1
|
Lam LY, Liang TR, Wu WJ, Lam HYP. Intestinal Lactobacillus johnsonii protects against neuroangiostrongyliasis in BALB/c mice through modulation of immune response. PLoS Negl Trop Dis 2025; 19:e0012977. [PMID: 40198714 PMCID: PMC11978024 DOI: 10.1371/journal.pntd.0012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
Neuroangiostrongyliasis is characterized by eosinophilic meningoencephalitis with a robust onset of severe neurological symptoms, by which immunological factors and peripheral metabolites have been postulated to affect the course of the disease. The gut-brain axis provides a bidirectional communication between the gut and the central nervous system, and therefore, understanding the gut microbiome may provide us with a deeper insight into the pathogenesis of angiostrongyliasis. Using 16S rRNA sequencing, we identified an increase in the abundance of different Lactobacillus species in Angiostrongylus cantonensis-infected mice, which was correlated to the disease severity. However, attempts to inoculate L. johnsonii into A. cantonensis-infected mice surprisingly revealed an improvement in neuroinflammation and prolonged survival. RNA sequencing suggested an immune-modulatory effect of L. johnsonii, which was confirmed by ELISA, showing increased levels of IL-10 and reduced levels of IL-2, IL-4, IL-5, and MCP-1 in the brain. Nevertheless, L. johnsonii-associated improvements were not associated with microbiome-related metabolites, as UHPLC-MS/MS analysis revealed no change in short-chain fatty acids, tryptophan metabolites, and bile acids. Our results suggest that while intestinal L. johnsonii appears to be linked to the progression of neuroangiostrongyliasis, these bacteria are likely attempting to modulate the dysregulated immune response to combat the disease. This is one of the first studies to investigate the gut microbiome in mice with A. cantonensis infection, which extends our knowledge from the microbiome-point-of-view of the pathogenesis of angiostrongyliasis and how the body defends against A. cantonensis. This work also extends to possible treatment approaches using L. johnsonii as probiotics.
Collapse
Affiliation(s)
- Long Yin Lam
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Ting-Ruei Liang
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Wen-Jui Wu
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
2
|
Liao W, Zhang X, Jia C, Chen W, Cai Y, Zhang H, Wei J, Chen T. Lactobacillus rhamnosus LC-STH-13 ameliorates the progression of SLE in MRL/lpr mice by inhibiting the TLR9/NF-κB signaling pathway. Food Funct 2025; 16:475-486. [PMID: 39744924 DOI: 10.1039/d4fo03966a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease often treated with glucocorticoids, which can lead to complications such as osteoporosis and an increased infection risk. Hence, identifying safe and effective treatment strategies is crucial. Lactobacillus has shown promise in improving immune disorders. We investigated Lactobacillus rhamnosus LC-STH-13 for its probiotic properties. Female MRL/lpr mice, prone to lupus, were used to assess its impact on SLE development. The results showed that the intervention with L. rhamnosus LC-STH-13 significantly reduced the level of circulating anti-autoantibodies (p < 0.05) and rebalanced Th17/Treg cells (p < 0.05). Kidney tissue analysis revealed reduced immune cell infiltration and immune complex deposition in glomeruli. L. rhamnosus LC-STH-13 mitigated kidney inflammation via the TLR9/NF-κB pathway (p < 0.05) and attenuated complement-induced renal damage (p < 0.05). Furthermore, 16S rRNA sequencing data analysis indicated that L. rhamnosus LC-STH-13 can restore intestinal microecological imbalance caused by the development of SLE. These findings suggested that L. rhamnosus LC-STH-13 improves the development of SLE by regulating the TLR9/NF-κB pathway and intestinal microbiota, offering a foundation for exploring safe and effective treatments.
Collapse
Affiliation(s)
- Wen Liao
- School of Life Sciences, Nanchang University, Nanchang 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Xinyi Zhang
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Chunjian Jia
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Wenjing Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yujie Cai
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Hongyan Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, China.
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Tingtao Chen
- School of Life Sciences, Nanchang University, Nanchang 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
3
|
Chathuranga K, Shin Y, Uddin MB, Paek J, Chathuranga WAG, Seong Y, Bai L, Kim H, Shin JH, Chang YH, Lee JS. The novel immunobiotic Clostridium butyricum S-45-5 displays broad-spectrum antiviral activity in vitro and in vivo by inducing immune modulation. Front Immunol 2023; 14:1242183. [PMID: 37881429 PMCID: PMC10595006 DOI: 10.3389/fimmu.2023.1242183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Clostridium butyricum is known as a probiotic butyric acid bacterium that can improve the intestinal environment. In this study, we isolated a new strain of C. butyricum from infant feces and evaluated its physiological characteristics and antiviral efficacy by modulating the innate immune responses in vitro and in vivo. The isolated C. butyricum S-45-5 showed typical characteristics of C. butyricum including bile acid resistance, antibacterial ability, and growth promotion of various lactic acid bacteria. As an antiviral effect, C. butyricum S-45-5 markedly reduced the replication of influenza A virus (PR8), Newcastle Disease Virus (NDV), and Herpes Simplex Virus (HSV) in RAW264.7 cells in vitro. This suppression can be explained by the induction of antiviral state in cells by the induction of antiviral, IFN-related genes and secretion of IFNs and pro-inflammatory cytokines. In vivo, oral administration of C. butyricum S-45-5 exhibited prophylactic effects on BALB/c mice against fatal doses of highly pathogenic mouse-adapted influenza A subtypes (H1N1, H3N2, and H9N2). Before challenge with influenza virus, C. butyricum S-45-5-treated BALB/c mice showed increased levels of IFN-β, IFN-γ, IL-6, and IL-12 in serum, the small intestine, and bronchoalveolar lavage fluid (BALF), which correlated with observed prophylactic effects. Interestingly, after challenge with influenza virus, C. butyricum S-45-5-treated BALB/c mice showed reduced levels of pro-inflammatory cytokines and relatively higher levels of anti-inflammatory cytokines at day 7 post-infection. Taken together, these findings suggest that C. butyricum S-45-5 plays an antiviral role in vitro and in vivo by inducing an antiviral state and affects immune modulation to alleviate local and systemic inflammatory responses caused by influenza virus infection. Our study provides the beneficial effects of the new C. butyricum S-45-5 with antiviral effects as a probiotic.
Collapse
Affiliation(s)
- Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yeseul Shin
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Md Bashir Uddin
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medicine, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jayoung Paek
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | | | - Yebin Seong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Lu Bai
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hongik Kim
- Research and Development Division, Vitabio, Inc., Daejeon, Republic of Korea
| | - Jeong Hwan Shin
- Department of Laboratory Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Young-Hyo Chang
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
4
|
Maciel-Fiuza MF, Muller GC, Campos DMS, do Socorro Silva Costa P, Peruzzo J, Bonamigo RR, Veit T, Vianna FSL. Role of gut microbiota in infectious and inflammatory diseases. Front Microbiol 2023; 14:1098386. [PMID: 37051522 PMCID: PMC10083300 DOI: 10.3389/fmicb.2023.1098386] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Thousands of microorganisms compose the human gut microbiota, fighting pathogens in infectious diseases and inhibiting or inducing inflammation in different immunological contexts. The gut microbiome is a dynamic and complex ecosystem that helps in the proliferation, growth, and differentiation of epithelial and immune cells to maintain intestinal homeostasis. Disorders that cause alteration of this microbiota lead to an imbalance in the host’s immune regulation. Growing evidence supports that the gut microbial community is associated with the development and progression of different infectious and inflammatory diseases. Therefore, understanding the interaction between intestinal microbiota and the modulation of the host’s immune system is fundamental to understanding the mechanisms involved in different pathologies, as well as for the search of new treatments. Here we review the main gut bacteria capable of impacting the immune response in different pathologies and we discuss the mechanisms by which this interaction between the immune system and the microbiota can alter disease outcomes.
Collapse
Affiliation(s)
- Miriãn Ferrão Maciel-Fiuza
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil
- Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Guilherme Cerutti Muller
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Daniel Marques Stuart Campos
- Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Perpétua do Socorro Silva Costa
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil
- Department of Nursing, Universidade Federal do Maranhão, Imperatriz, Brazil
| | - Juliano Peruzzo
- Dermatology Service of Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Medicine, Medical Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Renan Rangel Bonamigo
- Dermatology Service of Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Postgraduate Program in Medicine, Medical Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Postgraduate Program in Pathology, Universidade Federal De Ciências Da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Tiago Veit
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Microbiology, Immunology and Parasitology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Sales Luiz Vianna
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil
- Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Postgraduate Program in Medicine, Medical Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- *Correspondence: Fernanda Sales Luiz Vianna,
| |
Collapse
|
5
|
Liu FL, Chen CL, Huang CH. Preparation of fermented oat milk and evaluation of its modulatory effect on antigen-specific immune responses in ovalbumin-sensitized mice. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2120851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Fang-Ling Liu
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| | - Chien-Li Chen
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| | - Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| |
Collapse
|
6
|
Wang Y, Moon A, Huang J, Sun Y, Qiu HJ. Antiviral Effects and Underlying Mechanisms of Probiotics as Promising Antivirals. Front Cell Infect Microbiol 2022; 12:928050. [PMID: 35734576 PMCID: PMC9207339 DOI: 10.3389/fcimb.2022.928050] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics exert a variety of beneficial effects, including maintaining homeostasis and the balance of intestinal microorganisms, activating the immune system, and regulating immune responses. Due to the beneficial effects of probiotics, a wide range of probiotics have been developed as probiotic agents for animal and human health. Viral diseases cause serious economic losses to the livestock every year and remain a great challenge for animals. Moreover, strategies for the prevention and control of viral diseases are limited. Viruses enter the host through the skin and mucosal surface, in which are colonized by hundreds of millions of microorganisms. The antiviral effects of probiotics have been proved, including modulation of chemical, microbial, physical, and immune barriers through various probiotics, probiotic metabolites, and host signaling pathways. It is of great significance yet far from enough to elucidate the antiviral mechanisms of probiotics. The major interest of this review is to discuss the antiviral effects and underlying mechanisms of probiotics and to provide targets for the development of novel antivirals.
Collapse
Affiliation(s)
| | | | | | - Yuan Sun
- *Correspondence: Hua-Ji Qiu, ; Yuan Sun,
| | - Hua-Ji Qiu
- *Correspondence: Hua-Ji Qiu, ; Yuan Sun,
| |
Collapse
|
7
|
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
|
8
|
Cudrania tricuspidata Combined with Lacticaseibacillus rhamnosus Modulate Gut Microbiota and Alleviate Obesity-Associated Metabolic Parameters in Obese Mice. Microorganisms 2021; 9:microorganisms9091908. [PMID: 34576802 PMCID: PMC8468176 DOI: 10.3390/microorganisms9091908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/26/2021] [Accepted: 09/03/2021] [Indexed: 01/09/2023] Open
Abstract
The aim of the presented study was to investigate the synbiotic effects of L. rhamnosus 4B15 and C. tricuspidata extract administration on the gut microbiota and obesity-associated metabolic parameters in diet-induced obese mice. Thirty-one 6-week-old male C57BL/N6 mice were divided into five diet groups: normal diet (ND, n = 7) group; high-fat diet (HFD, n = 6) group; probiotic (PRO, n = 5) group; prebiotic (PRE, n = 7) group; and synbiotic (SYN, n = 6) group. After 10 weeks, the percent of fat mass, serum triglyceride, and ALT levels were significantly reduced in SYN-fed obese mice, compared with other treatments. SYN treatment also modulated the abundance of Desulfovibrio, Dorea, Adlercreutzia, Allobaculum, Coprococcus, unclassified Clostridiaceae, Lactobacillus, Helicobacter, Flexispira, Odoribacter, Ruminococcus, unclassified Erysipelotrichaceae, and unclassified Desulfovibrionaceae. These taxa showed a strong correlation with obesity-associated indices. Lastly, the SYN-supplemented diet upregulated metabolic pathways known to improve metabolic health. Further investigations are needed to understand the mechanisms driving the synbiotic effect of C. tricuspidata and L. rhamnosus 4B15.
Collapse
|
9
|
Ji L, Chen S, Gu G, Zhou J, Wang W, Ren J, Wu J, Yang D, Zheng Y. Exploration of Crucial Mediators for Carotid Atherosclerosis Pathogenesis Through Integration of Microbiome, Metabolome, and Transcriptome. Front Physiol 2021; 12:645212. [PMID: 34108883 PMCID: PMC8181762 DOI: 10.3389/fphys.2021.645212] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Background Carotid atherosclerosis (CAS) is an important cause of stroke. Although interactions between the gut microbiome and metabolome have been widely investigated with respect to the pathogenesis of cardiovascular diseases, information regarding CAS remains limited. Materials and Methods We utilized 16S ribosomal DNA sequencing and untargeted metabolomics to investigate the alterations in the gut microbiota and plasma metabolites of 32 CAS patients and 32 healthy controls. The compositions of the gut microbiota differed significantly between the two groups, and a total of 11 differentially enriched genera were identified. In the metabolomic analysis, 11 and 12 significantly changed metabolites were screened in positive (POS) and negative (NEG) modes, respectively. α-N-Phenylacetyl-L-glutamine was an upregulated metabolite in CAS patients detected in both POS and NEG modes and had the highest | log2(fold change)| in POS mode. In addition, transcriptomic analysis was performed using the GSE43292 dataset. Results A total of 132 differentially expressed genes (DEGs) were screened. Among the upregulated DEGs in CAS patients, FABP4 exhibited the highest | log2(fold change)|. Furthermore, FABP4 was positively associated with Acidaminococcus and had the highest Spearman’s correlation coefficient and the most significant p-value among the microbiota–DEG pairs. Conclusion In this study, we investigated the potential “microbiota–metabolite–gene” regulatory axis that may act on CAS, and our results may help to establish a theoretical basis for further specialized study of this disease.
Collapse
Affiliation(s)
- Lei Ji
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Siliang Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guangchao Gu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiawei Zhou
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wei Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jinrui Ren
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Chinese Academy of Medical Sciences, Peking Union Medical College, Institute of Medicinal Plant Development, Beijing, China
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Talepoor AG, Fouladseresht H, Khosropanah S, Doroudchi M. Immune-Inflammation in Atherosclerosis: A New Twist in an Old Tale. Endocr Metab Immune Disord Drug Targets 2020; 20:525-545. [DOI: 10.2174/1871530319666191016095725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022]
Abstract
Background and Objective:Atherosclerosis, a chronic and progressive inflammatory disease, is triggered by the activation of endothelial cells followed by infiltration of innate and adaptive immune cells including monocytes and T cells in arterial walls. Major populations of T cells found in human atherosclerotic lesions are antigen-specific activated CD4+ effectors and/or memory T cells from Th1, Th17, Th2 and Treg subsets. In this review, we will discuss the significance of T cell orchestrated immune inflammation in the development and progression of atherosclerosis.Discussion:Pathogen/oxidative stress/lipid induced primary endothelial wound cannot develop to a full-blown atherosclerotic lesion in the absence of chronically induced inflammation. While the primary inflammatory response might be viewed as a lone innate response, the persistence of such a profound response over time must be (and is) associated with diverse local and systemic T cell responses. The interplay between T cells and innate cells contributes to a phenomenon called immuneinflammation and has an impact on the progression and outcome of the lesion. In recent years immuneinflammation, an old term, has had a comeback in connecting the puzzle pieces of chronic inflammatory diseases.Conclusion:Taking one-step back and looking from afar at the players of immune-inflammation may help us provide a broader perspective of these complicated interactions. This may lead to the identification of new drug targets and the development of new therapies as well as preventative measures.
Collapse
Affiliation(s)
- Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahdad Khosropanah
- Department of Cardiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Wang G, Zhang M, Zhao J, Xia Y, Lai PFH, Ai L. A Surface Protein From Lactobacillus plantarum Increases the Adhesion of Lactobacillus Strains to Human Epithelial Cells. Front Microbiol 2018; 9:2858. [PMID: 30524417 PMCID: PMC6261977 DOI: 10.3389/fmicb.2018.02858] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022] Open
Abstract
Adhesion to epithelial cells is considered important for Lactobacillus to exert probiotic effects. In this study, we found that trypsin treatment decreased the adhesion ability of Lactobacillus plantarum AR326 and AR269, which exhibit good adhesion ability, and surface proteins extracts increased the adhesion of the strains with poor adhesion ability. By SDS–polyacrylamide gel electrophoresis and mass spectrometry analysis, the main component of the surface proteins was detected and identified as a protein of approximately 37 kDa. It was 100% homologous with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) from L. plantarum WCFS1. The adhesion of AR326 and AR269 was decreased significantly by blocking with the anti-GAPDH antibody, and GAPDH restored the adhesion of AR326 and AR269 treated with trypsin. In addition, purified GAPDH significantly increased the adhesion of the strains with poor adhesion ability. These results indicated that GAPDH mediates the adhesion of these highly adhesive lactobacilli to epithelial cells and can be used to improve the adhesion ability of probiotics or other bacteria of interest.
Collapse
Affiliation(s)
- Guangqiang Wang
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Minghui Zhang
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yongjun Xia
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Phoency F-H Lai
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Lianzhong Ai
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
12
|
Fuglsang E, Krych L, Lundsager MT, Nielsen DS, Frøkiaer H. Postnatal Administration of Lactobacillus rhamnosus HN001 Ameliorates Perinatal Broad-Spectrum Antibiotic-Induced Reduction in Myelopoiesis and T Cell Activation in Mouse Pups. Mol Nutr Food Res 2018; 62:e1800510. [PMID: 30211987 DOI: 10.1002/mnfr.201800510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/03/2018] [Indexed: 12/31/2022]
Abstract
SCOPE This study addresses whether administration of Lactobacillus rhamnosus HN001 could mitigate the effects of a compromised gut microbiota on the composition of mature leukocytes and granulocyte-macrophage progenitor cells (GMPs) in newborn mice. METHODS AND RESULTS Pregnant dams receive oral broad-spectrum antibiotics, which dramatically decrease the gut microbial composition analyzed by 16S rRNA sequencing. Perinatal antibiotic treatment decreases the proportions of bone marrow (BM) GMPs (postnatal day (PND2): 0.5% vs 0.8%, PND4: 0.2% to 0.6%) and mature granulocytes (33% vs 24% at PND2), and spleen granulocytes (7% vs 17% at PND2) and B cells (PND2:18% vs 28%, PND4:11% vs 22%). At PND35, T helper (Th) cells (20% vs 14%) and cytotoxic T (Tc) cells (10% vs 8%) decrease in the spleen. Oral administration of L. rhamnosus HN001 to neonatal pups (PND1-7) restores the antibiotic-induced changes of GMPs and granulocytes in BM and spleen, and further increases splenic granulocytes in control pups. At PND35, splenic proportions of B and Th but not Tc cells are restored. CONCLUSION Postnatal administration of a single bacterial strain efficiently restores granulopoiesis and most T cell activation in neonatal mice that suffer from a perinatal antibiotic-induced compromised gut microbiota at birth.
Collapse
Affiliation(s)
- Eva Fuglsang
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Mia Thorup Lundsager
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Hanne Frøkiaer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
13
|
Zhang Z, Lv J, Pan L, Zhang Y. Roles and applications of probiotic Lactobacillus strains. Appl Microbiol Biotechnol 2018; 102:8135-8143. [PMID: 30032432 DOI: 10.1007/s00253-018-9217-9] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022]
Abstract
Lactobacilli are recognized as probiotics on account of their health-promoting effects in the host. The aim of this review is to summarize current knowledge of the mechanisms of the adaption factors and main functions of lactobacilli that exert health-promoting effects in the host and to discuss important applications in animal and human health. The adaption mechanisms of lactobacilli facilitate interactions with the host and directly contribute to the beneficial nutritional, physiological, microbiological, and immunological effects in the host. Besides, the application of probiotic lactobacilli will increase our understanding of practical uses based on the roles of these organisms in immunoregulation, antipathogenic activities, and enhancement of the epithelial barrier.
Collapse
Affiliation(s)
- Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China. .,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China.,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| |
Collapse
|
14
|
Agah S, Alizadeh AM, Mosavi M, Ranji P, Khavari-Daneshvar H, Ghasemian F, Bahmani S, Tavassoli A. More Protection of Lactobacillus acidophilus Than Bifidobacterium bifidum Probiotics on Azoxymethane-Induced Mouse Colon Cancer. Probiotics Antimicrob Proteins 2018; 11:857-864. [DOI: 10.1007/s12602-018-9425-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Beta-Defensin-2 and Beta-Defensin-3 Reduce Intestinal Damage Caused by Salmonella typhimurium Modulating the Expression of Cytokines and Enhancing the Probiotic Activity of Enterococcus faecium. J Immunol Res 2017; 2017:6976935. [PMID: 29250559 PMCID: PMC5700477 DOI: 10.1155/2017/6976935] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/09/2017] [Accepted: 09/05/2017] [Indexed: 01/11/2023] Open
Abstract
The intestinal microbiota is a major factor in human health and disease. This microbial community includes autochthonous (permanent inhabitants) and allochthonous (transient inhabitants) microorganisms that contribute to maintaining the integrity of the intestinal wall, modulating responses to pathogenic noxae and representing a key factor in the maturation of the immune system. If this healthy microbiota is disrupted by antibiotics, chemotherapy, or a change in diet, intestinal colonization by pathogenic bacteria or viruses may occur, leading to disease. To manage substantial microbial exposure, epithelial surfaces of the intestinal tract produce a diverse arsenal of antimicrobial peptides (AMPs), including, of considerable importance, the β-defensins, which directly kill or inhibit the growth of microorganisms. Based on the literature data, the purpose of this work was to create a line of intestinal epithelial cells able to stably express gene encoding human β-defensin-2 (hBD-2) and human β-defensin-3 (hBD-3), in order to test their role in S. typhimurium infections and their interaction with the bacteria of the gut microbiota.
Collapse
|
16
|
Kwak JY, Lamousé-Smith ESN. Can probiotics enhance vaccine-specific immunity in children and adults? Benef Microbes 2017; 8:657-670. [PMID: 28856905 DOI: 10.3920/bm2016.0147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The growing use of probiotics by the general public has heightened the interest in understanding the role of probiotics in promoting health and preventing disease. General practitioners and specialists often receive inquiries from their patients regarding probiotic products and their use to ward off systemic infection or intestinal maladies. Enhanced immune function is among the touted health benefits conferred by probiotics but has not yet been fully established. Results from recent clinical trials in adults suggest a potential role for probiotics in enhancing vaccine-specific immunity. Although almost all vaccinations are given during infancy and childhood, the numbers of and results from studies using probiotics in pediatric subjects are limited. This review evaluates recent clinical trials of probiotics used to enhance vaccine-specific immune responses in adults and infants. We highlight meaningful results and the implications of these findings for designing translational and clinical studies that will evaluate the potential clinical role for probiotics. We conclude that the touted health claims of probiotics for use in children to augment immunity warrant further investigation. In order to achieve this goal, a consensus should be reached on common study designs that apply similar treatment timelines, compare well-characterised probiotic strains and monitor effective responses against different classes of vaccines.
Collapse
Affiliation(s)
- J Y Kwak
- 1 Department of Pediatrics, Columbia University Medical Center, PH17-105G, 622 West 168th Street, New York, NY 10032, USA
| | - E S N Lamousé-Smith
- 1 Department of Pediatrics, Columbia University Medical Center, PH17-105G, 622 West 168th Street, New York, NY 10032, USA
| |
Collapse
|
17
|
Vanhaecke T, Aubert P, Grohard PA, Durand T, Hulin P, Paul-Gilloteaux P, Fournier A, Docagne F, Ligneul A, Fressange-Mazda C, Naveilhan P, Boudin H, Le Ruyet P, Neunlist M. L. fermentum CECT 5716 prevents stress-induced intestinal barrier dysfunction in newborn rats. Neurogastroenterol Motil 2017; 29. [PMID: 28370715 DOI: 10.1111/nmo.13069] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal epithelial barrier (IEB) dysfunction plays a critical role in various intestinal disorders affecting infants and children, including the development of food allergies and colitis. Recent studies highlighted the role of probiotics in regulating IEB functions and behavior in adults, but their effects in the newborn remain largely unknown. We therefore characterized in rat pups, the impact of Lactobacillus fermentum CECT 5716 (L. fermentum) on stress-induced IEB dysfunction, systemic immune response and exploratory behavior. METHODS Newborn rats received daily by gavage either L. fermentum or water. Intestinal permeability to fluorescein sulfonic acid (FSA) and horseradish peroxidase (HRP) was measured following maternal separation (MS) and water avoidance stress (WAS). Immunohistochemical, transcriptomic, and Western blot analysis of zonula occludens-1 (ZO-1) distribution and expression were performed. Anxiety-like and exploratory behavior was assessed using the elevated plus maze test. Cytokine secretion of activated splenocytes was also evaluated. KEY RESULTS L. fermentum prevented MS and WAS-induced IEB dysfunction in vivo. L. fermentum reduced permeability to both FSA and HRP in the small intestine but not in the colon. L. fermentum increased expression of ZO-1 and prevented WAS-induced ZO-1 disorganization in ileal epithelial cells. L. fermentum also significantly reduced stress-induced increase in plasma corticosteronemia. In activated splenocytes, L. fermentum enhanced IFNγ secretion while it prevented IL-4 secretion. Finally, L. fermentum increased exploratory behavior. CONCLUSIONS & INFERENCES These results suggest that L. fermentum could provide a novel tool for the prevention and/or treatment of gastrointestinal disorders associated with altered IEB functions in the newborn.
Collapse
Affiliation(s)
- T Vanhaecke
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France.,Lactalis Recherche et Développement, Retiers, France
| | - P Aubert
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - P-A Grohard
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - T Durand
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - P Hulin
- Université de Nantes, Nantes, France.,MicroPICell - Cellular and Tissular Imaging Core Facility of Nantes, SFR Santé F. Bonamy-FED 4203/Inserm UMS016/CNRS UMS3556, Nantes, France
| | - P Paul-Gilloteaux
- Université de Nantes, Nantes, France.,MicroPICell - Cellular and Tissular Imaging Core Facility of Nantes, SFR Santé F. Bonamy-FED 4203/Inserm UMS016/CNRS UMS3556, Nantes, France
| | - A Fournier
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders (PhIND), Centre Cyceron, Caen, France
| | - F Docagne
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders (PhIND), Centre Cyceron, Caen, France
| | - A Ligneul
- Lactalis Recherche et Développement, Retiers, France
| | | | - P Naveilhan
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - H Boudin
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - P Le Ruyet
- Lactalis Recherche et Développement, Retiers, France
| | - M Neunlist
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| |
Collapse
|
18
|
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
|
19
|
Lactic acid bacteria as mucosal delivery vehicles: a realistic therapeutic option. Appl Microbiol Biotechnol 2016; 100:5691-701. [DOI: 10.1007/s00253-016-7557-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/11/2022]
|
20
|
Huang IF, Lin IC, Liu PF, Cheng MF, Liu YC, Hsieh YD, Chen JJ, Chen CL, Chang HW, Shu CW. Lactobacillus acidophilus attenuates Salmonella-induced intestinal inflammation via TGF-β signaling. BMC Microbiol 2015; 15:203. [PMID: 26446848 PMCID: PMC4596496 DOI: 10.1186/s12866-015-0546-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 10/02/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Salmonella is a common intestinal pathogen that causes acute and chronic inflammatory response. Probiotics reduce inflammatory cytokine production and serve as beneficial commensal microorganisms in the human gastrointestinal tract. TGF-β (transforming growth factor β)/SMAD and NF-κB signaling play important roles in inflammation in intestinal cells. However, the involvement of the signaling in regulating inflammation between Salmonella and probiotics is not fully understood. METHODS L. acidophilus and prebiotic inulin were used to treat human intestinal Caco-2 cells prior to infection with Salmonella. The cells were harvested to examine the cytokines and MIR21 expression with immunoblotting and real-time PCR. NF-κB and SMAD3/4 reporter vectors were transfected into cells to monitor inflammation and TGF-β1 signaling, respectively. RESULTS In this study, we showed that the probiotic L. acidophilus decreased Salmonella-induced NF-κB activation in human intestinal Caco-2 cells. Expression of the inflammatory cytokines, TNF-α and IL-8, in L. acidophilus-pretreated cells was also significantly lower than that in cells infected with Salmonella alone. Moreover, TGF-β1 and MIR21 expression was elevated in cells pretreated with L. acidophilus or synbiotic, a combination of inulin and L. acidophilus, compared to that in untreated cells or cells infected with S. typhimurium alone. By contrast, expression of SMAD7, a target of MIR21, was accordingly reduced in cells treated with L. acidophilus or synbiotics. Consistent with TGF-β1/MIR21 and SMAD7 expression, SMAD3/4 transcriptional activity was significantly higher in the cells treated with L. acidophilus or synbiotics. Furthermore, TGF-β1 antibody antagonized the SMAD3/4 and NF-κB transcriptional activity modulated by L. acidophilus in intestinal cells. CONCLUSION Our results suggest that the TGF-β1/MIR21 signaling pathway may be involved in the suppressive effects of L. acidophilus on inflammation caused by S. typhimurium in intestinal Caco-2 cells.
Collapse
Affiliation(s)
- I-Fei Huang
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.
- Chung Hwa University of Medical Technology, Tainan, Taiwan.
| | - I-Chun Lin
- Diet and Nutrition Section, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.
| | - Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
| | - Ming-Fang Cheng
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Yen-Chen Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
| | - Yao-Dung Hsieh
- Department of Dentistry, Kaohsiung Veterans General Hospital, Pingtung, Taiwan.
| | - Jih-Jung Chen
- Department of Pharmacy, Tajen University, Pingtung, Taiwan.
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
21
|
Ivanovic N, Minic R, Dimitrijevic L, Radojevic Skodric S, Zivkovic I, Djordjevic B. Lactobacillus rhamnosus LA68 and Lactobacillus plantarum WCFS1 differently influence metabolic and immunological parameters in high fat diet-induced hypercholesterolemia and hepatic steatosis. Food Funct 2015; 6:558-65. [DOI: 10.1039/c4fo00843j] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
TwoLactobacillusstrains were evaluated for their effects on high fat diet induced pathology in mice.
Collapse
Affiliation(s)
- Nevena Ivanovic
- Department of Bromatology
- Faculty of Pharmacy
- University of Belgrade
- Belgrade
- Serbia
| | - Rajna Minic
- Department of Research and Development
- Institute of Virology
- Belgrade
- Serbia
| | | | | | - Irena Zivkovic
- Department of Research and Development
- Institute of Virology
- Belgrade
- Serbia
| | - Brizita Djordjevic
- Department of Bromatology
- Faculty of Pharmacy
- University of Belgrade
- Belgrade
- Serbia
| |
Collapse
|