3,991
Views
56
CrossRef citations to date
0
Altmetric
Review

Roles of M cells in infection and mucosal vaccines

, , , &
Pages 3544-3551 | Received 03 Jul 2014, Accepted 21 Aug 2014, Published online: 10 Feb 2015

Abstract

The mucosal immune system plays a crucial part in the control of infection. Exposure of humans and animals to potential pathogens generally occurs through mucosal surfaces, thus, strategies that target the mucosa seem rational and efficient vaccination measures. Vaccination through the mucosal immune system can induce effective systemic immune responses simultaneously with mucosal immunity compared with parenteral vaccination. M cells are capable of transporting luminal antigens to the underlying lymphoid tissues and can be exploited by pathogens as an entry portal to invade the host. Therefore, targeting M-cell-specific molecules might enhance antigen entry, initiate the immune response, and induce protection against mucosal pathogens. Here, we outline our understanding of the distribution and function of M cells, and summarize the advances in mucosal vaccine strategies that target M cells.

Introduction

Despite continuous exposure to a broad range of biological stimuli, the mucosal surfaces provide a first line of defense against potential pathogens and nonself antigens, because most infections are initiated at mucosal sites. The mucosal immune system plays a vital role in maintaining mucosal homeostasis and consistently surveilling potential harmful agents. The mucosal immune system is essentially divided into 2 compartments known as inductive and effector sites, which work together to maintain the mucosal barrier.Citation1 The inductive sites mainly consist of mucosa-associated lymphoid tissue (MALT), including gut-associated lymphoid tissue (GALT), nasopharynx- or nose-associated lymphoid tissue (NALT), bronchus-associated lymphoid tissue (BALT), and genital-associated lymphoid tissue (GENALT), where antigen sampling occurs and stimulates cognate naive T and B lymphocytes. Although the effector sites are primarily constituted by the lamina propria of various mucosae, stroma of exocrine glands, and surface epithelia, at which antigen-specific antibodies and immune cells perform their specific action.Citation2

M cells (membranous epithelial, microfold or microvillous cells), first observed in 1965,Citation3 are functional units constituting inductive sites of the intestinal mucosal immune system. In the human gastrointestinal tract, M cells are predominantly present on the follicle-associated epithelium (FAE) which overlies the dome structure of Peyer's patches (PPs) in the small intestine,Citation4 and have been identified in the crypt epithelium of human tonsils and adenoidsCitation5 and colon and rectum of mice.Citation6 They are specialized epithelial cells of GALT, playing a crucial role in the intestinal immune system by sampling and uptake of antigens, and then transporting them from the gut lumen to the underlying immune system.Citation7 Gastrointestinal contents transported via M cells can be captured by dendritic cells (DCs) resident beneath M cells to initiate mucosal immune responses, leading to the production of antigen specific IgA by B cells.Citation8 M cells are among the most important epithelial cell types involved in the adhesion, uptake and sampling of antigens at mucosal surfaces, which facilitates M and adjacent cells working together to maintain mucosal homeostasis.

Distribution and function of M cells

Distribution of M cells

M cells exist principally in the FAE overlying both isolated (i.e. solitary nodules) and aggregated (i.e., PP) lymphoid folliclesCitation9 in GALT, and as well in NALTCitation10 and BALT.Citation11 They can be easily recognized and distinguished from neighboring enterocytes and goblet cells by their unique morphological characters, which include irregular brush border, high endocytic activity, reduced microvilli, thin glycocalyx, sparse lysosomes, and basolateral pockets containing mononuclear phagocytes and lymphocytes. They possess tight junctions and desmosomes that contact adjacent columnar cells and interdigitating latacoria (). These features are adaptions allowing materials (macromolecules and microorganisms) in the lumen to have easy access to the internalized apical surface of M cells and subsequently to be transported to the underlying tissues. However, the ultrastructure of M cells shows some variation between species and anatomical regions.Citation7 Therefore, identifying M cells needs morphological features at the light microscopic level and specific M cells markers to isolate or target them. Cellular morphology requires ultrastructural examination using scanning and transmission electron microscopy, which are labor intensive and not available to many researchers. Specific M cell markers are essential, whereas the expression of cellular antigens by M cells varies widely among species and anatomical sites.Citation7 Despite no universal M cell markers to date, many potential markers have been studied in humans and experimental animals in the past few years, using species- and tissue-specific M cell markers ().Citation12

Table 1. M cell markers in various species and MALT sites

Figure 1. Schematic diagram of intestinal epithelium showing M cells, PPs, intestinal epithelial cells and translocation mechanisms of several kinds of antigen. Gut-associated mucosal immune system has 2 parts, including inductive sites and effector sites. The inductive sites are mainly composed of MALT, and the effector sites are primarily constituted by the lamina propria of various mucosae, stroma of exocrine glands, and surface epithelia. In the FAE, luminal antigens are transported across the epithelium by M cells, where primary immune responses can be induced, and then phagocytosed by DCs present in the dome of the PPs, delivered to the mesenteric lymph nodes, which can directly prime T-cell responses to antigens. DCs also sample antigens through enterocytes. sIgA, secreted by mature plasma cells in the lamina propria, from epithelial cells into the gut lumen, may have a controlling role in bacterial persistence and uptake. M cells can transport bacteria, viruses, parasites and non-infectious particles through the apical membrane to the basolateral surface, exploiting many molecules and proteins to recognize and transport antigen. M cells lack microvilli and possess basolateral pockets containing mononuclear phagocytes and lymphocytes, which contribute antigens transported to the underlying tissues. PP, Peyer's patches; DC, dendritic cell; GP2, Glycoprotein 2; PrPC, cellular prion protein; C5aR, C5a receptor; FAE, follicle-associated epithelium; MALT, mucosa-associated lymphoid tissue.

Figure 1. Schematic diagram of intestinal epithelium showing M cells, PPs, intestinal epithelial cells and translocation mechanisms of several kinds of antigen. Gut-associated mucosal immune system has 2 parts, including inductive sites and effector sites. The inductive sites are mainly composed of MALT, and the effector sites are primarily constituted by the lamina propria of various mucosae, stroma of exocrine glands, and surface epithelia. In the FAE, luminal antigens are transported across the epithelium by M cells, where primary immune responses can be induced, and then phagocytosed by DCs present in the dome of the PPs, delivered to the mesenteric lymph nodes, which can directly prime T-cell responses to antigens. DCs also sample antigens through enterocytes. sIgA, secreted by mature plasma cells in the lamina propria, from epithelial cells into the gut lumen, may have a controlling role in bacterial persistence and uptake. M cells can transport bacteria, viruses, parasites and non-infectious particles through the apical membrane to the basolateral surface, exploiting many molecules and proteins to recognize and transport antigen. M cells lack microvilli and possess basolateral pockets containing mononuclear phagocytes and lymphocytes, which contribute antigens transported to the underlying tissues. PP, Peyer's patches; DC, dendritic cell; GP2, Glycoprotein 2; PrPC, cellular prion protein; C5aR, C5a receptor; FAE, follicle-associated epithelium; MALT, mucosa-associated lymphoid tissue.

Recently, the presence of intestinal villous M cells has challenged the notion of an exclusive location for M cells within the FAE region.Citation13 Intestinal villous M cells are located at the top of intestinal villous epithelium, forming small dense clusters or with diffuse distribution. They have a function similar to the PP M cells, and evidence from several studies shows that antigen-specific mucosal immune responses can be induced in GALT- and PP-deficient mice.Citation13,14

Function of M cells

M cells have the ability to transport bacteria,Citation15-18 viruses,Citation19-24 parasitesCitation25 and non-infectious particlesCitation26 through the apical membrane to the basolateral surface. When bacteria and large particles get into the mucosal lumen, M cells undergo apical membrane ruffling, actin cytoskeleton rearrangements,Citation27 and interdigitation, and then phagocytose these large particles. Viruses and small particles with adhesion properties undergo phagocytosis in clathrin-coated vesiclesCitation28 and nonadherent antigens undergo fluid-phase pinocytosis.Citation29 After transporting endocytotic vesicles to the endosomal compartment, internalization occurs quickly, and the vesicles are secreted by exocytosis to the basolateral membrane.Citation30

In contrast, M cells also provide an entry for many pathogens to invade the host and establish infection. Brucella abortus is internalized only in M cells among intestinal epithelial cells, demonstrating a role for M cells as an entry portal for B. abortus after oral infection. Researchers have also detected co-localization of cellular prion protein (PrPC) and the bacterium on the apical surface of M cells, indicating that PrPC on M cells serves as a major uptake receptor for B. abortus during oral infection.Citation31 As a result, the capacity of M cells for antigen sampling can accelerate invasion by potentially harmful enteric microorganisms.Citation32 An in vitro study in an M-like cell model that expressed caveolin-1 elucidated the role of caveolin-1 in the entry of Salmonella. Downregulation of caveolin-1 expression by siRNA reduced the level of Salmonella transcytosis across the M-like cells.Citation33 However, M cells deliver these pathogens directly to regions of the immune system fully equipped for coping with an emergency. The best evidence supporting this condition above is provided by studies carried on Shigella spp.,Citation18 Salmonella spp,Citation34 Yersinia spp.,Citation35 Vibrio cholera, reovirus type-1, poliovirus and HIV-1.36 It has been shown that M cells or antigen sampling by M cells can result in significant attenuation of the antigen-specific immune response in PPs in mice with oral administration of Salmonella enterica serovar Typhimurium.Citation37,38 In addition, M cells have the ability to distinguish among different commensal bacteria and alter subsequent immune responses.Citation39

It is particularly worth mentioning that M cells also have the capacity to transport proteins,Citation40 especially secretory IgA (SIgA). SIgA selectively interacts with M cells, targets DCs located in the sub-epithelial dome region of PP, resulting in limited mucosal and systemic immune responses against a non-self-associated protein antigen,Citation41 and exploits M cells as the primary route for delivery to the GALT. A recent study has demonstrated the mechanism by which SIgA is selectively bound and taken up. Both the Cɑ1 region and glycosylation, more particularly sialic acid residues, take part in M-cell-mediated reverse transcytosis, and M cells take up SIgA via the Dectin-1 receptor, in which Siglec-5 may act as a co-receptor.Citation42 There are several mechanisms of targeting M cells for induction of mucosal SIgA responses in animal models. For instance, it has been demonstrated that M cell targeting mediated by a Claudin-4-specific targeting peptide enhances mucosal IgA responses above the response to nontargeted mucosal antigen, which have promise in delivery of mucosal vaccination.Citation43

M cell-targeted mucosal immunization

For many years, researchers have been exploiting the potential of M-cell-specific mechanisms for drug and vaccine delivery to the mucosal immune system.Citation44 Many M-cell-targeted molecules have been used for development of mucosal vaccines ().

Table 2. M-cell-targeting molecules for mucosal vaccination

M-cell-specific molecules in mucosal vaccine development

M cells express a large amount of immune-surveillance receptors on the apical surfaces, contributing to the variety of microbial pathogens and antigens. They are provided with an array of molecules to present luminal antigens to the underlying mucosal lymphoid tissues. Therefore, identifying M-cell-specific targeting molecules has been a focus, by recognizing molecules exploited by pathogens to invade M cells.

Glycoprotein 2 (GP2)

GP2 is specially expressed on M cells.Citation45 This protein is highly expressed on the apical membranes of PP M cells, but not highly expressed on other enterocyte populations. Recent studies have revealed that GP2 acts as a transcytotic receptor, bound to FimH+ bacteria such as Escherichia coli and S. Typhmurium, by recognizing FimH, a major component of the type 1 pilus on the outer membrane of a subset of Gram-negative enterobacilli.Citation38 Thus, GP2 on M cells can act as a transcytotic receptor for bacterial antigens, and worthy of note, participate in the mucosal immune responses to these particular bacteria; a subset of commensal and pathogenic enterobacteria (E. coli and S. Typhmurium).Citation46,47 Other research has shown that a murine GP2 (mGP2)-specific aptamer, isolated using Systematic Evolution of Ligands by EXponential enrichment (SELEX), with a loop structure and the nucleotide sequence, AAAUA (both important for binding to mGP2), binds to mGP2 expressed on the cell surface, indicating that the aptamer serves as a promising tool for testing M-cell-targeted vaccine delivery in murine model systems.Citation48

PrPC

PrPC is highly expressed on the luminal side of the apical plasma membrane of murine M cells and co-localized with GP2, suggesting that it is an antigen receptor candidate on M cells.Citation49 PrPC interacts with heat shock protein (Hsp)60 of B. abortus, which had been recognized as an immunodominant antigen of many microbes.Citation50 Accumulated evidence suggests that PrPC on M cells is well placed to contribute to mucosal immunosurveillance by enhancing transcytosis of B. abortus or other exogenous antigens.Citation49

C5a receptor (C5aR)

The expression and nonredundant role of C5aR in human M-like cells and mouse M cells have been demonstrated, indicating the role of C5aR as a target receptor to induce the immune response. Sae-Hae Kim et al. verified phosphorylation of C5aR in vivo after oral infection of mice by Yersinia enterocolitica. They confirmed the expression of C5aR in the apical area of mouse M cells and human M-like cells by measuring the expression levels of mRNA and protein.Citation51 Sae-Hae Kim et al. also used the outer membrane protein H (OmpH) ligand of Y. enterocolitica, which acts as a targeting ligand to C5aR in M cells, to induce specific mucosal and systemic immunity against envelope domain III (EDIII) of dengue virus (DENV), suggesting OmpH-mediated targeting of antigens to M cells as an efficient oral vaccination against DENV infection.Citation52

Other specific molecules

There are other M-cell-specific molecules that may specifically bind to components of potential pathogenic organisms. Peptidoglycan recognition protein 1 is an innate recognition protein binding to bacterial peptidoglycan, and is also expressed highly in M cells.Citation53 Annexin (ANX) A5 expressed by M cellsCitation54 can bind to lipopolysaccharide (LPS) of Gram-negative bacteria and block endotoxin activity, suggesting that ANXA5 on M cells acts as an uptake receptor for Gram-negative bacteria.Citation55 The discovery of M-cell-targeting receptors using pathogen-exploited molecules could be a promising approach in the development of effective mucosal vaccines. Clusterin, fatty acid binding protein, cathepsin E, secretogranin V and other M-cell-expressed proteins may have potential roles in M cell functions, but these are less clearly understood.Citation56-59 The increasing evidences have demonstrated that M-cell-specific molecular antibody, which is conjugated with antigen protein or liver vector, can transport the antigen to mucosal tissues, leading to produce efficient immune responses. However, some molecules, selected as M-cell-specific molecules, are not uniquely expressed on M cells, resulting in producing a non-ideal oral delivery system for targeting M cells. With the development research on the mechanism of M cell differentiation, we can regulate the immune processes by means of artificial mediation of the M-cell-specific molecules gene expression. For instance, we can increase the efficiency of mucosal vaccination, through booting the expression of certain M-cell-specific molecules. Meanwhile, we can even inhibit the viral infection, by reducing the expression of some molecules, which are necessary for the entry of some virus particles.

M cell ligands as novel and effective mucosal vaccine targets

Many researchers have studied M cell ligands, in order to take advantage of the fact that targeting specific receptors on the apical membrane of M cells could specifically increase antigen uptake and presentation, evoking immune responses and providing protection against infection.Citation60

NKM 16-2-4

NKM 16–2–4 can be used as monoclonal antibody to target vaccine antigens to the M-cell-specific carbohydrate moiety.Citation61 It can distinguish α(1, 2)-fucosylated M cells from goblet cells containing abundant sialic acids neighboring the α(1, 2) fucose moiety and from non-α(1, 2)-fucosylated epithelial cells.Citation61 The use of NKM 16–2–4 targeting vaccine antigens to M cells could be effective for vaccine delivery into the intestinal mucosa. Oral vaccination using antigen-encapsulated liposomes coupled with UEA-1 could lead to increased uptake by PP M cells and the induction of higher antigen specific sIgA responses.Citation62

Co1 ligand

Many studies have investigated the M-cell-targeting ligand, Co1, selected from a phage display library against differentiated M-like cells, and have produced recombinant antigen fused to the selected ligands using the model antigen.Citation63-65 Co1 ligand promotes the uptake of fused antigen and enhances the immune response against the fused antigen, indicating that Co1 could be used as an adjuvant for targeted antigen delivery into the mucosal immune system to enhance immune induction.Citation64 Another promising approach used Co1 ligand to induce specific immune responses against a pathogenic viral antigen, EDIII of DENV. Efficient antigen delivery into PPs was observed and the antibodies induced by the Co1-ligand-conjugated EDIII antigen showed effective virus-neutralizing activity.Citation65 Taken together, these results reveal that M-cell-targeting ligands with adjuvant activity can be designed to exploit our knowledge of receptors expressed on the apical surface of M cells involved in pathogen invasion.Citation66,67

Caveolin-1

Caveolin-1 is the major structural component of caveolae. It was examined its expression in Caco-2-driven M-like cells, and was verified that co-culturing with B lymphocytes, caveolin-1 could increase the susceptibility of M cells to Salmonella infection.Citation68 Some recent studies have shown that caveolin-1 is not only a good marker of human M cells, but also a potent candidate for understanding M cell transcytosis as a novel target for mucosal immunity.

Ulex europaeus agglutinin (UEA)-1

UEA-1 has been confirmed as a specific ligand for α-l-fucose present on the apical membrane of M cells, anchored for selective delivery of antigen to GALT. Some researchers have used nanoparticles coated by UEA-1-conjugated alginate to induce immunological response in BALB/c mice, and compared them with aluminum hydroxide gel-based conventional vaccine. The results demonstrated that immunization with the former induced efficient systemic as well as mucosal immune responses against BSA compared to other formulations, which indicated the potential of UEA–alginate-coated nanoparticles as an effective oral delivery system.Citation69 However, UEA-1 lectin also reacted strongly with other issues, such as goblet cells and the mucus layer covering the intestinal epithelium.Citation70

Reovirus surface protein σ 1 (pσ1)

pσ1 has the ability to bind M cells, which facilitates reovirus infection via pσ1. A genetic fusion between ovalbumin (OVA) and pσ1 was applied nasally, to enhance tolerogen uptake. Studies showed that OVA– pσ1-mediated tolerance was lost in the absence of interleukin-10, demonstrating that the feasibility of using pσ1 as a mucosal delivery platform specifically for low-dose tolerance induction.Citation71 Another targeted transgene vaccination using pσ1 conjugated to polylysine through intranasal immunization, could induce mucosal immunity and enhance cell-mediated immunity, leading to prolong mucosal IgA and produce antigen-specific serum IgG.Citation72

The number of M cell receptors and their ligands that have been identified so far is limited, and most of them are not just expressed in M cells, but in neighboring enterocytes as well.Citation60 Toll-like receptor (TLR)-4 and α5β1 integrin, belonging to pathogen recognition receptors (PRRs), are expressed on the surface of human and mouse M cells. Interaction between these innate immune system molecules with pathogen-associated molecular patterns is essential for bacteria translocation across the lumen. Nevertheless, PRRs are also expressed in other enterocytes and not merely in M cells. For example, α5β1 integrin is both dispersed on the lateral and basolateral surfaces of enterocytes and on the apical surface of M cells, which is a challenge in targeting M cells alone.Citation60

M-cell-targeting ligands can enhance the uptake of oral vaccines by M cells and improve antigen-specific immune responses in both mucosal and systemic immunity. It seems that targeting ligand to antigen is a very promising approach in the development of efficient mucosal vaccine. However, simple targeting of antigen to M cells does not ensure the production of efficient protective immunity. We should pay more attention to the ligand study and find out the “optimal transporter," presenting antigens to M cells, leading to efficient immune responses.

Concluding Remarks

Compared with the parenteral vaccines, mucosal vaccines have many advantages, such as safety, low cost, convenient administration, being innocuous, and effectiveness due to induction of both local and systemic protective immune responses. Nevertheless, there are still some limits not to be ignored in mucosal vaccines. Mucosal tolerance can be induced by repeated oral administration of large doses of antigen in experimental animals.Citation73 Mucosal vaccines also should overcome 2 major obstacles, effectiveness and safety, because of the uniqueness of the mucosal environment.Citation74 The appearance of a new potent mucosal vaccine also brings other challenges, such as antigen degradation by proteolytic activities, low dose of antigen absorbed, and lack of potent mucosal adjuvants.Citation60 We need to probe oral vehicle delivery systems more deeply to overcome these issues. The capacity of M cells to sample antigens, transfer luminal bacteria, and stimulate mucosal immune responses, makes them fascinating targets for mucosal vaccine delivery. Results of previous studies imply that we require safe and effective oral mucosal vaccines with M-cell-targeting strategies, because the M-cell-targeting vaccines now available do not actively direct antigens to M cells. Recent studies have been exploited to search for specific M cell molecules to develop M-cell-targeted strategies, which will likely promote progress in mucosal vaccines. However, many mechanisms of M cells are still unclear and not known as well-known as those of DCs. Therefore, to develop M-cell-targeting strategies in mucosal vaccines, further understanding of mucosal immunology and M cell biology is required, and M cell function, and identification of more M-cell-specific molecules and effective mucosal adjuvants are crucial for further development of M-cell-targeted vaccines.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Funding

This study was financially supported by the Chinese “863” National Programs for High Technology Research and Development (Grant No.: 2011AA10A211), the National Pig Industrial System (CARS-36–06B), and the Special Fund for Agro-scientific Research in the Public Interest (201203039).

References

  • Otczyk DC, Cripps AW. Mucosal immunization A realistic alternative. Hum Vaccines 2010; 6:978-1006; PMID:21150284; http://dx.doi.org/10.4161/hv.6.12.13142
  • Brandtzaeg P, Kiyono H, Pabst R, Russell MW. Terminology: nomenclature of mucosa-associated lymphoid tissue. Mucosal Immunol 2008; 1:31-7; PMID:19079158; http://dx.doi.org/10.1038/mi.2007.9
  • Bockman DE, Cooper MD. Pinocytosis by epithelium associated with lymphoid follicles in the bursa of Fabricius, appendix, and Peyer's patches. An electron microscopic study. Am J Anat 1973; 136:455-77; PMID:4692973; http://dx.doi.org/10.1002/aja.1001360406
  • Makala LH, Suzuki N, Nagasawa H. Peyer's patches: organized lymphoid structures for the induction of mucosal immune responses in the intestine. Pathobiol: J Immunopathol, Mol Cell Biol 2002; 70:55-68; PMID:12476030; http://dx.doi.org/10.1159/000067305
  • Karchev T, Kabakchiev P. M-cells in the epithelium of the nasopharyngeal tonsil. Rhinology 1984; 22:201-10; PMID:6505520
  • Owen RL, Piazza AJ, Ermak TH. Ultrastructural and cytoarchitectural features of lymphoreticular organs in the colon and rectum of adult BALBc mice. Am J Anat 1991; 190:10-8; PMID:1984672; http://dx.doi.org/; http://dx.doi.org/10.1002/aja.1001900103
  • Miller H, Zhang J, Kuolee R, Patel GB, Chen W. Intestinal M cells: the fallible sentinels? World J Gastroenterol: WJG 2007; 13:1477-86; PMID:17461437; http://dx.doi.org/10.3748/wjg.v13.i10.1477
  • Neutra MR, Mantis NJ, Kraehenbuhl JP. Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nat Immunol 2001; 2:1004-9; PMID:11685223; http://dx.doi.org/10.1038/ni1101-1004
  • Sansonetti PJ, Phalipon A. M cells as ports of entry for enteroinvasive pathogens: mechanisms of interaction, consequences for the disease process. Semin Immunol 1999; 11:193-203; PMID:10381865; http://dx.doi.org/10.1006/smim.1999.0175
  • Kyd JM, Cripps AW. Functional differences between M cells and enterocytes in sampling luminal antigens. Vaccine 2008; 26:6221-4; PMID:18852006; http://dx.doi.org/10.1016/j.vaccine.2008.09.061
  • Gebert A, Pabst R. M cells at locations outside the gut. Semin Immunol 1999; 11:165-70; PMID:10381862; http://dx.doi.org/10.1006/smim.1999.0172
  • Kanaya T, Aso H, Miyazawa K, Kido T, Minashima T, Watanabe K, Ohwada S, Kitazawa H, Rose MT, Yamaguchi T. Staining patterns for actin and villin distinguish M cells in bovine follicle-associated epithelium. Res Veter Sci 2007; 82:141-9; PMID:16949627; http://dx.doi.org/10.1016/j.rvsc.2006.05.009
  • Jang MH, Kweon MN, Iwatani K, Yamamoto M, Terahara K, Sasakawa C, Suzuki T, Nochi T, Yokota Y, Rennert PD, et al. Intestinal villous M cells: an antigen entry site in the mucosal epithelium. Proc Nat Acad Sci U S A 2004; 101:6110-5; PMID:15071180; http://dx.doi.org/10.1073/pnas.0400969101
  • Yamamoto M, Rennert P, McGhee JR, Kweon MN, Yamamoto S, Dohi T, Otake S, Bluethmann H, Fujihashi K, Kiyono H. Alternate mucosal immune system: organized Peyer's patches are not required for IgA responses in the gastrointestinal tract. J Immunol 2000; 164:5184-91; PMID:10799877; http://dx.doi.org/10.4049/jimmunol.164.10.5184
  • Kerneis S, Bogdanova A, Kraehenbuhl JP, Pringault E. Conversion by Peyer's patch lymphocytes of human enterocytes into M cells that transport bacteria. Science 1997; 277:949-52; PMID:9252325; http://dx.doi.org/10.1126/science.277.5328.949
  • Owen RL, Pierce NF, Apple RT, Cray WC, Jr. M cell transport of Vibrio cholerae from the intestinal lumen into Peyer's patches: a mechanism for antigen sampling and for microbial transepithelial migration. J Infect Dis 1986; 153:1108-18; PMID:2422297; http://dx.doi.org/10.1093/infdis/153.6.1108
  • Walker RI, Schmauder-Chock EA, Parker JL, Burr D. Selective association and transport of Campylobacter jejuni through M cells of rabbit Peyer's patches. Can J Microbiol 1988; 34:1142-7; PMID:3196964; http://dx.doi.org/10.1139/m88-201
  • Sansonetti PJ, Arondel J, Cantey JR, Prevost MC, Huerre M. Infection of rabbit Peyer's patches by Shigella flexneri: effect of adhesive or invasive bacterial phenotypes on follicle-associated epithelium. Infect Immun 1996; 64:2752-64; PMID:8698505
  • Amerongen HM, Weltzin R, Farnet CM, Michetti P, Haseltine WA, Neutra MR. Transepithelial transport of HIV-1 by intestinal M cells: a mechanism for transmission of AIDS. J Acq Immune Def Synd 1991; 4:760-5; PMID:1856788
  • Karapetian O, Shakhov AN, Kraehenbuhl JP, Acha-Orbea H. Retroviral infection of neonatal Peyer's patch lymphocytes: the mouse mammary tumor virus model. J Exp Med 1994; 180:1511-6; PMID:7931081; http://dx.doi.org/10.1084/jem.180.4.1511
  • Sicinski P, Rowinski J, Warchol JB, Jarzabek Z, Gut W, Szczygiel B, Bielecki K, Koch G. Poliovirus type 1 enters the human host through intestinal M cells. Gastroenterology 1990; 98:56-8; PMID:2152776
  • Gonzalez-Hernandez MB, Liu T, Payne HC, Stencel-Baerenwald J, Ikizler M, Yagita H, Dermody TS, Williams IR, Wobus CE. Efficient norovirus and reovirus replication in the mouse intestine requires microfold (M) cells. J Virol 2014; 88:6934-43; PMID:24696493; http://dx.doi.org/10.1128/JVI.00204-14
  • Miyazawa K, Kanaya T, Takakura I, Tanaka S, Hondo T, Watanabe H, Rose MT, Kitazawa H, Yamaguchi T, Katamine S, et al. Transcytosis of murine-adapted bovine spongiform encephalopathy agents in an in vitro bovine M cell model. J Virol 2010; 84:12285-91; PMID:20861256; http://dx.doi.org/10.1128/JVI.00969-10
  • Gonzalez-Hernandez MB, Liu T, Blanco LP, Auble H, Payne HC, Wobus CE. Murine norovirus transcytosis across an in vitro polarized murine intestinal epithelial monolayer is mediated by M-like cells. J Virol 2013; 87:12685-93; PMID:24049163; http://dx.doi.org/10.1128/JVI.02378-13
  • Landsverk T. Cryptosporidiosis and the follicle-associated epithelium over the ileal Peyer's patch in calves. Res Veter Sci 1987; 42:299-306; PMID:3303206
  • Ermak TH, Dougherty EP, Bhagat HR, Kabok Z, Pappo J. Uptake and transport of copolymer biodegradable microspheres by rabbit Peyer's patch M cells. Cell Tissue Res 1995; 279:433-6; PMID:7895280; http://dx.doi.org/10.1007/BF00318501
  • Borghesi C, Regoli M, Bertelli E, Nicoletti C. Modifications of the follicle-associated epithelium by short-term exposure to a non-intestinal bacterium. J Pathol 1996; 180:326-32; PMID:8958813; http://dx.doi.org/10.1002/(SICI)1096-9896(199611)180:3%3c326::AID-PATH656%3e3.0.CO;2-6
  • Neutra MR, Phillips TL, Mayer EL, Fishkind DJ. Transport of membrane-bound macromolecules by M cells in follicle-associated epithelium of rabbit Peyer's patch. Cell Tissue Res 1987; 247:537-46; PMID:3568100; http://dx.doi.org/10.1007/BF00215747
  • Owen RL. Sequential uptake of horseradish peroxidase by lymphoid follicle epithelium of Peyer's patches in the normal unobstructed mouse intestine: an ultrastructural study. Gastroenterology 1977; 72:440-51; PMID:832793
  • Nicoletti C. Unsolved mysteries of intestinal M cells. Gut 2000; 47:735-9; PMID:11034595; http://dx.doi.org/10.1136/gut.47.5.735
  • Nakato G, Hase K, Suzuki M, Kimura M, Ato M, Hanazato M, Tobiume M, Horiuchi M, Atarashi R, Nishida N, et al. Cutting Edge: brucella abortus exploits a cellular prion protein on intestinal M cells as an invasive receptor. J Immunol 2012; 189:1540-4; PMID:22772447; http://dx.doi.org/10.4049/jimmunol.1103332
  • Neutra MR, Frey A, Kraehenbuhl JP. Epithelial M cells: gateways for mucosal infection and immunization. Cell 1996; 86:345-8; PMID:8756716; http://dx.doi.org/10.1016/S0092-8674(00)80106-3
  • Lim JS, Na HS, Lee HC, Choy HE, Park SC, Han JM, Cho KA. Caveolae-mediated entry of Salmonella typhimurium in a human M-cell model. Biochem Biophys Res Commun 2009; 390:1322-7; PMID:19879241; http://dx.doi.org/10.1016/j.bbrc.2009.10.145
  • Jones BD, Ghori N, Falkow S. Salmonella typhimurium initiates murine infection by penetrating and destroying the specialized epithelial M cells of the Peyer's patches. J Exp Med 1994; 180:15-23; PMID:8006579; http://dx.doi.org/10.1084/jem.180.1.15
  • Menard R, Dehio C, Sansonetti PJ. Bacterial entry into epithelial cells: the paradigm of Shigella. Trends Microbiol 1996; 4:220-6; PMID:8795157; http://dx.doi.org/10.1016/0966-842X(96)10039-1
  • KuoLee R, Chen WX. M cell-targeted delivery of vaccines and therapeutics. Expert Opin Drug Del 2008; 5:693-702; PMID:18532924; http://dx.doi.org/10.1517/17425247.5.6.693
  • Kanaya T, Hase K, Takahashi D, Fukuda S, Hoshino K, Sasaki I, Hemmi H, Knoop KA, Kumar N, Sato M, et al. The Ets transcription factor Spi-B is essential for the differentiation of intestinal microfold cells. Nat Immunol 2012; 13:729 -+; PMID:22706340; http://dx.doi.org/10.1038/ni.2352
  • Hase K, Kawano K, Nochi T, Pontes GS, Fukuda S, Ebisawa M, Kadokura K, Tobe T, Fujimura Y, Kawano S, et al. Uptake through glycoprotein 2 of FimH(+) bacteria by M cells initiates mucosal immune response. Nature 2009; 462:226-U101; PMID:19907495; http://dx.doi.org/10.1038/nature08529
  • Lapthorne S, MacSharry J, Scully P, Nally K, Shanahan F. Differential intestinal M-cell gene expression response to gut commensals. Immunology 2012; 136:312-24; PMID:22385384; http://dx.doi.org/10.1111/j.1365-2567.2012.03581.x
  • Gebert A. Identification of M-Cells in the Rabbit Tonsil by Vimentin Immunohistochemistry and in-Vivo Protein-Transport. Histochem Cell Biol 1995; 104:211-20; PMID:8542447; http://dx.doi.org/10.1007/BF01835154
  • Rey J, Garin N, Spertini F, Corthesy B. Targeting of secretory IgA to Peyer's patch dendritic and T cells after transport by intestinal M cells. J Immunol 2004; 172:3026-33; PMID:14978107; http://dx.doi.org/10.4049/jimmunol.172.5.3026
  • Rochereau N, Drocourt D, Perouzel E, Pavot V, Redelinghuys P, Brown GD, Tiraby G, Roblin X, Verrier B, Genin C, et al. Dectin-1 is essential for reverse transcytosis of glycosylated SIgA-antigen complexes by intestinal M cells. Plos Biol 2013; 11:e1001658; PMID:24068891; http://dx.doi.org/10.1371/journal.pbio.1001658
  • Lo DD, Ling J, Eckelhoefer AH. M cell targeting by a Claudin 4 targeting peptide can enhance mucosal IgA responses. Bmc Biotechnol 2012; 12:7; PMID:22413871; http://dx.doi.org/10.1186/1472-6750-12-7
  • Brayden DJ, Baird AW. Apical membrane receptors on intestinal M cells: potential targets for vaccine delivery. Adv Drug Deliver Rev 2004; 56:721-6; PMID:15063586; http://dx.doi.org/10.1016/j.addr.2003.10.036
  • Terahara K, Yoshida M, Igarashi O, Nochi T, Pontes GS, Hase K, Ohno H, Kurokawa S, Mejima M, Takayama N, et al. Comprehensive gene expression profiling of Peyer's patch M cells, villous M-like cells, and intestinal epithelial cells. J Immunol 2008; 180:7840-6; PMID:18523247; http://dx.doi.org/10.4049/jimmunol.180.12.7840
  • Ohno H, Hase K. Glycoprotein 2 (GP2): grabbing the FimH bacteria into M cells for mucosal immunity. Gut Microbes 2010; 1:407-10; PMID:21468225; http://dx.doi.org/10.4161/gmic.1.6.14078
  • Roggenbuck D, Reinhold D, Schierack P, Bogdanos DP, Conrad K, Laass MW. Crohn's disease specific pancreatic antibodies: clinical and pathophysiological challenges. Clin Chem Lab Med: CCLM FESCC 2014; 52:483-94
  • Hanazato M, Nakato G, Nishikawa F, Hase K, Nishikawa S, Ohno H. Selection of an aptamer against mouse GP2 by SELEX. Cell Struct Funct 2014; 39:23-9; PMID:24334484; http://dx.doi.org/10.1247/csf.13019
  • Nakato G, Fukuda S, Hase K, Goitsuka R, Cooper MD, Ohno H. New approach for m-cell-specific molecules screening by comprehensive transcriptome analysis. DNA Res: Int J Rapid Pub Rep Genes Genomes 2009; 16:227-35; PMID:19675110; http://dx.doi.org/10.1093/dnares/dsp013
  • Kaufmann SH. Heat shock proteins and the immune response. Immunol Today 1990; 11:129-36; PMID:2187470; http://dx.doi.org/10.1016/0167-5699(90)90050-J
  • Kim SH, Jung DI, Yang IY, Kim J, Lee KY, Nochi T, Kiyono H, Jang YS. M cells expressing the complement C5a receptor are efficient targets for mucosal vaccine delivery. Eur J Immunol 2011; 41:3219-29; PMID:21887786; http://dx.doi.org/10.1002/eji.201141592
  • Kim SH, Yang IY, Jang SH, Kim J, Truong TT, Pham TV, Truong NU, Lee KY, Jang YS. C5a receptor-targeting ligand-mediated delivery of dengue virus antigen to M cells evokes antigen-specific systemic and mucosal immune responses in oral immunization. Microbes Infect 2013; 15:895-902; PMID:23892099; http://dx.doi.org/10.1016/j.micinf.2013.07.006
  • Wang J, Gusti V, Saraswati A, Lo DD. Convergent and Divergent Development among M Cell Lineages in Mouse Mucosal Epithelium. J Immunol 2011; 187:5277-85; PMID:21984701; http://dx.doi.org/10.4049/jimmunol.1102077
  • Verbrugghe P, Waelput W, Dieriks B, Waeytens A, Vandesompele J, Cuvelier C. Murine m cells express annexin V specifically. J Pathol 2006; 209:240-9; PMID:16552796; http://dx.doi.org/10.1002/path.1970
  • Rand JH, Wu XX, Lin EY, Griffel A, Gialanella P, McKitrick JC. Annexin A5 binds to lipopolysaccharide and reduces its endotoxin activity. mBio 2012; 3:e00292-11; PMID:22415004; http://dx.doi.org/10.1128/mBio.00292-11
  • Hase K, Ohshima S, Kawano K, Hashimoto N, Matsumoto K, Saito H, Ohno H. Distinct gene expression profiles characterize cellular phenotypes of follicle-associated epithelium and M cells. DNA Res: Int J Rapid Pub Rep Genes Genomes 2005; 12:127-37; PMID:16303744; http://dx.doi.org/10.1093/dnares/12.2.127
  • Suzuki R, Nourani MR, Saino-Saito S, Abe H, Nochi T, Kiyono H, Spener F, Kondo H, Owada Y. Localization of fatty acid binding protein of epidermal type common to dendritic cells and presumptive macrophages in Peyer's patches and epithelial M cells of mouse intestine. Histochem Cell Biol 2009; 132:577-84; PMID:19787366; http://dx.doi.org/10.1007/s00418-009-0638-5
  • 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 2008; 129:311-20; PMID:18097679; http://dx.doi.org/10.1007/s00418-007-0369-4
  • Finzi G, Cornaggia M, Capella C, Fiocca R, Bosi F, Solcia E, Samloff IM. Cathepsin E in follicle associated epithelium of intestine and tonsils: localization to M cells and possible role in antigen processing. Histochemistry 1993; 99:201-11; PMID:8491674; http://dx.doi.org/10.1007/BF00269138
  • Azizi A, Kumar A, Diaz-Mitoma F, Mestecky J. Enhancing Oral Vaccine Potency by Targeting Intestinal M Cells. PLoS Pathogens 2010; 6:e1001147; PMID:21085599; http://dx.doi.org/10.1371/journal.ppat.1001147
  • Nochi T, Yuki Y, Matsumura A, Mejima M, Terahara K, Kim DY, Fukuyama S, Iwatsuki-Horimoto K, Kawaoka Y, Kohda T, et al. A novel M cell-specific carbohydrate-targeted mucosal vaccine effectively induces antigen-specific immune responses. J Exp Med 2007; 204:2789-96; PMID:17984304; http://dx.doi.org/10.1084/jem.20070607
  • Gupta PN, Vyas SP. Investigation of lectinized liposomes as M-cell targeted carrier-adjuvant for mucosal immunization. Colloid Surface B 2011; 82:118-25; PMID:20843665; http://dx.doi.org/10.1016/j.colsurfb.2010.08.027
  • Huy NX, Kim SH, Yang MS, Kim TG. Immunogenicity of a neutralizing epitope from porcine epidemic diarrhea virus: M cell targeting ligand fusion protein expressed in transgenic rice calli. Plant Cell Rep 2012; 31:1933-42; PMID:22736145; http://dx.doi.org/10.1007/s00299-012-1306-0
  • Kim SH, Seo KW, Kim J, Lee KY, Jang YS. The M cell-targeting ligand promotes antigen delivery and induces antigen-specific immune responses in mucosal vaccination. J Immunol 2010; 185:5787-95; PMID:20952686; http://dx.doi.org/10.4049/jimmunol.0903184
  • Kim SH, Jung DI, Yang IY, Jang SH, Kim J, Truong TT, Pham TV, Truong NU, Lee KY, Jang YS. Application of an M-cell-targeting ligand for oral vaccination induces efficient systemic and mucosal immune responses against a viral antigen. Int Immunol 2013; 25:623-32; PMID:23900425; http://dx.doi.org/10.1093/intimm/dxt029
  • Lycke N. Recent progress in mucosal vaccine development: potential and limitations. Nat Rev Immunol 2012; 12:592-605; PMID:22828912; http://dx.doi.org/10.1038/nri3251
  • Kunisawa J, Kurashima Y, Kiyono H. Gut-associated lymphoid tissues for the development of oral vaccines. Adv Drug Deliver Rev 2012; 64:523-30; PMID:21827802; http://dx.doi.org/10.1016/j.addr.2011.07.003
  • Lim JS, Na HS, Lee HC, Choy HE, Park SC, Han JM, Cho KA. Caveolae-mediated entry of Salmonella typhimurium in a human M-cell model. Biochem Biophys Res Commun 2009; 390:1322-7; PMID:19879241; http://dx.doi.org/10.1016/j.bbrc.2009.10.145
  • Malik B, Goyal AK, Markandeywar TS, Rath G, Zakir F, Vyas SP. Microfold-cell targeted surface engineered polymeric nanoparticles for oral immunization. J Drug Target 2012; 20:76-84; PMID:21942475; http://dx.doi.org/10.3109/1061186X.2011.611516
  • Kandori H, Hirayama K, Takeda M, Doi K. Histochemical, lectin-histochemical and morphometrical characteristics of intestinal goblet cells of germfree and conventional mice. Exp Anim Tokyo 1996; 45:155-60; PMID:8726140; http://dx.doi.org/10.1538/expanim.45.155
  • Rynda A, Maddaloni M, Mierzejewska D, Ochoa-Reparaz J, Maslanka T, Crist K, Riccardi C, Barszczewska B, Fujihashi K, McGhee JR, et al. Low-dose tolerance is mediated by the microfold cell ligand, reovirus protein sigma 1. J Immunol 2008; 180:5187-200; PMID:18390700; http://dx.doi.org/10.4049/jimmunol.180.8.5187
  • Wu Y, Wang X, Csencsits KL, Haddad A, Walters N, Pascual DW. M cell-targeted DNA vaccination. Proc Nat Acad Sci U S A 2001; 98:9318-23; PMID:11459939; http://dx.doi.org/10.1073/pnas.161204098
  • Faria AMC, Weiner HL. Oral tolerance. Immunol Rev 2005; 206:232-59; PMID:16048553; http://dx.doi.org/10.1111/j.0105-2896.2005.00280.x
  • Fujkuyama Y, Tokuhara D, Kataoka K, Gilbert RS, McGhee JR, Yuki Y, Kiyono H, Fujihashi K. Novel vaccine development strategies for inducing mucosal immunity. Expert Rev Vaccines 2012; 11:367-79; PMID:22380827; http://dx.doi.org/10.1586/erv.11.196
  • 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 2008; 129:311-20; PMID:18097679; http://dx.doi.org/10.1007/s00418-007-0369-4
  • Wong NA, Herriot M, Rae F. An immunohistochemical study and review of potential markers of human intestinal M cells. Eur J Histochem 2003; 47:143-50; PMID:12777211
  • Jepson MA, Mason CM, Bennett MK, Simmons NL, Hirst BH. Co-expression of vimentin and cytokeratins in M cells of rabbit intestinal lymphoid follicle-associated epithelium. Histochem J 1992; 24:33-9; PMID:1372597; http://dx.doi.org/10.1007/BF01043285
  • Gebert A, Hach G, Bartels H. Co-localization of vimentin and cytokeratins in M-cells of rabbit gut-associated lymphoid tissue (GALT). Cell Tissue Res 1992; 269:331-40; PMID:1384978; http://dx.doi.org/10.1007/BF00319625
  • Lelouard H, Reggio H, Mangeat P, Neutra M, Montcourrier P. Mucin-related epitopes distinguish M cells and enterocytes in rabbit appendix and Peyer's patches. Infect Immun 1999; 67:357-67; PMID:9864237
  • Clark MA, Hirst BH, Jepson MA. M-cell surface beta 1 integrin expression and invasin-mediated targeting of Yersinia pseudotuberculosis to mouse Peyer's patch M cells. Infect Immun 1998; 66:1237-43; PMID:9488419
  • Clark AM, Hirst BH. Expression of junction-associated proteins differentiates mouse intestinal M cells from enterocytes. Histochem Cell Biol 2002; 118:137-47; PMID:12189517
  • Rautenberg K, Cichon C, Heyer G, Demel M, Schmidt MA. Immunocytochemical characterization of the follicle-associated epithelium of Peyer's patches: anti-cytokeratin 8 antibody (clone 4.1.18) as a molecular marker for rat M cells. Eur J Cell Biol 1996; 71:363-70; PMID:8980907
  • Gebert A, Rothkotter HJ, Pabst R. Cytokeratin 18 is an M-cell marker in porcine Peyer's patches. Cell Tissue Res 1994; 276:213-21; PMID:7517344; http://dx.doi.org/10.1007/BF00306106
  • Hondo T, Kanaya T, Takakura I, Watanabe H, Takahashi Y, Nagasawa Y, Terada S, Ohwada S, Watanabe K, Kitazawa H, et al. Cytokeratin 18 is a specific marker of bovine intestinal M cell. Am J Physiol-Gastr L 2011; 300:G442-G53
  • Clark MA, Jepson MA, Simmons NL, Booth TA, Hirst BH. Differential expression of lectin-binding sites defines mouse intestinal M-cells. J Histochem Cytochem: Off J Histochem Soc 1993; 41:1679-87; PMID:7691933; http://dx.doi.org/10.1177/41.11.7691933
  • Clark MA, Jepson MA, Simmons NL, Hirst BH. Selective binding and transcytosis of Ulex europaeus 1 lectin by mouse Peyer's patch M-cells in vivo. Cell Tissue Res 1995; 282:455-61; PMID:8581939; http://dx.doi.org/10.1007/BF00318877
  • Foster N, Clark MA, Jepson MA, Hirst BH. Ulex europaeus 1 lectin targets microspheres to mouse Peyer's patch M-cells in vivo. Vaccine 1998; 16:536-41; PMID:9491509; http://dx.doi.org/10.1016/S0264-410X(97)00222-3
  • Terahara K, Nochi T, Yoshida M, Takahashi Y, Goto Y, Hatai H, Kurokawa S, Jang MH, Kweon MN, Domino SE, et al. Distinct fucosylation of M cells and epithelial cells by Fut1 and Fut2, respectively, in response to intestinal environmental stress. Biochem Biophys Res Commun 2011; 404:822-8; PMID:21172308; http://dx.doi.org/10.1016/j.bbrc.2010.12.067
  • Gupta PN, Khatri K, Goyal AK, Mishra N, Was SP. M-cell targeted biodegradable PLGA nanoparticles for oral immunization against hepatitis B. J Drug Target 2007; 15:701-13; PMID:18041638; http://dx.doi.org/10.1080/10611860701637982
  • Clark MA, Blair H, Liang L, Brey RN, Brayden D, Hirst BH. Targeting polymerised liposome vaccine carriers to intestinal M cells. Vaccine 2001; 20:208-17; PMID:11567766; http://dx.doi.org/10.1016/S0264-410X(01)00258-4
  • Wolf JL, Kauffman RS, Finberg R, Dambrauskas R, Fields BN, Trier JS. Determinants of reovirus interaction with the intestinal M cells and absorptive cells of murine intestine. Gastroenterology 1983; 85:291-300; PMID:6305756
  • Keely S, Glover LE, Weissmueller T, MacManus CF, Fillon S, Fennimore B, Colgan SP. Hypoxia-inducible factor-dependent regulation of platelet-activating factor receptor as a route for gram-positive bacterial translocation across pithelia. Mol Biol Cell 2010; 21:538-46; PMID:20032301; http://dx.doi.org/10.1091/mbc.E09-07-0573

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.