1
|
Lee AH, Rodriguez Jimenez DM, Meisel M. Limosilactobacillus reuteri - a probiotic gut commensal with contextual impact on immunity. Gut Microbes 2025; 17:2451088. [PMID: 39825615 DOI: 10.1080/19490976.2025.2451088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
The gut microbiome plays a key role in human health, influencing various biological processes and disease outcomes. The historical roots of probiotics are traced back to Nobel Laureate Élie Metchnikoff, who linked the longevity of Bulgarian villagers to their consumption of sour milk fermented by Lactobacilli. His pioneering work led to the global recognition of probiotics as beneficial supplements, now a multibillion-dollar industry. Modern probiotics have been extensively studied for their immunomodulatory effects. Limosilactobacillus reuteri (L. reuteri), a widely used probiotic, has garnered significant attention for its systemic immune-regulatory properties, particularly in relation to autoimmunity and cancer. This review delves into the role of L. reuteri in modulating immune responses, with a focus on its impact on systemic diseases.
Collapse
Affiliation(s)
- Amanda H Lee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Gao ZK, Fan CY, Zhang BW, Geng JX, Han X, Xu DQ, Arshad M, Sun HX, Li JY, Jin X, Mu XQ. Cardiac function of colorectal cancer mice is remotely controlled by gut microbiota: regulating serum metabolites and myocardial cytokines. Anim Microbiome 2025; 7:53. [PMID: 40448218 PMCID: PMC12123981 DOI: 10.1186/s42523-025-00405-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/06/2025] [Indexed: 06/02/2025] Open
Abstract
Several studies have indicated that the dysregulation of microbial metabolites and the inflammatory environment resulting from microbial dysbiosis may contribute to the occurrence and progression of cardiovascular diseases. Therefore, restoring the disordered gut microbiota in patients with colorectal cancer by fecal microbiota transplantation (FMT) has the potential to reduce the incidence of cardiac disease. In this study, we identified cardiac dysfunction in azomethane and dextran sodium sulfate-induced colorectal cancer mice. Intestinal microbes from healthy mice were transferred to colorectal cancer mice, which vastly reversed the disorder of the gut microbiota and effectively alleviated cardiac dysfunction. Moreover, FMT regulated the expression of serum metabolites such as uridine triphosphate (UTP), tiamulin, andrographolide, and N-Acetyl-D-glucosamine, as well as cytokines like TGF-β, IRF5, and β-MHC in the heart. These findings uncover that the disturbed gut microbiota causes cardiac dysfunction in colorectal cancer mice by modulating the expression of serum metabolites and cytokines, which could be alleviated by treatment with FMT.
Collapse
Affiliation(s)
- Zhan-Kui Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Chao-Yuan Fan
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Bo-Wen Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Jia-Xin Geng
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Xing Han
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Dan-Qi Xu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Muhammad Arshad
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Hao-Xuan Sun
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jiong-Yi Li
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiangyuan Jin
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Xiao-Qin Mu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China.
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
3
|
Sun W, Ma S, Meng D, Wang C, Zhang J. Advances in research on the intestinal microbiota in the mechanism and prevention of colorectal cancer (Review). Mol Med Rep 2025; 31:133. [PMID: 40116116 PMCID: PMC11948985 DOI: 10.3892/mmr.2025.13498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/19/2025] [Indexed: 03/23/2025] Open
Abstract
The intestinal microbiota represents a diverse population that serves a key role in colorectal cancer (CRC) and its treatment outcomes. Advancements in sequencing have revealed notable shifts in microbial composition and diversity among individuals with CRC. Concurrently, animal models have elucidated the involvement of specific microbes such as Lactobacillus fragilis, Escherichia coli and Fusobacterium nucleatum in the progression of CRC. The present review aimed to highlight contributions of intestinal microbiota to the pathogenesis of CRC, the effects of traditional treatments on intestinal microbiota and the potential for microbiota modulation as a therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Weitong Sun
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154000, P.R. China
| | - Shize Ma
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154000, P.R. China
| | - Dongdong Meng
- Department of Medical Services, Xuzhou Morning Star Women's and Children's Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Chaoxing Wang
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154000, P.R. China
| | - Jinbo Zhang
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154000, P.R. China
| |
Collapse
|
4
|
Sun M, Ma J, Zhang G, Song M, Lv R, Liang J, Shi Y, Zhao L. Brain Targeting Bacterial Extracellular Vesicles Enhance Ischemic Stroke Therapy via Efficient ROS Elimination and Suppression of Immune Infiltration. ACS NANO 2025; 19:15491-15508. [PMID: 40249658 DOI: 10.1021/acsnano.4c16161] [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: 04/20/2025]
Abstract
Ischemic stroke (IS) as a detrimental neurological disease is accompanied by oxidative-stress-induced injury, concurrent inflammatory response, overactivated brain immune microenvironment, and disruption of the blood-brain barrier (BBB). This cascade of events ultimately leads to neuronal death and significantly impairs the recovery of neurological function. In this study, we presented extracellular vesicles derived from the gut probiotic Lactobacillus reuteri (LrEVs) integrated with brain targeting, reactive oxygen species (ROS) scavenging, and reduced infiltration of immune cells for effective multiple therapeutic interventions of IS. LrEVs inherited peptidoglycan (PGN) specifically targeted upregulated toll-like receptor 2 (TLR2) in the injured region of the ischemic brain, achieving the effective penetration of the BBB and accumulation in the ischemic brain. In the meantime, LrEVs prevented neuronal apoptosis after stroke by scavenging ROS overproduction and modulating microglial polarization through inhibition of the MAPK and NF-κB pathways. Furthermore, LrEVs inhibited the aggregation of C-C motif chemokine ligand 2 (CCL2), reduced the infiltration of peripheral immune cells such as macrophages and neutrophils into ischemic brain tissue, and suppressed the impairment of BBB, thereby improving the overactivated brain immune microenvironment. The findings provide a vesicle that combines ROS scavenging and modulation of the immune microenvironment, showcasing the potential of gut-probiotic-derived vesicles to treat neurological damage.
Collapse
Affiliation(s)
- Mengdi Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jinghan Ma
- Institution of Life Science, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ge Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Mingzhu Song
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ruizhen Lv
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jia Liang
- Institution of Life Science, Jinzhou Medical University, Jinzhou 121000, PR China
- Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, PR China
- Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
- Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, PR China
- Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121000, PR China
| |
Collapse
|
5
|
Lu H, Zhang G, Wang K, Liu K, Gao Y, Chen J, Li Y, Yan J. The Role of Lactiplantibacillus plantarum CGMCC9513 in Alleviating Colitis by Synergistic Enhancement of the Intestinal Barrier Through Modulating Gut Microbiota and Activating the Aryl Hydrocarbon Receptor. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10551-0. [PMID: 40301232 DOI: 10.1007/s12602-025-10551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/01/2025]
Abstract
Ulcerative colitis (UC) has become a global health issue. This study evaluated whether administering Lactiplantibacillus plantarum CGMCC9513 to dextran sulfate sodium (DSS)-induced colitis mice could alleviate colitis by modulating gut microbiota imbalance and activating the aryl hydrocarbon receptor (AhR) to enhance intestinal barrier function. The anti-inflammatory effect and AhR activation ability of L. plantarum CGMCC9513 were evaluated with lipopolysaccharide (LPS)-induced cell inflammation model; 25 male BALB/c mice were divided into blank group (CNG), model group (DSS), L. plantarum CGMCC9513-treated group (LPDT), and L. plantarum CGMCC9513 control group (LP). The mice were pre-administered L. plantarum CGMCC9513 for 14 days and continued to receive it during DSS induction. Symptoms during induction, goblet cell count, expression of MUC2 and Occludin proteins, and changes in gut microbiota were observed. Subsequently, the expression of cytokines interleukin-10 (IL-10), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β) and AhR activation status was determined. The study found that L. plantarum CGMCC9513 could alleviate cell inflammation induced by LPS and activate AhR in vitro. For colitis mice, it could reduce colonic mucosal damage and enhance intestinal barrier function. Regarding gut microbiota changes, L. plantarum CGMCC9513 mainly downregulated Bacteroides, Blautia, Escherichia-Shigella, and Lachnospiraceae_ NK4A136_group and upregulated Firmicutes, Lactobacillus. It reduces the risk of bacterial translocation and increases beneficial gut bacteria. L. plantarum CGMCC9513 reduced the expression of pro-inflammatory cytokines TNF-α and IL-1β while increasing the expression of anti-inflammatory cytokine IL-10. Meanwhile, increased expression of AhR and Cytochrome P450 1A1 (CYP1A1) proteins indicated AhR activation by L. plantarum CGMCC9513. In conclusion, L. plantarum CGMCC9513 can synergistically enhance intestinal barrier alleviation in colitis mice by modulating gut microbiota imbalance and activating AhR.
Collapse
Affiliation(s)
- Hongyu Lu
- Medical School of Guangxi University, Nanning, 530004, China
| | - Guoqing Zhang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kaidi Wang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kefei Liu
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yingrui Gao
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Jinyan Chen
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yixiang Li
- Medical School of Guangxi University, Nanning, 530004, China.
| | - Jianhua Yan
- Medical School of Guangxi University, Nanning, 530004, China.
| |
Collapse
|
6
|
Jiang D, Zhang J, Ji Y, Dai Z, Yang Y, Wu Z. Glutamate Supplementation Regulates Nitrogen Metabolism in the Colon and Liver of Weaned Rats Fed a Low-Protein Diet. Nutrients 2025; 17:1465. [PMID: 40362775 PMCID: PMC12073364 DOI: 10.3390/nu17091465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Glutamate, a nutritionally non-essential amino acid, is a key intermediate in nitrogen metabolism. Despite more studies on its functional role in intestine health, it remains unknown how glutamate regulates nitrogen metabolism in animals fed a low-protein diet. Methods: Herein, we investigated the effects of glutamate supplementation on colonic amino acid transport, barrier protein expression, microbiota alterations, fecal nitrogen emissions, hepatic amino acid transport, and protein synthesis in weaned rats. Results: We found that protein restriction diminished the mucus thickness, reduced goblet cell numbers, and the expression of EAAT3, y+LAT2 in the colon. In contrast, glutamate supplementation reversed these effects, increasing the colon length and enhancing the expression of ZO-1, Occludin, and Claudin-1 in the colon. At the genus level, glutamate increased the abundance of Lactococcus and Clostridia_sensu_stricto_18. Additionally, glutamate supplementation resulted in an increased apparent nitrogen digestibility, reduced the ratio of fecal nitrogen to total nitrogen intake, and increased the ratio of fecal microbial nitrogen to total nitrogen intake. Protein restriction decreased the mRNA level of ATP1A1, EAAT3, SNAT9/2, and ASCT2, and the protein level of p-mTOR, mTOR, p-mTOR/mTOR, and p-p70S6K/p70S6K as well as p-4EBP1/4EBP1 in the liver. These effects were reversed by glutamate supplementation. Conclusions: In conclusion, glutamate supplementation upregulates amino acid transporters and barrier protein expression in the colon, modulates microbiota composition to reduce fecal nitrogen excretion, and enhances amino acid transport and protein synthesis in the liver by activating the mTOR/p70S6K/4EBP1 pathway, which influences nitrogen metabolism in weaned rats fed a low-protein diet.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (D.J.); (J.Z.); (Y.J.); (Z.D.); (Y.Y.)
| |
Collapse
|
7
|
Reis de Souza TC, Landín GM, Celis UM, Valeriano TH, Gómez-Soto JG, Briones CN. Supplementation with Potato Protein Concentrate and Saccharomyces boulardii to an Antibiotic-Free Diet Improves Intestinal Health in Weaned Piglets. Animals (Basel) 2025; 15:985. [PMID: 40218378 PMCID: PMC11988057 DOI: 10.3390/ani15070985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
Many countries have banned the use of antibiotics in livestock feed due to the development of antibiotic-resistant bacteria. Therefore, it is necessary to identify suitable alternatives to antibiotics in order to maintain intestinal health and improve piglet performance after weaning. The objective of this study was to evaluate how the intake of two functional foods-potato protein concentrate (PP) and Saccharomyces boulardii (Sb), either isolated or combined-would affect productive performance and some digestive morphophysiological responses after two weeks post-weaning. We evaluated five diets: SB, with Sb; PPC, with PP; PPC-SB, with a combination of PP and Sb; C-, without antibiotics, Sb, or PP; and C+, with antibiotics. Neither the absence of antibiotics in the diet nor the inclusion of PP and Sb affected performance or digestive organ development during the post-weaning phase. Piglets fed the C+ diet and the PP-Sb combination had higher concentrations of occludin proteins and longer villi in the jejunum compared to those fed C-, PPC, and SB. The use of Sb alone or in combination with PP increased fecal Lactobacillus, while antibiotics reduced coliform bacteria. The PP-Sb combination prevented the atrophy of the jejunal villi and promoted adhesion between enterocytes in the jejunum. Including these functional foods in piglet diets contributed to maintaining intestinal health during the critical post-weaning phase. However, further research is needed to validate and enhance the conclusions of this study.
Collapse
Affiliation(s)
- Tércia Cesária Reis de Souza
- Faculty of Natural Sciences, Autonomous University of Querétaro, Querétaro 76230, Mexico; (U.M.C.); (T.H.V.); (J.G.G.-S.); (C.N.B.)
| | - Gerardo Mariscal Landín
- National Institute of Agricultural and Livestock Forestry Research, National Center of Research in Animal Physiology, Ajuchitlán Colón, Querétaro 76280, Mexico;
| | - Ulisses Moreno Celis
- Faculty of Natural Sciences, Autonomous University of Querétaro, Querétaro 76230, Mexico; (U.M.C.); (T.H.V.); (J.G.G.-S.); (C.N.B.)
| | - Teresita Hijuitl Valeriano
- Faculty of Natural Sciences, Autonomous University of Querétaro, Querétaro 76230, Mexico; (U.M.C.); (T.H.V.); (J.G.G.-S.); (C.N.B.)
| | - José Guadalupe Gómez-Soto
- Faculty of Natural Sciences, Autonomous University of Querétaro, Querétaro 76230, Mexico; (U.M.C.); (T.H.V.); (J.G.G.-S.); (C.N.B.)
| | - Christian Narváez Briones
- Faculty of Natural Sciences, Autonomous University of Querétaro, Querétaro 76230, Mexico; (U.M.C.); (T.H.V.); (J.G.G.-S.); (C.N.B.)
| |
Collapse
|
8
|
Mercer SD, Doherty C, Singh G, Willmott T, Cheesapcharoen T, Teanpaisan R, O'Neill C, Ledder RG, McBain AJ. Lactobacillus lysates protect oral epithelial cells from pathogen-associated damage, increase secretion of pro-inflammatory cytokines and enhance barrier integrity. Sci Rep 2025; 15:5894. [PMID: 39966408 PMCID: PMC11836205 DOI: 10.1038/s41598-025-86914-y] [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: 07/26/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Periodontitis is a chronic gum disease characterised by inflammation and the loss of bone. We have explored the potential prophylactic effects of lysates from four Lactobacillus strains against the toxic effects of three periodontal pathogens (Porphyromonas gingivalis, Fusobacterium nucleatum, and Aggregatibacter actinomycetemcomitans). TR146 oral epithelial cells were pre-treated with Lactobacillus lysates (L. rhamnosus - GG, L. rhamnosus - SD11, L. reuteri and L. plantarum) and then challenged with pathogenic material (live cells, lysates, or supernatants). Cytokine analysis was performed on supernatants of cells treated with probiotic lysates from 1.5 h to 24 h. Effects of probiotic lysates on re-epithelialisation were determined using keratinocyte scratch assays, monitoring both migration and proliferation. Epithelial barrier function was observed after lysate addition by trans-epithelial electrical resistance (TEER) and by quantifying claudin-1 expression. Treatment of host cells with Lactobacillus lysates before pathogen exposure conferred significant protection against viability loss. Although extended pre-treatment did not generally increase protection, against live Aggregatibacter actinomycetemcomitans, significant increases in viability were seen after 24 h of pre-treatment for GG, SD11 and L. plantarum. Pro-inflammatory cytokines TNF-α, IP-10, IL-6, and IL-8 increased significantly with extended probiotic treatment, while IL-1β and IL-1α secretion significantly increased but remained constant over time. Secretion of the growth-promoting cytokine TGF-β increased after 3 h of treatment, however no increases in the regulatory cytokine IL-10 were recorded. Only exposure to SD11 significantly enhanced re-epithelialisation, TEER and claudin-1 expression while GG increased TEER but decreased claudin-1 expression. L. plantarum significantly inhibited re-epithelialisation but did not impact TEER or claudin-1 expression. All lysates significantly improved TEER in the presence of pathogenic material, demonstrating a protective effect on barrier function.
Collapse
Affiliation(s)
- Steven D Mercer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Division of Musculoskeletal and Dermatological Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Christopher Doherty
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Gurdeep Singh
- Division of Musculoskeletal and Dermatological Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Thomas Willmott
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Institute of Infection, Veterinary and Ecological Sciences, Clinical Infection, Microbiology & Immunology, University of Liverpool, Liverpool, UK
| | - Tanaporn Cheesapcharoen
- Department of Conservative Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat- Yai, Thailand
| | - Rawee Teanpaisan
- Medical Science Research and Innovation Institute, Prince of Songkla University, Hat-Yai, Thailand
| | - Catherine O'Neill
- Division of Musculoskeletal and Dermatological Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Ruth G Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
9
|
Xie Q, Yang M, Duanmu Q, Kang M, Wang J, Tan BE. Ningxiang pig-derived Lactobacillus reuteri improves the gut health of weaned piglets by regulating intestinal barrier function and cytokine profiles. Sci Rep 2025; 15:3993. [PMID: 39893246 PMCID: PMC11787358 DOI: 10.1038/s41598-025-87105-5] [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: 11/23/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
Weaning stress in piglets can induce intestinal damage, resulting in impaired growth performance. Probiotics have emerged as significant contributors to enhancing gut health in piglets. This study aimed to evaluate the effects of Lactobacillus reuteri (L. reuteri) supplementation on growth performance, intestinal immunity, and intestinal barrier integrity in weaned piglets. In this investigation, fourteen healthy weaned piglets of similar age and weight, were randomly assigned to two groups (n = 7), receiving either normal saline (Control group) or L. reuteri (L-treatment group) over a 16-day period. The findings revealed no significant impact of L. reuteri on growth performance compared to controls. However, it lowered serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels (P < 0.05) and elevated serum concentrations of Immunoglobulin A (IgA), immunoglobulin G (IgG), and secretory immunoglobulin A (sIgA) (P < 0.05). Additionally, L. reuteri notably enhanced the villus height-to-crypt depth ratio in the ileum (P < 0.05) and increased mRNA expression of zonula occludens-1 (ZO-1), Claudin-1, and ZO-1 in ileal tissue (P < 0.05). Furthermore, L. reuteri reduced mRNA expression of pro-inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) (P < 0.05) in the jejunum and colon while enhancing the expression of the anti-inflammatory cytokine interleukin-10 (IL-10) (P < 0.05) in both the ileum and colon. This study demonstrates that L. reuteri isolated from Ningxiang pigs can improve the intestinal health of weaned piglets by modulating gut barrier function and cytokine levels.
Collapse
Affiliation(s)
- Qian Xie
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Mei Yang
- Huizhou Engineering Vocational College, Huizhou, 516023, Guangdong, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Qing Duanmu
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Meng Kang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Jing Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Bi E Tan
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China.
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China.
| |
Collapse
|
10
|
Chen S, Niu X, Zhang Y, Wen J, Bao M, Li Y, Gao Y, Wang X, Liu X, Yong Y, Yu Z, Ma X, Eun JB, Shim JH, Abd El-Aty AM, Ju X. Butyrolactone-I from marine fungi alleviates intestinal barrier damage caused by DSS through regulating lactobacillus johnsonii and its metabolites in the intestine of mice. J Nutr Biochem 2025; 135:109786. [PMID: 39447992 DOI: 10.1016/j.jnutbio.2024.109786] [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: 02/23/2024] [Revised: 08/01/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Butyrolactone-I (BTL-1), a secondary metabolite from the marine fungus Aspergillus terreus, exhibits numerous biological activities. Previous research has indicated that Butyrolactone-I alleviates intestinal epithelial inflammation via the TLR4/NF-κB and MAPK pathways. However, the mechanisms underlying its protection against intestinal barrier damage remain unclear. This study aims to further elucidate these mechanisms. We observed that BTL-1 administration increased the abundance of Lactobacillus johnsonii (LJ) in both in vivo and in vitro experiments, prompting an investigation into the effects of LJ and its metabolites on DSS-induced inflammatory bowel disease (IBD). The results demonstrated that BTL-1 significantly upregulated tight junction (TJ) and adherens junction (AJ) proteins, maintained intestinal barrier integrity, and alleviated DSS-induced IBD in mice. These effects were associated with the proliferation of LJ and its metabolites, such as butyric and propionic acids, and the inhibition of the MAPK signaling pathway in the colon. Interestingly, administering LJ alone produced a protective effect against DSS-induced IBD similar to that observed with BTL-1. Furthermore, butyric acid, a metabolite of LJ, also upregulated TJ/AJ proteins in intestinal epithelial cells through the MAPK signaling pathway. Our findings suggest that BTL-1 regulates intestinal flora, promotes LJ proliferation, protects intestinal barrier integrity, increases the concentrations of butyric and propionic acids, and ultimately inhibits the activation of the MAPK signaling pathway in mice to alleviate IBD. Therefore, BTL-1 could potentially be used as a natural drug to prevent IBD and maintain intestinal flora balance. We explored how butyrolactone-I exerts a preventive effect on IBD through intestinal bacteria (Lactobacillus johnsonii).
Collapse
Affiliation(s)
- Shengwei Chen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Xueting Niu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Yi Zhang
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Jiaying Wen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Minglong Bao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Yin Li
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Yuan Gao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Xinchen Wang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China
| | - Xiaoxi Liu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Yanhong Yong
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Zhichao Yu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Xingbing Ma
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Jong-Bang Eun
- Department of Food Science and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Jae-Han Shim
- Natural Products Chemistry Laboratory, Biotechnology Research Institute, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Xianghong Ju
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China; Shenzheng Institute of Guangdong Ocean University, Organization X, Shenzheng, China.
| |
Collapse
|
11
|
Ma W, Wu Y, Lin X, Yang L, Huang L. Amelioration of inflammatory bowel disease by Bifidobacterium animalis subsp. lactis XLTG11 in combination with mesalazine. Front Microbiol 2024; 15:1472776. [PMID: 39697653 PMCID: PMC11652597 DOI: 10.3389/fmicb.2024.1472776] [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: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
The treatment of inflammatory bowel disease (IBD) remains challenging and significantly impacts both patients and their families. This study evaluated the role of Bifidobacterium animalis subsp. lacti XLTG11 (XLTG11) in combination with mesalazine (5-ASA) in the improvement of IBD. The results demonstrated that the XLTG11+5-ASA group exhibited superior recovery compared to both the XLTG11-only group and the 5-ASA-only group. The XLTG11+5-ASA group significantly reduced myeloperoxidase activity (MPO), attenuated colonic tissue damage, lowered the levels of lipopolysaccharides (LPS) and D-lactic acid (D-LA), and decreased intestinal permeability. Furthermore, it upregulated the mRNA expression of Claudin-1, Occludin, ZO-1, and MUC2, which contributed to the protective effect on intestinal barrier function. Additionally, the XLTG11+5-ASA group significantly increased the levels of anti-inflammatory cytokines while decreasing pro-inflammatory cytokine levels. Notably, treatment with the XLTG11+5-ASA group significantly increased levels of acetic, propionic, and butyric acids, as well as the relative abundance of beneficial bacteria such as Bifidobacterium and Lactobacillus, while decreasing the relative abundance of Enterococcus, Enterobacteriaceae, and Clostridium perfringens. The results indicate that the combination of XLTG11 and 5-ASA was more effective in treating IBD than either treatment alone, significantly improving IBD-related symptoms and providing a scientific basis for future clinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Lili Huang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
12
|
Butrungrod W, Chaiyasut C, Makhamrueang N, Peerajan S, Chaiyana W, Sirilun S. Postbiotic Metabolite Derived from Lactiplantibacillus plantarum PD18 Maintains the Integrity of Cell Barriers and Affects Biomarkers Associated with Periodontal Disease. Antibiotics (Basel) 2024; 13:1054. [PMID: 39596748 PMCID: PMC11591352 DOI: 10.3390/antibiotics13111054] [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: 10/08/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Periodontal disease is caused by oral infections, biofilms, persistent inflammation, and degeneration of cell barrier integrity, allowing pathogens to invade host cells. Probiotics have been extensively studied for the treatment of periodontal disease. However, research on the involvement of beneficial substances produced by probiotics, called "postbiotics," in periodontal diseases remains in its early stages. The present study aimed to evaluate the effect of a postbiotic metabolite (PM) from Lactiplantibacillus plantarum PD18 on immunomodulation and maintenance of cell barrier integrity related to periodontal disease. Method: The main substance in PM PD18 was analyzed by GC-MS. The cytotoxic effect of PM PD18 was performed using the MTT assay, wound healing through the scratch assay, cell permeability through TEER value, modulation of inflammatory cytokines through ELISA, and gene expression of inflammatory cytokines and tight junction protein was determined using qRT-PCR. Results: The main substance found in PM PD18 is 2,3,5,6-tetramethylpyrazine. PM PD18 did not exhibit cytotoxic effects on RAW 264.7 cells but promoted wound healing and had an antiadhesion effect on Porphyromonas gingivalis concerning SF-TY cells. This postbiotic could maintain cell barrier integrity by balancing transepithelial electrical resistance (TEER) and alkaline phosphatase (ALP) activity. In addition, the gene and protein expression levels of zonula occludens-1 (ZO-1) increased. PM PD18 was found to have immunomodulatory properties, as demonstrated by regulated anti- and pro-inflammatory cytokines. Interleukin-10 (IL-10) increased, while IL-6 and IL-8 were reduced. Conclusions: This study demonstrated that PM PD18 is efficient as a natural treatment for maintaining cell barrier integrity and balancing inflammatory responses associated with periodontal disease.
Collapse
Affiliation(s)
- Widawal Butrungrod
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (W.B.); (C.C.); (N.M.); (W.C.)
| | - Chaiyavat Chaiyasut
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (W.B.); (C.C.); (N.M.); (W.C.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Netnapa Makhamrueang
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (W.B.); (C.C.); (N.M.); (W.C.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Wantida Chaiyana
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (W.B.); (C.C.); (N.M.); (W.C.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasithorn Sirilun
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (W.B.); (C.C.); (N.M.); (W.C.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
13
|
Sheng T, Wang L, Yan S, Wei Q, Geng X, Lan W, Chen Y, Liu Y, Li N. Involvement of gut microbiota recovery and autophagy induction in Youhua Kuijie formula's protection against experimental ulcerative colitis. Exp Anim 2024; 73:357-369. [PMID: 38599877 PMCID: PMC11534492 DOI: 10.1538/expanim.23-0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/01/2024] [Indexed: 04/12/2024] Open
Abstract
Ulcerative colitis (UC) is characterized by overactive inflammatory response, impaired intestinal mucosal barrier and disrupted gut microbiota. Youhua Kuijie formula is a classic empirical prescription based on the pathogenesis of UC. The present study was designed to verify the protective effect of Youhua Kuijie formula on DSS-induced UC in mice and uncover the related mechanism. Youhua Kuijie formula were orally administrated to UC mice induced by DSS dissolved in drinking water for ten days. The protective effect of Youhua Kuijie formula was evidenced by reduced pathological symptoms accompanied by palliative inflammatory response and relatively intact intestinal barrier. The data from 16S rRNA gene sequencing and GC-MS untargeted metabolomics indicated that the supplement of Youhua Kuijie formula restructured gut microbiota community structure, and thereby modulated the metabolic profiles in UC mice. The analysis of pathway enrichment analysis suggested the major alterations in metabolic pathway were related to protein digestion and absorption. Besides, the results of the following experiments suggested that Youhua Kuijie formula treatment increased adenosine monophosphate-activated protein kinase (AMPK) activation, decreased mechanistic target of rapamycin (mTOR) phosphorylation, and thereby reversing autophagy deficiency in the intestinal tract of UC mice. Collectively, our results demonstrated that the regulation of AMPK/mTOR was involved in Youhua Kuijie formula administration mediated protective effect on UC.
Collapse
Affiliation(s)
- Tianjiao Sheng
- Graduate school, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshandong Road, Shenyang, Liaoning, 110847, P.R. China
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, No.83 Wenhua Road, Shenyang, Liaoning, 110016, P.R. China
| | - Lei Wang
- Department of anorectum, Hulunbuir Zhong Meng Hospital, No. 58 Xidajie Road, Hulunbuir, 021000, P.R. China
| | - Simeng Yan
- Department of 1st Area of Officers' Ward, General Hospital of Northern Theater Command, No.83 Wenhua Road, Shenyang, Liaoning, 110016, P.R. China
| | - Qiuyu Wei
- Graduate school, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshandong Road, Shenyang, Liaoning, 110847, P.R. China
| | - Xiao Geng
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, No.83 Wenhua Road, Shenyang, Liaoning, 110016, P.R. China
| | - Weiru Lan
- The third department of Anorectal hemorrhoids and Fistula, Liaoning University of Traditional Chinese Medicine Affiliated Third Hospital, No. 35, 11th Wei Road, Shenyang, Liaoning, 110003, P.R. China
| | - Yan Chen
- Graduate school, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshandong Road, Shenyang, Liaoning, 110847, P.R. China
| | - Yuedong Liu
- Graduate school, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshandong Road, Shenyang, Liaoning, 110847, P.R. China
| | - Na Li
- Department of Anorectal Surgery, Xianyang Central Hospital, No. 78 Renmin East Road, Xianyang, Shaanxi, 712000, P.R. China
| |
Collapse
|
14
|
García-Vicente EJ, Rey-Casero I, Martín M, Pérez A, Benito-Murcia M, Risco D. Oral supplementation with postbiotics modulates the immune response produced by myxomatosis vaccination in wild rabbits. Vaccine 2024; 42:125978. [PMID: 38760270 DOI: 10.1016/j.vaccine.2024.05.026] [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: 07/12/2023] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Rabbits (Oryctolagus cuniculus) are vitally important species in the Iberian Peninsula ecosystem. However, since 1950, there has been a significant population decline, with major repercussions. This situation is mainly due to the presence of infectious diseases, such as myxomatosis, which is expanding and is characterized by severe and fatal clinical manifestations. Current control measures, mainly those based on vaccinations, are ineffective. Therefore, new strategies need to be developed and implemented. This study aimed to evaluate whether supplementation with postbiotic products modulates the immune response in wild rabbits vaccinated against myxomatosis. For this purpose, two groups of rabbits were established: a control group fed with standard feed ad libitum from weaning (28 days) until two months of age, and a treated group, which was fed under the same conditions but supplemented with postbiotics (3 kg/Tm). All the studied rabbits were vaccinated against this disease during weaning. In addition, a blood samples were obtained from all animals immediately before vaccination and 30 days later, which allowed us to evaluate the level of antibodies against myxomatosis virus (ELISA detection) and the relative expression of gene encoding to cytokines related to the immune response (IL6, TNFα and IFNγ), at both times of the experience. Weight and length measurements were also taken at both times to calculate body index and mean daily gain (MDG). No statistically significant differences in growth parameters were observed. There were also no differences in the serological response among groups. However, a relative underexpression of gene codifying to TNFα (p-value = 0.03683) and a higher expression on IFNγ (p-value = 0.045) were observed in the treated group. This modulation in cytokines could lead to less severe lesions in wild rabbit naturally infected with myxomatosis virus.
Collapse
Affiliation(s)
- E J García-Vicente
- Department of Animal Medicine, Facultad de Veterinaria, Universidad de Extremadura, Av. de la Universidad s/n, 10003 Cáceres, Spain; Neobeitar S.L., Av. Alemania 6, 1° B, 10001, Cáceres, Spain.
| | - I Rey-Casero
- Neobeitar S.L., Av. Alemania 6, 1° B, 10001, Cáceres, Spain
| | - M Martín
- Neobeitar S.L., Av. Alemania 6, 1° B, 10001, Cáceres, Spain
| | - A Pérez
- Neobeitar S.L., Av. Alemania 6, 1° B, 10001, Cáceres, Spain
| | - M Benito-Murcia
- Neobeitar S.L., Av. Alemania 6, 1° B, 10001, Cáceres, Spain.
| | - D Risco
- Department of Animal Medicine, Facultad de Veterinaria, Universidad de Extremadura, Av. de la Universidad s/n, 10003 Cáceres, Spain.
| |
Collapse
|
15
|
Ebrahiminejad A, Sepahi AA, Yadegar A, Meyfour A. Pasteurized form of a potential probiotic lactobacillus brevis IBRC-M10790 exerts anti-inflammatory effects on inflammatory bowel disease in vitro. BMC Complement Med Ther 2024; 24:258. [PMID: 38987744 PMCID: PMC11234635 DOI: 10.1186/s12906-024-04576-1] [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: 03/16/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the gastrointestinal system. So far, no treatment has been identified that can completely cure IBD. Lactobacillus brevis is hypothesized to be beneficial in preventing inflammation. This study aimed to evaluate the potential probiotic effects of live and pasteurized L. brevis IBRC-M10790 on the in vitro cell co-culture model of IBD. METHODS An in vitro intestinal model was established using a transwell co-culture system of Caco-2 intestinal epithelial cells and RAW264.7 macrophages. Inflammatory conditions were induced in RAW264.7 cells using lipopolysaccharide. The effects of live and pasteurized L. brevis IBRC-M10790 on inflammatory mediators and epithelial barrier markers were investigated. RESULTS L. brevis IBRC-M10790 was able to significantly decrease the proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and increase the anti-inflammatory cytokine (IL-10) in the in vitro co-culture system. In addition, L. brevis increased adherens and tight junction (TJ) markers (ZO-1, E-cadherin, and Occludin) in Caco-2 intestinal epithelial cells. Based on the results, pasteurized L. brevis showed a higher protective effect than live L. brevis. CONCLUSIONS Our findings suggest that live and pasteurized forms of L. brevis possess probiotic properties and can mitigate inflammatory conditions in IBD.
Collapse
Affiliation(s)
- Ardeshir Ebrahiminejad
- Department of Microbiology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Abbas Akhavan Sepahi
- Department of Microbiology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Dmytriv TR, Storey KB, Lushchak VI. Intestinal barrier permeability: the influence of gut microbiota, nutrition, and exercise. Front Physiol 2024; 15:1380713. [PMID: 39040079 PMCID: PMC11260943 DOI: 10.3389/fphys.2024.1380713] [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: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
The intestinal wall is a selectively permeable barrier between the content of the intestinal lumen and the internal environment of the body. Disturbances of intestinal wall permeability can potentially lead to unwanted activation of the enteric immune system due to excessive contact with gut microbiota and its components, and the development of endotoxemia, when the level of bacterial lipopolysaccharides increases in the blood, causing chronic low-intensity inflammation. In this review, the following aspects are covered: the structure of the intestinal wall barrier; the influence of the gut microbiota on the permeability of the intestinal wall via the regulation of functioning of tight junction proteins, synthesis/degradation of mucus and antioxidant effects; the molecular mechanisms of activation of the pro-inflammatory response caused by bacterial invasion through the TLR4-induced TIRAP/MyD88 and TRAM/TRIF signaling cascades; the influence of nutrition on intestinal permeability, and the influence of exercise with an emphasis on exercise-induced heat stress and hypoxia. Overall, this review provides some insight into how to prevent excessive intestinal barrier permeability and the associated inflammatory processes involved in many if not most pathologies. Some diets and physical exercise are supposed to be non-pharmacological approaches to maintain the integrity of intestinal barrier function and provide its efficient operation. However, at an early age, the increased intestinal permeability has a hormetic effect and contributes to the development of the immune system.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
17
|
Zhang J, Zhang C, Yu L, Tian F, Chen W, Zhai Q. Analysis of the key genes of Lactobacillus reuteri strains involved in the protection against alcohol-induced intestinal barrier damage. Food Funct 2024; 15:6629-6641. [PMID: 38812427 DOI: 10.1039/d4fo01796j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Gastrointestinal inflammation and intestinal barrier function have important effects on human health. Alcohol, an important foodborne hazard factor, damages the intestinal barrier, increasing the risk of disease. Lactobacillus reuteri strains have been reported to reduce gastrointestinal inflammation and strengthen the intestinal barrier. In this study, we selected three anti-inflammatory L. reuteri strains to evaluate their role in the protection of the intestinal barrier and their immunomodulatory activity in a mouse model of gradient alcohol intake. Among the three strains tested (FSCDJY33M3, FGSZY33L6, and FCQHCL8L6), L. reuteri FSCDJY33M3 was found to protect the intestinal barrier most effectively, possibly due to its ability to reduce the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) and increase the expression of tight junction proteins (occludin, claudin-3). Genomic analysis suggested that the protective effects of L. reuteri FSCDJY33M3 may be related to functional genes and glycoside hydrolases associated with energy production and conversion, amino acid transport and metabolism, carbohydrate transport and metabolism, and DNA replication, recombination, and repair. These genes include COG2856, COG1804, COG2071, and COG1061, which encode adenine deaminase, acyl-CoA transferases, glutamine amidotransferase, RNA helicase, and glycoside hydrolases, including GH13_20, GH53, and GH70. Our results identified functional genes that may be related to protection against alcohol-induced intestinal barrier damage, which might be useful for screening lactic acid bacterial strains that can protect the intestinal barrier.
Collapse
Affiliation(s)
- Jiayi Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 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
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 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
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 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
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 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
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 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
| |
Collapse
|
18
|
Cao X, Wang B, Ren W, Wang J, Liu Q, Ren Y, Meng X. Mechanism of Ligilactobacillus salivarius GX118 in Regulating the Growth of Rainbow Trout (Oncorhynchus mykiss) and Resistance to Aeromonas salmonicida Infection. FISHES 2024; 9:157. [DOI: 10.3390/fishes9050157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Lactic acid bacteria Ligilactobacillus salivarius has been shown to be a substitute for antibiotics in the treatment of bacterial disease in high animals. However, its beneficial mechanism in fish farming is still unclear. This study evaluated the antagonistic effects of the Ligilactobacillus salivarius GX118 strain on Aeromonas salmonicida and its regulation of rainbow trout growth in vivo and in vitro. The results found that GX118 produces an antibacterial substance that can directly destroy the cell wall of A. salmonicida. Whole-genome sequencing of GX118 revealed that Enterolysin A is a type III bacteriocin with antibacterial properties. An in vivo experiment showed that the supplementation of GX118 in diet competitively inhibited the colonization of A. salmonicida in the intestine. In addition, it was able to improve the growth performance of rainbow trout within a 21-day feeding experiment. The supplementation of GX118 increased the diversity of gut microbiota, in which the abundance of Bacteroidota, Blautia, and Rhodobacteraceae increased. In addition, the use of GX118 activated the expression of IFN-γ and NF-κB genes and reduced the expression level of IL-6 and IL-8, thus exhibiting a certain effect on activating the immunity of rainbow trout. This study provides a scientific basis for the development of antibacterial probiotics in the healthy farming of rainbow trout.
Collapse
Affiliation(s)
- Xiyu Cao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Bowen Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Wenhao Ren
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Jiang Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Qi Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Yichao Ren
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Xianliang Meng
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266072, China
| |
Collapse
|
19
|
Li Y, Ma M, Wang X, Li J, Fang Z, Li J, Yang B, Lu Y, Xu X, Li Y. Celecoxib alleviates the DSS-induced ulcerative colitis in mice by enhancing intestinal barrier function, inhibiting ferroptosis and suppressing apoptosis. Immunopharmacol Immunotoxicol 2024; 46:240-254. [PMID: 38156770 DOI: 10.1080/08923973.2023.2300508] [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: 12/18/2022] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Ulcerative colitis (UC) is an inflammatory intestine disease characterized by dysfunction of the intestinal mucosal barrier, ferroptosis, and apoptosis. Previous researches suggest that celecoxib, a nonsteroidal anti-inflammatory drug, holds promise in alleviating inflammation in UC. Therefore, this study aims to investigate the effects and mechanisms of celecoxib in UC. METHODS To identify ferroptosis-related drugs and genes associated with UC, we utilized the Gene Expression Omnibus (GEO), FerrDb databases, and DGIdb database. Subsequently, we established a 2.5% DSS (Dextran sulfate sodium)-induced colitis model in mice and treated them with 10 mg/kg of celecoxib to validate the bioinformatics results. We evaluated histological pathologies, inflammatory response, intestinal barrier function, ferroptosis markers, and apoptosis regulators. RESULTS Celecoxib treatment significantly ameliorated DSS-induced UC in mice, as evidenced by the body weight change curve, colon length change curve, disease activity index (DAI) score, and histological index score. Celecoxib treatment reduced the level of pro-inflammatory factors and promoted the expressions of intestinal tight junction proteins such as Claudin-1 and Occludin, thereby restoring the integrity of the intestinal mucosal barrier. Furthermore, celecoxib treatment reversed the ferroptosis characteristics in DSS-induced mice by increasing glutathione (GSH), decreasing malondialdehyde (MDA), and increasing the expression of GPX-4 and xCT. Additionally, apoptosis was induced in mice with UC, as evidenced by increased Caspase3, BAD, P53, BAX, Caspase9 and Aifm1 production, and decreased expression of BCL-XL and BCL2. Celecoxib treatment significantly reversed the apoptotic changes in DSS-induced mice. CONCLUSION Our findings suggest that celecoxib effectively treats DSS-induced UC in mice by inhibiting ferroptosis and apoptosis.
Collapse
Affiliation(s)
- Yaxian Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengdi Ma
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaodong Wang
- The Robert Bosch Center for Tumor Diseases (RBCT), Stuttgart, Germany
| | - Jing Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziqing Fang
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianhui Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Yang
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yida Lu
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Xu
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongxiang Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Tang X. Probiotic Roles of Clostridium butyricum in Piglets: Considering Aspects of Intestinal Barrier Function. Animals (Basel) 2024; 14:1069. [PMID: 38612308 PMCID: PMC11010893 DOI: 10.3390/ani14071069] [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: 02/20/2024] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
China, as the global leader in pork production and consumption, is faced with challenges in ensuring sustainable and wholesome growth of the pig industry while also guaranteeing meat food safety amidst the ban on antibiotics usage in animal feed. The focus of the pig industry lies in guaranteeing piglet health and enhancing overall production performance through nutrition regulation. Clostridium butyricum (C. butyricum), a new type of probiotic, possesses characteristics such as heat resistance, acid resistance, and bile-salt tolerance, meaning it has potential as a feed additive. Previous studies have demonstrated that C. butyricum has a probiotic effect on piglets and can serve as a substitute for antibiotics. The objective of this study was to review the probiotic role of C. butyricum in the production of piglets, specifically focusing on intestinal barrier function. Through this review, we explored the probiotic effects of C. butyricum on piglets from the perspective of intestinal health. That is, C. butyricum promotes intestinal health by regulating the functions of the mechanical barrier, chemical barrier, immune barrier, and microbial barrier of piglets, thereby improving the growth of piglets. This review can provide a reference for the rational utilization and application of C. butyricum in swine production.
Collapse
Affiliation(s)
- Xiaopeng Tang
- State Engineering Technology Institute for Karst Desertification Control, School of Karst Science, Guizhou Normal University, Guiyang 550025, China
| |
Collapse
|
21
|
Wu Y, Liu X, Zou Y, Zhang X, Wang Z, Hu J, Han D, Zhao J, Dai Z, Wang J. Lactobacillus amylovorus Promotes Lactose Utilization in Small Intestine and Enhances Intestinal Barrier Function in Intrauterine Growth Restricted Piglets. J Nutr 2024; 154:535-542. [PMID: 38072153 DOI: 10.1016/j.tjnut.2023.12.004] [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: 07/31/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) resulted in high mortality and many physiological defects of piglets, causing huge economic loss in the swine industry. Lactobacillus amylovorus (L. amylovorus) was identified as one of the main differential bacteria between IUGR and normal piglets. However, the effects of L. amylovorus on the growth performance and intestinal health in IUGR piglets remained unclear. OBJECTIVES This study aimed to investigate the promoting effects of L. amylovorus Mafic1501, a new strain isolated from normal piglets, on the growth performance and intestinal barrier functions in IUGR piglets. METHODS Newborn mice or piglets were assigned into 3 groups: CON (normal birth weight, control), IUGR (low birth weight), and IUGR+L. amy (low birth weight), administered with sterile saline or L. amylovorus Mafic1501, respectively. Growth performance, lactose content in the digesta, intestinal lactose transporter, and barrier function parameters were profiled. IPEC-J2 cells were cultured to verify the effects of L. amylovorus Mafic1501 on lactose utilization and intestinal barrier functions. RESULTS L. amylovorus Mafic1501 elevated body weight and average daily gain of IUGR mice and piglets (P < 0.05). The lactose content in the ileum was decreased, whereas gene expression of glucose transporter 2 (GLUT2) was increased by L. amylovorus Mafic1501 in IUGR piglets during suckling period (P < 0.05). Besides, L. amylovorus Mafic1501 promoted intestinal barrier functions by increasing the villus height and relative gene expressions of tight junctions (P < 0.05). L. amylovorus Mafic1501 and its culture supernatant decreased the lactose level in the medium and upregulated gene expressions of transporter GLUT2 and tight junction protein Claudin-1 of IPEC-J2 cells (P < 0.05). CONCLUSION L. amylovorus Mafic1501 improved the growth performance of IUGR piglets by promoting the lactose utilization in small intestine and enhancing intestinal barrier functions. Our results provided the new evidence of L. amylovorus Mafic1501 for its application in the swine industry.
Collapse
Affiliation(s)
- Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiaoyi Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Youwei Zou
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jie Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
22
|
Kaur S, Sharma P, Mayer MJ, Neuert S, Narbad A, Kaur S. Beneficial effects of GABA-producing potential probiotic Limosilactobacillus fermentum L18 of human origin on intestinal permeability and human gut microbiota. Microb Cell Fact 2023; 22:256. [PMID: 38087304 PMCID: PMC10717626 DOI: 10.1186/s12934-023-02264-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) is a non-protein amino acid with neuroinhibitory, antidiabetic, and antihypertensive properties and is used as a drug for treating anxiety and depression. Some strains of lactobacilli are known to produce GABA and strengthen the gut barrier function which play an important role in ameliorating the effects caused by the pathogen on the gut barrier. The probiotic bacteria are also known to modulate the human fecal microbiota, however, the role of GABA-producing strains on the gut epithelium permeability and gut microbiota is not known. RESULTS In this study, we report the production of high levels of GABA by potential probiotic bacterium Limosilactobacillus fermentum L18 for the first time. The kinetics of the production of GABA by L18 showed that the maximum production of GABA in the culture supernatant (CS) occurred at 24 h, whereas in fermented milk it took 48 h of fermentation. The effect of L18 on the restoration of lipopolysaccharide (LPS)-disrupted intestinal cell membrane permeability in Caco-2 monolayers showed that it significantly restored the transepithelial electrical resistance (TEER) values, by significantly increasing the levels of junction proteins, occludin and E-cadherin in L18 and LPS-treated Caco-2 cells as compared to only LPS-treated cells. The effect of GABA-secreting L18 on the metataxonome of human stool samples from healthy individuals was investigated by a batch fermentor that mimics the conditions of the human colon. Although, no differences were observed in the α and β diversities of the L18-treated and untreated samples at 24 h, the relative abundances of bacterial families Lactobacillaceae and Bifidobacteriaceae increased in the L18-treated group, but both decreased in the untreated groups. On the other hand, the relative abundance of Enterobacteriaceae decreased in the L18 samples but it increased in the untreated samples. CONCLUSION These results indicate that Li. fermentum L18 is a promising GABA-secreting strain that strengthens the gut epithelial barrier by increasing junction protein concentrations and positively modulating the gut microbiota. It has the potential to be used as a psychobiotic or for the production of functional foods for the management of anxiety-related illnesses.
Collapse
Affiliation(s)
- Sumanpreet Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Medical Laboratory Sciences, Lovely Professional University, Jalandhar, India
| | - Preeti Sharma
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India
| | - Melinda J Mayer
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Saskia Neuert
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- East Genomics Laboratory Hub, Cambridge University Hospitals Genomic Laboratory, Hills Road, Cambridge, UK
| | - Arjan Narbad
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Sukhraj Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India.
| |
Collapse
|
23
|
Gong L, Mahmood T, Mercier Y, Xu H, Zhang X, Zhao Y, Luo Y, Guo Y. Dietary methionine sources and levels modulate the intestinal health status of broiler chickens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:242-255. [PMID: 38033606 PMCID: PMC10684994 DOI: 10.1016/j.aninu.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 12/02/2023]
Abstract
Given the key role of methionine in biological processes, adequate methionine should be provided to meet the nutritional requirements. DL-2-hydroxy-4-(methylthio)-butanoic acid (DL-HMTBA) has been considered as an important source of methionine. However, the effects of different sources and levels of methionine on the intestinal health status have not been clarified yet. An experiment was carried out to investigate the effects of different dietary sources and levels of methionine on the intestinal epithelial barrier, inflammatory cytokines expression, ileal morphology, microbiota composition, and cecal short chain fatty acids (SCFA) profiles. For this purpose, 720 male Arbor Acre broiler chicks at 1 d old were randomly assigned to a 2 × 3 factorial arrangement with 2 methionine sources (DL-methionine and DL-HMTBA) and 3 total sulfur amino acids (TSAA) levels (80%, 100%, and 120% of Arbor Acre recommendation). The results showed that DL-HMTBA supplementation promoted intestinal physical barrier at both gene expression level of claudin-1 and serum diamine oxidase level (P < 0.05), and the inflammatory cytokine IL-6 mRNA expression was down-regulated by dietary DL-HMTBA supplementation compared with the DL-methionine group (P < 0.05). Meanwhile, an upregulated gene expression of claudin-1 and zonula occluden-1 (ZO-1) were observed in the low-TSAA treatment on d 14 (P < 0.05), whereas this treatment increased the expression of IL-1β and IL-6 (P < 0.05). Villus height to crypt depth ratio was high (P < 0.05) in the middle-level TSAA group. Furthermore, DL-HMTBA supplementation optimized the microbiota of the ileum especially the relative abundance of Lactobacillus, where the digestion and absorption were completed, and elevated the concentrations of SCFA (acetate, propionate, and butyrate) in the cecal content on d 21 (P < 0.01). In conclusion, dietary DL-HMTBA supplementation improved the intestinal barrier function, immune homeostasis and optimized the microbiota to promote intestinal health status in broiler chickens.
Collapse
Affiliation(s)
- Lu Gong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | | | | | - Huiping Xu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaodan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yizhu Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yimeng Luo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
24
|
Zhang Y, Hu J, Song X, Dai J, Tang Z, Huang G, Jiao W, Wu Y, Wang C, Du L, Jin Y. The effects of Lactobacillus reuteri microcapsules on radiation-induced brain injury by regulating the gut microenvironment. Food Funct 2023; 14:10041-10051. [PMID: 37843434 DOI: 10.1039/d3fo03008c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
A radiation-induced brain injury (RIBI) is a major adverse event following radiotherapy of malignant tumors. RIBI would affect cognitive function, leading to a series of complications and even death. However, the pathogenesis of RIBI is still unclear, and it still lacks specific therapeutic drugs. The gut-brain bidirectional communication may be mediated by various microbiota and metabolites in the gastrointestinal tract. Probiotics are closely related to physiological health. The theory of the gut-brain axis provides us with a new idea to improve the gut microenvironment by supplementing probiotics against RIBI. Here, Lactobacillus reuteri microcapsules (LMCs) were prepared, which were predominantly irregular spheres with a rough surface under a scanning electron microscope and a narrow size distribution ranging from 20 to 700 μm. The transmission electron microscopy images showed that the structure of microcapsules containing Lactobacillus reuteri (L. reuteri) was a core and shell structure. The survival of L. reuteri in microcapsules was significantly more than that of free L. reuteri in the simulated stomach environment of pH 1.2. 16S rDNA sequencing showed that LMCs observably increased the relative abundance of Lactobacillus in RIBI mice. More importantly, compared with the RIBI model mice, the behavior of RIBI mice treated with LMCs was significantly improved. In addition, LMCs greatly alleviated the pathological damage of the hippocampus and intestines in the mice after irradiation and reduced the level of TNF-α and IL-6 in vivo. Generally, LMCs are a promising oral preparation, which provide new ideas and methods for the treatment of RIBI.
Collapse
Affiliation(s)
- Yizhi Zhang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jinglu Hu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
| | - Xingshuang Song
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing Dai
- Information Department, General Hospital of Western Zone, Chengdu 610083, China
| | - Ziyan Tang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Guiyu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
| | - Wencheng Jiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Hebei University, Baoding 071000, China
| | - Yanping Wu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chenyun Wang
- The Fourth Clinical Center Affiliated to Chinese PLA General Hospital, Beijing 100048, China
| | - Lina Du
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
- Hebei University, Baoding 071000, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
| |
Collapse
|
25
|
Yang W, Zhang X, Qi L, Wang Z, Wu W, Feng W, Gu Y. Colon-targeted EMSCs conditional medium hydrogel for treatment of ulcerative colitis in mice. Biomed Mater 2023; 18:065010. [PMID: 37722391 DOI: 10.1088/1748-605x/acfadb] [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: 03/22/2023] [Accepted: 09/18/2023] [Indexed: 09/20/2023]
Abstract
Oral ecto-mesenchymal stem cells-conditional medium (EMSCs-CM) is a promising strategy for treating ulcerative colitis (UC). However, this therapy is currently limited by the harsh gastrointestinal environment and poor colonic targeting ability. Herein, a glutamine transaminase 2 (TG2) crosslinked EMSCs-CM hydrogel (EMSCs-CM-Gel) was fabricated by combining EMSCs-CM with negatively chargedγ-polyglutamic acid (γ-PGA) hydrogel. Intestinal epithelial cell 6 (IEC-6) was applied to construct a cell model with lipopolysaccharide to evaluate the anti-inflammatory potential of EMSCs-CMin vitro. The crosslinked gel was orally administered to mice in liquid form to access the effects of EMSCs-CM-Gelin vivo. This study was based on the fact that the hydrogel containing EMSCs-CM has negative charges, which ensure it remains at the positively charged inflamed colon tissue. The EMSCs-CM could continuously be released in the damaged colon mucosa along with the degradation of theγ-PGA hydrogel. Immunofluorescence and western blot were performed to assess the effects of EMSCs-CM-Gel on mice. The resultsin vivoshowed that EMSCs-CM-Gel could significantly suppress the expression of inflammatory cytokines, prevent the shortening of the length of the intestine and repair the intestinal barrier. Collectively, our findings provided a novel colon-targeted strategy, hoping to benefit UC patients a lot.
Collapse
Affiliation(s)
- Wenjing Yang
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Xingxing Zhang
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Liuyao Qi
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Zhe Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Weijiang Wu
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Wenjing Feng
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Yahan Gu
- School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
| |
Collapse
|
26
|
Palkovicsné Pézsa N, Kovács D, Somogyi F, Karancsi Z, Móritz AV, Jerzsele Á, Rácz B, Farkas O. Effects of Lactobacillus rhamnosus DSM7133 on Intestinal Porcine Epithelial Cells. Animals (Basel) 2023; 13:3007. [PMID: 37835613 PMCID: PMC10571805 DOI: 10.3390/ani13193007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Antimicrobial resistance is one of the biggest health challenges nowadays. Probiotics are promising candidates as feed additives contributing to the health of the gastrointestinal tract. The beneficial effect of probiotics is species/strain specific; the potential benefits need to be individually assessed for each probiotic strain or species. We established a co-culture model, in which gastrointestinal infection was modeled using Escherichia coli (E. coli) and Salmonella enterica serovar Typhimurium (S. enterica serovar Typhimurium). Using intestinal porcine epithelial cells (IPEC-J2), the effects of pre-, co-, and post-treatment with Lactobacillus (L.) rhamnosus on the barrier function, intracellular (IC) reactive oxygen species (ROS) production, proinflammatory cytokine (IL-6 and IL-8) response, and adhesion inhibition were tested. E. coli- and S. Typhimurium-induced barrier impairment and increased ROS production could be counteracted using L. rhamnosus (p < 0.01). S. Typhimurium-induced IL-6 production was reduced via pre-treatment (p < 0.05) and post-treatment (p < 0.01); increased IL-8 secretion was decreased via pre-, co-, and post-treatment (p < 0.01) with L. rhamnosus. L. rhamnosus demonstrated significant inhibition of adhesion for both S. Typhimurium (p < 0.001) and E. coli (p < 0.001 in both pre-treatment and post-treatment; p < 0.05 in co-treatment). This study makes a substantial contribution to the understanding of the specific benefits of L. rhamnosus. Our findings can serve as a basis for further in vivo studies carried out in pigs and humans.
Collapse
Affiliation(s)
- Nikolett Palkovicsné Pézsa
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Dóra Kovács
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Fanni Somogyi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
| | - Zita Karancsi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Alma Virág Móritz
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary;
| | - Orsolya Farkas
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (D.K.); (F.S.); (Z.K.); (A.V.M.); (Á.J.); (O.F.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| |
Collapse
|
27
|
Kiernan DP, O’Doherty JV, Sweeney T. The Effect of Maternal Probiotic or Synbiotic Supplementation on Sow and Offspring Gastrointestinal Microbiota, Health, and Performance. Animals (Basel) 2023; 13:2996. [PMID: 37835602 PMCID: PMC10571980 DOI: 10.3390/ani13192996] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The increasing prevalence of antimicrobial-resistant pathogens has prompted the reduction in antibiotic and antimicrobial use in commercial pig production. This has led to increased research efforts to identify alternative dietary interventions to support the health and development of the pig. The crucial role of the GIT microbiota in animal health and performance is becoming increasingly evident. Hence, promoting an improved GIT microbiota, particularly the pioneer microbiota in the young pig, is a fundamental focus. Recent research has indicated that the sow's GIT microbiota is a significant contributor to the development of the offspring's microbiota. Thus, dietary manipulation of the sow's microbiota with probiotics or synbiotics, before farrowing and during lactation, is a compelling area of exploration. This review aims to identify the potential health benefits of maternal probiotic or synbiotic supplementation to both the sow and her offspring and to explore their possible modes of action. Finally, the results of maternal sow probiotic and synbiotic supplementation studies are collated and summarized. Maternal probiotic or synbiotic supplementation offers an effective strategy to modulate the sow's microbiota and thereby enhance the formation of a health-promoting pioneer microbiota in the offspring. In addition, this strategy can potentially reduce oxidative stress and inflammation in the sow and her offspring, enhance the immune potential of the milk, the immune system development in the offspring, and the sow's feed intake during lactation. Although many studies have used probiotics in the maternal sow diet, the most effective probiotic or probiotic blends remain unclear. To this extent, further direct comparative investigations using different probiotics are warranted to advance the current understanding in this area. Moreover, the number of investigations supplementing synbiotics in the maternal sow diet is limited and is an area where further exploration is warranted.
Collapse
Affiliation(s)
- Dillon P. Kiernan
- School of Veterinary Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, D04 C1P1 Dublin, Ireland;
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
| |
Collapse
|
28
|
Duarte ME, Garavito-Duarte Y, Kim SW. Impacts of F18 +Escherichia coli on Intestinal Health of Nursery Pigs and Dietary Interventions. Animals (Basel) 2023; 13:2791. [PMID: 37685055 PMCID: PMC10487041 DOI: 10.3390/ani13172791] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
This review focused on the impact of F18+E. coli on pig production and explored nutritional interventions to mitigate its deleterious effects. F18+E. coli is a primary cause of PWD in nursery pigs, resulting in substantial economic losses through diminished feed efficiency, morbidity, and mortality. In summary, the F18+E. coli induces intestinal inflammation with elevated IL6 (60%), IL8 (43%), and TNF-α (28%), disrupting the microbiota and resulting in 14% villus height reduction. Besides the mortality, the compromised intestinal health results in a 20% G:F decrease and a 10% ADFI reduction, ultimately culminating in a 28% ADG decrease. Among nutritional interventions to counter F18+E. coli impacts, zinc glycinate lowered TNF-α (26%) and protein carbonyl (45%) in jejunal mucosa, resulting in a 39% ADG increase. Lactic acid bacteria reduced TNF-α (36%), increasing 51% ADG, whereas Bacillus spp. reduced IL6 (27%), increasing BW (12%). Lactobacillus postbiotic increased BW (14%) and the diversity of beneficial bacteria. Phytobiotics reduced TNF-α (23%) and IL6 (21%), enhancing feed efficiency (37%). Additional interventions, including low crude protein formulation, antibacterial minerals, prebiotics, and organic acids, can be effectively used to combat F18+E. coli infection. These findings collectively underscore a range of effective strategies for managing the challenges posed by F18+E. coli in pig production.
Collapse
Affiliation(s)
| | | | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (M.E.D.); (Y.G.-D.)
| |
Collapse
|
29
|
He Z, Liu J, Liu Y. Daphnetin attenuates intestinal inflammation, oxidative stress, and apoptosis in ulcerative colitis via inhibiting REG3A-dependent JAK2/STAT3 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:2132-2142. [PMID: 37209277 DOI: 10.1002/tox.23837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Daphnetin is a natural coumarin compound with anti-inflammatory, anti-oxidant, and anti-apoptotic effects, which has been previously demonstrated to ameliorate DSS-induced ulcerative colitis (UC). However, the molecular mechanism involved in the daphnetin-mediated pathological process of UC remains unclarified. The current study used DSS-induced mice and LPS-challenged Caco-2 cells as UC models. Bodyweight, disease activity index (DAI) score, and colon length were used to evaluate the severity of colitis. The histological changes in colon tissues were observed using H&E and PAS staining. Protein levels were detected by western blot. The malondialdehyde (MDA) and superoxide dismutase (SOD) activities were used to assess oxidative stress. Inflammatory responses were evaluated by detecting the levels of inflammatory cytokines (IFN-r, IL-1β, IL-6, and TNF-α) using flow cytometry. CCK-8 and TUNEL assay were employed to determine cell growth and cell death, respectively. The results showed that daphnetin could ameliorate the severity of colitis and attenuate the damage to intestinal structure in DSS-induced mice. Compared with the DSS group, the expression of ZO-1, occludin, and anti-apoptotic protein (BCL-2) was increased while the level of pro-apoptotic proteins (Bax and cleaved caspase 3) was decreased in DSS + daphnetin group. The activity of MDA and SOD, as well as the levels of inflammatory cytokines were substantially suppressed by daphnetin. In consistency, in vitro assays indicated that daphnetin protected Caco-2 cells from LPS-stimulated viability impairment, apoptosis, oxidative stress, and inflammation. Furthermore, daphnetin suppressed the activity of JAK2/STAT signaling in LPS-induced Caco-2 cells in a REG3A-dependent manner. REG3A overexpression abated the ameliorative effects of daphnetin while JAK2/STAT signaling inhibition functioned synergically with daphnetin in LPS-stimulated Caco-2 cells. Collectively, this study deepened the understanding of the therapeutic effects of daphnetin on UC and uncovered for the first time that daphnetin functioned through REG3A-activated JAK2/STAT3 signaling in UC, which may provide novel insights for the treatment of UC.
Collapse
Affiliation(s)
- Zhi He
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jingjing Liu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yang Liu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
30
|
Luo Z, Chen A, Xie A, Liu X, Jiang S, Yu R. Limosilactobacillus reuteri in immunomodulation: molecular mechanisms and potential applications. Front Immunol 2023; 14:1228754. [PMID: 37638038 PMCID: PMC10450031 DOI: 10.3389/fimmu.2023.1228754] [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: 05/25/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Frequent use of hormones and drugs may be associated with side-effects. Recent studies have shown that probiotics have effects on the prevention and treatment of immune-related diseases. Limosilactobacillus reuteri (L. reuteri) had regulatory effects on intestinal microbiota, host epithelial cells, immune cells, cytokines, antibodies (Ab), toll-like receptors (TLRs), tryptophan (Try) metabolism, antioxidant enzymes, and expression of related genes, and exhibits antibacterial and anti-inflammatory effects, leading to alleviation of disease symptoms. Although the specific composition of the cell-free supernatant (CFS) of L. reuteri has not been clarified, its efficacy in animal models has drawn increased attention to its potential use. This review summarizes the effects of L. reuteri on intestinal flora and immune regulation, and discusses the feasibility of its application in atopic dermatitis (AD), asthma, necrotizing enterocolitis (NEC), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS), and provides insights for the prevention and treatment of immune-related diseases.
Collapse
Affiliation(s)
- Zichen Luo
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ailing Chen
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Anni Xie
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Xueying Liu
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Renqiang Yu
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
31
|
Zhang P, Chen J, Ming Y, Niu Y. Probiotics treatment ameliorated mycophenolic acid-induced colitis by enhancing intestinal barrier function and improving intestinal microbiota dysbiosis in mice. Front Microbiol 2023; 14:1153188. [PMID: 37533828 PMCID: PMC10390739 DOI: 10.3389/fmicb.2023.1153188] [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: 01/29/2023] [Accepted: 07/04/2023] [Indexed: 08/04/2023] Open
Abstract
Background Mycophenolic acid (MPA)-induced colitis was still a severe side effect and challenge faced by solid transplant recipients. We aimed to explore the function and mechanism of probiotics in the treatment of MPA-induced colitis. Methods In this study, 15 mice (C57BL/6) were randomly assigned into three groups: control (CNTL) group (n = 5), MPA group (n = 5) and the MPA + Probiotic group (n = 5). Bifid Triple Viable capsules, which were drugs for clinical use and consisted of Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis, were used in Probiotic group. Body weight change, stool scores, colon histopathology and morphology were used to evaluate the disease severity. The intestinal mucosal barrier function was assessed by measuring the expression level of secretory immunoglobulin A (sIgA), Zonula occludens-1 (ZO-1) and Occludin. Finally, 16S rDNA sequencing and bioinformatics analysis were performed on mice feces to compare the different intestinal microbial composition and diversity among three groups. Results Compared with the CNTL group, the mice in MPA group showed a significantly decreased body weight, increased stool scores, shortened colon length and severe colon inflammation. However, probiotics treated MPA mice reversed the disease severity, indicating that probiotics ameliorated MPA-induced colitis in mice. Mechanistically, probiotics improved the intestinal barrier function by up-regulating the expression of sIgA, ZO-1 and Occludin. Moreover, MPA-induced colitis led to intestinal microbiota dysbiosis, including the change of Firmicutes/Bacteroidetes ratio, α- and β-diversity. But probiotic treated group showed mild change in these microbial features. Additionally, we found that Clostridiales was the most significantly different microbiota flora in MPA group. Conclusion Probiotics treatment ameliorated MPA-induced colitis by enhancing intestinal barrier function and improving intestinal microbiota dysbiosis. Clostridiales might be the dominant functional intestinal microflora and serve as the potential therapy target in MPA-induced colitis.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jinwen Chen
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yingzi Ming
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying Niu
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
32
|
Du W, Wang X, Hu M, Hou J, Du Y, Si W, Yang L, Xu L, Xu Q. Modulating gastrointestinal microbiota to alleviate diarrhea in calves. Front Microbiol 2023; 14:1181545. [PMID: 37362944 PMCID: PMC10286795 DOI: 10.3389/fmicb.2023.1181545] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
The calf stage is a critical period for the development of heifers. Newborn calves have low gastrointestinal barrier function and immunity before weaning, making them highly susceptible to infection by various intestinal pathogens. Diarrhea in calves poses a significant threat to the health of young ruminants and may cause serious economic losses to livestock farms. Antibiotics are commonly used to treat diarrhea and promote calf growth, leading to bacterial resistance and increasing antibiotic residues in meat. Therefore, finding new technologies to improve the diarrhea of newborn calves is a challenge for livestock production and public health. The operation of the gut microbiota in the early stages after birth is crucial for optimizing immune function and body growth. Microbiota colonization of newborn animals is crucial for healthy development. Early intervention of the calf gastrointestinal microbiota, such as oral probiotics, fecal microbiota transplantation and rumen microbiota transplantation can effectively relieve calf diarrhea. This review focuses on the role and mechanisms of oral probiotics such as Lactobacillus, Bifidobacterium and Faecalibacterium in relieving calf diarrhea. The aim is to develop appropriate antibiotic alternatives to improve calf health in a sustainable and responsible manner, while addressing public health issues related to the use of antibiotics in livestock.
Collapse
|
33
|
Abstract
A wound is an injury to the skin or damage to the body tissue. The healing process differs between various kinds of wounds. Treatment of hard-to-heal (chronic) wounds becomes challenging for healthcare practitioners, especially if patients have underlying health complications such as diabetes. Infection of wounds is another factor that interferes with the healing process and extends its duration. Active research is being conducted into the development of advanced wound dressing technologies. These wound dressings are intended to manage the exudate, reduce bacterial infection and speed up the healing process. Probiotics have been receiving much attention because of their potential application in the clinical field, especially in diagnostics and treatment strategies of various infectious and non-infectious diseases. The host immune-modulatory response and antimicrobial activity of probiotics are expanding their role in the development of improved wound dressing technology.
Collapse
Affiliation(s)
- Shanmugaraja Meenakshi
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam-603103, Chennai, Tamil Nadu, India
| | - Ramadevi Santhanakumar
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam-603103, Chennai, Tamil Nadu, India
| |
Collapse
|
34
|
Luo D, Huang Z, Jia G, Zhao H, Liu G, Chen X. Naringin mitigates LPS-induced intestinal barrier injury in mice. Food Funct 2023; 14:1617-1626. [PMID: 36688440 DOI: 10.1039/d2fo03586c] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The aim of this study was to investigate the effect of naringin on lipopolysaccharide (LPS)-induced jejunal barrier function in mice. Forty-five 3-week-old healthy male Balb/c mice with similar body weights were randomly divided into control group, LPS group, LPS + naringin group, with 15 mice in each treatment group. The mice were intraperitoneally injected with the same dose of saline or LPS (10 mg per kg BW) at 43 d. The blood samples, liver and jejunal tissues were collected after 3 h of injection. The results showed that LPS significantly increased the serum diamine oxidase (DAO) activity, D-lactate (D-LA) concentration, and malondialdehyde (MDA) content in liver and jejunum, while decreased the activities of superoxide dismutase (SOD), glutathione peroxidase (Gpx) and catalase (CAT) in liver and jejunum. The LPS treatment caused an increase in the crypt depth and a decrease in the villus height and the ratio of villus height to crypt depth (V/C) of the jejunum. In addition, the LPS treatment significantly increased the mRNA expressions of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, toll-like receptor 4 (TLR4), p38-mitogen-activated protein kinase (p38 MAPK), nuclear factor-κB (NF-κB) and kelch-like ECH-associated protein 1 (Keap1), while decreased mRNA expressions of zonula occludens 1 (ZO-1), occludin, claudin, mucin 2 (MUC2) and junctional adhesion molecule 2 (JAM2), Gpx, SOD1, GST, CAT and nuclear factor-erythroid 2-related factor 2 (Nrf2). However, the naringin treatment mitigated these effects induced by LPS. Taken together, our findings suggested that naringin attenuates LPS-induced intestinal barrier damage by inhibiting inflammatory factors and improving antioxidant function and intestinal tight junction, which might be mediated by activating the Nrf2 signaling and suppressing the TLR4/p38 MAPK/NF-κB signaling.
Collapse
Affiliation(s)
- Diaoyun Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China.
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China.
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China.
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China.
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China.
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China.
| |
Collapse
|
35
|
Gao X, Yu J, Chang L, Wang Y, Sun X, Mu G, Qian F. In vitro antibacterial activity of Bacillus coagulans T242 on Caco-2 cells infected with Salmonella Typhimurium. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
36
|
Liu J, Ma X, Zhuo Y, Xu S, Hua L, Li J, Feng B, Fang Z, Jiang X, Che L, Zhu Z, Lin Y, Wu D. The Effects of Bacillus subtilis QST713 and β-mannanase on growth performance, intestinal barrier function, and the gut microbiota in weaned piglets. J Anim Sci 2023; 101:skad257. [PMID: 37583344 PMCID: PMC10449409 DOI: 10.1093/jas/skad257] [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: 05/17/2023] [Accepted: 08/08/2023] [Indexed: 08/17/2023] Open
Abstract
We investigated the effects of different Bacillus subtilis QST713 doses and a B. subtilis QST713 and β-mannanase mix on growth performance, intestinal barrier function, and gut microbiota in weaned piglets. In total, 320 healthy piglets were randomly assigned to four groups: 1) control group (basal diet), 2) BS100 group (basal diet plus 100 mg/kg B. subtilis QST713), 3) BS200 group (basal diet plus 200 mg/kg B. subtilis QST713), and 4) a BS100XT group (basal diet plus 100 mg/kg B. subtilis QST713 and 150 mg/kg β-mannanase). The study duration was 42 d. We showed that feed intake in weaned piglets on days 1 to 21 was increased in group BS100 (P < 0.05), and that the feed conversion ratio in group BS100XT animals decreased throughout the study (P < 0.05). In terms of microbial counts, the BS100XT group showed reduced Escherichia coli and Clostridium perfringens numbers on day 21 (P < 0.05). Moreover, no significant α-diversity differences were observed across all groups during the study (P > 0.05). However, principal coordinates analysis indicated clear separations in bacterial community structures across groups (analysis of similarities: P < 0.05) on days 21 and 42. Additionally, E-cadherin, occludin, and zonula occludens-1 (ZO-1) expression in piglet feces increased (P < 0.05) by adding B. subtilis QST713 and β-mannanase to diets. Notably, this addition decreased short-chain fatty acid concentrations. In conclusion, B. subtilis QST713 addition or combined B. subtilis QST713 plus β-mannanase effectively improved growth performance, intestinal barrier function, and microbial balance in weaned piglets.
Collapse
Affiliation(s)
- Junchen Liu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiangyuan Ma
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yong Zhuo
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Shengyu Xu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Lun Hua
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jian Li
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bin Feng
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhengfeng Fang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xuemei Jiang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Lianqiang Che
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zeyuan Zhu
- Elanco Animal Health, Mutiara Damansara, Selangor, Malaysia
| | - Yan Lin
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - De Wu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
37
|
Limosilactobacillus reuteri Regulating Intestinal Function: A Review. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation9010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Probiotics have extensive use in daily life, due to the function of the changing intestinal metabolism and material conversion processes, wherein they remodel the intestinal microbiota, regulate the intestinal function and affect the organism’s health. Limosilactobacillus reuteri (L. reuteri), originally discovered in breast milk and currently reported to be present within the gut of almost all vertebrates and mammals, is an intestinal probiotic with prebiotic efficacy. Most L. reuteri have good intestinal colonization and bacteriocin secretion abilities, which can increase the expression of the mucin (mucoprotein) genes 2 MUC2 and MUC13, which in turn promote the development and maturation of intestinal organoids, and augment mucin secretion. In enteritis patients, L. reuteri downregulates α Tumor necrosis factor-α, (TNF-α), Interleukin-6 (IL-6), IL-8, and IL-12 expression to attenuate inflammation. It also induces the host’s production of immunoglobulin A (IGA), which manipulates the intestinal microbial community, inhibiting the growth of pathogens. L. reuteri has been widely used in daily life. with in-depth studies having been conducted on the prebiotic effects of L. reuteri. However, the complexity of its application in a clinical setting is still unclear because the pathogenesis of various diseases still requires a large amount of data and theoretical support.
Collapse
|
38
|
Lee WJ, Ryu S, Kang AN, Song M, Shin M, Oh S, Kim Y. Molecular characterization of gut microbiome in weaning pigs supplemented with multi-strain probiotics using metagenomic, culturomic, and metabolomic approaches. Anim Microbiome 2022; 4:60. [PMID: 36434671 PMCID: PMC9700986 DOI: 10.1186/s42523-022-00212-w] [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: 05/02/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Probiotics have been reported to exhibit positive effects on host health, including improved intestinal barrier function, preventing pathogenic infection, and promoting nutrient digestion efficiency. These internal changes are reflected to the fecal microbiota composition and, bacterial metabolites production. In accordance, the application of probiotics has been broadened to industrial animals, including swine, which makes people to pursue better knowledge of the correlation between changes in the fecal microbiota and metabolites. Therefore, this study evaluated the effect of multi-strain probiotics (MSP) supplementation to piglets utilizing multiomics analytical approaches including metagenomics, culturomics, and metabolomics. RESULTS Six-week-old piglets were supplemented with MSP composed of Lactobacillus isolated from the feces of healthy piglets. To examine the effect of MSP supplement, piglets of the same age were selected and divided into two groups; one with MSP supplement (MSP group) and the other one without MSP supplement (Control group). MSP feeding altered the composition of the fecal microbiota, as demonstrated by metagenomics analysis. The abundance of commensal Lactobacillus was increased by 2.39%, while Clostridium was decreased, which revealed the similar pattern to the culturomic approach. Next, we investigated the microbial metabolite profiles, specifically SCFAs using HPLC-MS/MS and others using GC-MS, respectively. MSP supplement elevated the abundance of amino acids, including valine, isoleucine and proline as well as the concentration of acetic acid. According to the correlation analyses, these alterations were found out to be crucial in energy synthesizing metabolism, such as branched-chain amino acid (BCAA) metabolism and coenzyme A biosynthesis. Furthermore, we isolated commensal Lactobacillus strains enriched by MSP supplement, and analyzed the metabolites and evaluated the functional improvement, related to tight junction from intestinal porcine enterocyte cell line (IPEC-J2). CONCLUSIONS In conclusion, MSP administration to piglets altered their fecal microbiota, by enriching commensal Lactobacillus strains. This change contributed amino acid, acetic acid, and BCAA concentrations to be increased, and energy metabolism pathway was also increased at in vivo and in vitro. These changes produced by MSP supplement suggests the correlation between the various physiological energy metabolism functions induced by health-promoting Lactobacillus and the growth performance of piglets.
Collapse
Affiliation(s)
- Woong Ji Lee
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826 Korea
| | - Sangdon Ryu
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826 Korea
| | - An Na Kang
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826 Korea
| | - Minho Song
- grid.254230.20000 0001 0722 6377Division of Animal and Dairy Science, Chungnam National University, Daejeon, 34134 Korea
| | - Minhye Shin
- grid.202119.90000 0001 2364 8385Department of Microbiology, College of Medicine, Inha University, Incheon, 22212 Korea
| | - Sangnam Oh
- grid.411845.d0000 0000 8598 5806Department of Functional Food and Biotechnology, Jeonju University, Jeonju, 55069 Korea
| | - Younghoon Kim
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826 Korea
| |
Collapse
|
39
|
Cheng YC, Kim SW. Use of Microorganisms as Nutritional and Functional Feedstuffs for Nursery Pigs and Broilers. Animals (Basel) 2022; 12:3141. [PMID: 36428369 PMCID: PMC9686830 DOI: 10.3390/ani12223141] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The objectives of this review paper are to introduce the structures and composition of various microorganisms, to show some applications of single cells as alternative protein supplements or energy feeds in swine and poultry diets, and to discuss the functional effects of microorganisms as feed additives on the growth performance and intestinal health of nursery pigs and broilers. Microorganisms, including bacteria, yeasts, and microalgae, have been commonly supplemented in animal diets because they are cost-effective, stable, and have quantitative production that provides nutritional and functional benefits to pigs and broilers. Microorganisms could be alternative antibiotics to enhance intestinal health due to bioactive components from cell wall components, which interact with receptors on epithelial and immune cells. In addition, bioactive components could be digested by intestinal microbiota to produce short-chain fatty acids and enhance energy utilization. Otherwise, microorganisms such as single-cell protein (SCP) and single-cell oils (SCOs) are sustainable and economic choices to replace conventional protein supplements and energy feeds. Supplementing microorganisms as feedstuffs and feed additives improved the average daily gain by 1.83%, the daily feed intake by 0.24%, and the feed efficiency by 1.46% in pigs and broilers. Based on the properties of each microorganism, traditional protein supplements, energy feeds, and functional feed additives could be replaced by microorganisms, which have shown benefits to animal's growth and health. Therefore, specific microorganisms could be promising alternatives as nutritional and functional feedstuffs in animal diets.
Collapse
Affiliation(s)
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
40
|
Garbacz K. Anticancer activity of lactic acid bacteria. Semin Cancer Biol 2022; 86:356-366. [PMID: 34995799 DOI: 10.1016/j.semcancer.2021.12.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023]
Abstract
Lactic acid bacteria (LAB), a group of Gram-positive microorganisms naturally occurring in fermented food products and used as probiotics, have been gaining the interest of researchers for years. LAB are potent, albeit still not wholly understood, source of bioactive compounds with various functions and activity. Metabolites of LAB, among others, short-chain fatty acids, exopolysaccharides and bacteriocins have promising anticancer potential. Research on the interactions between the bioactive metabolites of LAB and immune mechanisms demonstrated that these substances could exert a strong immunomodulatory effect, which would explain their vast therapeutic potential. The anticancer activity of LAB was confirmed both in vitro and in animal models against cancer cells from various malignancies. LAB inhibit tumor growth through various mechanisms, including antiproliferative activity, induction of apoptosis, cell cycle arrest, as well as through antimutagenic, antiangiogenic and anti-inflammatory effects. The aim of this review was to summarize the most recent data about the anticancer activity of LAB, with particular emphasis on the most promising bioactive compounds with potential clinical application.
Collapse
Affiliation(s)
- Katarzyna Garbacz
- Department of Oral Microbiology, Medical Faculty, Medical University of Gdansk, 25 Dębowa Str., 80-204, Gdansk, Poland.
| |
Collapse
|
41
|
Zheng J, Ahmad AA, Yang Y, Liang Z, Shen W, Feng M, Shen J, Lan X, Ding X. Lactobacillus rhamnosus CY12 Enhances Intestinal Barrier Function by Regulating Tight Junction Protein Expression, Oxidative Stress, and Inflammation Response in Lipopolysaccharide-Induced Caco-2 Cells. Int J Mol Sci 2022; 23:ijms231911162. [PMID: 36232464 PMCID: PMC9569798 DOI: 10.3390/ijms231911162] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
The intestinal barrier is vital for preventing inflammatory bowel disease (IBD). The objectives of this study were to assess whether the Lactobacillus rhamnosus CY12 could alleviate oxidative stress, inflammation, and the disruption of tight junction (TJ) barrier functions induced by lipopolysaccharide (LPS), and therefore to explore the potential underlying molecular mechanisms. Our results showed that LPS-induced Cancer coli-2 (Caco-2) cells significantly increased the levels of reactive oxygen species (ROS), lactate dehydrogenase, inflammatory cytokines interleukin-1β, interleukin-6, interleukin-8, and tumor necrosis factor-α (IL-1β, IL-6, IL-8, and TNF-α), and the cell apoptosis rate while decreasing the levels of TJ proteins occludin, zonula occludens-1 (ZO-1), and claudin and antioxidant enzymes, such as catalase, superoxide dismutase, and glutathione peroxidase(CAT, SOD, and GSH-Px) (p < 0.05). However, Lactobacillus rhamnosus CY12 could relieve cytotoxicity, apoptosis, oxidative stress, and pro-inflammatory cytokine expressions, and also inhibit the Toll-like receptor 4/nuclear factor kappa-B(TLR4/NF-κB) signaling pathway. Furthermore, the gene expression of antioxidant enzymes, as well as the mRNA and protein expressions of TJ proteins, was improved. Particularly, the concentration of 108 cfu/mL significantly prevented the inflammatory injury induced by LPS in Caco-2 cells (p < 0.05). These findings support a potential application of Lactobacillus rhamnosus CY12 as a probiotic to prevent LPS-induced intestinal injury and treat intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Juanshan Zheng
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Anum Ali Ahmad
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- State Key Laboratory of Grassland Agro-Ecosystems, School of Life Sciences, Lanzhou University, Lanzhou 730050, China
| | - Yayuan Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zeyi Liang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Min Feng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jiahao Shen
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xianyong Lan
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
- Correspondence: (X.L.); (X.D.); Tel.: +86-931-211-5255 (X.D.)
| | - Xuezhi Ding
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Correspondence: (X.L.); (X.D.); Tel.: +86-931-211-5255 (X.D.)
| |
Collapse
|
42
|
Kaur H, Ali SA, Yan F. Interactions between the gut microbiota-derived functional factors and intestinal epithelial cells - implication in the microbiota-host mutualism. Front Immunol 2022; 13:1006081. [PMID: 36159834 PMCID: PMC9492984 DOI: 10.3389/fimmu.2022.1006081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/23/2022] [Indexed: 12/13/2022] Open
Abstract
Mutual interactions between the gut microbiota and the host play essential roles in maintaining human health and providing a nutrient-rich environment for the gut microbial community. Intestinal epithelial cells (IECs) provide the frontline responses to the gut microbiota for maintaining intestinal homeostasis. Emerging evidence points to commensal bacterium-derived components as functional factors for the action of commensal bacteria, including protecting intestinal integrity and mitigating susceptibility of intestinal inflammation. Furthermore, IECs have been found to communicate with the gut commensal bacteria to shape the composition and function of the microbial community. This review will discuss the current understanding of the beneficial effects of functional factors secreted by commensal bacteria on IECs, with focus on soluble proteins, metabolites, and surface layer components, and highlight the impact of IECs on the commensal microbial profile. This knowledge provides a proof-of-concept model for understanding of mechanisms underlying the microbiota-host mutualism.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Syed Azmal Ali
- German Cancer Research Center, Division of Proteomics of Stem Cell and Cancer, Heidelberg, Germany
| | - Fang Yan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States,*Correspondence: Fang Yan,
| |
Collapse
|
43
|
Lu Y, Zhao M, Mo J, Lan G, Liang J. Dietary supplementation ellagic acid on the growth, intestinal immune response, microbiota, and inflammation in weaned piglets. Front Vet Sci 2022; 9:980271. [PMID: 36118358 PMCID: PMC9478910 DOI: 10.3389/fvets.2022.980271] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Piglets are susceptible to weaning stress, which weakens the barrier and immune function of the intestinal mucosa, causes inflammation, and ultimately affects animal growth and development. Ellagic acid (EA) is a natural polyphenol dilactone with various biological functions. However, The mechanisms underlying the effects of EA on animal health are still poorly known. Herein, we examined whether dietary supplementation with EA has a positive effect on growth performance, intestinal health, immune response, microbiota, or inflammation in weaned piglets. Sixty weaned piglets (age, 30 days) were randomly divided into two groups: the control group (basic diet) and the test group (basic diet + 500 g/t EA). The pigs were fed for 40 days under the same feeding and management conditions, and the growth performance of each individual was measured. At the end of the feeding period, samples were collected from the small intestinal mucosa for further analysis. Using these tissues, the transcriptome sequences and intestinal microbial diversity were analyzed in both groups. An inflammation model using small intestinal mucosal epithelial cells (IPEC-J2) was also constructed. Dietary EA supplementation significantly increased the average daily weight gain (ADG) and reduced diarrhea rate and serum diamine oxidase (DAO) levels of weaned piglets. Transcriptome sequencing results revealed 401 differentially expressed genes in the jejunum mucosal tissue of pigs in the control and test groups. Of these, 163 genes were up-regulated and 238 were down-regulated. The down-regulated genes were significantly enriched in 10 pathways (false discovery rate < 0.05), including seven pathways related to immune response. The results of bacterial 16s rDNA sequencing show that EA affects the composition of the intestinal microbiota in the cecum and rectum, and reveal significant differences in the abundances of Prevotella_9, Lactobacillus delbrueckii, and Lactobacillus reuteri between the test and control groups (P < 0.05). Experiments using the inflammation model showed that certain doses of EA promote the proliferation of IPEC-J2 cells, increase the relative mRNA expression levels of tight junction-related proteins (ZO-1 and Occludin), improve the compactness of the intestine, reduce the expression of inflammatory factors TNF-α and IL-6, and significantly reduce LPS-induced inflammation in IPEC-J2 cells. In conclusion, we found for the first time that dietary supplementation of EA affects the gut immune response and promotes the beneficial gut microbiota in weaned piglets, reduces the occurrence of inflammatory responses, and thereby promotes the growth and intestinal health of piglets.
Collapse
Affiliation(s)
- Yujie Lu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Mingwei Zhao
- Guangxi Guiken Jinmao Animal Husbandry Co., Ltd., Nanning, China
| | - Jiayuan Mo
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ganqiu Lan
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jing Liang
- College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Jing Liang
| |
Collapse
|
44
|
Gou HZ, Zhang YL, Ren LF, Li ZJ, Zhang L. How do intestinal probiotics restore the intestinal barrier? Front Microbiol 2022; 13:929346. [PMID: 35910620 PMCID: PMC9330398 DOI: 10.3389/fmicb.2022.929346] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 12/14/2022] Open
Abstract
The intestinal barrier is a structure that prevents harmful substances, such as bacteria and endotoxins, from penetrating the intestinal wall and entering human tissues, organs, and microcirculation. It can separate colonizing microbes from systemic tissues and prevent the invasion of pathogenic bacteria. Pathological conditions such as shock, trauma, stress, and inflammation damage the intestinal barrier to varying degrees, aggravating the primary disease. Intestinal probiotics are a type of active microorganisms beneficial to the health of the host and an essential element of human health. Reportedly, intestinal probiotics can affect the renewal of intestinal epithelial cells, and also make cell connections closer, increase the production of tight junction proteins and mucins, promote the development of the immune system, regulate the release of intestinal antimicrobial peptides, compete with pathogenic bacteria for nutrients and living space, and interact with the host and intestinal commensal flora to restore the intestinal barrier. In this review, we provide a comprehensive overview of how intestinal probiotics restore the intestinal barrier to provide new ideas for treating intestinal injury-related diseases.
Collapse
Affiliation(s)
- Hong-Zhong Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yu-Lin Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Long-Fei Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhen-Jiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Lei Zhang,
| |
Collapse
|
45
|
Yang B, Liu C, Huang Y, Wu Q, Xiong Y, Yang X, Hu S, Jiang Z, Wang L, Yi H. The Responses of Lactobacillus reuteri LR1 or Antibiotic on Intestinal Barrier Function and Microbiota in the Cecum of Pigs. Front Microbiol 2022; 13:877297. [PMID: 35722272 PMCID: PMC9201390 DOI: 10.3389/fmicb.2022.877297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/04/2022] [Indexed: 12/29/2022] Open
Abstract
This study aimed to investigate responses of the Lactobacillus reuteri or an antibiotic on cecal microbiota and intestinal barrier function in different stages of pigs. A total of 144 weaned pigs (Duroc × Landrace × Yorkshire, 21 days of age) were randomly assigned to the control group (CON, fed with a basal diet), the antibiotic group (AO, fed with basal diet plus 100 mg/kg olaquindox and 75 mg/kg aureomycin), and the L. reuteri group (LR, fed with the basal diet + 5 × 1010 CFU/kg L. reuteri LR1) throughout the 164-d experiment. A total of 45 cecal content samples (5 samples per group) from different periods (14th, 42th, and 164th days) were collected for 16S rRNA gene amplification. The results revealed that although LR and AO did not change the diversity of cecal microbiota in pigs, the abundance of some bacteria at the genus level was changed with age. The proportion of Lactobacillus was increased by LR in early life, whereas it was decreased by AO compared with the control group. The relative abundance of Ruminococcaceae was increased along with age. In addition, the gas chromatography results showed that age, not AO or LR, has significant effects on the concentrations of SCFAs in the cecum of pigs (P < 0.05). However, the mRNA expression of tight junction proteins zonula occluden-1 (ZO-1) and occludin were increased by AO in the cecum of pigs on day 14, while LR increased the mRNA expression of intestinal barrier-related proteins ZO-1, occludin, mucin-1, mucin-2, PG1-5, and pBD2 in the cecum of pigs on days 14 and 164 (P < 0.05). In conclusion, LR and AO have different effects on the intestinal barrier function of the cecum, and neither LR nor AO damaged the intestinal barrier function of pig cecum. In addition, LR and AO have little effects on cecal microflora in different stages of the pigs. The microflora and their metabolite SCFAs were significantly changed along with age. These findings provide important information to understand the homeostasis of the cecum of pigs after antibiotic or probiotic treatment.
Collapse
Affiliation(s)
- Bijing Yang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chunyan Liu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yanna Huang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Qiwen Wu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yunxia Xiong
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xuefen Yang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Shenglan Hu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zongyong Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hongbo Yi
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
46
|
Effects of different probiotic strains B. lactis, L. rhamnosus and L. reuteri on brain-intestinal axis immunomodulation in an endotoxin-induced inflammation. Mol Neurobiol 2022; 59:5168-5178. [PMID: 35674863 DOI: 10.1007/s12035-022-02906-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/29/2022] [Indexed: 10/18/2022]
Abstract
The study evaluated the effects of supplementation with three different probiotic strains Bifidobacterium lactis (LACT GB™), Lactobacillus rhamnosus (RHAM GB™) and Lactobacillus reuteri (REUT GB™) on brain-intestinal immunomodulation in an animal model of LPS-induced inflammation. Fifty mice Balb/C were distributed into five groups: control; lipopolysaccharide (LPS); LPS + B. lactis (LACT GB™); LPS + L. rhamnosus (RHAM GB™); and LPS + L. reuteri (REUT GB™). The animals were supplemented with their respective probiotic microorganisms daily, for 30 days, at a concentration of 1 × 109 CFU/animal/day. After 30 days of supplementation, animals received the inflammatory insult by LPS (15 mg/kg). Behavioral tests, oxidative stress and inflammation were performed, as well as gut and brain histology. In the behavioral test, LPS + B. lactis group was less anxious than the other groups. Serum interleukin IL-1β and IL-6 levels increased in all groups that received the LPS insult, and there was a reduction in inflammation in the supplemented groups when compared to the LPS group in brain and gut. There is a reduction in myeloperoxidase activity and oxidative stress in groups supplemented with probiotics. In intestine histological analysis occurs damage to the tissue integrity in the LPS group, in the other hand, occurs preservation of integrity in the probiotic supplemented animals. In the brain, infiltrates of perivascular inflammatory cells can be seen in the LPS group. The three probiotic studies showed efficient immunomodulating activity and ensured integrity of the intestinal barrier function, even after the severe insult by LPS. These results show the important role of probiotics in the gut-brain axis. Graphical abstract illustratively represents the gut-brain axis and how different probiotic strains influence the immunomodulatory response releasing different pro- and anti-inflammatory cytokines, and their role in the balance of dysbiosis.
Collapse
|
47
|
Inhibitory effects of reuterin on biofilm formation, quorum sensing and virulence genes of Clostridium perfringens. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Zhou W, Xie M, Xie Y, Liang H, Li M, Ran C, Zhou Z. Effect of dietary supplementation of Cetobacterium somerae XMX-1 fermentation product on gut and liver health and resistance against bacterial infection of the genetically improved farmed tilapia (GIFT, Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2022; 124:332-342. [PMID: 35430347 DOI: 10.1016/j.fsi.2022.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 06/14/2023]
Abstract
The purpose of this study was to evaluate the effects of Cetobacterium somerae XMX-1 fermentation product on gut and liver health and resistance against bacterial infection in genetically improved farmed tilapia (GIFT, Oreochromis niloticus). Fingerling GIFTs (n = 120; initial weight 1.33 ± 0.00 g) were randomly assigned to twelve 90-L tanks (four tanks per diet, 10 fish per tank) with three groups: control group (basal high fat diet), 1% XMX-1 group and 2% XMX-1 group (basal diet supplemented with 10 and 20 g XMX-1/kg feed respectively). After 49 days feeding trial, the growth performance and gut and liver health parameters of tilapia were evaluated. Also the gut microbiota and virome were detected by sequencing. 2% XMX-1 fermentation product had no effect on growth performance. For gut health, the expression of hypoxia-inducible factor-lα (Hif-1α) tend to increase in 1% XMX-1 group (P = 0.053). The expression of intestinal interleukin-6 (IL-6) and tumor growth factor β (TGF-β) was significantly down-regulated in 1% and 2% XMX-1 groups (P < 0.05), and the intestinal expression of interleukin-1β (IL-1β) had a trend to decrease (P = 0.08) in 1% XMX-1 group versus control. 1% and 2% XMX-1 groups also increased the intestinal expression of tight junction genes Claudin (P = 0.06 and 0.07, respectively). For liver health, XMX-1 fermentation product significantly decreased liver TAG (P < 0.05). Furthermore, the hepatic expression of lipid synthesis gene fatty acid synthase (FAS) was significantly decreased and the expression of lipid catabolism related-gene uncoupling protein 2 (UCP2) was significantly increased in 1% XMX-1 and 2% XMX-1 groups (P < 0.01). And the hepatic expression of IL-1β and IL-6 significantly decreased in 1% XMX-1 and 2% XMX-1 groups (P < 0.05). XMX-1 fermentation product increased the abundance of Fusobacteria in the gut microbiota and 2% XMX-1 group led to alteration in the virome composition at family level. Lastly, the time of tilapia death post Aeromoans challenge was delayed in 1% XMX-1 and 2% XMX-1 groups compared with control. To sum up, our results show that the dietary supplementation of XMX-1 fermentation product can improve the gut and liver health as well as the resistance against pathogenic bacteria of tilapia.
Collapse
Affiliation(s)
- Wei Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Mingxu Xie
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yadong Xie
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hui Liang
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Ming Li
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Zhigang Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
49
|
Effects of Bacillus licheniformis and Bacillus subtilis on Gut Barrier Function, Proinflammatory Response, ROS Production and Pathogen Inhibition Properties in IPEC-J2—Escherichia coli/Salmonella Typhimurium Co-Culture. Microorganisms 2022; 10:microorganisms10050936. [PMID: 35630380 PMCID: PMC9145911 DOI: 10.3390/microorganisms10050936] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The emergence of antimicrobial resistance raises serious concerns worldwide. Probiotics offer a promising alternative to enhance growth promotion in farm animals; however, their mode of action still needs to be elucidated. The IPEC-J2 cell line (porcine intestinal epithelial cells) is an appropriate tool to study the effect of probiotics on intestinal epithelial cells. In our experiments, IPEC-J2 cells were challenged by two gastrointestinal (GI) infection causing agents, Escherichia coli (E. coli) or Salmonella enterica ser. Typhimurium (S. Typhimurium). We focused on determining the effect of pre-, co-, and post-treatment with two probiotic candidates, Bacillus licheniformis or Bacillus subtilis, on the barrier function, proinflammatory cytokine (IL-6 and IL-8) response, and intracellular reactive oxygen species (ROS) production of IPEC-J2 cells, in addition to the adhesion inhibition effect. Bacillus licheniformis (B. licheniformis) and Bacillus subtilis (B. subtilis) proved to be anti-inflammatory and had an antioxidant effect under certain treatment combinations, and further effectively inhibited the adhesion of pathogenic bacteria. Interestingly, they had little effect on paracellular permeability. Based on our results, Bacillus licheniformis and Bacillus subtilis are both promising candidates to contribute to the beneficial effects of probiotic multispecies mixtures.
Collapse
|
50
|
Ram AK, Vairappan B. Role of zonula occludens in gastrointestinal and liver cancers. World J Clin Cases 2022; 10:3647-3661. [PMID: 35647143 PMCID: PMC9100728 DOI: 10.12998/wjcc.v10.i12.3647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/08/2021] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
A growing body of evidence suggests that tight junction (TJ) proteins play a crucial role in the pathogenesis of various diseases, including gastrointestinal (GI) cancer and hepatocellular carcinoma (HCC). TJ proteins primarily maintain the epithelial and endothelial cells intact together through integral proteins however, recent reports suggest that they also regulate gene expression necessary for cell proliferation, angiogenesis, and metastasis through adapter proteins such as zonula occludens (ZO). ZO proteins are membrane-associated cytosolic scaffolding proteins that modulate cell proliferation by interacting with several transcription factors. Reduced ZO proteins in GI cancer and HCC are correlated with tumor development and poor prognosis. Pubmed has searched for using the keyword ZO and gastric cancer, ZO and cancer, and ZO and HCC for the last ten years to date. This review summarized the role of ZO proteins in cell proliferation and their expression in GI cancer and HCC. Furthermore, therapeutic interventions targeting ZO in GI and liver cancers are reviewed.
Collapse
Affiliation(s)
- Amit Kumar Ram
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, India
| | - Balasubramaniyan Vairappan
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, India
| |
Collapse
|