1
|
Saeed A, Yasmin A, Baig M, Ahmed MA, Farooqi ZUR. Streptococcus lactarius MB622 and Streptococcus salivarius MB620 isolated from human milk reduce chemokine IL-8 production in response to TNF-α in Caco-2 cell line, an exploratory study. Cytokine 2023; 168:156232. [PMID: 37224578 DOI: 10.1016/j.cyto.2023.156232] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023]
Abstract
Streptococci are a predominant genera of the human milk microbiome. Among different lactic acid bacteria (LAB) a few Streptococcal strains are also considered as probiotics. Probiotic bacteria are reported to modulate immunity when consumed in adequate amount and bacterial hydrophobicity can be considered as a preliminary experiment for the adhesive capability of probiotic bacteria to the epithelial cells. The present study aimed to investigate the probiotic, hydrophobic and immune modulation property of Streptococcus lactarius MB622 and Streptococcus salivarius MB620, isolated from human milk. S. lactarius MB622 and S. salivarius MB620 displayed higher hydrophobicity (78 % and 59 % respectively) in addition to intrinsic probiotic properties such as gram positive classification, catalase negative activity, resistance to artificially stimulated gastric juice and gastrointestinal bile salt concentration. In conclusion Streptococcus lactarius MB622 and Streptococcus salivarius MB620 isolated from human milk when administered in sufficient amount and for certain duration could be used to reduce inflammation inside the colon by reducing the production of inflammatory booster (IL-8) in diseased state.
Collapse
Affiliation(s)
- Ayesha Saeed
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University Rawalpindi, Pakistan.
| | - Azra Yasmin
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University Rawalpindi, Pakistan
| | - Mehreen Baig
- Surgical Unit II, Foundation University Islamabad, Pakistan
| | | | | |
Collapse
|
2
|
Rüb AM, Tsakmaklis A, Gräfe SK, Simon MC, Vehreschild MJ, Wuethrich I. Biomarkers of human gut microbiota diversity and dysbiosis. Biomark Med 2021; 15:137-148. [PMID: 33442994 DOI: 10.2217/bmm-2020-0353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
The association of gut microbiota dysbiosis with various human diseases is being substantiated with increasing evidence. Metabolites derived from both, microbiota and the human host play a central role in disease susceptibility and disease progression by extensively modulating host physiology and metabolism. Several of these metabolites have the potential to serve as diagnostic biomarkers for monitoring disease states in conjunction with intestinal microbiota dysbiosis. In this narrative review we evaluate the potential of trimethylamine-N-oxide, short-chain fatty acids, 3-indoxyl sulfate, p-cresyl sulfate, secondary bile acids, hippurate, human β-defensin-2, chromogranin A, secreted immunoglobulins and zonulin to serve as biomarkers for metabolite profiling and diagnostic suitability for dysbiosis and disease.
Collapse
Affiliation(s)
- Alina M Rüb
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Anastasia Tsakmaklis
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Stefanie K Gräfe
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Marie-Christine Simon
- Department of Nutrition & Food Sciences, Nutrition & Microbiota, University of Bonn, Bonn, Germany
| | - Maria Jgt Vehreschild
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Irene Wuethrich
- Department of Biosystems Science & Engineering, ETH Zurich, Basel, Switzerland
| |
Collapse
|
3
|
Wan MLY, Forsythe SJ, El-Nezami H. Probiotics interaction with foodborne pathogens: a potential alternative to antibiotics and future challenges. Crit Rev Food Sci Nutr 2018; 59:3320-3333. [PMID: 29993263 DOI: 10.1080/10408398.2018.1490885] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antibiotics are a key tool used nowadays in health care industry to fight against bacterial infections; however, repeated antibiotic use or misuses, have led to bacterial resistance, causing significant threats for many people with common bacterial infections. The use of probiotics to enhance gastrointestinal health has been proposed for many years. In recent years, there has been an increasing interest in the use of probiotic bacteria as alternatives for antibiotics for preventing or treating various intestinal infections. Several important underlying mechanisms responsible for the antagonistic effects of probiotics on different microorganisms include: (1) competitive exclusion for adhesion sites and nutritional sources; (2) secretion of antimicrobial substances; (3) enhancement of intestinal barrier function; and (4) immunomodulation. However, their mode of action is not very well understood and therefore a clearer understanding of these mechanisms is necessitated. This will enable appropriate probiotic strains to be selected for particular applications and may reveal new probiotic functions. The goal of this review was to highlight some studies from literature describing the probiotic interaction with several major foodborne pathogens, as well as explore the mechanisms for such probiotic-pathogen interaction. The review will conclude by presenting future perspective and challenges of probiotic application in food products.
Collapse
Affiliation(s)
- Murphy Lam Yim Wan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong S.A.R
| | | | - Hani El-Nezami
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong S.A.R.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
4
|
Dias Bastos PA, Lara Santos L, Pinheiro Vitorino RM. How are the expression patterns of gut antimicrobial peptides modulated by human gastrointestinal diseases? A bridge between infectious, inflammatory, and malignant diseases. J Pept Sci 2018. [PMID: 29542263 DOI: 10.1002/psc.3071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The human gut barrier is the tissue exposed to the highest load of microorganisms, harbouring 100 trillion bacteria. In addition, the gut's renewal rate outruns that of any other human tissue. Antimicrobial peptides (AMPs) are highly optimized defense molecules in the intestinal barrier optimized to maintain gastrointestinal homeostasis. Alterations in AMPs activity can lead to or result from human gastrointestinal diseases. In this review, unique, conserved, or otherwise regular alterations in the expression patterns of human AMPs across gastrointestinal inflammatory and infectious diseases were analyzed for pattern elucidation. Human gastrointestinal diseases are associated with alterations in gut AMPs' expression patterns in a peptide-specific, disease-specific, and pathogen-specific way, modulating human gastrointestinal functioning. Across diseases, there is a (i) marked reduction in otherwise constitutively expressed AMPs, leading to increased disease susceptibility, and a (ii) significant increase in the expression of inducible AMPs, leading to tissue damage and disease severity. Infections and inflammatory conditions are associated with altered gene expression in the gut, whose patterns may favour cellular metaplasia, mucosal dysfunction, and disease states. Altered expression of AMPs can thus thrive disease severity and evolution since its early stages. Nevertheless, the modulation of AMP expression patterns unveils promising therapeutic targets.
Collapse
Affiliation(s)
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Oncology Institute - Porto (IPO-Porto), Porto, Portugal.,Department of Surgical Oncology, Portuguese Oncology Institute - Porto (IPO-Porto), Porto, Portugal
| | - Rui Miguel Pinheiro Vitorino
- iBiMED, Institute for Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
5
|
Vijayan A, Rumbo M, Carnoy C, Sirard JC. Compartmentalized Antimicrobial Defenses in Response to Flagellin. Trends Microbiol 2018; 26:423-435. [PMID: 29173868 DOI: 10.1016/j.tim.2017.10.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/20/2017] [Accepted: 10/27/2017] [Indexed: 11/19/2022]
Abstract
Motility is often a pathogenicity determinant of bacteria targeting mucosal tissues. Flagella constitute the machinery that propels bacteria into appropriate niches. Besides motility, the structural component, flagellin, which forms the flagella, targets Toll-like receptor 5 (TLR5) to activate innate immunity. The compartmentalization of flagellin-mediated immunity and the contribution of epithelial cells and dendritic cells in detecting flagellin within luminal and basal sides are highlighted here, respectively. While a direct stimulation of the epithelium mainly results in recruitment of immune cells and production of antimicrobial molecules, TLR5 engagement on parenchymal dendritic cells can contribute to the stimulation of innate lymphocytes such as type 3 innate lymphoid cells, as well as T helper cells. This review, therefore, illustrates how the innate and adaptive immunity to flagellin are differentially regulated by the epithelium and the dendritic cells in response to pathogens that either colonize or invade mucosa.
Collapse
Affiliation(s)
- Aneesh Vijayan
- Université Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Martin Rumbo
- Instituto de Estudios Inmunológicos y Fisiopatológicos - CONICET - National Universtity of La Plata, 1900 La Plata, Argentina
| | - Christophe Carnoy
- Université Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France.
| | - Jean-Claude Sirard
- Université Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France.
| |
Collapse
|
6
|
Wang KC, Huang CH, Ding SM, Chen CK, Fang HW, Huang MT, Fang SB. Role of yqiC in the Pathogenicity of Salmonella and Innate Immune Responses of Human Intestinal Epithelium. Front Microbiol 2016; 7:1614. [PMID: 27777572 PMCID: PMC5056187 DOI: 10.3389/fmicb.2016.01614] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/27/2016] [Indexed: 01/12/2023] Open
Abstract
The yqiC gene of Salmonella enterica serovar Typhimurium (S. Typhimurium) regulates bacterial growth at different temperatures and mice survival after infection. However, the role of yqiC in bacterial colonization and host immunity remains unknown. We infected human LS174T, Caco-2, HeLa, and THP-1 cells with S. Typhimurium wild-type SL1344, its yqiC mutant, and its complemented strain. Bacterial colonization and internalization in the four cell lines significantly reduced on yqiC depletion. Post-infection production of interleukin-8 and human β-defensin-3 in LS174T cells significantly reduced because of yqiC deleted in S. Typhimurium. The phenotype of yqiC mutant exhibited few and short flagella, fimbriae on the cell surface, enhanced biofilm formation, upregulated type-1 fimbriae expression, and reduced bacterial motility. Type-1 fimbriae, flagella, SPI-1, and SPI-2 gene expression was quantified using real-time PCR. The data show that deletion of yqiC upregulated fimA and fimZ expression and downregulated flhD, fliZ, invA, and sseB expression. Furthermore, thin-layer chromatography and high-performance liquid chromatography revealed the absence of menaquinone in the yqiC mutant, thus validating the importance of yqiC in the bacterial electron transport chain. Therefore, YqiC can negatively regulate FimZ for type-1 fimbriae expression and manipulate the functions of its downstream virulence factors including flagella, SPI-1, and SPI-2 effectors.
Collapse
Affiliation(s)
- Ke-Chuan Wang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - Chih-Hung Huang
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology Taipei, Taiwan
| | - Shih-Min Ding
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of TechnologyTaipei, Taiwan
| | - Ching-Kuo Chen
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology Taipei, Taiwan
| | - Hsu-Wei Fang
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of TechnologyTaipei, Taiwan; Institute of Biomedical Engineering and Nanomedicine - National Health Research InstitutesZhunan, Taiwan
| | - Ming-Te Huang
- Department of Surgery, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - Shiuh-Bin Fang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| |
Collapse
|
7
|
Parks OB, Pociask DA, Hodzic Z, Kolls JK, Good M. Interleukin-22 Signaling in the Regulation of Intestinal Health and Disease. Front Cell Dev Biol 2016; 3:85. [PMID: 26793707 PMCID: PMC4710696 DOI: 10.3389/fcell.2015.00085] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/14/2015] [Indexed: 12/25/2022] Open
Abstract
Interleukin (IL)-22 is a member of the IL-10 family of cytokines that has been extensively studied since its discovery in 2000. This review article aims to describe the cellular sources and signaling pathways of this cytokine as well as the functions of IL-22 in the intestine. In addition, this article describes the roles of IL-22 in the pathogenesis of several gastrointestinal diseases, including inhibition of inflammation and barrier defense against pathogens within the intestine. Since many of the functions of IL-22 in the intestine are incompletely understood, this review is meant to assess our current understanding of the roles of IL-22 and provide new opportunities for inquiry to improve human intestinal health and disease.
Collapse
Affiliation(s)
- Olivia B Parks
- Department of Pediatrics, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Derek A Pociask
- Department of Pediatrics, University of Pittsburgh School of MedicinePittsburgh, PA, USA; Department of Pediatrics, Richard King Mellon Foundation Institute for Pediatric Research, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Zerina Hodzic
- Department of Pediatrics, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Jay K Kolls
- Department of Pediatrics, University of Pittsburgh School of MedicinePittsburgh, PA, USA; Department of Pediatrics, Richard King Mellon Foundation Institute for Pediatric Research, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Misty Good
- Department of Pediatrics, University of Pittsburgh School of MedicinePittsburgh, PA, USA; Division of Newborn Medicine, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| |
Collapse
|
8
|
Kim Y, Koh JH, Ahn YJ, Oh S, Kim SH. The Synergic Anti-inflammatory Impact of Gleditsia sinensis Lam. and Lactobacillus brevis KY21 on Intestinal Epithelial Cells in a DSS-induced Colitis Model. Korean J Food Sci Anim Resour 2015; 35:604-10. [PMID: 26761887 PMCID: PMC4670888 DOI: 10.5851/kosfa.2015.35.5.604] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/24/2015] [Accepted: 07/24/2015] [Indexed: 01/02/2023] Open
Abstract
We investigated the synergic anti-inflammatory activity of Gleditsia sinensis Lam. (GS) extract and Lactobacillus brevis KY21 both in vitro and in vivo. Western blot analysis and immunostaining showed that AKT phosphorylation that increased by the exposure of LPS were significantly decreased by the presence of either GS extract or L. brevis KY21. In addition, p65 intracellular transport was critically inhibited by GS extract and L. brevis KY21. We further studied these effects using an in vivo dextran sulfate sodium (DSS)-induced mouse model. Body weight, food intake, and clinical scores were dramatically decreased after treatment with DSS, whereas these effects were palliated by the addition of GS extract and L. brevis KY21. Importantly, transcription of genes encoding pro-inflammatory cytokines including IL-1β, TNF-α, and IFN-γ in mesenteric lymph nodes (MLN) and the spleen were increased by DSS treatment, whereas they were inhibited by the presence of GS extract and L. brevis KY21.
Collapse
Affiliation(s)
- Younghoon Kim
- BK21 Plus Graduate Program, Department of Animal Science and Institute Agricultural Science & Technology, Chonbuk National University, Jeonju 54896, Korea
| | - Ji Hoon Koh
- Division of Food Bioscience and Technology, Korea University, Seoul 02841, Korea
| | - Young Jun Ahn
- Division of Food Bioscience and Technology, Korea University, Seoul 02841, Korea
| | - Sejong Oh
- Division of Animal Science, Chonnam National University, Gwangju 61186, Korea
| | - Sea Hun Kim
- Division of Food Bioscience and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
9
|
Induction of human β-defensin-2 in airway epithelial cells by Streptococcus pneumoniae: involvement of IP3-dependent intracellular calcium release and NF-κB. CHINESE SCIENCE BULLETIN-CHINESE 2014. [DOI: 10.1007/s11434-014-0201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
10
|
Pathogenesis of human enterovirulent bacteria: lessons from cultured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev 2014; 77:380-439. [PMID: 24006470 DOI: 10.1128/mmbr.00064-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hosts are protected from attack by potentially harmful enteric microorganisms, viruses, and parasites by the polarized fully differentiated epithelial cells that make up the epithelium, providing a physical and functional barrier. Enterovirulent bacteria interact with the epithelial polarized cells lining the intestinal barrier, and some invade the cells. A better understanding of the cross talk between enterovirulent bacteria and the polarized intestinal cells has resulted in the identification of essential enterovirulent bacterial structures and virulence gene products playing pivotal roles in pathogenesis. Cultured animal cell lines and cultured human nonintestinal, undifferentiated epithelial cells have been extensively used for understanding the mechanisms by which some human enterovirulent bacteria induce intestinal disorders. Human colon carcinoma cell lines which are able to express in culture the functional and structural characteristics of mature enterocytes and goblet cells have been established, mimicking structurally and functionally an intestinal epithelial barrier. Moreover, Caco-2-derived M-like cells have been established, mimicking the bacterial capture property of M cells of Peyer's patches. This review intends to analyze the cellular and molecular mechanisms of pathogenesis of human enterovirulent bacteria observed in infected cultured human colon carcinoma enterocyte-like HT-29 subpopulations, enterocyte-like Caco-2 and clone cells, the colonic T84 cell line, HT-29 mucus-secreting cell subpopulations, and Caco-2-derived M-like cells, including cell association, cell entry, intracellular lifestyle, structural lesions at the brush border, functional lesions in enterocytes and goblet cells, functional and structural lesions at the junctional domain, and host cellular defense responses.
Collapse
|
11
|
Muniz LR, Knosp C, Yeretssian G. Intestinal antimicrobial peptides during homeostasis, infection, and disease. Front Immunol 2012; 3:310. [PMID: 23087688 PMCID: PMC3466489 DOI: 10.3389/fimmu.2012.00310] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/17/2012] [Indexed: 12/25/2022] Open
Abstract
Antimicrobial peptides (AMPs), including defensins and cathelicidins, constitute an arsenal of innate regulators of paramount importance in the gut. The intestinal epithelium is exposed to myriad of enteric pathogens and these endogenous peptides are essential to fend off microbes and protect against infections. It is becoming increasingly evident that AMPs shape the composition of the commensal microbiota and help maintain intestinal homeostasis. They contribute to innate immunity, hence playing important functions in health and disease. AMP expression is tightly controlled by the engagement of pattern recognition receptors (PRRs) and their impairment is linked to abnormal host responses to infection and inflammatory bowel diseases (IBD). In this review, we provide an overview of the mucosal immune barriers and the intricate crosstalk between the host and the microbiota during homeostasis. We focus on the AMPs and pay particular attention to how PRRs promote their secretion in the intestine. Furthermore, we discuss their production and main functions in three different scenarios, at steady state, throughout infection with enteric pathogens and IBD.
Collapse
Affiliation(s)
- Luciana R Muniz
- Department of Medicine, Immunology Institute, Mount Sinai School of Medicine New York, NY, USA
| | | | | |
Collapse
|
12
|
Bermudez-Brito M, Plaza-Díaz J, Muñoz-Quezada S, Gómez-Llorente C, Gil A. Probiotic mechanisms of action. ANNALS OF NUTRITION AND METABOLISM 2012; 61:160-74. [PMID: 23037511 DOI: 10.1159/000342079] [Citation(s) in RCA: 631] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 12/28/2022]
Abstract
Probiotics are live microorganisms that provide health benefits to the host when ingested in adequate amounts. The strains most frequently used as probiotics include lactic acid bacteria and bifidobacteria. Probiotics have demonstrated significant potential as therapeutic options for a variety of diseases, but the mechanisms responsible for these effects have not been fully elucidated yet. Several important mechanisms underlying the antagonistic effects of probiotics on various microorganisms include the following: modification of the gut microbiota, competitive adherence to the mucosa and epithelium, strengthening of the gut epithelial barrier and modulation of the immune system to convey an advantage to the host. Accumulating evidence demonstrates that probiotics communicate with the host by pattern recognition receptors, such as toll-like receptors and nucleotide-binding oligomerization domain-containing protein-like receptors, which modulate key signaling pathways, such as nuclear factor-ĸB and mitogen-activated protein kinase, to enhance or suppress activation and influence downstream pathways. This recognition is crucial for eliciting measured antimicrobial responses with minimal inflammatory tissue damage. A clear understanding of these mechanisms will allow for appropriate probiotic strain selection for specific applications and may uncover novel probiotic functions. The goal of this systematic review was to explore probiotic modes of action focusing on how gut microbes influence the host.
Collapse
Affiliation(s)
- Miriam Bermudez-Brito
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology José Mataix, Biomedical Research Center, University of Granada, Armilla, Spain
| | | | | | | | | |
Collapse
|
13
|
Kim JY, Park MS, Ji GE. Probiotic modulation of dendritic cells co-cultured with intestinal epithelial cells. World J Gastroenterol 2012; 18:1308-18. [PMID: 22493544 PMCID: PMC3319957 DOI: 10.3748/wjg.v18.i12.1308] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 09/30/2011] [Accepted: 01/22/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate cytokine production and cell surface phenotypes of dendritic cells (DC) in the presence of epithelial cells stimulated by probiotics.
METHODS: Mouse DC were cultured alone or together with mouse epithelial cell monolayers in normal or inverted systems and were stimulated with heat-killed probiotic bacteria, Bifidobacterium lactis AD011 (BL), Bifidobacterium bifidum BGN4 (BB), Lactobacillus casei IBS041 (LC), and Lactobacillus acidophilus AD031 (LA), for 12 h. Cytokine levels in the culture supernatants were determined by enzyme-linked immunosorbent assay and phenotypic analysis of DC was investigated by flow cytometry.
RESULTS: BB and LC in single-cultured DC increased the expression of I-Ad, CD86 and CD40 (I-Ad, 18.51 vs 30.88, 46.11; CD86, 62.74 vs 92.7, 104.12; CD40, 0.67 vs 6.39, 3.37, P < 0.05). All of the experimental probiotics increased the production of inflammatory cytokines, interleukin (IL)-6 and tumor necrosis factor (TNF)-α. However, in the normal co-culture systems, LC and LA decreased the expression of I-Ad (39.46 vs 30.32, 33.26, P < 0.05), and none of the experimental probiotics increased the levels of IL-6 or TNF-α. In the inverted co-culture systems, LC decreased the expression of CD40 (1.36 vs -2.27, P < 0.05), and all of the experimental probiotics decreased the levels of IL-6. In addition, BL increased the production of IL-10 (103.8 vs 166.0, P < 0.05) and LC and LA increased transforming growth factor-β secretion (235.9 vs 618.9, 607.6, P < 0.05).
CONCLUSION: These results suggest that specific probiotic strains exert differential immune modulation mediated by the interaction of dendritic cells and epithelial cells in the homeostasis of gastrointestinal tract.
Collapse
|
14
|
Gupta S, Ghosh SK, Scott ME, Bainbridge B, Jiang B, Lamont RJ, McCormick TS, Weinberg A. Fusobacterium nucleatum-associated beta-defensin inducer (FAD-I): identification, isolation, and functional evaluation. J Biol Chem 2010; 285:36523-31. [PMID: 20847052 DOI: 10.1074/jbc.m110.133140] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Human β-defensins (hBDs) are small, cationic antimicrobial peptides, secreted by mucosal epithelial cells that regulate adaptive immune functions. We previously reported that Fusobacterium nucleatum, a ubiquitous gram-negative bacterium of the human oral cavity, induces human β-defensin 2 (hBD2) upon contact with primary oral epithelial cells. We now report the isolation and characterization of an F. nucleatum (ATCC 25586)-associated defensin inducer (FAD-I). Biochemical approaches revealed a cell wall fraction containing four proteins that stimulated the production of hBD2 in human oral epithelial cells (HOECs). Cross-referencing of the N-terminal sequences of these proteins with the F. nucleatum genome revealed that the genes encoding the proteins were FadA, FN1527, FN1529, and FN1792. Quantitative PCR of HOEC monolayers challenged with Escherichia coli clones expressing the respective cell wall proteins revealed that FN1527 was most active in the induction of hBD2 and hence was termed FAD-I. We tagged FN1527 with a c-myc epitope on the C-terminal end to identify and purify it from the E. coli clone. Purified FN1527 (FAD-I) induced hBD2 mRNA and protein expression in HOEC monolayers. F. nucleatum cell wall and FAD-I induced hBD2 via TLR2. Porphorymonas gingivalis, an oral pathogen that does not induce hBD2 in HOECs, was able to significantly induce expression of hBD2 in HOECs only when transformed to express FAD-I. FAD-I or its derivates offer a potentially new paradigm in immunoregulatory therapeutics because they may one day be used to bolster the innate defenses of vulnerable mucosae.
Collapse
Affiliation(s)
- Sanhita Gupta
- Department of Biological Science, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Liu C, Zhang ZY, Dong K, Guo XK. Adhesion and immunomodulatory effects of Bifidobacterium lactis HN019 on intestinal epithelial cells INT-407. World J Gastroenterol 2010; 16:2283-90. [PMID: 20458767 PMCID: PMC2868223 DOI: 10.3748/wjg.v16.i18.2283] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To elucidate the adherence and immunomodulatory properties of a probiotic strain Bifidobacterium lactis (B. lactis) HN019.
METHODS: Adhesion assays of B. lactis HN019 and Salmonella typhimurium (S. typhimurium) ATCC 14028 to INT-407 cells were carried out by detecting copies of species-specific genes with real-time polymerase chain reaction. Morphological study was further conducted by transmission electron microscopy. Interleukin-1β (IL-1β), interleukin-8, and tumor necrosis factor-α (TNF-α) gene expression were assessed while enzyme linked immunosorbent assay was used to detect IL-8 protein secretion.
RESULTS: The attachment of S. typhimurium ATCC 14028 to INT407 intestinal epithelial cells was inhibited significantly by B. lactis HN019. B. lactis HN019 could be internalized into the INT-407 cells and attenuated IL-8 mRNA level at both baseline and S. typhimurium-induced pro-inflammatory responses. IL-8 secretion was reduced while IL-1β and TNF-α mRNA expression level remained unchanged at baseline after treated with B. lactis HN019.
CONCLUSION: B. lactis HN019 does not up-regulate the intestinal epithelium expressed pro-inflammatory cytokine, it showed the potential to protect enterocytes from an acute inflammatory response induced by enteropathogen.
Collapse
|
16
|
Han SH, Kim YE, Park JA, Park JB, Kim YS, Lee Y, Choi IG, Kwon HJ. Expression of human beta-defensin-2 gene induced by CpG-DNA in human B cells. Biochem Biophys Res Commun 2009; 389:443-8. [PMID: 19732743 DOI: 10.1016/j.bbrc.2009.08.162] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 08/29/2009] [Indexed: 11/20/2022]
Abstract
Defensins have a broad range of antimicrobial activity against bacteria, fungi, and viruses. The expression of human beta-defensin-2 (hBD-2) is prevalently observed in epithelial cells and is induced by bacterial infection. Here, we have shown that the expression of the hBD-2 gene and release of hBD-2 protein into the medium is up-regulated in response to CpG-DNA in human B cell line RPMI 8226. The induction of hBD-2 was dependent on CG sequence and phosphorothioate backbone-modification. This was also confirmed in primary human lymphocytes. To shed light on the molecular mechanism involved in hBD-2 induction by CpG-DNA, we examined the contribution of the NF-kappaB signaling pathway in RPMI 8226 cells. Suppression of MyD88 function and inhibition of NF-kappaB nuclear localization blocked hBD-2 induction. The NF-kappaB pathway inhibitors also abolished hBD-2 induction. These results may contribute to a better understanding on the therapeutic effects of CpG-DNA against infectious diseases.
Collapse
Affiliation(s)
- Su Ho Han
- Department of Microbiology, College of Medicine, Hallym University, Gangwon-do 200-702, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Mastroianni JR, Ouellette AJ. Alpha-defensins in enteric innate immunity: functional Paneth cell alpha-defensins in mouse colonic lumen. J Biol Chem 2009; 284:27848-27856. [PMID: 19687006 DOI: 10.1074/jbc.m109.050773] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Paneth cells are a secretory epithelial lineage that release dense core granules rich in host defense peptides and proteins from the base of small intestinal crypts. Enteric alpha-defensins, termed cryptdins (Crps) in mice, are highly abundant in Paneth cell secretions and inherently resistant to proteolysis. Accordingly, we tested the hypothesis that enteric alpha-defensins of Paneth cell origin persist in a functional state in the mouse large bowel lumen. To test this idea, putative Crps purified from mouse distal colonic lumen were characterized biochemically and assayed in vitro for bactericidal peptide activities. The peptides comigrated with cryptdin control peptides in acid-urea-PAGE and SDS-PAGE, providing identification as putative Crps. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry experiments showed that the molecular masses of the putative alpha-defensins matched those of the six most abundant known Crps, as well as N-terminally truncated forms of each, and that the peptides contain six Cys residues, consistent with identities as alpha-defensins. N-terminal sequencing definitively revealed peptides with N termini corresponding to full-length, (des-Leu)-truncated, and (des-Leu-Arg)-truncated N termini of Crps 1-4 and 6. Crps from mouse large bowel lumen were bactericidal in the low micromolar range. Thus, Paneth cell alpha-defensins secreted into the small intestinal lumen persist as intact and functional forms throughout the intestinal tract, suggesting that the peptides may mediate enteric innate immunity in the colonic lumen, far from their upstream point of secretion in small intestinal crypts.
Collapse
Affiliation(s)
- Jennifer R Mastroianni
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California 92697-4800
| | - André J Ouellette
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California 92697-4800; Department of Microbiology and Molecular Genetics, University of California, Irvine, California 92697-4800.
| |
Collapse
|
18
|
Diamond G, Beckloff N, Ryan LK. Host defense peptides in the oral cavity and the lung: similarities and differences. J Dent Res 2008; 87:915-27. [PMID: 18809744 DOI: 10.1177/154405910808701011] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Peptides with broad-spectrum antimicrobial activity are found in the mucosal surfaces at many sites in the body, including the airway, the oral cavity, and the digestive tract. Based on their in vitro antimicrobial and other immunomodulatory activities, these host defense peptides have been proposed to play an important role in the innate defense against pathogenic microbial colonization. The genes that encode these peptides are up-regulated by pathogens, further supporting their role in innate immune defense. However, the differences in the local microbial environments between the generally sterile airway and the highly colonized oral cavity suggest a more complex role for these peptides in innate immunity. For example, beta-defensin genes are induced in the airway by all bacteria and Toll-like receptor (TLR) agonists primarily through an NF-kappaB-mediated pathway. In contrast, the same genes are induced in the gingival epithelium by only a subset of bacteria and TLR ligands, via different pathways. Furthermore, the environments into which the peptides are secreted--specifically saliva, gingival crevicular fluid, and airway surface fluid--differ greatly and can effect their respective activities in host defense. In this review, we examine the differences and similarities between host defense peptides in the oral cavity and the airway, to gain a better understanding of their contributions to immunity.
Collapse
Affiliation(s)
- G Diamond
- Department of Oral Biology, UMDNJ-New Jersey Dental School, 185 South Orange Ave., Newark 07103, NJ 07101, USA.
| | | | | |
Collapse
|
19
|
Uchiya KI, Nikai T. Salmonella virulence factor SpiC is involved in expression of flagellin protein and mediates activation of the signal transduction pathways in macrophages. Microbiology (Reading) 2008; 154:3491-3502. [DOI: 10.1099/mic.0.2008/021667-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Kei-ichi Uchiya
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Toshiaki Nikai
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| |
Collapse
|
20
|
Interactions of the intestinal epithelium with the pathogen and the indigenous microbiota: a three-way crosstalk. Interdiscip Perspect Infect Dis 2008; 2008:626827. [PMID: 19259328 PMCID: PMC2648619 DOI: 10.1155/2008/626827] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 08/08/2008] [Indexed: 12/24/2022] Open
Abstract
The mucosal surfaces of the gastrointestinal tract harbor a vast number of commensal microbiota that have coevolved with the host, and in addition display one of the most complex relationships with the host. This relationship affects several important aspects of the biology of the host including the synthesis of nutrients, protection against infection, and the development of the immune system. On the other hand, despite the existence of several lines of mucosal defense mechanisms, pathogenic organisms such as Shigella and Salmonella have evolved sophisticated virulence strategies for breaching these barriers. The constant challenge from these pathogens and the attempts by the host to counter them set up a dynamic equilibrium of cellular and molecular crosstalk. Even slight perturbations in this equilibrium may be detrimental to the host leading to severe bacterial infection or even autoimmune diseases like inflammatory bowel disease. Several experimental model systems, including germ-free mice and antibiotic-treated mice, have been used by various researchers to study this complex relationship. Although it is only the beginning, it promises to be an exciting era in the study of these host-microbe relationships.
Collapse
|
21
|
Zheng Y, Valdez PA, Danilenko DM, Hu Y, Sa SM, Gong Q, Abbas AR, Modrusan Z, Ghilardi N, de Sauvage FJ, Ouyang W. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med 2008; 14:282-9. [PMID: 18264109 DOI: 10.1038/nm1720] [Citation(s) in RCA: 1497] [Impact Index Per Article: 93.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 01/04/2008] [Indexed: 11/09/2022]
Abstract
Infections by attaching and effacing (A/E) bacterial pathogens, such as Escherichia coli O157:H7, pose a serious threat to public health. Using a mouse A/E pathogen, Citrobacter rodentium, we show that interleukin-22 (IL-22) has a crucial role in the early phase of host defense against C. rodentium. Infection of IL-22 knockout mice results in increased intestinal epithelial damage, systemic bacterial burden and mortality. We also find that IL-23 is required for the early induction of IL-22 during C. rodentium infection, and adaptive immunity is not essential for the protective role of IL-22 in this model. Instead, IL-22 is required for the direct induction of the Reg family of antimicrobial proteins, including RegIIIbeta and RegIIIgamma, in colonic epithelial cells. Exogenous mouse or human RegIIIgamma substantially improves survival of IL-22 knockout mice after C. rodentium infection. Together, our data identify a new innate immune function for IL-22 in regulating early defense mechanisms against A/E bacterial pathogens.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Immunology, Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Eberhard J, Menzel N, Dommisch H, Winter J, Jepsen S, Mutters R. The stage of native biofilm formation determines the gene expression of human β-defensin-2, psoriasin, ribonuclease 7 and inflammatory mediators: a novel approach for stimulation of keratinocytes with in situ formed biofilms. ACTA ACUST UNITED AC 2007; 23:21-8. [DOI: 10.1111/j.1399-302x.2007.00385.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Chen YL, Chen YS, Lin HH, Chan CW, Chen SC, Chen CH. Immunostimulatory flagellin from Burkholderia pseudomallei effects on an increase in the intracellular calcium concentration and up-regulation of TNF-alpha by mononuclear cells. Microbiol Immunol 2007; 51:81-6. [PMID: 17237602 DOI: 10.1111/j.1348-0421.2007.tb03893.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Using flow cytometry analysis, the flagellin of Burkholderia pseudomallei acts as a signalling inducer, and evokes an increase in the intracellular calcium ion concentration ([Ca(2+)]i) in human peripheral blood mononuclear cells (PBMC). The cells with increased [Ca(2+)]i segregate into the live monocyte gate and not into the live lymphocyte gates. The stimulated [Ca(2+)]i increase can be neutralized with anti-flagellin antibodies. In the absence of [Ca(2+)], [Ca(2+)]i was increased rapidly in flagellin-treated cells compared to non-flagellin-treated cells only after the addition of 1 mM CaCl(2). Selective calcium antagonists were used to effectively block the [Ca(2+)]i signal, revealing that this signal was decreased by the addition of L-type calcium channel blockers (diltiazem, nifedipine and verapamil) and La(2+) but was not changed by the addition of a T-type calcium channel blocker (flunarizine). It seemed that flagellin facilitates [Ca(2+)]i influx via a La(2+) sensitive L-type cellular membrane channel. Furthermore, flagellin also acts as a TNF-alpha inducer in a time- and concentration-dependent manner when adhered mononuclear cells are treated with flagellin. This ability to induce TNF-alpha production was affected by the presence of [Ca(2+)] in the culture medium. It suggested that B. pseudomallei flagellin is an immuno-stimulatory molecule, causing an increase in [Ca(2+)]i and an up-regulation of TNF-alpha, which may play an important role in the inflammation process.
Collapse
Affiliation(s)
- Ya-Lei Chen
- Department of Biotechnology, National Kaohsiung Normal University
| | | | | | | | | | | |
Collapse
|
24
|
Dommisch H, Winter J, Willebrand C, Eberhard J, Jepsen S. Immune regulatory functions of human beta-defensin-2 in odontoblast-like cells. Int Endod J 2007; 40:300-7. [PMID: 17298411 DOI: 10.1111/j.0143-2885.2007.01228.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIM To investigate the effects of human beta-defensins on the expression of genes involved in the host immune response of the dental pulp. METHODOLOGY Human odontoblast-like cells were cultured in Dulbecco's modified Eagle's medium. Cells were stimulated by recombinant human beta-defensins (rhBDs) up to 4 h. RNA was extracted followed by cDNA synthesis (oligo-(dT)-primer). Samples were analysed by real-time polymerase chain reaction (PCR) technology. Genes of interest were: human beta-defensin-1, -2, interleukin (IL)-6, IL-8, tumour necrosis factor-alpha, cyclooxygenase-2, leukotriene-A4-hydrolase, cytosolic phospholipase-A-2 (cPLA(2)), and dentine sialophosphoprotein. Gene expression of beta-actin served as internal standard for normalizing real-time PCR data. Two-way anova and the paired t-test were applied for comparison of the gene expression. RESULTS In odontoblast-like cells rhBD-2 stimulation led to a down-regulation of the gene expression of hBD-1 (P < 0.05), whilst the mRNA expression of IL-6 (P < 0.05), IL-8 (P < 0.05) and cPLA(2) was increased in response to rhBD-2. CONCLUSION The results of the present study suggest immune regulatory functions of human beta-defensin-2 in odontoblast-like cells.
Collapse
Affiliation(s)
- H Dommisch
- Department of Periodontology, Operative and Preventative Dentistry, University Hospital Bonn, Bonn, Germany.
| | | | | | | | | |
Collapse
|
25
|
Abstract
The resident microbiota of the human intestine exerts a conditioning effect on intestinal homeostasis, delivering regulatory signals to the epithelium and instructing mucosal immune responses. Pattern recognition receptors are key mediators of innate host defense, and in healthy individuals, the mucosal immune system exhibits an exquisitely regulated restrained response to the resident microbiota. However, in genetically susceptible hosts, unrestrained mucosal immune activation in response to local bacterial signals can contribute to the pathogenesis of inflammatory bowel disease. Manipulation of the microbiota to enhance its beneficial components thus represents a potential therapeutic strategy for inflammatory bowel disease. Moreover, the microbiota might be a rich repository of metabolites that can be exploited for therapeutic benefit. Modern molecular techniques are facilitating improved understanding of host-microbe dialogue in health and in several disease processes, including inflammatory bowel disease. It follows that elucidating the molecular mechanisms of host-microbial interactions is now a prerequisite for a "bugs to drugs" program of discovery.
Collapse
Affiliation(s)
- Ann M O'Hara
- Alimentary Pharmabiotic Centre, University College Cork, National University of Ireland, Cork, Ireland
| | | |
Collapse
|
26
|
Adriaensen C, De Greve H, Tian JQ, De Craeye S, Gubbels E, Eeckhaut V, Van Immerseel F, Ducatelle R, Kumar M, Hernalsteens JP. A live Salmonella enterica serovar Enteritidis vaccine allows serological differentiation between vaccinated and infected animals. Infect Immun 2007; 75:2461-8. [PMID: 17261603 PMCID: PMC1865732 DOI: 10.1128/iai.01357-06] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Three precisely defined deletion mutants of Salmonella enterica serovar Enteritidis were constructed, a guanine auxotrophic DeltaguaB mutant, a nonflagellated DeltafliC mutant, and an auxotrophic and nonflagellated DeltaguaB DeltafliC double mutant. All three mutants were less invasive than the wild-type strain in primary chicken cecal epithelial cells and the human epithelial cell line T84 and less efficiently internalized in the chicken macrophage cell line HD11. The DeltafliC mutant was pathogenic in orally infected BALB/c mice, while the DeltaguaB mutant was attenuated and conferred protection against a challenge with the pathogenic parent strain. The DeltaguaB DeltafliC double mutant was totally asymptomatic and conferred better protection than the DeltaguaB mutant. This indicates that the major flagellar protein flagellin is not required for efficient vaccination of BALB/c mice against Salmonella infection. The DeltaguaB DeltafliC mutant was also safe for vaccination of 1-day-old chickens. After two immunizations, it induced statistically significant protection against infection of the internal organs of the birds by a virulent S. enterica serovar Enteritidis challenge strain but not against intestinal colonization. These data demonstrate that nonflagellated attenuated Salmonella mutants can be used as marker vaccines.
Collapse
Affiliation(s)
- Connie Adriaensen
- Viral Genetics Laboratory, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW To review recently published studies presenting novel and relevant information on antimicrobial peptides in gastrointestinal infections. RECENT FINDINGS Defensins and cathelicidins are important antimicrobial peptides expressed by the gastrointestinal epithelium. Their localization and regulation have been the focus of current research establishing the relevance of these peptides both in counteracting an attack by pathogens as well as in controlling the endogenous bacterial flora. In the small intestine, Paneth cell alpha-defensins maintain a low level of microorganisms and regulate the composition of the bacterial flora. In contrast, a constitutive beta-defensin can be found in nearly all gastrointestinal tissues. Other relevant beta-defensins as well as human cathelicidin are inducible by inflammation or infections. Thus Helicobacter pylori enhances defensin expression in the gastric mucosa and Campylobacter jejuni and Salmonella provoke a similar response in the colon. Other pathogenic bacteria may suppress the antimicrobial peptide response as an escape strategy. Notably, the therapeutic induction of cathelicidins alleviates experimental shigellosis, suggesting a future role of endogenous antibiotics in medical therapy. SUMMARY These recent findings together with a better understanding of underlying mechanisms involved in the regulation and biology of antimicrobial peptides will open up new therapeutic avenues to battle infections.
Collapse
|
28
|
O'Hara AM, O'Regan P, Fanning A, O'Mahony C, Macsharry J, Lyons A, Bienenstock J, O'Mahony L, Shanahan F. Functional modulation of human intestinal epithelial cell responses by Bifidobacterium infantis and Lactobacillus salivarius. Immunology 2006; 118:202-15. [PMID: 16771855 PMCID: PMC1782284 DOI: 10.1111/j.1365-2567.2006.02358.x] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intestinal epithelial cells (IECs) and dendritic cells (DCs) play a pivotal role in antigen sampling and the maintenance of gut homeostasis. However, the interaction of commensal bacteria with the intestinal surface remains incompletely understood. Here we investigated immune cell responses to commensal and pathogenic bacteria. HT-29 human IECs were incubated with Bifidobacterium infantis 35624, Lactobacillus salivarius UCC118 or Salmonella typhimurium UK1 for varying times, or were pretreated with a probiotic for 2 hr prior to stimulation with S. typhimurium or flagellin. Gene arrays were used to examine inflammatory gene expression. Nuclear factor (NF)-kappaB activation, interleukin (IL)-8 secretion, pathogen adherence to IECs, and mucin-3 (MUC3) and E-cadherin gene expression were assayed by TransAM assay, enzyme-linked immunosorbent assay (ELISA), fluorescence, and real-time reverse transcriptase-polymerase chain reaction (RT-PCR), respectively. IL-10 and tumour necrosis factor (TNF)-alpha secretion by bacteria-treated peripheral blood-derived DCs were measured using ELISA. S. typhimurium increased expression of 36 of the 847 immune-related genes assayed, including NF-kappaB and IL-8. The commensal bacteria did not alter expression levels of any of the 847 genes. However, B. infantis and L. salivarius attenuated both IL-8 secretion at baseline and S. typhimurium-induced pro-inflammatory responses. B. infantis also limited flagellin-induced IL-8 protein secretion. The commensal bacteria did not increase MUC3or E-cadherin expression, or interfere with pathogen binding to HT-29 cells, but they did stimulate IL-10 and TNF-alpha secretion by DCs. The data demonstrate that, although the intestinal epithelium is immunologically quiescent when it encounters B. infantis or L. salivarius, these commensal bacteria exert immunomodulatory effects on intestinal immune cells that mediate host responses to flagellin and enteric pathogens.
Collapse
Affiliation(s)
- Ann M O'Hara
- Alimentary Pharmabiotic Centre, University College Cork, National University of Ireland, Cork, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Liévin-Le Moal V, Servin AL. The front line of enteric host defense against unwelcome intrusion of harmful microorganisms: mucins, antimicrobial peptides, and microbiota. Clin Microbiol Rev 2006; 19:315-37. [PMID: 16614252 PMCID: PMC1471992 DOI: 10.1128/cmr.19.2.315-337.2006] [Citation(s) in RCA: 353] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The intestinal tract is a complex ecosystem that combines resident microbiota and the cells of various phenotypes with complex metabolic activities that line the epithelial wall. The intestinal cells that make up the epithelium provide physical and chemical barriers that protect the host against the unwanted intrusion of microorganisms that hijack the cellular molecules and signaling pathways of the host and become pathogenic. Some of the organisms making up the intestinal microbiota also have microbicidal effects that contribute to the barrier against enteric pathogens. This review describes the two cell lineages present in the intestinal epithelium: the goblet cells and the Paneth cells, both of which play a pivotal role in the first line of enteric defense by producing mucus and antimicrobial peptides, respectively. We also analyze recent insights into the intestinal microbiota and the mechanisms by which some resident species act as a barrier to enteric pathogens. Moreover, this review examines whether the cells producing mucins or antimicrobial peptides and the resident microbiota act in partnership and whether they function individually and/or synergistically to provide the host with an effective front line of defense against harmful enteric pathogens.
Collapse
Affiliation(s)
- Vanessa Liévin-Le Moal
- Unité 756 INSERM, Faculté de Pharmacie Paris XI, Signalisation et Physiopathologie des Cellules Epithéliales, Institut National de la Santé et de la Recherche Médicale, F-92296 Chātenay-Malabry, France
| | | |
Collapse
|
30
|
Wah J, Wellek A, Frankenberger M, Unterberger P, Welsch U, Bals R. Antimicrobial peptides are present in immune and host defense cells of the human respiratory and gastroinstestinal tracts. Cell Tissue Res 2006; 324:449-56. [PMID: 16501999 DOI: 10.1007/s00441-005-0127-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2005] [Accepted: 11/11/2005] [Indexed: 02/07/2023]
Abstract
Previous studies have implicated antimicrobial peptides in the host defense of the mammalian intestinal and respiratory tract. The aim of the present study has been to characterize further the expression of these molecules in non-epithelial cells of the human pulmonary and digestive systems by detailed immunohistochemical analysis of the small and large bowel and of the large airways and lung parenchyma. Additionally, cells obtained from bronchoalveolar lavage were analyzed by fluorescent activated cell sorting and immunostaining of cytospin preparations. hBD-1, hBD-2, and LL-37 were detected in lymphocytes and macrophages in the large airways, lung parenchyma, duodenum, and colon. Lymphocytes positive for the peptides revealed a staining pattern and distribution that largely matched that of CD3-positive and CD8-positive T-cells. Macrophages with positive staining for the antimicrobial peptides also stained positively for CD68 and CD74. In view of the morphology of the LL-37-positive and hBD-2-positive mucosal lymphocytes, they are probably also B-cells. Thus, antimicrobial peptides of the defensin and cathelicidin families are present in a variety of non-epithelial cells of mucosal organs. These findings confirm that antimicrobial peptides have multiple functions in the biology of the mucosa of these organs.
Collapse
Affiliation(s)
- Jenny Wah
- Department of Anatomy, Chair II, Ludwig Maximilian University, 80336 München, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Froy O. Regulation of mammalian defensin expression by Toll-like receptor-dependent and independent signalling pathways. Cell Microbiol 2005; 7:1387-97. [PMID: 16153239 DOI: 10.1111/j.1462-5822.2005.00590.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The immune system consists of innate and adaptive immune responses. The innate immune system confers non-specific protection against a large number of pathogens, hence, serving as the first line of defence. The innate immune system utilizes Toll-like receptors (TLRs) to recognize and bind pathogen-associated molecular patterns (PAMPs). Binding of PAMPs leads to TLR activation, which, in turn, initiates MAPK- or NF-kappaB-dependent cascades that culminate in a proinflammatory response. This response involves the secretion of cytokines, chemokines and broad-spectrum antibacterial substances, such as defensins. Increased defensin synthesis is also mediated by the activation of receptors other than TLRs, such as NOD2, IL-17R and PAR-2. This review summarizes the recently characterized signalling pathways leading to increased defensin synthesis as well as the pathway by which defensins activate TLRs on immature dendritic and memory T cells. Thus, not only do defensins eliminate pathogens, but they also recruit the adaptive immune system in instances of infection and/or inflammation.
Collapse
Affiliation(s)
- Oren Froy
- Institute of Biochemistry, Food Science and Nutrition, Faculty of Agricultural, Food, and Environmental Quality, The Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
32
|
Kumar A, Zhang J, Yu FSX. Toll-like receptor 2-mediated expression of beta-defensin-2 in human corneal epithelial cells. Microbes Infect 2005; 8:380-9. [PMID: 16242370 PMCID: PMC2666383 DOI: 10.1016/j.micinf.2005.07.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Revised: 06/28/2005] [Accepted: 07/04/2005] [Indexed: 11/20/2022]
Abstract
We previously showed that human corneal epithelial cells (HCECs) express Toll-like receptors (TLRs), which recognize gram-positive bacteria and respond to Staphylococcus aureus infection by the expression and secretion of proinflammatory cytokines and beta-defensin-2 (hBD2). In this study, we further elucidated the underlying mechanisms regulating hBD-2 expression and its role in innate defense in HCECs in response to S. aureus challenge. Exposure of HUCL cells, a telomerase-immortalized HCEC line, to S. aureus, its exoproducts (1:10 dilution), or synthetic lipopeptide Pam3Cys (10 microg/ml) resulted in the up-regulation of hBD-2, but not hBD1 and hBD3. Similar to HUCL cells, primary HCECs responded to S. aureus-exoproducts and Pam3Cys challenge by expressing hBD2 mRNA and secreting hBD2 into the culture media. Furthermore, these stimuli induced the expression of TLR2 at both mRNA and protein levels. Consistently with its role as a major pattern-recognizing receptor, TLR2 was located at the cell surface by cell surface biotinylation. The treatment of HUCL cells with TLR2 neutralizing antibody resulted in a significant decrease in Pam3Cys-induced hBD2 production as well as IL-6, IL-8, and TNF-alpha secretion. The Pam3Cys-induced hBD2 expression was completely blocked by NF-kappaB inhibitors and partially inhibited by p38 MAP kinase and the JNK inhibitors. Conditioned media derived from HCECs challenged with S. aureus-exoproducts or Pam3Cys exhibited antibacterial activity against S. aureus, Pseudomonas aeruginosa and Escherichia coli. These findings suggest that S. aureus induces hBD2 production through TLR2-mediated pathways in HCECs and that pathogen-challenged, TLR-activated HCECs possess antimicrobial activity. Thus, the epithelium might play a role in innate defense against bacterial infection by directly killing bacteria in the cornea.
Collapse
Affiliation(s)
| | | | - Fu-Shin X. Yu
- Corresponding author. Tel.: +1 313 577 1657; fax: +1 313 577 7781. E-mail address: (F.-S.X. Yu)
| |
Collapse
|
33
|
Dommisch H, Açil Y, Dunsche A, Winter J, Jepsen S. Differential gene expression of human beta-defensins (hBD-1, -2, -3) in inflammatory gingival diseases. ACTA ACUST UNITED AC 2005; 20:186-90. [PMID: 15836521 DOI: 10.1111/j.1399-302x.2005.00211.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antimicrobial peptides, like human beta-defensins, play an important role in the epithelial innate defense response. The aim of the present study was to investigate the quantitative expression of human beta-defensin-1, -2, and -3 in inflammatory gingival diseases. Gingival biopsies were obtained from patients with healthy gingiva (n = 10), patients with gingivitis (n = 10), and patients with periodontitis (n = 10). The clinical diagnosis was verified by histology. Gingival tissues were used for RNA extraction followed by reverse transcription. Gene expression was quantified by real-time polymerase chain reaction (normalization with GAP-DH). Comparing the tissues with different clinical stages of health and disease, no significant differences in mRNA expression were found for any of the beta-defensins studied. Similar levels of expression were found in healthy gingiva, whereas in gingivitis samples there was a significantly higher expression of hBD-2 compared to hBD-1 (P = 0.004) and hBD-3 (P = 0.016). Likewise, in periodontitis samples, hBD-2 expression was significantly higher than hBD-1 (P = 0.016); however, hBD-2 expression was comparable to hBD-3. In conclusion, the results of the present study showed a differential expression of human beta-defensins (hBD-1, -2, -3) in tissues with inflammatory gingival disease.
Collapse
Affiliation(s)
- H Dommisch
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany.
| | | | | | | | | |
Collapse
|
34
|
West AP, Dancho BA, Mizel SB. Gangliosides inhibit flagellin signaling in the absence of an effect on flagellin binding to toll-like receptor 5. J Biol Chem 2005; 280:9482-8. [PMID: 15632166 DOI: 10.1074/jbc.m411875200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A recent study (Ogushi, K., Wada, A., Niidome, T., Okuda, T., Llanes, R., Nakayama, M., Nishi, Y., Kurazono, H., Smith, K. D., Aderem, A., Moss, J., and Hirayama, T. (2004) J. Biol. Chem. 279, 12213-12219) concluded that gangliosides serve as co-receptors for flagellin signaling via toll-like receptor 5 (TLR5). In view of several findings in this study that were inconsistent with a role for gangliosides as co-receptors, we re-examined this important issue. Using TLR5-negative RAW 264.7 cells and a TLR5-enhanced yellow fluorescent protein chimera, we established an assay for specific binding of flagellin to cells. Inhibition of clatherin-mediated internalization of flagellin.TLR5-enhanced yellow fluorescent protein complexes did not impair flagellin activation of IRAK-1. Thus flagellin signal occurs at the cell surface and not intracellularly. Exogenous addition of mixed gangliosides (GM1, GD1a, and GT1b) as well as GD1a itself inhibited flagellin-induced interleukin-1 receptor-associated kinase activation as well as tumor necrosis factor alpha production in HeNC2, THP-1, and RAW 264.7 cells. Gangliosides inhibited flagellin signaling in the absence of an effect on flagellin binding to TLR5. Depletion of gangliosides in RAW 264.7 cells did not alter the concentration dependence or magnitude of flagellin signaling as measured by interleukin-1 receptor-associated kinase activation or tumor necrosis factor alpha production. Our findings are consistent with the conclusions that gangliosides are not essential co-receptors for flagellin and that the inhibitory effect of gangliosides is mediated by at least one mechanism that is distinct from any effect on the binding of flagellin to TLR5.
Collapse
Affiliation(s)
- A Phillip West
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | |
Collapse
|
35
|
Komatsuzawa H, Ouhara K, Yamada S, Fujiwara T, Sayama K, Hashimoto K, Sugai M. Innate defences against methicillin-resistantStaphylococcus aureus (MRSA) infection. J Pathol 2005; 208:249-60. [PMID: 16362993 DOI: 10.1002/path.1898] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The innate immune system is the primary defence against bacterial infection. Among the factors involved in innate defence, anti-microbial peptides produced by humans have recently attracted attention due to their relevance to some diseases and also to the development of new chemotherapeutic agents. Staphylococcus aureus is one of the major human pathogens, causing a variety of infections from suppurative disease to food poisoning. Methicillin-resistant S. aureus (MRSA) is a clinical problem and with the recent emergence of a vancomycin-resistant strain, this will pose serious problems in the near future. In investigating the molecular biology of S. aureus infections to develop new chemotherapeutic agents against MRSA infections, knowledge of the interaction of innate anti-microbial peptides with S. aureus is important. In vitro and in vivo experiments demonstrate that exposure of S. aureus to host cells can induce the anti-microbial peptides beta-defensin-2 (hBD2), hBD3, and LL37/CAP18. The induction level of these peptides differs among strains, as does the susceptibility of the strains, with MRSA strains exhibiting lower susceptibility. In summary, the susceptibility of S. aureus strains, including MRSA strains, to components of the innate immune system varies, with the MRSA strains showing more resistance to both innate immune factors and chemotherapeutic agents.
Collapse
Affiliation(s)
- Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical Sciences, Japan. hkomatsu@hiroshi,a-u.ac.jp
| | | | | | | | | | | | | |
Collapse
|
36
|
Servin AL. Antagonistic activities of lactobacilli and bifidobacteria against microbial pathogens. FEMS Microbiol Rev 2004; 28:405-40. [PMID: 15374659 DOI: 10.1016/j.femsre.2004.01.003] [Citation(s) in RCA: 740] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2003] [Revised: 11/19/2003] [Accepted: 01/28/2004] [Indexed: 12/16/2022] Open
Abstract
The gastrointestinal tract is a complex ecosystem that associates a resident microbiota and cells of various phenotypes lining the epithelial wall expressing complex metabolic activities. The resident microbiota in the digestive tract is a heterogeneous microbial ecosystem containing up to 1 x 10(14) colony-forming units (CFUs) of bacteria. The intestinal microbiota plays an important role in normal gut function and maintaining host health. The host is protected from attack by potentially harmful microbial microorganisms by the physical and chemical barriers created by the gastrointestinal epithelium. The cells lining the gastrointestinal epithelium and the resident microbiota are two partners that properly and/or synergistically function to promote an efficient host system of defence. The gastrointestinal cells that make up the epithelium, provide a physical barrier that protects the host against the unwanted intrusion of microorganisms into the gastrointestinal microbiota, and against the penetration of harmful microorganisms which usurp the cellular molecules and signalling pathways of the host to become pathogenic. One of the basic physiological functions of the resident microbiota is that it functions as a microbial barrier against microbial pathogens. The mechanisms by which the species of the microbiota exert this barrier effect remain largely to be determined. There is increasing evidence that lactobacilli and bifidobacteria, which inhabit the gastrointestinal microbiota, develop antimicrobial activities that participate in the host's gastrointestinal system of defence. The objective of this review is to analyze the in vitro and in vivo experimental and clinical studies in which the antimicrobial activities of selected lactobacilli and bifidobacteria strains have been documented.
Collapse
Affiliation(s)
- Alain L Servin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 510, Pathogénes et Fonctions des Cellules Epithéliales Polarisées, Faculté de Pharmacie Paris XI, F-92296 Châtenay-Malabry, France.
| |
Collapse
|
37
|
Vora P, Youdim A, Thomas LS, Fukata M, Tesfay SY, Lukasek K, Michelsen KS, Wada A, Hirayama T, Arditi M, Abreu MT. Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:5398-405. [PMID: 15494486 DOI: 10.4049/jimmunol.173.9.5398] [Citation(s) in RCA: 261] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The intestinal epithelium serves as a barrier to the intestinal flora. In response to pathogens, intestinal epithelial cells (IEC) secrete proinflammatory cytokines. To aid in defense against bacteria, IEC also secrete antimicrobial peptides, termed defensins. The aim of our studies was to understand the role of TLR signaling in regulation of beta-defensin expression by IEC. The effect of LPS and peptidoglycan on beta-defensin-2 expression was examined in IEC lines constitutively or transgenically expressing TLRs. Regulation of beta-defensin-2 was assessed using promoter-reporter constructs of the human beta-defensin-2 gene. LPS and peptidoglycan stimulated beta-defensin-2 promoter activation in a TLR4- and TLR2-dependent manner, respectively. A mutation in the NF-kappaB or AP-1 site within the beta-defensin-2 promoter abrogated this response. In addition, inhibition of Jun kinase prevents up-regulation of beta-defensin-2 protein expression in response to LPS. IEC respond to pathogen-associated molecular patterns with expression of the antimicrobial peptide beta-defensin-2. This mechanism may protect the intestinal epithelium from pathogen invasion and from potential invaders among the commensal flora.
Collapse
Affiliation(s)
- Puja Vora
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yu Y, Zeng H, Vijay-Kumar M, Neish AS, Merlin D, Sitaraman SV, Gewirtz AT. STAT Signaling Underlies Difference between Flagellin-induced and Tumor Necrosis Factor-α-induced Epithelial Gene Expression. J Biol Chem 2004; 279:35210-8. [PMID: 15199060 DOI: 10.1074/jbc.m404064200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Both bacterial flagellin and the cytokine tumor necrosis factor-alpha (TNFalpha) are potent activators of intestinal epithelial cell pro-inflammatory gene expression in general; nonetheless, there seem to be distinct differences in the specific patterns of gene expression induced by these agonists. The goal of this study was to define one such difference and elucidate the signaling mechanism responsible for such differential gene induction by these agonists. We observed that expression of inducible nitric-oxide synthase is substantially induced by flagellin but only minimally expressed in response to TNFalpha. This difference seemed to be underlain by differential induction of signal transducers and activators of transcription (STAT) activation in that, whereas flagellin and TNFalpha seemed to be equipotent activators of p38 mitogen-activated protein kinase and nuclear factor-kappaB, flagellin induced substantially higher levels of STAT-1 and -3 tyrosine phosphorylation. Such flagellin-induced STAT activation exhibited delayed kinetics and was ablated by treatment with cycloheximide. Flagellin-induced activation of STAT-3 was abolished via neutralizing antibodies to interleukin (IL)-6, but not interferon (IFN)beta nor IFNgamma; none of these neutralizing antibodies had any effect on flagellin-induced STAT-1 tyrosine phosphorylation. Flagellin induced substantially more IL-6 expression than did TNFalpha, but neither agonist elicited detectable levels of IFN expression. Flagellin-induced expression of inducible nitric-oxide synthase but not IL-6, was abolished by blocking STAT activation with AG490, and was reduced by blocking STAT-3 activation with anti-IL-6. Together, these results indicate that epithelial cell induction of flagellin-specific gene expression is mediated, in part, by STAT activation that results from autocrine activation via IL-6.
Collapse
Affiliation(s)
- Yimin Yu
- Department of Pathology, Division of Digestive Diseases, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Forsberg G, Fahlgren A, Hörstedt P, Hammarström S, Hernell O, Hammarström ML. Presence of bacteria and innate immunity of intestinal epithelium in childhood celiac disease. Am J Gastroenterol 2004; 99:894-904. [PMID: 15128357 DOI: 10.1111/j.1572-0241.2004.04157.x] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Exposure to gliadin and related prolamins and appropriate HLA-DQ haplotype are necessary but not sufficient for contracting celiac disease (CD). Aberrant innate immune reactions could be contributing risk factors. Therefore, jejunal biopsies were screened for bacteria and the innate immune status of the epithelium investigated. METHODS Children with untreated, treated, challenged CD, and controls were analyzed. Bacteria were identified by scanning electron microscopy. Glycocalyx composition and mucin and antimicrobial peptide production were studied by quantitative RT-PCR, antibody and lectin immunohistochemistry. RESULTS Rod-shaped bacteria were frequently associated with the mucosa of CD patients, with both active and inactive disease, but not with controls. The lectin Ulex europaeus agglutinin I (UEAI) stained goblet cells in the mucosa of all CD patients but not of controls. The lectin peanut agglutinin (PNA) stained glycocalyx of controls but not of CD patients. mRNA levels of mucin-2 (MUC2), alpha-defensins HD-5 and HD-6, and lysozyme were significantly increased in active CD and returned to normal in treated CD. Their expression levels correlated to the interferon-gamma mRNA levels in intraepithelial lymphocytes. MUC2, HD-5, and lysozyme proteins were seen in absorptive epithelial cells. beta-defensins hBD-1 and hBD-2, carcinoembryonic antigen (CEA), CEA cell adhesion molecule-1a (CEACAM1a), and MUC3 were not affected. CONCLUSIONS Unique carbohydrate structures of the glycocalyx/mucous layer are likely discriminating features of CD patients. These glycosylation differences could facilitate bacterial adhesion. Ectopic production of MUC2, HD-5, and lysozyme in active CD is compatible with goblet and Paneth cell metaplasia induced by high interferon-gamma production by intraepithelial lymphocytes.
Collapse
Affiliation(s)
- Göte Forsberg
- Department of Clinical Microbiology and Immunology, Umeå University, Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
40
|
Ogushi KI, Wada A, Niidome T, Okuda T, Llanes R, Nakayama M, Nishi Y, Kurazono H, Smith KD, Aderem A, Moss J, Hirayama T. Gangliosides act as co-receptors for Salmonella enteritidis FliC and promote FliC induction of human beta-defensin-2 expression in Caco-2 cells. J Biol Chem 2004; 279:12213-9. [PMID: 14707135 DOI: 10.1074/jbc.m307944200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Antimicrobial peptides such as defensins are crucial for host defense at mucosal surfaces. We reported previously that Salmonella enteritidis flagellin (FliC) induced human beta-defensin-2 (hBD-2) mRNA expression in Caco-2 cells via NF-kappaB activation (Ogushi, K., Wada, A., Niidome, T., Mori, N., Oishi, K., Nagatake, T., Takahashi, A., Asakura, H., Makino, S., Hojo, H., Nakahara, Y., Ohsaki, M., Hatakeyama, T., Aoyagi, H., Kurazono, H., Moss, J., and Hirayama, T. (2001) J. Biol. Chem. 276, 30521-30526). In this study, we examined the role of ganglioside as co-receptors with Toll-like receptor 5 (TLR5) on FliC induction of hBD-2 expression in Caco-2 cells. Exogenous gangliosides suppressed FliC induction of hBD-2 promoter activity and binding of FliC to Caco-2 cells. Incorporation of exogenous ganglioside GD1a into Caco-2 cell membranes increased the effect of FliC on hBD-2 promoter activity. In support of a role for endogenous gangliosides, incubation of Caco-2 cells with dl-threo-2-hexadecanoylamino-3-morpholino-1-phenylpropanol, a glucosylceramide synthase inhibitor, reduced FliC induction of hBD-2 promoter activity. GD1a-loaded CHO-K1-expressing TLR5 cells had a higher potential for hBD-2 induction following FliC stimulation than GD1a-loaded CHO-K1 cells not expressing TLR5. FliC increased phosphorylation of mitogen-activated protein kinase, p38, and ERK1/2. Exogenous gangliosides GD1a, GD1b, and GT1b each suppressed FliC induction of p38 and ERK1/2 phosphorylation. Furthermore, FliC did not enhance luciferase activity in Caco-2 cells transfected with a plasmid containing a mutated activator protein 1-binding site. These results suggest that gangliosides act as co-receptors with TLR5 for FliC and promote hBD-2 expression via mitogen-activated protein kinase.
Collapse
Affiliation(s)
- Ken-ichi Ogushi
- Department of Bacteriology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
O'Neil DA. Regulation of expression of beta-defensins: endogenous enteric peptide antibiotics. Mol Immunol 2003; 40:445-50. [PMID: 14568390 DOI: 10.1016/s0161-5890(03)00161-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Evidence for the central role that intestinal beta-defensins play in maintaining gut health continues to accumulate within the literature. Two epithelially-derived enteric beta-defensins, hBD1 and hBD2, have been identified thus far and the following chapter reviews our current understanding of how the expression and secretion of these endogenous antimicrobial, chemotactic and adjuvant peptides is regulated within the context of the most microbe-rich of mucosal environments, the gastrointestinal tract. The agonists and microbial moieties identified as being responsible for the direct receptor-mediated induction of enteric epithelial beta-defensins, the signaling and nuclear events that are triggered as a consequence and which drive defensin gene transcription, the potential antimicrobial and immunomodulatory consequences of beta-defensin release within the luminal and mucosal aspects of the alimentary tract thereafter and the validity of animal models for the study of these key immune effector molecules in vivo are discussed. These significant and recent discoveries have provided much in the way of momentum for the pace with which this exciting and dynamic area of mucosal immunology research continues to move forward.
Collapse
Affiliation(s)
- Deborah A O'Neil
- Gut Immunology Group, The Rowett Research Institute, Greenburn Road, Bucksburn, Aberdeen AB51 0UX, UK.
| |
Collapse
|
42
|
Cunliffe RN, Mahida YR. Expression and regulation of antimicrobial peptides in the gastrointestinal tract. J Leukoc Biol 2003; 75:49-58. [PMID: 14525966 DOI: 10.1189/jlb.0503249] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is exposed to a wide range of microorganisms. The expression of antimicrobial peptides has been demonstrated in different regions of the GI tract, predominantly in epithelial cells, which represent the first host cells with which the microorganisms have to interact for invasion. The intestinal epithelial monolayer is complex, consisting of different cell types, and most have a limited lifespan. Of the GI antimicrobial peptides, alpha- and beta-defensins have been studied the most and are expressed by distinct types of epithelial cells. Enteric alpha-defensin expression is normally restricted to Paneth and intermediate cells in the small intestine. However, there are important differences between mice and humans in the processing of the precursor forms of enteric alpha-defensins. Parasite infection induces an increase in the number of enteric alpha-defensin-expressing Paneth and intermediate cells in the murine small intestine. In the chronically inflamed colonic mucosa, metaplastic Paneth cells (which are absent in the normal colon) also express enteric alpha-defensins. Epithelial expression of beta-defensins may be constitutive or inducible by infectious and inflammatory stimuli. The production of some members of the beta-defensin family appears to be restricted to distinct parts of the GI tract. Recent studies using genetically manipulated rodents have demonstrated the likely in vivo importance of enteric antimicrobial peptides in innate host defense against microorganisms. The ability of these peptides to act as chemoattractants for cells of the innate- and adaptive-immune system may also play an important role in perpetuating chronic inflammation in the GI tract.
Collapse
Affiliation(s)
- R N Cunliffe
- Institute of Infection, Immunity and Inflammation and Division of Gastroenterology, University of Nottingham, United Kingdom
| | | |
Collapse
|
43
|
Ellermeier CD, Slauch JM. RtsA and RtsB coordinately regulate expression of the invasion and flagellar genes in Salmonella enterica serovar Typhimurium. J Bacteriol 2003; 185:5096-108. [PMID: 12923082 PMCID: PMC181000 DOI: 10.1128/jb.185.17.5096-5108.2003] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Salmonella enterica serovar Typhimurium encounters numerous host environments and defense mechanisms during the infection process. The bacterium responds by tightly regulating the expression of virulence genes. We identified two regulatory proteins, termed RtsA and RtsB, which are encoded in an operon located on an island integrated at tRNA(PheU) in S. enterica serovar Typhimurium. RtsA belongs to the AraC/XylS family of regulators, and RtsB is a helix-turn-helix DNA binding protein. In a random screen, we identified five RtsA-regulated fusions, all belonging to the Salmonella pathogenicity island 1 (SPI1) regulon, which encodes a type III secretion system (TTSS) required for invasion of epithelial cells. We show that RtsA increases expression of the invasion genes by inducing hilA expression. RtsA also induces expression of hilD, hilC, and the invF operon. However, induction of hilA is independent of HilC and HilD and is mediated by direct binding of RtsA to the hilA promoter. The phenotype of an rtsA null mutation is similar to the phenotype of a hilC mutation, both of which decrease expression of SPI1 genes approximately twofold. We also show that RtsA can induce expression of a SPI1 TTSS effector, slrP, independent of any SPI1 regulatory protein. RtsB represses expression of the flagellar genes by binding to the flhDC promoter region. Repression of the positive activators flhDC decreases expression of the entire flagellar regulon. We propose that RtsA and RtsB coordinate induction of invasion and repression of motility in the small intestine.
Collapse
Affiliation(s)
- Craig D Ellermeier
- Department of Microbiology, College of Medicine, Chemical and Life Sciences Laboratory, 601 S. Goodwin Avenue, University of Illinois, Urbana, IL 61801, USA
| | | |
Collapse
|
44
|
Abstract
This comprehensive review promotes the novel concept that a defensin deficiency, i.e. lack of mucosal peptide antibiotics, may play a pivotal role in the aetiopathogenesis of Crohn's disease. Such an impaired function of this chemical barrier is consistent with the epidemiological relationship of good domestic hygiene with the incidence of inflammatory bowel diseases. The disregulated adaptive immune system, formerly believed to be the major cause in the development of Crohn's disease, may reflect only the primary break of the mucosal defence since the immune response is mostly directed against lumenal bacteria. Recent work has identified five different defensins expressed in colonic mucosa. In contrast to ulcerative colitis, Crohn's disease is characterised by an impaired induction of human beta defensins 2 and 3. This deficient induction may be due to changes in the intracellular transcription by NFkappaB and the intracellular peptidoglycan receptor NOD2, mutated in Crohn's disease. These findings are consistent with the mucosal attachment of lumenal bacteria in inflammatory bowel diseases and the frequent occurrence of other infectious agents. The hypothesis of an impaired mucosal antibacterial activity is also consistent with the benefit from antibiotic or probiotic treatment in certain inflammatory bowel disease states.
Collapse
Affiliation(s)
- Klaus Fellermann
- Department of Internal Medicine I, Robert Bosch Krankenhaus, Stuttgart, Germany
| | | | | | | |
Collapse
|
45
|
Abstract
Antimicrobial peptides (AMPs) are effector molecules of the innate immune system. A variety of AMPs have been isolated from species of all kingdoms and are classified based on their structure and amino acid motifs. AMPs have a broad antimicrobial spectrum and lyse microbial cells by interaction with biomembranes. Besides their direct antimicrobial function, they have multiple roles as mediators of inflammation with impact on epithelial and inflammatory cells influencing diverse processes such as cell proliferation, immune induction, wound healing, cytokine release, chemotaxis and protease-antiprotease balance. AMPs qualify as prototypes of innovative drugs that may be used as antimicrobials, anti-lipopolysaccharide drugs or modifiers of inflammation. Several strategies have been followed to identify lead candidates for drug development, to modify the peptides' structures, and to produce sufficient amounts for pre-clinical and clinical studies. This review summarises the current knowledge about the basic and applied biology of AMPs.
Collapse
Affiliation(s)
- Andreas R Koczulla
- Department of Internal Medicine, Division of Pulmonary Medicine, Hospital of the University of Marburg, Philipps-University Marburg, Marburg, Germany
| | | |
Collapse
|
46
|
Szabó C. Role of flagellin in the pathogenesis of shock and acute respiratory distress syndrome: therapeutic opportunities. Crit Care Med 2003; 31:S39-45. [PMID: 12544975 DOI: 10.1097/00003246-200301001-00006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To provide an overview of the role of flagellin as an immunostimulatory and proinflammatory factor. DESIGN A systematic overview of the literature on this subject. SETTING An office equipped with a computer and Internet access to PubMed. SUBJECTS MEDLINE citations between 1960 and 2002. MAIN RESULTS Flagellin, a protein of 40-60 kD, is the principal constituent of the flagellum, a prominent surface structure found in motile bacteria. Recent work reveals that monomeric flagellin, a protein component of flagellated bacteria, can act as a soluble immunostimulatory and proinflammatory factor, activating the immune/inflammatory axis via the toll-like receptor 5-nuclear factor-kappaB axis. Monocytes, macrophages, and intestinal and pulmonary epithelial cells respond to monomeric flagellin at low concentrations. Monomeric flagellin can induce prominent local and systemic immune/inflammatory responses and. CONCLUSIONS Recognition of the flagellin-toll-like receptor 5 pathway offers novel opportunities for the experimental therapy of various forms of shock, sepsis, and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Csaba Szabó
- Inotek Pharmaceuticals Corporation, Beverly, MA, USA
| |
Collapse
|
47
|
Terebiznik MR, Vieira OV, Marcus SL, Slade A, Yip CM, Trimble WS, Meyer T, Finlay BB, Grinstein S. Elimination of host cell PtdIns(4,5)P(2) by bacterial SigD promotes membrane fission during invasion by Salmonella. Nat Cell Biol 2002; 4:766-73. [PMID: 12360287 DOI: 10.1038/ncb854] [Citation(s) in RCA: 220] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2002] [Revised: 06/09/2002] [Accepted: 08/05/2002] [Indexed: 01/26/2023]
Abstract
Salmonella invades mammalian cells by inducing membrane ruffling and macropinocytosis through actin remodelling. Because phosphoinositides are central to actin assembly, we have studied the dynamics of phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P(2)) in HeLa cells during invasion by Salmonella typhimurium. Here we show that the outermost parts of the ruffles induced by invasion show a modest enrichment in PtdIns(4,5)P(2), but that PtdIns(4,5)P(2) is virtually absent from the invaginating regions. Rapid disappearance of PtdIns(4,5)P(2) requires the expression of the Salmonella phosphatase SigD (also known as SopB). Deletion of SigD markedly delays fission of the invaginating membranes, indicating that elimination of PtdIns(4,5)P(2) may be required for rapid formation of Salmonella-containing vacuoles. Heterologous expression of SigD is sufficient to promote the disappearance of PtdIns(4,5)P(2), to reduce the rigidity of the membrane skeleton, and to induce plasmalemmal invagination and fission. Hydrolysis of PtdIns(4,5)P(2) may be a common and essential feature of membrane fission during several internalization processes including invasion, phagocytosis and possibly endocytosis.
Collapse
|