Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2004;4(9):699–710. doi: 10.1038/nri1439

NALT- versus PEYER'S-patch-mediated mucosal immunity

Hiroshi Kiyono 1,, Satoshi Fukuyama 1
PMCID: PMC7097243  PMID: 15343369

Key Points

  • The common mucosal immune system consists of organized inductive sites and diffuse effector sites for the generation of antigen-specific mucosal immunity.

  • In rodents, nasopharynx-associated lymphoid tissue (NALT) is found on both sides of the nasopharyngeal duct, dorsal to the cartilaginous soft palate, and it is considered to be analogous to Waldeyer's ring in humans.

  • NALT is a site of IgA class-switch recombination, and T helper 1 (TH1)- and TH2-cell generation. In this way, it is an important inductive site for the generation of mucosal immunity in the aero-digestive tract, similar to Peyer's patches in the intestine.

  • NALT-targeted immunization efficiently induces antigen-specific immune responses in both mucosal and systemic immune compartments, leading to a two-tiered immunological barrier.

  • NALT tissue genesis begins after birth, whereas the organogenesis of Peyer's patches commences during the gestational period.

  • A condition of 'programmed inflammation', such as that induced by the lymphotoxin-β receptor (LT-βR)–nuclear factor-κB-inducing kinase (NIK)-signalling cascade, is essential for the genesis of Peyer's patches, whereas NALT organogenesis is independent of this LT-βR–NIK-mediated inflammation.

  • NALT organogenesis is also independent of the interleukin-7 receptor (IL-7R)-mediated tissue-genesis programme that is essential for the development of Peyer's patches.

  • ID2 (inhibitor of DNA binding 2)-dependent, but not ROR-γ (retinoic-acid-receptor-related orphan receptor-γ)-dependent, CD3CD4+CD45+ inducer cells are essential for NALT organogenesis.

  • A thorough understanding of the unique molecular and cellular properties of NALT is important for the development of a successful intranasally administered vaccine.

Abstract

Recent studies indicate that the mechanism of nasopharynx-associated lymphoid tissue (NALT) organogenesis is different from that of other lymphoid tissues. NALT has an important role in the induction of mucosal immune responses, including the generation of T helper 1 and T helper 2 cells, and IgA-committed B cells. Moreover, intranasal immunization can lead to the induction of antigen-specific protective immunity in both the mucosal and systemic immune compartments. Therefore, a greater understanding of the differences between NALT and other organized lymphoid tissues, such as Peyer's patches, should facilitate the development of nasal vaccines.

Main

The mucosal immune system is responsible both for mediating the symbiotic relationship between the host and endogenous microorganisms (commensal bacteria), and for functioning as a first line of physical and immunological defence against invading pathogens1. Through innate and acquired immunity, the mucosal immune system maintains immunological homeostasis along the vast expanse of the epithelial surface area, ranging from the oral and nasal cavities to the respiratory, intestinal and genito-urinary tracts.

The initiation of antigen-specific immune responses occurs at special 'gateways', which comprise MICROFOLD (M) CELLS located in the epithelium overlying follicles of the mucosa-associated lymphoid tissues (MALT). These contain all of the immunocompetent cells that are required for the generation of an immune response (that is, T cells, B cells and antigen-presenting cells). Peyer's patches, in the gut, and nasopharynx-associated lymphoid tissue (NALT) — two of the main components of MALT — are important inductive tissues for the generation of mucosal immunity through the ingestion and inhalation of antigen in the intestinal and respiratory tracts respectively1 (Fig. 1). The COMMON MUCOSAL IMMUNE SYSTEM (CMIS) connects these inductive sites (that is, the Peyer's patches and NALT) with effector sites (such as the lamina propria of the intestinal and respiratory tracts, and glandular tissues) for the generation of antigen-specific T helper 2 (TH2)-cell-dependent IgA responses, and TH1-cell- and cytotoxic T lymphocyte (CTL)-dependent immune responses, which function as the first line of defence at mucosal surfaces1,2.

Figure 1. The common mucosal immune system.

Figure 1

Luminal antigens are transported to the nasopharynx-associated lymphoid tissue (NALT) and Peyer's patches through microfold (M) cells that are present in the epithelium overlying NALT and Peyer's-patch follicles. Dendritic cells process and present antigens to T cells in these lymphoid tissues. CD4+ T cells that are stimulated by dendritic cells then preferentially induce IgA-committed B-cell development in the germinal centre of the lymphoid follicle. After IgA class switching and affinity maturation, B cells rapidly migrate from NALT and Peyer's patches to the regional cervical lymph nodes and mesenteric lymph nodes respectively, through the efferent lymphatics. Finally, antigen-specific CD4+ T cells and IgA+ B cells migrate to effector sites (such as the nasal passage and intestinal lamina propria) through the thoracic duct and blood circulation. IgA+ B cells and plasmablasts then differentiate into IgA-producing plasma cells in the presence of cytokines (such as interleukin-5 (IL-5) and IL-6) that are produced by T helper 2 (TH2) cells, and they subsequently produce dimeric (or polymeric) forms of IgA. These dimeric forms of IgA then become secretory IgA by binding to polymeric Ig receptors (which become the secretory component in the process of secretory IgA formation) that are displayed on the monolayer of epithelial cells lining the mucosa. Secretory IgA is then released into the nasal passage and intestinal tract. TCR, T-cell receptor.

In this review, we discuss three issues concerning the biology of the NALT immune system: first, we focus on the unique characteristics of its tissue genesis compared with that of Peyer's patches; second, we examine the immunological function of NALT; and third, we discuss manipulation of the NALT immune system to develop mucosal vaccines.

Distinct features of NALT organogenesis

Despite the functional similarity of NALT and Peyer's patches in terms of their role as mucosal inductive sites, their programmes of lymphoid organogenesis are distinct. On the basis of recent studies, the unique characteristics of NALT development compared with those of Peyer's patches have become clear in terms of both kinetics and cytokine requirements.

Chronological development. In normal mice, NALT is a bell-shaped tissue that is characterized by an accumulation of lymphoid cells. In contrast to the HIGH ENDOTHELIAL VENULES (HEVs) of Peyer's patches, which express mucosal vascular addressin cell-adhesion molecule 1 (MADCAM1) (Ref. 3), NALT-associated HEVs express peripheral-node addressin (PNAD). Vascular cell-adhesion molecule 1 (VCAM1) has been shown to be associated with the tissue genesis of Peyer's patches, because a cluster of VCAM1+ stromal cells occurs at the site of Peyer's-patch development on day 15.5 after coitus4. To determine when NALT develops, we used immunohistochemistry to analyse PNAD expression in wild-type mice of various ages. NALT formation was not observed during embryogenesis or in newborn mice5 (Fig. 2), whereas Peyer's patches were already present in the embryo as dome-shaped lymphoid tissues6. Instead, PNAD+ HEVs with associated lymphocytes were first detected bilaterally in nasal tissue at 1 week after birth, and the complete formation of bell-shaped NALT (including lymphoid cells) was not observed until 5–8 weeks after birth5 (Fig. 2). In rats, the development of NALT is also observed postnatally as a small accumulation of lymphoid cells7. These findings indicate a prenatal initiation of lymphoid organogenesis for Peyer's patches and a postnatal initiation for NALT. An intriguing possibility is that the NALT-genesis programme is triggered after birth through stimulatory signals that are provided by environmental antigens and mitogens. This view is supported by the finding that nasal administration of cholera toxin, a well-known mucosal immunogen with adjuvant activity, resulted in the acceleration of NALT organogenesis and the development of the bell-shaped lymphoid tissue5. Therefore, environmental stimulation might be essential for NALT organogenesis, although we have observed the formation of NALT in adult mice that were born and raised under germ-free conditions (H.K. and S.F., unpublished observations). Nonetheless, it is a strong possibility that initiation of NALT genesis is programmed to be activated after birth, and the subsequent maturation process is controlled by environmental antigens.

Figure 2. Chronological differences between NALT- and Peyer's-patch tissue genesis.

Figure 2

a | Nasal tissue from newborn mice (day 0) is characterized by an absence of peripheral-node addressin (PNAD)-expressing high endothelial venules (HEVs). The NASOPHARYNX-ASSOCIATED LYMPHOID-TISSUE (NALT) ANLAGEN from one-week-old mice shows a small accumulation of lymphoid cells around a single PNAD-expressing HEV in the nasal tissue. In eight-week-old mice, NALT contains numerous PNAD-expressing HEVs. This figure is reproduced with permission from Ref. 5 © Elsevier (2002). b | The formation of NALT therefore starts after birth, whereas the development of Peyer's patches is initiated during embryogenesis. c | These kinetic differences in the initiation of tissue genesis of NALT and Peyer's patches are also supported by the appearance and frequency of CD3CD4+CD45+ inducer cells in nasal and intestinal tissues. The inducer cells accumulate postnatally at the site of NALT formation, whereas high numbers of these cells are observed in Peyer's patches during the gestational period. E, embryonic day.

Contribution of cytokines to Peyer's-patch and lymph-node organogenesis. To show that cytokine-mediated NALT organogenesis is unique, it is important to summarize the mechanisms of Peyer's-patch and lymph-node organogenesis for comparative purposes. A family of pro-inflammatory cytokines that consists of lymphotoxin (LT) and tumour-necrosis factor (TNF), and their corresponding receptors (LT-β receptor (LT-βR), TNF receptor p55 (TNFRp55) and TNFRp75), creates a condition of 'programmed inflammation', which controls secondary lymphoid-tissue genesis8,9 (Table 1). LTs are essential for secondary lymphoid-tissue organogenesis that is associated with the mucosal immune system, because deletion of either the genes that encode LT or the LT receptors, or artificial blockade of the interaction between the cytokine and its receptor during the embryonic period, results in the inhibition of both Peyer's-patch and peripheral lymph-node development8,10,11. For example, deletion of the Lt-α gene prevented Peyer's-patch formation and greatly limited the number of lymph nodes that developed8. LT-α forms LT-α1β2 heterotrimers that can transduce an activation signal through the LT-βR, contributing to the organization of secondary lymphoid tissues10. When an LT-βR–Ig fusion protein was infused to antagonize the biological function of the LT-α1β2 heterotrimer, lymphoid tissue formed at different anatomical locations depending on which embryonic stage was perturbed by introduction of the fusion protein10. This finding shows that the timing of secondary lymphoid-tissue development is regulated during embryogenesis10,12. We also found that the infusion of LT-βR–Ig between embryonic day (E) 15 and E17 suppressed Peyer's-patch development but had no effect on the formation of lymph nodes13. These studies clearly indicate the importance of the programmed inflammation that is mediated by LT-α1β2 and the LT-βR for the genesis of Peyer's patches (Table 1; Fig. 3), but it is also known that another membrane-bound member of the TNF family, LIGHT, can bind to the LT-βR14. However, lymph nodes and Peyer's patches develop in the absence of LIGHT15. These findings indicate that the LT-α1β2–LT-βR interaction is the essential component of programmed inflammation that initiates Peyer's-patch genesis at a particular time during the gestational period.

Table 1.

Unique organogenesis of NALT characterized by study of gene-manipulated mice

graphic file with name 41577_2004_Article_BFnri1439_Figa_HTML.jpg

Figure 3. Comparison of the organogenesis programme of NALT and Peyer's patches.

Figure 3

CD3CD4+CD45+ cells are considered to be the common inducers of secondary lymphoid tissue. ID2 (inhibitor of DNA binding 2) is indispensable for the induction and differentiation of these inducer cells from their fetal-liver precursors (which have the phenotype CD3CD4CD45+). a | For Peyer's patches, after activation through the interleukin-7 receptor (IL-7R) or TRANCE (tumour-necrosis-factor-related activation-induced cytokine), these CD3CD4+CD45+ cells express the lymphotoxin-α1β2 (LT-α1β2) heterotrimer, which then binds to the LT-β receptor (LT-βR) displayed on stromal cells and induces signal transduction through NIK (nuclear factor-κB (NF-κB)-inducing kinase). In turn, NIK promotes the expression of adhesion molecules and/or chemokines. These homing molecules trigger the accumulation of lymphoid cells at the site of Peyer's patches. So, the IL-7R- and LT-βR-mediated signals are essential for the tissue genesis of Peyer's patches. b | The development of CD3CD4+CD45+ cells in nasopharynx-associated lymphoid tissue (NALT) also requires ID2; however, the initiation of NALT organogenesis is independent of signalling that involves the IL-7R, LT-α1β2–LT-βR interactions and NIK. CCL, CC-chemokine ligand; CXCL, CXC-chemokine ligand; ICAM1, intercellular adhesion molecule 1; IKK-α, inhibitor of NF-κB (IκB) kinase-α; ROR-γ, retinoic-acid-receptor-related orphan receptor-γ; VCAM1, vascular cell-adhesion molecule 1.

An additional cytokine that is associated with the mucosal immune system, namely interleukin-7 (IL-7), also has a crucial role in the initiation of Peyer's-patch genesis. IL-7 is produced by both mouse and human intestinal epithelial cells16,17, and it provides stimulation and growth signals for neighbouring intestinal intraepithelial γδ T CELLS16,18. In mice that are deficient in the IL-7 receptor α-chain (Il-7rα−/−), only the formation of Peyer's patches, and not lymph nodes, was impaired19. Similarly, when IL-7Rα function was blocked by administration of a single injection of an antagonistic monoclonal antibody to pregnant mothers on E15.5, the resulting offspring were deficient in Peyer's patches but showed normal lymph-node development12. These findings further emphasize that the LT-βR- and IL-7R-mediated tissue-genesis programme is crucial for the initiation of Peyer's-patch formation at the appropriate stage of embryogenesis (E14–E17) (Figs 2,3).

Recently, a model that describes the development of Peyer's patches was proposed on the basis of this evidence. It was shown that lymphoid-lineage IL-7R+CD3CD4+CD45+ cells that are considered to be PEYER'S-PATCH INDUCERS express CXC-chemokine receptor 5 (CXCR5) and can produce membrane-associated LT-α1β2 heterotrimer, whereas mesenchymal-lineage VCAM1+ and intercellular adhesion molecule 1 (ICAM1)+ PEYER'S-PATCH ORGANIZERS express the LT-βR20,21 (Fig. 3). Following stimulatory signals that are provided through the IL-7R, Peyer's-patch inducers express LT-α1β2, which activates Peyer's-patch organizers through the LT-βR; and in turn, Peyer's-patch organizers produce chemokines, such as CXC-chemokine ligand 13 (CXCL13) and CC-chemokine ligand 19 (CCL19), which stimulate Peyer's-patch inducers through CXCR5 and CC-chemokine receptor 7 (CCR7) (Ref. 22). The reciprocal interaction between inducer and organizer cells through chemokine and cytokine receptors is essential for the formation of Peyer's patches (Fig. 3), and the loss of any component of either of the signalling programmes is sufficient to disrupt secondary lymphoid-tissue development, as indicated by the loss of Peyer's patches in LT-βR-deficient and IL-7Rα-deficient mice9,23. Furthermore, deletion of the gene that encodes CXCR5 partially reduces the formation and number of Peyer's patches24 (Table 1). The lack of Peyer's patches and lymph nodes in alymphoplasia (aly/aly) mice, which have defective NIK (nuclear factor-κB (NF-κB)-inducing kinase) function, also fits this model, because recent analyses have established that NIK is essential for the transduction of signals through the TNFR family, including those through the LT-βR25,26. So, aly/aly mice lack Peyer's patches because the NIK mutation inhibits the reciprocal interaction between Peyer's-patch inducers and organizers that is mediated through LT-α1β2 and the LT-βR27,28. Further evidence in support of this model comes from studies showing that mice that lack the CD3CD4+CD45+ inducer cells, owing to genetic deletion of the transcriptional regulators ID2 (inhibitor of DNA binding 2) or ROR-γ (retinoic-acid-receptor-related orphan receptor-γ), also lack Peyer's patches and lymph nodes29,30.

LT-βR- and IL-7R-independent NALT organogenesis. Because Peyer's-patch formation requires a cytokine-signalling cascade that involves the IL-7R and the LT-βR (Table 1; Fig. 3), we examined whether an identical receptor-signalling cascade would trigger NALT development. The formation of NALT was studied in mice lacking Peyer's patches and/or lymph nodes, including Lt-α−/−, Lt-β−/− and aly/aly mice, and mice that were treated in utero with the LT-βR–Ig fusion protein5 (Table 1). Nasal lymphoid tissue was detected in all mouse strains lacking Peyer's patches or both Peyer's patches and lymph nodes because of a deficiency in the LT-βR-mediated pro-inflammatory cytokine cascade5. A separate study by Harmsen and colleagues31 confirmed the formation of NALT in the absence of LT-βR-mediated signalling. The authors also showed that NALT formation was reconstituted in mice that were deficient in both TNF and LT-α by the adoptive transfer of wild-type bone marrow, even though Peyer's patches did not develop in these mice31. These findings further support the idea that NALT development does not conform with the model of programmed inflammation that is required for the genesis of Peyer's patches (Fig. 3).

Because Peyer's-patch formation has also been shown to require the IL-7R-mediated signalling pathway, in addition to the LT-βR cascade, NALT development was examined in IL-7R-deficient mice. NALT, but not Peyer's patches, was found to develop in IL-7R-deficient mice5,31. Taken together, these findings directly show that NALT formation is independent of IL-7R- and LT-βR-mediated tissue genesis (Fig. 3).

CD3CD4+CD45+ cells in NALT organogenesis. A unique subset of mononuclear cells that are characterized as being CD3CD4+CD45+ have been shown to function as inducer cells for the organogenesis of secondary lymphoid tissues, including Peyer's patches21. So, a high frequency of CD3CD4+CD45+ cells is observed in the intestinal tract at embryonic stages of development (Fig. 2). Furthermore, Id2 has been identified as one of the genes that is responsible for the induction of these CD3CD4+CD45+ inducer cells30. Not surprisingly, deletion of the Id2 gene completely impaired the genesis of all secondary lymphoid tissues, including both NALT and Peyer's patches5,30. CD3CD4+CD45+ inducer cells were shown to accumulate at the site of NALT formation after birth5 (Fig. 2), thereby clarifying the role of these cells in the induction of NALT development. To directly show that CD3CD4+CD45+ cells are responsible for the genesis of NALT, fetal liver cells were adoptively transferred from wild-type Id2+/+ mice to newborn Id2−/− mice. Seven days after this transfer, CD3CD4+ cells were observed to have migrated to the site of NALT formation, and 7 weeks after transfer, a NALT-like structure was detected5. These findings are the first to show directly in vivo that CD3CD4+CD45+ inducer cells are essential for the initiation of organogenesis of secondary lymphoid tissues (such as NALT).

The transcriptional regulator ROR-γ has also been shown to be required for the development of CD3CD4+CD45+ inducer cells29,32. Deletion of the gene that encodes ROR-γ suppressed Peyer's-patch and lymph-node organogenesis29,33. However, NALT development has been reported in ROR-γ-deficient mice31. This might indicate that although NALT and Peyer's patches have inducer cells of the same phenotype — that is, CD3CD4+CD45+ — those inducer cells can be classified into two distinct groups on the basis of their dependence on ROR-γ and ID2 (Fig. 4). We think that a population of IL-7R-expressing CD3CD4+CD45+ inducer cells that are essential for Peyer's-patch tissue genesis are regulated by both ROR-γ and ID2, whereas a subset of inducer cells that lack IL-7R expression and are required for NALT genesis are regulated by ID2 but not ROR-γ (Fig. 4) — although this has not been proven experimentally. So, the two CD3CD4+CD45+ inducer-cell populations for NALT and Peyer's-patch organogenesis might be determined or programmed at the level of the transcriptional regulator ROR-γ (Fig. 4). In addition, it is also possible that a population of CD3CD4+CD45+ inducer cells is absent in both Id2−/− mice and Ror-γ−/− mice, and instead, another as-yet-undefined cell population — which can substitute for the classical inducer cells during the formation of NALT — is present in Ror-γ−/− mice but not Id2−/− mice. Further studies are required to investigate these possibilities and others.

Figure 4. Model for the induction of organogenesis of NALT and Peyer's patches by two subsets of CD3CD4+CD45+ inducer cells.

Figure 4

CD3CD4+CD45+ cells differentiate from fetal-liver progenitors. We propose that both ROR-γ (retinoic-acid-receptor-related orphan receptor-γ) and ID2 (inhibitor of DNA binding 2) are essential for the generation of interleukin-7 receptor (IL-7R)-expressing CD3CD4+CD45+ cells for the induction of Peyer's-patch organogenesis. By contrast, the generation of the IL-7RCD3CD4+CD45+ inducer cells that are involved in nasopharynx-associated lymphoid tissue (NALT) organogenesis is regulated by ID2 but not ROR-γ. However, this model remains to be tested experimentally, and other possibilities exist (see main text). NALT genesis in ID2-deficient mice can be initiated by the adoptive transfer of CD3CD4+CD45+ cells from wild-type mice, but the maturation of NALT formation is incomplete. This indicates that other cell populations, or the endogenous expression of ID2 at the site of NALT, might be required for the full maturation of NALT.

Immunological features of NALT

In rodents, NALT is found on both sides of the nasopharyngeal duct, dorsal to the cartilaginous soft palate, and it is considered analogous to WALDEYER'S RING in humans34,35. Also, in a recent study, a NALT-like structure of lymphocyte aggregates that form follicles was identified in human nasal mucosa, particularly in the MIDDLE CONCHA of children less than two years of age36, indicating that an equivalent to mouse NALT can develop in humans. NALT consists of follicle-associated epithelium (FAE), HEVs, and T-cell- and B-cell-enriched areas. Antigen-sampling M cells are present in the epithelium of NALT, which is specialized for antigen uptake similar to the FAE of Peyer's patches7,37. Antigen-presenting cells, including dendritic cells (DCs) and macrophages, are also found in NALT38. So, NALT contains all of the lymphoid cells that are required for the induction and regulation of mucosal immune responses to antigens that are delivered to the nasal cavity. For example, the intranasal administration of reovirus resulted in the formation of germinal centres in NALT, leading to the clonal expansion of antigen-induced IgA+ B cells and the subsequent generation of reovirus-specific IgA in the respiratory and intestinal tracts39. Moreover, reovirus-specific CTLs were also induced in NALT with a high frequency. These findings show that NALT can be a potent inductive site for the mucosal immune system. In addition to the induction of positive immune responses, the nasal deposition of antigen has been shown to be effective for the induction of systemic unresponsiveness — a form of mucosally induced tolerance40. So, NALT has been shown to be involved in the generation of positive- and negative-regulatory signals for the induction of antigen-specific immunity and tolerance respectively. The cellular and molecular contributions of the immunocompetent cells present in NALT to the generation of tolerance to mucosally exposed antigens are unknown. Because little is known about the induction of nasally induced tolerance, we focus here on the role of NALT in the induction of protective immunity.

TH0 environment. Characterization of the mRNA that encodes TH1 and TH2 cytokines in CD4+ T cells isolated from mouse NALT revealed a dominant cytokine profile of TH0 CELLS, indicating that these T cells are capable of becoming TH1 or TH2 cells immediately after antigen exposure of the nasal tract41,42,43. CD4+ T cells isolated from NALT of naive wild-type mice are TH0 cells42, so they can become either TH1 or TH2 cells depending on the identity of the nasally administered antigen. Nasal delivery of protein antigens (such as bacterial cell-wall components or virus-associated antigens) together with cholera toxin as a mucosal adjuvant induces antigen-specific TH2-type responses that promote the generation of antigen-specific IgA-producing B cells, both in the nasal passages and at distant mucosal effector sites, including the genito-urinary, respiratory and intestinal tracts41,44,45. By contrast, intranasal vaccination with antigen-expressing recombinant Mycobacterium bovis bacillus Calmette-Guérin (rBCG) results in TH1-cell-mediated immunity43.

IgA class switching. Peyer's patches have long been thought to be the sites for the initiation of CLASS-SWITCH RECOMBINATION (CSR) of μ- to α-gene expression in the gastrointestinal tract, because they contain all of the cellular and microarchitectural elements that are required for the generation of IgA-committed B cells, including germinal-centre-containing B-cell follicles, a FOLLICULAR DC network and an interfollicular T-cell area1,46,47. The germinal-centre region contains a high frequency of IgM+B220+ B cells that express activation-induced cytidine deaminase (AID), which is essential for μ-to α-gene conversion48. In an early study, it was shown that incubation of IgM+IgA B cells isolated from Peyer's patches in the presence of the cytokine transforming growth factor-β (TGF-β) resulted in the generation of IgMIgA+ B cells49,50,51. These post-switch IgA-committed B cells then migrated to mucosal effector tissues (such as the intestinal lamina propria), a process mediated by a group of homing and chemokine receptors and their ligands (such as MADCAM1–α4β7-integrin and CCL25–CCR9 interactions)52,53(discussed later). In the intestinal lamina propria, these cells became IgA+ plasma cells in the presence of the IgA-enhancing cytokines IL-5, IL-6 and IL-10 (Refs 50,5458). So, it was generally accepted that organized lymphoid structures of MALT, such as Peyer's patches, function as the inductive sites for generating IgA-committed B cells through μ- to α-gene CSR, whereas the diffuse tissues of the intestinal lamina propria function as effector sites for the production of IgA1,2 (Fig. 1). However, the finding that IgA class switching can occur in the intestinal lamina propria without involvement of the Peyer's patches59 cast these assumptions into doubt. Stromal-cell-derived TGF-β present in the intestinal lamina propria was shown to trigger IgM+B220+ cells to switch to IgA+ B cells59. Mice with a deficiency in the programmed-inflammation-associated cytokine LT-α do not form Peyer's patches, so the levels of IgA responses are reduced compared with those of wild-type mice60. However, reconstitution of Lt-α−/− mice with LT-expressing bone-marrow cells, or transplantation of an intestinal segment from recombination-activating gene (Rag)−/− mice to Lt-α−/− mice, resulted in the recovery of IgA responses60. These findings imply that at least some IgA-committed B cells can develop, even in the absence of Peyer's patches. Lt-α−/− mice have also been shown to be capable of inducing antigen-specific IgA responses to orally administered Salmonella typhimurium, despite the absence of organized lymphoid tissues associated with the mucosal compartment61. Although these findings provide supporting evidence that the environment of the diffuse lamina propria region is self-sufficient for the μ- to α-gene CSRthat leads to the generation of IgA-committed B cells, it is also possible that unidentified, programmed-inflammation-independent miniature lymphoid aggregates, and recently characterized isolated lymphoid follicles62, have a role in mounting IgA responses in cases of Peyer's-patch deficiency.

The finding that IgA-specific CSR can occur in diffuse mucosal effector tissues indicates that organized mucosal tissue is not essential for the generation of IgA-committed B cells in the digestive tract59, although this new view is still controversial63,64. Because it has been shown that intestinal IgA is produced by two groups of cells, B1 cells and B2 CELLS63,65,66, an interesting possibility is that IgA-specific CSR of B1 cells does not require organized lymphoid structures, whereas these structures are essential for IgA-isotype switching in B2 cells. In support of this view, we have shown that most B cells in gut-associated lymphoid tissues are B2 cells, whereas B1 cells are located preferentially in the intestinal lamina-propria region66. So, we think that the initial antigenic stimulation for the triggering of IgA-isotype switching in B2 cells might be provided by antigen sampling through M cells that are located in the dome epithelium (or FAE) of Peyer's patches. By contrast, the IgA-specific CSR process that occurs for B1 cells might be triggered by antigens sampled through newly identified villous M cells that are located adjacent to lamina-propria regions that do not contain observable lymphoid-like structures67.

Because NALT is of similar importance for the initiation of IgA+ B-cell responses as Peyer's patches, we have investigated whether IgA-isotype switching can also occur in the diffuse tissue of the nasal passage. IgM+B220+ B cells, which are a prerequisite for CSR, were found in the organized inductive sites (NALT) but were mostly absent from the diffuse effector tissues (nasal passage) of the respiratory mucosal immune system68. Similarly, IgM+B220+ B cells were observed in the organized Peyer's patches of the intestinal tract but not in the intestinal lamina propria. So, in this study, IgM+B220+ B cells that are preconditioned to undergo IgA class switching are selectively located in the organized mucosa-associated inductive tissues of NALT and Peyer's patches68. This finding was confirmed by the molecular analysis of IgA CSR-associated mRNA specific for AID, Iα-Cμ CIRCULAR TRANSCRIPTS and Iμ-Cα TRANSCRIPTS. Because the expression of AID and the Iα-Cμ circular transcript are upregulated preferentially during μ- to α-gene conversion and then quickly downregulated, these molecular events are considered to be a hallmark of B cells that are undergoing IgA class switching69. The expression of Iμ-Cα transcripts indicates the completion of IgA-specific CSR59. This analysis showed that the expression of AID-, Iα-Cμ circular transcript- and Iμ-Cα transcript-specific mRNA was restricted to the organized mucosal inductive tissues of NALT and Peyer's patches but was not found in the diffuse effector tissues of the nasal passage and intestinal lamina propria68. Furthermore, these organized mucosal lymphoid tissues are known to be associated with B2 cells66. So, these findings indicate that the IgA class switching, at least for B2 cells, requires the organized lymphoid structures of NALT and Peyer's patches in the aero-digestive tract. NALT was also recently shown to be an important site for the generation of memory B cells, which produce high-affinity IgA70. Taken together, these findings show that NALT contains all of the immunocompetent cells that are required for the induction and regulation of antigen-specific TH1- or TH2-cell-mediated responses and B-cell immune responses.

Differences between NALT- and Peyer's-patch-initiated immune responses. NALT and Peyer's patches are thought to have similar immunological characteristics and biological functions, as well as to contain the same types of resident immunocompetent cell. So, similar to oral immunization, nasal immunization can stimulate antigen-specific TH1- or TH2-cell-mediated responses and IgA responses in distant mucosal effector tissues1,2,41,43,44,45. However, in general, NALT-targeted immunization effectively induces antigen-specific immunity in the respiratory and reproductive tissues, whereas Peyer's-patch-targeted immunization promotes the generation of protective immunity in the gastrointestinal-tract tissues1,2. Further support for a compartmentalized CMIS was provided recently when it was shown that nasal immunization induces the expression of high levels of CCR10 and α4β1-integrin by IgA-committed B cells, allowing them to efficiently traffic to the respiratory and genito-urinary tracts, which express the corresponding ligands, CCL28 and VCAM1 (Refs 71,72). By contrast, orally induced IgA-committed B cells express CCR9 and CCR10, as well as α4β7- and α4β1-integrins, so they migrate to sites such as the small intestine, which express CCL25 and/or CCL28 together with MADCAM1 and/or VCAM1 (Ref. 73).

So, despite NALT and Peyer's patches both belonging to the mucosal immune system, the subtle differences that we have discussed indicate that the tissue genesis and biological functions of NALT and Peyer's patches might differ because of their anatomically and environmentally distinct locations. Targeting the CMIS would therefore seem to be a logical choice for the development of a second generation mucosal (nasal or oral) vaccine to induce antigen-specific immune responses — such as a combination of TH1-cell or TH2-cell responses, CTL responses, and IgA and IgG responses — in the common and/or selective regions of the mucosal compartments, as well as at systemic sites, through the use of the NALT- and Peyer's-patch-initiated mucosal immune responses (Fig. 1).

NALT-based vaccine development

As we have described, NALT is one of the key components of the organized lymphoid tissue, and it contains all of the immunocompetent cells that are required for the induction of antigen-specific immune responses. It is therefore likely to have a central role in the development of a 'nasal vaccine'. Nasal vaccination has proven to be an effective regimen for the stimulation of the respiratory immune system1,2,41,45. Furthermore, this route of mucosal immunization can elicit both humoral and cell-mediated antigen-specific immune responses1,2,41,43,44,45. Another attractive feature of nasal immunization is that it requires a much smaller dose of antigen than does oral vaccination for the induction of antigen-specific mucosal and systemic immune responses, because the antigens are not exposed to degradation by digestive enzymes. Because intranasal administration of vaccine antigen alone has failed to fully stimulate NALT, researchers are attempting to develop an effective NALT-targeted vaccine-antigen delivery system and to develop a safe and effective immune-enhancing molecule (or adjuvant) for intranasal administration with the vaccine antigen.

NALT-targeted vaccine delivery. Antigens are known to be more immunogenic in particulate form than in soluble form, but they are vulnerable to antigen-degrading enzymes and acids that are associated with the mucosal environment. To overcome these obstacles, much effort has been focused on the creation of novel non-toxic and non-immunogenic vaccine vehicles that can effectively deliver even the soluble form of antigen to the organized mucosal inductive tissue. Such vehicles need to protect vaccine components from degradation, enhance their uptake from mucosal surfaces and perhaps function as an adjuvant. Among the various candidates for mucosal antigen delivery, Sendai-virus-associated fusion protein seems particularly suited to function as a molecule that guides antigen to the mucosal epithelium, because the Sendai virus itself uses this fusion protein for the invasion of respiratory epithelial cells74. A novel hybrid antigen-delivery vehicle has also been devised using this envelope fusion glycoprotein of Sendai virus (or using the haemagglutinating virus of Japan, HVJ); the fusion protein is displayed on the surface of liposomes (either fusogenic liposomes or HVJ liposomes) that contain the antigen of interest. When this delivery vehicle was used, intranasally administered antigen that was conjugated to green-fluorescent protein successfully reached the antigen-sampling M cells that are located in the epithelium of NALT75. Fusogenic liposomes were also found to effectively deliver antigen to epithelial cells and macrophages in both NALT and the nasal passages75. Furthermore, it was shown that an intranasally administered HVJ liposome containing the HIV glycoprotein 160 antigen (gp160; also known as env) was a powerful tool for inducing gp160-specific serum IgG, and gp160-specific mucosal IgA was also detected in nasal wash, saliva, faecal extract and vaginal wash76 (Table 2). These findings show that the novel hybrid antigen-delivery vehicle of fusogenic liposomes (or HVJ liposomes) effectively transports vaccine antigen to NALT for the initiation of antigen-specific IgA responses at distant mucosal effector sites. Furthermore, this immunization method can also induce antigen-specific immune responses (such as production of IgG) in the systemic compartment (Table 2).

Table 2.

Novel mucosal adjuvants and delivery systems for the development of nasal vaccines

graphic file with name 41577_2004_Article_BFnri1439_Figb_HTML.jpg

Because antigen-sampling M cells are scattered throughout the NALT epithelium77, it seems logical to develop an M-cell-targeted nasal vaccine. One promising approach has been to use a molecule that is involved in the normal course of invasion of an infectious agent. Reovirus, an enteric pathogen, is known to invade its host through M cells that are located in the epithelium of Peyer's patches78. The 45-kDa viral haemagglutinin σ1 protein of reovirus has a crucial role in its attachment to and entry into M cells79. The virus has been shown to recognize mouse M cells that are present in the airways80, and the recombinant form of the σ1 protein can bind to M cells that are associated with NALT epithelium80. On the basis of these findings, attempts have been made to develop an M-cell-targeted DNA vaccine using the σ1 protein as a guiding molecule81. When conjugated to a eukaryotic expression vector that encodes luciferase (known as pCMVLuc) and administered intranasally, the σ1 protein can specifically bind to the apical surface of M cells that are situated in the follicular epithelium of NALT; it then leads to the generation of luciferase-specific serum IgG and mucosal IgA responses80. A nasal vaccine assembled using the σ1 protein and gp160 resulted in gp160-specific CTL responses in various mucosa-associated and systemic immune compartments, including reproductive tissue and spleen respectively81. These findings further emphasize the efficacy of NALT-targeted immunization for the induction of humoral and/or cell-mediated antigen-specific immune responses in mucosal and systemic immune compartments.

Because it only infrequently causes serious complications, BCG, a commonly used vaccine for the control of tuberculosis, is considered to be a low-risk vaccine. The recombinant form of BCG is a useful vaccine-antigen delivery vehicle, because it has strong adjuvant activity that can induce both humoral and cell-mediated immune responses82. Indeed, systemic administration of rBCG that expresses HIV antigen has been shown to effectively induce cell-mediated immunity43,83. Our own studies have shown that intransal administration of rBCG that expresses V3J1, a neutralizing epitope of HIV, can induce V3-peptide-specific IgG that has neutralizing activity for more than 0.5–1 years in both normal and immunodeficient (interferon-γ-deficient or Il-4−/−) mice43 (Table 2). Furthermore, V3J1–rBCG-induced serum IgG has also been shown to effectively neutralize a homologous strain of HIV43. Accordingly, rBCG shows promise as an effective nasal-immunization vehicle for the induction of prolonged antigen-specific antibody responses.

Creation of safe toxin-based adjuvants. Both cholera toxin that is produced by Vibrio cholerae and the heat-labile enterotoxin of Escherichia coli function as adjuvants to enhance mucosal and serum antibody responses to co-administered protein antigens delivered by oral or nasal routes2,84. Unfortunately, despite their efficacy as mucosal adjuvants, the native forms of cholera toxin (nCT) and heat-labile enterotoxin (nLT) cause severe diarrhoea and so are unsuitable for use in humans. To overcome these hurdles, researchers have substituted a single amino acid to generate non-toxic mutant forms of cholera toxin (mCT) and heat-labile enterotoxin (mLT)85,86,87,88; these retain the adjuvanticity of the native forms but do not induce the ribosylation of ADP that is associated with toxic activity. Our efforts to devise a safe first generation toxin-based adjuvant have focused on mCT S61F (in which phenylalanine replaces serine at position 61) and mCT E112K (in which lysine replaces glutamic acid at position 112); these mutations were created by making a single amino-acid substitution in the active centre of the ADP-ribosyltransferase in the A subunit of cholera toxin89. The two mutant forms of cholera toxin have been shown to be safe by in vitro analyses of ADP-ribosylation activity and cyclic AMP formation, as well as by in vivo examination for diarrhoea-like symptoms. When pneumococcal surface-protein A (PspA) — a new candidate vaccine antigen for preventing infection with Streptococcus pneumoniae — was intranasally administered with mCT, antigen-specific mucosal IgA and systemic IgG responses were elicited90. Mice intranasally immunized with PspA and mCT were also protected against a lethal challenge with S. pneumoniae90. Interestingly, when the tetanus-toxoid vaccine (which is currently administered by injection) was intranasally administered with one of these two mCTs, it generated protective immunity against challenge with the toxin91. An independent study has also shown that mCT E112K is the safest and most effective of the currently available toxin-based mutant adjuvants92. Taken together, these findings support the idea that mCT is a strong candidate for an effective mucosal adjuvant to generate protective immunity by the nasal route of administration. Indeed, these findings indicate that the current preference for injection-type vaccines should be reconsidered, and in future, greater use should be made of spray-type vaccines that include mCT and other safe toxin-based adjuvants (Box 1).

To further enhance the efficacy of the mCT mucosal adjuvant, a second-generation, chimeric-type adjuvant was constructed from the A subunit of mCT (mCTA) and the B subunit of nLT (nLTB); therefore, the adjuvant has the immunobiological properties of both cholera toxin and heat-labile enterotoxin93. Nasal immunization with influenza-virus haemagglutinin plus the newly created chimeric mucosal adjuvant mCTA–nLTB resulted in significant haemagglutinin-specific serum IgG and IgA responses93 (Table 2). In addition, mice that were intranasally immunized with haemagglutinin and mCTA–nLTB showed high levels of haemagglutinin-specific IgA in nasal and lung washes and were protected from viral challenge93. These findings show that nasal vaccines containing mCT or mCTA–nLTB are effective for the induction of protective immunity. The goal of mucosal-vaccine development cannot be realized without the creation of such novel and safe mucosal adjuvants.

Recent progress in clinical application of nasal vaccination. Between the late 1950s and the early 1960s, the efficacy of immunization with an intranasally administered vaccine against infection with influenza virus was shown in a large clinical trial in Osaka, Japan, in which a nasal-spray vaccine containing live attenuated influenza virus was administered to more than 10,000 volunteers94,95. In recent years, two types of intranasally administered influenza vaccine, an inactivated form and a live attenuated form, were introduced in Switzerland and the United States respectively. Indeed, as early as 1997, an inactivated form of nasal vaccine containing a small amount of nLT as a mucosal adjuvant was introduced in Switzerland. However, this influenza vaccine was withdrawn from the market because of the development of BELL'S PALSY by some recipients after nasal vaccination. A causal relationship between the intranasally administered inactivated-influenza vaccine used in Switzerland and the incidence of Bell's palsy was formally established in a recent case–control study96. At this stage, the causes and pathogenesis of Bell's palsy remain unclear; however, because nLT has been shown to have pro-inflammatory properties and possible neurological toxicity97, the co-formulated nLT that is present in the inactivated-influenza vaccine is suspected to be the causative agent98. These findings highlight that the development of a safe mucosal adjuvant is crucial if progress is to be made towards a safe and effective mucosal vaccine.

In the past year, on the basis of promising clinical trials showing the induction of protective immunity, an intranasally administered cold-adapted influenza vaccine, known as 'FluMist', has been made available to healthy Americans of ages 5 to 49 (Refs 99,100). It should be noted that the concept of cold-adapted influenza virus was reported in 1967 (Ref. 101), so more than 35 years were required for this discovery to be translated into an intransal vaccine against infection with influenza virus. More recently, the concept of nasal immunization was adopted for the development of a vaccine against severe acute respiratory syndrome (SARS). An experimental nasally administered vaccine against SARS that consists of a recombinant attenuated parainfluenza virus expressing the envelope spike protein of the SARS coronavirus was able to induce protective immunity in African green monkeys, including SARS-coronavirus-specific neutralizing antibodies102. Although further experiments are essential before this finding can be applied to developing a vaccine against SARS for use in humans, the study emphasizes the usefulness of mucosal immunization for the immunoprophylaxis of infectious diseases.

Concluding remarks

The mucosal immune system is now recognized to be an important first line of defence against invading pathogens. NALTand Peyer's patches are important inductive sites for the initiation of antigen-specific mucosal IgA and serum IgG responses, as well as CTL immune responses, at both mucosal and systemic sites; in this way, both NALT and Peyer's patches function to maximize the two-tiered immunological barrier of the host. The respiratory mucosal immune system has several immunological characteristics that are distinct from those of the Peyer's-patch-centred intestinal mucosal immune system. Although the tissue-genesis programme for other secondary lymphoid tissues, including Peyer's patches, begins during embryonic life, the NALT-organogenesis programme is initiated only after birth. Lymphoid organogenesis of Peyer's patches requires cytokine-mediated programmed inflammation (through the LT-βR) and signalling through the IL-7R, whereas the initiation of NALT development seems to be independent of the IL-7R, LT-α1β2 and the LT-βR. Although the inducer cells for both NALT and Peyer's patches have a common phenotype, that is, CD3CD4+CD45+, NALT-inducer cells seem to be regulated by ID2 alone, whereas Peyer's-patch-inducer cells depend on both ID2 and ROR-γ. Because the organogenesis programme of NALT is different from that of other secondary lymphoid tissues, such as Peyer's patches, efforts should now be aimed at elucidating the distinct molecular characteristics of the NALT-genesis programme and the functional consequences of this. Clearly, it is important to have a thorough understanding of the unique molecular and cellular properties of the NALT-centred mucosal immune system for the development of a successful nasal vaccine.

Box 1 | Advantages and disadvantages of nasal vaccination.

Advantages

  • Is the most effective route to elicit optimal protective immunity in both mucosal and systemic immune compartments.

  • Can effectively induce antigen-specific immunity in the reproductive tract, as well as in the upper respiratory tract.

  • Can generate cross-protective immunity in the gut through the common mucosal immune system.

  • Can avoid degradation of vaccine antigen caused by digestive enzymes, so requires a smaller dose of antigen than oral immunization.

  • Does not require injection, so is less painful.

  • Does not require trained medical personnel for delivery.

Disadvantages

  • Possible deposition of antigen in the central nervous system through the olfactory bulbs and olfactory nerves; this requires further investigation.

  • Requires adjuvant safety to be clinically determined; clinical studies indicate that Bell's palsy is caused by influenza nasal vaccine that contains the native form of Escherichia coli heat-labile enterotoxin as a mucosal adjuvant.

Acknowledgements

Our work is supported by grants from the Core Research for Evolutional Science and Technology of Japan Science and Technology Corporation, the Ministry of Education, Science, Sports and Culture of Japan, the Ministry of Health, Labor, and Welfare of Japan, and the Uehara Memorial Foundation (Japan). We express appreciation of our present and former colleagues at the University of Tokyo, (Tokyo, Japan), Osaka University (Osaka, Japan) and the University of Alabama at Birmingham (United States) for their help and contributions to the generation of the results summarized in this review. Finally, our appreciation extends to K. K. McGhee and J. R. McGhee for editorial help and careful reading of the manuscript, respectively.

Glossary

MICROFOLD (M) CELLS

Specialized antigen-sampling cells that are located in the follicle-associated epithelium of the organized mucosa-associated lymphoid tissues. M cells deliver antigens by transepithelial vesicular transport from the aero-digestive lumen directly to the subepithelial lymphoid tissues of nasopharynx-associated lymphoid tissue and Peyer's patches.

COMMON MUCOSAL IMMUNE SYSTEM

(CMIS). An integrated pathway that allows communication between the organized mucosa-associated lymphoid tissues (inductive sites) and the diffuse mucosal tissues (effector sites), enabling the induction and regulation of host-protective immunity against pathogenic microorganisms.

HIGH ENDOTHELIAL VENULES

(HEVs). Venules (small veins that join capillaries to larger veins) that have a high-walled endothelium and are present in the paracortex of lymph nodes and tonsils, as well as in the interfollicular areas of Peyer's patches. HEVs are essential for lymphocyte homing to secondary lymphoid organs.

NASOPHARYNX-ASSOCIATED LYMPHOID-TISSUE (NALT) ANLAGEN

The site for the initiation of NALT development. At this site, the accumulation of CD3CD4+CD45+ cells and the expression of peripheral-node addressin (PNAD) by venules are observed in infant nasal tissues.

γδ T CELLS

T cells that express heterodimers consisting of the γ- and δ-chains of the T-cell receptor. They are present mainly in the intestinal epithelium as intraepithelial lymphocytes (IELs). Although the exact function of γδ T cells (or IELs) is still unknown, it has been suggested that mucosal γδ T cells are involved in the innate immune responses of the mucosal immune system.

PEYER'S-PATCH INDUCERS

CD3CD4+CD45+ cells that express the interleukin-7 receptor and lymphotoxin-α1β2. They differentiate from fetal liver cells and can induce Peyer's-patch formation during the embryonic stage.

PEYER'S-PATCH ORGANIZERS

Lymphotoxin-β-receptor-positive stromal cells that are present in the anlagen of Peyer's patches and also express both VCAM1 (vascular cell-adhesion molecule 1) and ICAM1 (intercellular adhesion molecule 1). Peyer's-patch development is initiated with the cooperation of Peyer's-patch inducers.

WALDEYER'S RING

Human nasopharynx-associated lymphoid tissues, including the palatine tonsils and adenoids, which are considered to have an important role in the induction and modulation of mucosal immunity in the upper respiratory tract.

MIDDLE CONCHA

Bony plate that extends from the central section of the lateral wall of the nasal cavity.

TH0 CELLS

Precursors of T helper 1 (TH1) cells and TH2 cells, which produce both interferon-γ and interleukin-4. This T-cell population has the capacity to become TH1- and/or TH2 cells.

CLASS-SWITCH RECOMBINATION

Molecular alteration of the constant-region gene of the immunoglobulin heavy chain (CH) that leads to a switch in expression from the Cμ (or Cδ) region to one of the other CH genes. This leads to a switch in the class of the immunoglobulin that is displayed on the cell-surface of the B cell (and that subsequently differentiating plasma cells produce) — from IgM (or IgD) to IgG, IgA or IgE — without altering the specificity of the immunoglobulin.

FOLLICULAR DC

(FDC). Cell with a dendritic morphology that is present in lymph nodes. These cells display on their surface intact antigens that are held in immune complexes, and B cells present in the lymph node can interact with these antigens. FDCs are of non-haematopoietic origin and are not related to dendritic cells.

B2 CELLS

IgMlowIgDhiMAC1B220hiCD23+ cells that originate from bone marrow and are distributed to mucosal and systemic immune compartments for the continuous secretion of antibodies with high affinity and fine specificity.

Iα-Cμ CIRCULAR TRANSCRIPTS

Circular DNA molecules that are present in activated B cells and consist of Iα (intervening region-α) and Cμ genes. They are a hallmark of B cells that are in the process of IgA class switching.

Iμ-Cα TRANSCRIPTS

Germline transcripts that can be detected in IgA-committed B cells after class switching.

BELL'S PALSY

Facial paralysis that is thought to be triggered by viral infection. The facial nerve is oedematous in patients suffering from this disease.

Biographies

Hiroshi Kiyono is currently Professor and Director of the Division of Mucosal Immunology in the Department of Microbiology and Immunology, at the Institute of Medical Science, the University of Tokyo, Tokyo, Japan. In 1977, he obtained his D.M.D. from Nihon University School of Dentistry, Matsudo, Japan. In 1983, he then obtained his Ph.D. in pathology from the University of Alabama at Birmingham (UAB), United States. His dissertation research focused on the characterization of Peyer's patches as the inductive site for the generation of T helper cells that promote the production of IgA. He then joined UAB as a faculty member, and in 1986, he worked at the Max-Planck-Institute for Biology in Tübingen, Germany. On the basis of his research contributions to the understanding of the unique molecular and cellular characteristics of the mucosal immune system, he was a recipient of the National Institutes of Health New Investigator Award (in 1984) and Research Career Development Award (in 1988). In 1991, he was promoted to the rank of Professor at UAB, and he became Co-director of the Immunobiology Vaccine Center. In 1994, he then joined the Research Institute for Microbial Diseases at Osaka University, Osaka, Japan, to build a programme in Mucosal Immunology, of which he was Professor and Chairman. His research efforts during the past 25 years have focused on elucidating the function and properties of the mucosal immune system with a view to the development of mucosal vaccines and mucosal immunotherapies.

Satoshi Fukuyama is a postdoctoral fellow in Hiroshi Kiyono's laboratory. In 1997, he obtained his M.D. from the School of Medicine at Kagoshima University, Kagoshima, Japan. In 1999, after his clinical training in otolaryngology, he joined the Kiyono laboratory at Osaka University as a graduate student, where he investigated the unique immunological characteristics of the respiratory-tract region of the mucosal immune system. He was awarded a Ph.D. in 2002, on the basis of his research showing that NALT organogenesis occurs through a programme that is distinct from the tissue-genesis programmes of other secondary lymphoid tissues. His current research is focused on the identification of novel tissue-genesis signalling molecules involved in NALT development.

Related links

DATABASES

Entrez Gene

CCL19

CCR7

CD3

CD4

CD45

CXCL13

CXCR5

gp160

ICAM1

ID2

IL-7

IL-7Rα

Lt-α

LT-βR

MADCAM1

NIK

ROR-γ

TNF

TNFRp55

TNFRp75

VCAM1

FURTHER INFORMATION

Hiroshi Kiyono's homepage

Competing interests

The authors declare no competing financial interests.

References

  • 1.Mestecky J, Blumberg R, Kiyono H, McGhee JR. Fundamental Immunology. 2003. pp. 965–1020. [Google Scholar]
  • 2.Yuki Y, Kiyono H. New generation of mucosal adjuvants for the induction of protective immunity. Rev. Med. Virol. 2003;13:293–310. doi: 10.1002/rmv.398. [DOI] [PubMed] [Google Scholar]
  • 3.Csencsits KL, Jutila MA, Pascual DW. Nasal-associated lymphoid tissue: phenotypic and functional evidence for the primary role of peripheral node addressin in naive lymphocyte adhesion to high endothelial venules in a mucosal site. J. Immunol. 1999;163:1382–1389. [PubMed] [Google Scholar]
  • 4.Adachi S, Yoshida H, Kataoka H, Nishikawa S. Three distinctive steps in Peyer's patch formation of murine embryo. Int. Immunol. 1997;9:507–514. doi: 10.1093/intimm/9.4.507. [DOI] [PubMed] [Google Scholar]
  • 5.Fukuyama S, et al. Initiation of NALT organogenesis is independent of the IL-7R, LTβR, and NIK signaling pathways but requires the Id2 gene and CD3−CD4+CD45+ cells. Immunity. 2002;17:31–40. doi: 10.1016/S1074-7613(02)00339-4. [DOI] [PubMed] [Google Scholar]
  • 6.Hashi H, et al. Compartmentalization of Peyer's patch anlagen before lymphocyte entry. J. Immunol. 2001;166:3702–3709. doi: 10.4049/jimmunol.166.6.3702. [DOI] [PubMed] [Google Scholar]
  • 7.Hameleers DM, van der Ende M, Biewenga J, Sminia T. An immunohistochemical study on the postnatal development of rat nasal-associated lymphoid tissue (NALT) Cell Tissue Res. 1989;256:431–438. doi: 10.1007/BF00218901. [DOI] [PubMed] [Google Scholar]
  • 8.De Togni P, et al. Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science. 1994;264:703–707. doi: 10.1126/science.8171322. [DOI] [PubMed] [Google Scholar]
  • 9.Futterer A, Mink K, Luz A, Kosco-Vilbois MH, Pfeffer K. The lymphotoxin β receptor controls organogenesis and affinity maturation in peripheral lymphoid tissues. Immunity. 1998;9:59–70. doi: 10.1016/S1074-7613(00)80588-9. [DOI] [PubMed] [Google Scholar]
  • 10.Rennert PD, Browning JL, Mebius R, Mackay F, Hochman PS. Surface lymphotoxin α/β complex is required for the development of peripheral lymphoid organs. J. Exp. Med. 1996;184:1999–2006. doi: 10.1084/jem.184.5.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Koni PA, et al. Distinct roles in lymphoid organogenesis for lymphotoxins α and β revealed in lymphotoxin β-deficient mice. Immunity. 1997;6:491–500. doi: 10.1016/S1074-7613(00)80292-7. [DOI] [PubMed] [Google Scholar]
  • 12.Yoshida H, et al. IL-7 receptor α+ CD3− cells in the embryonic intestine induces the organizing center of Peyer's patches. Int. Immunol. 1999;11:643–655. doi: 10.1093/intimm/11.5.643. [DOI] [PubMed] [Google Scholar]
  • 13.Yamamoto M, et al. Alternate mucosal immune system: organized Peyer's patches are not required for IgA responses in the gastrointestinal tract. J. Immunol. 2000;164:5184–5191. doi: 10.4049/jimmunol.164.10.5184. [DOI] [PubMed] [Google Scholar]
  • 14.Mauri DN, et al. LIGHT, a new member of the TNF superfamily, and lymphotoxin α are ligands for herpesvirus entry mediator. Immunity. 1998;8:21–30. doi: 10.1016/S1074-7613(00)80455-0. [DOI] [PubMed] [Google Scholar]
  • 15.Scheu S, et al. Targeted disruption of LIGHT causes defects in costimulatory T cell activation and reveals cooperation with lymphotoxin β in mesenteric lymph node genesis. J. Exp. Med. 2002;195:1613–1624. doi: 10.1084/jem.20020215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fujihashi K, et al. Interleukin 2 (IL-2) and interleukin 7 (IL-7) reciprocally induce IL-7 and IL-2 receptors on γδ T-cell receptor-positive intraepithelial lymphocytes. Proc. Natl Acad. Sci. USA. 1996;93:3613–3618. doi: 10.1073/pnas.93.8.3613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Watanabe M, et al. Interleukin 7 is produced by human intestinal epithelial cells and regulates the proliferation of intestinal mucosal lymphocytes. J. Clin. Invest. 1995;95:2945–2953. doi: 10.1172/JCI118002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fujihashi K, McGhee JR, Yamamoto M, Peschon JJ, Kiyono H. An interleukin-7 internet for intestinal intraepithelial T cell development: knockout of ligand or receptor reveal differences in the immunodeficient state. Eur. J. Immunol. 1997;27:2133–2138. doi: 10.1002/eji.1830270903. [DOI] [PubMed] [Google Scholar]
  • 19.Adachi S, et al. Essential role of IL-7 receptor α in the formation of Peyer's patch anlage. Int. Immunol. 1998;10:1–6. doi: 10.1093/intimm/10.1.1. [DOI] [PubMed] [Google Scholar]
  • 20.Mebius RE, Rennert P, Weissman IL. Developing lymph nodes collect CD4+CD3−LTβ+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity. 1997;7:493–504. doi: 10.1016/S1074-7613(00)80371-4. [DOI] [PubMed] [Google Scholar]
  • 21.Finke D, Acha-Orbea H, Mattis A, Lipp M, Kraehenbuhl J. CD4+CD3− cells induce Peyer's patch development: role of α4β1 integrin activation by CXCR5. Immunity. 2002;17:363–373. doi: 10.1016/S1074-7613(02)00395-3. [DOI] [PubMed] [Google Scholar]
  • 22.Honda K, et al. Molecular basis for hematopoietic/mesenchymal interaction during initiation of Peyer's patch organogenesis. J. Exp. Med. 2001;193:621–630. doi: 10.1084/jem.193.5.621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Peschon JJ, et al. Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice. J. Exp. Med. 1994;180:1955–1960. doi: 10.1084/jem.180.5.1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Forster R, et al. A putative chemokine receptor, BLR1, directs B cell migration to defined lymphoid organs and specific anatomic compartments of the spleen. Cell. 1996;87:1037–1047. doi: 10.1016/S0092-8674(00)81798-5. [DOI] [PubMed] [Google Scholar]
  • 25.Shinkura R, et al. Alymphoplasia is caused by a point mutation in the mouse gene encoding NF-κB-inducing kinase. Nature Genet. 1999;22:74–77. doi: 10.1038/8780. [DOI] [PubMed] [Google Scholar]
  • 26.Nakano H, et al. TRAF5, an activator of NF-κB and putative signal transducer for the lymphotoxin-β receptor. J. Biol. Chem. 1996;271:14661–14664. doi: 10.1074/jbc.271.25.14661. [DOI] [PubMed] [Google Scholar]
  • 27.Miyawaki S, et al. A new mutation, aly, that induces a generalized lack of lymph nodes accompanied by immunodeficiency in mice. Eur. J. Immunol. 1994;24:429–434. doi: 10.1002/eji.1830240224. [DOI] [PubMed] [Google Scholar]
  • 28.Yin L, et al. Defective lymphotoxin-β receptor-induced NFκB transcriptional activity in NIK-deficient mice. Science. 2001;291:2162–2165. doi: 10.1126/science.1058453. [DOI] [PubMed] [Google Scholar]
  • 29.Sun Z, et al. Requirement for RORγ in thymocyte survival and lymphoid organ development. Science. 2000;288:2369–2373. doi: 10.1126/science.288.5475.2369. [DOI] [PubMed] [Google Scholar]
  • 30.Yokota Y, et al. Development of peripheral lymphoid organs and natural killer cells depends on the helix-loop-helix inhibitor Id2. Nature. 1999;397:702–706. doi: 10.1038/17812. [DOI] [PubMed] [Google Scholar]
  • 31.Harmsen A, et al. Organogenesis of nasal-associated lymphoid tissue (NALT) occurs independently of lymphotoxin-α (LTα) and retinoic acid receptor-related orphan receptor-γ, but the organization of NALT is LTα dependent. J. Immunol. 2002;168:986–990. doi: 10.4049/jimmunol.168.3.986. [DOI] [PubMed] [Google Scholar]
  • 32.Eberl G, et al. An essential function for the nuclear receptor RORγ in the generation of fetal lymphoid tissue inducer cells. Nature Immunol. 2004;5:64–73. doi: 10.1038/ni1022. [DOI] [PubMed] [Google Scholar]
  • 33.Kurebayashi S, et al. Retinoid-related orphan receptor γ (RORγ) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. Proc. Natl Acad. Sci. USA. 2000;97:10132–10137. doi: 10.1073/pnas.97.18.10132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kuper CF, et al. Lymphoid and non-lymphoid cells in nasal-associated lymphoid tissue (NALT) in the rat. An immuno- and enzyme-histochemical study. Cell Tissue Res. 1990;259:371–377. doi: 10.1007/BF00318460. [DOI] [PubMed] [Google Scholar]
  • 35.Kuper CF, et al. The role of nasopharyngeal lymphoid tissue. Immunol. Today. 1992;13:219–224. doi: 10.1016/0167-5699(92)90158-4. [DOI] [PubMed] [Google Scholar]
  • 36.Debertin AS, et al. Nasal-associated lymphoid tissue (NALT): frequency and localization in young children. Clin. Exp. Immunol. 2003;134:503–507. doi: 10.1111/j.1365-2249.2003.02311.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Spit BJ, et al. Nose-associated lymphoid tissue (NALT) in the rat. Ultramicroscopy. 1987;21:201–204. doi: 10.1016/0304-3991(87)90130-6. [DOI] [Google Scholar]
  • 38.Porgador A, Staats HF, Itoh Y, Kelsall BL. Intranasal immunization with cytotoxic T-lymphocyte epitope peptide and mucosal adjuvant cholera toxin: selective augmentation of peptide-presenting dendritic cells in nasal mucosa-associated lymphoid tissue. Infect. Immun. 1998;66:5876–5881. doi: 10.1128/iai.66.12.5876-5881.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zuercher AZ, Coffin SE, Thumheer MC, Fundora P, Cebra JJ. Nasal-associated lymphoid tissue is a mucosal inductive site for virus-specific humoral and cellular immune responses. J. Immunol. 2002;168:1796–1803. doi: 10.4049/jimmunol.168.4.1796. [DOI] [PubMed] [Google Scholar]
  • 40.Prakken BJ, et al. Peptide-induced nasal tolerance for a mycobacterial heat shock protein 60 T cell epitope in rats suppresses both adjuvant arthritis and nonmicrobially induced experimental arthritis. Proc. Natl Acad. Sci. USA. 1997;94:3284–3289. doi: 10.1073/pnas.94.7.3284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yanagita M, et al. Nasopharyngeal-associated lymphoreticular tissue (NALT) immunity: fimbriae-specific TH1 and TH2 cell-regulated IgA responses for the inhibition of bacterial attachment to epithelial cells and subsequent inflammatory cytokine production. J. Immunol. 1999;162:3559–3565. [PubMed] [Google Scholar]
  • 42.Hiroi T, et al. Nasal immune system: distinctive TH0 and TH1/TH2 type environments in murine nasal-associated lymphoid tissues and nasal passage, respectively. Eur. J. Immunol. 1998;28:3346–3353. doi: 10.1002/(SICI)1521-4141(199810)28:10<3346::AID-IMMU3346>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 43.Hiroi T, et al. HIV mucosal vaccine: nasal immunization with rBCG-V3J1 induces a long term V3J1 peptide-specific neutralizing immunity in TH1- and TH2-deficient conditions. J. Immunol. 2001;167:5862–5867. doi: 10.4049/jimmunol.167.10.5862. [DOI] [PubMed] [Google Scholar]
  • 44.Imaoka K, et al. Nasal immunization of nonhuman primates with simian immunodeficiency virus p55gag and cholera toxin adjuvant induces TH1/TH2 help for virus-specific immune responses in reproductive tissues. J. Immunol. 1998;161:5952–5958. [PubMed] [Google Scholar]
  • 45.Kurono Y, et al. Nasal immunization induces Haemophilus influenzae-specific TH1 and TH2 responses with mucosal IgA and systemic IgG antibodies for protective immunity. J. Infect. Dis. 1999;180:122–132. doi: 10.1086/314827. [DOI] [PubMed] [Google Scholar]
  • 46.Spalding DM, Griffin JA. Different pathways of differentiation of pre-B cell lines are induced by dendritic cells and T cells from different lymphoid tissues. Cell. 1986;44:507–515. doi: 10.1016/0092-8674(86)90472-1. [DOI] [PubMed] [Google Scholar]
  • 47.Iwasaki A, Kelsall BL. Freshly isolated Peyer's patch, but not spleen, dendritic cells produce interleukin 10 and induce the differentiation of T helper type 2 cells. J. Exp. Med. 1999;190:229–239. doi: 10.1084/jem.190.2.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Muramatsu M, et al. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell. 2000;102:553–563. doi: 10.1016/S0092-8674(00)00078-7. [DOI] [PubMed] [Google Scholar]
  • 49.Coffman RL, Lebman DA, Shrader B. Transforming growth factor β specifically enhances IgA production by lipopolysaccharide-stimulated murine B lymphocytes. J. Exp. Med. 1989;170:1039–1044. doi: 10.1084/jem.170.3.1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sonoda E, et al. Transforming growth factor β induces IgA production and acts additively with interleukin 5 for IgA production. J. Exp. Med. 1989;170:1415–1420. doi: 10.1084/jem.170.4.1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ehrhardt RO, Strober W, Harriman GR. Effect of transforming growth factor (TGF)-β1 on IgA isotype expression. TGF-β1 induces a small increase in sIgA+ B cells regardless of the method of B cell activation. J. Immunol. 1992;148:3830–3836. [PubMed] [Google Scholar]
  • 52.Bowman EP, et al. The intestinal chemokine thymus-expressed chemokine (CCL25) attracts IgA antibody-secreting cells. J. Exp. Med. 2002;195:269–275. doi: 10.1084/jem.20010670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Youngman KR, et al. Correlation of tissue distribution, developmental phenotype, and intestinal homing receptor expression of antigen-specific B cells during the murine anti-rotavirus immune response. J. Immunol. 2002;168:2173–2181. doi: 10.4049/jimmunol.168.5.2173. [DOI] [PubMed] [Google Scholar]
  • 54.Murray PD, McKenzie DT, Swain SL, Kagnoff MF. Interleukin 5 and interleukin 4 produced by Peyer's patch T cells selectively enhance immunoglobulin A expression. J. Immunol. 1987;139:2669–2674. [PubMed] [Google Scholar]
  • 55.Coffman RL, Shrader B, Carty J, Mosmann TR, Bond MW. A mouse T cell product that preferentially enhances IgA production. I. Biologic characterization. J. Immunol. 1987;139:3685–3690. [PubMed] [Google Scholar]
  • 56.Beagley KW, et al. Recombinant murine IL-5 induces high rate IgA synthesis in cycling IgA-positive Peyer's patch B cells. J. Immunol. 1988;141:2035–2042. [PubMed] [Google Scholar]
  • 57.Beagley KW, et al. Interleukins and IgA synthesis. Human and murine interleukin 6 induce high rate IgA secretion in IgA-committed B cells. J. Exp. Med. 1989;169:2133–2148. doi: 10.1084/jem.169.6.2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Defrance T, et al. Interleukin 10 and transforming growth factor β cooperate to induce anti-CD40-activated naive human B cells to secrete immunoglobulin A. J. Exp. Med. 1992;175:671–682. doi: 10.1084/jem.175.3.671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fagarasan S, Kinoshita K, Muramatsu M, Ikuta K, Honjo T. In situ class switching and differentiation to IgA-producing cells in the gut lamina propria. Nature. 2001;413:639–643. doi: 10.1038/35098100. [DOI] [PubMed] [Google Scholar]
  • 60.Kang H-S, et al. Signaling via LTβR on the lamina propria stromal cells of the gut is required for IgA production. Nature Immunol. 2002;3:576–582. doi: 10.1038/ni795. [DOI] [PubMed] [Google Scholar]
  • 61.Davis IA, Knight KA, Rouse BT. The spleen and organized lymph nodes are not essential for the development of gut-induced mucosal immune responses in lymphotoxin-α deficient mice. Clin. Immunol. Immunopathol. 1998;89:150–159. doi: 10.1006/clin.1998.4601. [DOI] [PubMed] [Google Scholar]
  • 62.Hamada H, et al. Identification of multiple isolated lymphoid follicles on the antimesenteric wall of the mouse small intestine. J. Immunol. 2002;168:57–64. doi: 10.4049/jimmunol.168.1.57. [DOI] [PubMed] [Google Scholar]
  • 63.Brandtzaeg P, Baekkevold ES, Morton HC. From B to A the mucosal way. Nature Immunol. 2001;2:1093–1094. doi: 10.1038/ni1201-1093. [DOI] [PubMed] [Google Scholar]
  • 64.Fagarasan S, Honjo T. Intestinal IgA synthesis: regulation of front-line body defences. Nature Rev. Immunol. 2003;3:63–72. doi: 10.1038/nri982. [DOI] [PubMed] [Google Scholar]
  • 65.Kroese FG, de Waard R, Bos NA. B-1 cells and their reactivity with the murine intestinal microflora. Semin. Immunol. 1996;8:11–18. doi: 10.1006/smim.1996.0003. [DOI] [PubMed] [Google Scholar]
  • 66.Hiroi T, et al. Deficiency of IL-5 receptor α-chain selectively influences the development of the common mucosal immune system independent IgA-producing B-1 cell in mucosa-associated tissues. J. Immunol. 1999;162:821–828. [PubMed] [Google Scholar]
  • 67.Jang MH, et al. Intestinal villous M cell: an antigen entry site in the mucosal epithelium. Proc. Natl Acad. Sci. USA. 2004;101:6110–6115. doi: 10.1073/pnas.0400969101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shikina T, et al. IgA class switch occurs in the organized nasopharynx- and gut-associated lymphoid tissue, but not in the diffuse lamina propria of airways and gut. J. Immunol. 2004;172:6259–6264. doi: 10.4049/jimmunol.172.10.6259. [DOI] [PubMed] [Google Scholar]
  • 69.Iwasato T, Shimizu A, Honjo T, Yamagishi H. Circular DNA is excised by immunoglobulin class switch recombination. Cell. 1990;62:143–149. doi: 10.1016/0092-8674(90)90248-D. [DOI] [PubMed] [Google Scholar]
  • 70.Shimoda M, et al. Isotype-specific selection of high affinity memory B cells in nasal-associated lymphoid tissue. J. Exp. Med. 2001;194:1597–1607. doi: 10.1084/jem.194.11.1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lazarus NH, et al. A common mucosal chemokine (mucosae-associated epithelial chemokine/CCL28) selectively attracts IgA plasmablasts. J. Immunol. 2003;170:3799–3805. doi: 10.4049/jimmunol.170.7.3799. [DOI] [PubMed] [Google Scholar]
  • 72.Kunkel EJ, et al. CCR10 expression is a common feature of circulating and mucosal epithelial tissue IgA Ab-secreting cells. J. Clin. Invest. 2003;111:1001–1010. doi: 10.1172/JCI17244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kunkel EJ, Butcher EC. Plasma-cell homing. Nature Rev. Immunol. 2003;3:822–829. doi: 10.1038/nri1203. [DOI] [PubMed] [Google Scholar]
  • 74.Yonemitsu Y, et al. Efficient gene transfer to airway epithelium using recombinant Sendai virus. Nature Biotechnol. 2000;18:970–973. doi: 10.1038/79463. [DOI] [PubMed] [Google Scholar]
  • 75.Kunisawa J, et al. Sendai virus fusion protein mediates simultaneous induction of MHC class I/II-dependent mucosal and systemic immune responses via the nasopharyngeal-associated lymphoreticular tissue immune system. J. Immunol. 2001;167:1406–1412. doi: 10.4049/jimmunol.167.3.1406. [DOI] [PubMed] [Google Scholar]
  • 76.Sakaue G, et al. HIV mucosal vaccine: nasal immunization with gp160-encapsulated hemagglutinating virus of Japan-liposome induces antigen-specific CTLs and neutralizing antibody responses. J. Immunol. 2003;170:495–502. doi: 10.4049/jimmunol.170.1.495. [DOI] [PubMed] [Google Scholar]
  • 77.Park HS, Francis KP, Yu J, Cleary PP. Membranous cells in nasal-associated lymphoid tissue: a portal of entry for the respiratory mucosal pathogen group A Streptococcus. J. Immunol. 2003;171:2532–2537. doi: 10.4049/jimmunol.171.5.2532. [DOI] [PubMed] [Google Scholar]
  • 78.Wolf JL, et al. Intestinal M cells: a pathway for entry of reovirus into the host. Science. 1981;212:471–472. doi: 10.1126/science.6259737. [DOI] [PubMed] [Google Scholar]
  • 79.Lee PW, Hayes EC, Joklik WK. Protein σ1 is the reovirus cell attachment protein. Virology. 1981;108:156–163. doi: 10.1016/0042-6822(81)90535-3. [DOI] [PubMed] [Google Scholar]
  • 80.Wu Y, et al. M cell-targeted DNA vaccination. Proc. Natl Acad. Sci. USA. 2001;98:9318–9323. doi: 10.1073/pnas.161204098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang X, Hone DM, Haddad A, Shata MT, Pascual DW. M cell DNA vaccination for CTL immunity to HIV. J. Immunol. 2003;171:4717–4725. doi: 10.4049/jimmunol.171.9.4717. [DOI] [PubMed] [Google Scholar]
  • 82.Honda M, et al. Protective immune responses induced by secretion of a chimeric soluble protein from a recombinant Mycobacterium bovis bacillus Calmette-Guérin vector candidate vaccine for human immunodeficiency virus type 1 in small animals. Proc. Natl Acad. Sci. USA. 1995;92:10693–10697. doi: 10.1073/pnas.92.23.10693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Aldovini A, Young RA. Humoral and cell-mediated immune responses to live recombinant BCG–HIV vaccines. Nature. 1991;351:479–482. doi: 10.1038/351479a0. [DOI] [PubMed] [Google Scholar]
  • 84.Yamamoto M, McGhee JR, Hagiwara Y, Otake S, Kiyono H. Genetically manipulated bacterial toxin as a new generation mucosal adjuvant. Scand. J. Immunol. 2001;53:211–217. doi: 10.1046/j.1365-3083.2001.00883.x. [DOI] [PubMed] [Google Scholar]
  • 85.de Haan L, et al. Mutants of the Escherichia coli heat-labile enterotoxin with reduced ADP-ribosylation activity or no activity retain the immunogenic properties of the native holotoxin. Infect. Immun. 1996;64:5413–5416. doi: 10.1128/iai.64.12.5413-5416.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Douce G, et al. Mutants of Escherichia coli heat-labile toxin lacking ADP-ribosyltransferase activity act as nontoxic, mucosal adjuvants. Proc. Natl Acad. Sci. USA. 1995;92:1644–1648. doi: 10.1073/pnas.92.5.1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fontana MR, et al. Construction of nontoxic derivatives of cholera toxin and characterization of the immunological response against the A subunit. Infect. Immun. 1995;63:2356–2360. doi: 10.1128/iai.63.6.2356-2360.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Freytag LC, Clements JD. Bacterial toxins as mucosal adjuvants. Curr. Top. Microbiol. Immunol. 1999;236:215–236. doi: 10.1007/978-3-642-59951-4_11. [DOI] [PubMed] [Google Scholar]
  • 89.Yamamoto S, et al. Mutants in the ADP-ribosyltransferase cleft of cholera toxin lack diarrheagenicity but retain adjuvanticity. J. Exp. Med. 1997;185:1203–1210. doi: 10.1084/jem.185.7.1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yamamoto M, et al. A nontoxic adjuvant for mucosal immunity to pneumococcal surface protein A. J. Immunol. 1998;161:4115–4121. [PubMed] [Google Scholar]
  • 91.Yamamoto S, et al. A nontoxic mutant of cholera toxin elicits TH2-type responses for enhanced mucosal immunity. Proc. Natl Acad. Sci. USA. 1997;94:5267–5272. doi: 10.1073/pnas.94.10.5267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hagiwara Y, et al. Mutants of cholera toxin as an effective and safe adjuvant for nasal influenza vaccine. Vaccine. 1999;17:2918–2926. doi: 10.1016/S0264-410X(99)00135-8. [DOI] [PubMed] [Google Scholar]
  • 93.Kweon MN, et al. A nontoxic chimeric enterotoxin adjuvant induces protective immunity in both mucosal and systemic compartments with reduced IgE antibodies. J. Infect. Dis. 2002;186:1261–1269. doi: 10.1086/344526. [DOI] [PubMed] [Google Scholar]
  • 94.Nakamura K, Okuno Y. Vaccination with live attenuated influenza virus at the Osaka university hospital in 1960. Virus. 1961;11:349–354. doi: 10.2222/jsv.11.349. [DOI] [PubMed] [Google Scholar]
  • 95.Okuno Y, Nakamura K. Prophylactic effectiveness of live influenza vaccine in 1965. Biken J. 1966;9:89–95. [PubMed] [Google Scholar]
  • 96.Mutsch M, et al. Use of the inactivated intranasal influenza vaccine and the risk of Bell's palsy in Switzerland. N. Engl. J. Med. 2004;350:896–903. doi: 10.1056/NEJMoa030595. [DOI] [PubMed] [Google Scholar]
  • 97.Bourguignon P, et al. Molecular Approaches to Vaccine Design 23. 1999. [Google Scholar]
  • 98.Gluck U, Gebbers JO, Gluck R. Phase 1 evaluation of intranasal virosomal influenza vaccine with and without Escherichia coli heat-labile toxin in adult volunteers. J. Virol. 1999;73:7780–7786. doi: 10.1128/jvi.73.9.7780-7786.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.King JC, et al. Safety and immunogenicity of low and high doses of trivalent live cold-adapted influenza vaccine administered intranasally as drops or spray to healthy children. J. Infect. Dis. 1998;177:1394–1397. doi: 10.1086/517822. [DOI] [PubMed] [Google Scholar]
  • 100.Harper SA, Fukuda K, Cox NJ, Bridges CB. Using live, attenuated influenza vaccine for prevention and control of influenza: supplemental recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm. Rep. 2003;52(Suppl RR-13):1–8. [PubMed] [Google Scholar]
  • 101.Maassab HF. Adaptation and growth characteristics of influenza virus at 25°C. Nature. 1967;213:612–614. doi: 10.1038/213612a0. [DOI] [PubMed] [Google Scholar]
  • 102.Bukreyev A, et al. Mucosal immunisation of African green monkeys (Cercopithecus aethiops) with an attenuated parainfluenza virus expressing the SARS coronavirus spike protein for the prevention of SARS. Lancet. 2004;363:2122–2127. doi: 10.1016/S0140-6736(04)16501-X. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Nature Reviews. Immunology are provided here courtesy of Nature Publishing Group

RESOURCES