Вы находитесь на странице: 1из 7

Review

TRENDS in Immunology Vol.22 No.4 April 2001

205

IgA and the IgA Fc receptor


Marjolein van Egmond, Cora A. Damen, Annemiek B. van Spriel, Gestur Vidarsson, Evert van Garderen and Jan G.J. van de Winkel
IgA has traditionally been regarded a non-inflammatory antibody. This might indeed be true for secretory IgA (SIgA), which exerts its function at mucosal surfaces where commensal microorganisms and dietary antigens prevail. Serum IgA, however, potently triggers (pro)-inflammatory activity upon binding to the myeloid IgA receptor, FcRI. Here, new insights in the roles of IgA and FcRI are addressed and a model integrating the various functions of IgA in immunity is discussed.

Marjolein van Egmond Depts of Cell Biology and Immunology and Surgical Oncology, Vrije Universiteit, Van de Boechorststraat 7, 1081 BT Amsterdam, The Netherlands. Cora A. Damen Immunotherapy Laboratory, Dept of Immunology, Annemiek B. van Spriel Immunotherapy Laboratory, Dept of Immunology and Medarex Europe, Gestur Vidarsson Immunotherapy Laboratory, Dept of Immunology, Evert van Garderen Dept of Pathology, Faculty of Veterinary Medicine, Yalelaan 1, 3508 TD Utrecht, The Netherlands. Jan G.J. van de Winkel* Immunotherapy Laboratory, Dept of Immunology and Genmab, University Medical Center Utrecht, KC.02-085.2, Lundlaan 6, 3584 EA Utrecht, The Netherlands. *e-mail: j.vandewinkel@ azu.nl

IgA represents the most prominent antibody class at mucosal surfaces1 and the second prevalent antibody class in human serum. In fact, more IgA is produced daily than all other isotypes combined (66 mg kg1 day1)2. This abundance warrants further study and, although scientific interest has increased over the years, the biological role of IgA is still not fully understood. IgA that is excreted into mucosal secretions occurs predominantly as a dimeric complex containing two additional peptides, termed the J-chain and secretory component (SC). The functions of secretory IgA (SIgA) in protecting the mucosal wall are reasonably well understood. At these sites, it is crucial to maintain a balance between immunological responses against foreign pathogens, while reactions against the indigenous microflora and dietary antigens must be avoided. SIgA, therefore, serves as a first line of defense in mucosal areas by inhibiting adhesion of microorganisms. In addition, SIgA has been implicated in the removal of immune complexes, and in neutralization of intracellular viruses3. Because binding of SIgA to antigens does not trigger inflammatory processes, IgA is generally considered a non-inflammatory antibody. This makes sense if one reflects upon the role of SIgA: indeed, if SIgA triggered activatory responses after binding to the commensal microflora, an organism would be at risk of developing chronic inflammation in the intestinal tract. Although the function of serum IgA is incompletely understood, it has become increasingly clear that serum IgA triggers a plethora of effector functions. The description of a receptor for IgA (FcRI; CD89) that is expressed on myeloid cells and potently triggers activatory responses4 further challenges the view of IgA as a non-inflammatory molecule. Here, we aim to broaden the classical view of the role of IgA in immunity.
Structural features of IgA and FcRI Structure of IgA

IgA exists as a heterogeneous molecule and two subclasses termed IgA1 and IgA2 have been defined

that differ by the presence or absence of a 13amino-acid hinge region (Fig. 1a). This region, exclusively present in IgA1, has many O-linked glycosylation sites and is a target for at least two families of IgA1 bacterial proteases, expression of which has been linked to pathogenicity. IgA2 is not susceptible to proteolysis by such proteases and bears two additional N-linked carbohydrate chains. No functional implications of this heterogeneity have yet been shown, although glycosylation differences affect IgAreceptor interactions. For instance, uptake of IgA in the liver is dependent on N-linked glycosylation in mice. Consequently, IgA2 is more rapidly cleared via the asialoglycoprotein receptor on hepatocytes than IgA1 (Ref. 5). Whether this mechanism plays a role in humans remains to be established. The O-linked carbohydrate side-chains of IgA1 are bound by an IgD receptor on human T cells6. Additionally, IgA1 exists as a T-shaped structure, in contrast to the common Y-shape of other Igs7. In most animal species, IgA exists in the circulation as a polymer. In human serum, IgA is predominantly monomeric and constitutes 1520% of the total amount of Igs, whereas at mucosal sites IgA represents the principal antibody class. Here, IgA is expressed with an adjoining peptide, termed the J-chain, which stimulates dimerization2. By binding to the polymeric immunoglobulin receptor (pIgR) expressed on the basolateral surface of mucosal epithelial cells, dimeric IgA (dIgA) is actively transported through the cell8. At the apical membrane, the external domains of pIgR are cleaved off and the complex is released into secretions. The remainder of pIgR that binds covalently to dIgA is called the secretory component (SC), and the SCdIgA complex is referred to as SIgA (Fig. 1b)2. In rodents, pIgR is also expressed on hepatocytes. As a result, polymeric IgA is cleared through the liver by hepatobiliary transport and excreted into bile. In humans, however, this process represents only a minor pathway, owing to low hepatocyte expression levels of pIgR. pIgR transcytosis is tightly regulated, and several signal transduction pathways are involved. Although pIgR transcytosis occurs constitutively in the absence of IgA, binding of dIgA to pIgR augments the transcytosis rate. Two separate signals are required for IgA-induced transcytosis: the sensitization process is initiated by binding of dimeric IgA at the basolateral surface9 which

http://immunology.trends.com 1471-4906/01/$ see front matter 2001 Elsevier Science Ltd. All rights reserved. PII: S1471-4906(01)01873-7

206

Review

TRENDS in Immunology Vol.22 No.4 April 2001

(a)

Antigen-binding sites

(b)
Secretory component (SC)

Fab Hinge

VL C L VH CH1 CH2

O-linked glycosylation FcRIdocking site

VL C L VH CH1 CH2

J-chain

Fc CH3 Tailpiece IgA1

N-linked glycosylation

CH3

IgA2

Secretory IgA (SIgA)


TRENDS in Immunology

Fig. 1. (a) IgA isotypes. IgA is composed of two heavy (blue) and two light (green) chains. Heavy chains consist of three constant regions (CH1, CH2 and CH3) and one variable region (VH), whereas light chains are composed of one constant (CL) and one variable (VL) region. Positions of the FcRI docking site and the O- (), and N- (q) linked glycosylation sites are marked. IgA1 has a T-shaped structure. (b) Secretory IgA (SIgA) consist of two IgA molecules (one light blue, one green). The adjoining J-chain (orange) and secretory component (black) are indicated.

induces the pIgR dimerization necessary to stimulate transcytosis10. The complex is subsequently transported to postmicrotubular compartments, where the sensitized pIgR can respond to a second stimulation signal. The latter signal travels through the cell independently of pIgR movement and involves phosphorylation of the Src family protein tyrosine kinase (PTK) p62yes. This was demonstrated by significantly decreased levels of tyrosine kinase activity associated with pIgR in p62yes-deficient mice, and a reduction in transcytosed dIgA (Refs 11,12). The pIgR docking site on human dimeric IgA has been located in C3 (Ref. 13).
Molecular characteristics of FcRI

FcRI (CD89) is a transmembrane receptor belonging to the Ig receptor gene family, and is 5575 kDa on monocytes and neutrophils, and 70100 kDa on eosinophils, owing to heavy glycosylation4. It consists of a ligand-binding chain, which comprises two extracellular Ig-like domains, a transmembrane region, and a short cytoplasmic tail. Because the latter does not bear any known signaling motifs, FcRI must associate with the promiscuous FcR chain for signaling and function (Fig. 2). FcR chains bear an immunoreceptor tyrosine-based activation motif (ITAM) in their cytoplasmic region, essential for initiation of activatory signals14. This motif consists of two tyrosine-containing YXXL boxes interspaced by seven amino acids, and mutation of either of the tyrosines diminishes or abrogates signaling. FcRI crosslinking triggers ITAM phosphorylation by Src PTK p56lyn (Fig. 2)15. The next steps involve
http://immunology.trends.com

association of p72syk with phosphorylated ITAM, and modulation of adaptor complexes containing tyrosine-phosphorylated Cbl, Shc, SHIP, Grb2, SLP76 and CrkL . Formation of this complex catalyzes recruitment of Sos, followed by activation of Ras, Raf1MEKMAP and phosphoinositide 3kinase pathways16. Recently, FcRI was shown to relocate to specialized sphingolipidcholesterol microdomains in the plasma membrane following crosslinking17. These so-called RAFTs are rich in signaling molecules (including p56lyn) and might thus facilitate signaling18. The importance of the FcR chain for FcRI expression in vivo has been demonstrated in FcRI-transgenic (Tg) mice that were crossed with FcR-chain-deficient mice. Polymorphonuclear cells (PMNs) or monocytes/macrophages of these animals no longer expressed FcRI (Ref. 19). Recently, however, it was reported that some phagocytes express FcRI without the FcR chain20, which suggests that FcRI associates with other (as-yetundefined) molecules. With an affinity of ~106 M1, based upon biosensor analyses, FcRI is a low-affinity receptor21. Accordingly, FcRI binds complexed IgA, whereas monomeric IgA presumably interacts only transiently. Unusually for an FcR, the ligand-binding site for IgA has been located in the first extracellular domain of FcRI, whereas other leukocyte FcRs bind ligand in the second extracellular Ig-like domain2124. The FcRI docking site on IgA is located at or close to the C2/C3 boundary25.
Function of IgA and FcRI IgA-mediated protection at mucosal surfaces

By inhibiting adherence of pathogenic microorganisms to the mucosal wall, SIgA is the primary mediator of immunity in mucosal areas. SIgA is a hydrophilic, negatively charged molecule because of the predominance of hydrophilic amino acids in the Fc region of IgA, and abundant glycosylation of both IgA and SC (Ref. 2). As such, SIgA can surround microorganisms with a hydrophilic shell that is repelled by mucosal

Review

TRENDS in Immunology Vol.22 No.4 April 2001

207

Antigen-binding sites

VH

CH1 CH2 CH3

IgA

FcRI

EC1

EC2

FcR chain

Syk Syk p SLP-76 Cbl p Grb2 Cbl Shc Sos p SHIP

p p

Y Y

Y Y

p p

Syk Syk

Lyn

CrkL

Raf-1/MEK/MAP kinase
RAS

PI-3 kinase

Endocytosis Phagocytosis Antigen presentation Inflammatory mediator release ADCC


TRENDS in Immunology

Fig. 2. The leukocyte IgA receptor FcRI. FcRI is expressed as a transmembrane receptor complex with FcR chain subunits that is stabilized via a unique electrostatic interaction between a positively charged amino acid in FcRI (+) and a negatively charged () amino acid in FcR chains. The ligandbinding -chain consists of two extracellular (EC) Ig-like domains, of which EC1 contains the IgAbinding site (indicated by q). FcR chain bears an immunoreceptor tyrosine (Y)-based activatory motif (ITAM) in its cytoplasmic tail and is essential for signaling. Upon crosslinking of FcRI, the ITAM tyrosines become phosphorylated (P) by the p56lyn Src family kinase, whereupon p72syk associates with phosphorylated ITAM. Next, adaptor complexes, containing tyrosine-phosphorylated Cbl, Shc, SHIP , Grb2, SLP76 and CrkL are modulated, followed by recruitment of Sos and activation of the Ras kinase pathway. Then the Raf1MEKMAP and phosphoinositide 3-kinase (PI-3 kinase) pathways are activated, leading to effector functions. Abbreviation: ADCC, antibody-dependent cellular cytotoxicity.

surfaces. Other mechanisms include its ability to agglutinate microbes and interfere with bacterial motility by interacting with their flagella. In addition to binding to microorganisms, SIgA interacts with bacterial products such as enzymes and toxins, and neutralizes their actions (Fig. 3a). Because of the abundance of food components and microbial flora in the intestinal tract, antigens continuously reach the lamina propria through diffusion or transcytosis. Locally produced IgA interacts with these antigens and the resulting immune complexes are either taken up by phagocytes or transcytosed back to the lumen via the pIgR route (Fig. 3b). This immune elimination role of IgA might provide an effective means of ridding the mucosa of (excessive) immune complexes. Support for this phenomenon was obtained in pIgR/SC-deficient mice26, which are characterized by a complete lack of active IgA (and IgM) transport over the mucosal wall. Furthermore, increased IgG antibodies directed against Escherichia coli are present in the serum, indicating undue triggering of
http://immunology.trends.com

systemic immunity. Elevated levels of albumin in saliva and faeces of these mice reflect leakage of serum proteins, again suggesting a defective epithelial barrier. pIgR, therefore, probably plays a role in maintaining mucosal homeostasis. IgAdeficient individuals provide further support for the immune-exclusion role of IgA. Most patients do not suffer from serious complications, but are predisposed for allergies and autoimmune diseases27. This is presumably a consequence of diminished epithelial barrier function, leading to inappropriate responses against dietary components and indigenous bacterial flora. In several mucosal disorders, such as gluten-sensitive enteropathy and inflammatory bowel disease, this mechanism might play an important role, and increased frequencies of these diseases are observed in patients with selective IgA-deficiency27. An additional intriguing feature of IgA is its ability to neutralize viruses intracellularly. Whereas antibodies usually offer little protection against intracellular pathogens, IgA can intersect virus particles and interfere with virus replication or assembly when in transit through an infected epithelial cell. IgAvirus complexes might subsequently be excreted into the lumen (Fig. 3c). Evidence for this mechanism was initially observed using polarized cell monolayers, of which the apical surface was infected with Sendai virus. Addition of specific IgA anti-Sendai antibodies at the basolateral surface resulted in intracellular co-localization of IgA and Sendai virus3. IgA antibodies against rotavirus were effective in protecting mice only when given systemically, but not when presented at the luminal side of the intestinal tract, indicating that IgA transcytosis is required for viral inactivation in vivo28. In addition, HIV-specific dimeric IgA blocks infection of epithelial cells with HIV. Neutralization of virus was found to occur within apical recycling endosomes with subsequent recycling of immune complexes to the lumen29. Importantly, in a cohort study of Kenyan sex workers, 76% of HIV-resistant females expressed HIV-1-specific IgA in their genital tract, compared with 26% of infected females30. Furthermore, IgA from these so-called highly exposed, persistently seronegative individuals not only neutralizes HIV-1, but prevents its transcytosis across tight epithelial cell layers, supporting an important role for IgA in preventing sexual transmission of HIV-1 (Refs 31,32). Because no inflammation is triggered via the mechanisms mentioned above, IgA is considered a non-inflammatory antibody. This is further accentuated by the fact that IgA is a poor activator of complement. Although IgA has been shown to be capable of triggering the alternative complement pathway (under select conditions)33, IgA cannot bind C1q and hence is incapable of activating the classical pathway2. Recently, however, accumulating data support the capacity of IgA to trigger a plethora of

208

Review

TRENDS in Immunology Vol.22 No.4 April 2001

(a)
Lumen

(b)

(c)
Polymeric Ig receptor Dimeric IgA Secretory IgA

Epithelial cells N N N

Bacterium Bacterial toxins/enzymes Transcytosis IgA Transcytosis virus Environmental antigen

Lamina propria Plasma cell Plasma cell Plasma cell

Virus particle N Nucleus


TRENDS in Immunology

Fig. 3. Proposed functions of IgA in the mucosal area. Plasma cells in the lamina propria produce dimeric IgA (dIgA) that binds to the polymeric Ig receptor (pIgR), which is expressed on the basolateral membranes of epithelial cells. This complex is transported through epithelial cells to the lumen, where the pIgR is cleaved, releasing secretory IgA (SIgA). (a) SIgA shields the mucosa from bacterial penetration, agglutinates bacteria, interferes with their motility and neutralizes bacterial products. (b) Antigens in the lamina propria are bound by IgA and can be transported through epithelial cells via the same route as free dIgA, ridding the mucosa of excess antigens. (c) During transcytosis IgA can intercept virus antigens and interfere with viral synthesis and/or assembly, thus neutralizing viruses intracellularly.

inflammatory functions by interacting with the myeloid IgA receptor FcRI (Ref. 4).
Activatory properties of IgA and FcRI

FcRI is expressed on cells of the myeloid lineage, including neutrophils, monocytes, macrophages and eosinophils4. Several conflicting reports addressing the function of IgA receptors exist in the literature. Early reports demonstrated suppression of PMN chemotaxis by human myeloma IgA (Ref. 34). Together with studies in which IgA failed to induce phagocytosis by human PMNs, these findings led to the view of IgA receptors as anti-inflammatory molecules. However, in these experiments animal IgA, human SIgA or myeloma IgA was used. Weisbart et al. were among the first to document IgA-mediated phagocytosis by PMNs (Ref. 35) and, recently, extensive data were generated showing effective induction of phagocytosis, respiratory burst activity and release of inflammatory mediators and cytokines upon FcRI engagement4. Moreover, FcRI was shown to induce antibody-mediated tumor cell lysis by PMNs, monocytes and macrophages4,36. In fact, FcRI proved to represent the most effective leukocyte Fc receptor for initiation of CD20-targeted antibody therapy37. The capacity to lyse tumor cells was abolished in PMNs of FcRI Tg mice that were deficient in the 2 integrin Mac-1 (CR3; CD11b/CD18), which indicates that Mac-1 is essential for FcRI-mediated tumor cell killing19. Recent studies show that Mac-1/ PMNs cannot spread on antibody-coated tumor targets. Because impaired immunological synapse formation between
http://immunology.trends.com

FcRI/Mac-1/ PMNs and tumor cells was observed, extracellular lysis might be absent, owing to abnormal interactions between effector and target cells38. Furthermore, FcRI facilitates antigen presentation39, and FcRI expression was reported on monocyte-derived dendritic cells, which are located beneath the epithelium in vivo40. FcRI might, therefore, be involved in the capture of IgA immune complexes that arise from a diminished epithelial barrier, with subsequent processing and presentation to T cells. The ability of SIgA to trigger activatory functions is somewhat controversial. Although earlier reports suggested that SIgA induces phagocytosis, recent work clearly demonstrates that human serum IgA, but not human SIgA, initiates phagocytosis by either PMNs in vitro (Fig. 4), or Kupffer cells in vivo41. This is presumably owing to blockage of the FcRI-binding site located at or near the C2/C3 boundary on IgA by SC (Refs 25,42; G. Vidarsson et al., unpublished). However, SIgA can induce respiratory burst activity by PMNs (albeit less efficiently than serum IgA). Coating of SIgA on particles and/or surfaces too large to be phagocytosed might lead to a conformational change exposing the FcRI-binding site. SIgA might, therefore, play an important role in parasitic infections. This is further suggested by the ability of SIgA to trigger eosinophil degranulation. In fact, SIgA represents the most potent stimulus for eosinophil activation, which is owing (partly) to the presence of a unique receptor for SC on eosinophils. In addition, SIgA induces basophil degranulation in an FcRI-independent manner43. Basophils are active participants at sites of allergic inflammation, and the SIgA-induced release of inflammatory mediators might be implicated in the aggravation of allergic reactions.
Role of FcRI and IgA in immunity

Selective IgA deficiency represents the most common primary immune deficiency disease in humans,

Review

TRENDS in Immunology Vol.22 No.4 April 2001

209

Fig. 4. FcRI-expressing neutrophils phagocytose serum IgAopsonized Escherichia coli (a), but not SIgA-coated bacteria (b). Scale bar = 10 m. Modified, with permission, from Ref. 41.

affecting ~1:500 people. Although IgA deficiency has been associated with several diseases, including allergies, autoimmunity and recurrent infections of the upper respiratory tract, most individuals lacking IgA do not suffer from serious complications27. The relative importance of IgA in vivo is thus controversial. To investigate this in more detail, several animal models have been created. These include mice deficient in either the J-chain or pIgR, which prevents transport of IgA, and IgA-knockout mice26,44. Under laboratory settings these mice appear to be completely healthy, and challenge of IgA/ and IgA+/+ mice with influenza virus results in similar levels of pulmonary virus infection and mortality45. The absence of IgA in IgA/ mice, (a)
SIgA Bacterium

(b)

Mucosal sites

Mucosal sites

Gut To liver Serum IgA

Gut To liver

Kupffer cell

Hepatocyte

Liver

Liver
TRENDS in Immunology

Fig. 5. The role of IgA in mucosal immunity. Under physiological conditions (a) secretory IgA (SIgA) serves as the first line of defense by shielding the mucosa from bacterial penetration. SIgA, however, does not trigger deleterious inflammatory processes. Under pathological conditions (b), characterized by a defective mucosal barrier, pathogens can invade the underlying mucosal wall, and FcRI expression on Kupffer cells is induced by inflammatory mediators. Bacteria entering the liver via the circulation (and thus exposed to serum IgA) can then be filtered via (amongst others) FcRI-mediated phagocytosis by Kupffer cells, preventing septicemia and disease.

however, seems to be compensated by enhanced levels of IgM and IgG. If genetically normal mice are fed with chemically defined total parenteral nutrition, influenza-specific SIgA titers in the upper respiratory tract are reduced, whereas levels of influenza-specific IgG in serum remain unaffected. Thus, these mice show impaired mucosal immunity in the absence of compensating Igs. Challenge with influenza virus results in decreased nasal anti-influenza immunity in these mice, supporting a primary role for SIgA in protection46. The IgA-knockout model therefore allows the study of immunity in the absence of IgA, but might not provide information about the role of IgA in normal individuals. Furthermore, it should be noted that the systemic IgA system is substantially different in mice and man. Serum IgA is mostly monomeric in humans, and polymeric in mice. Clearance via the hepatobiliary route plays an important role in mice, but not in humans. Finally, although an Fc receptor for IgA has been identified on human myeloid cells (FcRI), no murine homolog has yet been defined. Two transgenic mouse models have been created to assess the role of FcRI in vivo. One model, in which the CD11b promoter was used, resulted in high human FcRI expression levels on monocytes and macrophages47. Another model was created with the use of a cosmid clone bearing the human FcRI gene. Like humans, these latter FcRI transgenic mice express the IgA receptor on cells of the myeloid lineage. Furthermore, regulation of FcRI expression mimics expression regulation in human cells, indicating that the FcRI gene is flanked by its own regulatory sequences19. Thus, the latter model closely parallels the human situation. Studying mice infected with Streptococcus pneumoniae showed that FcRI transgenic mice are protected against the development of pneumonia and sepsis by anti-S. pneumoniae IgA antibodies (G. Vidarsson and E. Saeland, unpublished). Furthermore, anti-Bordetella pertussis IgA decreased bacterial colonization in lungs of human FcRI transgenic mice, showing similar defense against infection with B. pertussis (S. Hellwig and A.B. van Spriel, unpublished). Although the precise mechanisms underlying protection in these bacterial models need to be addressed in more detail, the involvement of FcRI in IgA-mediated immunity is apparent. Another notable observation was the documentation of FcRI expression on Kupffer cells of transgenic mice, upon stimulation with cytokines such as granulocyte colony-stimulating factor or tumor necrosis factor (Ref. 41). Because Kupffer cells are essential for the maintenance of homeostasis by eliminating invasive bacteria that have entered via the gut, this finding directly links FcRI to mucosal immunity. Furthermore, in vivo FcRIexpressing Kupffer cells vigorously ingested E. coli

http://immunology.trends.com

210

Review

TRENDS in Immunology Vol.22 No.4 April 2001

Acknowledgements Owing to the broadness of the subject, combined with space limitations, coverage of all topics in IgA and FcRI biology was impossible. We apologize to all colleagues whose work could not be addressed and recommend Ref. 50 for further information.

that were opsonized with human serum IgA. By contrast, human SIgA could not initiate phagocytosis. Although it is not yet known whether this mechanism plays a role for other pathogens, in vitro experiments showed phagocytosis of human serum IgA-coated Staphylococcus aureus, Candida albicans, B. pertussis and S. pneumoniae by human or FcRI transgenic PMNs (Refs 48,49). In all cases, SIgA did not induce phagocytosis. Based on these results we propose the following model (Fig. 5). The primary role of SIgA might be to function as an antiseptic coating of the mucosa by preventing adherence and invasion of microorganisms (Fig. 5a). However, binding of antigens to SIgA does not initiate inflammatory processes, which qualifies SIgA as a noninflammatory antibody. Under pathological conditions, characterized by a defective mucosal barrier and production of inflammatory mediators, Kupffer cells express FcRI (Fig. 5b). Bacteria that have invaded the circulation (and are thus exposed to serum IgA), are subsequently phagocytosed by

FcRI-expressing Kupffer cells, hereby preventing septicemia and disease. Interactions between FcRI and serum IgA might therefore provide a second line of defense, designating serum IgA an inflammatory antibody.
Concluding remarks

There are still many questions concerning the role of IgA in immunity. Does the absence of serious complications in IgA-deficient patients imply that IgA is superfluous? Or are its functions of such importance that back-up mechanisms were developed to prevent lethality in the absence of IgA? The latter possibility is supported by both IgA-knockout mice and IgA-deficient individuals, in which increased levels of IgM and IgG are found. Furthermore, in IgA-deficient humans the severity of complications is associated with the absence of such compensatory mechanisms. Although further research is needed to solve these enigmas, it is clear that IgA can no longer be regarded merely as an antiseptic paint covering the mucosal walls.
extracellular domains. J. Exp. Med. 189, 17151722 Pleass, R.J. et al. (1999) Identification of residues in the CH2/CH3 domain interface of IgA essential for interaction with the human Fc receptor (FcR) CD89. J. Biol. Chem. 274, 2350823514 Johansen, F.E. et al. (1999) Absence of epithelial immunoglobulin A transport, with increased mucosal leakiness, in polymeric immunoglobulin receptor/secretory component-deficient mice. J. Exp. Med. 190, 915922 Burrows, P.D. and Cooper, M.D. (1997) IgAdeficiency. Adv. Immunol. 65, 245276 Burns, J.W. et al. (1996) Protective effect of rotavirus VP6-specific IgA monoclonal antibodies that lack neutralizing activity. Science 272, 104107 Bomsel, M. et al. (1998) Intracellular neutralization of HIV transcytosis across tight epithelial barriers by anti-HIV envelope protein dIgA or IgM. Immunity 9, 277287 Kaul, R. et al. (1999) HIV-1-specific mucosal IgA in a cohort of HIV-1-resistant Kenyan sex workers. AIDS 13, 2329 Devito, C. et al. (2000) Mucosal and plasma IgA from HIV-exposed seronegative individuals neutralize a primary HIV-1 isolate. AIDS 4, 19171920 Devito, C. et al. (2000) Mucosal and plasma IgA from HIV-1-exposed uninfected individuals inhibit HIV-1 transcytosis across human epithelial cells. J. Immunol. 165, 51705176 Janoff, E.N. et al. (1999) Killing of Streptococcus pneumoniae by capsular polysaccharide-specific polymeric IgA, complement, and phagocytes. J. Clin. Invest. 104, 11391147 Van Epps, D.E. and Williams, R.C., Jr (1976) Suppression of leukocyte chemotaxis by human IgA myeloma components. J. Exp. Med. 144, 12271242 Weisbart, R.H. et al. (1988) GM-CSF induces human neutrophil IgA-mediated phagocytosis by an IgA Fc receptor activation mechanism. Nature 332, 647648

References 1 Macpherson, A.J. et al. (2000) A primitive T cellindependent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science 288, 22222226 2 Kerr, M.A. (1990) The structure and function of human IgA. Biochem. J. 271, 285296 3 Mazanec, M.B. et al. (1992) Intracellular neutralization of virus by immunoglobulin A antibodies. Proc. Natl. Acad. Sci. U. S. A. 89, 69016905 4 Morton, H.C. et al. (1996) Structure and function of human IgA Fc receptors (FcR). Crit. Rev. Immunol. 16, 423440 5 Rifai, A. et al. (2000) The N-glycans determine the differential blood clearance and hepatic uptake of human immunoglobulin IgA1 and IgA2 isotypes. J. Exp. Med. 191, 21712182 6 Swenson, C.D. et al. (1998) Human T cell IgD receptors react with O-glycans on both human IgD and IgA1. Eur. J. Immunol. 28, 23662372 7 Boehm, M.K. et al. (1999) The Fab and Fc fragments of IgA1 exhibit a different arrangement from that in IgG: a study by X-ray and neutron solution scattering and homology modeling. J. Mol. Biol. 286, 14211447 8 Mostov, K.E. (1994) Transepithelial transport of immunoglobulins. Annu. Rev. Immunol. 12, 6384 9 Luton, F. and Mostov, K.E. (1999) Transduction of basolateral-to-apical signals across epithelial cells: ligand-stimulated transcytosis of the polymeric immunoglobulin receptor requires two signals. Mol. Biol. Cell 10, 14091427 10 Singer, K.L, and Mostov, K.E. (1998) Dimerization of the polymeric immunoglobulin receptor controls its transcytotic trafficking. Mol. Biol. Cell 9, 901915 11 Luton, F. et al. (1998) Role of tyrosine phosphorylation in ligand-induced regulation of transcytosis of the polymeric Ig receptor. Mol. Biol. Cell 9, 17871802

12 Luton, F. et al. (1999) The SRC family protein tyrosine kinase p62yes controls polymeric IgA transcytosis in vivo. Mol. Cell 4, 627632 13 Hexham, J.M. et al. (1999) A human immunoglobulin (Ig)A alpha3 domain motif directs polymeric Ig receptor-mediated secretion. J. Exp. Med. 189, 747752 14 Reth, M. (1989) Antigen receptor tail clue. Nature 338, 383384 15 Gulle, H. et al. (1998) Physical and functional association of FcR with protein tyrosine kinase Lyn. Blood 91, 383391 16 Park, R.K. et al. (1999) Role of Src in the modulation of multiple adaptor proteins in FcaRI oxidant signaling. Blood 94, 21122120 17 Lang, M.L. et al. (1999) -chain dependent recruitment of tyrosine kinases to membrane rafts by the human IgA receptor FcR. J. Immunol. 163, 53915398 18 Simons, K. and Ikonen, E. (1997) Functional rafts in cell membranes. Nature 387, 569572 19 Van Egmond, M. et al. (1999) Human immunoglobulin A receptor (FcRI, CD89) function in transgenic mice requires both FcR chain and CR3 (CD11b/CD18). Blood 93, 43874394 20 Launay, P. et al. (1999) Alternative endocytic pathway for immunoglobulin A Fc receptors (CD89) depends on the lack of FcR association and protects against degradation of bound ligand. J. Biol. Chem. 274, 72167225 21 Wines, B.D. et al. (1999) Identification of residues in the first domain of human Fc receptor essential for interaction with IgA. J. Immunol. 162, 21462153 22 Sondermann, P. et al. (2000) The 3.2- crystal structure of the human IgG1 Fc fragment-FcRIII complex. Nature 406, 267273 23 Garman, S.C. et al. (2000) Structure of the Fc fragment of human IgE bound to its high-affinity receptor FcRIa. Nature 406, 259266 24 Morton, H.C. et al. (1999) Immunoglobulinbinding sites of human FcRI (CD89) and bovine Fc2R are located in their membrane-distal

25

26

27 28

29

30

31

32

33

34

35

http://immunology.trends.com

Review

TRENDS in Immunology Vol.22 No.4 April 2001

211

36 Keler, T. et al. (2000) Differential effect of cytokine treatment on Fc receptor I- and Fc receptor Imediated tumor cytotoxicity by monocyte-derived macrophages. J. Immunol. 164, 57465752 37 Stockmeyer, B. et al. (2000) Triggering Fcreceptor I (CD89) recruits neutrophils as effector cells for CD20-directed antibody therapy. J. Immunol. 165, 59545961 38 Van Spriel, A.B. et al. Mac-1 (CD11b/CD18) is essential for Fc receptor-mediated neutrophil cytotoxicity and immunological synapse formation. Blood (in press) 39 Shen, L. et al. (2001) Presentation of ovalbumin internalized via the IgA Fc receptor (CD89) is enhanced through FcR chain signaling. Blood 97, 205213 40 Geissmann, F. et al. (2001) A subset of human dendritic cells expresses IgA Fc receptor (CD89), which mediates internalization and activation upon cross-linking by IgA complexes. J. Immunol. 166, 346352

41 Van Egmond, M. et al. (2000) FcRI-positive liver Kupffer cells: reappraisal of the function of immunoglobulin A in immunity. Nat. Med. 6, 680685 42 Crottet, P. and Corthesy, B. (1999) Mapping the interaction between murine IgA and murine secretory component carrying epitope substitutions reveals a role of domains II and III in covalent binding to IgA. J. Biol. Chem. 274, 3145631462 43 Iikura, M. et al. (1998) Secretory IgA induces degranulation of IL-3-primed basophils. J. Immunol. 161, 15101515 44 Harriman, G.R. et al. (1999) Targeted deletion of the IgA constant region in mice leads to IgA deficiency with alterations in expression of other Ig isotypes. J. Immunol. 162, 25212529 45 Mbawuike, I.N. et al. (1999) Mucosal immunity to influenza without IgA: an IgA knockout mouse model. J. Immunol. 162, 25302537

46 Renegar, K.B. et al. (2001) Impairment of mucosal immunity by total parenteral nutrition: requirement for IgA in murine nasotracheal anti-influenza immunity. J. Immunol. 166, 819825 47 Launay, P. et al. (2000) Fc receptor (CD89) mediates the development of immunoglobulin A (IgA) nephropathy (Bergers disease). Evidence for pathogenic soluble receptor-IgA complexes in patients and CD89 transgenic mice. J. Exp. Med. 191, 19992009 48 Van der Pol, L. et al. (2000) Pneumococcal capsular polysaccharide-specific IgA triggers efficient neutrophil effector functions via FcaRI (CD89). J. Infect. Dis. 182, 11391145 49 Van Spriel, A.B. et al. (1999) Effective phagocytosis and killing of Candida albicans via targeting FcRI (CD64) or FcRI (CD89) on neutrophils. J. Infect. Dis. 179, 661669 50 Ogra, P.L. et al., ed. (1999) In Mucosal Immunology (Chapters 1215), pp. 181251, Academic Press

VAP-1: an adhesin and an enzyme


Marko Salmi and Sirpa Jalkanen
Leukocyte extravasation from the blood into tissues is of paramount importance for normal immunosurveillance and in mounting adequate inflammatory responses. Multiple traditional adhesion molecules and chemoattractants on leukocytes and endothelial cells are involved in the emigration process. Vascular adhesion protein 1 (VAP-1) is a nonclassical inflammation-inducible endothelial molecule involved in leukocyte-subtypespecific rolling under physiological shear. Molecularly, VAP-1 belongs to a special class of cell surface amino oxidases. The enzymatic reaction itself and the biologically active end products can potentially regulate the adhesive status of the vessel wall. Thus, VAP-1 is an ectoenzyme that has inter-related adhesive and enzymatic functions in regulating physiological trafficking and inflammation.

events that lead to stable shear-resistant adhesion. This firm adhesion and the final diapedesis through the vascular wall relies heavily on leukocyte integrins and endothelial members of the immunoglobulin superfamily. Nevertheless, the currently known molecules cannot explain the full diversity, specificity and mechanics of immune cell trafficking, and newly discovered molecules are constantly refining the picture of extravasation.
Discovery of VAP-1

Marko Salmi* Sirpa Jalkanen MediCity Research Laboratory, Tykistkatu 6A, University of Turku, FIN-20520 Turku, Finland. *e-mail: marko.salmi@ utu.fi

In healthy adults there are some 10 billion lymphocytes and 15 billion granulocytes in the blood. Lymphocytes need to extravasate from the blood into secondary lymphoid tissues, such as lymph nodes, when patrolling the body in search of harmful antigens. Moreover, both lymphocytes and granulocytes must leave the blood to execute their effector functions in peripheral tissues during the defence reaction. Leukocytes normally emigrate from the blood using a multistep adhesion cascade13. There is an overwhelming amount of information available as to how these cellular interactions are governed by molecules both on the leukocyte surface and on the luminal side of the endothelial cell (Fig. 1). Thus, selectins and their carbohydrate ligands mediate the initial tethering of blood-borne leukocytes and are critical in the ensuing rolling phase. Chemoattractants and their serpentine receptors appear crucial for subsequent triggering

In the late 1980s, the endothelial determinants that direct lymphocyte homing to inflamed joints (a hallmark of arthritic disorders) were analyzed. At that time, two endothelial molecules, mucosal and peripheral addressins, were thought to act as tissuespecific address codes for gut-associated lymphatic tissues and peripheral lymph nodes (PLNs), respectively1. It had been shown that lymphocyte binding to synovial vessels differs from binding to either gut or PLN vessels and that healthy synovial tissue lining the joint cavity is practically devoid of lymphocytes. Therefore, it was hypothesized that a unique endothelial addressin might be expressed in inflamed synovial vessels4. A panel of monoclonal antibodies (mAbs) was produced against purified synovial vessels from a joint of a patient suffering from rheumatoid arthritis. One mAb was identified, 1B2, that reacted with the synovial vessels5. Importantly, the same antibody diminished binding of human peripheral blood lymphocytes to postcapillary venules in inflamed synovial

http://immunology.trends.com 1471-4906/01/$ see front matter 2001 Elsevier Science Ltd. All rights reserved. PII: S1471-4906(01)01870-1

Вам также может понравиться