1
|
Dillon A, Lo DD. M Cells: Intelligent Engineering of Mucosal Immune Surveillance. Front Immunol 2019; 10:1499. [PMID: 31312204 PMCID: PMC6614372 DOI: 10.3389/fimmu.2019.01499] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
M cells are specialized intestinal epithelial cells that provide the main machinery for sampling luminal microbes for mucosal immune surveillance. M cells are usually found in the epithelium overlying organized mucosal lymphoid tissues, but studies have identified multiple distinct lineages of M cells that are produced under different conditions, including intestinal inflammation. Among these lineages there is a common morphology that helps explain the efficiency of M cells in capturing luminal bacteria and viruses; in addition, M cells recruit novel cellular mechanisms to transport the particles across the mucosal barrier into the lamina propria, a process known as transcytosis. These specializations used by M cells point to a novel engineering of cellular machinery to selectively capture and transport microbial particles of interest. Because of the ability of M cells to effectively violate the mucosal barrier, the circumstances of M cell induction have important consequences. Normal immune surveillance insures that transcytosed bacteria are captured by underlying myeloid/dendritic cells; in contrast, inflammation can induce development of new M cells not accompanied by organized lymphoid tissues, resulting in bacterial transcytosis with the potential to amplify inflammatory disease. In this review, we will discuss our own perspectives on the life history of M cells and also raise a few questions regarding unique aspects of their biology among epithelia.
Collapse
Affiliation(s)
- Andrea Dillon
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
2
|
Saxena VK, Diaz A, Scheerlinck JPY. Identification and characterization of an M cell marker in nasopharynx- and oropharynx-associated lymphoid tissue of sheep. Vet Immunol Immunopathol 2019; 208:1-5. [DOI: 10.1016/j.vetimm.2018.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 01/10/2023]
|
3
|
Vigilance or Subversion? Constitutive and Inducible M Cells in Mucosal Tissues. Trends Immunol 2017; 39:185-195. [PMID: 28958392 DOI: 10.1016/j.it.2017.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 02/08/2023]
Abstract
Microfold (M) cells are epithelial cells present in mucosal tissues and specialized for the capture of luminal microparticles and their delivery to underlying immune cells; thus, they are crucial participants in mucosal immune surveillance. Multiple phenotypic subsets of M cells have now been described, all sharing a unique apical morphology that provides clues to their ability to capture microbial particles. The existence of diverse M cell phenotypes, especially inflammation-inducible M cells, provides an intriguing puzzle: some variants may augment luminal surveillance to boost mucosal immunity, while others may promote microbial access to tissues. Here, I consider the unique induction requirements of each M cell subset and functional differences, highlighting the potentially distinct consequences in mucosal immunity.
Collapse
|
4
|
Haines RA, Urbiztondo RA, Haynes RAH, Simpson E, Niewiesk S, Lairmore MD. Characterization of New Zealand White Rabbit Gut-Associated Lymphoid Tissues and Use as Viral Oncology Animal Model. ILAR J 2017; 57:34-43. [PMID: 27034393 DOI: 10.1093/ilar/ilw004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Rabbits have served as a valuable animal model for the pathogenesis of various human diseases, including those related to agents that gain entry through the gastrointestinal tract such as human T cell leukemia virus type 1. However, limited information is available regarding the spatial distribution and phenotypic characterization of major rabbit leukocyte populations in mucosa-associated lymphoid tissues. Herein, we describe the spatial distribution and phenotypic characterization of leukocytes from gut-associated lymphoid tissues (GALT) from 12-week-old New Zealand White rabbits. Our data indicate that rabbits have similar distribution of leukocyte subsets as humans, both in the GALT inductive and effector sites and in mesenteric lymph nodes, spleen, and peripheral blood. GALT inductive sites, including appendix, cecal tonsil, Peyer's patches, and ileocecal plaque, had variable B cell/T cell ratios (ranging from 4.0 to 0.8) with a predominance of CD4 T cells within the T cell population in all four tissues. Intraepithelial and lamina propria compartments contained mostly T cells, with CD4 T cells predominating in the lamina propria compartment and CD8 T cells predominating in the intraepithelial compartment. Mesenteric lymph node, peripheral blood, and splenic samples contained approximately equal percentages of B cells and T cells, with a high proportion of CD4 T cells compared with CD8 T cells. Collectively, our data indicate that New Zealand White rabbits are comparable with humans throughout their GALT and support future studies that use the rabbit model to study human gut-associated disease or infectious agents that gain entry by the oral route.
Collapse
Affiliation(s)
- Robyn A Haines
- Robyn A. Haines, DVM, was a PhD student; Rebeccah A. Urbiztondo, DVM, was a Master's student; Rashade A. H. Haynes, PhD, was a postdoctoral fellow; and Elaine Simpson, DVM, was a summer research student in the Department of Veterinary Biosciences at The Ohio State University in Columbus, Ohio. Stefan Niewiesk, PhD, DVM, is professor in the Department of Veterinary Biosciences at The Ohio State University is Columbus, Ohio. Michael D. Lairmore, DVM, PhD, is dean, in the School of Veterinary Medicine at the University of California, Davis in Davis, California
| | - Rebeccah A Urbiztondo
- Robyn A. Haines, DVM, was a PhD student; Rebeccah A. Urbiztondo, DVM, was a Master's student; Rashade A. H. Haynes, PhD, was a postdoctoral fellow; and Elaine Simpson, DVM, was a summer research student in the Department of Veterinary Biosciences at The Ohio State University in Columbus, Ohio. Stefan Niewiesk, PhD, DVM, is professor in the Department of Veterinary Biosciences at The Ohio State University is Columbus, Ohio. Michael D. Lairmore, DVM, PhD, is dean, in the School of Veterinary Medicine at the University of California, Davis in Davis, California
| | - Rashade A H Haynes
- Robyn A. Haines, DVM, was a PhD student; Rebeccah A. Urbiztondo, DVM, was a Master's student; Rashade A. H. Haynes, PhD, was a postdoctoral fellow; and Elaine Simpson, DVM, was a summer research student in the Department of Veterinary Biosciences at The Ohio State University in Columbus, Ohio. Stefan Niewiesk, PhD, DVM, is professor in the Department of Veterinary Biosciences at The Ohio State University is Columbus, Ohio. Michael D. Lairmore, DVM, PhD, is dean, in the School of Veterinary Medicine at the University of California, Davis in Davis, California
| | - Elaine Simpson
- Robyn A. Haines, DVM, was a PhD student; Rebeccah A. Urbiztondo, DVM, was a Master's student; Rashade A. H. Haynes, PhD, was a postdoctoral fellow; and Elaine Simpson, DVM, was a summer research student in the Department of Veterinary Biosciences at The Ohio State University in Columbus, Ohio. Stefan Niewiesk, PhD, DVM, is professor in the Department of Veterinary Biosciences at The Ohio State University is Columbus, Ohio. Michael D. Lairmore, DVM, PhD, is dean, in the School of Veterinary Medicine at the University of California, Davis in Davis, California
| | - Stefan Niewiesk
- Robyn A. Haines, DVM, was a PhD student; Rebeccah A. Urbiztondo, DVM, was a Master's student; Rashade A. H. Haynes, PhD, was a postdoctoral fellow; and Elaine Simpson, DVM, was a summer research student in the Department of Veterinary Biosciences at The Ohio State University in Columbus, Ohio. Stefan Niewiesk, PhD, DVM, is professor in the Department of Veterinary Biosciences at The Ohio State University is Columbus, Ohio. Michael D. Lairmore, DVM, PhD, is dean, in the School of Veterinary Medicine at the University of California, Davis in Davis, California
| | - Michael D Lairmore
- Robyn A. Haines, DVM, was a PhD student; Rebeccah A. Urbiztondo, DVM, was a Master's student; Rashade A. H. Haynes, PhD, was a postdoctoral fellow; and Elaine Simpson, DVM, was a summer research student in the Department of Veterinary Biosciences at The Ohio State University in Columbus, Ohio. Stefan Niewiesk, PhD, DVM, is professor in the Department of Veterinary Biosciences at The Ohio State University is Columbus, Ohio. Michael D. Lairmore, DVM, PhD, is dean, in the School of Veterinary Medicine at the University of California, Davis in Davis, California
| |
Collapse
|
5
|
Visualization of the entire differentiation process of murine M cells: suppression of their maturation in cecal patches. Mucosal Immunol 2015; 8:650-60. [PMID: 25336168 DOI: 10.1038/mi.2014.99] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 09/21/2014] [Indexed: 02/04/2023]
Abstract
The microfold (M) cell residing in the follicle-associated epithelium is a specialized epithelial cell that initiates mucosal immune responses by sampling luminal antigens. The differentiation process of M cells remains unclear due to limitations of analytical methods. Here we found that M cells were classified into two functionally different subtypes based on the expression of Glycoprotein 2 (GP2) by newly developed image cytometric analysis. GP2-high M cells actively took up luminal microbeads, whereas GP2-negative or low cells scarcely ingested them, even though both subsets equally expressed the other M-cell signature genes, suggesting that GP2-high M cells represent functionally mature M cells. Further, the GP2-high mature M cells were abundant in Peyer's patch but sparse in the cecal patch: this was most likely due to a decrease in the nuclear translocation of RelB, a downstream transcription factor for the receptor activator of nuclear factor-κB signaling. Given that murine cecum contains a protrusion of beneficial commensals, the restriction of M-cell activity might contribute to preventing the onset of any excessive immune response to the commensals through decelerating the M-cell-dependent uptake of microorganisms.
Collapse
|
6
|
|
7
|
Araújo F, Sarmento B. Towards the characterization of an in vitro triple co-culture intestine cell model for permeability studies. Int J Pharm 2013; 458:128-34. [DOI: 10.1016/j.ijpharm.2013.10.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 09/30/2013] [Accepted: 10/02/2013] [Indexed: 01/25/2023]
|
8
|
Casteleyn C, Van den Broeck W, Gebert A, Tambuyzer BR, Van Cruchten S, Van Ginneken C. M cell specific markers in man and domestic animals: Valuable tools in vaccine development. Comp Immunol Microbiol Infect Dis 2013; 36:353-64. [DOI: 10.1016/j.cimid.2013.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 03/01/2013] [Accepted: 03/21/2013] [Indexed: 12/13/2022]
|
9
|
Tahoun A, Mahajan S, Paxton E, Malterer G, Donaldson DS, Wang D, Tan A, Gillespie TL, O'Shea M, Roe AJ, Shaw DJ, Gally DL, Lengeling A, Mabbott NA, Haas J, Mahajan A. Salmonella transforms follicle-associated epithelial cells into M cells to promote intestinal invasion. Cell Host Microbe 2013; 12:645-56. [PMID: 23159054 DOI: 10.1016/j.chom.2012.10.009] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 07/25/2012] [Accepted: 10/02/2012] [Indexed: 12/16/2022]
Abstract
Salmonella Typhimurium specifically targets antigen-sampling microfold (M) cells to translocate across the gut epithelium. Although M cells represent a small proportion of the specialized follicular-associated epithelium (FAE) overlying mucosa-associated lymphoid tissues, their density increases during Salmonella infection, but the underlying molecular mechanism remains unclear. Using in vitro and in vivo infection models, we demonstrate that the S. Typhimurium type III effector protein SopB induces an epithelial-mesenchymal transition (EMT) of FAE enterocytes into M cells. This cellular transdifferentiation is a result of SopB-dependent activation of Wnt/β-catenin signaling leading to induction of both receptor activator of NF-κB ligand (RANKL) and its receptor RANK. The autocrine activation of RelB-expressing FAE enterocytes by RANKL/RANK induces the EMT-regulating transcription factor Slug that marks epithelial transdifferentiation into M cells. Thus, via the activity of a single secreted effector, S. Typhimurium transforms primed epithelial cells into M cells to promote host colonization and invasion.
Collapse
Affiliation(s)
- Amin Tahoun
- The Roslin Institute and Royal School of Veterinary Studies, University of Edinburgh, Edinburgh EH25 9RG, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
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: 1.2] [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
|
11
|
Goto Y, Kiyono H. Epithelial barrier: an interface for the cross-communication between gut flora and immune system. Immunol Rev 2012; 245:147-63. [PMID: 22168418 DOI: 10.1111/j.1600-065x.2011.01078.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Large numbers of environmental antigens, including commensal bacteria and food-derived antigens, constitutively interact with the epithelial layer of the gastrointestinal (GI) tract. Commensal bacteria peacefully cohabit with the host GI tract and exert multiple beneficial or destructive effects on their host. Intestinal epithelial cells (IECs) constitute the first physical and immunological protective wall against invasive pathogens and a cohabitation niche for commensal bacteria. As the physiological homeostasis of IECs is maintained by multiple biological processes such as apoptosis, autophagy, and the handling of endoplasmic reticulum stress, the aberrant kinetics of these biological events, which have genetic and environmental causes, leads to the development of host intestinal pathogenesis such as inflammatory bowel disease. In addition, IECs recognize and interact with commensal bacteria and give instructions to mucosal immune cells to initiate an immunological balance between active and quiescent conditions, eventually establishing intestinal homeostasis. The mucosal immune system regulates the homeostasis of gut microbiota by producing immunological molecules such as secretory immunoglobulin A, the production of which is mediated by IECs. IECs therefore play a central role in the creation and maintenance of a physiologically and immunologically stable intestinal environment.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
12
|
Bjerknes M, Khandanpour C, Möröy T, Fujiyama T, Hoshino M, Klisch TJ, Ding Q, Gan L, Wang J, Martín MG, Cheng H. Origin of the brush cell lineage in the mouse intestinal epithelium. Dev Biol 2011; 362:194-218. [PMID: 22185794 DOI: 10.1016/j.ydbio.2011.12.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 11/16/2011] [Accepted: 12/02/2011] [Indexed: 12/25/2022]
Abstract
Mix progenitors are short-lived multipotential cells formed as intestinal epithelial stem cells initiate a differentiation program. Clone dynamics indicates that various epithelial cell lineages arise from Mix via a sequence of progressively restricted progenitor states. Lateral inhibitory Notch signaling between the daughters of Mix (DOM) is thought to break their initial symmetry, thereby determining whether a DOM invokes a columnar (absorptive) or granulocytic (secretory) cell lineage program. This is supported by the absence of granulocytes following enforced Notch signaling or Atoh1 deletion. Conversely, granulocytes increase in frequency following inhibition of Notch signaling or Hes1 deletion. Thus reciprocal repression between Hes1 and Atoh1 is thought to implement a Notch signaling-driven cell-fate-determining binary switch in DOM. The brush (tuft) cells, a poorly understood chemosensory cell type, are not incorporated into this model. We report that brush cell numbers increase dramatically following conditional Atoh1-deletion, demonstrating that brush cell production, determination, differentiation and survival are Atoh1-independent. We also report that brush cells are derived from Gfi1b-expressing progenitors. These and related results suggest a model in which initially equivalent DOM progenitors have three metastable states defined by the transcription factors Hes1, Atoh1, and Gfi1b. Lateral inhibitory Notch signaling normally ensures that Hes1 dominates in one of the two DOMs, invoking a columnar lineage program, while either Atoh1 or Gfi1b dominates in the other DOM, invoking a granulocytic or brush cell lineage program, respectively, and thus implementing a cell fate-determining ternary switch.
Collapse
Affiliation(s)
- Matthew Bjerknes
- Department of Medicine, Clinical Science Division, University of Toronto, Toronto, Ontario, Canada M5S 1A8.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hondo T, Kanaya T, Takakura I, Watanabe H, Takahashi Y, Nagasawa Y, Terada S, Ohwada S, Watanabe K, Kitazawa H, Rose MT, Yamaguchi T, Aso H. Cytokeratin 18 is a specific marker of bovine intestinal M cell. Am J Physiol Gastrointest Liver Physiol 2011; 300:G442-53. [PMID: 21193527 DOI: 10.1152/ajpgi.00345.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Microfold (M) cells in the follicle-associated epithelium (FAE) of Peyer's patches have an important role in mucosal immune responses. A primary difficulty for investigations of bovine M cells is the lack of a specific molecular marker. To identify such a marker, we investigated the expression of several kinds of intermediate filament proteins using calf Peyer's patches. The expression patterns of cytokeratin (CK) 18 in jejunal and ileal FAE were very similar to the localization pattern of M cells recognized by scanning electron microscopy. Mirror sections revealed that jejunal CK18-positive cells had irregular and sparse microvilli, as well as pocket-like structures containing lymphocytes, typical morphological characteristic of M cells. However, CK18-negative cells had regular and dense microvilli on their surface, typical of the morphology of enterocytes. In contrast, CK20 immunoreactivity was detected in almost all villous epithelial cells and CK18-negative cells in the FAE. CK18-positive proliferating transit-amplifying cells in the crypt exchanged CK18 for CK20 above the mouth of the crypt and after moving to the villi; however, CK18-positive M cells in the crypt continued their expression of CK18 during movement to the FAE region. Terminal deoxynucleotidyl-transferase-mediated deoxyuridine-triphosphate-biotin nick-end labeling-positive apoptotic cells were specifically detected at the apical region of villi and FAE in the jejunum and ileum, and all were also stained for CK20. These data indicate that CK18 may be a molecular marker for bovine M cells in FAE and that M cells may transdifferentiate to CK20-positive enterocytes and die by apoptosis in the apex of the FAE.
Collapse
Affiliation(s)
- Tetsuya Hondo
- Cellular Biology Laboratory, Graduate School of Agricultural Science, Tohoku Univ., Sendai, Miyag, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Beyaz F, Ergün E, Bayraktaroğlu AG, Ergün L. Identification of intestinal M cells in isolated lymphoid follicles and Peyer’s patches of the Angora rabbit. Cell Tissue Res 2010; 341:417-27. [DOI: 10.1007/s00441-010-1005-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 06/08/2010] [Indexed: 02/08/2023]
|
15
|
The identification of intestinal M cells in the sacculus rotundus and appendix of the Angora rabbit. Vet Res Commun 2010; 34:255-65. [PMID: 20217227 DOI: 10.1007/s11259-010-9349-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2010] [Indexed: 12/18/2022]
Abstract
The present study was aimed at the immunohistochemical demonstration of M cells, found in the follicle-associated epithelium (FAE) of the sacculus rotundus (SR) and appendix of the Angora rabbit, using anti-vimentin primary antibodies, and at the determination of certain fine structural characteristics. Ten adult Angora rabbits constituted the material of the study. Immunohistochemical staining revealed that many cells composing the FAE, which covered the dome regions of the SR and appendix, reacted positively with vimentin. FAE contained two different types of vimentin-positive cells. The first type surrounded intraepithelial lymphocytes (IEL) with a basolateral invagination in the apex and periphery of the dome epithelium, whilst the second type consisted of columnar cells found in the FAE near crypts. The immunoreactivity of the cells found in the FAE covering the apex and periphery of the domes was observed particularly in the perinuclear cytoplasm and the cytoplasm surrounding the IEL. Electron microscopic examination demonstrated that the M cells found in the FAE covering the apex and periphery of the dome regions of the SR and appendix did not exhibit any microvilli on their apical surface. The FAE near crypts contained columnar cells, which resembled enterocytes. The apical membrane of these cells exhibited shorter and irregular microvilli, in contrast to neighbouring enterocytes. It was determined that M cells, found in the FAE of the SR and appendix in the Angora rabbit, displayed similarities in terms of localization and fine structure. This situation may be indicative of the two lymphoid structures with different localization having similar functional properties.
Collapse
|
16
|
Lelouard H, Henri S, De Bovis B, Mugnier B, Chollat-Namy A, Malissen B, Méresse S, Gorvel JP. Pathogenic bacteria and dead cells are internalized by a unique subset of Peyer's patch dendritic cells that express lysozyme. Gastroenterology 2010; 138:173-84.e1-3. [PMID: 19800337 DOI: 10.1053/j.gastro.2009.09.051] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 09/07/2009] [Accepted: 09/22/2009] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS Lysozyme has an important role in preventing bacterial infection. In the gastrointestinal tract, lysozyme is thought to be mainly expressed by Paneth cells of the crypt epithelium. We investigated its expression in the Peyer's patch, a major intestinal site of antigen sampling and pathogen entry. METHODS We performed immunostaining on normal and Salmonella Typhimurium-infected intestinal samples and analyzed them by confocal microscopy and flow cytometry. RESULTS In Peyer's patch of mouse, rat, and human, lysozyme was strongly expressed in the germinal center of follicles by tingible body macrophages and in the subepithelial dome by a subset of myeloid dendritic cells (DC). Among DC subsets from mouse Peyer's patches, these lysozyme-expressing DC displayed the highest surface expression of class II major histocompatibility complex and costimulatory molecules; they were the most efficient at capturing microspheres in vitro. Moreover, they were the main DC subset involved in bacterial pathogen uptake and in dead cell clearance, including M cells. CONCLUSIONS The subepithelial dome of Peyer's patches contains a unique population of intestinal DC that secretes high levels of lysozyme and internalizes bacteria and dead cells.
Collapse
Affiliation(s)
- Hugues Lelouard
- Aix Marseille Université, Faculté des Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Knoop KA, Kumar N, Butler BR, Sakthivel SK, Taylor RT, Nochi T, Akiba H, Yagita H, Kiyono H, Williams IR. RANKL is necessary and sufficient to initiate development of antigen-sampling M cells in the intestinal epithelium. THE JOURNAL OF IMMUNOLOGY 2009; 183:5738-47. [PMID: 19828638 DOI: 10.4049/jimmunol.0901563] [Citation(s) in RCA: 233] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microfold cells (M cells) are specialized epithelial cells situated over Peyer's patches (PP) and other organized mucosal lymphoid tissues that transport commensal bacteria and other particulate Ags into intraepithelial pockets accessed by APCs. The TNF superfamily member receptor activator of NF-kappaB ligand (RANKL) is selectively expressed by subepithelial stromal cells in PP domes. We found that RANKL null mice have <2% of wild-type levels of PP M cells and markedly diminished uptake of 200 nm diameter fluorescent beads. Ab-mediated neutralization of RANKL in adult wild-type mice also eliminated most PP M cells. The M cell deficit in RANKL null mice was corrected by systemic administration of exogenous RANKL. Treatment with RANKL also induced the differentiation of villous M cells on all small intestinal villi with the capacity for avid uptake of Salmonella and Yersinia organisms and fluorescent beads. The RANK receptor for RANKL is expressed by epithelial cells throughout the small intestine. We conclude that availability of RANKL is the critical factor controlling the differentiation of M cells from RANK-expressing intestinal epithelial precursor cells.
Collapse
Affiliation(s)
- Kathryn A Knoop
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
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: 1.2] [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
|
19
|
Kanaya T, Miyazawa K, Takakura I, Itani W, Watanabe K, Ohwada S, Kitazawa H, Rose MT, McConochie HR, Okano H, Yamaguchi T, Aso H. Differentiation of a murine intestinal epithelial cell line (MIE) toward the M cell lineage. Am J Physiol Gastrointest Liver Physiol 2008; 295:G273-84. [PMID: 18556421 DOI: 10.1152/ajpgi.00378.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
M cells are a kind of intestinal epithelial cell in the follicle-associated epithelium of Peyer's patches. These cells can transport antigens and microorganisms into underlying lymphoid tissues. Despite the important role of M cells in mucosal immune responses, the origin and mechanisms of differentiation as well as cell death of M cells remain unclear. To clarify the mechanism of M cell differentiation, we established a novel murine intestinal epithelial cell line (MIE) from the C57BL/6 mouse. MIE cells grow rapidly and have a cobblestone morphology, which is a typical feature of intestinal epithelial cells. Additionally, they express cytokeratin, villin, cell-cell junctional proteins, and alkaline phosphatase activity and can form microvilli. Their expression of Musashi-1 antigen indicates that they may be close to intestinal stem cells or transit-amplifying cells. MIE cells are able to differentiate into the M cell lineage following coculture with intestinal lymphocytes, but not with Peyer's patch lymphocytes (PPL). However, PPL costimulated with anti-CD3/CD28 MAbs caused MIE cells to display typical features of M cells, such as transcytosis activity, the disorganization of microvilli, and the expression of M cell markers. This transcytosis activity of MIE cells was not induced by T cells isolated from PPL costimulated with the same MAbs and was reduced by the depletion of the T cell population from PPL. A mixture of T cells treated with MAbs and B cells both from PPL led MIE cells to differentiate into M cells. We report here that MIE cells have the potential ability to differentiate into M cells and that this differentiation required activated T cells and B cells.
Collapse
Affiliation(s)
- Takashi Kanaya
- Cellular Biology Laboratory, Graduate School of Agricultural Science, Tohoku Univ., 1-1 Tsutsumidori Amamiyamachi, Aoba-ku, 981-8555 Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Corr S, Hill C, Gahan CGM. An in vitro cell-culture model demonstrates internalin- and hemolysin-independent translocation of Listeria monocytogenes across M cells. Microb Pathog 2006; 41:241-50. [PMID: 17049432 DOI: 10.1016/j.micpath.2006.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 08/28/2006] [Accepted: 08/28/2006] [Indexed: 01/02/2023]
Abstract
An ability to translocate the mucosal epithelia through M cells provides invasive pathogens with a rapid means of accessing the mucosal lymphoid tissues. In order to determine the role of M cells in Listeria monocytogenes infection, we initially assessed colonization of Peyer's patch (PP) epithelium in BALB/c mice by Vibrio cholerae Eltor, wild-type L. monocytogenes and an isogenic hemolysin mutant (LO28Deltahly). It was observed that both wild-type L. monocytogenes and Deltahly showed preferential colonization of PP epithelium in this model. Furthermore, a novel luciferase reporter system was used to show rapid site-specific localization of L. monocytogenes in intestinal Peyer's patches. To examine the role of M cells in transcytosis of L. monocytogenes we utilized an in vitro transwell model that mimics M-cell activity through differentiation of C2Bbe1 epithelial enterocytes via co-culture with murine Peyer's patch lymphocytes (PPL). It was shown that L. monocytogenes transits M cells at significantly increased rates compared to C2Bbe1 monocultures. In addition, M-cell transport occurred independently of bacterial hemolysin and internalin production. This study demonstrates rapid transcytosis of L. monocytogenes through M cells, a process that occurs independently of the action of classical virulence factors.
Collapse
Affiliation(s)
- Sinéad Corr
- Department of Microbiology and Alimentary Pharmabiotic Centre, University College Cork, Ireland
| | | | | |
Collapse
|
21
|
Blanco LP, DiRita VJ. Bacterial-associated cholera toxin and GM1 binding are required for transcytosis of classical biotype Vibrio cholerae through an in vitro M cell model system. Cell Microbiol 2006; 8:982-98. [PMID: 16681839 DOI: 10.1111/j.1462-5822.2005.00681.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To elucidate mechanisms involved in M cell uptake and transcytosis of Vibrio cholerae, we used an in vitro model of human M-like cells in a Caco-2 monolayer. Interspersed among the epithelial monolayer of Caco-2 cells we detect cells that display M-like features with or without prior lymphocyte treatment and we have established key parameters for V. cholerae transcytosis in this model. Cholera toxin (CT) mutants lacking the A subunit alone or both the A and B subunits were deficient for transcytosis. We explored this finding further and showed that expression of both subunits is required for binding by whole V. cholerae to immobilized CT receptor, the glycosphingolipid GM1. Confocal microscopy showed CT associated with transcytosing bacteria, and transcytosis was inhibited by pre-incubation with GM1 before infection. Finally, heat treatment of the bacterial cells caused a loss of binding to GM1 that was correlated with a significant decrease in uptake and transcytosis by the monolayer. Our data support a model in which the ability of bacteria to interact with GM1 in a CT-dependent fashion plays a critical role in transcytosis of V. cholerae by M cells.
Collapse
Affiliation(s)
- Luz P Blanco
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | | |
Collapse
|
22
|
De Coppi P, Pozzobon M, Piccoli M, Gazzola MV, Boldrin L, Slanzi E, Destro R, Zanesco L, Zanon GF, Gamba P. Isolation of mesenchymal stem cells from human vermiform appendix. J Surg Res 2006; 135:85-91. [PMID: 16650433 DOI: 10.1016/j.jss.2006.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 02/21/2006] [Accepted: 03/07/2006] [Indexed: 01/26/2023]
Abstract
BACKGROUND Recent findings have shown that pluripotent stem cells exist in areas outside the bone marrow (BM). Moreover, it has been demonstrated that the appendix is important for the development of mucosal gut immunity, and hematopoietic progenitors have been isolated from animal and human appendices. MATERIALS AND METHODS Non-inflamed appendices removed during laparotomy were processed and cultured until the appearance of adherent cells. Differentiations (performed under osteogenic, adipogenic, and myogenic conditions) were confirmed by immunohistochemistry and cytochemistry. Polymerase chain reaction and cytofluorimetric analyses were performed to evidence the presence of genes and protein specific lineages in appendix-derived mesenchymal stem cells (ADMCs). RESULTS ADMCs were present in non-inflamed appendices. ADMCs under osteogenic conditions differentiated in osteoblasts and showed increased alkaline phosphatase expression; at the gene level, we observed the expression of Core binding factor alpha 1 (Cbfa1) and osteocalcin in osteogenic induced ADMCs. Under adipogenic conditions, lipidic drops in the cytoplasm, expression of lipoprotein lipase (LpL), and peroxisome proliferator-activated receptor gamma were observed; under myogenic conditions, myotubes expressing muscle specific proteins like desmin were formed. Myogenic regulatory factor 4 and MyoD were selectively induced in the ADMCs under myogenic conditions. CONCLUSIONS This study shows for the first time that mesenchymal stem cells can be isolated from normal appendices obtained from a pediatric and adult age group (0-18 years of age). This finding not only may further knowledge of the maturation of the intestinal immunesystem but also could indicate a new physiological role of the human vermiform appendix.
Collapse
Affiliation(s)
- Paolo De Coppi
- Laboratorio Trapianto del Midollo Osseo and Banca del Sangue di Cordone Ombelicale, Dipartimento di Oncoematologia Pediatrica, Padova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Shakweh M, Ponchel G, Fattal E. Particle uptake by Peyer's patches: a pathway for drug and vaccine delivery. Expert Opin Drug Deliv 2005; 1:141-63. [PMID: 16296726 DOI: 10.1517/17425247.1.1.141] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Particle uptake by Peyer's patches offers the possibility of tailoring vaccines that can be delivered orally. However, particle uptake by the follicle-associated epithelium in the gastrointestinal tract depends on several different factors that are the physicochemical properties of the particles, the physiopathological state of the animal, the analytical method used to evaluate the uptake and finally the experimental model. These parameters do not allow a clear idea about the optimal conditions to target the Peyer's patches. The goal of this review is to clarify the role of each factor in this uptake.
Collapse
Affiliation(s)
- Monjed Shakweh
- University of Paris-South, Faculty of Pharmacy, UMR CNRS 8612, 5 rue Jean-Batiste Clement, 92290 Chatenay-Malabry Cedex, France
| | | | | |
Collapse
|
24
|
Miyazawa K, Aso H, Kanaya T, Kido T, Minashima T, Watanabe K, Ohwada S, Kitazawa H, Rose MT, Tahara K, Yamasaki T, Yamaguchi T. Apoptotic process of porcine intestinal M cells. Cell Tissue Res 2005; 323:425-32. [PMID: 16283391 DOI: 10.1007/s00441-005-0086-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2005] [Accepted: 09/12/2005] [Indexed: 12/20/2022]
Abstract
Membranous (M) cells of the follicle-associated epithelium (FAE) are believed to sample antigens from the gut lumen. However, the origin, differentiation mechanism, and cell death of M cells are still a matter of controversy. Therefore, we investigated the process of M cell differentiation and determined their fate in the intestine of three-way crossbred female pigs. We used anti-cytokeratin 18 and anti-PCNA antibodies to distinguish M cells and proliferative cells and performed immunohistochemistry, enzyme histochemistry, and scanning electron microscopy on fresh ileal Peyer's patches. Cell migration and apoptotic cells were detected by BrdU labeling and the TUNEL method, respectively. The turnover of the FAE was similar to that of the villi. M cells were mostly observed from the FAE crypt to the FAE periphery, but not in the FAE apex. As proliferative M cells (cytokeratin 18(+)/PCNA(+) cells) have previously been detected in the FAE crypt, porcine M cells may be directly derived from intestinal epithelial stem cells and committed as a distinct cell lineage in the crypts. M cells from the FAE periphery were unstained or only weakly stained for alkaline phosphatase, whereas cytokeratin 18(+)/alkaline phosphatase(+) cells lying near to the FAE apex showed a columnar shape similar to that of adjacent enterocytes. These data suggest that the committed M cells differentiate to mature M cells by contact with lymphocytes at the FAE periphery, and that they trans-differentiate to enterocytes and are finally excluded near the FAE apex.
Collapse
Affiliation(s)
- Kohtaro Miyazawa
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, 981-8555, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mahajan A, Naylor S, Mills AD, Low JC, Mackellar A, Hoey DEE, Currie CG, Gally DL, Huntley J, Smith DGE. Phenotypic and functional characterisation of follicle-associated epithelium of rectal lymphoid tissue. Cell Tissue Res 2005; 321:365-74. [PMID: 15965658 DOI: 10.1007/s00441-005-1080-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2004] [Accepted: 01/10/2005] [Indexed: 11/25/2022]
Abstract
Lymphoid follicles cluster in the terminal rectum of various animal species and of man and hence this site may be important in the development of immune responses to pathogens. For the induction of immune responses at mucosal sites, interplay is required between various cell types performing functions ranging from antigen-sampling cells via antigen-presenting cells to antigen-specific lymphocytes. Therefore, we have characterised the cell populations and relevant functioning of follicle-associated epithelium (FAE) and associated follicles in the terminal portion of rectum in cattle as a representative mammal. Immunohistochemical studies of this region identified immune cell subsets (CD4+, CD8+, WC 1+gammadelta, CD2+, CD 21+ and CD 40+ cells) characteristic of an immune-inductive site. Examination of FAE identified a subset of cells with structural and functional features of antigen-sampling M-cells. Cells of the FAE and adjacent follicle-associated crypts expressed vimentin and a subset of these cells internalised microparticles, a further attribute of M-cells. The FAE cells were phenotypically heterogeneous and therefore the function and phenotype of these cell subsets requires further characterisation, particularly with respect to their potentially important role in the interaction of hosts with pathogens and the development of immune responses.
Collapse
Affiliation(s)
- A Mahajan
- Zoonotic & Animal Pathogens Research Laboratory, Centre for Infectious Diseases, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Marchetti M, Sirard JC, Sansonetti P, Pringault E, Kernéis S. Interaction of pathogenic bacteria with rabbit appendix M cells: bacterial motility is a key feature in vivo. Microbes Infect 2005; 6:521-8. [PMID: 15158184 DOI: 10.1016/j.micinf.2004.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2003] [Accepted: 02/05/2004] [Indexed: 01/20/2023]
Abstract
Rabbit appendix consists mainly of lymphoid follicles (LF) covered by M cells, the specialized antigen-sampling cells of the mucosal immune system, and surrounded by glandular epithelium. Until now, these M cells have been characterized morphologically and histologically by using cellular markers. Here, the adhesion and transport of pathogenic bacteria were investigated to assess the function of M cells of the appendix. We used the enteroinvasive motile Salmonella typhimurium and the rabbit enteropathogenic non-motile Escherichia coli RDEC-1, which are known to target specifically rabbit M cells of Peyer's patches (PPs). We found that S. typhimurium efficiently attached and was transported through appendix M cells in vivo. In contrast to S. typhimurium, RDEC-1 targeted M cells only ex vivo, when bacteria were allowed to have direct contact with the surface of the follicle. The difference in interaction of the two bacteria with appendix M cells led us to investigate whether this could be correlated with the lack of motility of RDEC-1. We used an aflagellate mutant of S. typhimurium and found that it had the same infection phenotype as RDEC-1. Gene complementation restored the efficiency of infection to that of S. typhimurium wild-type strain. In conclusion, we show that M cells of the appendix display features of the canonical M cells of PP, since they efficiently sample luminal pathogenic bacteria. However, due to the morphology of the appendix, motile bacteria appear to be more potent in their interactions with appendix M cells.
Collapse
Affiliation(s)
- Marta Marchetti
- Laboratory of Lympho-epithelial Interactions, Department of Cell Biology and Infection, Pasteur Institute, 25-28, rue du Docteur Roux, 75724 Paris cedex 15, France.
| | | | | | | | | |
Collapse
|
27
|
|
28
|
Man AL, Prieto-Garcia ME, Nicoletti C. Improving M cell mediated transport across mucosal barriers: do certain bacteria hold the keys? Immunology 2004; 113:15-22. [PMID: 15312131 PMCID: PMC1782554 DOI: 10.1111/j.1365-2567.2004.01964.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Specialized microfold (M) cells of the follicle-associated epithelium (FAE) of the mucosal-associated lymphoid tissue (MALT) in gut and the respiratory system play an important role in the genesis of both mucosal and systemic immune responses by delivering antigenic substrate to the underlying lymphoid tissue where immune responses start. Although it has been shown that dendritic cells (DC) also have the ability to sample antigens directly from the gut lumen, M cells certainly remain the most important antigen-sampling cell to be investigated in order to devise novel methods to improve mucosal delivery of biologically active compounds. Recently, novel information on the interactions between bacteria and FAE have come to light that unveil further the complex cross-talk taking place at mucosal interfaces between bacteria, epithelial cells and the immune system and which are central to the formation and function of M cells. In particular, it has been shown that M cell mediated transport of antigen across the FAE is improved rapidly by exposure to certain bacteria, thus opening the way to identify new means to achieve a more effective mucosal delivery. Here, these novel findings and their potential in mucosal immunity are analysed and discussed, and new approaches to improve antigen delivery to the mucosal immune system are also proposed.
Collapse
Affiliation(s)
- Angela L Man
- Laboratory of Gut Immunology, Programme of Gastrointestinal Health and Function, Institute of Food Research, Norwich, UK
| | | | | |
Collapse
|
29
|
Jepson MA, Clark MA, Hirst BH. M cell targeting by lectins: a strategy for mucosal vaccination and drug delivery. Adv Drug Deliv Rev 2004; 56:511-25. [PMID: 14969756 DOI: 10.1016/j.addr.2003.10.018] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2003] [Accepted: 10/14/2003] [Indexed: 12/11/2022]
Abstract
Bioadhesins are a recognised method of enhancing the absorption of drugs and vaccines at mucosal surfaces. Additionally, bioadhesins allow for cell specific targeting. Lectin-mediated targeting and delivery exploits unique surface carbohydrates on mucosal epithelial cells. The antigen-sampling M cells offer a portal for absorption of colloidal and particulate delivery vehicles, including bacteria, viruses and inert microparticles. We review work supporting the use of lectins to aid targeting to intestinal M cells. Consideration is also given to lectin-mediated targeting in non-intestinal sites and to the potential application of other bioadhesins to enhance M cell transport. While substantial hurdles must be overcome before mucosal bioadhesins can guarantee consistent, safe, effective mucosal delivery, this strategy offers novel opportunities for drug and vaccine formulation.
Collapse
Affiliation(s)
- Mark A Jepson
- Cell Imaging Facility and Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| | | | | |
Collapse
|
30
|
Gebert A, Steinmetz I, Fassbender S, Wendlandt KH. Antigen transport into Peyer's patches: increased uptake by constant numbers of M cells. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:65-72. [PMID: 14695320 PMCID: PMC1602236 DOI: 10.1016/s0002-9440(10)63097-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Membranous (M) cells are specialized epithelial cells of the Peyer's patches that sample antigens from the gut lumen, thereby enabling the host to respond immunologically. Recent studies suggest that this transport can be up-regulated within hours by de novo formation of M cells from enterocytes. To test this hypothesis, we used an in vivo model and induced the transcytosis of tracers in Peyer's patches by application of Streptococcus pneumoniae R36a into the gut lumen. Using cell-type-specific markers, we quantified M cells in the Peyer's patch domes, lymphocytes associated with M cells, and the transport rate for experimentally applied microbeads after 3 hours of exposure to R36a. The transport of latex microbeads was significantly increased by +131% in the R36a-treated patches as compared to buffer controls (P < 0.001). While in controls, each M cell was associated with 2.05 +/- 0.64 lymphocytes, a significant increase (+55.1%; P < 0.001) was determined in the R36a-treated patches. However, no statistical difference was detected in the percentage of M cells in the dome epithelia (46.0 +/- 4.6% versus 45.5 +/- 3.8%). It is concluded that bacteria-induced up-regulation of particle transport in Peyer's patch domes is due to an increased transport rate of the M cells, but not to a de novo formation of M cells. The data support the hypothesis that M cells represent a separate cell lineage that does not derive from enterocytes on the domes.
Collapse
Affiliation(s)
- Andreas Gebert
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.
| | | | | | | |
Collapse
|
31
|
Ramirez C, Gebert A. Vimentin-positive cells in the epithelium of rabbit ileal villi represent cup cells but not M-cells. J Histochem Cytochem 2003; 51:1533-44. [PMID: 14566025 PMCID: PMC3957562 DOI: 10.1177/002215540305101113] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Membranous (M)-cells are specialized epithelial cells of the Peyer's patch domes that transport antigens from the intestinal lumen to the lymphoid tissue. Vimentin is a reliable marker for M-cells in rabbits. Using immunohistochemistry (IHC), a subpopulation of epithelial cells has recently been identified in ordinary rabbit ileal villi, which are vimentin-positive and share morphological characteristics with the M-cells of the domes. To test the hypothesis that these cells represent M-cells outside the organized lymphoid tissue, lectin labeling and tracer uptake experiments were performed. Lectins specific for N-acetyl-glucosamine oligomers selectively bound to the vimentin-positive villous cells but not to M-cells in the domes. Microbeads instilled into the ileal lumen were taken up by M-cells within 45 min but not by the vimentin-positive cells in the villi. Lectin-gold labeling on ultrathin sections revealed that the lectin binding sites were located in the brush border and in vesicles in the apical cytoplasm. The vimentin/lectin-positive cells shared ultrastructural characteristics with the so-called "cup cells." We conclude (a) that the vimentin-positive cells in ordinary villi represent cup cells but not M-cells, (b) that they are readily detectable by (GlucNAc)(N)-specific lectins, and (c) that they do not transcytose experimental tracers. Although the specific function of cup cells is still obscure, they most probably represent a cell type distinct from M-cells of the domes with respect to both function and expression of the two new markers.
Collapse
Affiliation(s)
| | - Andreas Gebert
- Institute of Anatomy, University of Lübeck, Lübeck, Germany (CR,AG)
- Correspondence to: Prof. Dr. A. Gebert, Inst. of Anatomy, University of Lübeck, 23538 Lübeck, Germany. E-mail:
| |
Collapse
|
32
|
Lo D, Tynan W, Dickerson J, Mendy J, Chang HW, Scharf M, Byrne D, Brayden D, Higgins L, Evans C, O'Mahony DJ. Peptidoglycan recognition protein expression in mouse Peyer's Patch follicle associated epithelium suggests functional specialization. Cell Immunol 2003; 224:8-16. [PMID: 14572796 DOI: 10.1016/s0008-8749(03)00155-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mammalian Peyer's Patches possess specialized epithelium, the follicle associated epithelium (FAE), and specialized cells called M cells which mediate transcytosis of antigens to underlying lymphoid tissue. To identify FAE specific genes, we used TOGA gene expression profiling of microdissected mouse Peyer's Patch tissue. We found expression of laminin beta3 across the FAE, and scattered expression of peptidoglycan recognition protein (PGRP)-S. Using the M cell specific lectin Ulex europaeus agglutinin 1 (UEA-1), PGRP-S expression was nearly exclusively co-localized with UEA-1+ M cells. By contrast, the related gene PGRP-L was expressed among a subset of UEA-1 negative FAE cells. Expression of these proteins in transfected cells demonstrated distinct subcellular localization. PGRP-S showed a vesicular pattern and extracellular secretion, while PGRP-L showed localization to both the cytoplasm and the cell surface. The potential function of these PGRP proteins as pattern recognition receptors and their distinctive cellular distribution suggests a complex coordination among specialized cells of the FAE in triggering mucosal immunity and innate immune responses.
Collapse
Affiliation(s)
- David Lo
- Digital Gene Technologies, Inc., La Jolla, CA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
M cells are located in the epithelia overlying mucosa-associated lymphoid tissues such as Peyer's patches where they function as the antigen sampling cells of the mucosal immune system. Paradoxically, some pathogens exploit M cells as a route of invasion. Here we review our current knowledge of intestinal M cells with particular emphasis on the mechanisms underlying bacterial infection of these atypical epithelial cells.
Collapse
Affiliation(s)
- M Ann Clark
- Department of Physiological Sciences, University of Newcastle, Newcastle upon Tyne NE2 4HH, UK
| | | |
Collapse
|
34
|
El Bahi S, Caliot E, Bens M, Bogdanova A, Kernéis S, Kahn A, Vandewalle A, Pringault E. Lymphoepithelial interactions trigger specific regulation of gene expression in the M cell-containing follicle-associated epithelium of Peyer's patches. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3713-20. [PMID: 11937521 DOI: 10.4049/jimmunol.168.8.3713] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the intestine, the follicle-associated epithelium (FAE) of Peyer's patches (PP) performs Ag sampling as the first step in developing immune responses. Depending on the species, this epithelium contains 10-50% of M cells, which act as regulated gates in epithelial barriers that can be used opportunistically by pathogens to invade their host. However, the mechanisms involved in the differentiation and uptake processes of M cells are not known, in part because their limited number in the intestinal mucosa has hampered molecular and biochemical studies. In this work we provide evidence that PP lymphocytes can themselves modulate gene expression in PP in vivo and in an in vitro model of FAE. Transgenic mice carrying a reporter gene under the control of a modified L-pyruvate kinase promoter (SVPK) exhibit strong transgene expression in PP and FAE, but not in the adjacent villous cells. We used the mouse intestinal epithelial cell line m-IC(cl2) transfected with the SVPK promoter fused to beta-galactosidase to investigate the direct effect of PP lymphocytes on SVPK promoter activity. beta-Galactosidase expression was 4.4-fold higher in transfected m-IC(cl2) cells when they were cultured with PP lymphocytes. Conversely, green fluorescent protein expression was 1.8-fold lower in stably transfected differentiated intestinal Caco-2(cl1) cells with the sucrase isomaltase promoter fused to green fluorescent protein cDNA when they were cultured with PP lymphocytes, indicating that the in vivo FAE down-regulation of sucrase isomaltase promoter is transcriptionally regulated.
Collapse
Affiliation(s)
- Sophia El Bahi
- Laboratoire des Interactions Lympho-Epithéliales, Département de Biologie Cellulaire et Infection, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Neutra MR, Mantis NJ, Kraehenbuhl JP. Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nat Immunol 2001; 2:1004-9. [PMID: 11685223 DOI: 10.1038/ni1101-1004] [Citation(s) in RCA: 387] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Immune surveillance of mucosal surfaces requires the delivery of intact macromolecules and microorganisms across epithelial barriers to organized mucosal lymphoid tissues. Transport, processing and presentation of foreign antigens, as well as local induction and clonal expansion of antigen-specific effector lymphocytes, involves a close collaboration between organized lymphoid tissues and the specialized follicle-associated epithelium. M cells in the follicle-associated epithelium transport foreign macromolecules and microorganisms to antigen-presenting cells within and under the epithelial barrier. Determination of the earliest cellular interactions that occur in and under the follicle-associated epithelium could greatly facilitate the design of effective mucosal vaccines in the future.
Collapse
Affiliation(s)
- M R Neutra
- Children's Hospital and Harvard Medical School, Enders 1220, 300 Longwood Avenue, Boston, MA 02115, USA.
| | | | | |
Collapse
|