1
|
Quansah M, David MA, Martins R, El-Omar E, Aliberti SM, Capunzo M, Jensen SO, Tayebi M. The Beneficial Effects of Lactobacillus Strains on Gut Microbiome in Alzheimer's Disease: A Systematic Review. Healthcare (Basel) 2025; 13:74. [PMID: 39791681 PMCID: PMC11720007 DOI: 10.3390/healthcare13010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND/OBJECTIVES Growing evidence suggests that the gut-brain axis influences brain function, particularly the role of intestinal microbiota in modulating cognitive processes. Probiotics may alter brain function and behavior by modulating gut microbiota, with implications for neurodegenerative diseases like Alzheimer's disease (AD). The purpose of this review is to systematically review the current literature exploring the effects of probiotic supplementation on gut microbiota and cognitive function in AD and mild cognitive impairment (MCI). METHODS A comprehensive literature search was conducted across PubMed/Medline, Embase, and Scopus to identify relevant randomized controlled trials (RCTs) from inception to 20 August 2024. The search focused on comparing outcomes between intervention and control/placebo groups. Data searches, article selection, data extraction, and risk of bias assessment were performed in accordance with Cochrane guidelines. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no: CRD42023446796. RESULTS Data from four RCTs involving 293 Individuals (AD and MCI patients) receiving mainly Lactobacillus and Bifidobacterium strains showed some beneficial effects on cognitive function, altered gut microbiota composition, and positively affected metabolic biomarkers. However, variability in microbiota assessment across studies limits the interpretation of results. The limited number and quality of the existing studies make it difficult to draw definitive conclusions from the data. Additional high-quality research is clearly needed. CONCLUSIONS Probiotics show promise as an adjunctive intervention for cognitive decline, but larger, long-term trials are needed to confirm their efficacy and clinical applicability in neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Michael Quansah
- Neuroimmunology Laboratory, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (M.Q.); (M.A.D.)
- Department of Medicine and Therapeutics, Medical School, University of Ghana, Accra LG25, Ghana
| | - Monique Antoinette David
- Neuroimmunology Laboratory, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (M.Q.); (M.A.D.)
| | - Ralph Martins
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia;
| | - Emad El-Omar
- Microbiome Research Centre, School of Clinical Medicine, UNSW Medicine & Health, St George & Sutherland Clinical Campuses, UNSW, Kogarah, NSW 2217, Australia;
| | - Silvana Mirella Aliberti
- Hygiene and Preventive Medicine Section, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (S.M.A.); (M.C.)
| | - Mario Capunzo
- Hygiene and Preventive Medicine Section, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (S.M.A.); (M.C.)
| | - Slade O. Jensen
- School of Medicine, Microbiology and Infectious Diseases, Ingham Institute for Applied Medical Research, Western Sydney University, Liverpool, NSW 1871, Australia;
| | - Mourad Tayebi
- Neuroimmunology Laboratory, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (M.Q.); (M.A.D.)
| |
Collapse
|
2
|
Yan R, Zeng X, Shen J, Wu Z, Guo Y, Du Q, Tu M, Pan D. New clues for postbiotics to improve host health: a review from the perspective of function and mechanisms. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6376-6387. [PMID: 38450745 DOI: 10.1002/jsfa.13444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 03/08/2024]
Abstract
Strain activity and stability severely limit the beneficial effects of probiotics in modulating host health. Postbiotics have emerged as a promising alternative as they can provide similar or even enhanced efficacy to probiotics, even under inactivated conditions. This review introduces the ingredients, preparation, and identification techniques of postbiotics, focusing on the comparison of the advantages and limitations between probiotics and postbiotics based on their mechanisms and applications. Inactivation treatment is the most significant difference between postbiotics and probiotics. We highlight the use of emerging technologies to inactivate probiotics, optimize process conditions to maintain the activity of postbiotics, or scale up their production. Postbiotics have high stability which can overcome unfavorable factors, such as easy inactivation and difficult colonization of probiotics after entering the intestine, and are rapidly activated, allowing continuous and rapid optimization of the intestinal microecological environment. They provide unique mechanisms, and multiple targets act on the gut-organ axis, co-providing new clues for the study of the biological functions of postbiotics. We summarize the mechanisms of action of inactivated lactic acid bacteria, highlighting that the NF-κB and MAPK pathways can be used as immune targeting pathways for postbiotic modulation of host health. Generally, we believe that as the classification, composition, and efficacy mechanism of postbiotics become clearer they will be more widely used in food, medicine, and other fields, greatly enriching the dimensions of food innovation. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ruonan Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Xiaoqun Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Jiamin Shen
- Zhejiang Shenjinji Food Technology Co., LTD, Huzhou, China
| | - Zhen Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Yuxing Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Qiwei Du
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Maolin Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Luongo D, De Sena V, Maurano F, Rossi M. Modulation of Mouse Dendritic Cells In Vitro by Lactobacillus gasseri Postbiotic Proteins. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10292-6. [PMID: 38836988 DOI: 10.1007/s12602-024-10292-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/06/2024]
Abstract
Different lactobacilli are probiotics for their beneficial effects that confer to the host. Recently, some of these effects were associated with released metabolic products/constituents (postbiotics). In the present study, the potential immunomodulatory capacity of the probiotic Lactobacillus gasseri OLL2809 cell-free supernatant (sup) was investigated in murine bone marrow-derived dendritic cells (DCs). Bacteria induced significantly higher expression of all examined cytokines than those induced by the stimulatory lipopolysaccharide (LPS) itself. On the contrary, sup only induced the anti-inflammatory IL-10 similarly to LPS, whereas IL-12 and IL-6 secretions were stimulated at a lower level. Moreover, sup reduced the surface expression of the analyzed co-stimulatory markers CD40, CD80, and CD86. Treatments of sup with different digestive enzymes indicated the proteinaceous nature of these immunomodulatory metabolites. Western blot and immunoadsorption analyzes revealed cross-reactivity of sup with the surface-layer proteins (SLPs) isolated from OLL2809. Therefore, we directly tested the ability of OLL2809 SLPs to stimulate specifically cytokine expression in iDCs. Interestingly, we found that all tested cytokines were induced by SLPs and in a dose-dependent manner. In conclusion, our results highlighted distinct immune properties between L. gasseri OLL2809 and its metabolites, supporting the concept that bacterial viability is not an essential prerequisite to exert immunomodulatory effects.
Collapse
Affiliation(s)
- Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy.
| | - Vincenzo De Sena
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy
| |
Collapse
|
4
|
Rocchetti MT, Russo P, De Simone N, Capozzi V, Spano G, Fiocco D. Immunomodulatory Activity on Human Macrophages by Cell-Free Supernatants to Explore the Probiotic and Postbiotic Potential of Lactiplantibacillus plantarum Strains of Plant Origin. Probiotics Antimicrob Proteins 2024; 16:911-926. [PMID: 37202651 PMCID: PMC11126452 DOI: 10.1007/s12602-023-10084-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/20/2023]
Abstract
Upon dietary administration, probiotic microorganisms can reach as live cells the human gut, where they interact with the microbiota and host cells, thereby exerting a beneficial impact on host functions, mainly through immune-modulatory activities. Recently, attention has been drawn by postbiotics, i.e. non-viable probiotic microbes, including their metabolic products, which possess biological activities that benefit the host. Lactiplantibacillus plantarum is a bacterial species that comprises recognised probiotic strains. In this study, we investigated in vitro the probiotic (and postbiotic) potential of seven L. plantarum strains, including five newly isolated from plant-related niches. The strains were shown to possess some basic probiotic attributes, including tolerance to the gastrointestinal environment, adhesion to the intestinal epithelium and safety. Besides, their cell-free culture supernatants modulated cytokine patterns in human macrophages in vitro, promoting TNF-α gene transcription and secretion, while attenuating the transcriptional activation and secretion of both TNF-α and IL-8 in response to a pro-inflammatory signal, and enhancing the production of IL-10. Some strains induced a high IL-10/IL-12 ratio that may correlate to an anti-inflammatory capacity in vivo. Overall, the investigated strains are good probiotic candidates, whose postbiotic fraction exhibits immunomodulatory properties that need further in vivo studies. The main novelty of this work consists in the polyphasic characterisation of candidate beneficial L. plantarum strains obtained from relatively atypical plant-associated niches, by an approach that explores both probiotic and postbiotic potentials, in particular studying the effect of microbial culture-conditioned media on cytokine pattern, analysed at both transcriptional and secretion level in human macrophages.
Collapse
Affiliation(s)
| | - Pasquale Russo
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Nicola De Simone
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, Foggia, Italy
| | - Vittorio Capozzi
- Institute of Sciences of Food Production, National Research Council (CNR) of Italy, C/O CS-DAT, Foggia, Italy
| | - Giuseppe Spano
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, Foggia, Italy
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
5
|
Rahman Z, Padhy HP, Dandekar MP. Cell-Free Supernatant of Lactobacillus rhamnosus and Bifidobacterium breve Ameliorates Ischemic Stroke-Generated Neurological Deficits in Rats. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10256-w. [PMID: 38656733 DOI: 10.1007/s12602-024-10256-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] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
The beneficial effects of probiotics, postbiotics, and paraprobiotics have already been registered in managing ischemic stroke-generated neuroinflammation and gut dysbiosis. Herein, we examined the impact of cell-free supernatant (CFS) obtained from probiotics (Lactobacillus rhamnosus UBLR-58 and Bifidobacterium breve UBBr-01) in a rat transient middle cerebral artery occlusion (MCAO) model of focal cerebral injury. Pre-MCAO supplementation of probiotics (2 × 109 CFU/mL) for 21 days or CFS (1 mL/rat) for 7 days protect the MCAO-induced somatosensory and motor impairments recorded at 24 h and 72 h after reperfusion in foot-fault, rotarod, adhesive removal, and vibrissae-evoked forelimb placing tests. We also noted the reduced infarct area and neuronal degradation in the right hemisphere of probiotics- and CFS-recipient MCAO-operated animals. Moreover, MCAO-induced altered concentrations of glial-fibrillary acidic protein, NeuN, zonula occludens-1 (ZO-1), TLR4, IL-1β, IL-6, and TNF-α, as well as matrix metalloproteinase-9 (MMP9) were reversed in the treatment groups. Probiotics and CFS treatment ameliorated the elevated levels of IL-6, IL-1β, and MMP9 in the blood plasma of rats. The disrupted microbial phyla, Firmicutes-to-Bacteroides ratio, villi/crypt ratio, and decreased mucin-producing goblet cells, ZO-1, and occludin in the colon of MCAO-operated rats were recovered following probiotics and CFS treatment. NMR characterization of CFS and rat blood plasma revealed the presence of several important bacterial metabolites. These findings suggest that the CFS obtained from Lactobacillus rhamnosus UBLR-58 and Bifidobacterium breve UBBr-01 has the propensity to improve MCAO-generated neurological dysfunctions in rats by dampening neuroinflammation and modulating the gut-brain axis modulators.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, PIN 500037, Telangana, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, PIN 500037, Telangana, India.
| |
Collapse
|
6
|
Xie W, Zhong YS, Li XJ, Kang YK, Peng QY, Ying HZ. Postbiotics in colorectal cancer: intervention mechanisms and perspectives. Front Microbiol 2024; 15:1360225. [PMID: 38450163 PMCID: PMC10914944 DOI: 10.3389/fmicb.2024.1360225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
Colorectal cancer (CRC) is a common malignancy affecting the gastrointestinal tract worldwide. The etiology and progression of CRC are related to factors such as environmental influences, dietary structure, and genetic susceptibility. Intestinal microbiota can influence the integrity of the intestinal mucosal barrier and modulate intestinal immunity by secreting various metabolites. Dysbiosis of the intestinal microbiota can affect the metabolites of the microbial, leading to the accumulation of toxic metabolites, which can trigger chronic inflammation or DNA damage and ultimately lead to cellular carcinogenesis and the development of CRC. Postbiotics are preparations of inanimate microorganisms or their components that are beneficial to the health of the host, with the main components including bacterial components (e.g., exopolysaccharides, teichoic acids, surface layer protein) and metabolites (e.g., short-chain fatty acids, tryptophan metabolite, bile acids, vitamins and enzymes). Compared with traditional probiotics, it has a more stable chemical structure and higher safety. In recent years, it has been demonstrated that postbiotics are involved in regulating intestinal microecology and improving the progression of CRC, which provides new ideas for the prevention and diagnosis of CRC. In this article, we review the changes in intestinal microbiota in different states of the gut and the mechanisms of anti-tumor activity of postbiotic-related components, and discuss the potential significance of postbiotics in the diagnosis and treatment of CRC. This reviews the changes and pathogenesis of intestinal microbiota in the development of CRC, and summarizes the relevant mechanisms of postbiotics in resisting the development of CRC in recent years, as well as the advantages and limitations of postbiotics in the treatment process of CRC.
Collapse
Affiliation(s)
| | | | | | | | | | - Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
7
|
Heldner A, Heath MD, Schnautz B, Kotz S, Chaker A, Kramer MF, Jakwerth CA, Zissler UM, Schmidt-Weber CB, Blank S. Ex Vivo Immunomodulatory Effects of Lactobacillus-, Lacticaseibacillus-, and Bifidobacterium-Containing Synbiotics on Human Peripheral Blood Mononuclear Cells and Monocyte-Derived Dendritic Cells in the Context of Grass Pollen Allergy. Probiotics Antimicrob Proteins 2023; 15:868-879. [PMID: 35113319 PMCID: PMC10393851 DOI: 10.1007/s12602-022-09920-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Abstract
Sensing of the intestinal microbiota by the host immune system is important to induce protective immune responses. Hence, modification of the gut microbiota might be able to prevent or treat allergies, mediated by proinflammatory Th2 immune responses. The aim was to investigate the ex vivo immunomodulatory effects of the synbiotics Pollagen® and Kallergen®, containing the probiotic bacterial strains Lactobacillus, Lacticaseibacillus and Bifidobacterium, in the context of grass pollen allergy. Peripheral blood mononuclear cells (PBMCs) from grass pollen-allergic patients and healthy controls were stimulated with grass pollen extract (GPE) and synbiotics and Gata3 expression and cytokine secretion analyzed. Monocyte-derived dendritic cells (MoDCs) cells were matured in the presence of GPE and synbiotics, co-cultured with autologous naïve T cells and maturation markers and cytokine secretion analyzed. GPE stimulation of PBMCs from grass pollen-allergic patients resulted in a significant higher production of the Th2 cytokines IL-4, IL-5, IL-9 and IL-13 compared to healthy controls. Gata3+CD4+ T cell induction was independent of the allergic status. The synbiotics promoted IL-10 and IFN-γ secretion and downregulated the GPE-induced Th2-like phenotype. Co-culturing naïve T cells with MoDCs, matured in the presence of GPE and synbiotics, shifted the GPE-induced Th2 cytokine release towards Th1-Th17-promoting conditions in allergic subjects. The investigated synbiotics are effective in downregulating the GPE-induced Th2 immune response in PBMCs from grass pollen-allergic patients as well as in autologous MoDC-T cell stimulation assays. In addition to increased IL-10 release, the data indicates a shift from a Th2- to a more Th1- and Th17-like phenotype.
Collapse
Affiliation(s)
- Alexander Heldner
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | | | - Benjamin Schnautz
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Sebastian Kotz
- Faculty of Medicine, Department of Otolaryngology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Adam Chaker
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
- Faculty of Medicine, Department of Otolaryngology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Matthias F Kramer
- Allergy Therapeutics PLC, Worthing, UK
- Bencard Allergie GmbH, Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Simon Blank
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany.
| |
Collapse
|
8
|
Rosas-Val P, Adhami M, Brotons-Canto A, Gamazo C, Irache JM, Larrañeta E. 3D printing of microencapsulated Lactobacillus rhamnosus for oral delivery. Int J Pharm 2023; 641:123058. [PMID: 37207858 DOI: 10.1016/j.ijpharm.2023.123058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
3D Printing is an innovative technology within the pharma and food industries that allows the design and manufacturing of novel delivery systems. Orally safe delivery of probiotics to the gastrointestinal tract faces several challenges regarding bacterial viability, in addition to comply with commercial and regulatory standpoints. Lactobacillus rhamnosus CNCM I-4036 (Lr) was microencapsulated in generally recognised as safe (GRAS) proteins, and then assessed for robocasting 3D printing. Microparticles (MP-Lr) were developed and characterised, prior to being 3D printed with pharmaceutical excipients. MP-Lr showed a size of 12.3 ± 4.1 µm and a non-uniform wrinkled surface determined by Scanning Electron Microscopy (SEM). Bacterial quantification by plate counting accounted for 8.68 ±0.6 CFU/g of live bacteria encapsulated within. Formulations were able to keep the bacterial dose constant upon contact with gastric and intestinal pH. Printlets consisted in oval-shape formulations (15 mm × 8 mm × 3.2 mm) of ca. 370 mg of total weight, with a uniform surface. After the 3D printing process, bacterial viability remained even as MP-Lr protected bacteria alongside the process (log reduction of 0.52, p>0.05) in comparison with non-encapsulated probiotic (log reduction of 3.05). Moreover, microparticle size was not altered during the 3D printing process. We confirmed the success of this technology for developing an orally safe formulation, GRAS category, of microencapsulated Lr for gastrointestinal vehiculation.
Collapse
Affiliation(s)
- Pablo Rosas-Val
- Nucaps Nanotechnology S.L., Spain; Department of Microbiology & Parasitology, University of Navarra, Spain
| | | | | | - Carlos Gamazo
- Department of Microbiology & Parasitology, University of Navarra, Spain
| | - Juan M Irache
- Department of Technology & Pharmaceutical Chemistry, University of Navarra, Spain
| | | |
Collapse
|
9
|
Vale GC, Mota BIS, Ando-Suguimoto ES, Mayer MPA. Effect of Probiotics Lactobacillus acidophilus and Lacticaseibacillus rhamnosus on Antibacterial Response Gene Transcription of Human Peripheral Monocytes. Probiotics Antimicrob Proteins 2023; 15:264-274. [PMID: 34405373 DOI: 10.1007/s12602-021-09832-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 12/23/2022]
Abstract
Periodontitis and related systemic inflammatory diseases are characterized by imbalanced ratio between pro- and anti-inflammatory factors. Probiotics may control inflammation by altering the inflammatory phenotype of defense cells. We aimed to evaluate the gene transcription of the antibacterial response of monocytes to exposure to probiotic lactobacilli. CD14 + monocytes were obtained by positive selection from peripheral blood mononuclear cells from healthy donors (5 × 104 CD14 + /mL) and cultured with probiotic strains of Lacticaseibacillus rhamnosus (LR-32) and Lactobacillus acidophilus (LA-5) at a 1:10 multiplicity of infection in 24-well plates for 12 h. The gene expression analysis was performed by RT-qPCR using the Kit RT2 human antibacterial response, and in the supernatant, the cytokines were determined by ELISA. Tukey's post hoc test following an ANOVA with a p value of 5% was used for statistical analysis. Both probiotic strains increased the levels of cytokines TNF-α and CXCL-8 in the supernatant compared to the control of non-challenged cells (p < 0.05), but for IL-1Β and IL-6, this effect was observed only for LA-5 (p < 0.05). The fold-regulation values for the following genes for LA-5 and LR-32 were, respectively, IL-12B (431.94 and 33.30), IL-1Β (76.73 and 17.14), TNF-α (94.63 and 2.49), CXCL-8 (89.59 and 4.18), and TLR-2 (49.68 and 3.40). Likewise, most of the other genes evaluated showed greater expression for LA-5 compared to LR-32 (p < 0.05). The positive regulation of inflammatory factors such as IL-1β promoted by L. acidophilus LA-5 may increase the antibacterial activity of innate defense in periodontal tissues. However, this property may be deleterious by increasing inflammatory response.
Collapse
Affiliation(s)
- Glauber Campos Vale
- Restorative Dentistry Department, Federal University of Piauí, Teresina, Brazil.
| | | | | | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Álvarez-Mercado AI, Plaza-Díaz J, de Almagro MC, Gil Á, Moreno-Muñoz JA, Fontana L. Bifidobacterium longum subsp. infantis CECT 7210 Reduces Inflammatory Cytokine Secretion in Caco-2 Cells Cultured in the Presence of Escherichia coli CECT 515. Int J Mol Sci 2022; 23:10813. [PMID: 36142723 PMCID: PMC9503999 DOI: 10.3390/ijms231810813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Previous works have described the activity of Bifidobacterium longum subsp. infantis CECT 7210 (also commercially named B. infantis IM-1®) against rotavirus in mice and intestinal pathogens in piglets, as well as its diarrhea-reducing effect on healthy term infants. In the present work, we focused on the intestinal immunomodulatory effects of B. infantis IM-1® and for this purpose we used the epithelial cell line isolated from colorectal adenocarcinoma Caco-2 and a co-culture system of human dendritic cells (DCs) from peripheral blood together with Caco-2 cells. Single Caco-2 cultures and Caco-2: DC co-cultures were incubated with B. infantis IM-1® or its supernatant either in the presence or absence of Escherichia coli CECT 515. The B. infantis IM-1® supernatant exerted a protective effect against the cytotoxicity caused by Escherichia coli CECT 515 on single cultures of Caco-2 cells as viability reached the values of untreated cells. B. infantis IM-1® and its supernatant also decreased the secretion of pro-inflammatory cytokines by Caco-2 cells and the co-cultures incubated in the presence of E. coli CECT 515, with the response being more modest in the latter, which suggests that DCs modulate the activity of Caco-2 cells. Overall, the results obtained point to the immunomodulatory activity of this probiotic strain, which might underlie its previously reported beneficial effects.
Collapse
Affiliation(s)
- Ana I. Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, 18016 Armilla, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | | | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, 18016 Armilla, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Instituto de Salud Carlos III, CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
| | | | - Luis Fontana
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, 18016 Armilla, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| |
Collapse
|
11
|
Charalambous A, Grivogiannis E, Dieronitou I, Michael C, Rahme L, Apidianakis Y. Proteobacteria and Firmicutes Secreted Factors Exert Distinct Effects on Pseudomonas aeruginosa Infection under Normoxia or Mild Hypoxia. Metabolites 2022; 12:449. [PMID: 35629953 PMCID: PMC9146490 DOI: 10.3390/metabo12050449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Microbiota may alter a pathogen's virulence potential at polymicrobial infection sites. Here, we developed a multi-modal Drosophila assay, amenable to the assessment of human bacterial interactions using fly survival or midgut regeneration as a readout, under normoxia or mild hypoxia. Deploying a matrix of 12 by 33 one-to-one Drosophila co-infections via feeding, we classified bacterial interactions as neutral, synergistic, or antagonistic, based on fly survival. Twenty six percent of these interactions were antagonistic, mainly occurring between Proteobacteria. Specifically, Pseudomonas aeruginosa infection was antagonized by various Klebsiella strains, Acinetobacter baumannii, and Escherichia coli. We validated these interactions in a second screen of 7 by 34 one-to-one Drosophila co-infections based on assessments of midgut regeneration, and in bacterial co-culture test tube assays, where antagonistic interactions depended on secreted factors produced upon high sugar availability. Moreover, Enterococci interacted synergistically with P. aeruginosa in flies and in test tubes, enhancing the virulence and pyocyanin production by P. aeruginosa. However, neither lactic acid bacteria nor their severely hypoxic culture supernatants provided a survival benefit upon P. aeruginosa infection of flies or mice, respectively. We propose that at normoxic or mildly hypoxic sites, Firmicutes may exacerbate, whereas Proteobacteria secreted factors may ameliorate, P. aeruginosa infections.
Collapse
Affiliation(s)
- Anna Charalambous
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Evangelos Grivogiannis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Irene Dieronitou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Christina Michael
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| | - Laurence Rahme
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (A.C.); (E.G.); (I.D.); (C.M.)
| |
Collapse
|
12
|
Exploring the Probiotic Potential of Dairy Industrial-Relevant Lactobacilli. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12104989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Usually, the search for new candidate probiotics starts from strain isolation, followed by genotypic and phenotypic characterisations. For the best candidates, the final selection criteria, i.e., an efficient biomass production and the survival to stressful conservation processes, may often represent a bottleneck. The aim of this study is to reverse this classic bottom-up approach, thereby evaluating the in vitro probiotic properties of microbes that are already commercialized and employed in the dairy sector. The major advantage of reversing the traditional scheme is to deal with strains that are already suitable for the scale-up at the industrial level. In this work, four lactobacilli strains were analysed, belonging to the species of Lactiplantibacillus plantarum (strains PLA and PLA2) and Lacticaseibacillus rhamnosus (strains PAR4 and RHM). Both L. plantarum strains showed the best survival under simulated oro-gastrointestinal stress; PLA and PAR4 had the strongest inhibitory activity against all the tested harmful bacteria, with the latter strain showing also the highest percentage of Caco-2 adhesion; RHM was the best biofilm producer on abiotic surface. Finally, cell-free surnatants from all the strain cultures exhibited anti-inflammatory action on THP-1 macrophages. For all the studied strains, it is possible to claim beneficial functional properties other than the technological ones for which they are already marketed. The possible use of the four strains in a mixture could represent a strategy to diversify and maximize their beneficial potential. Nonetheless, future studies are necessary to validate in vivo the observed beneficial properties and to evaluate any effect of the vehicle product on the probiotic aptitude.
Collapse
|
13
|
Lacticaseibacillus rhamnosus: A Suitable Candidate for the Construction of Novel Bioengineered Probiotic Strains for Targeted Pathogen Control. Foods 2022; 11:foods11060785. [PMID: 35327208 PMCID: PMC8947445 DOI: 10.3390/foods11060785] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics, with their associated beneficial effects, have gained popularity for the control of foodborne pathogens. Various sources are explored with the intent to isolate novel robust probiotic strains with a broad range of health benefits due to, among other mechanisms, the production of an array of antimicrobial compounds. One of the shortcomings of these wild-type probiotics is their non-specificity. A pursuit to circumvent this limitation led to the advent of the field of pathobiotechnology. In this discipline, specific pathogen gene(s) are cloned and expressed into a given probiotic to yield a novel pathogen-specific strain. The resultant recombinant probiotic strain will exhibit enhanced species-specific inhibition of the pathogen and its associated infection. Such probiotics are also used as vehicles to deliver therapeutic agents. As fascinating as this approach is, coupled with the availability of numerous probiotics, it brings a challenge with regard to deciding which of the probiotics to use. Nonetheless, it is indisputable that an ideal candidate must fulfil the probiotic selection criteria. This review aims to show how Lacticaseibacillus rhamnosus, a clinically best-studied probiotic, presents as such a candidate. The objective is to spark researchers’ interest to conduct further probiotic-engineering studies using L. rhamnosus, with prospects for the successful development of novel probiotic strains with enhanced beneficial attributes.
Collapse
|
14
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Jastrząb R, Graczyk D, Siedlecki P. Molecular and Cellular Mechanisms Influenced by Postbiotics. Int J Mol Sci 2021; 22:ijms222413475. [PMID: 34948270 PMCID: PMC8707144 DOI: 10.3390/ijms222413475] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, commensal bacteria colonizing the human body have been recognized as important determinants of health and multiple pathologic conditions. Among the most extensively studied commensal bacteria are the gut microbiota, which perform a plethora of functions, including the synthesis of bioactive products, metabolism of dietary compounds, and immunomodulation, both through attenuation and immunostimulation. An imbalance in the microbiota population, i.e., dysbiosis, has been linked to many human pathologies, including various cancer types and neurodegenerative diseases. Targeting gut microbiota and microbiome-host interactions resulting from probiotics, prebiotics, and postbiotics is a growing opportunity for the effective treatment of various diseases. As more research is being conducted, the microbiome field is shifting from simple descriptive analysis of commensal compositions to more molecular, cellular, and functional studies. Insight into these mechanisms is of paramount importance for understanding and modulating the effects that microbiota, probiotics, and their derivatives exert on host health.
Collapse
|
16
|
Díaz‐Garrido N, Badia J, Baldomà L. Microbiota-derived extracellular vesicles in interkingdom communication in the gut. J Extracell Vesicles 2021; 10:e12161. [PMID: 34738337 PMCID: PMC8568775 DOI: 10.1002/jev2.12161] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
The intestine is fundamental in controlling human health. Intestinal epithelial and immune cells are continuously exposed to millions of microbes that greatly impact on intestinal epithelial barrier and immune function. This microbial community, known as gut microbiota, is now recognized as an important partner of the human being that actively contribute to essential functions of the intestine but also of distal organs. In the gut ecosystem, bidirectional microbiota-host communication does not involve direct cell contacts. Both microbiota and host-derived extracellular vesicles (EVs) are key players of such interkingdom crosstalk. There is now accumulating body of evidence that bacterial secreted vesicles mediate microbiota functions by transporting and delivering into host cells effector molecules that modulate host signalling pathways and cell processes. Consequently, vesicles released by the gut microbiota may have great influence on health and disease. Here we review current knowledge on microbiota EVs and specifically highlight their role in controlling host metabolism, intestinal barrier integrity and immune training.
Collapse
Affiliation(s)
- Natalia Díaz‐Garrido
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i FisiologiaFacultat de Farmàcia i Ciències de l'AlimentacióUniversitat de BarcelonaBarcelonaSpain
- Institut de Recerca Sant Joan de Déu (IRSJD)Institut de Biomedicina de la Universitat de Barcelona (IBUB)BarcelonaSpain
| | - Josefa Badia
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i FisiologiaFacultat de Farmàcia i Ciències de l'AlimentacióUniversitat de BarcelonaBarcelonaSpain
- Institut de Recerca Sant Joan de Déu (IRSJD)Institut de Biomedicina de la Universitat de Barcelona (IBUB)BarcelonaSpain
| | - Laura Baldomà
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i FisiologiaFacultat de Farmàcia i Ciències de l'AlimentacióUniversitat de BarcelonaBarcelonaSpain
- Institut de Recerca Sant Joan de Déu (IRSJD)Institut de Biomedicina de la Universitat de Barcelona (IBUB)BarcelonaSpain
| |
Collapse
|
17
|
Bosman-Schluep D, de Pril R, Verbaken B, Legent A, Stallen J, de Jong EC, Janssen RAJ. siRNA-based identification of IBD-related targets in human monocyte-derived dendritic cells. J Immunol Methods 2021; 494:113058. [PMID: 33891922 DOI: 10.1016/j.jim.2021.113058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/31/2021] [Accepted: 04/16/2021] [Indexed: 01/09/2023]
Abstract
Inflammatory bowel disease (IBD) is thought to be caused by an aberrant host response to the commensal enteric flora in genetically susceptible individuals. Dendritic cells (DCs) play a key role in the regulation of this response as they sample gut commensals. In healthy individuals DCs actively contribute to tolerance upon recognition of these resident bacteria, whereas in individuals with IBD, DCs will initiate an inflammatory response. To mimic the disease response in vitro, human monocyte-derived DCs were matured with E. coli causing the cells to produce high levels of the pro-inflammatory cytokine IL-12/IL-23p40 (p40) and low levels of the anti-inflammatory cytokine IL-10. A siRNA-based screening assay was developed and screened to identify potential therapeutic targets that shift this balance towards an immunosuppressive state with lower levels of p40 and higher levels of IL-10. The screening assay was optimized and quality controlled using non-targeting controls and positive control siRNAs targeting IL12B and TLR4 transcripts. In the primary screen, smartpool siRNAs were screened for reduction in p40 expression, induction of IL-10 levels, or increase in IL-10:p40 ratios without affecting cell viability. All potential targets were taken forward into a confirmation screen in a different DC donor in which four individual siRNAs per target were screened. At least two siRNAs per target should have an effect to be considered a valid target. This screen resulted in a concise list of ten genes, of which their role in DC maturation is currently being investigated.
Collapse
Affiliation(s)
| | | | | | | | | | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam UMC, the Netherlands
| | | |
Collapse
|
18
|
Sanborn V, Azcarate-Peril MA, Updegraff J, Manderino L, Gunstad J. Randomized Clinical Trial Examining the Impact of Lactobacillus rhamnosus GG Probiotic Supplementation on Cognitive Functioning in Middle-aged and Older Adults. Neuropsychiatr Dis Treat 2020; 16:2765-2777. [PMID: 33223831 PMCID: PMC7671471 DOI: 10.2147/ndt.s270035] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/02/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE The gut microbiome has been linked to cognitive function and appears to worsen with aging. Probiotic supplementation has been found to improve the health of the gut microbiome. As such, it is possible that probiotic supplementation may protect the aging brain. The current study examined the cognitive benefits of probiotic supplementation (Lactobacillus rhamnosus GG) in healthy middle-aged and older adults. MATERIALS AND METHODS The study was a double-blind, placebo-controlled, randomized clinical trial. Two hundred community-dwelling adults aged 52-75 were enrolled (mean age=64.3, SD=5.52). A three-month intervention involved daily consumption of probiotic or placebo. Independent sample t-tests, chi-squared tests, and repeated measure ANOVAs compared groups and examined changes over time. Primary outcome was change in NIH Toolbox Total Cognition Score from baseline to follow-up. RESULTS A total of 145 participants were examined in primary analyses (probiotic=77, placebo=68) and excluded persons due to discontinuation, low adherence, missing data, or outlier values. Established criteria (ie ≥1 subtest t-scores ≤35; n=19, n=23) were used to operationally define cognitive impairment. Repeated measures ANOVAs revealed that persons with cognitive impairment who consumed probiotics exhibited a greater total cognition score improvement than persons with cognitive impairment in the placebo group and cognitively intact persons in probiotic or placebo groups. CONCLUSION Lactobacillus rhamnosus GG probiotic supplementation was associated with improved cognitive performance in middle-aged and older adults with cognitive impairment. Probiotic supplementation may be a novel method for protecting cognitive health in aging.
Collapse
Affiliation(s)
- Victoria Sanborn
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| | - M Andrea Azcarate-Peril
- Department of Cell Biology and Physiology and Microbiome Core Facility, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - John Updegraff
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| | - Lisa Manderino
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| | - John Gunstad
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
19
|
Lactic acid bacteria secrete toll like receptor 2 stimulating and macrophage immunomodulating bioactive factors. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103783] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
20
|
Xu H, Wang J, Cai J, Feng W, Wang Y, Liu Q, Cai L. Protective Effect of Lactobacillus rhamnosus GG and its Supernatant against Myocardial Dysfunction in Obese Mice Exposed to Intermittent Hypoxia is Associated with the Activation of Nrf2 Pathway. Int J Biol Sci 2019; 15:2471-2483. [PMID: 31595164 PMCID: PMC6775312 DOI: 10.7150/ijbs.36465] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/25/2019] [Indexed: 02/06/2023] Open
Abstract
Prolonged intermittent hypoxia (IH) has been shown to impair myocardial function (mainly via oxidative stress and inflammation) and modify gut microbiota in mice. Gut microbiota plays an important role in health and disease, including obesity and cardiovascular disease (CVD). Probiotics refer to live microorganisms that confer health benefits on the host after administration in adequate amounts. Research on novel probiotics related therapies has evoked much attention. In our previous study, both Lactobacillus rhamnosus GG (LGG) and LGG cell-free supernatant (LGGs) were found to protect against alcohol-induced liver injury and steatosis; however, the effects of LGG and LGGs on cardiac tissues of obese mice exposed to IH have not been determined. Here we exposed high-fat high-fructose diet (HFHFD)-induced obese mice to IH, to establish a model of obesity with obstructive sleep apnea (OSA). Mice were divided into four groups: (1) HFHFD for 15 weeks; (2) HFHFD for 15 weeks with IH in the last 12 weeks (HFHFD/IH); (3) and (4) HFHFD/IH plus oral administration of either LGG (109 CFU bacteria/day) or LGGs (dose equivalent to 109 CFU bacteria/day) over the 15 weeks, respectively. Compared to HFHFD mice, HFHFD/IH-mice showed heart dysfunction with significant cardiac remodeling and inflammation; all these pathological and functional alterations were prevented by treatment with both LGG and LGGs (no significant difference between LGG and LGGs in this respect). The cardioprotective effect of LGG and LGGs against IH/HFHFD was associated with up-regulation of nuclear factor erythroid 2-related factor 2(Nrf2)-mediated antioxidant pathways. Our findings suggest a cardioprotective effect of LGG and LGGs in obese mice with OSA.
Collapse
Affiliation(s)
- Hui Xu
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, 130021 China
- Pediatric Research Institute, Department of Pediatrics, the University of Louisville, Norton Healthcare, Louisville, KY 40202, USA
| | - Jiqun Wang
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, 130021 China
- Pediatric Research Institute, Department of Pediatrics, the University of Louisville, Norton Healthcare, Louisville, KY 40202, USA
| | - Jun Cai
- Pediatric Research Institute, Department of Pediatrics, the University of Louisville, Norton Healthcare, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, the University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Wenke Feng
- Department of Pharmacology and Toxicology, the University of Louisville School of Medicine, Louisville, KY 40202, USA
- Division of Gastroenterology, Department of Medicine, the University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Yonggang Wang
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, 130021 China
| | - Quan Liu
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, 130021 China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, the University of Louisville, Norton Healthcare, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, the University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
21
|
Frickmann H, Klenk C, Warnke P, Redanz S, Podbielski A. Influence of Probiotic Culture Supernatants on In Vitro Biofilm Formation of Staphylococci. Eur J Microbiol Immunol (Bp) 2018; 8:119-127. [PMID: 30719328 PMCID: PMC6348700 DOI: 10.1556/1886.2018.00022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 08/27/2018] [Indexed: 01/15/2023] Open
Abstract
Background The effects of cell-free culture supernatants of probiotic Lactobacillus rhamnosus GG and Streptococcus salivarius K12 on replication and biofilm forming of Staphylococcus aureus and S. epidermidis were assessed in vitro. Methods S. aureus and S. epidermidis strains were exposed to cell-free culture supernatants of L. rhamnosus GG and S. salivarius K12 at different concentrations starting at 0, 4, and 24 h after the onset of incubation. Bacterial amplification was measured on microplate readers, as well as biofilm growth after safranine staining. Scanning electron microscopy was performed for visualization of biofilm status. Results The S. salivarius K12 culture supernatant not only reduced or prevented the formation and maturation of fresh biofilms but even caused a reduction of preformed S. epidermidis biofilms. The L. rhamnosus GG culture supernatant did not show clear inhibitory effects regardless of concentration or time of addition of supernatant, and even concentration-depending promotional effects on the planktonic and biofilm growth of S. aureus and S. epidermidis were observed. Conclusion In particular, the inhibitory effects of the S. salivarius K12 culture supernatant on the formation of staphylococcal biofilms are of potential relevance for biofilm-associated diseases and should be further assessed by in vivo infection models.
Collapse
Affiliation(s)
- Hagen Frickmann
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany.,Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, Hamburg, Germany
| | - Caroline Klenk
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Philipp Warnke
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Sylvio Redanz
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany.,Kreth Lab, Department of Restorative Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Andreas Podbielski
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
22
|
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
|
23
|
Zinöcker MK, Lindseth IA. The Western Diet-Microbiome-Host Interaction and Its Role in Metabolic Disease. Nutrients 2018; 10:E365. [PMID: 29562591 PMCID: PMC5872783 DOI: 10.3390/nu10030365] [Citation(s) in RCA: 427] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/06/2018] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
The dietary pattern that characterizes the Western diet is strongly associated with obesity and related metabolic diseases, but biological mechanisms supporting these associations remain largely unknown. We argue that the Western diet promotes inflammation that arises from both structural and behavioral changes in the resident microbiome. The environment created in the gut by ultra-processed foods, a hallmark of the Western diet, is an evolutionarily unique selection ground for microbes that can promote diverse forms of inflammatory disease. Recognizing the importance of the microbiome in the development of diet-related disease has implications for future research, public dietary advice as well as food production practices. Research into food patterns suggests that whole foods are a common denominator of diets associated with a low level of diet-related disease. Hence, by studying how ultra-processing changes the properties of whole foods and how these foods affect the gut microbiome, more useful dietary guidelines can be made. Innovations in food production should be focusing on enabling health in the super-organism of man and microbe, and stronger regulation of potentially hazardous components of food products is warranted.
Collapse
Affiliation(s)
- Marit K Zinöcker
- Department of Nutrition, Bjørknes University College, Lovisenberggata 13, 0456 Oslo, Norway.
| | | |
Collapse
|
24
|
Wu J, Yang K, Wu W, Tang Q, Zhong Y, Gross G, Lambers TT, van Tol EAF, Cai W. Soluble Mediators From Lactobacillus rhamnosus Gorbach-Goldin Support Intestinal Barrier Function in Rats After Massive Small-Bowel Resection. JPEN J Parenter Enteral Nutr 2018; 42:1026-1034. [PMID: 30133842 DOI: 10.1002/jpen.1044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/18/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal barrier plays an essential role in maintaining gastrointestinal health. This study aimed to explore the effects of a soluble mediator preparation derived from Lactobacillus rhamnosus Gorbach-Goldin (LGG) on intestinal barrier function in a rat model of short bowel syndrome (SBS). METHODS Six-week-old male Sprague-Dawley rats underwent 80% small-bowel resection (SBR) and then were supplemented with water (SBS), 5 × 108 colony-forming unit viable LGG (SBS+LGG), or the LGG soluble mediators (SBS+LSM) in an equivalent dose to LGG by intragastric gavage daily from day 2 throughout day 14 after operation. Rats that underwent bowel transection and reanastomosis were used as the sham group. Body weight, ileum histology, intestinal permeability and bacterial translocation, inflammatory cytokines, and tight junction protein expressions of ileum were evaluated. RESULTS Animals undergoing SBR showed higher intestinal permeability and decreased expression of tight junction proteins in the ileum than sham group. Both SBS+LGG and SBS+LSM groups had reduced bacterial translocation and intestinal permeability as compared with the SBS group, with lower levels of serum endotoxin and tumor necrotizing factor alpha in ileum tissues. Moreover, the SBS+LSM group showed better body weight gain, lower endotoxin and FD-40 levels, and higher expressions of claudin-1 and claudin-4 in ileum than the SBS+LGG group. CONCLUSION Enteral supplementation of LSMs or viable LGG can ameliorate intestinal barrier disruption in a rat model of SBS. The LSM preparation not only mimicked biological effects of viable LGG but also was revealed to be more effective in reducing inflammation and supporting intestinal barrier function.
Collapse
Affiliation(s)
- Jiang Wu
- Department of Clinical Nutrition, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Kefeng Yang
- Department of Nutrition, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjie Wu
- Department of Pediatric Surgery, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingya Tang
- Department of Clinical Nutrition, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yan Zhong
- Mead Johnson Pediatric Nutrition Institute, Shanghai, China
| | - Gabriele Gross
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Tim T Lambers
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Eric A F van Tol
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Wei Cai
- Department of Clinical Nutrition, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Department of Nutrition, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Pediatric Surgery, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Plaza-Díaz J, Ruiz-Ojeda FJ, Gil-Campos M, Gil A. Immune-Mediated Mechanisms of Action of Probiotics and Synbiotics in Treating Pediatric Intestinal Diseases. Nutrients 2018; 10:42. [PMID: 29303974 PMCID: PMC5793270 DOI: 10.3390/nu10010042] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 12/24/2017] [Accepted: 01/03/2018] [Indexed: 12/21/2022] Open
Abstract
The pediatric population is continually at risk of developing infectious and inflammatory diseases. The treatment for infections, particularly gastrointestinal conditions, focuses on oral or intravenous rehydration, nutritional support and, in certain case, antibiotics. Over the past decade, the probiotics and synbiotics administration for the prevention and treatment of different acute and chronic infectious diseases has dramatically increased. Probiotic microorganisms are primarily used as treatments because they can stimulate changes in the intestinal microbial ecosystem and improve the immunological status of the host. The beneficial impact of probiotics is mediated by different mechanisms. These mechanisms include the probiotics' capacity to increase the intestinal barrier function, to prevent bacterial transferation and to modulate inflammation through immune receptor cascade signaling, as well as their ability to regulate the expression of selected host intestinal genes. Nevertheless, with respect to pediatric intestinal diseases, information pertaining to these key mechanisms of action is scarce, particularly for immune-mediated mechanisms of action. In the present work, we review the biochemical and molecular mechanisms of action of probiotics and synbiotics that affect the immune system.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
| | - Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
| | - Mercedes Gil-Campos
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition CB12/03/30028), Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research (IMIBIC), Av. Menendez Pidal s/n, 14010 Córdoba, Spain.
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, 18016 Granada, Spain.
- Instituto de Investigación Biosanitaria ibs GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition CB12/03/30028), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
26
|
Gong J, Bai T, Zhang L, Qian W, Song J, Hou X. Inhibition effect of Bifidobacterium longum, Lactobacillus acidophilus, Streptococcus thermophilus and Enterococcus faecalis and their related products on human colonic smooth muscle in vitro. PLoS One 2017; 12:e0189257. [PMID: 29216305 PMCID: PMC5720742 DOI: 10.1371/journal.pone.0189257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 11/22/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To investigate the effects of four strains, generally used in clinic, including Bifidobacterium longum, Lactobacillus acidophilus, Streptococcus thermophilus and Enterococcus faecalis, and their related products on human colonic smooth muscle in vitro. METHODS Human colonic circular muscle strips obtained from disease-free margins of resected segments from 25 patients with colorectal cancer were isometrically examined in a constant-temperature organ bath and exposed to different concentrations of living bacteria, sonicated cell fractions and cell-free supernatant (CFS). The area under the curve (AUC) representing the contractility of smooth muscle strips was calculated. RESULTS (1) The four living probiotics inhibited the contractility of human colonic muscle strips only at high concentration (1010 CFUs/mL, all P<0.05). (2) The sonicated cell fractions from the four probiotics obviously inhibited human colonic smooth muscle strips in a dose-dependent manner (P<0.01). (3) The CFS from the four probiotics also inhibited colonic smooth muscle strips in a dose-dependent manner (all P<0.05). (4) The inhibition effect of CFS from Streptococcus thermophilus and Enterococcus faecalis decreased obviously when pretreated with NG-nitro-L-arginine (L-NNA, 10-5 mol/L) (P<0.05), but not the Bifidobacterium longum and Lactobacillus acidophilus (P>0.05). CONCLUSION Four common probiotics related products, including the sonicated cell fractions and the CFS, obviously inhibited human colonic smooth muscles strips contraction in a dose-dependent manner. Only high concentration living probiotics (1010 CFUs/mL) can inhibit the colonic smooth muscles strips contraction. The NO pathway may be partly involved in the inhibitory effect of CFS from Streptococcus thermophilus and Enterococcus faecalis.
Collapse
Affiliation(s)
- Jing Gong
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Llewellyn A, Foey A. Probiotic Modulation of Innate Cell Pathogen Sensing and Signaling Events. Nutrients 2017; 9:E1156. [PMID: 29065562 PMCID: PMC5691772 DOI: 10.3390/nu9101156] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 02/07/2023] Open
Abstract
There is a growing body of evidence documenting probiotic bacteria to have a beneficial effect to the host through their ability to modulate the mucosal immune system. Many probiotic bacteria can be considered to act as either immune activators or immune suppressors, which have appreciable influence on homeostasis, inflammatory- and suppressive-immunopathology. What is becoming apparent is the ability of these probiotics to modulate innate immune responses via direct or indirect effects on the signaling pathways that drive these activatory or suppressive/tolerogenic mechanisms. This review will focus on the immunomodulatory role of probiotics on signaling pathways in innate immune cells: from positive to negative regulation associated with innate immune cells driving gut mucosal functionality. Research investigations have shown probiotics to modulate innate functionality in many ways including, receptor antagonism, receptor expression, binding to and expression of adaptor proteins, expression of negative regulatory signal molecules, induction of micro-RNAs, endotoxin tolerisation and finally, the secretion of immunomodulatory proteins, lipids and metabolites. The detailed understanding of the immunomodulatory signaling effects of probiotic strains will facilitate strain-specific selective manipulation of innate cell signal mechanisms in the modulation of mucosal adjuvanticity, immune deviation and tolerisation in both healthy subjects and patients with inflammatory and suppressive pathology.
Collapse
Affiliation(s)
- Amy Llewellyn
- School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine & Dentistry, Drake Circus, Plymouth PL4 8AA, UK.
- Menzies School of Health Research, John Mathews Building (Building 58), Royal Darwin Hospital Campus, PO Box 41096, Casuarina NT0811, Australia.
| | - Andrew Foey
- School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine & Dentistry, Drake Circus, Plymouth PL4 8AA, UK.
| |
Collapse
|
28
|
Ding YH, Qian LY, Pang J, Lin JY, Xu Q, Wang LH, Huang DS, Zou H. The regulation of immune cells by Lactobacilli: a potential therapeutic target for anti-atherosclerosis therapy. Oncotarget 2017; 8:59915-59928. [PMID: 28938693 PMCID: PMC5601789 DOI: 10.18632/oncotarget.18346] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/22/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is an inflammatory disease regulated by several immune cells including lymphocytes, macrophages and dendritic cells. Gut probiotic bacteria like Lactobacilli have been shown immunomodificatory effects in the progression of atherogenesis. Some Lactobacillus stains can upregulate the activity of regulatory T-lymphocytes, suppress T-lymphocyte helper (Th) cells Th1, Th17, alter the Th1/Th2 ratio, influence the subsets ratio of M1/M2 macrophages, inhibit foam cell formation by suppressing macrophage phagocytosis of oxidized low-density lipoprotein, block the activation of the immune system with dendritic cells, which are expected to suppress the atherosclerosis-related inflammation. However, various strains can have various effects on inflammation. Some other Lactobacillus strains were found have potential pro-atherogenic effect through promote Th1 cell activity, increase pro-inflammatory cytokines levels as well as decrease anti-inflammatory cytokines levels. Thus, identifying the appropriate strains is essential to the therapeutic potential of Lactobacilli as an anti-atherosclerotic therapy.
Collapse
Affiliation(s)
- Ya-Hui Ding
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Lin-Yan Qian
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Jie Pang
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Jing-Yang Lin
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Qiang Xu
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Li-Hong Wang
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Dong-Sheng Huang
- People's Hospital of Hangzhou Medical College, Hangzhou 310014, China.,Department of Hepatobiliary Surgery, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310014, Chinaa.,People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
29
|
Plaza-Díaz J, Ruiz-Ojeda FJ, Vilchez-Padial LM, Gil A. Evidence of the Anti-Inflammatory Effects of Probiotics and Synbiotics in Intestinal Chronic Diseases. Nutrients 2017; 9:555. [PMID: 28555037 PMCID: PMC5490534 DOI: 10.3390/nu9060555] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/18/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022] Open
Abstract
Probiotics and synbiotics are used to treat chronic diseases, principally due to their role in immune system modulation and the anti-inflammatory response. The present study reviewed the effects of probiotics and synbiotics on intestinal chronic diseases in in vitro, animal, and human studies, particularly in randomized clinical trials. The selected probiotics exhibit in vitro anti-inflammatory properties. Probiotic strains and cell-free supernatants reduced the expression of pro-inflammatory cytokines via action that is principally mediated by toll-like receptors. Probiotic administration improved the clinical symptoms, histological alterations, and mucus production in most of the evaluated animal studies, but some results suggest that caution should be taken when administering these agents in the relapse stages of IBD. In addition, no effects on chronic enteropathies were reported. Probiotic supplementation appears to be potentially well tolerated, effective, and safe in patients with IBD, in both CD and UC. Indeed, probiotics such as Bifidobacterium longum 536 improved the clinical symptoms in patients with mild to moderate active UC. Although it has been proposed that probiotics can provide benefits in certain conditions, the risks and benefits should be carefully assessed before initiating any therapy in patients with IBD. For this reason, further studies are required to understand the precise mechanism by which probiotics and synbiotics affect these diseases.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada 18071, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, Granada 18016, Spain.
- Instituto de Investigación Biosanitaria ibs., GRANADA, Complejo Hospitalario Universitario de Granada, Granada 18014, Spain.
| | - Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada 18071, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, Granada 18016, Spain.
- Instituto de Investigación Biosanitaria ibs., GRANADA, Complejo Hospitalario Universitario de Granada, Granada 18014, Spain.
| | - Laura Maria Vilchez-Padial
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, Granada 18016, Spain.
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada 18071, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, University of Granada, Armilla, Granada 18016, Spain.
- Instituto de Investigación Biosanitaria ibs., GRANADA, Complejo Hospitalario Universitario de Granada, Granada 18014, Spain.
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain.
| |
Collapse
|
30
|
MacPherson CW, Shastri P, Mathieu O, Tompkins TA, Burguière P. Genome-Wide Immune Modulation of TLR3-Mediated Inflammation in Intestinal Epithelial Cells Differs between Single and Multi-Strain Probiotic Combination. PLoS One 2017; 12:e0169847. [PMID: 28099447 PMCID: PMC5242491 DOI: 10.1371/journal.pone.0169847] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/22/2016] [Indexed: 12/27/2022] Open
Abstract
Genome-wide transcriptional analysis in intestinal epithelial cells (IEC) can aid in elucidating the impact of single versus multi-strain probiotic combinations on immunological and cellular mechanisms of action. In this study we used human expression microarray chips in an in vitro intestinal epithelial cell model to investigate the impact of three probiotic bacteria, Lactobacillus helveticus R0052 (Lh-R0052), Bifidobacterium longum subsp. infantis R0033 (Bl-R0033) and Bifidobacterium bifidum R0071 (Bb-R0071) individually and in combination, and of a surface-layer protein (SLP) purified from Lh-R0052, on HT-29 cells' transcriptional profile to poly(I:C)-induced inflammation. Hierarchical heat map clustering, Set Distiller and String analyses revealed that the effects of Lh-R0052 and Bb-R0071 diverged from those of Bl-R0033 and Lh-R0052-SLP. It was evident from the global analyses with respect to the immune, cellular and homeostasis related pathways that the co-challenge with probiotic combination (PC) vastly differed in its effect from the single strains and Lh-R0052-SLP treatments. The multi-strain PC resulted in a greater reduction of modulated genes, found through functional connections between immune and cellular pathways. Cytokine and chemokine analyses based on specific outcomes from the TNF-α and NF-κB signaling pathways revealed single, multi-strain and Lh-R0052-SLP specific attenuation of the majority of proteins measured (TNF-α, IL-8, CXCL1, CXCL2 and CXCL10), indicating potentially different mechanisms. These findings indicate a synergistic effect of the bacterial combinations relative to the single strain and Lh-R0052-SLP treatments in resolving toll-like receptor 3 (TLR3)-induced inflammation in IEC and maintaining cellular homeostasis, reinforcing the rationale for using multi-strain formulations as a probiotic.
Collapse
Affiliation(s)
- Chad W. MacPherson
- Lallemand Health Solutions Inc., 6100 avenue Royalmount, Montreal, QC, Canada
- * E-mail:
| | - Padmaja Shastri
- University of Ontario Institute of Technology, Oshawa, Canada
| | - Olivier Mathieu
- Lallemand Health Solutions Inc., 6100 avenue Royalmount, Montreal, QC, Canada
| | - Thomas A. Tompkins
- Lallemand Health Solutions Inc., 6100 avenue Royalmount, Montreal, QC, Canada
| | - Pierre Burguière
- Lallemand Health Solutions Inc., 6100 avenue Royalmount, Montreal, QC, Canada
| |
Collapse
|
31
|
Ren C, Zhang Q, de Haan BJ, Zhang H, Faas MM, de Vos P. Identification of TLR2/TLR6 signalling lactic acid bacteria for supporting immune regulation. Sci Rep 2016; 6:34561. [PMID: 27708357 PMCID: PMC5052581 DOI: 10.1038/srep34561] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/13/2016] [Indexed: 02/07/2023] Open
Abstract
Although many lactic acid bacteria (LAB) influence the consumer’s immune status it is not completely understood how this is established. Bacteria-host interactions between bacterial cell-wall components and toll-like receptors (TLRs) have been suggested to play an essential role. Here we investigated the interaction between LABs with reported health effects and TLRs. By using cell-lines expressing single or combination of TLRs, we show that LABs can signal via TLR-dependent and independent pathways. The strains only stimulated and did not inhibit TLRs. We found that several strains such as L. plantarum CCFM634, L. plantarum CCFM734, L. fermentum CCFM381, L. acidophilus CCFM137, and S. thermophilus CCFM218 stimulated TLR2/TLR6. TLR2/TLR6 is essential in immune regulatory processes and of interest for prevention of diseases. Specificity of the TLR2/TLR6 stimulation was confirmed with blocking antibodies. Immunomodulatory properties of LABs were also studied by assessing IL-10 and IL-6 secretion patterns in bacteria-stimulated THP1-derived macrophages, which confirmed species and strain specific effects of the LABs. With this study we provide novel insight in LAB specific host-microbe interactions. Our data demonstrates that interactions between pattern recognition receptors such as TLRs is species and strain specific and underpins the importance of selecting specific strains for promoting specific health effects.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Qiuxiang Zhang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Bart J de Haan
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| | - Hao Zhang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| |
Collapse
|
32
|
Dutra V, Silva AC, Cabrita P, Peres C, Malcata X, Brito L. Lactobacillus plantarum LB95 impairs the virulence potential of Gram-positive and Gram-negative food-borne pathogens in HT-29 and Vero cell cultures. J Med Microbiol 2016; 65:28-35. [DOI: 10.1099/jmm.0.000196] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Virna Dutra
- LEAF (Linking Landscape, Environment, Agriculture and Food)/DRAT (Departamento dos Recursos Naturais, Ambiente e Território), Instituto Superior de Agronomia, University of Lisbon, 1349-017 Lisbon, Portugal
| | - Ana Carla Silva
- LEAF (Linking Landscape, Environment, Agriculture and Food)/DRAT (Departamento dos Recursos Naturais, Ambiente e Território), Instituto Superior de Agronomia, University of Lisbon, 1349-017 Lisbon, Portugal
| | - Paula Cabrita
- LEAF (Linking Landscape, Environment, Agriculture and Food)/DRAT (Departamento dos Recursos Naturais, Ambiente e Território), Instituto Superior de Agronomia, University of Lisbon, 1349-017 Lisbon, Portugal
- Instituto Nacional de Investigação Agrária e Veterinária, IP, Av. República, Quinta do Marquês, Nova Oeiras, 2784-505 Oeiras, Portugal
| | - Cidália Peres
- Instituto Nacional de Investigação Agrária e Veterinária, IP, Av. República, Quinta do Marquês, Nova Oeiras, 2784-505 Oeiras, Portugal
| | - Xavier Malcata
- LEPABE (Laboratory for Process Engineering, Environment, Biotechnology and Energy), Rua Dr Roberto Frias, 4200-264 Porto, Portugal
- Department of Chemical Engineering, University of Porto, Rua Dr Roberto Frias, 4200-264 Porto, Portugal
| | - Luisa Brito
- LEAF (Linking Landscape, Environment, Agriculture and Food)/DRAT (Departamento dos Recursos Naturais, Ambiente e Território), Instituto Superior de Agronomia, University of Lisbon, 1349-017 Lisbon, Portugal
| |
Collapse
|
33
|
Meshkibaf S, Fritz J, Gottschalk M, Kim SO. Preferential production of G-CSF by a protein-like Lactobacillus rhamnosus GR-1 secretory factor through activating TLR2-dependent signaling events without activation of JNKs. BMC Microbiol 2015; 15:238. [PMID: 26502905 PMCID: PMC4623291 DOI: 10.1186/s12866-015-0578-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023] Open
Abstract
Background Different species and strains of probiotic bacteria confer distinct immunological responses on immune cells. Lactobacillus rhamnosus GR-1 (GR-1) is a probiotic bacterial strain found in both the intestinal and urogenital tracts, and has immunomodulatory effects on several cell types including macrophages. However, detailed immunological responses and the signaling mechanism involved in the response are largely unknown. Results We examined the production of GR-1-induced cytokines/chemokines and signaling events in macrophages. Among 84 cytokines and chemokines examined, GR-1 discretely induced granulocyte colony-stimulating factor (G-CSF) mRNA at highest levels (>60-fold) without inducing other cytokines such as IL-1α, IL-1β, IL-6 and TNF-α (<5-fold). The toll-like receptor (TLR) 2/6-agonist PAM2CSK4, TLR2/1-agonist PAM3CSK4 and TLR4-agonist lipopolysaccharide induced all of these inflammatory cytokines at high levels (>50-fold). The TLR2 ligand lipoteichoic acid activated all mitogen-activated kinases, Akt and NF-κB; whereas, GR-1 selectively activated extracellular regulated kinases and p38, NF-κB and Akt, but not c-Jun N-terminal kinases (JNKs) in a TLR2-dependent manner. Using specific inhibitors, we demonstrated that lack of JNKs activation by GR-1 caused inefficient production of pro-inflammatory cytokines but not G-CSF production. A secreted heat-labile protein-like molecule, 30–100 kDa in size, induced the preferential production of G-CSF. Conclusion This study elucidated unique signaling events triggered by GR-1, resulting in selective production of the immunomodulatory cytokine G-CSF in macrophages. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0578-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shahab Meshkibaf
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, Western University, London, ON, N6G 2 V4, Canada. .,Center for Human Immunology, Western University, London, ON, N6G 2 V4, Canada.
| | - Jӧrg Fritz
- Department of Microbiology, McGill University, Montreal, QC, H3G 0B1, Canada.
| | - Marcelo Gottschalk
- Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC, J2S 2 M2, Canada.
| | - Sung Ouk Kim
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, Western University, London, ON, N6G 2 V4, Canada. .,Center for Human Immunology, Western University, London, ON, N6G 2 V4, Canada.
| |
Collapse
|
34
|
Bermudez-Brito M, Sahasrabudhe NM, Rösch C, Schols HA, Faas MM, de Vos P. The impact of dietary fibers on dendritic cell responses in vitro is dependent on the differential effects of the fibers on intestinal epithelial cells. Mol Nutr Food Res 2015; 59:698-710. [PMID: 25620425 DOI: 10.1002/mnfr.201400811] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/15/2015] [Accepted: 01/16/2015] [Indexed: 12/19/2022]
Abstract
SCOPE In the present study, the direct interaction of commonly consumed fibers with epithelial or dendritic cells (DCs) was studied. METHODS AND RESULTS The fibers were characterized for their sugar composition and chain length profile. When in direct contact, fibers activate DCs only mildly. This was different when DCs and fibers were co-cultured together with supernatants from human epithelial cells (Caco spent medium). Caco spent medium enhanced the production of IL-12, IL-1Ra, IL-6, IL-8, TNF-α, MCP-1 (monocyte chemotactic protein), and MIP-1α but this was strongly attenuated by the dietary fibers. This attenuating effect on proinflammatory cytokines was dependent on the interaction of the fibers with Toll-like receptors as it was reduced by Pepinh-myd88. The interaction of galacto-oligosaccharides, chicory inulin, wheat arabinoxylan, barley β-glucan with epithelial cells and DCs led to changes in the production of the Th1 cytokines in autologous T cells, while chicory inulin, and barley β-glucan reduced the Th2 cytokine IL-6. The Treg-promoting cytokine IL-10 was induced by galacto-oligosaccharides whereas chicory inulin decreased the IL-10 production. CONCLUSIONS Our results suggest that dietary fibers can modulate the host immune system not only by the recognized mechanism of effects on microbiota but also by direct interaction with the consumer's mucosa. This modulation is dietary fiber type dependent.
Collapse
Affiliation(s)
- Miriam Bermudez-Brito
- Top Institute Food and Nutrition, Wageningen, The Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
35
|
Yang S, Reid G, Challis JRG, Kim SO, Gloor GB, Bocking AD. Is there a role for probiotics in the prevention of preterm birth? Front Immunol 2015; 6:62. [PMID: 25741339 PMCID: PMC4330906 DOI: 10.3389/fimmu.2015.00062] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/01/2015] [Indexed: 01/12/2023] Open
Abstract
Preterm birth (PTB) continues to be a global health challenge. An over-production of inflammatory cytokines and chemokines, as well as an altered maternal vaginal microbiome has been implicated in the pathogenesis of inflammation/infection-associated PTB. Lactobacillus represents the dominant species in the vagina of most healthy pregnant women. The depletion of Lactobacillus in women with bacterial vaginosis (BV) has been associated with an increased risk of PTB. It remains unknown at what point an aberrant vaginal microbiome composition specifically induces the cascade leading to PTB. The ability of oral or vaginal lactobacilli probiotics to reduce BV occurrence and/or dampen inflammation is being considered as a means to prevent PTB. Certain anti-inflammatory properties of lactobacilli suggest potential mechanisms. To date, clinical studies have not been powered with sufficiently high rates of PTB, but overall, there is merit in examining this promising area of clinical science.
Collapse
Affiliation(s)
- Siwen Yang
- Department of Physiology, Obstetrics and Gynecology, University of Toronto , Toronto, ON , Canada ; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital , Toronto, ON , Canada
| | - Gregor Reid
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Lawson Health Research Institute , London, ON , Canada
| | - John R G Challis
- Department of Physiology, Obstetrics and Gynecology, University of Toronto , Toronto, ON , Canada ; Department of Obstetrics and Gynecology, The University of Western Australia , Perth, WA , Australia
| | - Sung O Kim
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Lawson Health Research Institute , London, ON , Canada
| | - Gregory B Gloor
- Department of Biochemistry, Western University , London, ON , Canada
| | - Alan D Bocking
- Department of Physiology, Obstetrics and Gynecology, University of Toronto , Toronto, ON , Canada ; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital , Toronto, ON , Canada
| |
Collapse
|