1
|
Montorsi L, Pitcher MJ, Zhao Y, Dionisi C, Demonti A, Tull TJ, Dhami P, Ellis RJ, Bishop C, Sanderson JD, Jain S, D'Cruz D, Gibbons DL, Winkler TH, Bemark M, Ciccarelli FD, Spencer J. Double-negative B cells and DNASE1L3 colocalise with microbiota in gut-associated lymphoid tissue. Nat Commun 2024; 15:4051. [PMID: 38744839 PMCID: PMC11094119 DOI: 10.1038/s41467-024-48267-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Intestinal homeostasis is maintained by the response of gut-associated lymphoid tissue to bacteria transported across the follicle associated epithelium into the subepithelial dome. The initial response to antigens and how bacteria are handled is incompletely understood. By iterative application of spatial transcriptomics and multiplexed single-cell technologies, we identify that the double negative 2 subset of B cells, previously associated with autoimmune diseases, is present in the subepithelial dome in health. We show that in this location double negative 2 B cells interact with dendritic cells co-expressing the lupus autoantigens DNASE1L3 and C1q and microbicides. We observe that in humans, but not in mice, dendritic cells expressing DNASE1L3 are associated with sampled bacteria but not DNA derived from apoptotic cells. We propose that fundamental features of autoimmune diseases are microbiota-associated, interacting components of normal intestinal immunity.
Collapse
Affiliation(s)
- Lucia Montorsi
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Michael J Pitcher
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Yuan Zhao
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Chiara Dionisi
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Alicia Demonti
- School of Immunology and Microbial Sciences, King's College London, London, UK
- École Normale Supérieure de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Thomas J Tull
- St. John's Institute of Dermatology, King's College London, London, UK
| | - Pawan Dhami
- Genomics Research Platform and Single Cell Laboratory at Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Richard J Ellis
- Advanced Cytometry Platform (Flow Core), Research and Development Department at Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Cynthia Bishop
- Advanced Cytometry Platform (Flow Core), Research and Development Department at Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jeremy D Sanderson
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Gastroenterology, Guy's and St Thomas' Foundation Trust, London, UK
| | - Sahil Jain
- Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - David D'Cruz
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Louise Coote Lupus Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Deena L Gibbons
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Thomas H Winkler
- Division of Genetics, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Mats Bemark
- Department of Translational Medicine - Human Immunology, Lund University, Malmö, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Jo Spencer
- School of Immunology and Microbial Sciences, King's College London, London, UK.
| |
Collapse
|
2
|
Didriksen BJ, Eshleman EM, Alenghat T. Epithelial regulation of microbiota-immune cell dynamics. Mucosal Immunol 2024; 17:303-313. [PMID: 38428738 PMCID: PMC11412483 DOI: 10.1016/j.mucimm.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
The mammalian gastrointestinal tract hosts a diverse community of trillions of microorganisms, collectively termed the microbiota, which play a fundamental role in regulating tissue physiology and immunity. Recent studies have sought to dissect the cellular and molecular mechanisms mediating communication between the microbiota and host immune system. Epithelial cells line the intestine and form an initial barrier separating the microbiota from underlying immune cells, and disruption of epithelial function has been associated with various conditions ranging from infection to inflammatory bowel diseases and cancer. From several studies, it is now clear that epithelial cells integrate signals from commensal microbes. Importantly, these non-hematopoietic cells also direct regulatory mechanisms that instruct the recruitment and function of microbiota-sensitive immune cells. In this review, we discuss the central role that has emerged for epithelial cells in orchestrating intestinal immunity and highlight epithelial pathways through which the microbiota can calibrate tissue-intrinsic immune responses.
Collapse
Affiliation(s)
- Bailey J Didriksen
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily M Eshleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
3
|
Goto Y. Epithelial Cells as a Transmitter of Signals From Commensal Bacteria and Host Immune Cells. Front Immunol 2019; 10:2057. [PMID: 31555282 PMCID: PMC6724641 DOI: 10.3389/fimmu.2019.02057] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Intestinal epithelial cells (IECs) are non-hematopoietic cells that form a physical barrier against external antigens. Recent studies indicate that IECs have pleiotropic functions in the regulation of luminal microbiota and the host immune system. IECs produce various immune modulatory cytokines and chemokines in response to commensal bacteria and contribute to developing the intestinal immune system. In contrast, IECs receive cytokine signals from immune cells and produce various immunological factors against luminal bacteria. This bidirectional function of IECs is critical to regulate homeostasis of microbiota and the host immune system. Disruption of the epithelial barrier leads to detrimental host diseases such as inflammatory bowel disease, colonic cancer, and pathogenic infection. This review provides an overview of the functions and physiology of IECs and highlights their bidirectional functions against luminal bacteria and immune cells, which contribute to maintaining gut homeostasis.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan.,Division of Mucosal Symbiosis, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Gut Permeability and Glucose Absorption Are Affected at Early Stages of Graft Rejection in a Small Bowel Transplant Rat Model. Transplant Direct 2017; 3:e220. [PMID: 29184909 PMCID: PMC5682765 DOI: 10.1097/txd.0000000000000718] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 02/02/2023] Open
Abstract
Supplemental digital content is available in the text. Background Intestinal transplantation (ITx) faces many challenges due to the complexity of surgery and to the multiple immunological reactions that lead to the necessity of rigorous follow-up for early detection of acute cellular rejection (ACR). Our aim was to determine the kinetics of ACR using an experimental ITx model, with emphasis in the characterization of the process using different approaches, including the use of functional assays of absorptive and barrier function. Methods ITx in rats conducting serial sampling was performed. Clinical monitoring, graft histology, proinflammatory gene expression, and nitrosative stress determination were performed. Also, glucose absorption, barrier function using ovalbumin translocation, and contractile function were analyzed. Results The model used reproduced the different stages of ACR. Allogeneic ITx recipients showed signs of rejection from postoperative day (POD) 5, with increasing severity until 12 POD. Histological evaluation showed mild rejection in early sampling and severe rejection at late stages, with alterations in all graft layers. IL-6, CXCL 10, IFNg, and nitrite plasmas levels showed behavior coincident with histopathology. Remarkably, allogeneic grafts showed a marked alteration of glucose absorptive capacity from POD 5 that was sustained until endpoint. Coincidently, barrier function alteration was evidenced by luminal ovalbumin translocation to serum. Contractile function was progressively impaired along ACR. Conclusions Glucose absorption and barrier function are altered at early stages of ACR when histological alterations or gene expression changes were much subtle. This observation may provide simple evaluation tools that could be eventually translated to the clinics to contribute to early ACR diagnosis.
Collapse
|
5
|
Suff N, Waddington SN. The power of bioluminescence imaging in understanding host-pathogen interactions. Methods 2017; 127:69-78. [PMID: 28694065 DOI: 10.1016/j.ymeth.2017.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 01/06/2023] Open
Abstract
Infectious diseases are one of the leading causes of death worldwide. Modelling and understanding human infection is imperative to developing treatments to reduce the global burden of infectious disease. Bioluminescence imaging is a highly sensitive, non-invasive technique based on the detection of light, produced by luciferase-catalysed reactions. In the study of infectious disease, bioluminescence imaging is a well-established technique; it can be used to detect, localize and quantify specific immune cells, pathogens or immunological processes. This enables longitudinal studies in which the spectrum of the disease process and its response to therapies can be monitored. Light producing transgenic rodents are emerging as key tools in the study of host response to infection. Here, we review the strategies for identifying biological processes in vivo, including the technology of bioluminescence imaging and illustrate how this technique is shedding light on the host-pathogen relationship.
Collapse
Affiliation(s)
- Natalie Suff
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, United Kingdom.
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, United Kingdom
| |
Collapse
|
6
|
Wood MB, Rios D, Williams IR. TNF-α augments RANKL-dependent intestinal M cell differentiation in enteroid cultures. Am J Physiol Cell Physiol 2016; 311:C498-507. [PMID: 27413168 PMCID: PMC5129760 DOI: 10.1152/ajpcell.00108.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/12/2016] [Indexed: 01/13/2023]
Abstract
Microfold (M) cells are phagocytic intestinal epithelial cells in the follicle-associated epithelium of Peyer's patches that transport particulate antigens from the gut lumen into the subepithelial dome. Differentiation of M cells from epithelial stem cells in intestinal crypts requires the cytokine receptor activator of NF-κB ligand (RANKL) and the transcription factor Spi-B. We used three-dimensional enteroid cultures established with small intestinal crypts from mice as a model system to investigate signaling pathways involved in M cell differentiation and the influence of other cytokines on RANKL-induced M cell differentiation. Addition of RANKL to enteroids induced expression of multiple M cell-associated genes, including Spib, Ccl9 [chemokine (C-C motif) ligand 9], Tnfaip2 (TNF-α-induced protein 2), Anxa5 (annexin A5), and Marcksl1 (myristoylated alanine-rich protein kinase C substrate) in 1 day. The mature M cell marker glycoprotein 2 (Gp2) was strongly induced by 3 days and expressed by 11% of cells in enteroids. The noncanonical NF-κB pathway was required for RANKL-induced M cell differentiation in enteroids, as addition of RANKL to enteroids from mice with a null mutation in the mitogen-activated protein kinase kinase kinase 14 (Map3k14) gene encoding NF-κB-inducing kinase failed to induce M cell-associated genes. While the cytokine TNF-α alone had little, if any, effect on expression of M cell-associated genes, addition of TNF-α to RANKL consistently resulted in three- to sixfold higher levels of multiple M cell-associated genes than RANKL alone. One contributing mechanism is the rapid induction by TNF-α of Relb and Nfkb2 (NF-κB subunit 2), genes encoding the two subunits of the noncanonical NF-κB heterodimer. We conclude that endogenous activators of canonical NF-κB signaling present in the gut-associated lymphoid tissue microenvironment, including TNF-α, can play a supportive role in the RANKL-dependent differentiation of M cells in the follicle-associated epithelium.
Collapse
Affiliation(s)
- Megan B Wood
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel Rios
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ifor R Williams
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
7
|
Bolla P, Abraham A, Pérez P, de los Angeles Serradell M. Kefir-isolated bacteria and yeasts inhibit Shigella flexneri invasion and modulate pro-inflammatory response on intestinal epithelial cells. Benef Microbes 2016; 7:103-110. [DOI: 10.3920/bm2015.0061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The aim of this work was to evaluate the ability of a kefir-isolated microbial mixture containing three bacterial and two yeast strains (MM) to protect intestinal epithelial cells against Shigella flexneri invasion, as well as to analyse the effect on pro-inflammatory response elicited by this pathogen. A significant decrease in S. flexneri strain 72 invasion was observed on both HT-29 and Caco-2 cells pre-incubated with MM. Pre-incubation with the individual strains Saccharomyces cerevisiae CIDCA 8112 or Lactococcus lactis subsp. lactis CIDCA 8221 also reduced the internalisation of S. flexneri into HT-29 cells although in a lesser extent than MM. Interestingly, Lactobacillus plantarum CIDCA 83114 exerted a protective effect on the invasion of Caco-2 and HT-29 cells by S. flexneri. Regarding the pro-inflammatory response on HT-29 cells, S. flexneri infection induced a significant activation of the expression of interleukin 8 (IL-8), chemokine (C-C motif) ligand 20 (CCL20) and tumour necrosis factor alpha (TNF-α) encoding genes (P<0.05), whereas incubation of cells with MM did not induce the expression of any of the mediators assessed. Interestingly, pre-incubation of HT-29 monolayer with MM produced an inhibition of S. flexneri-induced IL-8, CCL20 and TNF-α mRNA expression. In order to gain insight on the effect of MM (or the individual strains) on this pro-inflammatory response, a series of experiments using a HT-29-NF-κB-hrGFP reporter system were performed. Pre-incubation of HT-29-NF-κB-hrGFP cells with MM significantly dampened Shigella-induced activation. Our results showed that the contribution of yeast strain Kluyveromyces marxianus CIDCA 8154 seems to be crucial in the observed effect. In conclusion, results presented in this study demonstrate that pre-treatment with a microbial mixture containing bacteria and yeasts isolated from kefir, resulted in inhibition of S. flexneri internalisation into human intestinal epithelial cells, along with the inhibition of the signalling via NF-κB that in turn led to the attenuation of the inflammatory response.
Collapse
Affiliation(s)
- P.A. Bolla
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, CP 1900, Argentina
- División Química Analítica, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, CP 1900, Argentina
| | - A.G. Abraham
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT-La Plata, CONICET, 47 y 116, La Plata, CP 1900, Argentina
- Área de Bromatología y Control de Alimentos, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, CP 1900, Argentina
| | - P.F. Pérez
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, CP 1900, Argentina
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT-La Plata, CONICET, 47 y 116, La Plata, CP 1900, Argentina
| | - M. de los Angeles Serradell
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, CP 1900, Argentina
| |
Collapse
|
8
|
Romanin DE, Llopis S, Genovés S, Martorell P, Ramón VD, Garrote GL, Rumbo M. Probiotic yeast Kluyveromyces marxianus CIDCA 8154 shows anti-inflammatory and anti-oxidative stress properties in in vivo models. Benef Microbes 2015; 7:83-93. [PMID: 26565081 DOI: 10.3920/bm2015.0066] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inflammatory bowel diseases (IBDs) are complex affections with increasing incidence worldwide. Multiple factors are involved in the development and maintenance of the symptoms including enhanced oxidative stress in intestinal mucosa. The conventional therapeutic approaches for IBDs are based on the use anti-inflammatory drugs with important collateral effects and partial efficacy. In the present work we tested the anti-inflammatory capacity of Kluyveromyces marxianus CIDCA 8154 in different models. In vitro, we showed that the pretreatment of epithelial cells with the yeast reduce the levels of intracellular reactive oxygen species. Furthermore, in a murine model of trinitro benzene sulfonic acid-induced colitis, yeast-treated animals showed a reduced histopathological score (P<0.05) and lower levels of circulating interleukin 6 (P<0.05). The capacity to modulate oxidative stress in vivo was assessed using a Caenorhabditis elegans model. The yeast was able to protect the nematodes from oxidative stress by modulating the SKN-1 transcription factor trough the DAF-2 pathway. These results indicate that K. marxianus CIDCA 8154 could control the intestinal inflammation and cellular oxidative stress. Deciphering the mechanisms of action of different probiotics might be useful for the rational formulation of polymicrobial products containing microorganisms targeting different anti-inflammatory pathways.
Collapse
Affiliation(s)
- D E Romanin
- 1 Instituto de Estudios en Inmunología y Fisiopatología (IIFP, UNLP-CONICET), Calle 47 y 115, 1900 La Plata, Argentina
| | - S Llopis
- 2 Laboratorio Biología Celular. Departamento Biotecnología Agroalimentaria, Biópolis, S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino 9, edificio 2, 46980 Paterna, Spain
| | - S Genovés
- 2 Laboratorio Biología Celular. Departamento Biotecnología Agroalimentaria, Biópolis, S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino 9, edificio 2, 46980 Paterna, Spain
| | - P Martorell
- 2 Laboratorio Biología Celular. Departamento Biotecnología Agroalimentaria, Biópolis, S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino 9, edificio 2, 46980 Paterna, Spain
| | - V D Ramón
- 2 Laboratorio Biología Celular. Departamento Biotecnología Agroalimentaria, Biópolis, S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino 9, edificio 2, 46980 Paterna, Spain
| | - G L Garrote
- 3 Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA, UNLP-CONICET), Calle 47 y 116, 1900 La Plata, Argentina
| | - M Rumbo
- 1 Instituto de Estudios en Inmunología y Fisiopatología (IIFP, UNLP-CONICET), Calle 47 y 115, 1900 La Plata, Argentina
| |
Collapse
|
9
|
Zambernardi A, Chiodetti A, Meier D, Cabanne A, Nachman F, Solar H, Rumbo C, Gondolesi GE, Rumbo M. Immunosuppressive therapies after intestinal transplant modulate the expression of Th1 signature genes during acute cellular rejection. Implications in the search for rejection biomarkers. Clin Transplant 2014; 28:1365-71. [DOI: 10.1111/ctr.12464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Agustina Zambernardi
- Laboratorio de Investigaciones del Sistema Inmune (LISIN); Facultad de Cs. Exactas; Universidad Nacional de La Plata; La Plata Argentina
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Ana Chiodetti
- Laboratorio de Investigaciones del Sistema Inmune (LISIN); Facultad de Cs. Exactas; Universidad Nacional de La Plata; La Plata Argentina
| | - Dominik Meier
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Ana Cabanne
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Fabio Nachman
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Héctor Solar
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Carolina Rumbo
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Gabriel E. Gondolesi
- Instituto de Transplante Multiorgánico; Fundación Favaloro; Buenos Aires Argentina
| | - Martin Rumbo
- Laboratorio de Investigaciones del Sistema Inmune (LISIN); Facultad de Cs. Exactas; Universidad Nacional de La Plata; La Plata Argentina
| |
Collapse
|
10
|
Iraporda C, Romanin DE, Rumbo M, Garrote GL, Abraham AG. The role of lactate on the immunomodulatory properties of the nonbacterial fraction of kefir. Food Res Int 2014. [DOI: 10.1016/j.foodres.2014.03.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
11
|
Airway structural cells regulate TLR5-mediated mucosal adjuvant activity. Mucosal Immunol 2014; 7:489-500. [PMID: 24064672 DOI: 10.1038/mi.2013.66] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/16/2013] [Accepted: 08/13/2013] [Indexed: 02/04/2023]
Abstract
Antigen-presenting cell (APC) activation is enhanced by vaccine adjuvants. Most vaccines are based on the assumption that adjuvant activity of Toll-like receptor (TLR) agonists depends on direct, functional activation of APCs. Here, we sought to establish whether TLR stimulation in non-hematopoietic cells contributes to flagellin's mucosal adjuvant activity. Nasal administration of flagellin enhanced T-cell-mediated immunity, and systemic and secretory antibody responses to coadministered antigens in a TLR5-dependent manner. Mucosal adjuvant activity was not affected by either abrogation of TLR5 signaling in hematopoietic cells or the presence of flagellin-specific, circulating neutralizing antibodies. We found that flagellin is rapidly degraded in conducting airways, does not translocate into lung parenchyma and stimulates an early immune response, suggesting that TLR5 signaling is regionalized. The flagellin-specific early response of lung was regulated by radioresistant cells expressing TLR5 (particularly the airway epithelial cells). Flagellin stimulated the epithelial production of a small set of mediators that included the chemokine CCL20, which is known to promote APC recruitment in mucosal tissues. Our data suggest that (i) the adjuvant activity of TLR agonists in mucosal vaccination may require TLR stimulation of structural cells and (ii) harnessing the effect of adjuvants on epithelial cells can improve mucosal vaccines.
Collapse
|
12
|
Susceptibility to Salmonella carrier-state: a possible Th2 response in susceptible chicks. Vet Immunol Immunopathol 2014; 159:16-28. [PMID: 24694400 DOI: 10.1016/j.vetimm.2014.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 01/13/2014] [Accepted: 03/02/2014] [Indexed: 11/23/2022]
Abstract
Infection of chicken with Salmonella may lead to a carrier-state characterized by the persistence of bacteria in the ceca for a long period of time and result in their excretion in feces. This excretion is the source of contamination of their congeners and food. During infection, enterocytes are the primary target cells for Salmonella, the producers of soluble factors which launch immune response and cells which are reciprocally responsive to surrounding immune cells. This study used microarrays to compare the gene expression profile during carrier-state of enterocytes purified from infected and control chicks which are either resistant or susceptible to Salmonella Enteritidis carrier-state. In total, we identified 271 genes significantly differentially expressed with an absolute fold change greater than 1.5. A global analysis determined interaction networks between differentially regulated genes. Using an a priori approach, our analyses focused on differentially expressed genes which were transcriptionally linked to cytokines playing a major role in the fate of the immune response. The expression of genes transcriptionally linked to type I interferon and TGF-β was down-regulated in infected chicks from both lines. Gene expression linked to the Th1 axis suggests the latter is inhibited in both lines. Finally, the expression of genes linked to IL-4, IL-5 and IL-13 indicates that susceptibility to carrier-state could be associated with a Th2 bias. Overall, these results highlight that the response to Salmonella during the acute phase and carrier-state is different and that enterocytes play a central role in this response.
Collapse
|
13
|
Crispo M, Van Maele L, Tabareau J, Cayet D, Errea A, Ferreira AM, Rumbo M, Sirard JC. Transgenic mouse model harboring the transcriptional fusion ccl20-luciferase as a novel reporter of pro-inflammatory response. PLoS One 2013; 8:e78447. [PMID: 24265691 PMCID: PMC3827052 DOI: 10.1371/journal.pone.0078447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/10/2013] [Indexed: 12/12/2022] Open
Abstract
The chemokine CCL20, the unique ligand of CCR6 functions as an attractant of immune cells. Expression of CCL20 is induced by Toll-like Receptor (TLR) signaling or proinflammatory cytokine stimulation. However CCL20 is also constitutively produced at specific epithelial sites of mucosa. This expression profile is achieved by transcriptional regulation. In the present work we characterized regulatory features of mouse Ccl20 gene. Transcriptional fusions between the mouse Ccl20 promoter and the firefly luciferase (luc) encoding gene were constructed and assessed in in vitro and in vivo assays. We found that liver CCL20 expression and luciferase activity were upregulated by systemic administration of the TLR5 agonist flagellin. Using shRNA and dominant negative form specific for mouse TLR5, we showed that this expression was controlled by TLR5. To address in situ the regulation of gene activity, a transgenic mouse line harboring a functional Ccl20-luc fusion was generated. The luciferase expression was highly concordant with Ccl20 expression in different tissues. Our data indicate that the transgenic mouse model can be used to monitor activation of innate response in vivo.
Collapse
Affiliation(s)
- Martina Crispo
- Unidad de Animales Transgénicos y de Experimentación – Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Laurye Van Maele
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
| | - Julien Tabareau
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
| | - Delphine Cayet
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
| | - Agustina Errea
- Laboratorio de Investigaciones del Sistema Inmune (LISIN) – National University of La Plata, Provincia de Buenos Aires, Argentina
| | - Ana María Ferreira
- Catedra de Inmunologia, Facultad de Ciencias/Facultad de Quimica, Universidad de la República, Montevideo, Uruguay
| | - Martin Rumbo
- Laboratorio de Investigaciones del Sistema Inmune (LISIN) – National University of La Plata, Provincia de Buenos Aires, Argentina
| | - Jean Claude Sirard
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
- * E-mail:
| |
Collapse
|
14
|
Franco MC, Golowczyc MA, De Antoni GL, Pérez PF, Humen M, Serradell MDLA. Administration of kefir-fermented milk protects mice against Giardia intestinalis infection. J Med Microbiol 2013; 62:1815-1822. [PMID: 24072759 DOI: 10.1099/jmm.0.068064-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Giardiasis, caused by the protozoan Giardia intestinalis, is one of the most common intestinal diseases worldwide and constitutes an important problem for the public health systems of various countries. Kefir is a probiotic drink obtained by fermenting milk with 'kefir grains', which consist mainly of bacteria and yeasts that coexist in a complex symbiotic association. In this work, we studied the ability of kefir to protect mice from G. intestinalis infection, and characterized the host immune response to this probiotic in the context of the intestinal infection. Six- to 8-week-old C75BL/6 mice were separated into four groups: controls, kefir mice (receiving 1 : 100 dilution of kefir in drinking water for 14 days), Giardia mice (infected orally with 4×10(7) trophozoites of G. intestinalis at day 7) and Giardia-kefir mice (kefir-treated G. intestinalis-infected mice), and killed at 2 or 7 days post-infection. Kefir administration was able to significantly reduce the intensity of Giardia infection at 7 days post-infection. An increase in the percentage of CD4(+) T cells at 2 days post-infection was observed in the Peyer's patches (PP) of mice belonging to the Giardia group compared with the control and kefir groups, while the percentage of CD4(+) T cells in PP in the Giardia-kefir group was similar to that of controls. At 2 days post-infection, a reduction in the percentage of B220-positive major histocompatibility complex class II medium cells in PP was observed in infected mice compared with the other groups. At 7 days post-infection, Giardia-infected mice showed a reduction in RcFcε-positive cells compared with the control group, suggesting a downregulation of the inflammatory response. However, the percentages of RcFcε-positive cells did not differ from controls in the kefir and Giardia-kefir groups. An increase in IgA-positive cells was observed in the lamina propria of the kefir group compared with controls at 2 days post-infection. Interestingly, the diminished number of IgA-positive cells registered in the Giardia group at 7 days post-infection was restored by kefir feeding, although the increase in IgA-positive cells was no longer observed in the kefir group at that time. No significant differences in CXCL10 expression were registered between groups, in concordance with the absence of inflammation in small-intestinal tissue. Interestingly, a slight reduction in CCL20 expression was observed in the Giardia group, suggesting that G. intestinalis might downregulate its expression as a way of evading the inflammatory immune response. On the other hand, a trend towards an increase in TNF-α expression was observed in the kefir group, while the Giardia-kefir group showed a significant increase in TNF-α expression. Moreover, kefir-receiving mice (kefir and Giardia-kefir groups) showed an increase in the expression of IFN-γ, the most relevant Th1 cytokine, at 2 days post-infection. Our results demonstrate that feeding mice with kefir reduces G. intestinalis infection and promotes the activation of different mechanisms of humoral and cellular immunity that are downregulated by parasitic infection, thus contributing to protection.
Collapse
Affiliation(s)
- Mariana Correa Franco
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, Argentina.,Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT La Plata - CONICET, 47 y 116, La Plata, Argentina
| | - Marina A Golowczyc
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT La Plata - CONICET, 47 y 116, La Plata, Argentina
| | - Graciela L De Antoni
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, Argentina.,Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT La Plata - CONICET, 47 y 116, La Plata, Argentina
| | - Pablo F Pérez
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, Argentina.,Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT La Plata - CONICET, 47 y 116, La Plata, Argentina
| | - Martín Humen
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA), CCT La Plata - CONICET, 47 y 116, La Plata, Argentina
| | - María de Los Angeles Serradell
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, Argentina
| |
Collapse
|
15
|
Tozaki K, Kimura J, Yasuda M, Ryu N, Nasu T, Pernthaner A, Hein WR. C6, a new monoclonal antibody, reacts with the follicle-associated epithelium of calf ileal Peyer's patches. J Vet Sci 2013; 14:1-6. [PMID: 23388432 PMCID: PMC3615225 DOI: 10.4142/jvs.2013.14.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/08/2012] [Indexed: 11/20/2022] Open
Abstract
The follicle-associated epithelium (FAE) of Peyer's patches (PPs) contains M cells that are important for reducing mucosal immune responses by transporting antigens into the underlying lymphoid tissue. We generated a monoclonal antibody (C6) that reacted with the FAE of calf ileal PPs, and analyzed the characteristics of C6 using immunohistochemistry and Western blotting. FAE of the ileal PP was stained with C6 during both late fetal developmental and postnatal stages. Neither the villous epithelial cell nor intestinal crypt basal cells were stained at any developmental stage. During the prenatal stages, FAE of the jejunal PP was C6-negative. However, a few C6-positive cells were distributed diffusely in some FAE of the jejunal PPs during the postnatal stages. The protein molecular weight of the antigen recognized by C6 was approximately 45 kDa. These data show that C6 is useful for identifying the FAE in ileal PPs and further suggest that differentiation of the FAE in these areas is independent of external antigens.
Collapse
Affiliation(s)
- Kana Tozaki
- Department of Veterinary Anatomy, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Intestinal epithelial cells as mediators of the commensal-host immune crosstalk. Immunol Cell Biol 2013; 91:204-14. [PMID: 23318659 DOI: 10.1038/icb.2012.80] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Commensal bacteria regulate the homeostasis of host effector immune cell subsets. The mechanisms involved in this commensal-host crosstalk are not well understood. Intestinal epithelial cells (IECs) not only create a physical barrier between the commensals and immune cells in host tissues, but also facilitate interactions between them. Perturbations of epithelial homeostasis or function lead to the development of intestinal disorders such as inflammatory bowel diseases (IBD) and intestinal cancer. IECs receive signals from commensals and produce effector immune molecules. IECs also affect the function of immune cells in the lamina propria. Here we discuss some of these properties of IECs that define them as innate immune cells. We focus on how IECs may integrate and transmit signals from individual commensal bacteria to mucosal innate and adaptive immune cells for the establishment of the unique mucosal immunological equilibrium.
Collapse
|
17
|
Pang G, Xie J, Chen Q, Hu Z. How functional foods play critical roles in human health. FOOD SCIENCE AND HUMAN WELLNESS 2012. [DOI: 10.1016/j.fshw.2012.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Hsieh EH, Lo DD. Jagged1 and Notch1 help edit M cell patterning in Peyer's patch follicle epithelium. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 37:306-312. [PMID: 22504165 PMCID: PMC3374009 DOI: 10.1016/j.dci.2012.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/06/2012] [Accepted: 04/07/2012] [Indexed: 05/31/2023]
Abstract
Mucosal epithelium M cells are dispersed across Peyer's patch follicle associated epithelium (PPFAE) with minimal clustering. Since Notch signaling can influence patterning in epithelia, we examined its influence on PPFAE M cell distribution. Conditional deletion of Notch1 in intestinal epithelium increased PPFAE M cells and also increased M cell clustering, implying a role for Notch in both M cell numbers and lateral inhibition. By contrast, conditional deletion of the ligand Jagged1 also increased M cell clustering, but with a paradoxical decrease in M cell density. In vitro, inhibition of Notch signaling reduced expression of an M cell associated gene CD137, consistent with cis-promoting effects on M cell development. Thus, Jagged1 may have a cis-promoting role in committed M cells, but a trans-inhibitory effect on neighboring cells. In sum, Jagged1-Notch signaling may edit the pattern of M cells across the PPFAE, which may help optimize mucosal immune surveillance.
Collapse
Affiliation(s)
- En-Hui Hsieh
- Division of Biomedical Sciences, University of California Riverside, CA 92521, United States
| | | |
Collapse
|
19
|
Naturally occurring motility-defective mutants of Salmonella enterica serovar Enteritidis isolated preferentially from nonhuman rather than human sources. Appl Environ Microbiol 2011; 77:7740-8. [PMID: 21926214 DOI: 10.1128/aem.05318-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonellosis represents a worldwide health problem because it is one of the major causes of food-borne disease. Although motility is postulated as an important Salmonella virulence attribute, there is little information about variation in motility in natural isolates. Here we report the identification of a point mutation (T551 → G) in motA, a gene essential for flagellar rotation, in several Salmonella enterica serovar Enteritidis field isolates. This mutation results in bacteria that can biosynthesize structurally normal but paralyzed flagella and are impaired in their capacity to invade human intestinal epithelial cells. Introduction of a wild-type copy of motA into one of these isolates restored both motility and cell invasiveness. The motA mutant triggered higher proinflammatory transcriptional responses than an aflagellate isolate in differentiated Caco-2 cells, suggesting that the paralyzed flagella are able to signal through pattern recognition receptors. A specific PCR was designed to screen for the T551 → G mutation in a collection of 266 S. Enteritidis field isolates from a nationwide epidemic, comprising 194 from humans and 72 from other sources. We found that 72 of the 266 (27%) isolates were nonmotile, including 24.7% (48/194) of human and 33.3% (24/72) of food isolates. Among nonmotile isolates, 15 carried the T551 → G mutation and, significantly, 13 were recovered from food, including 7 from eggs, but only 2 were from human sources. These results suggest that the presence of paralyzed flagella may impair the ability of S. Enteritidis to cause disease in the human host but does not prevent its ability to colonize chickens and infect eggs.
Collapse
|
20
|
CCR6 as a mediator of immunity in the lung and gut. Exp Cell Res 2011; 317:613-9. [PMID: 21376174 DOI: 10.1016/j.yexcr.2010.12.018] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 12/20/2010] [Accepted: 12/20/2010] [Indexed: 12/11/2022]
Abstract
Chemokines are key mediators of leukocyte recruitment during pathogenic insult and also play a prominent role in homeostasis. While most chemokine receptors bind to multiple chemokines, CCR6 is unique in that this receptor is one of only a few that can bind only a single chemokine ligand, CCL20. CCR6 is an important receptor that is involved in regulating several aspects of mucosal immunity, including the ability to mediate the recruitment of immature dendritic cells (DCs) and mature DCs, and professional antigen presenting cells (APCs) to the sites of epithelial inflammation. Further, CCR6 mediates the homing of both CD4(+) T (T-helper; Th) cells and DCs to the gut mucosal lymphoid tissue. DCs, which are known to be essential immune cells in innate immunity and in the initiation of adaptive immunity, play a central role in initiating a primary immune response. Herein, we summarize the role of CCR6 in immune responses at epithelial and mucosal sites in both the lung and gut based on a review of the current literature.
Collapse
|
21
|
Hiriart Y, Errea A, González Maciel D, Lopez JC, Rumbo M. A method for the purification of bacterial flagellin that allows simple upscaling. World J Microbiol Biotechnol 2011; 28:15-21. [DOI: 10.1007/s11274-011-0786-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 05/09/2011] [Indexed: 10/25/2022]
|
22
|
Yim L, Betancor L, Martinez A, Giossa G, Bryant C, Maskell D, Chabalgoity JA. Differential phenotypic diversity among epidemic-spanning Salmonella enterica serovar enteritidis isolates from humans or animals. Appl Environ Microbiol 2010; 76:6812-20. [PMID: 20802078 PMCID: PMC2953042 DOI: 10.1128/aem.00497-10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 08/14/2010] [Indexed: 11/20/2022] Open
Abstract
Nontyphoidal salmonellae are major causes of food-borne disease worldwide. In Uruguay, Salmonella enterica serovar Enteritidis was the most commonly isolated serovar throughout the last decade, with a marked epidemic period between 1995 and 2004. In a previous study, we conducted comparative genomics of 29 epidemic-spanning S. Enteritidis field isolates, and here we evaluated the pathogenic potential of the same set of isolates using several phenotypic assays. The sample included 15 isolates from human gastroenteritis, 5 from invasive disease, and 9 from nonhuman sources. Contrary to the genetic homogeneity previously observed, we found great phenotypic variability among these isolates. One-third of them were defective in at least one assay, namely, 10 isolates were defective in motility, 8 in invasion of Caco-2 cells, and 10 in survival in egg albumen. Twelve isolates were tested for invasiveness in 3-day-old chickens, and five of these were significantly less invasive than the reference strain. The two oldest preepidemic isolates were reduced in fitness in all assays, providing a plausible explanation for the previous negligible incidence of S. Enteritidis in Uruguay and supporting the view that the introduction or emergence of a more virulent strain was responsible for the marked rise of this serovar. Further, we found differences in fitness among the isolates which depended on the source of isolation. A total of 1 out of 14 isolates from human gastroenteritis, but 6 out of 13 isolates from other sources, was impaired in at least two assays, suggesting enhanced fitness among strains able to cause intestinal disease in humans.
Collapse
Affiliation(s)
- Lucía Yim
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Laura Betancor
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Arací Martinez
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Gerardo Giossa
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Clare Bryant
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Duncan Maskell
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Jose A. Chabalgoity
- Department of Biotechnology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Bacteriology and Virology, School of Medicine, Universidad de la República, Instituto de Higiene, Av. A. Navarro 3051, CP 11600, Montevideo, Uruguay, Department of Avian Pathology, Veterinary School, Universidad de la República, Montevideo, Uruguay, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
23
|
Hamm CM, Reimers MA, McCullough CK, Gorbe EB, Lu J, Gu CC, Li E, Dieckgraefe BK, Gong Q, Stappenbeck TS, Stone CD, Dietz DW, Hunt SR. NOD2 status and human ileal gene expression. Inflamm Bowel Dis 2010; 16:1649-57. [PMID: 20155851 PMCID: PMC4002261 DOI: 10.1002/ibd.21208] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND NOD2 single nucleotide polymorphisms have been associated with increased risk of ileal Crohn's disease (CD). This exploratory study was conducted to compare ileal mucosal gene expression in CD patients with and without NOD2 risk alleles. METHODS Ileal samples were prospectively collected from 18 nonsmoking CD patients not treated with anti-TNF-α biologics and 9 nonsmoking control patients without inflammatory bowel disease undergoing initial resection and genotyped for the 3 major NOD2 risk alleles (Arg702Trp, Gly908Arg, Leu1007fs). Microarray analysis was performed in samples from 4 NOD2(R) (at least 1 risk allele) CD patients, 4 NOD2(NR) (no risk alleles) CD patients, and 4 NOD2(NR) controls. Candidate genes selected by significance analysis of microarrays (SAM) were confirmed by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assays of all the samples. RESULTS SAM detected upregulation of 18 genes in affected ileum in NOD2(R) compared to NOD2(NR) CD patients, including genes related to lymphocyte activation. SAM also detected altered ileal gene expression in unaffected NOD2(NR) ileal mucosal CD samples compared to NOD2(NR) control samples. qRT-PCR conducted on all the samples confirmed that increased CD3D expression in affected samples was associated with NOD2(R) status, and that increased MUC1, DUOX2, DMBT1 and decreased C4orf7 expression in unaffected samples was associated with CD, independent of NOD2 status. CONCLUSIONS The results support the concept that NOD2 risk alleles contribute to impaired regulation of inflammation in the ileum. Furthermore, altered ileal gene expression, independent of NOD2 status, is detected in the unaffected proximal margin of resected ileum from CD patients.
Collapse
Affiliation(s)
- Christina M Hamm
- Division of Gastroenterology, Washington University, St Louis School of Medicine, St Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hsieh EH, Fernandez X, Wang J, Hamer M, Calvillo S, Croft M, Kwon BS, Lo DD. CD137 is required for M cell functional maturation but not lineage commitment. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:666-76. [PMID: 20616340 DOI: 10.2353/ajpath.2010.090811] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mucosal immune surveillance depends on M cells that reside in the epithelium overlying Peyer's patch and nasopharyngeal associated lymphoid tissue to transport particles to underlying lymphocytes. M cell development is associated with B lymphocytes in a basolateral pocket, but the interactions between these cells are poorly understood. In a cell culture model of M cell differentiation, we found lymphotoxin/tumor necrosis factor alpha induction of CD137 (TNFRSF9) protein on intestinal epithelial cell lines, raising the possibility that CD137 on M cells in vivo might interact with CD137L expressed by B cells. Accordingly, while CD137-deficient mice produced UEA-1+ M cell progenitors in nasopharyngeal associated lymphoid tissue and Peyer's patch epithelium, they showed an abnormal morphology, including the absence of basolateral B cell pockets. More important, CD137-deficient nasopharyngeal associated lymphoid tissue M cells were defective in microparticle transcytosis. Bone marrow irradiation chimeras confirmed that while induction of UEA-1+ putative M cell precursors was not CD137-dependent, full M cell transcytosis function required expression of CD137 by radioresistant stromal cells as well as by bone marrow-derived cells. These results are consistent with a two-step model of M cell differentiation, with initial CD137-independent commitment to the M cell lineage followed by a CD137-CD137L interaction of M cells with CD137-activated B lymphocytes or dendritic cells for functional maturation.
Collapse
Affiliation(s)
- En Hui Hsieh
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Down-regulation of intestinal epithelial innate response by probiotic yeasts isolated from kefir. Int J Food Microbiol 2010; 140:102-8. [DOI: 10.1016/j.ijfoodmicro.2010.04.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 04/13/2010] [Accepted: 04/15/2010] [Indexed: 12/30/2022]
|
26
|
Blázquez AB, Knight AK, Getachew H, Bromberg JS, Lira SA, Mayer L, Berin MC. A functional role for CCR6 on proallergic T cells in the gastrointestinal tract. Gastroenterology 2010; 138:275-84.e1-4. [PMID: 19782082 PMCID: PMC2813342 DOI: 10.1053/j.gastro.2009.09.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 09/02/2009] [Accepted: 09/11/2009] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS CCL20 is a chemokine that regulates the homeostatic and inflammatory trafficking of leukocytes to the small intestine and regulates the development of the gastrointestinal lymphoid architecture. T cells expressing T helper cell (Th) 2 cytokines are critical for experimental food allergy, and we hypothesized that CCL20 is involved in the localization of these cells to the gut. METHODS We evaluated the role of CCR6 in allergic diarrhea induced by sensitization and oral challenge with ovalbumin (OVA) using CCR6(+/+) and CCR6(-/-) mice. RESULTS CCR6(-/-) mice were protected from OVA-induced diarrhea but surprisingly were not impaired in mastocytosis or allergen-specific immunoglobulin E. CCR6(-/-) mice were also protected from T cell-mediated diarrhea induced by anti-CD3 antibody. Allergic diarrhea was associated with an increased expression of Th2 cytokines within the intestinal mucosa that was significantly reduced in CCR6(-/-) mice. Inhibition of lymphocyte homing by treatment with FTY720 did not impair allergic diarrhea, indicating that reactivation of T cells could occur locally within the small intestine. Finally, T-cell transfer studies demonstrated that CCR6 was required both on the transferred T cells and in the recipient mouse to manifest allergic disease in the gastrointestinal tract. CONCLUSIONS These studies highlight a mast cell- and immunoglobulin E-independent role for CCR6-bearing T cells in the pathogenesis of gastrointestinal allergic disease.
Collapse
Affiliation(s)
- Ana Belén Blázquez
- Jaffe Food Allergy Institute, Mount Sinai School of Medicine, New York, NY
| | - Adina Kay Knight
- Jaffe Food Allergy Institute, Mount Sinai School of Medicine, New York, NY
| | | | | | - Sergio A. Lira
- Immunology Institute, Mount Sinai School of Medicine, New York, NY
| | - Lloyd Mayer
- Immunology Institute, Mount Sinai School of Medicine, New York, NY
| | - M. Cecilia Berin
- Jaffe Food Allergy Institute, Mount Sinai School of Medicine, New York, NY, Immunology Institute, Mount Sinai School of Medicine, New York, NY
| |
Collapse
|
27
|
Wang J, Lopez-Fraga M, Rynko A, Lo DD. TNFR and LTbetaR agonists induce follicle-associated epithelium and M cell specific genes in rat and human intestinal epithelial cells. Cytokine 2009; 47:69-76. [PMID: 19501526 PMCID: PMC2756805 DOI: 10.1016/j.cyto.2009.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 04/13/2009] [Accepted: 05/07/2009] [Indexed: 02/08/2023]
Abstract
M cells assist mucosal immune surveillance by transcytosis of particles to underlying lymphoid tissue, but the mechanisms of M cell differentiation are poorly understood. To develop a better defined cell culture model of M cell differentiation, we treated human (Caco-2BBe) and rat (IEC-6) intestinal epithelial cell lines with lymphotoxin beta receptor (LTbetaR) and TNF receptor (TNFR) agonists. Treated cells were studied for regulation of genes associated with M cell and follicle-associated epithelium (FAE). We found that LTbetaR and TNFR agonists induce transcription of FAE-specific genes (Ccl20 and Lamb3) in Caco2-BBe cells and IEC-6 cells as well as rodent M cell specific genes such as Sgne-1/Scg5, Cldn4, and Gp2. The cytokines have distinct but complementary effects; TNFR agonists mainly induced FAE-specific genes, while the LTbetaR agonist induced M cell specific genes. The combination of cytokines showed additive induction of the FAE-associated Ccl20, Lamb3 and a surprising induction of CD137/Tnfrsf9. On the other hand TNF agonists appeared to suppress expression of some LTbetaR-induced genes. Functionally, cytokine treatment led to the reorganization of microvilli and Claudin-4 redistribution. These studies suggest complex interactions between these cytokines in the context of either inflammation or tissue differentiation.
Collapse
Affiliation(s)
- Jing Wang
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
| | - Marta Lopez-Fraga
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
- Sylentis, Madrid, Spain
| | - Abby Rynko
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jollla, CA 92037
| | - David D. Lo
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jollla, CA 92037
| |
Collapse
|
28
|
Finke D. Induction of intestinal lymphoid tissue formation by intrinsic and extrinsic signals. Semin Immunopathol 2009; 31:151-69. [PMID: 19506873 DOI: 10.1007/s00281-009-0163-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 05/20/2009] [Indexed: 12/20/2022]
Abstract
Since the discovery of inducer cells as a separate lineage for organogenesis of Peyer's patches in the small intestine of fetal mice, a lot of progress has been made in understanding the molecular pathways involved in the generation of lymphoid tissue and the maintenance of the lymphoid architecture. The findings that inducer cells also exist in adult mice and in humans, have a lineage relationship to natural killer cells, and can be stimulated during infections highlight their possible role in establishing innate and adaptive immune responses. Novel concepts in the development of intestinal lymphoid tissues have been made in the past few years suggesting that lymphoid organs are more plastic as previously thought and depend on antigenic stimulation. In addition, the generation of novel lymphoid organs in the gut under inflammatory conditions indicates a function in chronic diseases. The present review summarizes current knowledge on the basic framework of signals required for developing lymphoid tissue under normal and inflammatory conditions.
Collapse
Affiliation(s)
- Daniela Finke
- Department of Biomedicine, Developmental Immunology, University of Basel, Basel, Switzerland.
| |
Collapse
|
29
|
Sirard JC, Didierlaurent A, Cayet D, Sierro F, Rumbo M. Toll-like receptor 5- and lymphotoxin beta receptor-dependent epithelial Ccl20 expression involves the same NF-kappaB binding site but distinct NF-kappaB pathways and dynamics. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2009; 1789:386-94. [PMID: 19303953 DOI: 10.1016/j.bbagrm.2009.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 02/24/2009] [Accepted: 03/06/2009] [Indexed: 10/21/2022]
Abstract
Canonical and alternative NF-kappaB pathways depend on distinct NF-kappaB members and regulate expression of different gene subset in inflammatory and steady state conditions, respectively. In intestinal epithelial cells, both pathways control the transcription of the gene coding the CCL20 chemokine. Lymphotoxin beta receptor (LTbetaR) mediates long lasting CCL20 expression whereas Toll-like receptor 5 (TLR5) signals promote inducible and transient activation. Here, we investigated whether the regulation of ccl20 expression involves different promoter sites and NF-kappaB molecules in response to TLR5 and LTbetaR stimulation. In epithelial cells, both stimulation required the same promoter regions, especially the NF-kappaB binding site but involved different NF-kappaB isoforms: p65/p50 and p52/RelB, for TLR5 and LTbetaR-dependent activation, respectively. The dynamic of activation and interaction with CCL20-specific NF-kappaB site correlated with gene transcription. Similar Ccl20 expression and NF-kappaB activation was found in the small intestine of mice stimulated with TLR5 and LTbetaR agonists. In summary, different NF-kappaB pathways modulate CCL20 transcription by operating on the same NF-kappaB binding site in the same cell type.
Collapse
Affiliation(s)
- Jean-Claude Sirard
- Institut National de la Santé et de la Recherche Médicale U801, Institut Pasteur de Lille, Université de Lille 2, Lille, France
| | | | | | | | | |
Collapse
|
30
|
Contribution of flagellin pattern recognition to intestinal inflammation during Salmonella enterica serotype typhimurium infection. Infect Immun 2009; 77:1904-16. [PMID: 19237529 DOI: 10.1128/iai.01341-08] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serotype Typhimurium causes acute inflammatory diarrhea in humans. Flagella contribute to intestinal inflammation, but the mechanism remains unclear since most mutations abrogating pattern recognition of flagellin also prevent motility and reduce bacterial invasion. To determine the contribution of flagellin pattern recognition to the generation of innate immune responses, we compared in two animal models a nonmotile, but flagellin-expressing and -secreting serotype Typhimurium strain (flgK mutant) to a nonmotile, non-flagellin-expressing strain (flgK fliC fljB mutant). In vitro, caspase-1 can be activated by cytosolic delivery of flagellin, resulting in release of the interferon gamma inducing factor interleukin-18 (IL-18). Experiments with streptomycin-pretreated caspase-1-deficient mice suggested that induction of gamma interferon expression in the murine cecum early (12 h) after serotype Typhimurium infection was caspase-1 dependent but independent of flagellin pattern recognition. In addition, mRNA levels of the CXC chemokines macrophage inflammatory protein 2 and keratinocyte-derived chemokine were markedly increased early after serotype Typhimurium infection of streptomycin-pretreated wild-type mice regardless of flagellin expression. In contrast, in bovine ligated ileal loops, flagellin pattern recognition contributed to increased mRNA levels of macrophage inflammatory protein 3alpha and more fluid accumulation at 2 h after infection. Collectively, our data suggest that pattern recognition of flagellin contributes to early innate host responses in the bovine ileal mucosa but not in the murine cecal mucosa.
Collapse
|
31
|
Verbrugghe P, Kujala P, Waelput W, Peters PJ, Cuvelier CA. Clusterin in human gut-associated lymphoid tissue, tonsils, and adenoids: localization to M cells and follicular dendritic cells. Histochem Cell Biol 2007; 129:311-20. [PMID: 18097679 DOI: 10.1007/s00418-007-0369-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2007] [Indexed: 01/29/2023]
Abstract
The follicle-associated epithelium (FAE) overlying the follicles of mucosa-associated lymphoid tissue is a key player in the initiation of mucosal immune responses. We recently reported strong clusterin expression in the FAE of murine Peyer's patches. In this study, we examined the expression of clusterin in the human gut-associated lymphoid tissue (GALT) and Waldeyer's ring. Immunohistochemistry for clusterin in human Peyer's patches, appendix and colon lymphoid follicles revealed expression in M cells and in follicular dendritic cells (FDCs). Using cryo-immunogold electron microscopy in Peyer's patches, we observed cytosolic immunoreactivity in M cells and labeling in the ER/Golgi biosynthetic pathway in FDCs. In palatine tonsils and adenoids, we demonstrated clusterin expression in germinal centers and in the lymphoepithelium in the crypts where M cells are localized. In conclusion, clusterin is expressed in M cells and follicular dendritic cells at inductive sites of human mucosa-associated lymphoid tissue suggesting a role for this protein in innate immune responses. Moreover, the use of clusterin as a human M cell marker could prove to be a valuable tool in future M cell research.
Collapse
Affiliation(s)
- Phebe Verbrugghe
- Department of Pathology, University Hospital Ghent, De Pintelaan 185, Ghent, Belgium
| | | | | | | | | |
Collapse
|
32
|
Ott LW, Resing KA, Sizemore AW, Heyen JW, Cocklin RR, Pedrick NM, Woods HC, Chen JY, Goebl MG, Witzmann FA, Harrington MA. Tumor Necrosis Factor-alpha- and interleukin-1-induced cellular responses: coupling proteomic and genomic information. J Proteome Res 2007; 6:2176-85. [PMID: 17503796 PMCID: PMC2877378 DOI: 10.1021/pr060665l] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pro-inflammatory cytokines, Tumor Necrosis Factor-alpha (TNFalpha) and Interleukin-1 (IL-1) mediate the innate immune response. Dysregulation of the innate immune response contributes to the pathogenesis of cancer, arthritis, and congestive heart failure. TNFalpha- and IL-1-induced changes in gene expression are mediated by similar transcription factors; however, TNFalpha and IL-1 receptor knock-out mice differ in their sensitivities to a known initiator (lipopolysaccharide, LPS) of the innate immune response. The contrasting responses to LPS indicate that TNFalpha and IL-1 regulate different processes. A large-scale proteomic analysis of TNFalpha- and IL-1-induced responses was undertaken to identify processes uniquely regulated by TNFalpha and IL-1. When combined with genomic studies, our results indicate that TNFalpha, but not IL-1, mediates cell cycle arrest.
Collapse
Affiliation(s)
- Lee W Ott
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, MS 4053, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The CCR6 chemokine receptor is expressed by most B cells and subsets of T cells and dendritic cells (DCs) found in the gut mucosal immune system. CCL20, the single chemokine ligand for CCR6, is selectively made by the follicle-associated epithelium (FAE) overlying Peyer's patches (PPs) and isolated lymphoid follicles (ILFs). CCL20 contributes to the recruitment of CCR6-expressing B cells to these structures. CCL20 expression by the intestinal epithelium is also highly inducible in response to inflammatory stimuli. Thus, CCL20 functions as both an inflammatory and homeostatic chemokine. Interactions between CCR6 and CCL20 play a role at several stages in the development of intestinal lymphoid structures. A subset of the c-kit(+) lymphoid precursors found in cryptopatches (CPs) expresses CCR6. Recruitment of B cells to CPs and the subsequent expansion and organization of these B cells allows differentiation of some of these structures into ILFs. In CCR6 knockout mice, PPs are smaller with fewer follicles and the development of ILFs is compromised. These defects in the development of mucosal inductive sites in CCR6-deficient mice are responsible for decreased IgA production to oral antigens. CCR6 can be included with CXCR5 and CCR7 in a list of chemokine receptors that participate in shaping the organized lymphoid structures that are part of the intestinal immune system.
Collapse
Affiliation(s)
- Ifor R Williams
- Department of Pathology, Emory University School of Medicine, Whitehead 105D, 615 Michael Street, Atlanta, GA 30322, USA.
| |
Collapse
|
34
|
Gullberg E, Söderholm JD. Peyer's Patches and M Cells as Potential Sites of the Inflammatory Onset in Crohn's Disease. Ann N Y Acad Sci 2006; 1072:218-32. [PMID: 17057202 DOI: 10.1196/annals.1326.028] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Clinical observations suggest that the sites of initial inflammation in ileal Crohn's disease (CD) are the lymphoid follicles, where the aphtoid lesions originate from small erosions of the follicle-associated epithelium (FAE). Lymphoid follicles and Peyer's patches (PPs) consist of a number of B-cell follicles with intervening T cell areas. The T cell follicular area is also populated by dendritic cells (DCs) and macrophages. A single layer of epithelial cells covering each follicle forms a dome between the surrounding villi. This FAE differs from normal villus epithelium in several ways that make the epithelial cells of the FAE more exposed to the luminal contents, more accessible to antigens, and in closer contact with the immune system. The most prominent feature is the presence of specialized M cells, which are optimized for antigen adherence and transport. M cells play an important role in the surveillance of the intestinal lumen, but also provide a route of entry for various pathogens. In this article we review the current knowledge on the epithelial phenotype of the human FAE, and changes of the FAE and M cells in intestinal inflammation, leading to a hypothesis of the role of the FAE and M cells in the pathogenesis of CD.
Collapse
Affiliation(s)
- Elisabet Gullberg
- Colorectal Surgery Unit, Department of Surgery, University Hospital, SE-581 85 Linköping, Sweden
| | | |
Collapse
|
35
|
Rumbo M, Nempont C, Kraehenbuhl JP, Sirard JC. Mucosal interplay among commensal and pathogenic bacteria: lessons from flagellin and Toll-like receptor 5. FEBS Lett 2006; 580:2976-84. [PMID: 16650409 DOI: 10.1016/j.febslet.2006.04.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Accepted: 04/08/2006] [Indexed: 12/19/2022]
Abstract
Toll-like receptors (TLR) detect pathogen-associated molecular patterns (PAMP) and play a crucial role in triggering immunity. Due to their large surfaces in direct contact with the environment, mucosal tissues are the major sites of PAMP-TLR signalling. How innate and adaptive immunity are triggered through flagellin-TLR5 interaction is the main focus of the review. In view of recent reports on genetic polymorphism, we will summarize the impact of TLR5 on the susceptibility to mucosal infections and on various immuno-pathologies. Finally, the contribution of TLRs in the induction and maintenance of mucosal homeostasis and commensal discrimination is discussed.
Collapse
Affiliation(s)
- Martin Rumbo
- UNLP, Laboratorio de Investigaciones en el Sistema Inmune, Facultad de Ciencias Exactas, La Plata, Argentina
| | | | | | | |
Collapse
|
36
|
Hase K, Murakami T, Takatsu H, Shimaoka T, Iimura M, Hamura K, Kawano K, Ohshima S, Chihara R, Itoh K, Yonehara S, Ohno H. The membrane-bound chemokine CXCL16 expressed on follicle-associated epithelium and M cells mediates lympho-epithelial interaction in GALT. THE JOURNAL OF IMMUNOLOGY 2006; 176:43-51. [PMID: 16365394 DOI: 10.4049/jimmunol.176.1.43] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The recently identified CXCL16 has dual functions as a transmembrane adhesion molecule and a soluble chemokine. In this study we found that CXCL16 mRNA and protein were expressed constitutively on the follicle-associated epithelium covering Peyer's patches (PPs), isolated lymphoid follicles, and cecal patches, but minimally on the villous epithelium in the murine gastrointestinal tract. The CXCL16 receptor CXCR6/Bonzo was constitutively expressed on subpopulations of CD4+ and CD8+ T cells isolated from PPs. The expression of CXCR6/Bonzo on the PP T cells was up-regulated after stimulation with anti-CD3 and anti-CD28 mAbs. The activated PP T cells showed chemotactic migration in response to the soluble N-terminal chemokine domain of CXCL16. Furthermore, the activated PP T cells selectively adhered to cells expressing murine CXCL16. To determine the physiological role of CXCL16 in GALT, we first carefully analyzed T cell distribution in PPs. T cells localized not only in the interfollicular region but also at a lesser frequency in the subepithelial dome (SED) and in the germinal center of lymphoid follicles. Consistently, the majority of the adoptive transferred activated T cells migrated into the SED and the interfollicular region. However, the neutralization of CXCL16 specifically reduced the migration of the adoptive, transferred, activated T cells into the SED of PPs. These data suggest that CXCL16 expressed on the follicle-associated epithelium plays an important role in the recruitment and retention of activated T cells in the SED and should, at least partially, be responsible for lymphocyte compartmentalization in GALT.
Collapse
Affiliation(s)
- Koji Hase
- Research Center for Allergy and Immunology, RIKEN, Yokohama, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|