1
|
Loupy KM, Dawud LM, Zambrano CA, Lee T, Heinze JD, Elsayed AI, Hassell JE, D'Angelo HM, Frank MG, Maier SF, Brenner LA, Lowry CA. Effects of Oral Administration of the Probiotic Lactobacillus rhamnosus GG on the Proteomic Profiles of Cerebrospinal Fluid and Immunoregulatory Signaling in the Hippocampus of Adult Male Rats. Neuroimmunomodulation 2025; 32:94-109. [PMID: 40031897 DOI: 10.1159/000544842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/08/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION The microbiome-gut-brain axis, by modulating bidirectional immune, metabolic, and neural signaling pathways in the host, has emerged as a target for the prevention and treatment of psychiatric and neurological disorders. Oral administration of the probiotic bacterium Lactobacillus rhamnosus GG (LGG; ATCC 53103) exhibits anti-inflammatory effects, although the precise mechanisms by which LGG benefits host physiology and behavior are not known. The goal of this study was to explore the general effects of LGG on the cerebrospinal fluid (CSF) proteome and a biological signature of anti-inflammatory signaling in the central nervous system (CNS) of undisturbed, adult male rats. METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were conducted using CSF samples collected after 21 days of oral treatment with live LGG (3.34 × 107 colony-forming units (CFU)/mL in the drinking water (resulting in an estimated delivery of ∼1.17 × 109 CFU/day/rat) or water vehicle. Gene enrichment analysis (using DAVID, v. 6.8) and protein-protein interactions (using STRING, v. 11) were used to explore physiological network changes in CSF. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR) was performed to assess gene expression changes of anti-inflammatory cytokines in the hippocampus. Genes associated with anti-inflammatory signaling that were analyzed included Il10, Tgfb1, Il4, and IL-4-responsive genes, Cd200, Cd200r1, and Mrc1 (Cd206). RESULTS Oral LGG administration altered the abundance of CSF proteins, increasing the abundance of five proteins (cochlin, NPTXR, reelin, Sez6l, and VPS13C) and decreasing the abundance of two proteins (CPQ, IGFBP-7) in the CSF. Simultaneously, LGG increased the expression of Il10 mRNA, encoding the anti-inflammatory cytokine interleukin 10, in the hippocampus. CONCLUSION Oral LGG altered the abundance of CSF proteins associated with extracellular scaffolding, synaptic plasticity, and glutamatergic signaling. These data are consistent with the hypothesis that oral administration of LGG improves memory and cognition, and promotes a physiological resilience to neurodegenerative disease, by increasing glutamatergic signaling and promoting an anti-inflammatory environment in the brain.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lamya'a M Dawud
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado, USA
| | - Jared D Heinze
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lisa A Brenner
- Rocky Mountain Regional VA Medical Center (RMRVAMC), Aurora, Colorado, USA
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, Colorado, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Rocky Mountain Regional VA Medical Center (RMRVAMC), Aurora, Colorado, USA
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, Colorado, USA
- Center for Microbial Exploration, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
2
|
Jang SB, Kim Y, Yeo HC, Kang GH, An BC, Ryu Y, Chung MJ, Cho SG. Probiotic-Derived P8 Protein: Promoting Proliferation and Migration in Stem Cells and Keratinocytes. Int J Stem Cells 2025; 18:87-98. [PMID: 39491493 PMCID: PMC11867908 DOI: 10.15283/ijsc24107] [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: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Probiotics exert various effects on the body and provide different health benefits. Previous reports have demonstrated that the P8 protein (P8), isolated from Lactobacillus rhamnosus, has anticancer properties. However, its efficacy in stem cells and normal cells has not been reported. In this study, the effect of P8 on cell proliferation and wound healing was evaluated, investigating its underlying mechanism. Based on scratch assay results, we demonstrated that P8 treatment significantly increases wound healing by activating the cell cycle and promoting stem cell stemness. Cellular mechanisms were further investigated by culturing stem cells in a medium containing Lactobacillus-derived P8 protein, revealing its promotion of cell proliferation and migration. Also, it is found that P8 enhances the expression of stemness markers, such as OCT4 and SOX2, along with activation of the mitogen-activated protein kinase (MAPK) signaling and Hippo pathways. These results indicate that P8 can promote cell growth by increasing stem cell proliferation, migration, and stemness in a manner associated with MAPK and Hippo signaling, which could contribute to the increased wound healing after P8 treatment. Furthermore, P8 could promote wound healing in keratinocytes by activating the MAPK signaling pathways. These results suggest that P8 might be a promising candidate to enhance stem cell culture efficiency by activating cell proliferation, and enhance therapeutic effects in skin diseases.
Collapse
Affiliation(s)
- Soo Bin Jang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Han Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | | | | | - Yongku Ryu
- R&D Center, Cell Biotech Co., Ltd., Gimpo, Korea
| | | | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| |
Collapse
|
3
|
Khan MN, Bashir S, Afzal A, Andleeb N, Krych L, Nielsen DS, Imran M. Probiotic characteristics and survival of a multi-strain lactic acid bacteria consortium in simulated gut model. Folia Microbiol (Praha) 2025:10.1007/s12223-025-01244-w. [PMID: 39924639 DOI: 10.1007/s12223-025-01244-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/15/2025] [Indexed: 02/11/2025]
Abstract
Dahi, a traditional yet underexplored fermented milk product from Pakistan, harbors diverse lactic acid bacteria (LAB) that have potential as probiotics. These bacteria could be used for therapeutic purposes, beneficial modulation of gut microbiota, and in the formulation of functional foods and feeds. This study aimed to isolate and characterize probiotic LAB from dahi, assess their survival in simulated gastrointestinal conditions, and evaluate their safety and probiotic potential, both phenotypically and genotypically. A total of 143 isolates from 37 samples were evaluated for probiotic traits, including acid and bile tolerance, antibacterial activity, cholesterol-lowering capacity, and antioxidant activity. The strains were also tested for antibiotic sensitivity and safety through in vitro tests and genomic analysis. A multi-strain probiotic consortium was developed and tested for enhanced functionality. Out of 143 isolates, 62 LAB strains were identified. These strains demonstrated significant survival under acidic (pH 2) and bile conditions. Antibacterial activity against pathogens ranged from 51 to 88%. The strains exhibited high cholesterol removal (up to 98%) and antioxidant activity (up to 76%). Genomic analysis revealed the presence of key probiotic-related genes, including those for acid resistance, bile salt hydrolase, and adhesion. All strains were sensitive to EFSA-recommended antibiotics and exhibited no hemolytic or DNase activity, confirming their safety. The multi-strain consortium showed superior probiotic potential and survival in simulated gastrointestinal conditions. LAB strains isolated from dahi possess strong probiotic potential, confirmed through in vitro and genomic safety assessments. The multi-strain consortium holds promise for applications.
Collapse
Affiliation(s)
- Muhammad Nadeem Khan
- Faculty of Biological Sciences, Department of Microbiology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
- Department of Cell Biology & Genetics, Shantou University Medical College, Shantou, 51504, China
- Department of Food Science, Section for Food Microbiology and Fermentation, University of Copenhagen, Copenhagen, Denmark
| | - Saeeda Bashir
- Faculty of Biological Sciences, Department of Microbiology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
| | - Afifa Afzal
- Faculty of Biological Sciences, Department of Microbiology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
| | - Naghmana Andleeb
- Faculty of Biological Sciences, Department of Microbiology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
| | - Lukasz Krych
- Department of Food Science, Section for Food Microbiology and Fermentation, University of Copenhagen, Copenhagen, Denmark
| | - Dennis Sandris Nielsen
- Department of Food Science, Section for Food Microbiology and Fermentation, University of Copenhagen, Copenhagen, Denmark
| | - Muhammad Imran
- Faculty of Biological Sciences, Department of Microbiology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan.
| |
Collapse
|
4
|
Karim A. Unveiling the Potential of Probiotics in Osteoarthritis Management. Curr Rheumatol Rep 2024; 27:2. [PMID: 39579259 DOI: 10.1007/s11926-024-01166-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA), a highly prevalent degenerative joint disease, is of increasing concern due to its debilitating nature and negative impact on quality of life. Recent investigations have explored the therapeutic potential of probiotics to alleviate OA. This review summarizes the emerging evidence for the potential role of probiotics in managing OA symptoms and disease progression. The link between gut dysbiosis and chronic inflammation, a key player in OA progression is discussed in this review. RECENT FINDINGS Probiotics may modulate gut microbiota composition, potentially reducing systemic inflammation and alleviating OA symptoms, including joint pain and function. Possible mechanisms through which probiotics may exert these effects, including dampening inflammatory pathways and enhancing intestinal barrier integrity have been highlighted. Promising results from preclinical and clinical studies investigating the specific beneficial effects of specific probiotic strain(s) for OA management have been highlighted. Finally, limitations in current research and future directions, emphasizing the need for well-designed, large-scale clinical trials to definitively establish the therapeutic potential of probiotics in OA treatment have been discussed.
Collapse
Affiliation(s)
- Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates.
- Iron Biology Research Group, Sharjah Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
5
|
Lakshmanan DK, Ravichandran G, Elangovan A, D AA, Thilagar S. Mechanisms and Intervention of Prebiotic Foods in Musculoskeletal Health. J Nutr 2024; 154:2628-2639. [PMID: 39004225 DOI: 10.1016/j.tjnut.2024.07.009] [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: 03/05/2024] [Revised: 06/25/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024] Open
Abstract
The review focuses primarily on collating and analyzing the mechanistic research data that discusses the function of prebiotics to halt the frailty of musculoskeletal system. Musculoskeletal diseases (MSDs) are frequently reported to co-occur within their own categories of conditions, such as osteoarthritis, rheumatoid arthritis, gouty arthritis, and psoriatic arthritis owing to their overlapping pathogenesis. Consequently, the same drugs are often used to manage the complications of most types. A few recent studies have addressed the therapeutic functions of gut microbes toward those commonly shared MSD pathway targets. Improving microbial diversity and enriching their population in the gut would promote the regeneration and recovery of the musculoskeletal system. Prebiotics are usually nondigestible substrates that are selectively used or digested by the gut microbes conferring health promotion. The microbial fermentation of prebiotics generates numerous host-beneficial therapeutic molecules. This study inspects the presumptive functions of plant-derived prebiotics for the growth and restoration of intestinal microbiota and the consequent improvement of skeletal health. The review also highlights the discrete functions of prebiotics against inflammation, autoimmunity, infection, physiologic overloading mechanism, and aging-associated loss of metabolism in MSD.
Collapse
Affiliation(s)
- Dinesh Kumar Lakshmanan
- Department of Biotechnology, Bannari Amman Institute of Technology, Sathyamangalam, Tamil Nadu 638402, India; Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Guna Ravichandran
- Centre for Biomedical and Molecular Biology Research, Vinayaka Mission's Medical College & Hospital (VMMCH), Vinayaka Mission's Research Foundation (VMRF), Karaikal, India
| | - Abbirami Elangovan
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Arul Ananth D
- Department of Biotechnology, The American College Madurai, Tamil Nadu, India
| | - Sivasudha Thilagar
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India.
| |
Collapse
|
6
|
Wang P, Wang S, Wang D, Li Y, Yip RCS, Chen H. Postbiotics-peptidoglycan, lipoteichoic acid, exopolysaccharides, surface layer protein and pili proteins-Structure, activity in wounds and their delivery systems. Int J Biol Macromol 2024; 274:133195. [PMID: 38885869 DOI: 10.1016/j.ijbiomac.2024.133195] [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: 03/20/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
Chronic wound healing is a pressing global public health concern. Abuse and drug resistance of antibiotics are the key problems in the treatment of chronic wounds at present. Postbiotics are a novel promising strategy. Previous studies have reported that postbiotics have a wide range of biological activities including antimicrobial, immunomodulatory, antioxidant and anti-inflammatory abilities. However, several aspects related to these postbiotic activities remain unexplored or poorly known. Therefore, this work aims to outline general aspects and emerging trends in the use of postbiotics for wound healing, such as the production, characterization, biological activities and delivery strategies of postbiotics. In this review, a comprehensive overview of the physiological activities and structures of postbiotic biomolecules that contribute to wound healing is provided, such as peptidoglycan, lipoteichoic acid, bacteriocins, exopolysaccharides, surface layer proteins, pili proteins, and secretory proteins (p40 and p75 proteins). Considering the presence of readily degradable components in postbiotics, potential natural polymer delivery materials and delivery systems are emphasized, followed by the potential applications and commercialization prospects of postbiotics. These findings suggest that the treatment of chronic wounds with postbiotic ingredients will help provide new insights into wound healing and better guidance for the development of postbiotic products.
Collapse
Affiliation(s)
- Pu Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Shuxin Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Donghui Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Yuanyuan Li
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Stocking Hall, 411 Tower Road, Ithaca, NY 14853, USA.
| | - Ryan Chak Sang Yip
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord St, Toronto, ON M5S 3G5, Canada.
| | - Hao Chen
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| |
Collapse
|
7
|
Saito S, Cao DY, Maekawa T, Tsuji NM, Okuno A. Lactococcus lactis subsp. cremoris C60 Upregulates Macrophage Function by Modifying Metabolic Preference in Enhanced Anti-Tumor Immunity. Cancers (Basel) 2024; 16:1928. [PMID: 38792006 PMCID: PMC11120145 DOI: 10.3390/cancers16101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Lactococcus lactis subsp. cremoris C60 is a probiotic strain of lactic acid bacteria (LAB) which induces various immune modifications in myeloid lineage cells. These modifications subsequently regulate T cell function, resulting in enhanced immunity both locally and systemically. Here, we report that C60 suppresses tumor growth by enhancing macrophage function via metabolic alterations, thereby increasing adenosine triphosphate (ATP) production in a murine melanoma model. Intragastric (i.g.) administration of C60 significantly reduced tumor volume compared to saline administration in mice. The anti-tumor function of intratumor (IT) macrophage was upregulated in mice administered with C60, as evidenced by an increased inflammatory phenotype (M1) rather than an anti-inflammatory/reparative (M2) phenotype, along with enhanced antigen-presenting ability, resulting in increased tumor antigen-specific CD8+ T cells. Through this functional modification, we identified that C60 establishes a glycolysis-dominant metabolism, rather than fatty acid oxidation (FAO), in IT macrophages, leading to increased intracellular ATP levels. To address the question of why orally supplemented C60 exhibits functions in distal places, we found a possibility that bacterial cell wall components, which could be distributed throughout the body from the gut, may induce stimulatory signals in peripheral macrophages via Toll-like receptors (TLRs) signaling activation. Thus, C60 strengthens macrophage anti-tumor immunity by promoting a predominant metabolic shift towards glycolysis upon TLR-mediated stimulation, thereby increasing substantial energy production.
Collapse
Affiliation(s)
- Suguru Saito
- Department of Infection and Immunity, Division of Virology, Faculty of Medicine, Jichi Medical University, Shimotsuke, Tochigi 3290431, Japan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Toshio Maekawa
- iFoodMed Inc., Tsuchiura, Ibaraki 3000873, Japan;
- Department of Pathology and Microbiology, Division of Immune Homeostasis, Nihon University School of Medicine, Itabashi, Tokyo 1738610, Japan;
| | - Noriko M. Tsuji
- Department of Pathology and Microbiology, Division of Immune Homeostasis, Nihon University School of Medicine, Itabashi, Tokyo 1738610, Japan;
- Department of Pathology and Microbiology, Division of Microbiology, Nihon University School of Medicine, Itabashi, Tokyo 1738610, Japan
- Department of Food Science, Jumonji University, Niiza, Saitama 3528510, Japan
| | - Alato Okuno
- Department of Health and Nutrition, Faculty of Human Design, Shibata Gakuen University, Hirosaki, Aomori 0368530, Japan
| |
Collapse
|
8
|
Cappellucci G, Baini G, Miraldi E, Pauletto L, De Togni H, Raso F, Biagi M. Investigation on the Efficacy of Two Food Supplements Containing a Fixed Combination of Selected Probiotics and β-Glucans or Elderberry Extract for the Immune System: Modulation on Cytokines Expression in Human THP-1 and PBMC. Foods 2024; 13:458. [PMID: 38338593 PMCID: PMC10855234 DOI: 10.3390/foods13030458] [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: 12/29/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Several herbal and other natural products are used as ingredients in food supplements to strengthen immunity even if, very often, marketed products are proposed without a clear rationale or experimental evidence. In this study, we aimed to investigate the effect on human monocytes (THP-1) and on ex vivo human peripheral blood mononuclear cells (PBMC) of two formulations, one containing Bifidobacterium animalis subsp. lactis Bl-04® with β-glucans (for adults) and one containing Lactobacillus rhamnosus CRL1505 with elderberry extract (for children). We compared formulations with single ingredients, with bacterial lipopolysaccharide (LPS) and the drug pidotimod; cytokines expression level was evaluated testing different concentrations of samples at two exposure times. As expected, LPS caused a non-specific huge upregulation of cytokines expression both in THP-1 and in PBMC, whereas pidotimod mainly upregulated IL-2 in PBMC and IL-8 in THP-1. The two formulations showed a difference between a pro-inflammatory stimulus such as LPS, and also from an immunostimulant drug, such as pidotimod, as they mainly upregulated the expression of IL-6 and IL-10 in PBMC but not in THP-1, in a concentration-dependent mode. Probiotics were shown to play a major role, but β-glucans and elderberry extract exerted a synergistic activity. This work demonstrated that combining selected probiotics with other natural products having immunomodulatory properties is an interesting strategy to develop innovative formulations in the sector of food supplements.
Collapse
Affiliation(s)
- Giorgio Cappellucci
- Department of Physics, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.C.); (G.B.); (E.M.)
| | - Giulia Baini
- Department of Physics, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.C.); (G.B.); (E.M.)
| | - Elisabetta Miraldi
- Department of Physics, Earth and Environmental Sciences, University of Siena, 53100 Siena, Italy; (G.C.); (G.B.); (E.M.)
| | - Lara Pauletto
- Scientific Affairs Department Schwabe Pharma Italia, 39044 Egna, Italy; (L.P.); (H.D.T.); (F.R.)
| | - Heide De Togni
- Scientific Affairs Department Schwabe Pharma Italia, 39044 Egna, Italy; (L.P.); (H.D.T.); (F.R.)
| | - Floriana Raso
- Scientific Affairs Department Schwabe Pharma Italia, 39044 Egna, Italy; (L.P.); (H.D.T.); (F.R.)
| | - Marco Biagi
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
9
|
Magryś A, Pawlik M. Postbiotic Fractions of Probiotics Lactobacillus plantarum 299v and Lactobacillus rhamnosus GG Show Immune-Modulating Effects. Cells 2023; 12:2538. [PMID: 37947616 PMCID: PMC10648844 DOI: 10.3390/cells12212538] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Probiotic bacteria belonging to Lactobacillus spp. are important producers of bioactive molecules, known as postbiotics, that play essential roles in the immunological support of the intestinal mucosa. In this study, the system of co-culture of intestinal epithelial cells with macrophage cells in vitro was used to study the potential effect of postbiotic fractions of L. rhamonosus and L. plantarum on the modulation of the immune response induced by pro-inflammatory stimuli. This study's results revealed that the presence of probiotic bacterial components on the mucosal surface in the early and late stage of inflammatory conditions is based on cellular interactions that control inflammation and consequent damage to the intestinal epithelium. In our studies, heat killed fractions of probiotic bacteria and their extracted proteins showed a beneficial effect on controlling inflammation, regardless of the strain tested, consequently protecting intestinal barrier damage. In conclusion, the presented results emphasize that the fractions of probiotic bacteria of L. plantarum and L. rhamnosus may play a significant role in the regulation of LPS-mediated cytotoxic activity in intestinal epithelial cells. The fractions of probiotic strains of L. rhamnosus and L. plantarum showed the potential to suppress inflammation, effectively activating the anti-inflammatory cytokine IL-10 and modulating the IL-18-related response.
Collapse
Affiliation(s)
- Agnieszka Magryś
- Chair and Department of Medical Microbiology, Medical University of Lublin, ul. Chodźki 1, 20-093 Lublin, Poland
| | | |
Collapse
|
10
|
Kwak MJ, Kim SH, Kim HH, Tanpure R, Kim JI, Jeon BH, Park HK. Psychobiotics and fecal microbial transplantation for autism and attention-deficit/hyperactivity disorder: microbiome modulation and therapeutic mechanisms. Front Cell Infect Microbiol 2023; 13:1238005. [PMID: 37554355 PMCID: PMC10405178 DOI: 10.3389/fcimb.2023.1238005] [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: 06/10/2023] [Accepted: 06/30/2023] [Indexed: 08/10/2023] Open
Abstract
Dysbiosis of the gut microbiome is thought to be the developmental origins of the host's health and disease through the microbiota-gut-brain (MGB) axis: such as immune-mediated, metabolic, neurodegenerative, and neurodevelopmental diseases. Autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD) are common neurodevelopmental disorders, and growing evidence indicates the contribution of the gut microbiome changes and imbalances to these conditions, pointing to the importance of considering the MGB axis in their treatment. This review summarizes the general knowledge of gut microbial colonization and development in early life and its role in the pathogenesis of ASD/ADHD, highlighting a promising therapeutic approach for ASD/ADHD through modulation of the gut microbiome using psychobiotics (probiotics that positively affect neurological function and can be applied for the treatment of psychiatric diseases) and fecal microbial transplantation (FMT).
Collapse
Affiliation(s)
- Min-jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Hoo Hugo Kim
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Rahul Tanpure
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Johanna Inhyang Kim
- Department of Psychiatry, Hanyang University Medical Center, Seoul, Republic of Korea
- Clinical Research Institute of Developmental Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Clinical Research Institute of Developmental Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
11
|
Loganathan T, Priya Doss C G. The influence of machine learning technologies in gut microbiome research and cancer studies - A review. Life Sci 2022; 311:121118. [DOI: 10.1016/j.lfs.2022.121118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
|
12
|
Kang SJ, Jun JS, Hong KW. Transcriptome Analysis Reveals Immunomodulatory Effect of Spore-Displayed p75 on Human Intestinal Epithelial Caco-2 Cells. Int J Mol Sci 2022; 23:ijms232314519. [PMID: 36498846 PMCID: PMC9739243 DOI: 10.3390/ijms232314519] [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: 10/13/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Lacticaseibacillus rhamnosus GG (LGG) can promote intestinal health by modulating the immune responses of the gastrointestinal tract. However, knowledge about the immunomodulatory action of LGG-derived soluble factors is limited. In our previous study, we have displayed LGG-derived p75 protein on the spore surface of Bacillus subtilis. The objective of this study was to determine the effect of spore-displayed p75 (CotG-p75) on immune system by investigating transcriptional response of Caco-2 cells stimulated by CotG-p75 through RNA-sequencing (RNA-seq). RNA-seq results showed that CotG-p75 mainly stimulated genes involved in biological processes, such as response to stimulus, immune regulation, and chemotaxis. KEGG pathway analysis suggested that many genes activated by CotG-p75 were involved in NF-ĸB signaling and chemokine signaling pathways. CotG-p75 increased cytokines and chemokines such as CXCL1, CXCL2, CXCL3, CXCL8, CXCL10, CCL20, CCL22, and IL1B essential for the immune system. In particular, CotG-p75 increased the expression levels of NF-ĸB-related genes such as NFKBIA, TNFAIP3, BIRC3, NFKB2, and RELB involved in immune and inflammatory responses. This study provides genes and pathways involved in immune responses influenced by CotG-p75. These comprehensive transcriptome profiling could be used to elucidate the immunomodulatory action of CotG-p75.
Collapse
|
13
|
Quazi S. Anti-cancer activity of human gastrointestinal bacteria. Med Oncol 2022; 39:220. [PMID: 36175586 DOI: 10.1007/s12032-022-01771-3] [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: 02/13/2022] [Accepted: 06/14/2022] [Indexed: 06/16/2023]
Abstract
Malignant neoplasm is one of the most incurable diseases among inflammatory diseases. Researchers have been studying for decades to win over this lethal disease and provide the light of hope to humankind. The gastrointestinal bacteria of human hold a complex ecosystem and maintain homeostasis. One hundred trillion microbes are residing in the gastrointestinal tract of human. Disturbances in the microbiota of human's gastrointestinal tract can create immune response against inflammation and also can develop diseases, including cancer. The bacteria of the gastrointestinal tract of human can secrete a variety of metabolites and bioproducts which aid in the preservation of homeostasis in the host and gut. During pathogenic dysbiosis, on the other hand, numerous microbiota subpopulations may increase and create excessive levels of toxins, which can cause inflammation and cancer. Furthermore, the immune system of host and the epithelium cell can be influenced by gut microbiota. Probiotics, which are bacteria that live in the gut, have been protected against tumor formation. Probiotics are now studied to see if they can help fight dysbiosis in cancer patients undergoing chemotherapy or radiotherapy because of their capacity to maintain gut homeostasis. Countless numbers of gut bacteria have demonstrated anti-cancer efficiency in cancer treatment, prevention, and boosting the efficiency of immunotherapy. The review article has briefly explained the anti-cancer immunity of gut microbes and their application in treating a variety of cancer. This review paper also highlights the pre-clinical studies of probiotics against cancer and the completed and ongoing clinical trials on cancers with the two most common and highly effective probiotics Lactobacillus and Bacillus spp.
Collapse
Affiliation(s)
- Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore, 560043, Karnataka, India.
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, UK.
| |
Collapse
|
14
|
Li Z, Zhou Q, Qingsong Q, Liao Y, Yang F, Sheng M, Feng L, Shi X. Effect of Maifan Stone on the Growth of Probiotics and Regulation of Gut Microbiota. Lett Appl Microbiol 2022; 75:1423-1432. [PMID: 35975465 DOI: 10.1111/lam.13809] [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: 03/21/2022] [Revised: 08/02/2022] [Accepted: 08/07/2022] [Indexed: 11/29/2022]
Abstract
Maifan stone is a kind of mineral medicine in Chinese medicine, which has good adsorption, dissolution, mineralization, and biological activity. It has an excellent therapeutic effect on livestock, poultry, and aquatic animals suffering from intestinal diseases. This study explored the effect of Maifan stone on the growth ability of Lacticaseibacillus rhamnosus GG (L. rhamnosus GG) and the effect of Maifan stone-L. rhamnosus GG fermented product on the intestinal inflammation and gut microbiota. We find that Maifan stone can adsorb L. rhamnosus GG to form a carrier bacteria. Maifan stone has the characteristics of acid tolerance and bile salt tolerance and can also improve the activity of L. rhamnosus GG in artificial gastrointestinal juice. The fermented product can reduce the degree of diarrhea and colon pathology in rats to a certain extent and significantly improve intestinal inflammatory factors and gut microbiota. This study improves the application effect of L. rhamnosus GG in the prevention and treatment of diarrhea animals and provide a scientific basis for the rational development of Maifan stone resources.
Collapse
Affiliation(s)
- ZhiXun Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qing Zhou
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qu Qingsong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yuyao Liao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Fang Yang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mengke Sheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Lei Feng
- Inner Mongolia Yougaoya Health Technology Co., Ltd., Inner Mongolia, 028399, China
| | - Xinyuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.,Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing, 100029, China
| |
Collapse
|
15
|
Colonization of Lactobacillus rhamnosus GG in Cirrhinus molitorella (Mud Carp) Fingerling: Evidence for Improving Disease Resistance and Growth Performance. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of probiotic bacteria can not only enhance the nutritional utilization of fish feeds to produce more biomass but can also provide a practically “safer” alternative to the fish farming industry to reduce the abuse of antibiotics and drugs. This study investigated the possibility of colonizing Lactobacillus rhamnosus strain GG (LGG) to the intestine of Cirrhinus molitorella (mud carp) fingerling. Colonization of LGG was observed in gut tissue after 14 days of administration with a diet supplemented with 1 × 108 CFU/mL LGG. Moreover, growth performance parameters of the LGG-supplemented diet group, including relative weight gain, feed conversion ratio and feed efficiency, were found about two-fold higher than the control group after 60 days. In addition, fish fed with an LGG-supplemented diet for 60 days showed substantial resistance against the infection of pathogenic bacterial Aeromonas hydrophila, with a relative survival rate of up to 57% compared to the control group. In summary, the results indicated that LGG as dietary supplement for mud carp fingerling can enhance nutrition utilization and better protect fish against the infection of Aeromonas hydrophila. The results provide an insight to the fish farming industry, encouraging a reduction in the use of antibiotics and drugs and the production of “safer” mud carp for the market at a manageable cost.
Collapse
|
16
|
Stanojević S, Blagojević V, Ćuruvija I, Vujić V. Lactobacillus rhamnosus Affects Rat Peritoneal Cavity Cell Response to Stimulation with Gut Microbiota: Focus on the Host Innate Immunity. Inflammation 2021; 44:2429-2447. [PMID: 34505975 DOI: 10.1007/s10753-021-01513-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022]
Abstract
Gut microbiota contribute to shaping the immune repertoire of the host, whereas probiotics may exert beneficial effects by modulating immune responses. Having in mind the differences in both the composition of gut microbiota and the immune response between rats of Albino Oxford (AO) and Dark Agouti (DA) rat strains, we investigated if intraperitoneal (i.p.) injection of live Lactobacillus rhamnosus (LB) may influence peritoneal cavity cell response to in vitro treatments with selected microbiota in the rat strain-dependent manner. Peritoneal cavity cells from AO and DA rats were lavaged two (d2) and seven days (d7) following i.p. injection with LB and tested for NO, urea, and H2O2 release basally, or upon in vitro stimulation with autologous E.coli and Enterococcus spp. Whereas the single i.p. injection of LB nearly depleted resident macrophages and increased the proportion of small inflammatory macrophages and monocytes on d2 in both rat strains, greater proportion of MHCIIhiCD163- and CCR7+ cells and increased NO/diminished H2O2 release in DA compared with AO rats suggest a more intense inflammatory priming by LB in this rat strain. Even though E.coli- and/or Enterococcus spp.-induced rise in H2O2 release in vitro was abrogated by LB in cells from both rat strains, LB prevented microbiota-induced increase in NO/urea ratio only in cells from AO and augmented it in cells from DA rats. Thus, the immunomodulatory properties may not be constant for particular probiotic bacteria, but shaped by innate immunity of the host.
Collapse
Affiliation(s)
- Stanislava Stanojević
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia. .,Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia.
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia
| | - Ivana Ćuruvija
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia
| | - Vesna Vujić
- Department of Chemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Probiotic-Induced Tolerogenic Dendritic Cells: A Novel Therapy for Inflammatory Bowel Disease? Int J Mol Sci 2021; 22:ijms22158274. [PMID: 34361038 PMCID: PMC8348973 DOI: 10.3390/ijms22158274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are immune-mediated, chronic relapsing diseases with a rising prevalence worldwide in both adult and pediatric populations. Treatment options for immune-mediated diseases, including IBDs, are traditional steroids, immunomodulators, and biologics, none of which are capable of inducing long-lasting remission in all patients. Dendritic cells (DCs) play a fundamental role in inducing tolerance and regulating T cells and their tolerogenic functions. Hence, modulation of intestinal mucosal immunity by DCs could provide a novel, additional tool for the treatment of IBD. Recent evidence indicates that probiotic bacteria might impact immunomodulation both in vitro and in vivo by regulating DCs’ maturation and producing tolerogenic DCs (tolDCs) which, in turn, might dampen inflammation. In this review, we will discuss this evidence and the mechanisms of action of probiotics and their metabolites in inducing tolDCs in IBDs and some conditions associated with them.
Collapse
|
18
|
The Conditioned Medium of Lactobacillus rhamnoides GG Regulates Microglia/Macrophage Polarization and Improves Functional Recovery after Spinal Cord Injury in Rats. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3376496. [PMID: 34337004 PMCID: PMC8289592 DOI: 10.1155/2021/3376496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/25/2021] [Indexed: 11/18/2022]
Abstract
Lactobacillus rhamnoides, a human intestinal colonizer, can act through various pathways to induce microglia/macrophages to produce cytokines and to polarize microglia/macrophages to different phenotypes to reduce the inflammatory response. In this article, we evaluated the treatment potential of the Lactobacillus rhamnoides GG conditioned medium (LGG-CM) in rat model with SCI (acute spinal cord injury), including functional, neurophysiological, and histological outcomes and the underlying neuroprotective mechanisms. In our experiment, LGG-CM (30 mg/kg) was injected directly into the injury site in rats immediately after SCI. Measured by the BBB scale (Basso, Beattie, and Bresnahan locomotor rating scale) and inclined plane test, rats in the LGG-CM-treated group showed better locomotor scores. Moreover, compared to the vehicle treatment group, LGG-CM increased the mRNA level of the M2 marker (CD206), and decreased that of the M1 marker (iNOS). Western blot assays showed that LGG-CM-treated SCI rats had a higher grayscale ratio of p65 and a lower ratio of p-IκBα/IκBα. Our study shows that local injection of LGG-CM after acute SCI can inhibit inflammatory responses and improve motor function recovery. These effects may be related with the inhibition to the NF-κB (The nuclear factor-kappa B) signal pathway which leads to M2 microglia/macrophage polarization.
Collapse
|
19
|
Oral Probiotic Vaccine Expressing Koi Herpesvirus (KHV) ORF81 Protein Delivered by Chitosan-Alginate Capsules Is a Promising Strategy for Mass Oral Vaccination of Carps against KHV Infection. J Virol 2021; 95:JVI.00415-21. [PMID: 33827944 DOI: 10.1128/jvi.00415-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023] Open
Abstract
Koi herpesvirus (KHV) is highly contagious and lethal to cyprinid fish, causing significant economic losses to the carp aquaculture industry, particularly to koi carp breeders. Vaccines delivered through intramuscular needle injection or gene gun are not suitable for mass vaccination of carp. So, the development of cost-effective oral vaccines that are easily applicable at a farm level is highly desirable. In this study, we utilized chitosan-alginate capsules as an oral delivery system for a live probiotic (Lactobacillus rhamnosus) vaccine, pYG-KHV-ORF81/LR CIQ249, expressing KHV ORF81 protein. The tolerance of the encapsulated recombinant Lactobacillus to various digestive environments and the ability of the probiotic strain to colonize the intestine of carp was tested. The immunogenicity and the protective efficacy of the encapsulated probiotic vaccine was evaluated by determining IgM levels, lymphocyte proliferation, expression of immune-related genes, and viral challenge to vaccinated fish. It was clear that the chitosan-alginate capsules protected the probiotic vaccine effectively against extreme digestive environments, and a significant level (P < 0.01) of antigen-specific IgM with KHV-neutralizing activity was detected, which provided a protection rate of ca. 85% for koi carp against KHV challenge. The strategy of using chitosan-alginate capsules to deliver probiotic vaccines is easily applicable for mass oral vaccination of fish.IMPORTANCE An oral probiotic vaccine, pYG-KHV-ORF81/LR CIQ249, encapsulated by chitosan-alginate capsules as an oral delivery system was developed for koi carp against koi herpesvirus (KHV) infection. This encapsulated probiotic vaccine can be protected from various digestive environments and maintain effectively high viability, showing a good tolerance to digestive environments. This encapsulated probiotic vaccine has a good immunogenicity in koi carp via oral vaccination, and a significant level of antigen-specific IgM was effectively induced after oral vaccination, displaying effective KHV-neutralizing activity. This encapsulated probiotic vaccine can provide effective protection for koi carp against KHV challenge, which is handling-stress free for the fish, cost effective, and suitable for the mass oral vaccination of koi carp at a farm level, suggesting a promising vaccine strategy for fish.
Collapse
|
20
|
Jang HJ, Son S, Kim JA, Jung MY, Choi YJ, Kim DH, Lee HK, Shin D, Kim Y. Characterization and Functional Test of Canine Probiotics. Front Microbiol 2021; 12:625562. [PMID: 33763044 PMCID: PMC7982664 DOI: 10.3389/fmicb.2021.625562] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/17/2021] [Indexed: 02/01/2023] Open
Abstract
Probiotics can modulate the composition of gut microbiota and benefit the host animal health in multiple ways. Lactic acid bacteria (LAB), mainly Lactobacillus and Bifidobacterium species, are well-known microbes with probiotic potential. In the present study, 88 microbial strains were isolated from canine feces and annotated. Among these, the four strains CACC517, 537, 558, and 566 were tested for probiotic characteristics, and their beneficial effects on hosts were evaluated both in vitro and in vivo; these strains exhibited antibiosis, antibiotic activity, acid and bile tolerance, and relative cell adhesion to the HT-29 monolayer cell line. Byproducts of these strains increased the viability and decreased oxidative stress in mouse and dog cell lines (RAW264.7 and DH82, respectively). Subsequently, when the probiotics were applied to the clinical trial, changes in microbial composition and relative abundance of bacterial strains were clearly observed in the experimental animals. Experimental groups before and after the application were obviously separated from PCA analysis of clinical results. Conclusively, these results could provide comprehensive understanding of the effects of probiotic strains (CACC517, 537, 558, and 566) and their industrial applications.
Collapse
Affiliation(s)
- Hyun-Jun Jang
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| | - Seungwoo Son
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju-si, South Korea.,The Animal Molecular Genetics & Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| | - Jung-Ae Kim
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea.,Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju-si, South Korea
| | - Min Young Jung
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| | - Yeon-Jae Choi
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| | - Dae-Hyuk Kim
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea.,Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju-si, South Korea.,Department of Molecular Biology, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju-si, South Korea
| | - Hak Kyo Lee
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju-si, South Korea.,The Animal Molecular Genetics & Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| | - Donghyun Shin
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju-si, South Korea.,The Animal Molecular Genetics & Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| | - Yangseon Kim
- Department of Research and Development, Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, South Korea
| |
Collapse
|
21
|
Celebioglu HU. Effects of potential synbiotic interaction between Lactobacillus rhamnosus GG and salicylic acid on human colon and prostate cancer cells. Arch Microbiol 2021; 203:1221-1229. [PMID: 33620523 DOI: 10.1007/s00203-021-02200-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/03/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022]
Abstract
Salicylic acid, widely distributed in the whole plant kingdom, is a benzoic acid derivative acting as a signal substance in plants, but could be related to differences in cancer incidence, as many herbs and spices contain high amounts. Lactobacillus rhamnosus GG (LGG) is one of the best-known lactic acid bacteria that has been studied for over 30 years. Probiotic and/or commensal bacteria of the human microbiota are known to respond to diet constituents. Therefore, the present study aims at investigating the possible effects of salicylic acid on the probiotic properties of LGG, and in vitro cytotoxic effects of combination of salicylic acid and LGG on human colon and prostate cancer cells. Salicylic acid significantly (p < 0.05) increased co-aggregation of LGG with E. coli (~ twofold) and anti-oxidant properties. Furthermore, it also induced the cytotoxic effects of LGG against human colon cancer cells. These results suggest that interaction of LGG with salicylic acid can exert more probiotic properties.
Collapse
|
22
|
Lactobacillus rhamnosus GG soluble mediators ameliorate early life stress-induced visceral hypersensitivity and changes in spinal cord gene expression. Neuronal Signal 2020; 4:NS20200007. [PMID: 33343931 PMCID: PMC7726314 DOI: 10.1042/ns20200007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Visceral hypersensitivity is a hallmark of many functional and stress-related gastrointestinal disorders, and there is growing evidence that the gut microbiota may play a role in its pathophysiology. It has previously been shown that early life stress-induced visceral sensitivity is reduced by various probiotic strains of bacteria (including Lactobacillus rhamnosus GG (LGG)) alone or in combination with prebiotic fibres in rat models. However, the exact mechanisms underpinning such effects remain unresolved. Here, we investigated if soluble mediators derived from LGG can mimic the bacteria's effects on visceral hypersensitivity and the microbiota-gut-brain axis. Rats were exposed to maternal separation (MS) from postnatal days 2-12. From weaning onwards both non-separated (NS) and MS offspring were provided drinking water with or without supplementation of standardized preparations of the LGG soluble mediators (LSM). Our results show that MS led to increased visceral sensitivity and exaggerated corticosterone plasma levels following restraint stress in adulthood, and both of these effects were ameliorated through LSM supplementation. Differential regulation of various genes in the spinal cord of MS versus NS rats was observed, 41 of which were reversed by LSM supplementation. At the microbiota composition level MS led to changes in beta diversity and abundance of specific bacteria including parabacteroides, which were ameliorated by LSM. These findings support probiotic soluble mediators as potential interventions in the reduction of symptoms of visceral hypersensitivity.
Collapse
|
23
|
Jeong† M, Kim† JH, Lee JS, Kang SD, Shim S, Jung MY, Yang H, Byun S, Lee KW. Heat-Killed Lactobacillus brevis Enhances Phagocytic Activity and Generates Immune-Stimulatory Effects through Activating the TAK1 Pathway. J Microbiol Biotechnol 2020; 30:1395-1403. [PMID: 32627755 PMCID: PMC9728231 DOI: 10.4014/jmb.2002.02004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/26/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022]
Abstract
There is an increasing interest in using inactivated probiotics to modulate the host immune system and protect against pathogens. As the immunomodulatory function of heat-killed Lactobacillus brevis KCTC 12777BP (LBB) and its mechanism is unclear, we investigated the effect of LBB on immune response based on the hypothesis that LBB might exert stimulatory effects on immunity. In the current study, we demonstrate that administration of LBB can exert immune-stimulatory effects and promote clearance of foreign matters through enhancing phagocytosis. Treatment with LBB induced the production of TNF-α, IL-6, and nitric oxide in macrophages. Importantly, LBB directly increased the phagocytic activity of macrophages against bacterial particles. LBB was able to promote the production of TNF-α in bone marrow-derived macrophages and splenocytes and also increase the proliferation rate of splenocytes, suggesting that the immune-stimulating activity of LBB can be observed in primary immune cells. Investigation into the molecular mechanism responsible revealed that LBB upregulates TAK1 activity and its downstream ERK, p38, and JNK signaling pathways. To further confirm the immunomodulatory capability of LBB in vivo, we orally administered LBB to mice and assessed the effect on primary splenocytes. Splenocytes isolated from LBB-treated mice exhibited higher TNF-α expression and proliferative capacity. These results show that heat-killed L. brevis, a wildly consumed probiotic, may provide protection against pathogens through enhancing host immunity.
Collapse
Affiliation(s)
- Minju Jeong†
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Hwan Kim†
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Su Lee
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Shin Dal Kang
- Research Institute of Food and Biotechnology, SPC Group, Seoul 151742, Republic of Korea
| | - Sangmin Shim
- Research Institute of Food and Biotechnology, SPC Group, Seoul 151742, Republic of Korea
| | - Moon Young Jung
- Research Institute of Food and Biotechnology, SPC Group, Seoul 151742, Republic of Korea
| | - Hee Yang
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea,Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - Sanguine Byun
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea,Corresponding authors K.W.L. Phone: +82-2-880-4662 E-mail:
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea,Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea,Corresponding authors K.W.L. Phone: +82-2-880-4662 E-mail:
| |
Collapse
|
24
|
Sireswar S, Biswas S, Dey G. Adhesion and anti-inflammatory potential of Lactobacillus rhamnosus GG in a sea buckthorn based beverage matrix. Food Funct 2020; 11:2555-2572. [DOI: 10.1039/c9fo02249j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A seabuckthorn based beverage matrix retains the functionality of L. rhamnosus GG and exhibits enhanced anti-inflammatory effects against LPS-induced inflammation in zebrafish.
Collapse
Affiliation(s)
- Srijita Sireswar
- School of Biotechnology
- Kalinga Institute of Industrial Technology
- Deemed to be University
- Bhubaneswar
- India
| | | | - Gargi Dey
- School of Biotechnology
- Kalinga Institute of Industrial Technology
- Deemed to be University
- Bhubaneswar
- India
| |
Collapse
|
25
|
Kumperscak HG, Gricar A, Ülen I, Micetic-Turk D. A Pilot Randomized Control Trial With the Probiotic Strain Lactobacillus rhamnosus GG (LGG) in ADHD: Children and Adolescents Report Better Health-Related Quality of Life. Front Psychiatry 2020; 11:181. [PMID: 32256407 PMCID: PMC7092625 DOI: 10.3389/fpsyt.2020.00181] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/25/2020] [Indexed: 12/28/2022] Open
Abstract
Objectives: This double-blind pilot randomized placebo-controlled trial examined the possible effect of the probiotic strain Lactobacillus rhamnosus GG ATCC53103 (LGG) on symptoms of attention-deficit/hyperactivity disorder (ADHD), health-related quality of life (QoL), and serum levels of cytokines in children and adolescents with ADHD. Methods: This trial evaluated 32 drug-naive children and adolescents aged between four and 17 years with a diagnosis of ADHD. The study subjects were randomly assigned to either the group that received LGG or the group that received the placebo. Assessments, comprising the ADHD Parent-Report Rating Scale-IV: Home Version; the Child Self-Report and Parent Proxy-Report of the Pediatric Quality of Life Inventory TM (PedsQL TM ) 4.0 Generic Core Scale; the Parent Form (CBCL/6-18) and the Teacher Report Form (TRF) of the Child Behavior Checklist (CBCL) for ages 6-18 of the Achenbach System of Empirically Based Assessment (ASEBA); and the serum cytokines; were compared between the groups at the baseline and after 3 months. Results: Thirty-five participants were randomized, with 32 completing the study (91.4% retention). There was a significant improvement in the PedsQL Child Self-Report Total Score after 3 months of treatment in the probiotic (p = 0.021, d = 0.53), whereas there was no significant improvement in the placebo group (p = 0.563, d = 0.04). The results of psychometric parameters assessed by parents and teachers were not so straightforward. There were statistically significant differences in the levels of serum cytokines between the groups after the 3-month treatment period: IL-6 in both the probiotic (p = 0.004, d = 0.73) and the placebo groups (p = 0.035, d = 0.94); IL-10 (p = 0.035, d = 0.6); IL-12 p70 (p = 0.025, d = 0.89); and TNF-α (p = 0.046, d = 0.64) in the probiotic group only. Conclusions: Children and adolescents with ADHD who received LGG supplementation reported better health-related QoL compared to their peers who received the placebo. This suggests that LGG supplementation could be beneficial. But results with psychometric tests conducted by parents and teachers as well as differences in the levels of inflammatory cytokines were ambiguous. Based on these results, we propose some study modifications: a longer observation period (6-12 months); inclusion of more children's self-report assessments; recruitment of non-drug naive patients and the possible omission of serum cytokines measurements. Clinical Trial Registration: Medical Ethics Committee (UKC-MB-KME-19-06/16).
Collapse
Affiliation(s)
- Hojka Gregoric Kumperscak
- Pediatric Clinic, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Alja Gricar
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Ina Ülen
- Community Health Center Dr. Adolf Drolc, Maribor, Slovenia
| | | |
Collapse
|
26
|
Li C, Bei T, Niu Z, Guo X, Wang M, Lu H, Gu X, Tian H. Adhesion and Colonization of the Probiotic Lactobacillus rhamnosus Labeled by Dsred2 in Mouse Gut. Curr Microbiol 2019; 76:896-903. [PMID: 31115599 DOI: 10.1007/s00284-019-01706-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022]
Abstract
The health-promoting effects of the probiotic strain Lactobacillus rhamnosus are based on its adherence and colonization ability. However, little is known about its adhesion and colonization rates. Lactobacillus rhamnosus in mouse intestinal mucosa a mutant of the red fluorescence protein (RFP) DSred2 was used to tag L. rhamnosus to observe the adhesion and distribution of L. rhamnosus in mouse intestinal mucosa. A mutant of the red fluorescence protein (RFP) Dsred2 was used to tag L. rhamnosus to allow us to observe and distinguish it in the mouse intestine. Seven-week-old female BALB/c mice were fed once (at day 0) with an oral administration of the labeled L. rhamnosus, and the number of labeled bacteria was detected in different regions of the intestinal tract at 3 h and at day 1, 2, 3, 4, 5, 6, 7, and 15 after administration. The labeling process changed the morphology of L. rhamnosus, as it appeared after observation under the microscope, but did not change its basic probiotic properties in vitro. In vivo, labeled L. rhamnosus reached the colonization peak at the fourth day after gavage. From the distribution point of view, the number of colonization strains increased from the proximal to the distal small intestine (duodenum < jejunum < ileum) and the number of strains in the colon was less than the distal small intestine (ileum). The labeling protocol actually allowed the detection of the distribution and adhesion of this bacterium to the intestine, thus demonstrating that the health-promoting effects of this probiotic are satisfied. This study provides a scientific basis in the use of probiotics such as L. rhamnosus in functional foods.
Collapse
Affiliation(s)
- Chen Li
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Tingting Bei
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Zhihua Niu
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Xin Guo
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Miaoshu Wang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, China
| | - Haiqiang Lu
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Xinxi Gu
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Hongtao Tian
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, 071000, China.
| |
Collapse
|
27
|
Abstract
Lactobacillus rhamnosus GG (LGG) was the first strain belonging to the genus Lactobacillus to be patented in 1989 thanks to its ability to survive and to proliferate at gastric acid pH and in medium containing bile, and to adhere to enterocytes. Furthermore LGG is able to produces both a biofilm that can mechanically protect the mucosa, and different soluble factors beneficial to the gut by enhancing intestinal crypt survival, diminishing apoptosis of the intestinal epithelium, and preserving cytoskeletal integrity. Moreover LGG thanks to its lectin-like protein 1 and 2 inhibits some pathogens such as Salmonella species. Finally LGG is able to promote type 1 immune-responsiveness by reducing the expression of several activation and inflammation markers on monocytes and by increasing the production of interleukin-10, interleukin-12 and tumor necrosis factor-α in macrophages. A large number of research data on Lactobacillus GG is the basis for the use of this probiotic for human health. In this review we have considered predominantly randomized controlled trials, meta-analysis, Cochrane Review, guide lines of Scientific Societies and anyway studies whose results were evaluated by means of relative risk, odds ratio, weighted mean difference 95% confidence interval. The effectiveness of LGG in gastrointestinal infections and diarrhea, antibiotic and Clostridium difficile associated diarrhea, irritable bowel syndrome, inflammatory bowel disease, respiratory tract infections, allergy, cardiovascular diseases, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, cystic fibrosis, cancer, elderly end sport were analyzed.
Collapse
|
28
|
Gorreja F. Gene expression changes as predictors of the immune-modulatory effects of probiotics: Towards a better understanding of strain-disease specific interactions. NFS JOURNAL 2019. [DOI: 10.1016/j.nfs.2019.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, Stefani S, Torino F, Banna GL, Tonini G, Libra M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers (Basel) 2019; 11:38. [PMID: 30609850 PMCID: PMC6356461 DOI: 10.3390/cancers11010038] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer is a multifactorial pathology and it represents the second leading cause of death worldwide. In the recent years, numerous studies highlighted the dual role of the gut microbiota in preserving host's health. Gut resident bacteria are able to produce a number of metabolites and bioproducts necessary to protect host's and gut's homeostasis. Conversely, several microbiota subpopulations may expand during pathological dysbiosis and therefore produce high levels of toxins capable, in turn, to trigger both inflammation and tumorigenesis. Importantly, gut microbiota can interact with the host either modulating directly the gut epithelium or the immune system. Numerous gut populating bacteria, called probiotics, have been identified as protective against the genesis of tumors. Given their capability of preserving gut homeostasis, probiotics are currently tested to help to fight dysbiosis in cancer patients subjected to chemotherapy and radiotherapy. Most recently, three independent studies show that specific gut resident species may potentiate the positive outcome of anti-cancer immunotherapy. The highly significant studies, uncovering the tight association between gut microbiota and tumorigenesis, as well as gut microbiota and anti-cancer therapy, are here described. The role of the Lactobacillus rhamnosus GG (LGG), as the most studied probiotic model in cancer, is also reported. Overall, according to the findings here summarized, novel strategies integrating probiotics, such as LGG, with conventional anti-cancer therapies are strongly encouraged.
Collapse
Affiliation(s)
- Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Rossella Salemi
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, 95123 Catania, Italy.
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, 95123 Catania, Italy.
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, Tor Vergata University of Rome, 00133 Rome, Italy.
| | | | - Giuseppe Tonini
- Department of Medical Oncology, University Campus Bio-Medico of Rome, 00128 Rome, Italy.
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
30
|
Jeffrey MP, Strap JL, Jones Taggart H, Green-Johnson JM. Suppression of Intestinal Epithelial Cell Chemokine Production by Lactobacillus rhamnosus R0011 and Lactobacillus helveticus R0389 Is Mediated by Secreted Bioactive Molecules. Front Immunol 2018; 9:2639. [PMID: 30524427 PMCID: PMC6262363 DOI: 10.3389/fimmu.2018.02639] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Host intestinal epithelial cells (IEC) present at the gastrointestinal interface are exposed to pathogenic and non-pathogenic bacteria and their products. Certain probiotic lactic acid bacteria (LAB) have been associated with a range of host-immune modulatory activities including down-regulation of pro-inflammatory gene expression and cytokine production by IEC, with growing evidence suggesting that these bacteria secrete bioactive molecules with immunomodulatory activity. The aim of this study was to determine whether two lactobacilli with immunomodulatory activity [Lactobacillus rhamnosus R0011 (Lr) and Lactobacillus helveticus R0389 (Lh)], produce soluble mediators able to influence IEC responses to Pattern Recognition Receptor (PRR) ligands and pro-inflammatory cytokines [Tumor Necrosis Factor α (TNFα), Interleukin-1β (IL-1β)], signals inducing IEC chemokine production during infection. To this end, the effects of cell-free supernatants (CFS) from Lr and Lh on IEC production of the pro-inflammatory chemokines interleukin (IL)-8 and cytokine-induced neutrophil chemoattractant 1 (CINC-1) induced by a range of host- or pathogen-derived pro-inflammatory stimuli were determined, and the impact on human HT-29 IEC and a primary IEC line (rat IEC-6) was compared. The Lr-CFS and Lh-CFS did not significantly modulate basal IL-8 production from HT-29 IECs or CINC-1 production from IEC-6 cells. However, both Lr-CFS and Lh-CFS significantly down-regulated IL-8 production from HT-29 IECs challenged with varied PRR ligands. Lr-CFS and Lh-CFS had differential effects on PRR-induced CINC-1 production by rat IEC-6 IECs, with no significant down-regulation of CINC-1 observed from IEC-6 IECs cultured with Lh-CFS. Further analysis of the Lr-CFS revealed down-regulation of IL-8 production induced by the pro-inflammatory cytokines IL-1β and TNFα Preliminary characterization of the bioactive constituent(s) of the Lr-CFS indicates that it is resistant to treatment with DNase, RNase, and an acidic protease, but is sensitive to alterations in pH. Taken together, these results indicate that these lactobacilli secrete bioactive molecules of low molecular weight that may modulate host innate immune activity through interactions with IEC.
Collapse
Affiliation(s)
- Michael P Jeffrey
- Applied Bioscience Graduate Program, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | - Janice L Strap
- Applied Bioscience Graduate Program, University of Ontario Institute of Technology, Oshawa, ON, Canada.,Faculty of Science, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | - Holly Jones Taggart
- Applied Bioscience Graduate Program, University of Ontario Institute of Technology, Oshawa, ON, Canada.,Faculty of Health Sciences, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | - Julia M Green-Johnson
- Applied Bioscience Graduate Program, University of Ontario Institute of Technology, Oshawa, ON, Canada.,Faculty of Science, University of Ontario Institute of Technology, Oshawa, ON, Canada
| |
Collapse
|
31
|
Ludwig IS, Broere F, Manurung S, Lambers TT, van der Zee R, van Eden W. Lactobacillus rhamnosus GG-Derived Soluble Mediators Modulate Adaptive Immune Cells. Front Immunol 2018; 9:1546. [PMID: 30042761 PMCID: PMC6048560 DOI: 10.3389/fimmu.2018.01546] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022] Open
Abstract
Probiotics and probiotic-related nutritional interventions have been described to have beneficial effects on immune homeostasis and gut health. In previous studies, Lactobacillus rhamnosus GG (LGG) soluble mediators (LSM) have been demonstrated to exert beneficial effects in preclinical models of allergic sensitization, bacterial infection, and intestinal barrier function. In the context of allergic diseases, differentiation of dendritic cells (DCs) and their interactions with T cell populations are crucial for driving tolerogenic responses. In this study, we set out to evaluate whether these LSM can modulate DC maturation and have an impact on prompting protective and/or tolerogenic T cell responses. Monocytes were isolated from PBMC of healthy blood donors and cultured in the presence of GM-CSF, IL-4, and LSM or unconditioned bacterial culture medium control (UCM) during 6 days to induce DC differentiation. Subsequently, these DCs were matured in the presence of TNF-α for 1 day and analyzed for their phenotype and ability to induce autologous T cell activation and differentiation to model recall antigens. After 7 days of co-culture, T cells were analyzed for activation and differentiation by flow cytometry of intracellular cytokines (IFN-γ, IL-2, IL-10, and IL-17A), activation markers (CD25), and Foxp3+ expression. LSM did not alter DC numbers or maturation status. However, these DCs did show improved capacity to induce a T cell response as shown by increased IL-2 and IFN-γ producing T cell populations upon stimulation with recall antigens. These enhanced recall responses coincided with enhanced Foxp3+ expression that was not observed when T cells were cultured in the presence of UCM-treated DCs. By contrast, the number of activated T cells (determined by CD25 expression) was only slightly increased. In conclusion, this study reveals that LSM can influence adaptive immune responses as shown by the modulation of DC functionality. These mechanisms might contribute to previous observed effects in animal models in vivo. Altogether, these results suggest that LSM may provide an alternative to live probiotics in case life bacteria may not be used because of health conditions, although further clinical testing is needed.
Collapse
Affiliation(s)
- Irene S. Ludwig
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Femke Broere
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | | | - Tim T. Lambers
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, Netherlands
| | - Ruurd van der Zee
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
32
|
Two-step production of anti-inflammatory soluble factor by Lactobacillus reuteri CRL 1098. PLoS One 2018; 13:e0200426. [PMID: 29979794 PMCID: PMC6034873 DOI: 10.1371/journal.pone.0200426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/26/2018] [Indexed: 01/28/2023] Open
Abstract
We have demonstrated previously that a soluble factor (LrS) produced by Lactobacillus (L.) reuteri CRL 1098 modulates the inflammatory response triggered by lipopolysaccharide. In this study, the production of LrS by L. reuteri CRL 1098 was realized through two steps: i) bacterial biomass production, ii) LrS production, where the bacterial biomass was able to live but did not proliferate. Therefore, the simultaneous evaluation of the effect of different factors on the growth and LrS production was performed. Biomass production was found to be dependent mainly on culture medium, while LrS production with anti-inflammatory activity depended on culture conditions of the biomass such as pH, agitation and growth phase. The L. reuteri CRL 1098 biomass and LrS production in the optimized culture media designed for this work reduced the complete process cost by approximately 95%, respectively to laboratory scale cost.
Collapse
|
33
|
Probiotics and antibiotic-associated diarrhea in children: A review and new evidence on Lactobacillus rhamnosus GG during and after antibiotic treatment. Pharmacol Res 2018; 128:63-72. [DOI: 10.1016/j.phrs.2017.08.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 12/13/2022]
|
34
|
Jatzlauk G, Bartel S, Heine H, Schloter M, Krauss-Etschmann S. Influences of environmental bacteria and their metabolites on allergies, asthma, and host microbiota. Allergy 2017; 72:1859-1867. [PMID: 28600901 DOI: 10.1111/all.13220] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2017] [Indexed: 02/07/2023]
Abstract
The prevalence of allergic diseases and asthma has dramatically increased over the last decades, resulting in a high burden for patients and healthcare systems. Thus, there is an unmet need to develop preventative strategies for these diseases. Epidemiological studies show that reduced exposure to environmental bacteria in early life (eg, birth by cesarean section, being formula-fed, growing up in an urban environment or with less contact to various persons) is associated with an increased risk to develop allergies and asthma later in life. Conversely, a reduced risk for asthma is consistently found in children growing up on traditional farms, thereby being exposed to a wide spectrum of microbes. However, clinical studies with bacteria to prevent allergic diseases are still rare and to some extent contradicting. A detailed mechanistic understanding of how environmental microbes influence the development of the human microbiome and the immune system is important to enable the development of novel preventative approaches that are based on the early modulation of the host microbiota and immunity. In this mini-review, we summarize current knowledge and experimental evidence for the potential of bacteria and their metabolites to be used for the prevention of asthma and allergic diseases.
Collapse
Affiliation(s)
- G. Jatzlauk
- Division of Early Life Origins of Chronic Lung Diseases; Priority Area Asthma and Allergy; Research Center Borstel; Leibniz-Center for Medicine and Biosciences; Member of the Airway Research Center North (ARCN); German Center for Lung Research (DZL); Borstel Germany
| | - S. Bartel
- Division of Early Life Origins of Chronic Lung Diseases; Priority Area Asthma and Allergy; Research Center Borstel; Leibniz-Center for Medicine and Biosciences; Member of the Airway Research Center North (ARCN); German Center for Lung Research (DZL); Borstel Germany
| | - H. Heine
- Division of Innate Immunity; Priority Area Asthma and Allergy; Research Center Borstel; Leibniz-Center for Medicine and Biosciences; Member of the Airway Research Center North (ARCN); German Center for Lung Research (DZL); Borstel Germany
| | - M. Schloter
- Research Unit Environmental Genomics; Helmholtz Zentrum München; Oberschleißheim Germany
| | - S. Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases; Priority Area Asthma and Allergy; Research Center Borstel; Leibniz-Center for Medicine and Biosciences; Member of the Airway Research Center North (ARCN); German Center for Lung Research (DZL); Borstel Germany
- Institute for Experimental Medicine; Christian-Albrechts-Universität zu Kiel; Kiel Germany
| |
Collapse
|
35
|
Kwak JY, Lamousé-Smith ESN. Can probiotics enhance vaccine-specific immunity in children and adults? Benef Microbes 2017; 8:657-670. [PMID: 28856905 DOI: 10.3920/bm2016.0147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The growing use of probiotics by the general public has heightened the interest in understanding the role of probiotics in promoting health and preventing disease. General practitioners and specialists often receive inquiries from their patients regarding probiotic products and their use to ward off systemic infection or intestinal maladies. Enhanced immune function is among the touted health benefits conferred by probiotics but has not yet been fully established. Results from recent clinical trials in adults suggest a potential role for probiotics in enhancing vaccine-specific immunity. Although almost all vaccinations are given during infancy and childhood, the numbers of and results from studies using probiotics in pediatric subjects are limited. This review evaluates recent clinical trials of probiotics used to enhance vaccine-specific immune responses in adults and infants. We highlight meaningful results and the implications of these findings for designing translational and clinical studies that will evaluate the potential clinical role for probiotics. We conclude that the touted health claims of probiotics for use in children to augment immunity warrant further investigation. In order to achieve this goal, a consensus should be reached on common study designs that apply similar treatment timelines, compare well-characterised probiotic strains and monitor effective responses against different classes of vaccines.
Collapse
Affiliation(s)
- J Y Kwak
- 1 Department of Pediatrics, Columbia University Medical Center, PH17-105G, 622 West 168th Street, New York, NY 10032, USA
| | - E S N Lamousé-Smith
- 1 Department of Pediatrics, Columbia University Medical Center, PH17-105G, 622 West 168th Street, New York, NY 10032, USA
| |
Collapse
|
36
|
Gonzalez-Perez G, Lamousé-Smith ESN. Gastrointestinal Microbiome Dysbiosis in Infant Mice Alters Peripheral CD8 + T Cell Receptor Signaling. Front Immunol 2017; 8:265. [PMID: 28337207 PMCID: PMC5340779 DOI: 10.3389/fimmu.2017.00265] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 02/23/2017] [Indexed: 01/08/2023] Open
Abstract
We recently reported that maternal antibiotic treatment (MAT) of mice in the last days of pregnancy and during lactation dramatically alters the density and composition of the gastrointestinal microbiota of their infants. MAT infants also exhibited enhanced susceptibility to a systemic viral infection and altered adaptive immune cell activation phenotype and function. CD8+ effector T cells from MAT infants consistently demonstrate an inability to sustain interferon gamma (IFN-γ) production in vivo following vaccinia virus infection and in vitro upon T cell receptor (TCR) stimulation. We hypothesize that T cells developing in infant mice with gastrointestinal microbiota dysbiosis and insufficient toll-like receptor (TLR) exposure alters immune responsiveness associated with intrinsic T cell defects in the TCR signaling pathway and compromised T cell effector function. To evaluate this, splenic T cells from day of life 15 MAT infant mice were stimulated in vitro with anti-CD3 and anti-CD28 antibodies prior to examining the expression of ZAP-70, phosphorylated ZAP-70, phospho-Erk-1/2, c-Rel, total protein tyrosine phosphorylation, and IFN-γ production. We determine that MAT infant CD8+ T cells fail to sustain total protein tyrosine phosphorylation and Erk1/2 activation. Lipopolysaccharide treatment in vitro and in vivo, partially restored IFN-γ production in MAT effector CD8+ T cells and reduced mortality typically observed in MAT mice following systemic viral infection. Our results demonstrate a surprising dependence on the gastrointestinal microbiome and TLR ligand stimulation toward shaping optimal CD8+ T cell function during infancy.
Collapse
Affiliation(s)
- Gabriela Gonzalez-Perez
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Medical Center , New York, NY , USA
| | - Esi S N Lamousé-Smith
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Medical Center , New York, NY , USA
| |
Collapse
|
37
|
Lactobacillus rhamnosus GG supernatant enhance neonatal resistance to systemic Escherichia coli K1 infection by accelerating development of intestinal defense. Sci Rep 2017; 7:43305. [PMID: 28262688 PMCID: PMC5338013 DOI: 10.1038/srep43305] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
The objective of this study was to determine whether Lactobacillus rhamnosus GG culture supernatant (LCS) has a preventive effect against gut-derived systemic neonatal Escherichia coli (E. coli) K1 infection. The preventive effects were evaluated in human colonic carcinoma cell line Caco-2 and neonatal rat models. Our in vitro results showed that LCS could block adhesion, invasion and translocation of E. coli K1 to Caco-2 monolayer via up-regulating mucin production and maintaining intestinal integrity. In vivo experiments revealed that pre-treatment with LCS significantly decrease susceptibility of neonatal rats to oral E. coli K1 infection as reflected by reduced bacterial intestinal colonization, translocation, dissemination and systemic infections. Further, we found that LCS treated neonatal rats have higher intestinal expressions of Ki67, MUC2, ZO-1, IgA, mucin and lower barrier permeability than those in untreated rats. These results indicated that LCS could enhance neonatal resistance to systemic E. coli K1 infection via promoting maturation of neonatal intestinal defense. In conclusions, our findings suggested that LCS has a prophylactic effect against systemic E. coli K1 infection in neonates. Future studies aimed at identifying the specific active ingredients in LCS will be helpful in developing effective pharmacological strategies for preventing neonatal E. coli K1 infection.
Collapse
|
38
|
曾 庆, 何 肖, 肖 汉, 杜 蕾, 李 雨, 陈 乐, 田 慧, 黄 胜, 曹 虹. [Lactobacillus rhamnosus GG conditioned medium prevents E. coli meningitis by inhibiting nuclear factor-κB pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:24-29. [PMID: 28109094 PMCID: PMC6765744 DOI: 10.3969/j.issn.1673-4254.2017.01.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To investigate whether Lactobacillus rhamnosus GG conditioned medium(LGG-CM)has preventive effect against E. coli K1-induced neuropathogenicity in vitro by inhibiting nuclear factor-κB (NF-κB) signaling pathway. METHODS An in vitro blood-brain barrier (BBB) model was constructed using human brain microvascular endothelial cells (HBMECs). The effect of LGG-CM on E. coli-actived NF-κB signaling pathway was assayed using Western blotting. Invasion assay and polymorphonuclear leukocyte (PMN) transmigration assay were performed to explore whether LGG-CM could inhibit E. coli invasion and PMN transmigration across the BBB in vitro. The expressions of ZO-1 and CD44 were detected using Western blotting and immunofluorescence. The changes of trans-epithelial electric resistance (TEER) and bacterial translocation were determined to evaluate the BBB permeability. RESULTS Pre-treament with LGG-CM inhibited E. coli-activated NF-κB signaling pathway in HBMECs and decreased the invasion of E. coli K1 and transmigration of PMN. Western blotting showed that LGG-CM could alleviate E. coli-induced up-regulation of CD44 and down-regulation of ZO-1 expressions in HBMECs. In addition, pre-treatment with LGG-CM alleviated E. coli K1-induced reduction of TEER and suppressed bacterial translocation across the BBB in vitro. CONCLUSION LGG-CM can block E. coli-induced activation of NF-κB signaling pathway and thereby prevents E. coli K1-induced neuropathogenicity by decreasing E. coli K1 invasion rates and PMN transmigration.
Collapse
Affiliation(s)
- 庆 曾
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| | - 肖龙 何
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| | - 汉森 肖
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| | - 蕾 杜
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| | - 雨静 李
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| | - 乐程 陈
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| | - 慧文 田
- 南方医科大学 第一临床医学院, 广东 广州 510515First College of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - 胜和 黄
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
- 南加州大学洛杉矶儿童医院Saban研究所, 美国 洛杉矶 90027Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, 90027, USA
| | - 虹 曹
- 南方医科大学 公共卫生学院//广东省热带病研究重点实验室, 微生物学系, 广东 广州 510515Department of Microbiology, School of Public healthy
| |
Collapse
|
39
|
Johansson MA, Björkander S, Mata Forsberg M, Qazi KR, Salvany Celades M, Bittmann J, Eberl M, Sverremark-Ekström E. Probiotic Lactobacilli Modulate Staphylococcus aureus-Induced Activation of Conventional and Unconventional T cells and NK Cells. Front Immunol 2016; 7:273. [PMID: 27462316 PMCID: PMC4939411 DOI: 10.3389/fimmu.2016.00273] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/29/2016] [Indexed: 12/17/2022] Open
Abstract
Lactobacilli are probiotic commensal bacteria and potent modulators of immunity. When present in the gut or supplemented as probiotics, they beneficially modulate ex vivo immune responsiveness. Further, factors derived from several lactobacilli strains act immune regulatory in vitro. In contrast, Staphylococcus aureus (S. aureus) is known to induce excessive T cell activation. In this study, we aimed to investigate S. aureus-induced activation of human mucosal-associated invariant T cells (MAIT cells), γδ T cells, NK cells, as well as of conventional CD4+ and CD8+ T cells in vitro. Further, we investigated if lactobacilli-derived factors could modulate their activation. PBMC were cultured with S. aureus 161:2 cell-free supernatants (CFS), staphylococcal enterotoxin A or CD3/CD28-beads alone, or in combination with Lactobacillus rhamnosus GG-CFS or Lactobacillus reuteri DSM 17938-CFS and activation of T and NK cells was evaluated. S. aureus-CFS induced IFN-γ and CD107a expression as well as proliferation. Costimulation with lactobacilli-CFS dampened lymphocyte-activation in all cell types analyzed. Preincubation with lactobacilli-CFS was enough to reduce subsequent activation, and the absence of APC or APC-derived IL-10 did not prevent lactobacilli-mediated dampening. Finally, lactate selectively dampened activation of unconventional T cells and NK cells. In summary, we show that molecules present in the lactobacilli-CFS are able to directly dampen in vitro activation of conventional and unconventional T cells and of NK cells. This study provides novel insights on the immune-modulatory nature of probiotic lactobacilli and suggests a role for lactobacilli in the modulation of induced T and NK cell activation.
Collapse
Affiliation(s)
- Maria A Johansson
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Sophia Björkander
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Manuel Mata Forsberg
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Khaleda Rahman Qazi
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Maria Salvany Celades
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Julia Bittmann
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Eva Sverremark-Ekström
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| |
Collapse
|
40
|
Iqbal J, Yuen T, Sun L, Zaidi M. From the gut to the strut: where inflammation reigns, bone abstains. J Clin Invest 2016; 126:2045-8. [PMID: 27111233 DOI: 10.1172/jci87430] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In this issue of the JCI, Li et al. show that germ-free mice, when chemically castrated, do not lose bone - a finding that unequivocally establishes a role of gut microbiota in mediating hypogonadal bone loss. Additionally and not unexpectedly, probiotics reversed hypogonadal osteopenia in sex steroid-deficient mice by preventing the disruption of gut barrier function and dampening cytokine-induced inflammation. The authors propose that TNFα is a key mediator; however, it is very likely that other molecules - including IL-1, IL-6, IL-17, RANKL, OPG, and CCL2 - modulate probiotic action. The results of this study highlight the potential for repurposing probiotics for the therapy of osteoporosis. Future placebo-controlled clinical trials will be required to establish safety and efficacy of probiotics in reducing fracture risk in people.
Collapse
|