1
|
Smolinska S, Popescu FD, Zemelka-Wiacek M. A Review of the Influence of Prebiotics, Probiotics, Synbiotics, and Postbiotics on the Human Gut Microbiome and Intestinal Integrity. J Clin Med 2025; 14:3673. [PMID: 40507435 PMCID: PMC12156228 DOI: 10.3390/jcm14113673] [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: 02/21/2025] [Revised: 05/01/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
OBJECTIVE This review aims to comprehensively evaluate the current evidence on the role of prebiotics, probiotics, synbiotics, and postbiotics-collectively referred to as "biotics"-in modulating the human gut microbiota and enhancing intestinal epithelial integrity. FINDINGS Biotics exert their beneficial effects through several mechanisms, including by promoting the growth of beneficial microbes, producing short-chain fatty acids (SCFAs), strengthening the gut barrier, and regulating immune responses. Prebiotics selectively stimulate beneficial bacteria, probiotics introduce live microorganisms with therapeutic functions, synbiotics combine the strengths of both, and postbiotics offer non-viable microbial components and metabolites that mimic probiotic benefits with enhanced safety profiles. Each type of biotic demonstrates unique and complementary effects across a range of conditions, such as inflammatory bowel disease, irritable bowel syndrome, obesity, constipation, and antibiotic-associated diarrhea. IMPLICATIONS As disruptions in the gut microbiota and intestinal barrier are increasingly linked to chronic and immune-mediated diseases, leveraging biotics offers promising avenues for personalized nutrition, preventive healthcare, and adjunct therapies. The integration of biotics into clinical and dietary strategies may significantly contribute to improving gastrointestinal and systemic health.
Collapse
Affiliation(s)
- Sylwia Smolinska
- Department of Clinical Immunology, Faculty of Medicine, Wroclaw Medical University, 51-616 Wroclaw, Poland;
| | - Florin-Dan Popescu
- Department of Allergology, Nicolae Malaxa Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Magdalena Zemelka-Wiacek
- Department of Clinical Immunology, Faculty of Medicine, Wroclaw Medical University, 51-616 Wroclaw, Poland;
| |
Collapse
|
2
|
Shi Y, Liu J, Zhou H, Wu Z, Qiu Y, Ye C. Dihydromyricetin alleviates ETEC K88-induced intestinal inflammatory injury by inhibiting quorum sensing-related virulence factors. BMC Microbiol 2025; 25:201. [PMID: 40205366 PMCID: PMC11980137 DOI: 10.1186/s12866-025-03879-8] [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: 01/16/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) is responsible for piglet diarrhea and causes substantial economic loss in the pig industry. Along with the restriction of antibiotics, natural compounds targeting bacterial virulence factors are supposed to be efficacious and attractive alternatives for controlling ETEC infection. This study aimed to investigate the influence of dihydromyricetin (DMY), a natural flavonoid compound, on the expression of virulence factors of ETEC and intestinal inflammatory injury. RESULTS DMY interfered with the quorum sensing (QS) of ETEC K88 since it decreased AI-2 secretion and downregulated the expression of LuxS and Pfs, which dominate AI-2 production, and decreased the expression mRNA level of genes (lsrA, lsrB, lsrC, lsrD, lsrK, and lsrR) that are involved in AI-2 internalization and signal transduction. Additionally, DMY markedly dampened the expression of QS-related virulence genes (elt-1, estB, fliC, faeG), biofilm formation, cell adhesion, and stress tolerance of ETEC K88. Furthermore, DMY treatment applied to the ETEC K88 infection in mice model resulted in decreased amount of heat-labile (LT) and heat-stable (ST) enterotoxins, reduced production of cAMP and cGMP, downregulated protein level of CFTR and upregulated expression of NHE3 in the ileum. In addition, the mRNA expression of proinflammatory cytokines (TNF-α, IL-1β, and IL-6) and histological damage in the ileum were significantly decreased by DMY treatment. CONCLUSIONS DMY can inhibit the AI-2 QS and virulence factor expression, thereby attenuating the virulence of ETEC and alleviating intestinal inflammatory damage in ETEC K88-challenged mice. This study indicated that DMY has the potential to be a promising antivirulence agent for combating ETEC infection.
Collapse
Affiliation(s)
- Yaqian Shi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Jin Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Hualin Zhou
- Agricultural College, Xiangyang Polytechnic, Xiangyang, China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China.
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China.
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
- Wuhan Engineering and Technology Research Center of Animal Disease-resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
3
|
Hou XW, Meng J, Chen XT, Zhao JX, Shang KM, Wei YJ, Liu R. Bacillus safensis M01 reversed the inflammatory injury of mice jejunum caused by enterotoxigenic Escherichia coli K88. Arch Microbiol 2025; 207:87. [PMID: 40087175 DOI: 10.1007/s00203-025-04287-2] [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: 01/14/2025] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major pathogen causing neonatal diarrhea in livestock, with antibiotics commonly used for control. However, antibiotic overuse has led to issues such as residues and bacterial resistance, underscoring the need for alternative prevention strategies. This study investigated the potential of Bacillus safensis (B. safensis) M01, isolated from healthy porcine feces in Shandong, China, to prevent ETEC infections. M01 exhibited over 80% inhibition of ETEC in vitro and was selected for further analysis. Pre-treatment of IPEC-J2 cells with M01 significantly reduced ETEC-induced cellular damage, enhanced cell viability, and inhibited bacterial adhesion. It modulated inflammatory responses by down-regulating IL-1β and TNF-α while up-regulating IL-10. Additionally, M01 promoted the expression of tight junction proteins, including Claudin-1, Occludin, and ZO-1. In the C57BL/6 mouse model, pre-feeding with M01 for 14 days improved jejunal injury caused by ETEC, as indicated by increased villus height/crypt depth ratios. Similar to in vitro findings, M01 reduced IL-1β and TNF-α expression while enhancing tight junction protein levels. These results suggest that B. safensis M01 is a promising probiotic candidate for preventing ETEC infections in livestock, offering an effective alternative to antibiotics.
Collapse
Affiliation(s)
- Xin-Wen Hou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Jinxin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Xiao-Tong Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China.
| |
Collapse
|
4
|
Jin Y, Gong T, Lu X, Wang Y, Cheng Y. Effects of gut microbiota and metabolites on the host defense peptide expression. Appl Microbiol Biotechnol 2025; 109:10. [PMID: 39825892 PMCID: PMC11742865 DOI: 10.1007/s00253-024-13400-2] [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: 11/27/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/20/2025]
Abstract
The widespread use of antibiotics has led to the emergence of multidrug-resistant bacteria, which pose significant threats to animal health and food safety. Host defense peptides (HDPs) have emerged as promising alternatives because of their unique antimicrobial properties and minimal resistance induction. However, the high costs associated with HDP production and incorporation into animal management practices hinder their widespread application. Alternatively, promoting endogenous HDP expression has gained attention as a sustainable and cost-effective approach. This study summarizes the latest research findings on the modulation of HDP expression by the gut microbiota and its metabolites. By exploring the intricate relationships among the gut microbiota, metabolites, and HDP expression, this study aims to provide a theoretical foundation for the development of targeted strategies to increase endogenous HDP production, thereby promoting animal health and resistance to infectious diseases. KEY POINTS: • Host defense peptides (HDPs) are expressed via various factors, such as nutrients, the gut microbiota, and microbial metabolites. • Recent trends include mechanisms among the gut microbiota, microbiota metabolites, and the intestine on HDP expression. • A comprehensive overview of mechanisms of HDP expression and gut microbiota-host interaction is provided.
Collapse
Affiliation(s)
- Yuanli Jin
- National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Hangzhou, 310058, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Hangzhou, 310058, Zhejiang, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, Zhejiang, China
| | - Tao Gong
- National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Hangzhou, 310058, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Hangzhou, 310058, Zhejiang, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, Zhejiang, China
| | - Xiaoxi Lu
- National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Hangzhou, 310058, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Hangzhou, 310058, Zhejiang, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, Zhejiang, China
| | - Yizhen Wang
- National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Hangzhou, 310058, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Hangzhou, 310058, Zhejiang, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, Zhejiang, China
| | - Yuanzhi Cheng
- National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Hangzhou, 310058, Zhejiang, China.
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Hangzhou, 310058, Zhejiang, China.
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, Zhejiang, China.
| |
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
|
Varada VV, Kumar S, Balaga S, Thanippilly AJ, Pushpadass HA, M RH, Jangir BL, Tyagi N, Samanta AK. Oral delivery of electrohydrodynamically encapsulated Lactiplantibacillus plantarum CRD7 modulates gut health, antioxidant activity, and cytokines-related inflammation and immunity in mice. Food Funct 2024; 15:10761-10781. [PMID: 39390885 DOI: 10.1039/d4fo02732a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The current study aimed to evaluate the effects of L. plantarum CRD7 on performance and gut health biomarkers in a Swiss albino mouse model. The results showed that supplementation with non-encapsulated (NLP) and electrohydrodyanamically encapsulated L. plantarum CRD7 (ELP) for four weeks significantly increased (P < 0.05) body weight and weekly feed intake of mice. Specifically, these interventions strengthened the gut barrier functions, as evidenced by the increased expression of tight junction proteins (claudin-1, ZO-1, and occludin), inhibiting pro-inflammatory factors (TNF-α, MCP-1, and IL-6), and promoting short-chain fatty acid production. Histopathological examination revealed no probiotic-related adverse effects in liver and intestinal tissues. Furthermore, ELP and NLP possess the ability to regulate immunity and antioxidant capacity in mice. Notably, the supplementation of ELP modified the gut microbiota by promoting beneficial bacteria (Lactobacillus and Bifibacterium) and suppressing pathogenic bacteria (E. coli and C. perfringens), thereby restoring a balanced gut microbiota. Taken together, oral delivery of encapsulated L. plantarum CRD7 can modify the composition of the gut microbiota, fortify the intestinal barrier functions, maintain the gastrointestinal equilibrium, and augment the immune and antioxidant capacity. This comprehensive study provides valuable insights for the potential application of encapsulated probiotic products in food and feed formulations aimed at alleviating gut diseases.
Collapse
Affiliation(s)
- Vinay Venkatesh Varada
- Rumen Biotechnology Lab., Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| | - Sachin Kumar
- Rumen Biotechnology Lab., Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| | - Sravani Balaga
- Rumen Biotechnology Lab., Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| | - Antony Johnson Thanippilly
- Rumen Biotechnology Lab., Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| | - Heartwin A Pushpadass
- Dairy Engineering Section, ICAR-National Dairy Research Institute, Southern Regional Station, Bengaluru, India.
| | - Rashmi H M
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| | - Babu Lal Jangir
- Department of Veterinary Pathology, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar-125004, India.
| | - Nitin Tyagi
- Rumen Biotechnology Lab., Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| | - Ashish Kumar Samanta
- Rumen Biotechnology Lab., Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| |
Collapse
|
7
|
Yoon KN, Yang J, Yeom SJ, Kim SS, Park JH, Song BS, Eun JB, Park SH, Lee JH, Kim HB, Lee JH, Kim JK. Lactiplantibacillus argentoratensis AGMB00912 protects weaning mice from ETEC infection and enhances gut health. Front Microbiol 2024; 15:1440134. [PMID: 39318427 PMCID: PMC11420142 DOI: 10.3389/fmicb.2024.1440134] [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: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Maintaining a healthy intestinal environment, optimal epithelial barrier integrity, and balanced gut microbiota composition are essential for the growth performance of weaning pigs. We identified Lactiplantibacillus argentoratensis AGMB00912 (LA) in healthy porcine feces as having antimicrobial activity against pathogens and enhanced short-chain fatty acid (SCFA) production. Herein, we assess the protective role of LA using a weaning mouse model with enterotoxigenic Escherichia coli (ETEC) infection. LA treatment improves feed intake and weight gain and alleviates colon shortening. Furthermore, LA inhibits intestinal damage, increases the small intestine villus height compared with the ETEC group, and enhances SCFA production. Using the Kyoto Encyclopedia of Genes and Genomes and other bioinformatic tools, including InterProScan and COGNIZER, we validated the presence of SCFA-producing pathways of LA and Lactiplantibacillus after whole genome sequencing. LA mitigates ETEC-induced shifts in the gut microbiota, decreasing the proportion of Escherichia and Enterococcus and increasing SCFA-producing bacteria, including Kineothrix, Lachnoclostridium, Roseuburia, Lacrimispora, Jutongia, and Blautia. Metabolic functional prediction analysis revealed enhanced functions linked to carbohydrate, amino acid, and vitamin biosynthesis, along with decreased functions associated with infectious bacterial diseases compared to the ETEC group. LA mitigates the adverse effects of ETEC infection in weaning mice, enhances growth performance and intestinal integrity, rebalances gut microbiota, and promotes beneficial metabolic functions. These findings validate the functionality of LA in a small animal model, supporting its potential application in improving the health and growth performance of weaning pigs.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Jihye Yang
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Sang-Su Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Heum Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Beom-Seok Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Ju-Hoon Lee
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| |
Collapse
|
8
|
Filidou E, Kandilogiannakis L, Shrewsbury A, Kolios G, Kotzampassi K. Probiotics: Shaping the gut immunological responses. World J Gastroenterol 2024; 30:2096-2108. [PMID: 38681982 PMCID: PMC11045475 DOI: 10.3748/wjg.v30.i15.2096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/28/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
Probiotics are live microorganisms exerting beneficial effects on the host's health when administered in adequate amounts. Among the most popular and adequately studied probiotics are bacteria from the families Lactobacillaceae, Bifidobacteriaceae and yeasts. Most of them have been shown, both in vitro and in vivo studies of intestinal inflammation models, to provide favorable results by means of improving the gut microbiota composition, promoting the wound healing process and shaping the immunological responses. Chronic intestinal conditions, such as inflammatory bowel diseases (IBD), are characterized by an imbalance in microbiota composition, with decreased diversity, and by relapsing and persisting inflammation, which may lead to mucosal damage. Although the results of the clinical studies investigating the effect of probiotics on patients with IBD are still controversial, it is without doubt that these microorganisms and their metabolites, now named postbiotics, have a positive influence on both the host's microbiota and the immune system, and ultimately alter the topical tissue microenvironment. This influence is achieved through three axes: (1) By displacement of potential pathogens via competitive exclusion; (2) by offering protection to the host through the secretion of various defensive mediators; and (3) by supplying the host with essential nutrients. We will analyze and discuss almost all the in vitro and in vivo studies of the past 2 years dealing with the possible favorable effects of certain probiotic genus on gut immunological responses, highlighting which species are the most beneficial against intestinal inflammation.
Collapse
Affiliation(s)
- Eirini Filidou
- Faculty of Medicine, Laboratory of Pharmacology, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Leonidas Kandilogiannakis
- Faculty of Medicine, Laboratory of Pharmacology, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Anne Shrewsbury
- Department of Surgery, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| | - George Kolios
- Faculty of Medicine, Laboratory of Pharmacology, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Katerina Kotzampassi
- Department of Surgery, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| |
Collapse
|
9
|
Li H, Li H, Stanton C, Ross RP, Zhao J, Chen W, Yang B. Exopolysaccharides Produced by Bifidobacterium longum subsp. longum YS108R Ameliorates DSS-Induced Ulcerative Colitis in Mice by Improving the Gut Barrier and Regulating the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7055-7073. [PMID: 38520351 DOI: 10.1021/acs.jafc.3c06421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
Ulcerative colitis (UC) is a major disease that has endangered human health. Our previous study demonstrated that Bifidobacterium longum subsp. longum YS108R, a ropy exopolysaccharide (EPS)-producing bacterium, could alleviate UC in mice, but it is unclear whether EPS is the key substance responsible for its action. In this study, we proposed to investigate the remitting effect of EPS from B. longum subsp. longum YS108R on UC in a DSS-induced UC mouse model. Water extraction and alcohol precipitation were applied to extract EPS from the supernatant of B. longum subsp. longum YS108R culture. Then the animal trial was performed, and the results indicated that YS108R EPS ameliorated colonic pathological damage and the intestinal barrier. YS108R EPS suppressed inflammation via NF-κB signaling pathway inhibition and attenuated oxidative stress via the Nrf2 signaling pathway activation. Remarkably, YS108R EPS regulated gut microbiota, as evidenced by an increase in short-chain fatty acid (SCFA)-producing bacteria and a decline in Gram-negative bacteria, resulting in an increase of propionate and butyrate and a reduction of lipopolysaccharide (LPS). Collectively, YS108R EPS manipulated the intestinal microbiota and its metabolites, which further improved the intestinal barrier and inhibited inflammation and oxidative stress, thereby alleviating UC.
Collapse
Affiliation(s)
- Huizhen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
| | - R Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
10
|
Fuochi V, Furneri PM. Applications of Probiotics and Their Potential Health Benefits. Int J Mol Sci 2023; 24:15915. [PMID: 37958897 PMCID: PMC10650470 DOI: 10.3390/ijms242115915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Probiotics have garnered significant attention in recent years due to their potential health benefits and their role in promoting a balanced gut microbiome [...].
Collapse
Affiliation(s)
- Virginia Fuochi
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95124 Catania, Italy;
| | | |
Collapse
|
11
|
Du Z, Li J, Li W, Fu H, Ding J, Ren G, Zhou L, Pi X, Ye X. Effects of prebiotics on the gut microbiota in vitro associated with functional diarrhea in children. Front Microbiol 2023; 14:1233840. [PMID: 37720150 PMCID: PMC10502507 DOI: 10.3389/fmicb.2023.1233840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Purpose Diarrhea is among the top five causes of morbidity and mortality in children. Dysbiosis of the gut microbiota is considered the most important risk factor for diarrhea. Prebiotics have shown efficacy in treating diarrhea by regulating the balance of the gut microbiota in vivo. Methods In this study, we used an in vitro fermentation system to prevent the interference of host-gut microbe interactions during in vivo examination and investigated the effect of fructo-oligosaccharides (FOS) on gut microbiota composition and metabolism in 39 pediatric patients with functional diarrhea. Results 16S rRNA sequencing revealed that FOS significantly improved α- and β-diversity in volunteers with pediatric diarrhea (p < 0.05). This improvement manifested as a significant increase (LDA > 2, p < 0.05) in probiotic bacteria (e.g., Bifidobacterium) and a significant inhibition (LDA > 2, p < 0.05) of harmful bacteria (e.g., Escherichia-Shigella). Notably, the analysis of bacterial metabolites after FOS treatment showed that the decrease in isobutyric acid, isovaleric acid, NH3, and H2S levels was positively correlated with the relative abundance of Lachnoclostridium. This decrease also showed the greatest negative correlation with the abundance of Streptococcus. Random forest analysis and ROC curve validation demonstrated that gut microbiota composition and metabolites were distinct between the FOS treatment and control groups (area under the curve [AUC] > 0.8). Functional prediction using PICRUSt 2 revealed that the FOS-induced alteration of gut microbiota was most likely mediated by effects on starch and sucrose metabolism. Conclusion This study is the first to evince that FOS can modulate gut microbial disorders in children with functional diarrhea. Our findings provide a framework for the application of FOS to alleviate functional diarrhea in children and reduce the use of antibiotics for managing functional diarrhea-induced disturbances in the gut microbiota.
Collapse
Affiliation(s)
- Zhi Du
- Department of Pharmacy, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiabin Li
- Department of Pharmacy, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Li
- Department of Clinical Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Hao Fu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jieying Ding
- Department of Pharmacy, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guofei Ren
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang, China
| | - Linying Zhou
- People's Hospital of Longquan City, Longquan, China
| | - Xionge Pi
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Xiaoli Ye
- Department of Medical Administration, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Lin Q, Hao WJ, Zhou RM, Huang CL, Wang XY, Liu YS, Li XZ. Pretreatment with Bifidobacterium longum BAA2573 ameliorates dextran sulfate sodium (DSS)-induced colitis by modulating gut microbiota. Front Microbiol 2023; 14:1211259. [PMID: 37346749 PMCID: PMC10280014 DOI: 10.3389/fmicb.2023.1211259] [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: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Objectives Inflammatory bowel disease (IBD) is a chronic lifelong inflammatory disease. Probiotics such as Bifidobacterium longum are considered to be beneficial to the recovery of intestinal inflammation by interaction with gut microbiota. Our goals were to define the effect of the exclusive use of BAA2573 on dextran sulfate sodium (DSS)-induced colitis, including improvement of symptoms, alleviation of histopathological damage, and modulation of gut microbiota. Methods In the present study, we pretreated C57BL/6J mice with Bifidobacterium longum BAA2573, one of the main components in an over-the-counter (OTC) probiotic mixture BIFOTO capsule, before modeling with DSS. 16S rDNA sequencing and liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based non-targeted metabolomic profiling were performed with the collected feces. Results We found that pretreatment of Bifidobacterium longum BAA2573 given by gavage significantly improved symptoms and histopathological damage in DSS-induced colitis mice. After the BAA2573 intervention, 57 genera and 39 metabolites were significantly altered. Pathway enrichment analysis demonstrated that starch and sucrose metabolism, vitamin B6 metabolism, and sphingolipid metabolism may contribute to ameliorating colitis. Moreover, we revealed that the gut microbiome and metabolites were interrelated in the BAA2573 intervention group, while Alistipes was the core genus. Conclusion Our study demonstrates the impact of BAA2573 on the gut microbiota and reveals a possible novel adjuvant therapy for IBD patients.
Collapse
Affiliation(s)
- Qiong Lin
- Nephrology and Immunology Department, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Digestive, Affiliated Children's Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Wu-Juan Hao
- Department of Digestive, Affiliated Children's Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Ren-Min Zhou
- Department of Digestive, Affiliated Children's Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | | | - Xu-Yang Wang
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan-Shan Liu
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xiao-Zhong Li
- Nephrology and Immunology Department, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|