Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Microbes and Infection, 3, 2001, 1021−1035

© 2001 Éditions scientifiques et médicales Elsevier SAS. All rights reserved


S1286457901014605/REV

Review

IgA responses in the intestinal mucosa against


pathogenic and non-pathogenic microorganisms
Andrew J. Macpherson*, Lukas Hunziker, Kathy McCoy, Alain Lamarre
Institute of Experimental Immunology, Universitätsspital, Schmelzbergstrasse 12, CH8091, Zürich, Switzerland

ABSTRACT – IgA is the most abundant immunoglobulin produced in mammals; most is secreted
as a dimer across mucous membranes. This review discusses the different mechanisms of induction of
IgA, and its role in protecting mucosal surfaces against pathogenic and non-pathogenic
microorganisms. © 2001 Éditions scientifiques et médicales Elsevier SAS
IgA / bacteria / viruses / intestinal mucosa

1. Introduction intestinal epithelial cells [2] and transferred to the intesti-


nal lumen (figure 2). The translocation mechanism depends
The intestinal immune system has the delicate task of on an 80 000 Mr transmembrane glycoprotein expressed
tolerating colonisation by a prodigious commensal flora, by intestinal epithelial cells termed the polymeric immu-
whilst remaining able to mount a rapid and specific noglobulin receptor (pIgR [3]). Dimeric IgA is bound to the
response to invading pathogens. In mammals, commensal receptor at the basolateral surface of enterocytes by cova-
bacteria colonise the intestinal tract rapidly after birth lent linkage between this protein and the CH2 domain of
(although the diversity and density of bacterial species one of the alpha heavy chains. Subsequently the entire
involved is reduced in the neonate during the period of complex is endocytosed into a vesicle with the IgA
exclusive lactation [1]). Commensal bacteria are an advan- attached. The vesicle is then delivered to the apical (lumi-
tage to the host animal since they allow energy from nal) surface, whereupon proteolysis cuts the receptor and
otherwise indigestible carbohydrates to be salvaged, and releases the N-terminal part of the receptor protein (secre-
the biofilms of relatively innocent microorganisms in the tory component), free or attached to IgA (reviewed in [4]).
intestinal mucus limit access to pathogens. However, This mechanism functions across the epithelial layer
despite the lack of toxins and epithelial membrane attach- throughout the small and large intestines (and other
ment devices in commensal organisms, they possess a mucous membranes), and in man approximately 3 g of IgA
large array of immunostimulatory compounds which must is delivered each day into the intestinal lumen [5].
be largely confined to the lumen of the intestine if intesti-
This transport system is more than just a means of
nal and systemic immunopathology is not to ensue. This
delivery of dimeric IgA from the basolateral surface of
review will focus on the role of secreted IgA in responses
epithelia into the lumen. First, it is able to transport IgA in
to non-pathogenic intestinal bacteria and to intestinal
an immune complex with antigen so that antigens that
pathogenic microorganisms.
leak through the epithelial barrier can be cleared back into
1.1. Secretion of IgA across intestinal mucous membranes the lumen, and monomeric IgA or IgG antibodies, which
are not themselves substrates for the pIgR, can be trans-
IgA is the most abundantly produced immunoglobulin
ported as part of these immune complexes [6]. Secondly, it
in mammals; it constitutes a small component of serum
has been shown that specific IgA can even neutralise viral
immunoglobulin, and most IgA is secreted across mucous
replication within the epithelial cells themselves during
membranes of the intestinal, respiratory, biliary and geni-
the transit process. The evidence for this comes from
tal tracts. In the intestine, the secretory form of IgA is
experiments with polarised MDCK cell monolayers in
synthesised by plasma cells in the lamina propria (figure
culture which had been stably transfected to express the
1): this is a dimer of two IgA molecules covalently linked
pIgR [7]. These were infected with Sendai virus on the
together through a J chain molecule between the alpha
luminal surface, and anti-Sendai haemagglutinin-
heavy chains. The complex is then translocated through
neuraminidase monoclonal IgA, or irrelevant IgA was
applied to the basolateral surface (figure 3). After 4 h both
*Correspondence and reprints. surfaces were treated with trypsin to remove adherent
E-mail address: amacpher@pathol.unizh.ch (A.J. Macpherson). antibody or virus; the incubation was then continued for a

Microbes and Infection 1021


2001, 1021-0
Review Macpherson et al.

Figure 1. Recirculation of B cells through the


lymph and the blood following triggering in
mucosal inductive sites. B1 cells from the perito-
neum are also triggered to produce IgA (indepen-
dently of help from T cells: see Section 5.3),
although whether they participate in a similar
recirculation process is unclear. The lower small
intestine and the colon contain an enormous load
of commensal bacteria which are normally not
pathogenic; this comprises about 1 000 species
with up to 1012 organisms/g of intestinal contents.

Figure 2. Transport of dimeric IgA


through the epithelial cell layer into the
intestinal lumen. In this process IgA is
covalently linked to the polymeric immu-
noglobulin receptor within the vesicular
membrane as the vesicles cross the cell. At
the apical surface IgA is released by pro-
teolysis of the receptor, part of which (secre-
tory component) remains attached to the
free immunoglobulin.

1022 Microbes and Infection


2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

production and transepithelial transport. The first is through


secretion into bile, which is then passed into the intestine
through the ampulla of Vater in the upper small intestine
(duodenum). To reach the bile, polymeric IgA is either
transported across hepatocytes [11, 12] (which in rodents
express the pIgR [13–15]) into biliary canaliculi, or in
humans (whose hepatocytes lack pIgR [16, 17]) transport
is across the biliary epithelia of the bile ducts and gall
bladder [18–21]. Another way in which IgA is delivered
into the intestine is through secretion of IgA into the
maternal milk, which is then fed to the infant. IgA is the
predominant immunoglobulin in milk [22]. Early in lacta-
tion most of the IgA is polymeric IgA transported from the
serum across the mammary epithelia, later local IgA-
producing plasma cells (including those induced in intes-
tinal lymphoid tissue) are found in the mammary gland
[23–25]. The possible relevance of milk IgA to the neonate
is discussed further in Section 5.2.

1.4. Serum IgA

IgA is also a component of serum, although here it is


quantitatively less than IgM or IgG and its constituent
isotypes. In humans there are two α heavy chains, giving
rise to different IgA1 and IgA2 isoforms. (Multiple α chain
isotypes are also seen in non-human primates and in
Figure 3. Set-up of the experiment by Kaetzel et al. [7], showing rabbits; other mammals including mice have a single α
that there is intracellular neutralisation of virus by specific IgA gene.) Of these, IgA1 is almost exclusively present as a
during transport through epithelial cells. Specific IgA applied to monomeric form in human serum. The in-vivo function of
the basolateral surface of a polarised epithelial cell layer is able to serum IgA remains very poorly understood.
prevent intracellular viral replication of Sendai virus infecting
from the apical surface. Neither non-specific IgA (which is trans- In humans there is a Fcα receptor (CD89 or FcαRI) on
ported, but does not bind virus) nor specific IgG (which is not the surface of eosinophils, neutrophils, monocytes and
transported) can do this. An in vivo version of this experiment, macrophages [26, 27] which can trigger responses mea-
carried out by Burns et al. [95] is described in Section 4.1. surable in vitro, including phagocytosis [26], antibody-
dependent cellular cytotoxicity [28] and secretion of
inflammatory mediators [29]. These responses seem curi-
further 20 h, when the apical supernatant was removed ous compared with the usual view of IgA as a relatively
and the cells were lysed. The titres of virus were found to non-inflammatory immunoglobulin, yet their significance
be lower both in the apical supernatants and lysates of in vivo is hard to define because no mouse CD89 homo-
monolayers treated with specific IgA, compared with those logue has yet been described. However, a transgenic
treated with non-specific IgA or specific IgG (which is not mouse in which human CD89 (together with its own
transported through the cells because it does not bind to promoter and regulatory elements) has been generated in
the pIgR). The finding of less virus within the cells (i.e. the which FcαRI expression is on the same cell types found
lysates) indicated that specific IgA had interacted with naturally in humans, and in vitro responses (phagocytosis
viral proteins during transit through the cells and inhibited etc.) are similar. When these animals are not manipulated,
replication of the virus. expression of the FcαRI is restricted to cells of the myeloid
lineage in the bone marrow and periphery. However, in
1.2. Predominant importance of IgA the presence of inflammatory mediators (such as experi-
as a secretory immunoglobulin mental treatment with granulocyte colony-stimulating fac-
IgA is overwhelmingly the most important immunoglo- tor) FcαRI was expressed on Kupffer cells in the liver [30].
bulin in the intestine and at other mucosal surfaces, and These cells were then able to phagocytose bacteria coated
relatively little (< 10%) of the other isotypes (IgM, IgG or with serum (but not secretory) IgA. This implies that mono-
IgE) are locally produced or secreted unless: i) the tissues meric serum IgA may form a second-line defence system
are inflamed or diseased [8]; or ii) there is a deficiency of to clear bacteria that escape intestinal mucosal barriers,
IgA (found in about 1:500 Caucasian human subjects), in which works by coating these bacteria in the portal circu-
which case mucosal IgM production and secretion partly lation, allowing hepatocyte phagocytosis to be triggered.
compensates for the lack of IgA [9, 10]. Whilst this work is important as a first glimpse at the
possible purpose of having IgA in the serum, rather than
1.3. IgA in bile and milk just being secreted at mucosal surfaces, whether such a
There are two other important ways in which IgA may pathway is functional in normal humans or non-transgenic
be delivered into the intestine other than by local mucosal animals has yet to be shown.

Microbes and Infection 1023


2001, 1021-0
Review Macpherson et al.

2. The immunologic anatomy


of IgA production
2.1. IgA+ B-cell triggering in Peyer’s patches
and recirculation through the lymph and the blood
Early experiments showed that if radiolabelled large
lymphocytes from the thoracic duct lymph or mesenteric
lymph nodes of adult donors were injected intravenously
they would preferentially migrate into the intestine of adult
recipients or neonatal animals [31]. Migration could even
occur into foetal intestine grafted under the kidney cap-
sule of adult mice, showing that antigen is not required for
the homing mechanism [32]. This recirculation mecha-
nism applies to both B- and T-cell blasts, since both appear
in the recipient mucosal tissues [33]. Transfer of Peyer’s
patch cells into irradiated recipient animals showed that
these were a primary source of IgA+ B cells [34]. The
recirculation required for the induction of IgA was inves-
tigated in the classical isolated loop experiments of Gow-
ans and his colleagues. These established that IgA is
induced in intestinal Peyer’s patches and that after trigger-
ing, B cells pass through the mesenteric lymph nodes and
circulate via the lymphatic system to enter the subclavian
vein from the thoracic duct, then they home back to a
mucosal site (e.g., the intestinal lamina propria) through
the arterial blood. The key evidence for this was obtained
by constructing intestinal (Thiry Vella) loops in rats, which
retained a lymphatic and blood supply, but were discon-
Figure 4. Set-up of the experiment by Husband and Gowans
nected from the intestinal stream, with both ends having
[35] in which isolated (‘Thiry-Vella’) loops of small intestine
an external opening onto the skin (figure 4). When two
were constructed in rats. From the position marked ‘xxxx’ a
separate loops were constructed and antigen was intro-
segment of small intestine had been isolated, preserving its
duced into one of them, secretory IgA was subsequently
vascular and lymphatic supply, and both ends were connected to
observed both in the immunised loop and non-immunised
the skin of the animal. The ends of the remaining bowel were
loop, although the specific response was always greater in
anastomosed (at ‘xxxx’) to preserve intestinal continuity. The
the immunised loop [35]. Work with the powerful mucosal
effects of local immunisation into the intestine could now be
immunogen cholera toxin allowed plasmablasts to be
compared with the effects at distant sites by administering immu-
tracked as they passed through the thoracic lymph, and
nogen (cholera toxin) either into the loop, or into the main
later appeared in the intestinal lamina propria [36]. Experi-
intestine. In some animals two loops were constructed. This led
ments in rats where either Peyer’s patches or the mesen-
to the finding that local immunisation has an effect at distant
teric lymph nodes had been excised showed that Peyer’s
intestinal sites, which is an important part of the evidence that
patches are principally required for this process [35].
IgA is induced in lymphoid follicles (Peyer’s patches) in the
The existence of specialised mucosal inductive sites
intestine, and then cells recirculate through the lymph and blood
and their separation by the recirculation process from the
to diffusely populate the intestinal mucosa (and other mucosal
remainder of the mucosa was thus established. This also
tissues). See also figure 1.
provides the mucosal immune system with the ability to
induce responses at sites that are distant from the imme-
diate inductive environment of the Peyer’s patch, or even
in different mucosal tissues (leading to the concept of lymph nodes selectively home to the mammary glands
generalised functioning of mucosal tissues with some cross during lactation [24]. This appears to tailor the protection
talk between them). However, the local response of delivered to the neonatal intestine to the current antigens
mucosal immunisation is consistently greater than the sensed in the maternal intestinal immune system (see also
diffuse response [35], either as a result of some direct Section 5.2).
migration or of selective homing to the area of the intestine
2.2. The B1 and B2 lymphocytes and their origins
where induction first occurred.
The composition of milk IgA is probably an important There is a further issue concerning the immunological
example of how systemic recirculation is important fol- anatomy of mucosal antibody production, which is the
lowing B-cell induction in the intestine. Certainly milk IgA site of origin of the B cells that are induced to switch to IgA
limits the exposure of the neonate to the intestinal bacte- production. The earliest site of B-cell production is the
rial flora (which is rapidly acquired after birth) [37]. It has foetal liver, but after birth, B cells are produced both in the
been shown that milk contains IgA against antigens of the bone marrow and the pleuropericardial cavities [38]. It is
intestinal flora, and that IgA+ B cells from the mesenteric extremely controversial whether these two physical sites
1024 Microbes and Infection
2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

represent different lineages of B cells [39], although the


progeny of bone marrow and pleuroperitoneal percursors
certainly have different levels of surface marker expres-
sion, thus the pleuropericardial cells (called B1) stain
strongly with antibodies against surface IgM, Mac-1 and
CD5, and weakly with antibodies against B220 (CD45R)
and IgD, whereas in bone marrow-derived (B2) B cells the
situation is reversed (strong B1 markers stain weakly, and
vice versa) [40–42].
Both B1 and B2 lineages contribute to IgA production in
mice, and the evidence for this is discussed below. B1 cells
also give rise to IgM that is relatively undiversified (lacking
the molecular hallmarks of having been through a germi-
nal centre reaction; mutations in the complementarity-
determining regions of the antibody variable chains [43,
44]), and the resultant IgM has specificites against micro-
bial antigens. This IgM has been considered ‘natural’
antibody, because it is produced in germ-free conditions
when the cognate antigen(s) are presumed to be absent
[45], however, there is evidence that environmental [46]
or endogenous [47] antigens are required to positively
select or expand particular B1 cell specificities. The rel-
evant questions for intestinal IgA are: 1) how much secre-
tory IgA can also be considered B1-derived antibody,
produced without germinal centre-dependent pathways Figure 5. Set-up of radiation chimaera experiment [52]. The
of IgA induction; and 2) what is its functional importance? two classes of B lymphocytes (B1 cells from the peritoneum and
B2 cells from the bone marrow) are taken from different strains of
No significant population of IgA-staining B1 cells is
inbred mice in which the IgA molecules have (slight) ‘allotypic’
found in preparations isolated from the peritoneal cavity
differences that can be detected in immunological (ELISA and
(these are IgM+ cells which have yet to switch to IgA
ELISPOT) assays. In adult mice B1 cells are absent from the bone
production), and we have very sketchy knowledge about
marrow. A mouse that has had its own lymphocytes destroyed by
where these cells are triggered. B1 cells with the charac-
irradiation is reconstituted with the two different sorts of B
teristic surface markers are not readily detected in Peyer’s
lymphocytes. Once the situation in the recipient mouse has
patches, although they can be seen in animals with a
stabilised (after 3–6 months), one can determine the relative
targeted B-cell deficiency of the transforming growth fac-
proportions of IgA that are derived from the bone marrow (B2) or
tor (TGF-β) receptor (TβRIIBcell–/–) [48]. How then do we
peritoneal (B1) subsets by assaying for the different allotypes.
know that B1 cells can end up as intestinal IgA-producing
Normally about half IgA is B1 derived. From the results of this
lamina propria cells? The answer lies in making chimaeras
experiment, where the donor and recipient animals lack T cells
from lethally irradiated animals so their own lymphocyte
(TCRβ–/– δ–/–) and only FACS purified B cells are given from the
populations are destroyed, and afterwards they are recon-
peritoneum, one can show that most of the T-independent IgA is
stituted with bone marrow cells from an animal that will
B1 derived.
produce immunoglobulins of one allotype, and with peri-
toneal cells that will produce immunoglobulins of a differ-
ent allotype (see example in figure 5) [49]. The reconsti-
tuted animal is now left to recover from the effects of B cells bearing the CD5 surface marker also constitute
manipulation, and after about 3–4 months, the allotypic 10–25% of circulating and splenic B lymphocytes in adult
markers for the different sorts of immunoglobulin can be humans, and form the majority of B cells in human foetal
analysed in the serum and the intestinal (or other) secre- spleen and in umbilical cord blood [53, 54]. They are also
tions. In wild-type mice this shows that much of the serum increased in patients with rheumatoid arthritis, in whom
IgM and about half the intestinal IgA is derived from B1 they are a source of low-affinity polyreactive antibodies
cells [50]. The functional details of the B1 contribution to [55]. Thus there is also a B1 phenotype in humans. The
IgA have been explored in MHC class II-deficient mice extent of the B1 contribution to human intestinal IgA can
that also have the xid mutation [51]. Whereas class II–/– obviously not be determined by radiation chimaera tech-
mice have a relatively normal IgA content (despite a niques. An indirect approach has therefore been taken. It
deficiency of CD4+ T cells), the presence of the xid lesion is clear that most B1-derived immunoglobulin in mice is in
abolishes peritoneal B1 cells and the intestinal IgA content an unmutated germline configuration of the constituent V,
becomes sharply reduced. We have studied mice that are (D), J and C segments [43, 44], whereas B2-derived immu-
T-cell-deficient because of targeted lesions in both the noglobulin following immunisation is characterised by
beta and delta T-cell receptor genes: radiation chimaeras multiple somatic mutations focused on the complementa-
made in these animals show directly that the remaining rity determining regions that encode the binding site of the
IgA produced in the intestine is of B1 origin [52]. variable chains. Therefore immunoglobulin variable genes

Microbes and Infection 1025


2001, 1021-0
Review Macpherson et al.

from human CD5+ cells have been examined to determine isolated from intestinal lymphoid follicles or other lym-
whether they have somatic mutations or remain in germ- phoid tissues have been cultured in vitro to determine
line configuration to see if they have been through an whether the origins of accessory cells have a critical
antigen-driven process. Problems with this approach are: bearing on IgA induction. This has shown that the natural
i) that comparisons are usually made with database V, D, source of Peyer’s patch B [66], T and dendritic cells [67] is
(J), C sequences from different individuals, and polymor- more efficient for IgA production than when these cells are
phisms may be incorrectly assigned as mutations; and ii) derived from a non-mucosal site (such as the spleen). The
that analysis of cDNA clones even in sorted populations of relative importance of having just one of these compo-
B1 cells may be biased by higher mRNA expression levels nents (B, T or dendritic cells) from the Peyer’s patches has
in the contaminating B2 cells (e.g., reference [56] gives also been addressed (i.e. Peyer’s patch T cells cultured
examples of these problems). with splenic B and dendritic cells or other permutations):
Sequencing of rearranged IgA immunoglobulin V the interpretation of the results of different experiments is
regions has been carried out from intestinal biopsies to complicated by technical considerations of cell purifica-
look at mutational frequencies using both a reverse tran- tion, but B cells probably become committed to IgA switch-
scription PCR and rearranged DNA approach. In adults ing within the Peyer’s patches as a result of the unique
there certainly seems to be a high mutational frequency cytokine environment (especially TGF-β) generated by the
[56, 57], which is less in children [58], although insuffi- T, antigen-presenting and stromal cells there [66].
cient material from young children has been studied at the
single cell level. Whilst it is possible that B1 lymphocytes 3.3. Requirements for IgA induction in vivo
make a similar contribution to intestinal IgA in both To understand the functional requirements for different
humans and mice, this has not been resolved. cell types, cytokines and accessory molecules in vivo,
gene-targeted mice have been studied. To measure spe-
cific IgA induction in these strains, extensive use has been
3. Mechanisms of IgA induction made of ’oral adjuvants’, of which the most popular has
in model systems been cholera toxin. When given into the stomach of mice
in three successive doses (of 15 µg) at 10-day intervals,
Early experiments with nude mice (which have a very cholera toxin does not cause diarrhoea, but both intestinal
restricted population of T cells) showed that much of the and serum IgA are induced against cholera toxin itself
IgA production was dependent on T-cell help [59, 60]. As [68]. It is termed an oral adjuvant because when a soluble
shown below for both intestinal pathogens and commen- protein is given orally with the cholera toxin, a similar
sal bacteria, this is only part of the story, and there is also response is mounted against this protein, whereas nor-
an important T-independent pathway of IgA induction. mally giving a soluble protein alone into the intestine
There are four different experimental approaches, dis- would not immunise but rather induce tolerance to subse-
cussed in the following sections, that have been taken to quent systemic immunisation protocols [69].
study the mechanisms of IgA induction further. It is important to realise that this method of IgA induc-
tion may be rather a special case, because serum IgG and
3.1. IgA switching and secretion in cell culture IgE are also induced [68, 70]. Nonetheless, the model has
The cytokine requirements and the details of the been a powerful tool to demonstrate that a memory IgA
IgM→IgA switching mechanism have been addressed by response can be seen in mice 2 years after the initial
experiments on B-cell cultures in vitro. These have used B immunisation with cholera toxin [71], and it has been a
cells purified ex-vivo free from other cell types (derived good system to look at the requirements for induction of
either from the spleen or Peyer’s patches), or B-cell lines, IgA in vivo in different gene-targeted mice. It is also
which have been cultured in media supplemented with possible to study functional protection from cholera toxin
lipopolysaccharide as a non-specific stimulant and vari- in experiments in which a segment of mid small intestine is
ous combinations of cytokines. The object has been to isolated in anaesthetised mice by ligatures tied around the
look at the proportion of B cells that switch to IgA produc- bowel about 6–8 cm apart [72]. If cholera toxin is then
tion. This approach showed that TGF-β and interleukin injected into the lumen of this isolated segment, after 4 h
(IL)-4 promoted the switch from IgM→IgA surface expres- the fluid accumulation/cm length of the segment can be
sion and subsequent IgA secretion in splenocytes that measured. The fluid accumulation response is consider-
were depleted of T cells [61, 62]. The effect was increased ably reduced in animals where specific secretory IgA has
in the presence of IL-2, but IL-2 had little effect on its own been induced previously by immunisation with cholera
[61]. Once the switch IgM→IgA has taken place, IL-5 [62, toxin, but if IgA secretion is defective (for example in
63] and IL-6 [63, 64] then act to enhance secretion of IgA. J-chain-deficient mice) protection is impaired [73].
IL-10 also synergises with TGF-β to increase the efficiency Oral immunisation with cholera toxin is CD4+ T-cell
of IgA switching [65]. dependent in vivo (since it is defective in CD4–/– mice [72]
and MHC class II-deficient mice [51]). There is also a
3.2. Cells required for the IgM→IgA switch process requirement for IL-4, as responses to cholera toxin are also
Experiments have also been carried out to attempt to reduced in IL-4–/– mice; interestingly, where cholera toxin
reconstitute the different cell types that are required (in is used as an adjuvant in IL-4–/– mice for oral immunisation
addition to B cells) to allow the IgA switch to take place. with a soluble protein, the response to the soluble protein
For this, combinations of B, T and antigen-presenting cells itself is completely absent [74]. Immunisation is also inef
1026 Microbes and Infection
2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

fective in CTLA4-Hγ1 transgenic mice: these express a cination [84] and it depends on mucosal exposure to
CTLA4 protein construct under the control of the immu- vaccine [85]. A requirement for mucosal vaccination for
noglobulin heavy chain promoter which blocks the CD28- secretory immunity was also found for rubella [86].
CD80/86 costimulation signals between antigen-
presenting cells and T cells [75]. Despite this, 4.1. Rotaviruses
paradoxically, the intestinal IgA content in both CD4–/– Rotaviruses have been studied extensively in relation to
and CTLA4-Hγ1 mice is normal, indicating that induction the functional importance of the mucosal IgA response
of IgA (by other antigens) may be independent of these (reviewed in [87]). These cause the death of 800 000
T-B-cell interactions. infants per year worldwide, and the impetus to develop a
The in vivo importance of TGF-β, IL-2 and IL-10 have vaccine led to development of animal models. Rotavirus
been more difficult to assess, because mice with targeted infections in calves and piglets were used as models of
deletions at these loci have generalised (TGF-β [76]) or human disease, but had the disadvantage that germ-free
intestinal (IL-2, IL-10) chronic inflammation [77, 78], conditions were often required. Rotavirus was also shown
which interferes with assessment of unmanipulated IgA to cause epidemic diarrhoea in young mice (postnatal
levels and the results of immunisation protocols. How- days 4–14). Certain strains are also capable of infecting
ever, an inducible targeted deletion of TGF-β receptor II adult mice; although they do not get diarrhoea, there is
confined to B cells has recently been reported, these mice subclinical fluid malabsorption at the time of peak viral
do not have generalised systemic or intestinal inflamma- load [88]. The mouse rotavirus model has thus allowed the
tion, but there are low IgA levels combined with a consid- immune mechanisms resulting in viral clearance to be
erably expanded population of peritoneal B1 cells and explored in detail.
hypertrophied Peyer’s patches [48]. Whether the intestinal Clearance of virus after primary infection, and subse-
secretory IgA is also reduced by this selective induced quent protection against reinfection (seen for at least
lesion in TGF-β signalling (as found in TGF-β–/– mice [79]) 1 year) correlates well with the production of mucosal and
has not yet been reported; if so the activation of Peyer’s serum IgA [88, 89]. BALB/c or C57BL/6x129 strains of
patches and the peritoneal B1 compartment may represent severe combined immunodeficiency (SCID) or RAG–/–
compensatory changes as a result of increased exposure to mice (both of which lack both B and T cells) become
antigens at the mucosal surfaces in the absence of secre- persistently infected, so adaptive immunity induced fol-
tory IgA function. lowing infection is clearly important for viral clearance
[90, 91]. However, innate mechanisms also play a role, as
3.4. Mechanisms of in-vivo induction of IgA
40% of C57BL/6 SCID mice can also clear the virus. The
to intestinal pathogens or non-pathogenic
commensal intestinal bacteria relative importance of the T- and B-cell responses in the
adaptive response has been extensively examined by infec-
Experiments have also addressed the mechanisms of tion of strains: i) after immunodepletion of T or B cells [92];
IgA induction to microbial pathogens or non-pathogenic ii) following transfers into (SCID) animals [90]; by studies
intestinal commensal intestinal flora. It is now clear that in mice: iii) deficient in B cells (JH–/– mice [91, 92]); or iv)
although IgA induction by cholera toxin is very dependent selectively deficient in IgA [93]); v) those deficient in both
on T-cell help, cognate T-cell help is certainly not obliga- CD4 and CD8 T cells (nude mice or T-cell receptor β–/–
tory for IgA induction by pathogens (Section 4.1) or non- δ–/– [94]); or vi) deficient in cytotoxic T cells alone (β2
pathogenic commensal intestinal bacteria (Section 5.3). microglobulin–/– mice or mice depleted of CD8+ cells [91,
Of course, this is an explanation of the paradox in CD4–/– 94]). The overall conclusions from these experiments are
and CTLA4-Hγ1 animals described above, where T-cell that B cells which permit antibody production are protec-
help or co-stimulatory responses are defective, yet the tive both in the initial infection and in rechallenge, whereas
unmanipulated content of intestinal IgA is normal. CTL deficiency delays viral clearance.
Other strong antigens (recombinant Salmonella, As described in Section 3, the induction of IgA has
ISCOMS) can also induce IgA in IL-4–/– mice [80, 81]. In usually been thought highly dependent on the presence of
vivo investigations of isolated deficiency of cytokines that specific T-cell help. Thus it was a surprise when both nude
act in vitro to enhance IgA secretion after the switch mice (having a very restricted T-cell compartment) and
process has taken place shows no defect in IL-5–/– mice, T-cell receptor β–/– δ–/– mice (which lack T cells) were
but conflicting results with IL-6–/– mice [82, 83]. capable of viral clearance even on the C57BL/6x129
background, where innate mechanisms are relatively inef-
fective [94]. Moreover, both IgM (known to be T-cell-
4. Protection by IgA independent) and small amounts of specific IgA were
from intestinal infections produced in the serum and secretions of these mice. Since
protection from virus correlated with T-cell-independent
IgA has been shown to neutralise viruses in culture, and IgA production, induction of some protective IgA can
to form part of the protective response in vivo. The success probably occur in response to pathogens independently of
of mucosal vaccination against polio with live attenuated T-cell help.
virus has been one of the great achievements of modern To look at the protective effect of secreted IgA per se,
healthcare; it was clear from early studies that the route of Burns et al. investigated mice that carried a ’backpack’
immunisation was critical, secretory IgA was induced far hybridoma secreting monoclonal IgAs directed against
more effectively after mucosal rather than parenteral vac- one of the outer rotaviral capsid proteins haemagglutinin

Microbes and Infection 1027


2001, 1021-0
Review Macpherson et al.

(VP4) or glycoprotein (VP7), or the inner capsid protein Vibrio cholera [106], Shigella (through the bronchial
VP6 [95]. Two monoclonal antibodies to VP6 secreted mucosa [107]) and the low-grade pathogen Helicobacter
from backpack tumours were able to protect BALB/c mice felis [108].
from rotaviral infection, or to clear the virus from prein- Potentially therefore, IgA in sufficient quantities can be
fected SCID mice. Strangely, these monoclonal antibodies shown to be protective in at least some models. However,
were not neutralising in vitro (i.e. they do not protect cells evidence from the strain of mice which selectively lack
in culture from becoming infected with virus as seen with IgA (but not other immunoglobulin isotypes) owing to a
some antibodies against VP4 or VP7). Moreover, they were targeted genetic lesion, suggests that IgA is often not
not active in vivo when present on the luminal side of the obligatory for mucosal protection.
gastrointestinal tract, so the in vivo effect in the mucosa
(which presumably depends on intracellular transport of
IgA across epithelial cells by the pIgR) works through an
intracellular mechanism. The probable physiological
5. Interactions between IgA
importance of IgA in protecting animals from mucosal and the commensal intestinal flora
infections was reinforced by the observation that a back-
pack hybridoma secreting large amounts of IgG antibody 5.1. Effects of colonisation of intestines
with commensal bacterial flora on intestinal IgA
that did effectively neutralise in vitro (against VP7) could
neither prevent nor resolve rotavirus infection in vivo. As noted at the start of this review, the intestinal tract of
However, whilst IgA is sufficient for protection, it is not mammals is colonised by a dense bacterial flora of (usu-
obligatory, and other mechanisms (probably mainly IgM) ally) non-pathogenic organisms, which confer advantages
can compensate for its absence in IgA–/– mice [93]. to the host in terms of metabolism of otherwise indigest-
ible carbohydrates, and thus allow the host animal to
4.2. Other mucosal infections salvage energy from the diet. The presence of the com-
mensal flora also competes with pathogenic organisms,
The role of secreted IgA has also been studied in and limits their ability to colonise/infect the intestinal
relation to mucosal infection with influenza. Whilst the tract. The commensal flora has a remarkable density: there
influenza models depend on respiratory tract challenge are 1012 bacteria/g faeces in the colon, and approximately
with pathogen, they are discussed here because of the 103 known species, with anaerobes predominating.
relevance of potential functional protection by IgA.
A useful tool to understand the impact of the non-
After transferring polymeric or monomeric monoclonal pathogenic commensals on the immune system has been
IgA, or monomeric monoclonal IgG, each specific for the the use of germ-free (gnotobiotic) mice. A colony of germ-
haemagglutinin of PR8 influenza virus, Renegar and Small free animals can initially be set up by delivering the pups
were able to show that polymeric IgA was selectively through Caesarian section and hand-rearing them in a
transported into nasal secretions and bile, and that poly- sterile isolator. Such hand-rearing is extremely demand-
meric IgA (but not IgG) transfer protected mice against ing, but subsequently it is much easier to interbreed the
subsequent nasal challenge with live virus. This protection offspring within the isolator, keeping the conditions
could be abrogated by intranasal administration of anti- (including the food and water) sterile. Germ-free animals
IgA antiserum [96]. Similarly, mice that had recovered can be moved between isolators to separate some animals
from an influenza infection 4–6 weeks previously could from the sterile breeding colony for experiments, such as
be reinfected intranasally if the nasopharynx was treated recolonisation of the intestine with defined bacteria. Also,
with anti-IgA [97]. Studies of cross-protection between different strains of mice can be derived germ-free without
different strains of influenza infecting mice through a nasal the need for hand-rearing, by transferring embryos from
route also correlate better with sIgA production than cyto- that strain into a germ-free recipient foster-mother.
toxic T-cell reactivity [98, 99]; CD8+ CTLs do contribute to Germ-free mice have extremely small Peyer’s patches
recovery from infection, but are not strong mediators of and a very small number of lamina propria IgA-producing
resistance to reinfection [100–103]. plasma cells (figure 6). The number of CD4+ T cells in the
Despite the data above that show that that passive intestinal lamina propria, and αβ T-cell receptor CD8+
transfer of immune immunoglobulins can protect against cells in the intraepithelial compartment is also reduced,
subsequent influenza infection, it has not been straightfor- although the γδ T-cell receptor CD8+ intraepithelial com-
ward to show increased susceptibility to (intranasal) influ- ponent is essentially normal.
enza infection in mice that are selectively deficient in IgA Cebra and his colleagues have carried out experiments
[104] or J chain [103] as a result of a targeted genetic to define the sequence of changes in the mucosal immune
lesion. This appears to be partly due to the experimental system when germ-free mice are recolonised with a single
protocols used: in the protection study of the IgA–/– mice species of Gram-negative aerobic bacteria [109]. Their
[104], the influenza subunit vaccine was given with chol- results are illustrated in figure 7. Initially, there was very
era toxin; when this strong mucosal adjuvant is omitted, low production of intestinal IgA, and the recolonising
the protection is much weaker in IgA–/– than in wild-type commensal bacteria were not successfully confined to the
animals [105]. intestine. Organisms could be cultured from the spleen
Backpack models have been used with other experi- and mesenteric lymph nodes for up to 100 days, having
mental pathogens to show that specific IgA produced in a penetrated or ’translocated’ from the intestinal lumen to
hybridoma is protective against challenge; these include underlying tissues. The reduction and disappearance of
1028 Microbes and Infection
2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

Figure 7. Set-up of the experiment by Shroff et al. [109] in


which germ-free mice were recolonised by a single Gram-negative
commensal organism Morganella morganii. The mice were kept in
an isolator throughout to avoid contamination with other organ-
isms. Initially the bacteria penetrated into the body and could be
cultured from the mesenteric lymph nodes and the spleen. How-
ever, once IgA had been induced (and other adaptive changes had
occurred in the mucosal immune system), the bacteria were
successfully confined to the intestine.
Figure 6. Histology of intestinal sections from germ-free mice
and mice with a normal (specific pathogen-free, SPF) intestinal
bacterial flora. A) IgA-secreting cells are very sparse in germ-free
mice compared with B) SPF mice. C) The Peyer’s patches of sal bacteria, which depends on penetration of the com-
germ-free mice are macroscopically very small: microscopically mensal organisms, is much more likely in IgA–/– animals
they have relatively few germinal centres and IgA+ B cells; D) [52].
SPF Peyer’s patch shown for comparison. E) The number of CD4+
cells in the lamina propria is also less in germ-free mice compared 5.2. Functional effects of milk IgA
with F) SPF mice, but the numbers of CD8+ intrapepithelial cells on the neonatal mucosal immune system
in germ-free animals (G), are comparable with those with an SPF
flora (H). In another series of experiments carried out by Cebra
and his colleagues, the immune value of the mothers’ milk
was determined. IgA in milk is initially derived from the
translocation coincided with the appearance of secreted serum, and later is produced by plasma cells within the
intestinal IgA, so it is possible that IgA, once induced, mammary tissue. IgA in the milk can also be shown to bind
prevents the commensal bacteria from leaving the lumen to commensal bacteria [37], and it is clear that during the
of the intestine and reaching extraluminal tissues. Whilst period of exclusive lactation the number of species of the
this is probably broadly correct (see Section 5.3 below), commensal flora and their density are restricted, with a
IgA is not the only factor that limits the translocation of normal adult flora composition being rapidly established
commensal bacteria, as other changes also occur after once other nutritional sources (formula milk/weaning to
recolonisation, including increased content of lamina pro- solids) are introduced. To look at the impact of immuno-
pria CD4 and αβTCR CD8 cells, and increased macro- globulins in the milk on the development of the mucosal
phage activation, so the exact contribution of secreted immune system, the results of immunoincompetent (SCID)
intestinal IgA in preventing the penetration of commensal dams nursing immunocompetent (scid/+) pups were com-
organisms from the lumen of the bowel has not been pared with immunocompetent dams nursing immuno-
formally shown. However, we have found that spontane- competent pups. Since SCID dams have the innate immune
ous priming of the systemic immune system by commen- components present in milk, these experiments reveal the

Microbes and Infection 1029


2001, 1021-0
Review Macpherson et al.

effects of adaptive components, principally the immuno- 5.3. Intestinal IgA directed against
globulins. Those pups nursed by SCID mothers showed the commensal intestinal bacterial flora
early induction of germinal centres in the Peyer’s patches Despite the immense load of commensal intestinal
at postnatal day 15, rather than day 21 when the nursing bacteria, normal mice are systemically immunologically
mother was immunocompetent [37]. ignorant of these organisms when kept in a pathogen-free
environment. We showed this by studying serum IgG-
Using this system the responses of neonatal mice to
binding to proteins of Enterobacter cloacae (the principal
infection with reovirus were also studied [110]. They
aerobe in the intestinal flora): whilst normal mice had no
found that the neonatal mice were quite able to mount
binding IgG, this was easily induced 14 days after an
specific mucosal responses to reovirus (from postnatal day
intravenous infection with 106 cultured bacteria. Some of
10), and to generate specific IgA in the Peyer’s patches.
this protection is due to the barrier of secreted IgA, because
Moreover, when litter swap experiments were carried out,
IgA–/– mice are much more likely to have spontaneous
an immunocompetent pup lactating (since birth) on a
priming of a systemic immune response (i.e. without intra-
non-immune nurse dam mounted a mucosal response to a
venous infection) despite being kept in pathogen-free con-
reovirus infection whether or not it had been born to a ditions [52].
reovirus immune (immunocompetent) mother. The reverse Although wild-type mice have no serum IgG- (or serum
situation, where an immunocompetent pup was lactated IgA-) binding to commensal bacteria, Western blot experi-
by an immune dam, did not mount a response, whether or ments show that intestinal secretory IgA is spontaneously
not it had been born to an immune dam. Therefore the induced by the presence of commensal organisms in the
adaptive immunity of milk (in mice) is critical in conferring intestine. Thus germ-free mice do not bind proteins of
immune protection against oral challenge with reovirus E. cloacae (even allowing for the reduced intestinal IgA
independently of placental immunoglobulin transfer. content). That intestinal secretory IgA is responsive to
In mice, transfer of maternal IgG continues from birth changes in the intestinal flora can also be shown by
by uptake from the milk in the duodenum (which is largely colonising the intestines of mice with bacteria that express
complete by postnatal day 10) [111, 112]. (This mecha- a novel protein in vivo; following colonisation sIgA bind-
nism plays a much smaller role in humans, although some ing to the new commensal bacterial protein is induced
colostral IgG can probably be transferred through the [52].
neonatal intestine [112,113]). Whilst adaptive responses Experiments with cholera toxin (described in Section
are therefore the key to explaining the premature induc- 3.3) have shown that induction of specific intestinal IgA is
tion of the natural responses (presumably to commensal very dependent on T-cell help, yet the total content of IgA
bacteria) in the mucosal immune system, and protection is not reduced in circumstances where T-cell help or
from infection in neonatal mice, one cannot say what role costimulation are lacking, when cholera toxin induction is
IgA and IgG have individually in the effects. However, the ineffective. Using mice with targeted lesion of the β and δ
resources to resolve this experimentally are available. chains of the T-cell receptor (TCRβ–/– δ–/–), we showed that
intestinal IgA content was reduced, but only to about a
In humans (where maternofoetal immunoglobulin trans- quarter of wild-type levels. In agreement with this, the
fer is largely placental, so benefits of lactation are on induction of intestinal IgA was found to be unimpaired in
limiting the exposure of the neonate to commensal bacte- tumour necrosis factor receptor (TNFR)-I–/– mice, which
ria or to pathogens) it is clear that breast milk can contain are characterised by almost no Peyer’s patches, and those
antibodies to pathogenic intestinal bacteria recently that are present lack the normal follicular structure and
encountered by the mother. Epidemiological studies have germinal centres. Moreover, the ability of the T-cell-
consistently demonstrated that the benefits of breast- deficient (or TNFR-I–/–) mice to mount intestinal sIgA adap-
feeding are enormous, and that breast-fed infants in devel- tive responses to changes in the antigenic content of the
oping countries are protected from death from infectious intestinal flora was unimpaired. Thus IgA is only partly
causes (especially dehydrating diarrhoea), although a com- dependent on T-cell help for induction, and in particular
ponent of this benefit is improved nutrition [114, 115]. In the responses to the commensal intestinal flora are much
developed countries, where nutrition from natural and more T-independent than responses to cholera toxin. This
formula milk is similar, there is also an association with appears to make sense, since protection from the intestinal
reduced gastrointestinal illness [115]. There is also some flora would presumably require a broad-based secretory
evidence that the benefits of adequate breast-feeding can IgA response, whereas the production of neutralising activ-
persist many years after weaning, with significant reduc- ity against intestinal bacterial toxins needs a narrow speci-
tions in childhood respiratory illness and other allergic ficity, high-affinity IgA.
disorders [116–118]. Exclusively breast-fed infants have a It has been known that IgA hybridomas from B1 cells
different composition of the commensal intestinal bacte- produce antibodies that bind to intestinal bacterial deter-
rial flora compared with exclusively or partially formula- minants [119]. Using the radiation chimaera approach
fed infants [1] (as simple examination of the nappies will described in Section 2.2, and illustrated in figure 5, we
confirm). The details of the in vivo mechanisms by which showed that much of the T-independent intestinal IgA was
innate and adaptive responses in the milk influence the of B1 lymphocyte origin. The T-independent intestinal IgA
intestinal flora of the neonate and impact on later mucosal derived from B1 cells is not ’natural’ antibody in the sense
and systemic immunity remain unclear and need further that it is present in the absence of intestinal bacteria
study. themselves; T-cell-deficient mice recolonised with bacte
1030 Microbes and Infection
2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

ria producing a novel protein in the intestine in vivo mount [5] Conley M.E., Delacroix D.L., Intravascular and mucosal
an IgA response to that protein, so as with some serum immunoglobulin A: two separate but related systems of
B1-derived antibodies [46, 47, the T-independent system immune defense? Ann. Intern. Med. 106 (1987) 892–899.
is moulded (positively selected) by changes in the intesti- [6] Kaetzel C.S., Robinson J.K., Lamm M.E., Epithelial trans-
nal flora. The fact that this occurs even in mice which cytosis of monomeric IgA and IgG cross-linked through
already have a flora indicates that this is not just a non- antigen to polymeric IgA. A role for monomeric antibod-
specific immunostimulatory effect of the presence of intes- ies in the mucosal immune system, J. Immunol. 152
tinal bacteria per se, but the system can respond to alter- (1994) 72–76.
ations in the content and diversity of the intestinal flora. [7] Kaetzel C.S., Robinson J.K., Chintalacharuvu K.R., Vaer-
The precise role of these adaptive IgA responses in provid- man J.P., Lamm M.E., The polymeric immunoglobulin
ing a barrier against penetration of commensal organisms receptor (secretory component) mediates transport of
from the intestine into the body remains to be determined. immune complexes across epithelial cells: a local defense
function for IgA, Proc. Natl. Acad. Sci. USA 88 (1991)
8796–8800.
6. Conclusions [8] Macpherson A., Khoo U.Y., Forgacs I., Philpott-
Howard J., Bjarnason I., Mucosal antibodies in inflamma-
Although we now know that IgA can be induced both tory bowel disease are directed against intestinal bacteria,
by T-dependent and T-independent mechanisms, and it Gut 38 (1996) 365–375.
can bind bacteria of the commensal intestinal flora, we
[9] Friman V., Quiding M., Czerkinsky C., Nordstrom I.,
still know comparatively little about its influence on the
Larsson L., Ericson D., Bjorkander J., Theman K., Kilan-
density and composition of the flora. This is not an easy
der A., Holmgren J., et al., Intestinal and circulating
area to address, because intestinal bacteria exist in mucus
antibody-forming cells in IgA-deficient individuals after
biofilms adjacent to the epithelial cell layer as well as the
oral cholera vaccination, Clin. Exp. Immunol. 95 (1994)
free-living (planktonic) forms present in the intestinal
222–226.
lumen. It is the organised biofilm colonies that are likely to
be the most relevant for stimulation of the mucosal and [10] Harriman G.R., Bogue M., Rogers P., Finegold M.,
systemic immune systems, and penetration into the body Pacheco S., Bradley A., Zhang Y., Mbawuike I.N., Tar-
when the intestinal barrier is breached. The fact that the geted deletion of the IgA constant region in mice leads to
bacterial flora is limited in neonates by adaptive and IgA deficiency with alterations in expression of other
innate immune factors in maternal milk has poorly under- immunoglobulin isotypes, J. Immunol. 162 (1999)
stood consequences on the developing immune system, 2521–2529.
but epidemiological evidence in formula-fed infants in the [11] Orlans E., Peppard J., Reynolds J., Hall J., Rapid active
developing and developed world suggests that adaptive transport of immunoglobulin A from blood to bile, J. Exp.
immunity transmitted from the intestine to the mammary Med. 147 (1978) 588–592.
gland may have significant (and possibly long-term) con- [12] Jackson G.D., Lemaitre-Coelho I., Vaerman J.P.,
sequences for human health. Bazin H., Beckers A., Rapid disappearance from serum of
intravenously injected rat myeloma IgA and its secretion
into bile, Eur. J. Immunol. 8 (1978) 123–126.
Acknowledgments [13] Socken D.J., Jeejeebhoy K.N., Bazin H., Underdown B.J.,
Identification of secretory component as an IgA receptor
on rat hepatocytes, J. Exp. Med. 150 (1979) 1538–1548.
We would like to thank Professors Rolf Zinkernagel and
Hans Hengartner for their encouragement and support. [14] Orlans E., Peppard J., Fry J.F., Hinton R.H., Mul-
lock B.M., Secretory component as the receptor for poly-
meric IgA on rat hepatocytes, J. Exp. Med. 150 (1979)
1577–1581.
References [15] Kuhn L.C., Kraehenbuhl J.P., The membrane receptor for
polymeric immunoglobulin is structurally related to secre-
[1] Mackie R., Sghir A., Gaskins H.R., Developmental micro- tory component. Isolation and characterization of mem-
bial ecology of the neonatal gastrointestinal tract, Am. brane secretory component from rabbit liver and mam-
J. Clin. Nutr. 69 (1999) 1035S–1045S. mary gland, J. Biol. Chem. 256 (1981) 12490–12495.
[2] Brandtzaeg P., Prydz H., Direct evidence for an integrated [16] Delacroix D.L., Hodgson H.J., McPherson A., Dive C.,
function of J chain and secretory component in epithelial Vaerman J.P., Selective transport of polymeric immuno-
transport of immunoglobulins, Nature 311 (1984) 71–73. globulin A in bile. Quantitative relationships of mono-
[3] Mostov K.E., Transepithelial transport of immunoglobu- meric and polymeric immunoglobulin A, immunoglobu-
lins, Annu. Rev. Immunol. 12 (1994) 63–84. lin M, and other proteins in serum, bile, and saliva, J. Clin.
[4] Brandtzaeg P., Krajci P., Lamm M.E., Kaetzel C.S., in: Invest. 70 (1982) 230–241.
Ogra P.L., Mestecky J., Lamm M.E., Strober W., [17] Dooley J.S., Potter B.J., Thomas H.C., Sherlock S., A
McGhee J.R., Bienenstock J. (Eds.), Handbook of Mucosal comparative study of the biliary secretion of human
Immunology, Academic Press, San Diego, 1994, dimeric and monomeric IgA in the rat and in man, Hepa-
pp. 113–126. tology 2 (1982) 323–327.

Microbes and Infection 1031


2001, 1021-0
Review Macpherson et al.

[18] Nagura H., Smith P.D., Nakane P.K., Brown W.R., IgA [33] Guy-Grand D., Griscelli C., Vassalli P., The gut-associated
in human bile and liver, J. Immunol. 126 (1981) 587–595. lymphoid system: nature and properties of the large divid-
[19] Nagura H., Tsutsumi Y., Hasegawa H., Watanabe K., ing cells, Eur. J. Immunol. 4 (1974) 435–443.
Nakane P.K., Brown W.R., IgA plasma cells in biliary [34] Craig S.W., Cebra J.J., Peyer’s patches: an enriched source
mucosa: a likely source of locally synthesized IgA in of precursors for IgA-producing immunocytes in the rab-
human hepatic bile, Clin. Exp. Immunol. 54 (1983) bit, J. Exp. Med. 134 (1971) 188–200.
671–680. [35] Husband A.J., Gowans J.L., The origin and antigen-
[20] Vuitton D.A., Seilles E., Claude P., Sava P., Delac- dependent distribution of IgA-containing cells in the
roix D.L., Gall bladder: the predominant source of bile intestine, J. Exp. Med. 148 (1978) 1146–1160.
IgA in man? Clin. Exp. Immunol. 62 (1985) 185–192. [36] Pierce N.F., Gowans J.L., Cellular kinetics of the intesti-
[21] Tomana M., Kulhavy R., Mestecky J., Receptor-mediated nal immune response to cholera toxoid in rats, J. Exp.
binding and uptake of immunoglobulin A by human Med. 142 (1975) 1550–1563.
liver, Gastroenterology 94 (1988) 762–770. [37] Kramer D.R., Cebra J.J., Early appearance of ‘natural’
[22] Weisz-Carrington P., Roux M.E., Lamm M.E., Plasma mucosal IgA responses and germinal centers in suckling
cells and epithelial immunoglobulins in the mouse mam- mice developing in the absence of maternal antibodies,
mary gland during pregnancy and lactation, J. Immunol. J. Immunol. 154 (1995) 2051–2062.
119 (1977) 1306–1307. [38] Melchers F., Rolink A., in: Paul W.E. (Ed.), Fundamental
[23] Halsey J.F., Mitchell C., Meyer R., Cebra J.J., Metabolism Immunology, Lippincott-Raven, Philadelphia, 1999,
of immunoglobulin A in lactating mice: origins of immu- pp. 183–261.
noglobulin A in milk, Eur. J. Immunol. 12 (1982) [39] Lam K.P., Rajewsky K., B cell antigen receptor specificity
107–112. and surface density together determine B-1 versus B-2 cell
[24] Roux M.E., McWilliams M., Phillips-Quagliata J.M., development, J. Exp. Med. 190 (1999) 471–478.
Weisz-Carrington P., Lamm M.E., Origin of IgA-secreting [40] Hayakawa K., Hardy R.R., Parks D.R., Herzenberg L.A.,
plasma cells in the mammary gland, J. Exp. Med. 146 The ‘Ly-1 B’ cell subpopulation in normal immunodefec-
(1977) 1311–1322. tive, and autoimmune mice, J. Exp. Med. 157 (1983)
[25] Weisz-Carrington P., Roux M.E., McWilliams M., JM P.- 202–218.
Q., Lamm M.E., Organ and isotype distribution of plasma
[41] Hayakawa K., Hardy R.R., Herzenberg L.A., Progenitors
cells producing specific antibody after oral immunization:
for Ly-1 B cells are distinct from progenitors for other B
evidence for a generalized secretory immune system,
cells, J. Exp. Med. 161 (1985) 1554–1568.
J. Immunol. 123 (1979) 1705–1708.
[42] Hayakawa K., Hardy R.R., Normal, autoimmune, and
[26] Weisbart R.H., Kacena A., Schuh A., Golde D.W.,
malignant CD5+ B cells: the Ly-1 B lineage? Annu. Rev.
GM-CSF induces human neutrophil IgA-mediated phago-
Immunol. 6 (1988) 197–218.
cytosis by an IgA Fc receptor activation mechanism,
Nature 332 (1988) 647–648. [43] Förster I., Gu H., Rajemsky K., Germline antibody V
[27] Monteiro R.C., Kubagawa H., Cooper M.D., Cellular regions as determinants of clonal persistence and malig-
distribution, regulation, and biochemical nature of an Fc nant growth in the B cell compartment, EMBO J. 7
alpha receptor in humans, J. Exp. Med. 171 (1990) (1988) 3693.
597–613. [44] Tarlinton D., Stall A.M., Herzenberg L.A., Repetitive
[28] Deo Y.M., Sundarapandiyan K., Keler T., Wallace P.K., usage of immunoglobulin VH and D gene segments in
Graziano R.F., Bispecific molecules directed to the Fc CD5+ Ly-1 B clones of (NZB x NZW)F1 mice, EMBO J.
receptor for IgA (Fc alpha RI, CD89) and tumor antigens 7 (1988) 3705–3710.
efficiently promote cell-mediated cytotoxicity of tumor [45] Haury M., Sundblad A., Grandien A., Barreau C.,
targets in whole blood, J. Immunol. 160 (1998) Coutinho A., Nobrega A., The repertoire of IgM in nor-
1677–1686. mal mice is largely independent of external antigenic
[29] Patry C., Herbelin A., Lehuen A., Bach J.F., Mon- contact, Eur. J. Immunol. 27 (1997) 1557–1563.
teiro R.C., Fc alpha receptors mediate release of tumour [46] Murakami M., Nakajima K., Yamazaki K., Muraguchi T.,
necrosis factor-alpha and interleukin-6 by human mono- Serikawa T., Honjo T., Effects of breeding environments
cytes following receptor aggregation, Immunology 86 on generation and activation of autoreactive B-1 cells in
(1995) 1–5. anti-red blood cell autoantibody transgenic mice, J. Exp.
[30] van Egmond M., van Garderen E., van Spriel A.B., Med. 185 (1997) 791–794.
Damen C.A., van Amersfoort E.S., van Zandbergen G., [47] Hayakawa K., Asano M., Shinton S.A., Gui M., All-
van Hattum J., Kuiper J., van de Winkel J.G., FcalphaRI- man D., Stewart C.L., Silver J., Hardy R.R., Positive
positive liver Kupffer cells: reappraisal of the function of selection of natural autoreactive B cells, Science 285 (1999)
immunoglobulin A in immunity, Nat. Med. 6 (2000) 113–116.
680–685. [48] Cazac B.B., Roes J., TGF-beta receptor controls B cell
[31] Gowans J.L., Knight E.J., The route of recirculation of responsiveness and induction of IgA in vivo, Immunity 13
lymphocytes in the rat, Proc. R. Soc. Lond. B. Biol. Sci. (2000) 443–451.
159 (1964) 257–282. [49] Hayakawa K., Hardy R.R., Stall A.M., Herzenberg L.A.,
[32] Parrott D.M., Ferguson A., Selective migration of lym- Immunoglobulin-bearing B cells reconstitute and main-
phocytes within the mouse small intestine, Immunology tain the murine Ly-1 B cell lineage, Eur. J. Immunol. 16
26 (1974) 571–588. (1986) 1313–1316.
1032 Microbes and Infection
2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

[50] Kroese F.G.M., Butcher E.C., Stall A.M., Lalor P.A., [65] Defrance T., Vanbervliet B., Briere F., Durand I., Rous-
Adams S., Herzenberg L.A., Many of the IgA producing set F., Banchereau J., Interleukin 10 and transforming
plasma cells in the murine gut are derived from self- growth factor beta cooperate to induce anti- CD40-
replenishing precursors in the peritoneal cavity, Int. activated naive human B cells to secrete immunoglobulin,
Immunol. 1 (1988) 75–84. J. Exp. Med. 175 (1992) 671–682.
[51] Snider D.P., Liang H., Switzer I., Underdown B.J., IgA [66] Weinstein P.D., Schweitzer P.A., Cebra-Thomas A.,
production in MHC class II-deficient mice is primarily a Cebra J.J., Molecular genetic features reflecting the pref-
function of B-1a cells, Int. Immunol. 11 (1999) 191–198. erence for isotype switching to IgA expression by Peyer’s
[52] Macpherson A.J., Gatto D., Sainsbury E., Harriman G.R., patch germinal center B cells, Int. Immunol. 3 (1991)
Hengartner H., Zinkernagel R.M., A primitive T cell- 1253–1263.
independent mechanism of intestinal mucosal IgA [67] Schrader C.E., Cebra J.J., Dendritic cell dependent expres-
responses to commensal bacteria, Science 288 (2000) sion of IgA by clones in T/B microcultures, Adv. Exp.
2222–2226. Med. Biol. 329 (1993) 59–64.
[53] Antin J.H., Emerson S.G., Martin P., Gadol N., [68] Elson C.O., Ealding W., Generalized systemic and mucosal
Ault K.A., Leu-1+ (CD5+) B cells. A major lymphoid immunity in mice after mucosal stimulation with cholera
subpopulation in human fetal spleen: phenotypic and toxin, J. Immunol. 132 (1984) 2736–2741.
functional studies, J. Immunol. 136 (1986) 505–510. [69] Elson C.O., Ealding W., Cholera toxin feeding did not
[54] Casali P., Notkins A.L., CD5+ B lymphocytes, polyreac- induce oral tolerance in mice and abrogated oral tolerance
tive antibodies and the human B-cell repertoire, Immu- to an unrelated protein antigen, J. Immunol. 133 (1984)
nol. Today 10 (1989) 364–368. 2892–2897.
[55] Hardy R.R., Hayakawa K., Shimizu M., Yamasaki K., [70] Snider D.P., Marshall J.S., Perdue M.H., Liang H., Pro-
Kishimoto T., Rheumatoid factor secretion from human duction of IgE antibody and allergic sensitization of intes-
Leu-1+ B cells, Science 236 (1987) 81–83. tinal and peripheral tissues after oral immunization with
[56] Fischer M., Küppers R., Human IgA- and IgM-secreting protein Ag and cholera toxin, J. Immunol. 153 (1994)
intestinal plasma cells carry heavily mutated Vh region 647–657.
genes, Eur. J. Immunol. 28 (1998) 2971–2977. [71] Lycke N., Holmgren J., Intestinal mucosal memory and
[57] Boursier L., Dunn-Walters D.K., Spencer J., Characteris- presence of memory cells in lamina propria and Peyer’s
tics of IgVH genes used by human intestinal plasma cells patches in mice 2 years after oral immunization with
from childhood, Immunology 97 (1999) 558–564. cholera toxin, Scand. J. Immunol. 23 (1986) 611–616.
[58] Dunn-Walters D.K., Boursier L., Spencer J., Hypermuta- [72] Hörnquist C.E., Ekman L., Grdic K.D., Schön K.,
tion, diversity and dissemination of human intestinal Lycke N.Y., Paradoxical IgA immunity in CD4-deficient
lamina propria plasma cells, Eur. J. Immunol. 27 (1997) mice, J. Immunol. 155 (1995) 2877–2887.
2959–2964.
[73] Lycke N., Erlandsson L., Ekman L., Schon K., Leander-
[59] Guy-Grand D., Griscelli C., Vassalli P., Peyer’s patches,
son T., Lack of J chain inhibits the transport of gut IgA
gut IgA plasma cells and thymic function: study in nude
and abrogates the development of intestinal antitoxic
mice bearing thymic grafts, J. Immunol. 115 (1975)
protection, J. Immunol. 163 (1999) 913–919.
361–364.
[74] Vajdy M., Kosco-Vilbois M.H., Kopf M., Kohler G.,
[60] Lycke N., Eriksen L., Holmgren J., Protection against
Lycke N., Impaired mucosal immune responses in inter-
cholera toxin after oral immunisation is thymus depen-
leukin 4-targeted mice, J. Exp. Med. 181 (1995) 41–53.
dent and associated with intestinal production of neutral-
ising IgA antitoxin, Scand. J. Immunol. 25 (1987) [75] Gardby E., Lane P., Lycke N.Y., Requirements for
413–419. B7-CD28 costimulation in mucosal IgA responses: para-
[61] Coffman R.L., Lebman D.A., Shrader B., Transforming doxes observed in CTLA4-H gamma 1 transgenic mice,
growth factor β specifically enhances IgA production by J. Immunol. 161 (1998) 49–59.
lipopolysaccharide stimulated murine B lymphocytes, [76] Shull M.M., Ormsby I., Kier A.B., Pawlowski S., Die-
J. Exp. Med. 170 (1989) 1039–1044. bold R.J., Yin M., Allen R., Sidman C., Proetzel G.,
[62] Kunimoto D.Y., Harriman G.R., Strober W., Regulation Calvin D., et al., Targeted disruption of the mouse trans-
of IgA differentiation in CH12LX B cells by lymphokines: forming growth factor-beta 1 gene results in multifocal
IL-4 induces membrane IgM-positive CH12LX cells to inflammatory disease, Nature 359 (1992) 693–699.
express membrane IgA and IL-5 induces membrane IgA- [77] Kuhn R., Lohler J., Rennick D., Rajewsky K., Muller W.,
positive CH12LX cells to secrete IgA, J. Immunol. 141 Interleukin-10-deficient mice develop chronic enterocoli-
(1988) 713–720. tis, Cell 75 (1993) 263–274.
[63] Beagley K.W., Eldridge J.H., Kiyono H., Everson M.P., [78] Sadlack B., Merz H., Schorle H., Schimpl A., Feller A.C.,
Koopman W.J., Honjo T., McGhee J.R., Recombinant Horak I., Ulcerative colitis-like disease in mice with a
murine IL-5 induces high rate IgA synthesis in cycling disrupted interleukin-2 gene, Cell 75 (1993) 253–261.
IgA-positive Peyer’s patch B cells, J. Immunol. 141 (1988) [79] van Ginkel F.W., Wahl S.M., Kearney J.F., Kweon M.N.,
2035–2042. Fujihashi K., Burrows P.D., Kiyono H., McGhee J.R.,
[64] Kunimoto D.Y., Nordan R.P., Strober W., IL-6 is a potent Partial IgA-deficiency with increased Th2-type cytokines
cofactor of IL-1 in IgM synthesis and of IL-5 in IgA in TGF-beta 1 knockout mice, J. Immunol. 163 (1999)
synthesis, J. Immunol. 143 (1989) 2230–2235. 1951–1957.

Microbes and Infection 1033


2001, 1021-0
Review Macpherson et al.

[80] Okahashi N., Yamamoto M., Vancott J.L., Chatfield S.N., [95] Burns J.W., Siadat-Pajouh M., Krishnaney A.A., Green-
Roberts M., Bluethmann H., Hiroi T., Kiyono H., berg H.B., Protective effect of rotavirus VP6-specific IgA
McGhee J.R., Oral immunization of interleukin-4 (IL-4) monoclonal antibodies that lack neutralizing activity, Sci-
knockout mice with a recombinant Salmonella strain or ence 272 (1996) 104–107.
cholera toxin reveals that CD4+ Th2 cells producing IL-6 [96] Renegar K.B., Small P.A., Passive transfer of local immu-
and IL-10 are associated with mucosal immunoglobulin A nity to influenza-virus infection by IgA antibody, J. Immu-
responses, Infect. Immun. 64 (1996) 1516–1525. nol. 146 (1991) 1972–1978.
[81] Mowat A.M., Smith R.E., Donachie A.M., Furrie E., [97] Renegar K.B., Small P.A., Immunoglobulin A mediation
Grdic D., Lycke N., Oral vaccination with immune stimu- of murine nasal anti-influenza virus immunity, J. Virol. 65
lating complexes, Immunol. Lett. 65 (1999) 133–140. (1991) 2146–2148.
[82] Ramsay A.J., Husband A.J., Ramshaw I.A., Bao S., Mat- [98] Takase H., Murakami Y., Endo A., Ikeuchi T., Antibody
thaei K.I., Koehler G., Kopf M., The role of interleukin-6 responses and protection in mice immunized orally against
in mucosal IgA antibody responses in vivo, Science 264 influenza virus, Vaccine 14 (1996) 1651–1656.
(1994) 561–563. [99] Tamura S., Funato H., Hirabayashi Y., Suzuki Y., Naga-
[83] Bromander A.K., Ekman L., Kopf M., Nedrud J.G., mine T., Aizawa C., Kurata T., Cross-protection against
Lycke N.Y., IL-6-deficient mice exhibit normal mucosal influenza-A virus-infection by passively transferred
IgA responses to local immunizations and Helicobacter felis respiratory-tract IgA antibodies to different hemaggluti-
infection, J. Immunol. 156 (1996) 4290–4297. nin molecules, Eur. J. Immunol. 21 (1991) 1337–1344.
[84] Ogra P.L., Karzon D.T., Poliovirus antibody response in [100] Mackenzie C.D., Taylor P.M., Askonas B.A., Rapid recov-
serum and nasal secretions following intranasal inocula- ery of lung histology correlates with clearance of influenza
tion with inactivated poliovaccine, J. Immunol. 102 virus by specific CD8+ cytotoxic T cells, Immunology 67
(1969) 15–23. (1989) 375–381.
[85] Herremans T., Reimerink J.H.J., Buisman A.M., Kim-
[101] Lightman S., Cobbold S., Waldmann H., Askonas B.A.,
man T.G., Koopmans M.P.G., Induction of mucosal
Do L3T4+ T cells act as effector cells in protection against
immunity by inactivated poliovirus vaccine is dependent
influenza virus infection, Immunology 62 (1987)
on previous mucosal contact with live virus, J. Immunol.
139–144.
162 (1999) 5011–5018.
[102] Epstein S.L., Lo C.Y., Misplon J.A., Bennink J.R., Mecha-
[86] Ogra P.L., Kerr-Grant D., Umana G., Dzierba J., Wein-
nism of protective immunity against influenza virus infec-
traub D., Antibody response in serum and nasopharynx
tion in mice without antibodies, J. Immunol. 160 (1998)
after naturally acquired and vaccine-induced infection
322–327.
with rubella virus, N. Engl. J. Med. 285 (1971)
1333–1339. [103] Epstein S.L., Lo C.Y., Misplon J.A., Lawson C.M., Hen-
[87] Franco M.A., Greenberg H.B., Immunity to rotavirus drickson B.A., Max E.E., Subbarao K., Mechanisms of
infection in mice, J. Infect. Dis. 179 (1999) S466–S469. heterosubtypic immunity to lethal influenza A virus infec-
[88] Burns J.W., Krishnaney A.A., Vo P.T., Rouse R.V., Ander- tion in fully immunocompetent, T cell-depleted, beta2-
son L.J., Greenberg H.B., Analyses of homologous rotavi- microglobulin-deficient, and J chain-deficient mice,
rus infection in the mouse model, Virology 207 (1995) J. Immunol. 158 (1997) 1222–1230.
143–153. [104] Mbawuike I.N., Pacheco S., Acuna C.L., Switzer K.C.,
[89] Feng N., Burns J.W., Bracy L., Greenberg H.B., Compari- Zhang Y.X., Harriman G.R., Mucosal immunity to influ-
son of mucosal and systemic humoral immune responses enza without IgA: An IgA knockout mouse model,
and subsequent protection in mice orally inoculated with J. Immunol. 162 (1999) 2530–2537.
a homologous or a heterologous rotavirus, J. Virol. 68 [105] Arulanandam B.P., Raeder R.H., Nedrud J.G.,
(1994) 7766–7773. Bucher D.J., Le J., Metzger D.W., IgA immunodeficiency
[90] Dharakul T., Rott L., Greenberg H.B., Recovery from leads to inadequate Th cell priming and increased suscep-
chronic rotavirus infection in mice with severe combined tibility to influenza virus infection, J. Immunol. 166
immunodeficiency: virus clearance mediated by adoptive (2001) 226–231.
transfer of immune CD8+ T lymphocytes, J. Virol. 64 [106] Winner L.D., Mack J., Weltzin R., Mekalanos J.J., Krae-
(1990) 4375–4382. henbuhl J.P., Neutra M.R., New model for analysis of
[91] Franco M.A., Greenberg H.B., Role of B cells and cyto- mucosal immunity: intestinal secretion of specific mono-
toxic T lymphocytes in clearance of and immunity to clonal immunoglobulin A from hybridoma tumors pro-
rotavirus infection in mice, J. Virol. 69 (1995) 7800–7806. tects against Vibrio cholerae infection, Infect. Immun. 59
[92] McNeal M.M., Barone K.S., Rae M.N., Ward R.L., Effec- (1991) 977–982.
tor functions of antibody and CD8+ cells in resolution of [107] Phalipon A., Kaufmann M., Michetti P., Cavaillon J.M.,
rotavirus infection and protection against reinfection in Huerre M., Sansonetti P., Kraehenbuhl J.P., Monoclonal
mice, Virology 214 (1995) 387–397. immunoglobulin-A antibody-directed against serotype-
[93] O’Neal C.M., Harriman G.R., Conner M.E., Protection of specific epitope of Shigella flexneri lipopolysaccharide pro-
the villus epithelial cells of the small intestine from rotavi- tects against murine experimental shigellosis, J. Exp.
rus infection does not require immunoglobulin A, Med. 182 (1995) 769–778.
J. Virol. 74 (2000) 4102–4109. [108] Czinn S.J., Cai A., Nedrud J.G., Protection of germ-free
[94] Franco M., Greenberg H.B., Immunity to rotavirus in T mice from infection by Helicobacter felis after active oral or
cell deficient mice, Virology 238 (1997) 169–179. passive IgA immunization, Vaccine 11 (1993) 637–642.
1034 Microbes and Infection
2001, 1021-0
IgA induction by pathogenic or non-pathogenic microorganisms Review

[109] Shroff K.E., Meslin K., Cebra J.J., Commensal enteric Gigante L.P., Barros F.C., Evidence for protection by
bacteria engender a self-limiting humoral mucosal breast-feeding against infant deaths from infectious dis-
immune response while permanently colonizing the gut, eases in Brazil, Lancet 2 (1987) 319–322.
Infect. Immun. 63 (1995) 3904–3913. [115] Fuchs S.C., Victora C.G., Martines J., Case-control study
[110] Kramer D.R., Cebra J.J., Role of maternal antibody in the of risk of dehydrating diarrhoea in infants in vulnerable
induction of virus specific and bystander IgA responses in period after full weaning, Br. Med. J. 313 (1996) 391–394.
Peyer’s patches of suckling mice, Int. Immunol. 7 (1995) [116] Howie P.W., Forsyth J.S., Ogston S.A., Clark A., Flo-
911–918. rey C.D., Protective effect of breast feeding against infec-
[111] Jacobowitz Israel E., Patel V.K., Taylor S.F., Marshak- tion, Br. Med. J. 300 (1990) 11–16.
Rothstein A., Simister N.E., Requirement for a beta [117] Lucas A., Brooke O.G., Morley R., Cole T.J., Bam-
2-microglobulin-associated Fc receptor for acquisition of ford M.F., Early diet of preterm infants and development
maternal IgG by fetal and neonatal mice, J. Immunol. 154 of allergic or atopic disease: randomised prospective study,
(1995) 6246–6251. Br. Med. J. 300 (1990) 837–840.
[112] Ghetie V., Ward E.S., Multiple roles for the major histo- [118] Wilson A.C., Forsyth J.S., Greene S.A., Irvine L., Hau C.,
compatibility complex Class I related receptor FcRN, Howie P.W., Relation of infant diet to childhood health:
Annu. Rev. Immunol. 18 (2000) 739–766. seven year follow up of cohort of children in Dundee infant
[113] Dickinson B.L., Badizadegan K., Wu Z., Ahouse J.C., feeding study, Br. Med. J. 316 (1998) 21–25.
Zhu X., Simister N.E., Blumberg R.S., Lencer W.I., Bidi- [119] Bos N.A., Bun J.C., Popma S.H., Cebra E.R.,
rectional FcRn-dependent IgG transport in a polarized Deenen G.J., van der Cammen M.J., Kroese F.G.,
human intestinal epithelial cell line, J. Clin. Inv. 104 Cebra J.J., Monoclonal immunoglobulin A derived from
(1999) 903–911. peritoneal B cells is encoded by both germ line and
[114] Victora C.G., Smith P.G., Vaughan J.P., Nobre L.C., somatically mutated VH genes and is reactive with com-
Lombardi C., Teixeira A.M., Fuchs S.M., Moreira L.B., mensal bacteria, Infect. Immun. 64 (1996) 616–623.

Microbes and Infection 1035


2001, 1021-1035

You might also like