb12 induces IL-10 through cell membrane-associated components via TLR2 in swine. J Appl Microbiol 2018; 125:1881-1889. [PMID: 30106205 PMCID: PMC7166459 DOI: 10.1111/jam.14069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/06/2018] [Accepted: 08/09/2018] [Indexed: 01/22/2023]
Abstract
AIM To investigate the role of Toll-like receptor 2 (TLR2) in interleukin-10 (IL-10) production induced by Bifidobacterium animalis ssp. lactis Bb12 (Bb12) in swine immune cells. METHODS AND RESULTS Blood-monocytes and cells from mesenteric lymph nodes were obtained from pigs and cultured with live Bb12 for 4 and 12 h. Transcript levels of IL-10 and TLR2 were analysed. Furthermore, TLR2 was blocked to determine its participation in IL-10 production. TLR2 blockade was achieved with neutralizing antibodies, followed by stimulation with Bb12. Bifidobacteria induced IL-10 production in both swine monocytes and mesenteric cells. Monocytes with TLR2 blockade had a decrease in IL-10 transcripts, while mesenteric cells did not. Bacterial cell wall components were responsible for Bb12-induced IL-10 production since no IL-10 was detected in the culture supernatant. CONCLUSIONS We demonstrated that IL-10 production is largely mediated through the recognition of Bb12 structures by TLR2, as bacterial metabolites in the culture supernatant failed to induce IL-10 expression. SIGNIFICANCE AND IMPACT OF THE STUDY The present study provides evidence for the potential use of Bb12 in the swine industry; these bacteria can also be used as additional method to treat intestinal inflammation and enhance intestinal health in pigs.
Collapse
Affiliation(s)
- M. Arenas‐Padilla
- Department of Food Science, Microbiology and ImmunologyCentro de Investigación en Alimentación y Desarrollo, A. C.HermosilloMéxico
| | - J.L. Duarte‐Gutiérrez
- Department of Food Science, Microbiology and ImmunologyCentro de Investigación en Alimentación y Desarrollo, A. C.HermosilloMéxico
| | - V. Mata‐Haro
- Department of Food Science, Microbiology and ImmunologyCentro de Investigación en Alimentación y Desarrollo, A. C.HermosilloMéxico
| |
Collapse
|
11
|
Abstract
Intensive farming may involve the use of diets, environments or management practices that impose physiological and psychological stressors on the animals. In particular, early weaning is nowadays a common practice to increase the productive yield of pig farms. Still, it is considered one of the most critical periods in swine production, where piglet performance can be seriously affected and where they are predisposed to the overgrowth of opportunistic pathogens. Pig producers nowadays face the challenge to overcome this situation in a context of increasing restrictions on the use of antibiotics in animal production. Great efforts are being made to find strategies to help piglets overcome the challenges of early weaning. Among them, a nutritional strategy that has received increasing attention in the last few years is the use of probiotics. It has been extensively documented that probiotics can reduce digestive disorders and improve productive parameters. Still, research in probiotics so far has also been characterized as being inconsistent and with low reproducibility from farm to farm. Scientific literature related to probiotic effects against gastrointestinal pathogens will be critically examined in this review. Moreover, the actual practical approach when using probiotics in these animals, and potential strategies to increase consistency in probiotic effects, will be discussed. Thus, considering the boost in probiotic research observed in recent years, this paper aims to provide a much-needed, in-depth review of the scientific data published to-date. Furthermore, it aims to be useful to swine nutritionists, researchers and the additive industry to critically consider their approach when developing or using probiotic strategies in weaning piglets.
Collapse
|
12
|
Mach N, Fuster-Botella D. Endurance exercise and gut microbiota: A review. JOURNAL OF SPORT AND HEALTH SCIENCE 2017; 6:179-197. [PMID: 30356594 PMCID: PMC6188999 DOI: 10.1016/j.jshs.2016.05.001] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/25/2016] [Accepted: 03/14/2016] [Indexed: 05/17/2023]
Abstract
BACKGROUND The physiological and biochemical demands of intense exercise elicit both muscle-based and systemic responses. The main adaptations to endurance exercise include the correction of electrolyte imbalance, a decrease in glycogen storage and the increase of oxidative stress, intestinal permeability, muscle damage, and systemic inflammatory response. Adaptations to exercise might be influenced by the gut microbiota, which plays an important role in the production, storage, and expenditure of energy obtained from the diet as well as in inflammation, redox reactions, and hydration status. METHODS A systematic and comprehensive search of electronic databases, including MEDLINE, Scopus, ClinicalTrials.gov, ScienceDirect, Springer Link, and EMBASE was done. The search process was completed using the keywords: "endurance", "exercise", "immune response", "microbiota", "nutrition", and "probiotics". RESULTS Reviewed literature supports the hypothesis that intestinal microbiota might be able to provide a measureable, effective marker of an athlete's immune function and that microbial composition analysis might also be sensitive enough to detect exercise-induced stress and metabolic disorders. The review also supports the hypothesis that modifying the microbiota through the use of probiotics could be an important therapeutic tool to improve athletes' overall general health, performance, and energy availability while controlling inflammation and redox levels. CONCLUSION The present review provides a comprehensive overview of how gut microbiota may have a key role in controlling the oxidative stress and inflammatory responses as well as improving metabolism and energy expenditure during intense exercise.
Collapse
Affiliation(s)
- Núria Mach
- Health Science Department, International Graduate Institute of the Open University of Catalonia (UOC), Barcelona 08035, Spain
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParis Tech, University of Paris-Saclay, Jouy-en-Josas 78350, France
- Corresponding author.
| | - Dolors Fuster-Botella
- Health Science Department, International Graduate Institute of the Open University of Catalonia (UOC), Barcelona 08035, Spain
| |
Collapse
|
13
|
Barba-Vidal E, Castillejos L, López-Colom P, Rivero Urgell M, Moreno Muñoz JA, Martín-Orúe SM. Evaluation of the Probiotic Strain Bifidobacterium longum subsp. Infantis CECT 7210 Capacities to Improve Health Status and Fight Digestive Pathogens in a Piglet Model. Front Microbiol 2017; 8:533. [PMID: 28443068 PMCID: PMC5386966 DOI: 10.3389/fmicb.2017.00533] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 03/14/2017] [Indexed: 01/12/2023] Open
Abstract
Probiotics have been demonstrated to be useful to enhance gut health and prevent gastrointestinal infections. The objective of this study is to demonstrate the potential of the probiotic strain Bifidobacterium longum subsp. infantis CECT 7210 (B. infantis IM1) to prevent and fight intestinal disease by using a Salmonella Typhimurium (Trial 1) or an enterotoxigenic Escherichia coli K88 (Trial 2) oral challenge in a weaning piglet model. Seventy-two piglets were used in each trial. After an adaptation period, animals were orally challenged. One animal per pen was euthanized at Days 4 and 8/9 (Trial 1/Trial 2) post-inoculation (PI). Animal performance, clinical signs, pathogen excretion, fermentation, immune response, and intestinal morphology were evaluated. In Trial 1, most parameters responded to the challenge, whereas, in Trial 2, effects were much milder. Consistent effects of the probiotic were detected in both experiments: Reduction of pathogen excretion (P = 0.043 on Day 3 PI, Trial 1) or ileal colonization (33% reduction of animals with countable coliforms; P = 0.077, Trial 2); increases in intraepithelial lymphocytes (P = 0.002 on Day 8 PI in Trial 1, P = 0.091 on Day 4 PI in Trial 2), and improvement of the fermentation profile by increasing butyric acid in non-challenged animals [P challenge × probiotic (interaction) = 0.092 in Trial 1 and P = 0.056 in Trial 2] concomitant with an enhancement of the villus:crypt ratio on Day 8/9 PI (P interaction = 0.091 for Trial 1 and P = 0.006 for Trial 2). Challenged animals treated with the probiotic showed reduced feed intakes (P interaction = 0.019 in Trial 1 and P = 0.020 in Trial 2) and had lower short-chain fatty acid concentrations in the colon (P interaction = 0.008 in Trial 1 and P = 0.082 in Trial 2). In conclusion, this probiotic demonstrated potential to reduce the intestinal colonization by pathogens and to stimulate local immune response. However, effects on feed intake, microbial fermentation, and intestinal architecture showed a differential pattern between challenged and non-challenged animals. Effects of the probiotic intervention were dependent on the structure of the ecosystem in which it was applied.
Collapse
Affiliation(s)
- Emili Barba-Vidal
- Animal Nutrition and Welfare Service, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de BarcelonaBellaterra, Spain
| | - Lorena Castillejos
- Animal Nutrition and Welfare Service, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de BarcelonaBellaterra, Spain
| | - Paola López-Colom
- Animal Nutrition and Welfare Service, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de BarcelonaBellaterra, Spain
| | | | | | - Susana M Martín-Orúe
- Animal Nutrition and Welfare Service, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de BarcelonaBellaterra, Spain
| |
Collapse
|
14
|
Larskaya IA, Gorshkova TA. Plant oligosaccharides - outsiders among elicitors? BIOCHEMISTRY (MOSCOW) 2016; 80:881-900. [PMID: 26542002 DOI: 10.1134/s0006297915070081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This review substantiates the need to study the plant oligoglycome. The available information on oligosaccharins - physiologically active fragments of plant cell wall polysaccharides - is summarized. The diversity of such compounds in chemical composition, origin, and proved biological activity is highlighted. At the same time, plant oligosaccharides can be considered as outsiders among elicitors of various natures in research intensity of recent decades. This review discusses the reasons for such attitude towards these regulators, which are largely connected with difficulties in isolation and identification. Together with that, approaches are suggested whose potentials can be used to study oligosaccharins. The topics of oligosaccharide metabolism in plants, including the ways of formation, transport, and inactivation are presented, together with data on biological activity and interaction with plant hormones. The current viewpoints on the mode of oligosaccharin action - perception, signal transduction, and possible "targets" - are considered. The potential uses of such compounds in medicine, food industry, agriculture, and biotechnology are discussed.
Collapse
Affiliation(s)
- I A Larskaya
- Kazan Institute of Biochemistry and Biophysics, Kazan Scientific Center, Russian Academy of Sciences, Kazan, 420111, Russia.
| | | |
Collapse
|
15
|
Wang H, Geier MS, Howarth GS. Prebiotics: A Potential Treatment Strategy for the Chemotherapy-damaged Gut? Crit Rev Food Sci Nutr 2016; 56:946-956. [PMID: 25162145 DOI: 10.1080/10408398.2012.741082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mucositis, characterized by ulcerative lesions along the alimentary tract, is a common consequence of many chemotherapy regimens. Chemotherapy negatively disrupts the intestinal microbiota, resulting in increased numbers of potentially pathogenic bacteria, such as Clostridia and Enterobacteriaceae, and decreased numbers of "beneficial" bacteria, such as Lactobacilli and Bifidobacteria. Agents capable of restoring homeostasis in the bowel microbiota could, therefore, be applicable to mucositis. Prebiotics are indigestible compounds, commonly oligosaccharides, that seek to reverse chemotherapy-induced intestinal dysbiosis through selective colonization of the intestinal microbiota by probiotic bacteria. In addition, evidence is emerging that certain prebiotics contribute to nutrient digestibility and absorption, modulate intestinal barrier function through effects on mucin expression, and also modify mucosal immune responses, possibly via inflammasome-mediated processes. This review examines the known mechanisms of prebiotic action, and explores their potential for reducing the severity of chemotherapy-induced mucositis in the intestine.
Collapse
Affiliation(s)
- Hanru Wang
- a School of Animal and Veterinary Sciences, University of Adelaide , Roseworthy Campus , South Australia
| | - Mark S Geier
- a School of Animal and Veterinary Sciences, University of Adelaide , Roseworthy Campus , South Australia
- b South Australian Research and Development Institute, Pig and Poultry Production Institute, Nutrition Research Laboratory , Roseworthy , South Australia
| | - Gordon S Howarth
- a School of Animal and Veterinary Sciences, University of Adelaide , Roseworthy Campus , South Australia
- c Centre for Paediatric and Adolescent Gastroenterology, Children, Youth and Women's Health Service , North Adelaide , South Australia
| |
Collapse
|
16
|
Priori D, Trevisi P, Mazzoni M, Messori S, Bosi P. Effect of fasting and refeeding on expression of genes involved in the gastric nutrient sensing and orexigenic control of pigs. J Anim Physiol Anim Nutr (Berl) 2014; 99:692-700. [DOI: 10.1111/jpn.12265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 09/22/2014] [Indexed: 01/20/2023]
Affiliation(s)
- D. Priori
- Agri-Food Science & Technology Department; University of Bologna; Reggio Emilia Italy
| | - P. Trevisi
- Agri-Food Science & Technology Department; University of Bologna; Reggio Emilia Italy
| | - M. Mazzoni
- Department of Veterinary Medical Sciences; University of Bologna; Ozzano Emilia Bologna Italy
| | - S. Messori
- Agri-Food Science & Technology Department; University of Bologna; Reggio Emilia Italy
| | - P. Bosi
- Agri-Food Science & Technology Department; University of Bologna; Reggio Emilia Italy
| |
Collapse
|
17
|
Plaza-Diaz J, Gomez-Llorente C, Fontana L, Gil A. Modulation of immunity and inflammatory gene expression in the gut, in inflammatory diseases of the gut and in the liver by probiotics. World J Gastroenterol 2014; 20:15632-15649. [PMID: 25400447 PMCID: PMC4229528 DOI: 10.3748/wjg.v20.i42.15632] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/04/2014] [Accepted: 06/21/2014] [Indexed: 02/06/2023] Open
Abstract
The potential for the positive manipulation of the gut microbiome through the introduction of beneficial microbes, as also known as probiotics, is currently an active area of investigation. The FAO/WHO define probiotics as live microorganisms that confer a health benefit to the host when administered in adequate amounts. However, dead bacteria and bacterial molecular components may also exhibit probiotic properties. The results of clinical studies have demonstrated the clinical potential of probiotics in many pathologies, such as allergic diseases, diarrhea, inflammatory bowel disease and viral infection. Several mechanisms have been proposed to explain the beneficial effects of probiotics, most of which involve gene expression regulation in specific tissues, particularly the intestine and liver. Therefore, the modulation of gene expression mediated by probiotics is an important issue that warrants further investigation. In the present paper, we performed a systematic review of the probiotic-mediated modulation of gene expression that is associated with the immune system and inflammation. Between January 1990 to February 2014, PubMed was searched for articles that were published in English using the MeSH terms "probiotics" and "gene expression" combined with "intestines", "liver", "enterocytes", "antigen-presenting cells", "dendritic cells", "immune system", and "inflammation". Two hundred and five original articles matching these criteria were initially selected, although only those articles that included specific gene expression results (77) were later considered for this review and separated into three major topics: the regulation of immunity and inflammatory gene expression in the gut, in inflammatory diseases of the gut and in the liver. Particular strains of Bifidobacteria, Lactobacilli, Escherichia coli, Propionibacterium, Bacillus and Saccharomyces influence the gene expression of mucins, Toll-like receptors, caspases, nuclear factor-κB, and interleukins and lead mainly to an anti-inflammatory response in cultured enterocytes. In addition, the interaction of commensal bacteria and probiotics with the surface of antigen-presenting cells in vitro results in the downregulation of pro-inflammatory genes that are linked to inflammatory signaling pathways, whereas other anti-inflammatory genes are upregulated. The effects of probiotics have been extensively investigated in animal models ranging from fish to mice, rats and piglets. These bacteria induce a tolerogenic and hyporesponsive immune response in which many genes that are related to the immune system, in particular those genes expressing anti-inflammatory cytokines, are upregulated. By contrast, information related to gene expression in human intestinal cells mediated by the action of probiotics is scarce. There is a need for further clinical studies that evaluate the mechanism of action of probiotics both in healthy humans and in patients with chronic diseases. These types of clinical studies are necessary for addressing the influence of these microorganisms in gene expression for different pathways, particularly those that are associated with the immune response, and to better understand the role that probiotics might have in the prevention and treatment of disease.
Collapse
|
18
|
Age-related expression of the polymeric immunoglobulin receptor (pIgR) in the gastric mucosa of young pigs. PLoS One 2013; 8:e81473. [PMID: 24236214 PMCID: PMC3827463 DOI: 10.1371/journal.pone.0081473] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/23/2013] [Indexed: 11/19/2022] Open
Abstract
To date few studies have addressed the development and function of the porcine gastric mucosal immune system and this is a major limitation to understanding the immunopathogenesis of infections occurring in young pigs. The polymeric immunoglobulin receptor (pIgR) mediates the transport of secretory immunoglobulins until luminal surface of the gut mucosa and the aim of this study was to investigate the time course of pIgR expression and to determine its localization in three functionally different porcine gastric sites during the suckling period and after weaning. An additional goal was to investigate the time course expression of toll-like receptors (TLRs) in relation to pIgR expression. Gastric samples were collected from the cardiac-to-oxyntic transition (Cd), the oxyntic (Ox), and the pyloric (Py) regions in 84 pigs, slaughtered before weaning (14, 21 and 28 days of age; 23, 23 and 19 pigs, respectively) and 14 days post-weaning (42 days of age, 23 pigs). PIgR was expressed in the mucosa of all the three gastric sites, and its transcript levels were modulated during suckling and after weaning, with regional differences. PIgR expression increased linearly during suckling (P=0.019) and also increased post-weaning (P=0.001) in Cd, it increased post-weaning in Py (P=0.049) and increased linearly during suckling in Ox (P=0.036). TLRs expression was also modulated during development: in Cd, TLR2 increased linearly during suckling (P=0.003); in Ox, TLR2 decreased after weaning (P=0.038) while TLR4 increased linearly during suckling(P=0.008). The expression of TLR2, 3 and 4 in Ox was positively correlated with pIgR expression (P<0.001). Importantly, both pIgR protein and mRNA were localized, by immunohistochemistry and in situ hybridization, respectively, in the gastric glands of the lamina propria. These results indicate that pIgR is actively synthesized in the gastric mucosa and suggest that pIgR could play a crucial role in gastric mucosal immune defense of growing pigs.
Collapse
|
19
|
Latest concepts on the association between nonsteroidal anti-inflammatory drug-induced small intestinal injury and intestinal bacterial flora. Clin J Gastroenterol 2013; 6:345-51. [DOI: 10.1007/s12328-013-0424-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 08/23/2013] [Indexed: 12/16/2022]
|
20
|
Kavanagh K, Wylie AT, Tucker KL, Hamp TJ, Gharaibeh RZ, Fodor AA, Cullen JMC. Dietary fructose induces endotoxemia and hepatic injury in calorically controlled primates. Am J Clin Nutr 2013; 98:349-57. [PMID: 23783298 PMCID: PMC3712547 DOI: 10.3945/ajcn.112.057331] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Controversy exists regarding the causative role of dietary fructose in obesity and fatty liver diseases. Clinical trials have indicated that negative health consequences may occur only when fructose is consumed within excess calories. Animal studies have suggested that fructose impairs intestinal integrity and leads to hepatic steatosis (HS). OBJECTIVES We assessed nonhuman primates after chronic ad libitum and short-term calorically controlled consumption of a high-fructose (HFr), low-fat diet (24% of calories). Microbial translocation (MT), microbiome, and metabolic health indexes were evaluated. DESIGN Seventeen monkeys fed 0.3–7 y of an HFr ad libitum diet were compared with 10 monkeys fed a low-fructose, low-fat diet (control). Ten middle-aged, weight-stable, fructose-naive monkeys were stratified into HFr and control groups fed for 6 wk at caloric amounts required to maintain weight stability. Metabolic endpoints, feces, liver, small and large intestinal biopsies, and portal blood samples were collected. RESULTS Monkeys allowed ad libitum HFr developed HS in contrast to the control diet, and the extent of ectopic fat was related to the duration of feeding. Diabetes incidence also increased. Monkeys that consumed calorically controlled HFr showed significant increases in biomarkers of liver damage, endotoxemia, and MT indexes and a trend for greater hepatitis that was related to MT; however, HS did not develop. CONCLUSIONS Even in the absence of weight gain, fructose rapidly causes liver damage that we suggest is secondary to endotoxemia and MT. HS relates to the duration of fructose consumption and total calories consumed. These data support fructose inducing both MT and ectopic fat deposition in primates.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Department of Pathology Section on Comparative Medicine and Lipid Sciences, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27127, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Pang G, Xie J, Chen Q, Hu Z. How functional foods play critical roles in human health. FOOD SCIENCE AND HUMAN WELLNESS 2012. [DOI: 10.1016/j.fshw.2012.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
22
|
Abstract
The delivery of certain living microorganisms in food has long been suggested as having positive health effects in humans. This practice has extended into food animal production, with a variety of microorganisms being used; lactic acid bacteria, various Bacillus species and the yeast Saccharomyces cerevisiae have been particularly used in the pig industry. The increased interest in probiotics is essentially due to the problem of microbial resistance to antibiotics and following the ban of the use of antibiotics in animal production, probiotics being considered an alternative means to reduce pathogen infection and improve animal health especially around the time of weaning. However, there is still a need to clarify the probiotic effectiveness in pigs, and the underlying mechanisms. When assessing the efficacy of probiotics one must consider the particular strain of organism being used and the production stage of the pigs being treated. The reproducible delivery of probiotics in industrial pig production is problematic as maintenance of viability is key to their beneficial activity, but difficult to achieve with commonly used feed processing technologies. One specific context where probiotics organisms may be reliably delivered is in systems utilising fermented liquid feeds. Liquid feed may be fermented by the activity of wild lactic acid bacteria or may be stimulated using specific isolates as 'starters'; the latter system has advantages in terms of reproducibility and speed of fermentation. The farm context in which the organism is used is likely to be critical; the use of probiotics is more likely to result in measurable economic gains in animals living in sub-optimal conditions rather than in those reared in the highest welfare and husbandry conditions. The establishment of a beneficial lactic acid bacteria population at birth may lead to healthier animals, this may be most effectively achieved by treating sows, which provide an amplification step and flood the neonatal pigs' environment with desirable bacterial strains. In contrast, it may be sufficient to provide a supportive, protective microbiota around the time of weaning as this is a time of major crisis with instability and loss of certain bacterial populations.
Collapse
|
23
|
Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science 2012; 336:1262-7. [PMID: 22674330 DOI: 10.1126/science.1223813] [Citation(s) in RCA: 3170] [Impact Index Per Article: 243.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The composition and activity of the gut microbiota codevelop with the host from birth and is subject to a complex interplay that depends on the host genome, nutrition, and life-style. The gut microbiota is involved in the regulation of multiple host metabolic pathways, giving rise to interactive host-microbiota metabolic, signaling, and immune-inflammatory axes that physiologically connect the gut, liver, muscle, and brain. A deeper understanding of these axes is a prerequisite for optimizing therapeutic strategies to manipulate the gut microbiota to combat disease and improve health.
Collapse
Affiliation(s)
- Jeremy K Nicholson
- Biomolecular Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | | | | | | | | | | | | |
Collapse
|
24
|
Heo JM, Opapeju FO, Pluske JR, Kim JC, Hampson DJ, Nyachoti CM. Gastrointestinal health and function in weaned pigs: a review of feeding strategies to control post-weaning diarrhoea without using in-feed antimicrobial compounds. J Anim Physiol Anim Nutr (Berl) 2012; 97:207-37. [PMID: 22416941 DOI: 10.1111/j.1439-0396.2012.01284.x] [Citation(s) in RCA: 498] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
For the last several decades, antimicrobial compounds have been used to promote piglet growth at weaning through the prevention of subclinical and clinical disease. There are, however, increasing concerns in relation to the development of antibiotic-resistant bacterial strains and the potential of these and associated resistance genes to impact on human health. As a consequence, European Union (EU) banned the use of antibiotics as growth promoters in swine and livestock production on 1 January 2006. Furthermore, minerals such as zinc (Zn) and copper (Cu) are not feasible alternatives/replacements to antibiotics because their excretion is a possible threat to the environment. Consequently, there is a need to develop feeding programs to serve as a means for controlling problems associated with the weaning transition without using antimicrobial compounds. This review, therefore, is focused on some of nutritional strategies that are known to improve structure and function of gastrointestinal tract and (or) promote post-weaning growth with special emphasis on probiotics, prebiotics, organic acids, trace minerals and dietary protein source and level.
Collapse
Affiliation(s)
- J M Heo
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | |
Collapse
|
25
|
Burcelin R, Serino M, Chabo C, Blasco-Baque V, Amar J. Gut microbiota and diabetes: from pathogenesis to therapeutic perspective. Acta Diabetol 2011; 48:257-273. [PMID: 21964884 PMCID: PMC3224226 DOI: 10.1007/s00592-011-0333-6] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 09/01/2011] [Indexed: 12/13/2022]
Abstract
More than several hundreds of millions of people will be diabetic and obese over the next decades in front of which the actual therapeutic approaches aim at treating the consequences rather than causes of the impaired metabolism. This strategy is not efficient and new paradigms should be found. The wide analysis of the genome cannot predict or explain more than 10-20% of the disease, whereas changes in feeding and social behavior have certainly a major impact. However, the molecular mechanisms linking environmental factors and genetic susceptibility were so far not envisioned until the recent discovery of a hidden source of genomic diversity, i.e., the metagenome. More than 3 million genes from several hundreds of species constitute our intestinal microbiome. First key experiments have demonstrated that this biome can by itself transfer metabolic disease. The mechanisms are unknown but could be involved in the modulation of energy harvesting capacity by the host as well as the low-grade inflammation and the corresponding immune response on adipose tissue plasticity, hepatic steatosis, insulin resistance and even the secondary cardiovascular events. Secreted bacterial factors reach the circulating blood, and even full bacteria from intestinal microbiota can reach tissues where inflammation is triggered. The last 5 years have demonstrated that intestinal microbiota, at its molecular level, is a causal factor early in the development of the diseases. Nonetheless, much more need to be uncovered in order to identify first, new predictive biomarkers so that preventive strategies based on pre- and probiotics, and second, new therapeutic strategies against the cause rather than the consequence of hyperglycemia and body weight gain.
Collapse
Affiliation(s)
- Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1048, Toulouse, France.
- Université de Toulouse, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), 31432, Toulouse cedex 4, France.
| | - Matteo Serino
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1048, Toulouse, France
- Université de Toulouse, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), 31432, Toulouse cedex 4, France
| | - Chantal Chabo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1048, Toulouse, France
- Université de Toulouse, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), 31432, Toulouse cedex 4, France
| | - Vincent Blasco-Baque
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1048, Toulouse, France
- Université de Toulouse, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), 31432, Toulouse cedex 4, France
| | - Jacques Amar
- Department of Therapeutics, Rangueil Hospital, Toulouse, France
| |
Collapse
|
26
|
Hummelen R, Vos AP, van't Land B, van Norren K, Reid G. Altered host-microbe interaction in HIV: a target for intervention with pro- and prebiotics. Int Rev Immunol 2011; 29:485-513. [PMID: 20839912 DOI: 10.3109/08830185.2010.505310] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The intestinal immune system is severely affected by HIV and circulating microbial products from the intestinal tract that provide an ongoing source of systemic inflammation and concomitant viral replication. In addition, HIV-infected individuals can have a deregulated immune response that may hamper the anti-viral capacity of the host. Various probiotic organisms and prebiotic agents have been shown to enhance intestinal epithelial barrier functions, reduce inflammation, and support effective Th-1 responses. As these characteristics may benefit HIV patients, this review aims to provide a theoretical framework for the development of probiotic and prebiotic interventions specifically for this population.
Collapse
Affiliation(s)
- Ruben Hummelen
- Department of Public Health, Erasmus MC, University Medical Centre Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
27
|
Bosi P, Trevisi P. New topics and limits related to the use of beneficial microbes in pig feeding. Benef Microbes 2010; 1:447-54. [DOI: 10.3920/bm2010.0036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Reports highlighting the positive effects of probiotics on the performance of pigs or on in vitro traits are now quite frequent, but the use of probiotics in feed compounds has not been widespread. Prerequisites for the healthy and efficient growth of young pigs are the rapid maturation of the gut mucosa and the mucosa-associated lymphoid tissue, and the formation of a local stable and complex bacterial community. In neonatal pigs, suckling and the maternal environment shape the gut microbiota. Later, when weaning stress causes a transient drop in favourable bacteria, the oral supply of microbes could contribute to re-establish the microbiota balance. Some strains isolated from piglets were tested for their ability to settle in the intestine. After weaning, piglets experience new and often unfavourable bacteria. Probiotics have been investigated to contrast the enteropathogens, owing to their properties (production of antibacterial molecules, competition on adhesion sites, stimulation of immune response, etc.). Data in general show that their oral administration can be favourable or, at least, innocuous. However, two cases are presented here, where a probiotic given to pigs already combating enteropathogens impaired pig health, and this could be explained by their effect on the immune response. A more tolerogenic response of the host is expected when beneficial bacteria directly contrast the pathogens, probiotics are claimed to directly modulate or even activate the immune system. For one probiotic divergent effects on growth and health are presented, and these differences may be due to different experimental details or different starting microbiological environments. Scarce data are available on specific immune responses induced by commensal microbes in pigs, and on the interaction of resident microbiota with orally supplied probiotics. Increased knowledge of the role of commensal microbiota in the gut and in the pig metabolism, helps in selecting the best bacteria and in designing the best feeding strategies for improving the efficacy and the reliability of their oral use.
Collapse
Affiliation(s)
- P. Bosi
- DIPROVAL, University of Bologna, Via F.lli Rosselli 107, 42100 Reggio Emilia, Italy
| | - P. Trevisi
- DIPROVAL, University of Bologna, Via F.lli Rosselli 107, 42100 Reggio Emilia, Italy
| |
Collapse
|
28
|
Sanz Y, De Palma G. Gut microbiota and probiotics in modulation of epithelium and gut-associated lymphoid tissue function. Int Rev Immunol 2010; 28:397-413. [PMID: 19954356 DOI: 10.3109/08830180903215613] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intestinal tract mucosa is exposed to a vast number of environmental antigens and a large community of commensal bacteria. The mucosal immune system has to provide both protection against pathogens and tolerance to harmless bacteria. Immune homeostasis depends on the interaction of indigenous commensal and transient bacteria (probiotics) with various components of the epithelium and the gut-associated lymphoid tissue. Herein, an update is given of the mechanisms by which the gut microbiota and probiotics are translocated through the epithelium, sensed via pattern-recognition receptors, and activate innate and adaptive immune responses.
Collapse
Affiliation(s)
- Yolanda Sanz
- Microbial Ecophysiology and Nutrition Group, Institute of Agrochemistry and Food Technology (IATA), Spanish National Research Council (CSIC), Burjassot-Valencia, Spain.
| | | |
Collapse
|
29
|
Courtois J. Oligosaccharides from land plants and algae: production and applications in therapeutics and biotechnology. Curr Opin Microbiol 2009; 12:261-73. [DOI: 10.1016/j.mib.2009.04.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 04/21/2009] [Accepted: 04/27/2009] [Indexed: 01/02/2023]
|
30
|
Fujimori S, Gudis K, Mitsui K, Seo T, Yonezawa M, Tanaka S, Tatsuguchi A, Sakamoto C. A randomized controlled trial on the efficacy of synbiotic versus probiotic or prebiotic treatment to improve the quality of life in patients with ulcerative colitis. Nutrition 2009; 25:520-525. [PMID: 19201576 DOI: 10.1016/j.nut.2008.11.017] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 11/08/2008] [Accepted: 11/11/2008] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Studies suggest that synbiotic therapy could prove more effective in the treatment of ulcerative colitis (UC) than therapies limited to probiotics or prebiotics. This study compared the effect of each of these therapies in the treatment of UC. METHODS One hundred twenty outpatients with UC were randomly sorted into three groups of 40 patients each for probiotic, prebiotic, or synbiotic therapy. The probiotic group ingested one daily capsule consisting of Bifidobacterium longum 2 x 10(9) colony-forming units and the prebiotic group ingested daily 8.0-g doses of psyllium. The synbiotic group underwent both treatments. All patients completed Inflammatory Bowel Disease Questionnaires (IBDQs) at the onset of the trial, at the 2-wk midpoint, and at the 4-wk end of the trial. Blood variables were also evaluated in a subset of 32 patients randomly selected from all groups and values were compared with IBDQ scores. RESULTS Thirty-one patients in the probiotic group, 31 in the prebiotic group, and 32 in the synbiotic group qualified for analyses. The remaining 26 patients had incomplete questionnaires. Total IBDQ scores improved within groups by the end of the trial (probiotics 162 to 169, NS; prebiotics 174 to 182, NS; synbiotics 168 to 176, P = 0.03). Individual scores improved as follows: probiotics, emotional function (P = 0.03); prebiotics, bowel function (P = 0.04); and synbiotics, systemic and social functions (P = 0.008 and P = 0.02). C-reactive protein decreased significantly only with synbiotic therapy (from 0.59 to 0.14 mg/dL, P = 0.04). There were no adverse events. CONCLUSION Patients with UC on synbiotic therapy experienced greater quality-of-life changes than patients on probiotic or prebiotic treatment. These data suggest that synbiotic therapy may have a synergistic effect in the treatment of UC.
Collapse
Affiliation(s)
- Shunji Fujimori
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|