Immune Netw.  2012 Oct;12(5):165-175. 10.4110/in.2012.12.5.165.

Mucosal Immune System and M Cell-targeting Strategies for Oral Mucosal Vaccination

Affiliations
  • 1Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Korea. yongsuk@jbnu.ac.kr
  • 2Department of Oral Microbiology and Institute of Oral Bioscience, Chonbuk National University, Jeonju 561-756, Korea.

Abstract

Vaccination is one of the most effective methods available to prevent infectious diseases. Mucosa, which are exposed to heavy loads of commensal and pathogenic microorganisms, are one of the first areas where infections are established, and therefore have frontline status in immunity, making mucosa ideal sites for vaccine application. Moreover, vaccination through the mucosal immune system could induce effective systemic immune responses together with mucosal immunity in contrast to parenteral vaccination, which is a poor inducer of effective immunity at mucosal surfaces. Among mucosal vaccines, oral mucosal vaccines have the advantages of ease and low cost of vaccine administration. The oral mucosal immune system, however, is generally recognized as poorly immunogenic due to the frequent induction of tolerance against orally-introduced antigens. Consequently, a prerequisite for successful mucosal vaccination is that the orally introduced antigen should be transported across the mucosal surface into the mucosa-associated lymphoid tissue (MALT). In particular, M cells are responsible for antigen uptake into MALT, and the rapid and effective transcytotic activity of M cells makes them an attractive target for mucosal vaccine delivery, although simple transport of the antigen into M cells does not guarantee the induction of specific immune responses. Consequently, development of mucosal vaccine adjuvants based on an understanding of the biology of M cells has attracted much research interest. Here, we review the characteristics of the oral mucosal immune system and delineate strategies to design effective oral mucosal vaccines with an emphasis on mucosal vaccine adjuvants.

Keyword

Adjuvant; Complement 5a receptor; Mucosal immune system; Vaccine

MeSH Terms

Biology
Communicable Diseases
Immune System
Immunity, Mucosal
Lymphoid Tissue
Mucous Membrane
Receptor, Anaphylatoxin C5a
Vaccination
Vaccines
Receptor, Anaphylatoxin C5a
Vaccines

Figure

  • Figure 1 Gut-associated mucosal immune system. In the intestinal immune system, Peyer's patches and isolated lymphoid follicles are inductive sties, while the lamina propria is the effector site. T cell-dependent activation of IgA+ B cells is induced in Peyer's patches. Dendritic cells located beneath the specialized M cells take up and process antigens, and naïve CD4+ T cells are primed. Follicular helper T cells expressing Bcl-6 and CXCR5 interact with B cells in the follicular dendritic cell network and aid in IgA isotype switching in the germinal center. IgA+ long-lived plasma cells and memory B cells migrate to effector sites in the lamina propria (9). SIgA plays critical roles in mucosal immune responses such as immune exclusion, antigen excretion, and intracellular virus neutralization.

  • Figure 2 The Co1 M cell-targeting ligand is able to induce antigen-specific immune responses after oral administration of ligand-conjugated antigen. We found that the M cell-targeting ligand Co1, which is selected by M-like cell-specific binding (A), is able to deliver conjugated antigen into mouse M cells and induce antigen-specific immune responses in systemic and mucosal compartments (B). A targeting receptor on M cells for the Co1 ligand was expected to be C5aR because the Co1 ligand has 60% amino acid sequence homology with OmpH α1 helix of Y. enterocolitica, which is similar in sequence to skp of E. coli, which is a ligand for C5aR. We verified that C5aR is expressed on both mouse M cells and human M-like cells and that it interacts with Y. enterocolitica (C).


Cited by  4 articles

Targeted Delivery of VP1 Antigen of Foot-and-mouth Disease Virus to M Cells Enhances the Antigen-specific Systemic and Mucosal Immune Response
Sae-Hae Kim, Ha-Yan Lee, Yong-Suk Jang
Immune Netw. 2013;13(4):157-162.    doi: 10.4110/in.2013.13.4.157.

Expression of the ATP-gated P2X7 Receptor on M Cells and Its Modulating Role in the Mucosal Immune Environment
Sae-Hae Kim, Ha-Yan Lee, Yong-Suk Jang
Immune Netw. 2015;15(1):44-49.    doi: 10.4110/in.2015.15.1.44.

N-terminal Domain of the Spike Protein of Porcine Epidemic Diarrhea Virus as a New Candidate Molecule for a Mucosal Vaccine
Sae-Hae Kim, Byeol-Hee Cho, Kyung-Yeol Lee, Yong-Suk Jang
Immune Netw. 2018;18(3):e21.    doi: 10.4110/in.2018.18.e21.

N-terminal Domain of the Spike Protein of Porcine Epidemic Diarrhea Virus as a New Candidate Molecule for a Mucosal Vaccine
Sae-Hae Kim, Byeol-Hee Cho, Kyung-Yeol Lee, Yong-Suk Jang
Immune Netw. 2018;18(3):.    doi: 10.4110/in.2018.18.e21.


Reference

1. Almond JW. Vaccine renaissance. Nat Rev Microbiol. 2007. 5:478–481.
Article
2. Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-first century society. Nat Rev Immunol. 2011. 11:865–872.
Article
3. Mitragotri S. Immunization without needles. Nat Rev Immunol. 2005. 5:905–916.
Article
4. Chen K, Cerutti A. Vaccination strategies to promote mucosal antibody responses. Immunity. 2010. 33:479–491.
Article
5. Shin S, Desai SN, Sah BK, Clemens JD. Oral vaccines against cholera. Clin Infect Dis. 2011. 52:1343–1349.
Article
6. Czerkinsky C, Holmgren J. Enteric vaccines for the developing world: a challenge for mucosal immunology. Mucosal Immunol. 2009. 2:284–287.
Article
7. Pasetti MF, Simon JK, Sztein MB, Levine MM. Immunology of gut mucosal vaccines. Immunol Rev. 2011. 239:125–148.
Article
8. Weiner HL, da Cunha AP, Quintana F, Wu H. Oral tolerance. Immunol Rev. 2011. 241:241–259.
Article
9. Lycke N. Recent progress in mucosal vaccine development: potential and limitations. Nat Rev Immunol. 2012. 12:592–605.
Article
10. Ogra PL. Mucosal immunity: some historical perspective on host-pathogen interactions and implications for mucosal vaccines. Immunol Cell Biol. 2003. 81:23–33.
Article
11. Artis D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol. 2008. 8:411–420.
Article
12. Wells JM, Rossi O, Meijerink M, van Baarlen P. Epithelial crosstalk at the microbiota-mucosal interface. Proc Natl Acad Sci USA. 2011. 108 Suppl 1:4607–4614.
Article
13. Iwasaki A. Mucosal dendritic cells. Annu Rev Immunol. 2007. 25:381–418.
Article
14. McGuckin MA, Lindén SK, Sutton P, Florin TH. Mucin dynamics and enteric pathogens. Nat Rev Microbiol. 2011. 9:265–278.
Article
15. Garrett WS, Gordon JI, Glimcher LH. Homeostasis and inflammation in the intestine. Cell. 2010. 140:859–870.
Article
16. Heazlewood CK, Cook MC, Eri R, Price GR, Tauro SB, Taupin D, Thornton DJ, Png CW, Crockford TL, Cornall RJ, Adams R, Kato M, Nelms KA, Hong NA, Florin TH, Goodnow CC, McGuckin MA. Aberrant mucin assembly in mice causes endoplasmic reticulum stress and spontaneous inflammation resembling ulcerative colitis. PLoS Med. 2008. 5:e54.
Article
17. Biton M, Levin A, Slyper M, Alkalay I, Horwitz E, Mor H, Kredo-Russo S, Avnit-Sagi T, Cojocaru G, Zreik F, Bentwich Z, Poy MN, Artis D, Walker MD, Hornstein E, Pikarsky E, Ben-Neriah Y. Epithelial microRNAs regulate gut mucosal immunity via epithelium-T cell crosstalk. Nat Immunol. 2011. 12:239–246.
Article
18. Bevins CL, Salzman NH. Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nat Rev Microbiol. 2011. 9:356–368.
Article
19. Kaser A, Tomczak M, Blumberg RS. "ER stress(ed out)!": Paneth cells and ischemia-reperfusion injury of the small intestine. Gastroenterology. 2011. 140:393–396.
Article
20. Niederreiter L, Kaser A. Endoplasmic reticulum stress and inflammatory bowel disease. Acta Gastroenterol Belg. 2011. 74:330–333.
21. Iliev ID, Mileti E, Matteoli G, Chieppa M, Rescigno M. Intestinal epithelial cells promote colitis-protective regulatory T-cell differentiation through dendritic cell conditioning. Mucosal Immunol. 2009. 2:340–350.
Article
22. Rimoldi M, Chieppa M, Salucci V, Avogadri F, Sonzogni A, Sampietro GM, Nespoli A, Viale G, Allavena P, Rescigno M. Intestinal immune homeostasis is regulated by the crosstalk between epithelial cells and dendritic cells. Nat Immunol. 2005. 6:507–514.
Article
23. He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, Shan M, Chadburn A, Villanacci V, Plebani A, Knowles DM, Rescigno M, Cerutti A. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of the cytokine APRIL. Immunity. 2007. 26:812–826.
Article
24. Kyd JM, Cripps AW. Functional differences between M cells and enterocytes in sampling luminal antigens. Vaccine. 2008. 26:6221–6224.
Article
25. Jang MH, Kweon MN, Iwatani K, Yamamoto M, Terahara K, Sasakawa C, Suzuki T, Nochi T, Yokota Y, Rennert PD, Hiroi T, Tamagawa H, Iijima H, Kunisawa J, Yuki Y, Kiyono H. Intestinal villous M cells: an antigen entry site in the mucosal epithelium. Proc Natl Acad Sci USA. 2004. 101:6110–6115.
Article
26. Corr SC, Gahan CC, Hill C. M-cells: origin, morphology and role in mucosal immunity and microbial pathogenesis. FEMS Immunol Med Microbiol. 2008. 52:2–12.
Article
27. Owen RL, Jones AL. Epithelial cell specialization within human Peyer's patches: an ultrastructural study of intestinal lymphoid follicles. Gastroenterology. 1974. 66:189–203.
Article
28. Heath JP. Epithelial cell migration in the intestine. Cell Biol Int. 1996. 20:139–146.
Article
29. Gebert A, Fassbender S, Werner K, Weissferdt A. The development of M cells in Peyer's patches is restricted to specialized dome-associated crypts. Am J Pathol. 1999. 154:1573–1582.
Article
30. Hsieh EH, Fernandez X, Wang J, Hamer M, Calvillo S, Croft M, Kwon BS, Lo DD. CD137 is required for M cell functional maturation but not lineage commitment. Am J Pathol. 2010. 177:666–676.
Article
31. 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. J Immunol. 2009. 183:5738–5747.
Article
32. Mach J, Hshieh T, Hsieh D, Grubbs N, Chervonsky A. Development of intestinal M cells. Immunol Rev. 2005. 206:177–189.
Article
33. Clark MA, Jepson MA. Intestinal M cells and their role in bacterial infection. Int J Med Microbiol. 2003. 293:17–39.
Article
34. Pickard JM, Chervonsky AV. Sampling of the intestinal microbiota by epithelial M cells. Curr Gastroenterol Rep. 2010. 12:331–339.
Article
35. Azizi A, Kumar A, Diaz-Mitoma F, Mestecky J. Enhancing oral vaccine potency by targeting intestinal M cells. PLoS Pathog. 2010. 6:e1001147.
Article
36. 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. 99:201–211.
Article
37. Kernéis S, Bogdanova A, Kraehenbuhl JP, Pringault E. Conversion by Peyer's patch lymphocytes of human enterocytes into M cells that transport bacteria. Science. 1997. 277:949–952.
Article
38. Gullberg E, Leonard M, Karlsson J, Hopkins AM, Brayden D, Baird AW, Artursson P. Expression of specific markers and particle transport in a new human intestinal M-cell model. Biochem Biophys Res Commun. 2000. 279:808–813.
Article
39. Brandtzaeg P, Kiyono H, Pabst R, Russell MW. Terminology: nomenclature of mucosa-associated lymphoid tissue. Mucosal Immunol. 2008. 1:31–37.
Article
40. Brandtzaeg P. Mucosal immunity: induction, dissemination, and effector functions. Scand J Immunol. 2009. 70:505–515.
Article
41. Otczyk DC, Cripps AW. Mucosal immunization: a realistic alternative. Hum Vaccin. 2010. 6:978–1006.
Article
42. Bemark M, Boysen P, Lycke NY. Induction of gut IgA production through T cell-dependent and T cell-independent pathways. Ann N Y Acad Sci. 2012. 1247:97–116.
Article
43. Strugnell RA, Wijburg OL. The role of secretory antibodies in infection immunity. Nat Rev Microbiol. 2010. 8:656–667.
Article
44. Cerutti A, Chen K, Chorny A. Immunoglobulin responses at the mucosal interface. Annu Rev Immunol. 2011. 29:273–293.
Article
45. Hoft DF, Brusic V, Sakala IG. Optimizing vaccine development. Cell Microbiol. 2011. 13:934–942.
Article
46. McAleer JP, Kolls JK. Mechanisms controlling Th17 cytokine expression and host defense. J Leukoc Biol. 2011. 90:263–270.
Article
47. Blaschitz C, Raffatellu M. Th17 cytokines and the gut mucosal barrier. J Clin Immunol. 2010. 30:196–203.
Article
48. Sheridan BS, Lefrançois L. Regional and mucosal memory T cells. Nat Immunol. 2011. 12:485–491.
Article
49. Borges O, Lebre F, Bento D, Borchard G, Junginger HE. Mucosal vaccines: recent progress in understanding the natural barriers. Pharm Res. 2010. 27:211–223.
Article
50. Cho KA, Cha JE, Woo SY. Oral tolerance increased the proportion of CD8+ T cells in mouse intestinal lamina propria. Immune Netw. 2008. 8:46–52.
Article
51. Rescigno M, Lopatin U, Chieppa M. Interactions among dendritic cells, macrophages, epithelial cells in the gut: implications for immune tolerance. Curr Opin Immunol. 2008. 20:669–675.
Article
52. Czerkinsky C, Holmgren J. Mucosal delivery routes for optimal immunization: targeting immunity to the right tissues. Curr Top Microbiol Immunol. 2012. 354:1–18.
Article
53. Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, Otipoby KL, Yokota A, Takeuchi H, Ricciardi-Castagnoli P, Rajewsky K, Adams DH, von Andrian UH. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science. 2006. 314:1157–1160.
Article
54. Iwata M, Hirakiyama A, Eshima Y, Kagechika H, Kato C, Song SY. Retinoic acid imprints gut-homing specificity on T cells. Immunity. 2004. 21:527–538.
Article
55. Pavot V, Rochereau N, Genin C, Verrier B, Paul S. New insights in mucosal vaccine development. Vaccine. 2012. 30:142–154.
Article
56. Mestecky J, Nguyen H, Czerkinsky C, Kiyono H. Oral immunization: an update. Curr Opin Gastroenterol. 2008. 24:713–719.
Article
57. Sturm JT, Carr ME, Luxenberg MG, Swoyer JK, Cicero JJ. The prevalence of Neisseria gonorrhoeae and Chlamydia trachomatis in victims of sexual assault. Ann Emerg Med. 1990. 19:587–590.
Article
58. Streatfield SJ, Howard JA. Plant-based vaccines. Int J Parasitol. 2003. 33:479–493.
Article
59. Walmsley AM, Arntzen CJ. Plants for delivery of edible vaccines. Curr Opin Biotechnol. 2000. 11:126–129.
Article
60. Haq TA, Mason HS, Clements JD, Arntzen CJ. Oral immunization with a recombinant bacterial antigen produced in transgenic plants. Science. 1995. 268:714–716.
Article
61. Yuki Y, Tokuhara D, Nochi T, Yasuda H, Mejima M, Kurokawa S, Takahashi Y, Kataoka N, Nakanishi U, Hagiwara Y, Fujihashi K, Takaiwa F, Kiyono H. Oral MucoRice expressing double-mutant cholera toxin A and B subunits induces toxin-specific neutralising immunity. Vaccine. 2009. 27:5982–5988.
Article
62. Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010. 33:492–503.
Article
63. Reed SG, Bertholet S, Coler RN, Friede M. New horizons in adjuvants for vaccine development. Trends Immunol. 2009. 30:23–32.
Article
64. Babai I, Samira S, Barenholz Y, Zakay-Rones Z, Kedar E. A novel influenza subunit vaccine composed of liposome-encapsulated haemagglutinin/neuraminidase and IL-2 or GM-CSF. II. Induction of TH1 and TH2 responses in mice. Vaccine. 1999. 17:1239–1250.
Article
65. Kuolee R, Chen W. M cell-targeted delivery of vaccines and therapeutics. Expert Opin Drug Deliv. 2008. 5:693–702.
Article
66. Gebert A, Rothkötter HJ, Pabst R. M cells in Peyer's patches of the intestine. Int Rev Cytol. 1996. 167:91–159.
Article
67. 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. 16:536–541.
Article
68. Clark MA, Jepson MA, Simmons NL, Hirst BH. Differential surface characteristics of M cells from mouse intestinal Peyer's and caecal patches. Histochem J. 1994. 26:271–280.
Article
69. Gupta PN, Khatri K, Goyal AK, Mishra N, Vyas SP. M-cell targeted biodegradable PLGA nanoparticles for oral immunization against hepatitis B. J Drug Target. 2007. 15:701–713.
Article
70. Clark MA, Blair H, Liang L, Brey RN, Brayden D, Hirst BH. Targeting polymerised liposome vaccine carriers to intestinal M cells. Vaccine. 2001. 20:208–217.
Article
71. Devriendt B, De Geest BG, Goddeeris BM, Cox E. Crossing the barrier: Targeting epithelial receptors for enhanced oral vaccine delivery. J Control Release. 2012. 160:431–439.
Article
72. Nochi T, Yuki Y, Matsumura A, Mejima M, Terahara K, Kim DY, Fukuyama S, Iwatsuki-Horimoto K, Kawaoka Y, Kohda T, Kozaki S, Igarashi O, Kiyono H. A novel M cell-specific carbohydrate-targeted mucosal vaccine effectively induces antigen-specific immune responses. J Exp Med. 2007. 204:2789–2796.
Article
73. Hirabayashi J, Hashidate T, Arata Y, Nishi N, Nakamura T, Hirashima M, Urashima T, Oka T, Futai M, Muller WE, Yagi F, Kasai K. Oligosaccharide specificity of galectins: a search by frontal affinity chromatography. Biochim Biophys Acta. 2002. 1572:232–254.
Article
74. 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 epithelia. Mol Biol Cell. 2010. 21:538–546.
Article
75. Tyrer P, Foxwell AR, Cripps AW, Apicella MA, Kyd JM. Microbial pattern recognition receptors mediate M-cell uptake of a gram-negative bacterium. Infect Immun. 2006. 74:625–631.
Article
76. 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. 185:5787–5795.
Article
77. Clark MA, Hirst BH, Jepson MA. M-cell surface beta1 integrin expression and invasin-mediated targeting of Yersinia pseudotuberculosis to mouse Peyer's patch M cells. Infect Immun. 1998. 66:1237–1243.
Article
78. 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.
Article
79. Hase K, Kawano K, Nochi T, Pontes GS, Fukuda S, Ebisawa M, Kadokura K, Tobe T, Fujimura Y, Kawano S, Yabashi A, Waguri S, Nakato G, Kimura S, Murakami T, Iimura M, Hamura K, Fukuoka S, Lowe AW, Itoh K, Kiyono H, Ohno H. Uptake through glycoprotein 2 of FimH(+) bacteria by M cells initiates mucosal immune response. Nature. 2009. 462:226–230.
Article
80. Nakato G, Hase K, Suzuki M, Kimura M, Ato M, Hanazato M, Tobiume M, Horiuchi M, Atarashi R, Nishida N, Watarai M, Imaoka K, Ohno H. Cutting Edge: Brucella abortus exploits a cellular prion protein on intestinal M cells as an invasive receptor. J Immunol. 2012. 189:1540–1544.
Article
81. 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. 41:3219–3229.
Article
82. Kunisawa J, Kurashima Y, Kiyono H. Gut-associated lymphoid tissues for the development of oral vaccines. Adv Drug Deliv Rev. 2012. 64:523–530.
Article
Full Text Links
  • IN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr