251
|
Santiago AF, Fernandes RM, Santos BP, Assis FA, Oliveira RP, Carvalho CR, Faria AMC. Role of mesenteric lymph nodes and aging in secretory IgA production in mice. Cell Immunol 2008; 253:5-10. [PMID: 18632091 DOI: 10.1016/j.cellimm.2008.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 05/21/2008] [Accepted: 06/04/2008] [Indexed: 12/12/2022]
Abstract
Although it is known that Peyer's patches are the major inductive site for S-IgA production and B1 cells contribute to half of the IgA plasma cells detected in the gut lamina propria, the type of contribution of mesenteric lymph nodes to the process is still unclear. Cytokines such as TGF-beta, IL-10, IL-4, IL-5, and IL-6, are required to promote IgA class switching and IgA synthesis. Aging-related alterations in T and B cells and in cytokine production are already known. Some reports have also proposed that S-IgA production might be altered in aged animals. Herein, we investigated the role of MLN and aging in S-IgA production. Two- to 18-month-old BALB/c mice were used to evaluate aging-related alterations and MLN were removed to study its role in S-IgA production. We found that MLN are important, although not essential for S-IgA production. In addition, we showed that production of IgA-related cytokines are well preserved in MLN but not in PP of aged mice and that S-IgA levels are not affected by aging. Our results suggest that MLN may play a complementary role in S-IgA production mostly in aged animals.
Collapse
Affiliation(s)
- A F Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | |
Collapse
|
252
|
Bock JM, Trask DK. Coblation-Assisted Lingual Tonsillectomy for Dysphagia Secondary to Tongue Base Hypertrophy. Ann Otol Rhinol Laryngol 2008; 117:506-9. [PMID: 18700425 DOI: 10.1177/000348940811700706] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objectives: Lingual tonsillar hypertrophy is an underappreciated cause of dysphagia and is believed to impede swallowing function by inhibition of laryngeal elevation and epiglottic inversion due to mechanical interference by bulky tongue base tissue. We present a case of severe dysphagia secondary to idiopathic tongue base hypertrophy that was treated with coblation lingual tonsillectomy and tongue base reduction. Methods: We report a case and discuss the relevant literature regarding tongue base hypertrophy and surgical interventions to treat the enlarged base of the tongue. Results: Symptoms of dysphagia and globus sensation and signs of decreased epiglottic inversion and laryngeal penetration improved markedly after surgical reduction of hypertrophied lingual tonsillar tissue using coblation. Preoperative and postoperative clinical imaging and radiographs are presented to show the reduction of tongue base size, correlated with the patient's improved clinical function. Conclusions: Coblation-assisted lingual tonsillectomy and tongue base reduction can successfully treat dysphagia secondary to tongue base hypertrophy.
Collapse
Affiliation(s)
- Jonathan M Bock
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
253
|
Distinctive homing profile of pathogen-specific activated lymphocytes in human urinary tract infection. Clin Immunol 2008; 128:427-34. [PMID: 18585960 DOI: 10.1016/j.clim.2008.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 03/24/2008] [Accepted: 05/06/2008] [Indexed: 11/23/2022]
Abstract
In contrast to other mucosal sites, information on migration/homing of lymphocytes activated in the human urinary tract is lacking. The expression of lymphocyte homing receptors (HR) on pathogen-specific antibody-secreting cells (ASC) originating from the urinary tract (patients with pyelonephritis, PN) was compared to that on antigen-specific ASC originating from the intestine (patients with gastroenteritis) or from a parenteral site (tetanus toxoid-immunized volunteers). In the PN group, 61% of ASC expressed the gut HR, alpha(4)beta(7,) 52% the peripheral lymph node HR, L-selectin, and 13% the skin HR, CLA. This homing profile of urinary tract-originating lymphocytes was found to differ from both of the two major vaccination routes, intestinal (less gut-targeting) or parenteral (more gut-targeting, less targeting to parenteral sites). This information on targeting of the immune response may prove useful when developing vaccines against urinary tract infection (UTI).
Collapse
|
254
|
Shimomura Y, Ogawa A, Kawada M, Sugimoto K, Mizoguchi E, Shi HN, Pillai S, Bhan AK, Mizoguchi A. A unique B2 B cell subset in the intestine. ACTA ACUST UNITED AC 2008; 205:1343-55. [PMID: 18519649 PMCID: PMC2413032 DOI: 10.1084/jem.20071572] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Over 80% of the body's activated B cells are located in mucosal sites, including the intestine. The intestine contains IgM+ B cells, but these cells have not been characterized phenotypically or in terms of their developmental origins. We describe a previously unidentified and unique subset of immunoglobulin M+ B cells that present with an AA4.1−CD21−CD23− major histocompatibility complex class IIbright surface phenotype and are characterized by a low frequency of somatic hypermutation and the potential ability to produce interleukin-12p70. This B cell subset resides within the normal mucosa of the large intestine and expands in response to inflammation. Some of these intestinal B cells originate from the AA4.1+ immature B2 cell pool in the steady state and are also recruited from the recirculating naive B cell pool in the context of intestinal inflammation. They develop in an antigen-independent and BAFF-dependent manner in the absence of T cell help. Expansion of these cells can be induced in the absence of the spleen and gut-associated lymphoid tissues. These results describe the existence of an alternative pathway of B cell maturation in the periphery that gives rise to a tissue-specific B cell subset.
Collapse
Affiliation(s)
- Yasuyo Shimomura
- Experimental Pathology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
255
|
Abstract
The immune system faces a considerable challenge in its efforts to maintain tissue homeostasis in the intestinal mucosa. It is constantly confronted with a large array of antigens, and has to prevent the dissemination and proliferation of potentially harmful agents while sparing the vital structures of the intestine from immune-mediated destruction. Complex interactions between the highly adapted effector cells and mechanisms of the innate and adaptive immune system generally prevent the luminal microflora from penetrating the intestinal mucosa and from spreading systemically. Non-haematopoietic cells critically contribute to the maintenance of local tissue homeostasis in an antigen-rich environment by producing protective factors (e.g. production of mucus by goblet cells, or secretion of microbicidal defensins by Paneth cells) and also through interactions with the adaptive and innate immune system (such as the production of chemotactic factors that lead to the selective recruitment of immune cell subsets). The complexity of the regulatory mechanisms that control the local immune response to luminal antigens is also reflected in the observation that mutations in immunologically relevant genes often lead to the development of uncontrolled inflammatory reactions in the microbially colonized intestine of experimental animals.
Collapse
Affiliation(s)
- Mirjam Schenk
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
256
|
Petri WA, Miller M, Binder HJ, Levine MM, Dillingham R, Guerrant RL. Enteric infections, diarrhea, and their impact on function and development. J Clin Invest 2008; 118:1277-90. [PMID: 18382740 DOI: 10.1172/jci34005] [Citation(s) in RCA: 294] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Enteric infections, with or without overt diarrhea, have profound effects on intestinal absorption, nutrition, and childhood development as well as on global mortality. Oral rehydration therapy has reduced the number of deaths from dehydration caused by infection with an enteric pathogen, but it has not changed the morbidity caused by such infections. This Review focuses on the interactions between enteric pathogens and human genetic determinants that alter intestinal function and inflammation and profoundly impair human health and development. We also discuss specific implications for novel approaches to interventions that are now opened by our rapidly growing molecular understanding.
Collapse
Affiliation(s)
- William A Petri
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | |
Collapse
|
257
|
Yuan L, Wen K, Azevedo MSP, Gonzalez AM, Zhang W, Saif LJ. Virus-specific intestinal IFN-gamma producing T cell responses induced by human rotavirus infection and vaccines are correlated with protection against rotavirus diarrhea in gnotobiotic pigs. Vaccine 2008; 26:3322-31. [PMID: 18456375 DOI: 10.1016/j.vaccine.2008.03.085] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 03/26/2008] [Accepted: 03/31/2008] [Indexed: 11/17/2022]
Abstract
We examined rotavirus-specific IFN-gamma producing CD4+, CD8+ and CD4+CD8+ T cell responses in gnotobiotic pigs infected with a virulent human rotavirus (VirHRV) or vaccinated with an attenuated (Att) HRV vaccine (AttHRV3x or AttHRV2x) or an AttHRV oral priming and 2/6-virus-like particle (VLP) intranasal boosting (AttHRV-2/6VLP) regimen. In VirHRV infected pigs, HRV-specific IFN-gamma producing T cells reside primarily in ileum. AttHRV-2/6VLP induced similar frequencies of intestinal IFN-gamma producing T cells as the VirHRV, whereas AttHRV3x or 2x vaccines were less effective. Protection rates against rotavirus diarrhea upon VirHRV challenge significantly correlated (r=0.97-1.0, p<0.005) with frequencies of intestinal IFN-gamma producing T cells, suggesting their role in protective immunity.
Collapse
Affiliation(s)
- Lijuan Yuan
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 44691, USA.
| | | | | | | | | | | |
Collapse
|
258
|
Novak N, Haberstok J, Bieber T, Allam JP. The immune privilege of the oral mucosa. Trends Mol Med 2008; 14:191-8. [PMID: 18396104 DOI: 10.1016/j.molmed.2008.03.001] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 02/25/2008] [Accepted: 03/04/2008] [Indexed: 01/22/2023]
Abstract
Despite high bacterial colonization and frequent allergen contact, acute inflammatory and allergic reactions are rarely seen in the oral mucosa. Therefore we assert that immune tolerance predominates at this site and antigen presenting cells, such as dendritic cells and different T cell subtypes, serve as key players in oral mucosal tolerance induction. In this article we describe the mechanisms that lead to tolerance induced in the oral mucosa and how they differ from tolerance induced in the lower gastrointestinal tract. Furthermore we discuss ways in which novel nonparenteral approaches for immune intervention, such as allergen-specific immunotherapy applied by way of the sublingual route, might be improved to target the tolerogenic properties of the sophisticated oral mucosal immune network.
Collapse
Affiliation(s)
- Natalija Novak
- Department of Dermatology and Allergy, University of Bonn, Sigmund-Freud-Str 25, 53105 Bonn, Germany.
| | | | | | | |
Collapse
|
259
|
Pabst R, Russell MW, Brandtzaeg P. Tissue distribution of lymphocytes and plasma cells and the role of the gut. Trends Immunol 2008; 29:206-8; author reply 209-10. [PMID: 18394963 DOI: 10.1016/j.it.2008.02.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 01/14/2008] [Accepted: 02/04/2008] [Indexed: 10/22/2022]
|
260
|
Hermsen JL, Gomez FE, Maeshima Y, Sano Y, Kang W, Kudsk KA. Decreased enteral stimulation alters mucosal immune chemokines. JPEN J Parenter Enteral Nutr 2008; 32:36-44. [PMID: 18165445 DOI: 10.1177/014860710803200136] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Migration of lymphocytes into and through the mucosal immune system depends upon adhesion molecules to attract circulating cells and chemokines to stimulate diapedesis into tissues. Decreased enteral stimulation significantly reduces mucosal addressin cellular adhesion molecule-1 (MAdCAM-1) levels, an adhesion molecule critical for homing of T and B cells to Peyer's patches (PP), which reduces PP and intestinal T and B cells. We studied the effect of type and route of nutrition on tissue specific chemokines in PP (CXCL-12, -13 and CCL-19, -20 and -21), small intestine (SI; CCL-20, -25 and -28) and lung (CXCL-12, CCL-28). METHODS Intravenously cannulated male Institute of Cancer Research (ICR) mice were randomized to chow or parenteral nutrition (PN) for 5 days. PP, SI, and lung chemokine mRNA levels were measured using real-time qRT-polymerase chain reaction, and analyzed semiquantitatively by the DeltaDeltaCt method. Protein levels were quantified using enzyme-linked immunosorbent assay (ELISA) techniques, and groups compared using Student's t-test. RESULTS PP CXCL13 protein significantly decreased, whereas CCL21 protein increased significantly in the parenterally fed group. Parenteral feeding significantly decreased SI CCL20 and CCL 25 protein levels. CCL28 decreased significantly in the SI and lung of intravenously fed animals. mRNA levels changed in the opposite direction (compared with protein) for all chemokines except CCL28. CONCLUSIONS Decreased enteral stimulation significantly alters key mucosal immune chemokine protein levels at multiple sites. In general, PN (and concomitant lack of enteral stimulation) results in decreased levels of chemokines that control lymphocyte migration within the mucosal immune system.
Collapse
Affiliation(s)
- Joshua L Hermsen
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53792-7375, USA
| | | | | | | | | | | |
Collapse
|
261
|
Bessa J, Schmitz N, Hinton HJ, Schwarz K, Jegerlehner A, Bachmann MF. Efficient induction of mucosal and systemic immune responses by virus-like particles administered intranasally: implications for vaccine design. Eur J Immunol 2008; 38:114-26. [PMID: 18081037 DOI: 10.1002/eji.200636959] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intranasal (i.n.) immunization aims to induce local as well as systemic immune responses. In the present study, we assessed a vaccine platform based on virus-like particles (VLP) derived from the RNA phage Qbeta for i.n. immunization. We found that both i.n. and subcutaneous (s.c.) administration of Qbeta-VLP elicited strong and comparable specific IgG responses in serum and lung. Surprisingly, both routes also induced high levels of specific IgA in serum. In contrast, only i.n. administration of Qbeta-VLP resulted in local IgA production in the lung. Efficient induction of B cell responses by i.n. administration of VLP was further supported by the presence of large numbers of germinal centers (GC) as well as memory B cells in the spleen and plasma cells in the bone marrow. Results obtained for the VLP itself could be extended to an antigen covalently attached to it. Specifically, i.n. immunization of mice with VLP displaying the influenza virus derived ectodomain of the M2 protein resulted in strong M2-specific antibody responses as well as anti-viral protection. In contrast, i.n. immunization with VLP displaying p33 peptide, the major CTL epitope of lymphocytic choriomeningitis virus, induced relatively inefficient cytotoxic T cell responses, resulting in low numbers of specific T cells and poor effector cell differentiation. Taken together, these results suggest that effective antibody-based vaccines are achievable by i.n. administration of Qbeta-VLP displaying specific antigens.
Collapse
Affiliation(s)
- Juliana Bessa
- Cytos Biotechnology AG, Zürich-Schlieren, Switzerland
| | | | | | | | | | | |
Collapse
|
262
|
Kadaoui KA, Corthésy B. Secretory IgA mediates bacterial translocation to dendritic cells in mouse Peyer's patches with restriction to mucosal compartment. THE JOURNAL OF IMMUNOLOGY 2008; 179:7751-7. [PMID: 18025221 DOI: 10.4049/jimmunol.179.11.7751] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In addition to fulfilling its function of immune exclusion at mucosal surfaces, secretory IgA (SIgA) Ab exhibits the striking feature to adhere selectively to M cells in the mouse and human intestinal Peyer's patches (PPs). Subsequent uptake drives the SIgA Ab to dendritic cells (DCs), which become partially activated. Using freshly isolated mouse DCs, we found that the interaction with SIgA was tissue and DC subtype dependent. Only DCs isolated from PPs and mesenteric lymph nodes interacted with the Ab. CD11c(+)CD11b(+) DCs internalized SIgA, while CD11c(+)CD19(+) DCs only bound SIgA on their surface, and no interaction occurred with CD11c(+)CD8alpha(+) DCs. We next examined whether SIgA could deliver a sizeable cargo to PP DCs in vivo by administering SIgA-Shigella flexneri immune complexes into a mouse ligated intestinal loop containing a PP. We found that such immune complexes entered the PPs and were internalized by subepithelial dome PP DCs, in contrast to S. flexneri alone that did not penetrate the intestinal epithelium in mice. Dissemination of intraepithelial S. flexneri delivered as immune complexes was limited to PPs and mesenteric lymph nodes. We propose that preexisting SIgA Abs associated with microbes contribute to mucosal defense by eliciting responses that prevent overreaction while maintaining productive immunity.
Collapse
Affiliation(s)
- Khalil A Kadaoui
- R&D Laboratory of the Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon, Switzerland
| | | |
Collapse
|
263
|
Abstract
The production of immunoglobulin A (IgA) in mammals exceeds all other isotypes, and it is mostly exported across mucous membranes. The discovery of IgA and the realization that it dominates humoral mucosal immunity, in contrast to the IgG dominance of the systemic immune system, was early evidence for the distinct nature of mucosal immunology. It is now clear that IgA can function in high-affinity modes for neutralization of toxins and pathogenic microbes, and as a low-affinity system to contain the dense commensal microbiota within the intestinal lumen. The basic map of induction of IgA B cells in the Peyer's patches, which then circulate through the lymph and bloodstream to seed the mucosa with precursors of plasma cells that produce dimeric IgA for export through the intestinal epithelium, has been known for more than 30 years. In this review, we discuss the mechanisms underlying selective IgA induction of mucosal B cells for IgA production and the immune geography of their homing characteristics. We also review the functionality of secretory IgA directed against both commensal organisms and pathogens.
Collapse
|
264
|
Brandtzaeg P, Kiyono H, Pabst R, Russell MW. Terminology: nomenclature of mucosa-associated lymphoid tissue. Mucosal Immunol 2008; 1:31-7. [PMID: 19079158 DOI: 10.1038/mi.2007.9] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Stimulation of mucosal immunity has great potential in vaccinology and immunotherapy. However, the mucosal immune system is more complex than the systemic counterpart, both in terms of anatomy (inductive and effector tissues) and effectors (cells and molecules). Therefore, immunologists entering this field need a precise terminology as a crucial means of communication. Abbreviations for mucosal immune-function molecules related to the secretory immunoglobulin A system were defined by the Society for Mucosal Immunolgy Nomenclature Committee in 1997, and are briefly recapitulated in this article. In addition, we recommend and justify standard nomenclature and abbreviations for discrete mucosal immune-cell compartments, belonging to, and beyond, mucosa-associated lymphoid tissue.
Collapse
Affiliation(s)
- P Brandtzaeg
- Department of Pathology, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | | | |
Collapse
|
265
|
Secretory Immunity Following Mutans Streptococcal Infection or Immunization. Curr Top Microbiol Immunol 2008; 319:131-56. [DOI: 10.1007/978-3-540-73900-5_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
266
|
Koskinen K, Nevalainen S, Karikoski M, Hänninen A, Jalkanen S, Salmi M. VAP-1-deficient mice display defects in mucosal immunity and antimicrobial responses: implications for antiadhesive applications. THE JOURNAL OF IMMUNOLOGY 2007; 179:6160-8. [PMID: 17947691 DOI: 10.4049/jimmunol.179.9.6160] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
VAP-1, an ecto-enzyme expressed on the surface of endothelial cells, is involved in leukocyte trafficking between the blood and tissues under physiological and pathological conditions. In this study, we used VAP-1-deficient mice to elucidate whether absence of VAP-1 alters the immune system under normal conditions and upon immunization and microbial challenge. We found that VAP-1-deficient mice display age-dependent paucity of lymphocytes, in the Peyer's patches of the gut. IgA concentration in serum was also found to be lower in VAP-1(-/-) animals than in wild-type mice. Although there were slightly less CD11a on B and T cells isolated from VAP-1-deficient mice than on those from wild-type mice, there were no differences in the expression of gut-homing-associated adhesion molecules or chemokine receptors. Because anti-VAP-1 therapies are being developed for clinical use to treat inflammation, we determined the effect of VAP-1 deletion on useful immune responses. Oral immunization with OVA showed defective T and B cell responses in VAP-1-deficient mice. Antimicrobial immune responses against Staphylococcus aureus and coxsackie B4 virus were also affected by the absence of VAP-1. Importantly, when the function of VAP-1 was acutely neutralized using small molecule enzyme inhibitors and anti-VAP-1 Abs rather than by gene deletion, no significant impairment in antimicrobial control was detected. In conclusion, VAP-1-deficient mice have mild deviations in the mucosal immune system and therapeutic targeting of VAP-1 does not appear to cause a generalized increase in the risk of infection.
Collapse
|
267
|
Mestecky J, Russell MW, Elson CO. Perspectives on mucosal vaccines: is mucosal tolerance a barrier? THE JOURNAL OF IMMUNOLOGY 2007; 179:5633-8. [PMID: 17947632 DOI: 10.4049/jimmunol.179.9.5633] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mucosal administration of Ags induces specific Abs in external secretions and systemic unresponsiveness termed oral or mucosal tolerance. The dominant response depends on the species studied, the nature, dose, frequency, route of Ag application, and the use of adjuvants. The temporal sequence of Ag exposure determines the quality of the ensuing immune response; although initial mucosal Ag exposure results in systemic T cell hyporesponsiveness, pre-existing systemic responses are refractory to the tolerizing effects of mucosal Ag encounter. Mucosal and systemic humoral responses may be induced concomitantly with diminished systemic T cell responses, thereby permitting Ab-mediated containment of mucosal Ags without stimulation of the systemic immune compartment. B cell Ig isotype switching and differentiation toward IgA production share common regulatory mechanisms with the suppression of T cells. Optimization of mucosal vaccination strategies has the potential for enhancing protective immune responses and suppressing systemic responses to autoantigens desirable for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Jiri Mestecky
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | | |
Collapse
|
268
|
Hystad ME, Myklebust JH, Bø TH, Sivertsen EA, Rian E, Forfang L, Munthe E, Rosenwald A, Chiorazzi M, Jonassen I, Staudt LM, Smeland EB. Characterization of early stages of human B cell development by gene expression profiling. THE JOURNAL OF IMMUNOLOGY 2007; 179:3662-71. [PMID: 17785802 DOI: 10.4049/jimmunol.179.6.3662] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have characterized several stages of normal human B cell development in adult bone marrow by gene expression profiling of hemopoietic stem cells, early B (E-B), pro-B, pre-B, and immature B cells, using RNA amplification and Lymphochip cDNA microarrays (n = 6). Hierarchical clustering of 758 differentially expressed genes clearly separated the five populations. We used gene sets to investigate the functional assignment of the differentially expressed genes. Genes involved in VDJ recombination as well as B lineage-associated transcription factors (TCF3 (E2A), EBF, BCL11A, and PAX5) were turned on in E-B cells, before acquisition of CD19. Several transcription factors with unknown roles in B lymphoid cells demonstrated interesting expression patterns, including ZCCHC7 and ZHX2. Compared with hemopoietic stem cells and pro-B cells, E-B cells had increased expression of 18 genes, and these included IGJ, IL1RAP, BCL2, and CD62L. In addition, E-B cells expressed T/NK lineage and myeloid-associated genes including CD2, NOTCH1, CD99, PECAM1, TNFSF13B, and MPO. Expression of key genes was confirmed at the protein level by FACS analysis. Several of these Ags were heterogeneously expressed, providing a basis for further subdivision of E-B cells. Altogether, these results provide new information regarding expression of genes in early stages of human B cell development.
Collapse
Affiliation(s)
- Marit E Hystad
- Department of Immunology, Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Centre, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
269
|
Goetz M, Atreya R, Ghalibafian M, Galle PR, Neurath MF. Exacerbation of ulcerative colitis after rituximab salvage therapy. Inflamm Bowel Dis 2007; 13:1365-8. [PMID: 17604367 DOI: 10.1002/ibd.20215] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND B-cells are considered to play a pathogenic role in human ulcerative colitis (UC) by producing autoantibodies that cause epithelial cell damage. Here we report on a patient with intractable UC who suffered from a severe exacerbation of UC after salvage therapy with rituximab, a B-cell-depleting anti-CD20-antibody. METHODS A 58-year-old patient with active long-standing UC and unresponsiveness or adverse events to mesalamine, corticosteroids, azathioprine, methotrexate, infliximab, leukapheresis, mycophenolate mofetil, and adalimumab received 375 mg/m(2) rituximab. RESULTS A severe exacerbation of UC activity was noted upon therapy that required hospitalization. Subsequent studies showed a complete depletion of CD20-positive mucosal B-cells associated with a suppression of local IL-10 production. CONCLUSIONS In contrast to rheumatoid arthritis patients, rituximab had deleterious effects in our UC patient by blocking IL-10 producing B-cells. Our data suggest an important anti- rather than proinflammatory role of B-cells in UC.
Collapse
|
270
|
Kato A, Schleimer RP. Beyond inflammation: airway epithelial cells are at the interface of innate and adaptive immunity. Curr Opin Immunol 2007; 19:711-20. [PMID: 17928212 DOI: 10.1016/j.coi.2007.08.004] [Citation(s) in RCA: 254] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 08/21/2007] [Accepted: 08/23/2007] [Indexed: 12/19/2022]
Abstract
It has become increasingly clear that airway epithelial cells are central participants in innate and adaptive immune responses as well as mucosal inflammation. Epithelial cells produce antimicrobial host defense molecules, proinflammatory cytokines and chemokines in response to activation via pathogen recognition receptors. Recruitment of immune cells including dendritic cells, T cells and B cells into the proximity of epithelium results in the enhancement of adaptive immunity through interactions with epithelial cells. Newly identified epithelial-derived cytokines, including TSLP, IL-33 and BAFF, help to shape the local accumulation and activation of Th2 responses and B cell immunoglobulin production. Epithelial cells are also downstream targets of molecules that activate IL-13R and EGFR and are responsible for mucus production in both protective immune responses and allergic airway inflammatory diseases. Improved understanding of epithelial immune and inflammatory responses will hopefully suggest new strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Atsushi Kato
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | |
Collapse
|
271
|
Naito T, Suda T, Suzuki K, Nakamura Y, Inui N, Sato J, Chida K, Nakamura H. Lung dendritic cells have a potent capability to induce production of immunoglobulin A. Am J Respir Cell Mol Biol 2007; 38:161-7. [PMID: 17709597 DOI: 10.1165/rcmb.2007-0237oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The mucosal immune system provides the first line of defense against inhaled pathogens in the lung. This system is largely mediated by immunoglobulin A (IgA) locally produced by plasma cells, which originate from homing IgA-committed B cells. It has not been determined what types of antigen-presenting cells (APCs) primarily induce B cell differentiation for IgA production in the lung. In addition, although mucosal dendritic cells (DCs) are functionally distinct from DCs in other tissues, it is unclear whether IgA-inducing capability differs between mucosal lung DCs (LDCs) and nonmucosal DCs. The present study was conducted to identify APCs principally responsible for IgA induction in the lung, and to determine potential differences in IgA-inducing capacity between LDCs and nonmucosal DCs. We measured immunoglobulin and cytokine production in a coculture system containing naive IgD(+) B cells, naive T cells from ovalbumin-specific T cell-receptor transgenic mice, and APCs including LDCs, alveolar macrophages (AMs), or spleen DCs (SDCs). LDCs induced significantly greater levels of IgA, IgG1, IL-6, and TGF-beta than AMs and SDCs, whereas no differences were found in the production of IgM or IgG2a. In addition, the IgA percentage of total class-switched immunoglobulin was highest in cocultures with LDCs (38.4%) when compared with those with AMs (15.1%) and SDCs (22.7%). Neutralizing TGF-beta, but not IL-6, significantly decreased IgA induction by LDCs and SDCs, but not by AMs. This study suggests that LDCs are the primary APCs introducing IgA to the lung, and have a more potent IgA-inducing capacity than nonmucosal DCs.
Collapse
Affiliation(s)
- Tateaki Naito
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine,1-20-1 Handayama, Higashiku, Hamamatsu, 431-3192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
272
|
Stoel M, Evenhuis WNH, Kroese FGM, Bos NA. Rat salivary gland reveals a more restricted IgA repertoire than ileum. Mol Immunol 2007; 45:719-27. [PMID: 17692918 DOI: 10.1016/j.molimm.2007.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 07/03/2007] [Indexed: 10/23/2022]
Abstract
Secretory IgA is the most abundantly produced Ig in different mucosal tissues, such as the gastrointestinal tract and the salivary glands. These mucosal tissues are considered to be part of the common mucosal immune system. The specificity and immunoglobulin (Ig) VH gene repertoire of the IgA producing cells of both tissues is still largely unknown. To investigate the diversity of the antibody repertoire of IgA producing cells at different mucosal effector sites, we analysed used Ig VH genes by H-CDR3 spectrotyping and VH gene sequencing of both ileum and salivary gland IgA producing cells of PVG rats. Both types of tissues showed a limited diversity for the two major VH gene families, J558 and PC7183. The salivary gland showed even less diversity than the ileum of the same rat. Cloning and sequencing of used IgA VH genes confirmed the very restricted usage of VH genes since multiple sets of clonally related sequences in both types of tissues were found. More clones were found in salivary gland than in ileum and both tissues did not have shared VDJ joining regions. IgA derived from salivary gland used germline or near germline VH genes, whereas the ileal VH genes contained more mutations. Furthermore, clonal evolution patterns from all analyzed VH gene sequences of the salivary gland IgA producing cells show mainly randomly acquired somatic mutations, in contrast to the clonal evolution patterns often observed as a consequence of affinity maturation in germinal center reactions in peripheral lymphoid organs and Peyer's patches. Our results imply that IgA producing cells in the salivary gland are neither induced at the same place nor selected in the same way as the IgA producing cells in the ileum. The function of the IgA secreted by salivary gland is very likely a first line of defense with (near) germline encoded IgA, whereas in the intestine the majority of utilized IgA VH genes show evidence of somatic hypermutation.
Collapse
Affiliation(s)
- Maaike Stoel
- Department of Cell Biology, Immunology Section, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | |
Collapse
|
273
|
Rahman A, Fahlgren A, Sitohy B, Baranov V, Zirakzadeh A, Hammarström S, Danielsson A, Hammarström ML. Beta-defensin production by human colonic plasma cells: a new look at plasma cells in ulcerative colitis. Inflamm Bowel Dis 2007; 13:847-55. [PMID: 17387677 DOI: 10.1002/ibd.20141] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Previously, we showed that colonic epithelium of ulcerative colitis (UC) patients expresses increased levels of mRNA for 3 antimicrobial peptides, human beta-defensin 2 (hBD-2), hBD-3, and hBD-4 compared to controls. METHODS Human colon mucosa was analyzed using double immunofluorescence staining, in situ hybridization, immunoelectron microscopy, and quantitative real-time reverse-transcriptase polymerase chain reaction (qRT-PCR) with specific antibodies and probes in the respective assays. RESULTS We demonstrate that lamina propria in colon from UC patients, Crohn's colitis patients, and controls contain cells that express hBD-2. These cells were identified as mature plasma cells by the highly specific CD138 marker, by their prominent IgA or IgG expression, and by their ultrastructural characteristics. By immunoelectron microscopy it was furthermore shown that the hBD-2 peptide was expressed in rough endoplasmic reticulum, the Golgi complex, and cytoplasmic vesicles, reflecting consecutive steps of synthesis and transport for secretion. Plasma cells were 2-3 times more abundant in UC colon than in control colon and Crohn's colitis. Moreover, plasma cells in UC colon expressed hBD-3 and hBD-4 mRNA. Additionally, hBD-2 mRNA expression was demonstrated in 3 out of 4 well-characterized plasma cell lines. CONCLUSIONS Mature colonic plasma cells can express multiple beta-defensins. In UC, defensin production by plasma cells is probably clinically relevant since plasma cells accumulate in large numbers between the distorted crypts and muscularis mucosae, first focally than diffusely, so as to protect against microbial attack.
Collapse
Affiliation(s)
- Arman Rahman
- Department of Clinical Microbiology, Immunology, Umeå University, Umeå, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
274
|
Brandtzaeg P. Induction of secretory immunity and memory at mucosal surfaces. Vaccine 2007; 25:5467-84. [PMID: 17227687 DOI: 10.1016/j.vaccine.2006.12.001] [Citation(s) in RCA: 346] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 11/08/2006] [Accepted: 12/01/2006] [Indexed: 11/27/2022]
Abstract
Mucosal epithelia comprise an extensive vulnerable barrier which is reinforced by numerous innate defence mechanisms cooperating intimately with adaptive immunity. Local generation of secretory IgA (SIgA) constitutes the largest humoral immune system of the body. Secretory antibodies function both by performing antigen exclusion at mucosal surfaces and by virus and endotoxin neutralization within epithelial cells without causing tissue damage. SIgA is thus persistently containing commensal bacteria outside the epithelial barrier but can also target invasion of pathogens and penetration of harmful antigens. Resistance to toxin-producing bacteria such as Vibrio cholerae and enterotoxigenic Escherichia coli appears to depend largely on SIgA, and so does herd protection against horizontal faecal-oral spread of enteric pathogens under naïve or immunized conditions--with a substantial innate impact both on cross-reactivity and memory. Like natural infections, live mucosal vaccines or adequate combinations of non-replicating vaccines and mucosal adjuvants, give rise not only to SIgA antibodies but also to longstanding serum IgG and IgA responses. However, there is considerably disparity with regard to migration of memory/effector cells from mucosal inductive sites to secretory effector sites and systemic immune organs. Also, although immunological memory is generated after mucosal priming, this may be masked by a self-limiting response protecting the inductive lymphoid tissue in the gut. The intranasal route of vaccine application targeting nasopharynx-associated lymphoid tissue may be more advantageous for certain infections, but only if successful stimulation is achieved without the use of toxic adjuvants that might reach the central nervous system. The degree of protection obtained after mucosal vaccination ranges from reduction of symptoms to complete inhibition of re-infection. In this scenario, it is often difficult to determine the relative importance of SIgA versus serum antibodies, but infection models in knockout mice strongly support the notion that SIgA exerts a decisive role in protection and cross-protection against a variety of infectious agents. Nevertheless, relatively few mucosal vaccines have been approved for human use, and more basic work is needed in vaccine and adjuvant design, including particulate or live-vectored combinations.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology, Institute and Department of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Centre, N-0027 Oslo, Norway.
| |
Collapse
|
275
|
Jarillo-Luna A, Rivera-Aguilar V, Garfias HR, Lara-Padilla E, Kormanovsky A, Campos-Rodríguez R. Effect of repeated restraint stress on the levels of intestinal IgA in mice. Psychoneuroendocrinology 2007; 32:681-92. [PMID: 17590521 DOI: 10.1016/j.psyneuen.2007.04.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2006] [Revised: 04/14/2007] [Accepted: 04/19/2007] [Indexed: 10/23/2022]
Abstract
The effects of restraint stress on the intestinal humoral immune system, particularly those about intestinal IgA production, have not been explored in detail. Thus, the purpose of this study was to assess the effect of restraint stress on the production and secretion of intestinal IgA as well as on the number of IgA+ cells in the intestinal lamina propria. The involvement of glucocorticoids and catecholamines were also evaluated. Mice were exposed to 1 or 4 h restraint stress for 4 d. The intestinal IgA concentration was quantified by ELISA and the number of IgA containing cells in the lamina propria was determined by immunohistochemistry. The effects of restraint were also analyzed in mice submitted to different procedures: adrenalectomy, chemical sympathectomy, treatment with a glucocorticoid antagonist (RU486), dexamethasone and epinephrine. The main findings were that (1) chronic restraint-stress reduced the intestinal IgA concentration without changing the number of IgA+ cells in lamina propria; (2) adrenalectomy restored the production of IgA in stressed mice; (3) RU486 and chemical sympathectomy partially blocked the decrease in intestinal IgA in stressed mice; and (4) pharmacological doses of dexamethasone and epinephrine significantly reduced the intestinal IgA concentration and the number of IgA+ cells. The restraint stress probably reduced the intestinal IgA concentration through the effects of glucocorticoids and catecholamines.
Collapse
Affiliation(s)
- Adriana Jarillo-Luna
- Departamento de Morfología, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, CP 11340, México, DF
| | | | | | | | | | | |
Collapse
|
276
|
Abstract
The internal surfaces of the human body are covered by distinct types of epithelial cells and mucus-secreting cells. The mucosal surfaces serve many vital functions, such as respiration (nasal passage and lung), absorption (gastrointestinal tract), excretion (lung, urinary tract, large intestine), and reproduction (reproductive tract). In performing these functions, the host is inevitably exposed to environmental antigens, food particles, commensal flora, and pathogens. Mucosal surfaces contain specialized dendritic cells (DCs) capable of sensing these external stimuli and mounting appropriate local responses depending on the nature of the elements they encounter. In the absence of pathogens, mucosal DCs either ignore the antigen or induce regulatory responses. Upon recognition of microorganisms that invade the mucosal barrier, mucosal DCs mount robust protective immunity. This review highlights progress in our understanding of how mucosal DCs process external information and direct appropriate responses by mobilizing various cells of the innate and adaptive immune systems to achieve homeostasis and protection.
Collapse
Affiliation(s)
- Akiko Iwasaki
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| |
Collapse
|
277
|
Budec M, Koko V, Todorović V, Marković D, Postić M, Drndarević N, Spasić A, Mitrović O. Possible mechanism of acute effect of ethanol on intestinal IgA expression in rat. Int Immunopharmacol 2007; 7:858-63. [PMID: 17466919 DOI: 10.1016/j.intimp.2007.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Revised: 02/14/2007] [Accepted: 02/14/2007] [Indexed: 12/23/2022]
Abstract
The purpose of this study was to investigate the possible mechanism of acute effect of ethanol on IgA expression in rat intestine. To this end, adult female Wistar rats showing diestrus day 1 were treated with (a) ethanol (2 or 4 g/kg, i.p.); (b) N omega-nitro-L-arginine-methyl ester (L-NAME), which inhibits the activity of all isoforms of nitric oxide synthase, (30 mg/kg, s.c.) followed by ethanol 3 h later; and (c) L-NAME (30 mg/kg, s.c.) followed by saline 3 h later. Saline-injected and untreated rats were used as controls. The animals were sacrificed 0.5 h after ethanol administration. Intestinal expression of IgA was evaluated by both immunohistochemistry and Western immunoblotting. Morphometric analysis showed that acute ethanol treatment increased the number of IgA-immunoreactive cells in a dose-dependent manner. Pretreatment with L-NAME abolished this action of alcohol. Injection of L-NAME followed by saline had no influence on the number of IgA+cells. The results, obtained by Western immunoblotting, paralleled our immunohistochemical findings. Taken together, these data suggest that acute effect of ethanol on intestinal IgA might be mediated by endogenous nitric oxide.
Collapse
Affiliation(s)
- Mirela Budec
- University of Belgrade, Institute for Medical Research, Belgrade, Serbia.
| | | | | | | | | | | | | | | |
Collapse
|
278
|
Abstract
Two major antibody classes operate in saliva: secretory IgA (SIgA) and IgG. The former is synthesized as dimeric IgA by plasma cells (PCs) in salivary glands and is exported by the polymeric Ig receptor (pIgR). Most IgG in saliva is derived from serum (mainly via gingival crevices), although some is locally produced. Gut-associated lymphoid tissue (GALT) and nasopharynx-associated lymphoid tissue (NALT) do not contribute equally to mucosal PCs throughout the body. Thus, enteric immunostimulation is an inadequate mode of stimulating salivary IgA antibodies, which are poorly associated with the intestinal SIgA response, for instance after enteric cholera vaccination. Nevertheless, the IgA response in submandibular/sublingual glands is better related to B cell induction in GALT than the parotid response. Such disparity is suggested by the elevated levels of IgA in submandibular secretions of AIDS patients, paralleling their highly upregulated intestinal IgA system. Moreover, in patients with active celiac disease, IgA antibodies to disease-precipitating gliadin are reliably represented in whole saliva but not in parotid secretion. Parotid SIgA may be more consistently linked to immune induction in palatine tonsils and adenoids (human NALT), as supported by the homing molecule profile of NALT-derived B cell blasts. Also several other variables influence the levels of antibodies in oral secretions. These include difficulties with reproducibility and standardization of immunoassays, the impact of flow rate, acute or chronic stress, protein loss during sample handling, and uncontrolled admixture of serum-derived IgG and monomeric IgA. Despite such problems, saliva remains an interesting biological fluid with great scientific and clinical potentials.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology, Department and Institute of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Center, Norway.
| |
Collapse
|
279
|
Hinkula J. Clarification of how HIV-1 DNA and protein immunizations may be better used to obtain HIV-1-specific mucosal and systemic immunity. Expert Rev Vaccines 2007; 6:203-12. [PMID: 17408370 DOI: 10.1586/14760584.6.2.203] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
More focused research on a mucosal HIV-1 vaccine is needed urgently. An increasing amount of collected data, using heterologous multimodality prime-booster strategies, suggest that an efficient and protective HIV-1 vaccine must generate broad, long-lasting HIV-specific CD8(+) cytotoxic T-lymphocyte and neutralizing antibody responses. In the mucosa, these responses would be most effective if a preferential stimulus of HIV-1 neutralizing secretory immunoglobulin A and G were obtained. The attractive property of mucosal immunization is the obtained mucosal and systemic immunity, whereas systemic immunization induces a more limited immunity, predominantly in systemic sites. These objectives will require new vaccine regimens, such as multiclade HIV DNA and protein vaccines (nef, tat, gag and env expressed in DNA plasmids) delivered onto mucosal surfaces with needle-free delivery methods, such as nasal drop, as well as oral and rectal/vaginal delivery, and should merit clinical trials.
Collapse
Affiliation(s)
- Jorma Hinkula
- Department of Molecular Virology, Linkoping University, Linkoping, Sweden.
| |
Collapse
|
280
|
Abstract
Traditionally, the function of immunoglobulins A (IgA), the major type of secreted antibodies, has been thought to be restricted to binding antigens outside the epithelium basal membrane. Therefore, effector mechanisms eliminating IgA-opsonized targets have not been investigated so far. However, some indirect observations of infectious agents penetrating into tissues and blood from the environment suggest such mechanisms (analogous to IgG/IgM-dependent activation of complement and natural killers). In the present review, we examine details of IgA structure that might contribute to elucidation of IgA-dependent effector functions in human and animal immunity. Special attention is given to a putative transduction of signal about antigen binding in the active center of IgA from the Fab- to the Fc-superdomain via intramolecular conformational rearrangements. Different structure of the IgA subclasses (IgA1 and IgA2) is examined taking into account probable divergence of their functions in immune response.
Collapse
Affiliation(s)
- T N Kazeeva
- Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow 119071, Russia
| | | |
Collapse
|
281
|
Corthésy B. Roundtrip ticket for secretory IgA: role in mucosal homeostasis? THE JOURNAL OF IMMUNOLOGY 2007; 178:27-32. [PMID: 17182536 DOI: 10.4049/jimmunol.178.1.27] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
An important activity of mucosal surfaces is the production of Ab referred to as secretory IgA (SIgA). SIgA serves as the first line of defense against microorganisms through a mechanism called immune exclusion. In addition, SIgA adheres selectively to M cells in intestinal Peyer's patches, thus mediating the transepithelial transport of the Ab molecule from the intestinal lumen to underlying gut-associated organized lymphoid tissue. In Peyer's patches, SIgA binds and is internalized by dendritic cells in the subepithelial dome region. When used as carrier for Ags in oral immunization, SIgA induces mucosal and systemic responses associated with production of anti-inflammatory cytokines and limits activation of dendritic cells. In terms of humoral immunity at mucosal surfaces, SIgA appears thus to combine properties of a neutralizing agent (immune exclusion) and of a mucosal immunopotentiator inducing effector immune responses in a noninflammatory context favorable to preserve local homeostasis of the gastrointestinal tract.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory of the Division of Immunology and Allergy, State University Hospital (Centré Hospitalìer Universitaire Vandois), Rue du Bugnon, BH 19-650, 1011 Lausanne, Switzerland.
| |
Collapse
|
282
|
Källenius G, Pawlowski A, Brandtzaeg P, Svenson S. Should a new tuberculosis vaccine be administered intranasally? Tuberculosis (Edinb) 2007; 87:257-66. [PMID: 17321797 DOI: 10.1016/j.tube.2006.12.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Revised: 12/14/2006] [Accepted: 12/21/2006] [Indexed: 12/22/2022]
Abstract
Most of the world's population is vaccinated with the only available vaccine against tuberculosis (TB), the Bacillus Calmette-Guérin (BCG) vaccine that was developed almost a century ago. Despite the wide coverage of the BCG vaccine, there are great variations in protective efficacy among different study populations. BCG vaccination protects against childhood forms of TB, but this immunity wanes with age, resulting in none, or insufficient, protection against adult pulmonary TB (PTB). PTB is the major disease manifestation of TB in adults and it causes death at the most productive age, further adding to poverty in already impoverished countries. Therefore, new more effective vaccines and novel immunisation strategies are urgently needed. The most common route of TB is by inhalation of tubercle bacilli leading to the establishment of a primary infection in the lung. Immunising through the nasal mucosal surface should therefore have advantage over other routes, as such vaccine administration elicits protective immune responses also in the lung, i.e. at the site of primary infection. Several new TB-vaccine candidates have been evaluated for their protective efficacy in animal models using the mucosal route of immunisation. In formulating such vaccines, the adjuvants and delivery systems are crucially important.
Collapse
Affiliation(s)
- Gunilla Källenius
- Department of Bacteriology, Swedish Institute for Infectious Disease Control, 17182 Solna, Sweden.
| | | | | | | |
Collapse
|
283
|
Nagasawa T, Kiji M, Yashiro R, Hormdee D, Lu H, Kunze M, Suda T, Koshy G, Kobayashi H, Oda S, Nitta H, Ishikawa I. Roles of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin in periodontal health and disease. Periodontol 2000 2007; 43:65-84. [PMID: 17214836 DOI: 10.1111/j.1600-0757.2006.00185.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Toshiyuki Nagasawa
- Division of Periodontology, Department of Hard Tissue Engineering, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
284
|
Schmidt LD, Xie Y, Lyte M, Vulchanova L, Brown DR. Autonomic neurotransmitters modulate immunoglobulin A secretion in porcine colonic mucosa. J Neuroimmunol 2007; 185:20-8. [PMID: 17320195 PMCID: PMC1913560 DOI: 10.1016/j.jneuroim.2006.10.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 10/18/2006] [Accepted: 10/18/2006] [Indexed: 10/23/2022]
Abstract
Secretory immunoglobulin A (sIgA) plays a crucial role in mucosal surface defense. We tested the hypothesis that colonic sIgA secretion is under enteric neural control. Immunohistochemistry of the porcine distal colonic mucosa revealed presumptive cholinergic and adrenergic nerve fibers apposed to secretory component (SC)-positive crypt epithelial cells and neighboring IgA(+) plasmacytes. The cholinomimetic drug carbamylcholine elicited rapid, atropine-sensitive IgA secretion into the luminal fluid bathing mucosal explants mounted in Ussing chambers. The adrenergic receptor agonist norepinephrine also increased IgA secretion, an action inhibited by phentolamine. These effects were independent of agonist-induced anion secretion. In Western blots of luminal fluid, both agonists increased the density of protein bands co-immunoreactive for IgA and SC. Mucosal exposure to enterohemorrhagic Escherichia coli did not affect IgA secretion, and carbamylcholine treatment did not affect mucosal adherence of this enteropathogen. Acetylcholine and norepinephrine, acting respectively through muscarinic cholinergic and alpha-adrenergic receptors in the colonic mucosa, stimulate sIgA secretion and may enhance mucosal defense in vivo.
Collapse
Affiliation(s)
- Lisa D. Schmidt
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1988 Fitch Avenue, St. Paul, Minnesota 55108-6010
| | - Yonghong Xie
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1988 Fitch Avenue, St. Paul, Minnesota 55108-6010
| | - Mark Lyte
- Department of Pharmacy Practice, Texas Tech University Health Sciences Center, 3601 Fourth Street, MS 8162, Lubbock, Texas 79430
| | - Lucy Vulchanova
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1988 Fitch Avenue, St. Paul, Minnesota 55108-6010
| | - David R. Brown
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1988 Fitch Avenue, St. Paul, Minnesota 55108-6010
- Corresponding author: David R. Brown, Ph.D., Department of Veterinary and Biomedical Sciences, University of Minnesota, 1988 Fitch Avenue, St. Paul, Minnesota 55108-6010, Telephone: (612) 624-0713; FAX: (612) 625-0204; E-mail:
| |
Collapse
|
285
|
Braathen R, Hohman VS, Brandtzaeg P, Johansen FE. Secretory Antibody Formation: Conserved Binding Interactions between J Chain and Polymeric Ig Receptor from Humans and Amphibians. THE JOURNAL OF IMMUNOLOGY 2007; 178:1589-97. [PMID: 17237408 DOI: 10.4049/jimmunol.178.3.1589] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abs of the secretory Ig (SIg) system reinforce numerous innate defense mechanisms to protect the mucosal surfaces against microbial penetration. SIgs are generated by a unique cooperation between two distinct cell types: plasma cells that produce polymers of IgA or IgM (collectively called pIgs) and polymeric Ig receptor (pIgR)-expressing secretory epithelial cells that mediate export of the pIgs to the lumen. Apical delivery of SIgs occurs by cleavage of the pIgR to release its extracellular part as a pIg-bound secretory component, whereas free secretory components are derived from an unoccupied receptor. The joining chain (J chain) is crucial in pIg/SIg formation because it serves to polymerize Igs and endows them with a binding site for the pIgR. In this study, we show that the J chain from divergent tetrapods including mammals, birds, and amphibians efficiently induced polymerization of human IgA, whereas the J chain from nurse shark (a lower vertebrate) did not. Correctly assembled polymers showed high affinity to human pIgR. Sequence analysis of the J chain identified two regions, conserved only in tetrapods, which by mutational analysis were found essential for pIgA-pIgR complexing. Furthermore, we isolated and characterized pIgR from the amphibian Xenopus laevis and demonstrated that its pIg binding domain showed high affinity to human pIgA. These results showed that the functional site of interaction between pIgR, J chain and Ig H chains is conserved in these species and suggests that SIgs originated in an ancestor common to tetrapods.
Collapse
Affiliation(s)
- Ranveig Braathen
- Laboratory for Immunohistochemistry and Immunopathology, Institute of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway
| | | | | | | |
Collapse
|
286
|
Bergqvist P, Gärdby E, Stensson A, Bemark M, Lycke NY. Gut IgA class switch recombination in the absence of CD40 does not occur in the lamina propria and is independent of germinal centers. THE JOURNAL OF IMMUNOLOGY 2007; 177:7772-83. [PMID: 17114448 DOI: 10.4049/jimmunol.177.11.7772] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Conflicting findings have recently been presented as to the sites and sources of B cells that undergo class switch recombination (CSR) to IgA in the gut. In this study we provide compelling evidence in CD40(-/-) mice demonstrating that IgA CSR can be independent of CD40 signaling and germinal center formation and does not occur in the gut lamina propria (LP) itself. We found that CD40(-/-) mice had near normal levels of gut total IgA despite lacking germinal centers and completely failing to raise specific responses against the T cell-dependent Ags cholera toxin and keyhole limpet hemocyanin. The Peyer's patches in CD40(-/-) mice expressed unexpectedly high levels of activation-induced cytidine deaminase mRNA and germline alpha transcripts, but few postswitch circular DNA transcripts, arguing against significant IgA CSR. Moreover and more surprisingly, wild-type mice exhibited no to low IgA CSR in mesenteric lymph nodes or isolated lymphoid follicles. Importantly, both strains failed to demonstrate any of the molecular markers for IgA CSR in the gut LP itself. Whereas all of the classical sites for IgA CSR in the GALT in CD40(-/-) mice appeared severely compromised for IgA CSR, B cells in the peritoneal cavity demonstrated the expression of activation-induced cytidine deaminase mRNA comparable to that of wild-type mice. However, peritoneal cavity B cells in both strains expressed intermediate levels of the germinal center marker GL7 and exhibited no germline alpha transcripts, and only three of 51 mice analyzed showed the presence of postswitch circular DNA transcripts. Taken together, these findings strongly argue for alternative inductive sites for gut IgA CSR against T cell-independent Ags outside of the GALT and the nonorganized LP.
Collapse
Affiliation(s)
- Peter Bergqvist
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, Göteborg University, 405 30 Göteborg, Sweden
| | | | | | | | | |
Collapse
|
287
|
Kurtz CC, Carey HV. Seasonal changes in the intestinal immune system of hibernating ground squirrels. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2007; 31:415-28. [PMID: 16930701 DOI: 10.1016/j.dci.2006.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 06/23/2006] [Accepted: 07/03/2006] [Indexed: 05/06/2023]
Abstract
Hibernation is associated with a prolonged fast (5-8 mo) which has the potential to affect intestinal immunity. We examined several aspects of the intestinal immune system in summer (non-hibernating) and hibernating ground squirrels. Peyer's patches were largely unaffected by hibernation, but numbers of intraepithelial lymphocytes (IEL) and lamina propria leukocytes (LPL) were greater in hibernators compared with summer. Hibernator IEL were less mature as demonstrated by low numbers of cells expressing activation-associated markers and co-receptors. Compared with summer, the percentage of B cells was higher and percentage of T cells was lower in the hibernator LPL. Hibernation was associated with greater mucosal levels of IFN-gamma, TNF-alpha, IL-10 and IL-4, but IL-6 and TGF-beta were unchanged. Mucosal IgA levels were greater in entrance and torpid hibernators compared with summer. The results suggest that modifications of the intestinal immune system during hibernation may help preserve gut integrity throughout the winter fast.
Collapse
Affiliation(s)
- Courtney C Kurtz
- Department of Comparative Biosciences, University of Wisconsin, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI 53706, USA
| | | |
Collapse
|
288
|
Kaminski DA, Stavnezer J. Enhanced IgA class switching in marginal zone and B1 B cells relative to follicular/B2 B cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:6025-9. [PMID: 17056527 DOI: 10.4049/jimmunol.177.9.6025] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mouse splenic marginal zone (MZ) B cells and B1 B cells enriched in the peritoneal cavity respond preferentially to T cell-independent Ags compared with follicular (FO)/B2 B cells. Despite the differential responses of B cell subsets to various stimuli, and despite the need for multiple stimuli to induce IgA class switching, the relative contribution of B cell subpopulations to IgA production is unknown. By culturing purified B cell populations, we find that MZ and peritoneal B1 cells switch more readily to IgA than do splenic FO or peritoneal B2 cells in BLyS/LPS/TGF-beta. Addition of IL-4, IL-5, and anti-IgD dextran to the cultures enhances IgA switching in FO/B2 and MZ B cells to a similar frequency, but this treatment suppresses IgA class switching in B1 cells. Thus, IgA switching differs among purified B cell subsets, suggesting that individual B cell populations could contribute differentially to IgA expression in vivo, depending on available stimuli.
Collapse
Affiliation(s)
- Denise A Kaminski
- Immunology and Virology Program, Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | |
Collapse
|
289
|
Carlsen HS, Yamanaka T, Scott H, Rugtveit J, Brandtzaeg P. The proportion of CD40+ mucosal macrophages is increased in inflammatory bowel disease whereas CD40 ligand (CD154)+ T cells are relatively decreased, suggesting differential modulation of these costimulatory molecules in human gut lamina propria. Inflamm Bowel Dis 2006; 12:1013-24. [PMID: 17075342 DOI: 10.1097/01.mib.0000234135.43336.72] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Signal transduction through binding of CD40 on antigen-presenting cells and CD40 ligand (CD154) on T cells appears to be crucial for mutual cellular activation. Antibodies aimed at blocking the CD40-CD154 costimulatory pathway dampen the severity of experimental colitis. To elucidate the microanatomical basis for signaling through this costimulatory pathway in human inflammatory bowel disease, we studied in situ the cellular distribution of these 2 molecules on lamina propria macrophages and T cells, respectively. METHODS Colonic specimens from 8 patients with ulcerative colitis and 8 with Crohn's disease, 8 small bowel specimens of Crohn's disease, and histologically normal control samples (6 from colon and 6 from small bowel) were included. Multicolor immunofluorescence in situ staining was performed to determine the percentage of subepithelial macrophages expressing CD40 and that of lamina propria T cells expressing CD154 while avoiding cells in lymphoid aggregates. RESULTS The proportion of subepithelial CD40CD68 macrophages was significantly increased in normal colon compared with normal small bowel and showed further elevation in both colon and small bowel afflicted with inflammatory bowel disease. In addition, on a per-CD68-cell basis, CD40 expression was significantly increased in severely inflamed compared with moderately inflamed colonic specimens. Conversely, the proportion of CD154 T cells was similar in colon and small bowel, and interestingly, it was significantly reduced in colonic inflammatory bowel disease. CONCLUSIONS Our findings suggested that modulation of CD40 expression by subepithelial macrophages and CD154 by lamina propria T cells is inversely modulated in the human gut.
Collapse
Affiliation(s)
- Hege S Carlsen
- Laboratory for Immunohistochemistry and Immunopathology, Department of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway.
| | | | | | | | | |
Collapse
|
290
|
Yoshida M, Masuda A, Kuo TT, Kobayashi K, Claypool SM, Takagawa T, Kutsumi H, Azuma T, Lencer WI, Blumberg RS. IgG transport across mucosal barriers by neonatal Fc receptor for IgG and mucosal immunity. ACTA ACUST UNITED AC 2006; 28:397-403. [PMID: 17051393 DOI: 10.1007/s00281-006-0054-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Accepted: 10/03/2006] [Indexed: 12/21/2022]
Abstract
Mucosal secretions of the human gastrointestinal, respiratory, and genital tracts contain significant quantities of IgG. The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. The FcRn can then recycle the IgG/antigen complex back across the intestinal barrier into the lamina propria for processing by dendritic cells and presentation to CD4(+) T cells in regional organized lymphoid structures. FcRn, through its ability to secrete and absorb IgG, thus integrates luminal antigen encounters with systemic immune compartments and, as such, provides essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Frontier Medical Science in Gastroenterology, ICMRT, Kobe University School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
291
|
Yoshida M, Kobayashi K, Kuo TT, Bry L, Glickman JN, Claypool SM, Kaser A, Nagaishi T, Higgins DE, Mizoguchi E, Wakatsuki Y, Roopenian DC, Mizoguchi A, Lencer WI, Blumberg RS. Neonatal Fc receptor for IgG regulates mucosal immune responses to luminal bacteria. J Clin Invest 2006; 116:2142-2151. [PMID: 16841095 PMCID: PMC1501111 DOI: 10.1172/jci27821] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Accepted: 05/16/2006] [Indexed: 01/22/2023] Open
Abstract
The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. Selective expression of FcRn in the epithelium is shown here to be associated with secretion of IgG into the lumen that allows for defense against an epithelium-associated pathogen (Citrobacter rodentium). This pathway of host resistance to a bacterial pathogen as mediated by FcRn involves retrieval of bacterial antigens from the lumen and initiation of adaptive immune responses in regional lymphoid structures. Epithelial-associated FcRn, through its ability to secrete and absorb IgG, may thus integrate luminal antigen encounters with systemic immune compartments and as such provide essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Kanna Kobayashi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Timothy T. Kuo
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Lynn Bry
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Jonathan N. Glickman
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Steven M. Claypool
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Arthur Kaser
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Takashi Nagaishi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Darren E. Higgins
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Emiko Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Yoshio Wakatsuki
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Derry C. Roopenian
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Atsushi Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Wayne I. Lencer
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| |
Collapse
|
292
|
Wrackmeyer U, Hansen GH, Seya T, Danielsen EM. Intelectin: a novel lipid raft-associated protein in the enterocyte brush border. Biochemistry 2006; 45:9188-97. [PMID: 16866365 DOI: 10.1021/bi060570x] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intelectin is a mammalian Ca2+-dependent, D-galactosyl-specific lectin expressed in Paneth and goblet cells of the small intestine and proposed to serve a protective role in the innate immune response to parasite infection. In addition, it is structurally identical to the intestinal lactoferrin receptor known to reside in the enterocyte brush border. To clarify this apparent discrepancy with regard to localization, the aim of this work was to study the cellular and subcellular distribution of small intestinal intelectin by immunofluorescence and immunogold electron microscopy. Secretory granules of lysozyme-positive Paneth cells in the bottom of the crypts as well as goblet cells along the crypt-villus axis were intensively labeled with intelectin antibodies, but quantitatively, the major site of intelectin deposition was the enterocyte brush border. This membrane is organized in stable glycolipid-based lipid raft microdomains, and like the divalent lectin galectin-4, intelectin was enriched in microvillar "superrafts", i.e., membranes that resist solubilization with Triton X-100 at 37 degrees C. This strategic localization suggests that the trimeric intelectin, like galectin-4, serves as an organizer and stabilizer of the brush border membrane, preventing loss of digestive enzymes to the gut lumen and protecting the glycolipid microdomains from pathogens.
Collapse
Affiliation(s)
- Uta Wrackmeyer
- Department of Medical Biochemistry and Genetics, Panum Institute, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | | | | | | |
Collapse
|
293
|
Ganley-Leal LM, Liu X, Wetzler LM. Toll-like receptor 2-mediated human B cell differentiation. Clin Immunol 2006; 120:272-84. [PMID: 16766226 DOI: 10.1016/j.clim.2006.04.571] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 04/12/2006] [Accepted: 04/20/2006] [Indexed: 10/24/2022]
Abstract
Human B cells likely have a major role in the adjuvant activity of Toll-like receptor (TLR) 9 agonists by enhancing innate and adaptive immune responses. As several TLR2 ligands are promising vaccine adjuvant candidates, our aim was to characterize the effects of TLR2 stimulation on human B cell activation and differentiation using cells derived from healthy peripheral blood (PB), spleen, and diseased tonsils. We found a subset of partially differentiated TLR2+ PB and splenic B cells which responds to TLR2 agonists by mediating events involved in germinal center formation, such as upregulating CD77 and secreting chemokines. Furthermore, we show that TLR2-activated monocytes induce B cells to secrete significant quantities of IgM. Finally, activated TLR2+ B cells from tonsils are induced to secrete IgM directly by TLR2 ligands. Thus, TLR2 is likely involved in specific B cell-mediated functions and may be a viable vaccine adjuvant target in humans.
Collapse
Affiliation(s)
- Lisa M Ganley-Leal
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Evans Biomedical Research Center, 650 Albany Street, Boston, MA 02118, USA.
| | | | | |
Collapse
|
294
|
Guarner F, Bourdet-Sicard R, Brandtzaeg P, Gill HS, McGuirk P, van Eden W, Versalovic J, Weinstock JV, Rook GAW. Mechanisms of disease: the hygiene hypothesis revisited. ACTA ACUST UNITED AC 2006; 3:275-84. [PMID: 16673007 DOI: 10.1038/ncpgasthep0471] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Accepted: 02/14/2006] [Indexed: 12/13/2022]
Abstract
In industrialized countries the incidence of diseases caused by immune dysregulation has risen. Epidemiologic studies initially suggested this was connected to a reduction in the incidence of infectious diseases; however, an association with defects in immunoregulation is now being recognized. Effector T(H)1 and T(H)2 cells are controlled by specialized subsets of regulatory T cells. Some pathogens can induce regulatory cells to evade immune elimination, but regulatory pathways are homeostatic and mainly triggered by harmless microorganisms. Helminths, saprophytic mycobacteria, bifidobacteria and lactobacilli, which induce immunoregulatory mechanisms in the host, ameliorate aberrant immune responses in the setting of allergy and inflammatory bowel disease. These organisms cause little, if any, harm, and have been part of human microecology for millennia; however, they are now less frequent or even absent in the human environment of westernized societies. Deficient exposure to these 'old friends' might explain the increase in immunodysregulatory disorders. The use of probiotics, prebiotics, helminths or microbe-derived immunoregulatory vaccines might, therefore, become a valuable approach to disease prevention.
Collapse
|
295
|
Abstract
The varied interaction of the Fc region of IgA with receptors confers this antibody class with many of its unique properties. The epithelial polymeric Ig receptor on mucosal epithelial cells transports polymeric immunoglobulin A (pIgA) produced by mucosal B cells to the mucosal surface where, in complex with the secretory component (SC), this secretory immunoglobulin A (SIgA) excludes the multitude of dietary, environmental, and microbial antigens that continuously bombard the mucosae. In health, this IgA-mediated exclusion not only forms the initial defence against infection, it also spares the systemic immune system from potentially deleterious responses to innocuous antigens which can otherwise culminate in inflammatory bowel disease or asthma. Beyond antigen exclusion, in closer encounters with antigens, IgA receptors play roles in protective immunity and disease. FcaRI is the principal myeloid IgA receptor and is responsible for differing IgA-mediated effector responses such as respiratory burst, degranulation, and phagocytosis variously by granulyoctes, monocytes, and macrophages. Furthermore an unknown IgA receptor specific for the secretory component (SC) elicits powerful effector responses from eosinophils. On dendritic cells, FcaRI participates in antigen presentation while on microfold cells, key cells in mucosal antigen presentation, another unknown IgA receptor functions in the transport of antigens across the mucosal epithelial barrier. The activity of another uncharacterized IgA1/IgD receptor on T cells may affect autoimmune disorders. The interplay of different IgA receptors affects immune complex deposition in the common renal disease immunoglobulin A nephropathy (IgAN). Finally, the therapeutic application of various IgA receptors has been sought in the areas of infectious disease, vaccines, and cancer.
Collapse
Affiliation(s)
- Bruce D Wines
- Helen Macpherson Smith Trust Inflammatory Disease Laboratory, The Macfarlane Burnet Institute for Medical Research and Public Health, Austin Health Campus, Heidelberg, Victoria, Australia.
| | | |
Collapse
|
296
|
Hansen GH, Niels-Christiansen LL, Immerdal L, Danielsen EM. Antibodies in the small intestine: mucosal synthesis and deposition of anti-glycosyl IgA, IgM, and IgG in the enterocyte brush border. Am J Physiol Gastrointest Liver Physiol 2006; 291:G82-90. [PMID: 16565420 DOI: 10.1152/ajpgi.00021.2006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Synthesis and deposition of immunoglobulins in the brush border was studied in organ-cultured pig small intestinal mucosal explants. Surprisingly, comparable amounts of IgM and IgA were synthesized during a 6-h pulse, and also newly made IgG was detected in media and explants, including the microvillar fraction. For IgA and IgM, this subcellular distribution is consistent with basolateral-to-apical transcytosis, mediated by the polymeric immunoglobulin receptor. IgG is a ligand for the Fc receptor FcRn, and beta2-microglobulin, the light chain of FcRn, coclustered in immunogold double labeling with IgG in subapical endosomes and in the basolateral membrane of enterocytes. In addition, beta2-microglobulin was copurified with IgG on protein G-Sepharose. Apical endocytosis of IgG, as judged by internalization of fluorescent protein G, was not detectable except in a few isolated cells. This suggests that IgG in the adult small intestine is transported across the enterocyte mainly in the basolateral to apical direction. Significant fractions of all immunoglobulins bound to lactoseagarose, indicating that "anti-glycosyl" antibodies, raised against commensal gut bacteria, are synthesized locally in the small intestine. By partial deposition in the brush border, these antibodies therefore may have a protective function by preventing lectin-like pathogens from gaining access to the brush border surface.
Collapse
Affiliation(s)
- Gert H Hansen
- Department of Medical Biochemistry and Genetics, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | | | | | | |
Collapse
|
297
|
Feng N, Jaimes MC, Lazarus NH, Monak D, Zhang C, Butcher EC, Greenberg HB. Redundant role of chemokines CCL25/TECK and CCL28/MEC in IgA+ plasmablast recruitment to the intestinal lamina propria after rotavirus infection. THE JOURNAL OF IMMUNOLOGY 2006; 176:5749-59. [PMID: 16670280 DOI: 10.4049/jimmunol.176.10.5749] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rotaviruses (RV) are the most important cause of severe childhood diarrheal disease. In suckling mice, infection with RV results in an increase in total and virus-specific IgA(+) plasmablasts in the small intestinal lamina propria (LP) soon after infection, providing a unique opportunity to study the mechanism of IgA(+) cell recruitment into the small intestine. In this study, we show that the increase in total and RV-specific IgA(+) plasmablasts in the LP after RV infection can be blocked by the combined administration of Abs against chemokines CCL25 and CCL28, but not by the administration of either Ab alone. RV infection in CCR9 knockout mice still induced a significant accumulation of IgA(+) plasmablasts in the LP, which was blocked by the addition of anti-CCL28 Ab, confirming the synergistic role of CCL25 and CCL28. The absence of IgA(+) plasmablast accumulation in LP following combined anti-chemokine treatment was not due to changes in proliferation or apoptosis in these cells. We also found that coadministration of anti-CCL25 and anti-CCL28 Abs with the addition of anti-alpha(4) Ab did not further inhibit IgA(+) cell accumulation in the LP and that the CCL25 receptor, CCR9, was coexpressed with the intestinal homing receptor alpha(4)beta(7) on IgA(+) plasmablasts. Finally, we showed that RV infection was associated with an increase in both CCL25 and CCL28 in the small intestine. Hence, our findings indicate that alpha(4)beta(7) along with either CCR9 or CCR10 are sufficient for mediating the intestinal migration of IgA(+) plasmablasts during RV infection.
Collapse
Affiliation(s)
- Ningguo Feng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
298
|
Moser K, Tokoyoda K, Radbruch A, MacLennan I, Manz RA. Stromal niches, plasma cell differentiation and survival. Curr Opin Immunol 2006; 18:265-70. [PMID: 16616478 DOI: 10.1016/j.coi.2006.03.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Accepted: 03/27/2006] [Indexed: 01/21/2023]
Abstract
Contacts made with other cells and stroma have a major impact on proliferation, differentiation, survival, migration and immunoglobulin class switching of plasma cell precursors as well as on the lifespan of the antibody-secreting cells. Induction of tissue-specific chemokine receptors and adhesion molecules directs migratory plasma cell precursors to tissues close to those in which the original immune stimulation occurred. This mechanism focuses the production of specific antibodies within a particular type of tissue, thus providing a means for the most efficient protection against tissue-specific pathogens. Relocation does not apply to long-lived plasma cells responsible for sustained titers of high-affinity systemic antibody. These are formed in germinal centers and migrate to specific niches in the bone marrow that support their further differentiation and long-term survival.
Collapse
Affiliation(s)
- Katrin Moser
- Department for Humoral Immunology, German Arthritis Research Centre, Berlin, Schumannstrasse 21/22, D-10117 Berlin, Germany
| | | | | | | | | |
Collapse
|
299
|
Abstract
Due to their vast surface area, the mucosal surfaces of the body represent a major site of potential attack by invading pathogens. The secretions that bathe mucosal surfaces contain significant levels of immunoglobulins (Igs), which play key roles in immune defense of these surfaces. IgA is the predominant antibody class in many external secretions and has many functional attributes, both direct and indirect, that serve to prevent infective agents such as bacteria and viruses from breaching the mucosal barrier. This review details current understanding of the structural and functional characteristics of IgA, including interaction with specific receptors (such as Fc(alpha)RI, Fc(alpha)/microR, and CD71) and presents examples of the means by which certain pathogens circumvent the protective properties of this important Ig.
Collapse
Affiliation(s)
- Jenny M Woof
- Division of Pathology and Neuroscience, University of Dundee Medical School, Ninewells Hospital, Dundee, UK.
| | | |
Collapse
|