151
|
Lin M, Du L, Brandtzaeg P, Pan-Hammarström Q. IgA subclass switch recombination in human mucosal and systemic immune compartments. Mucosal Immunol 2014; 7:511-20. [PMID: 24064668 DOI: 10.1038/mi.2013.68] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/24/2013] [Accepted: 08/14/2013] [Indexed: 02/04/2023]
Abstract
Human immunoglobulin A (IgA) comprises two IgA subclasses, IgA1 and IgA2, whose distribution has been shown by immunohistochemistry to be different in various body compartments. In comparison with systemic immune compartments, we investigated the IgA switch profiles at the molecular level in salivary and lacrimal glands, nasal mucosa, and proximal and distal gut mucosa. Direct switching from IgM to IgA1 or IgA2 predominated in all immune compartments analyzed. Similar composition of the Sμ-Sα1 and Sμ-Sα2 junctions was observed, including microhomology usage, which suggested that there is no major difference in the actual recombination mechanism utilized during IgA subclass switching. The proportion of IgA1/IgA2 switch recombination events largely paralleled the previously published immunohistochemical representation of IgA1(+) and IgA2(+) plasma cells, implying that the local subclass distribution generally reflects precommitted memory/effector B cells that have undergone IgA subclass switching before extravasation at the effector site. The extremely low or undetectable levels of activation-induced cytidine deaminase (AID) and Iα-Cμ circle transcripts in intestinal lamina propria samples as compared with Peyer's patches suggest that the cellular IgA subclass distribution outside of organized gut-associated lymphoid tissue is only to a minor extent, if at all, influenced by in situ switching.
Collapse
Affiliation(s)
- M Lin
- 1] Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden [2] State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - L Du
- Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo, and Department of Pathology, Oslo University Hospital, Rikshopitalet, Norway
| | - Q Pan-Hammarström
- Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
152
|
Rouaud P, Saintamand A, Saad F, Carrion C, Lecardeur S, Cogné M, Denizot Y. Elucidation of the enigmatic IgD class-switch recombination via germline deletion of the IgH 3' regulatory region. ACTA ACUST UNITED AC 2014; 211:975-85. [PMID: 24752300 DOI: 10.1084/jem.20131385] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Classical class-switch recombination (cCSR) substitutes the Cμ gene with Cγ, Cε, or Cα, thereby generating IgG, IgE, or IgA classes, respectively. This activation-induced deaminase (AID)-driven process is controlled by the IgH 3' regulatory region (3'RR). Regulation of rare IgD CSR events has been enigmatic. We show that μδCSR occurs in mouse mesenteric lymph node (MLN) B cells and is AID-dependent. AID attacks differ from those in cCSR because they are not accompanied by extensive somatic hypermutation (SHM) of targeted regions and because repaired junctions exhibit features of the alternative end-joining (A-EJ) pathway. In contrast to cCSR and SHM, μδCSR is 3'RR-independent, as its absence affects neither breakpoint locations in Sμ- and Sδ-like (σ(δ)) nor mutation patterns at Sμ-σ(δ) junctions. Although mutations occur in the immediate proximity of the μδ junctions, SHM is absent distal to the junctions within both Sμ and rearranged VDJ regions. In conclusion, μδCSR is active in MLNs, occurs independently of 3'RR-driven assembly, and is even dramatically increased in 3'RR-deficient mice, further showing that its regulation differs from cCSR.
Collapse
Affiliation(s)
- Pauline Rouaud
- UMR CNRS 7276, Centre National de la Recherche Scientifique, Université de Limoges, 87025 Limoges, France
| | | | | | | | | | | | | |
Collapse
|
153
|
Changes in nephritogenic serum galactose-deficient IgA1 in IgA nephropathy following tonsillectomy and steroid therapy. PLoS One 2014; 9:e89707. [PMID: 24586974 PMCID: PMC3931817 DOI: 10.1371/journal.pone.0089707] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/23/2014] [Indexed: 02/01/2023] Open
Abstract
Background Recent studies have shown that galactose-deficient IgA1 (GdIgA1) has an important role in the pathogenesis of IgA nephropathy (IgAN). Although emerging data suggest that serum GdIgA1 can be a useful non-invasive IgAN biomarker, the localization of nephritogenic GdIgA1-producing B cells remains unclear. Recent clinical and experimental studies indicate that immune activation tonsillar toll-like receptor (TLR) 9 may be involved in the pathogenesis of IgAN. Here we assessed the possibility of GdIgA1 production in the palatine tonsils in IgAN patients. Methods We assessed changes in serum GdIgA1 levels in IgAN patients with clinical remission of hematuria and proteinuria following combined tonsillectomy and steroid pulse therapy. Further, the association between clinical outcome and tonsillar TLR9 expression was evaluated. Results Patients (n = 37) were divided into two groups according to therapy response. In one group, serum GdIgA1 levels decreased after tonsillectomy (59%) alone, whereas in the other group most levels only decreased after the addition of steroid pulse therapy to tonsillectomy (41%). The former group showed significantly higher tonsillar TLR9 expression and better improvement in hematuria immediately after tonsillectomy than the latter group. Conclusions The present study indicates that the palatine tonsils are probably a major sites of GdIgA1-producing cells. However, in some patients these cells may propagate to other lymphoid organs, which may partially explain the different responses observed to tonsillectomy alone. These findings help to clarify some of the clinical observations in the management of IgAN, and may highlight future directions for research.
Collapse
|
154
|
|
155
|
Oral administration of Shiga toxin-producing Escherichia coli induces intestinal and systemic specific immune response in mice. Med Microbiol Immunol 2014; 203:145-54. [DOI: 10.1007/s00430-013-0325-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022]
|
156
|
Influence of a probiotic lactobacillus strain on the intestinal ecosystem in a stress model mouse. Brain Behav Immun 2014; 35:77-85. [PMID: 24016865 DOI: 10.1016/j.bbi.2013.08.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 08/29/2013] [Accepted: 08/30/2013] [Indexed: 12/13/2022] Open
Abstract
Daily exposure to stressful situations affects the health of humans and animals. It has been shown that psychological stress affects the immune system and can exacerbate diseases. Probiotics can act as biological immunomodulators in healthy people, increasing both intestinal and systemic immune responses. The use of probiotics in stress situations may aid in reinforcing the immune system. The aim of this study was to evaluate the effect of a probiotic bacterium on the gut immune system of mice that were exposed to an experimental model of stress induced by food and mobility restriction. The current study focused on immune cells associated with the lamina propria of the intestine, including CD4+ and CD8+ T lymphocytes, CD11b+ macrophages, CD11c+ dendritic cells, and IgA+ B lymphocytes, as well as the concentrations of secretory IgA (S-IgA) and cytokine interferon gamma (INF-γ in intestinal fluid. We also evaluated the probiotic's influence on the gut microbiota. Probiotic administration increased IgA producing cells, CD4+ cells in the lamina propria of the small intestine, and S-IgA in the lumen; it also reduced the levels of IFN-γ that had increased during stress and improved the intestinal microbiota as measured by an increase in the lactobacilli population. The results obtained from administration of the probiotic to stressed mice suggest that the use of food containing these microorganisms may work as a palliative to reinforce the immune system.
Collapse
|
157
|
Pierre JF, Heneghan AF, Wang X, Roenneburg DA, Groblewski GE, Kudsk KA. Bombesin improves adaptive immunity of the salivary gland during parenteral nutrition. JPEN J Parenter Enteral Nutr 2013; 39:190-9. [PMID: 24121183 DOI: 10.1177/0148607113507080] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The parotid and submandibular salivary glands are gut-associated lymphoid tissues (GALTs) that secrete immune compounds into the oral cavity. Parenteral nutrition (PN) without enteral stimulation decreases GALT function, including intestinal lymphocyte counts and secretory immunoglobulin A (sIgA) levels. Since the neuropeptide bombesin (BBS), a gastrin-releasing peptide analogue, stimulates intestinal function and restores GALT parameters, we hypothesized that PN + BBS would stimulate parotid and salivary gland IgA levels, T lymphocytes, and IgA plasma cell counts compared with PN alone. METHODS Male (Institute of Cancer Research) ICR mice received intravenous catheters and were randomized to chow with saline, PN, or PN + BBS (15 µg/tid/mouse) for 5 days (8/group), 2 days after cannulation. Salivary glands were weighed and either frozen for IgA and amylase analysis or fixed for histological analysis of acinar cells, IgA+ plasma cells, and T lymphocytes. Small intestinal wash fluid was collected for IgA regression analysis with salivary glands. RESULTS PN reduced organ weight, acinar cell size, and amylase activity compared with chow; BBS had no significant effects on these parameters. Compared with chow, PN significantly reduced salivary gland IgA levels, IgA+ plasma cells, and T lymphocytes. PN + BBS significantly elevated IgA and restored cellularity compared with PN. Salivary gland tissue homogenate IgA levels significantly correlated with intestinal fluid IgA levels. CONCLUSIONS Compared with chow, PN results in atrophy of the salivary glands characterized by reduced amylase, IgA, and immune cellularity. BBS has no effect on acinar cells or amylase activity compared with PN but maintains tissue IgA and plasma cells and T-lymphocyte numbers compared with chow.
Collapse
Affiliation(s)
- Joseph F Pierre
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, WI Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Aaron F Heneghan
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, WI Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Xinying Wang
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI Department of Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Drew A Roenneburg
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Guy E Groblewski
- Department of Nutritional Science, University of Wisconsin-Madison, Madison, WI
| | - Kenneth A Kudsk
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, WI Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
158
|
Brandtzaeg P. Secretory IgA: Designed for Anti-Microbial Defense. Front Immunol 2013; 4:222. [PMID: 23964273 PMCID: PMC3734371 DOI: 10.3389/fimmu.2013.00222] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/16/2013] [Indexed: 01/30/2023] Open
Abstract
Prevention of infections by vaccination remains a compelling goal to improve public health. Mucosal vaccines would make immunization procedures easier, be better suited for mass administration, and most efficiently induce immune exclusion - a term coined for non-inflammatory antibody shielding of internal body surfaces, mediated principally by secretory immunoglobulin A (SIgA). The exported antibodies are polymeric, mainly IgA dimers (pIgA), produced by local plasma cells (PCs) stimulated by antigens that target the mucose. SIgA was early shown to be complexed with an epithelial glycoprotein - the secretory component (SC). A common SC-dependent transport mechanism for pIgA and pentameric IgM was then proposed, implying that membrane SC acts as a receptor, now usually called the polymeric Ig receptor (pIgR). From the basolateral surface, pIg-pIgR complexes are taken up by endocytosis and then extruded into the lumen after apical cleavage of the receptor - bound SC having stabilizing and innate functions in the secretory antibodies. Mice deficient for pIgR show that this is the only receptor responsible for epithelial export of IgA and IgM. These knockout mice show a variety of defects in their mucosal defense and changes in their intestinal microbiota. In the gut, induction of B-cells occurs in gut-associated lymphoid tissue, particularly the Peyer's patches and isolated lymphoid follicles, but also in mesenteric lymph nodes. PC differentiation is accomplished in the lamina propria to which the activated memory/effector B-cells home. The airways also receive such cells from nasopharynx-associated lymphoid tissue but by different homing receptors. This compartmentalization is a challenge for mucosal vaccination, as are the mechanisms used by the mucosal immune system to discriminate between commensal symbionts (mutualism), pathobionts, and overt pathogens (elimination).
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
159
|
|
160
|
Moldt B, Saye-Francisco K, Schultz N, Burton DR, Hessell AJ. Simplifying the synthesis of SIgA: combination of dIgA and rhSC using affinity chromatography. Methods 2013; 65:127-32. [PMID: 23811333 DOI: 10.1016/j.ymeth.2013.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/17/2013] [Accepted: 06/18/2013] [Indexed: 01/30/2023] Open
Abstract
The mucosal epithelia together with adaptive immune responses, such as local production and secretion of dimeric and polymeric immunoglobulin A (IgA), are a crucial part of the first line of defense against invading pathogens. IgA is primarily secreted as SIgA and plays multiple roles in mucosal defense. The study of SIgA-mediated protection is an important area of research in mucosal immunity but an easy, fast and reproducible method to generate pathogen-specific SIgA in vitro has not been available. We report here a new method to produce SIgA by co-purification of dimeric IgA, containing J chain, and recombinant human SC expressed in CHO cells. We previously reported the generation, production and characterization of the human recombinant monoclonal antibody IgA2 b12. This antibody, derived from the variable regions of the neutralizing anti-HIV-1 mAb IgG1 b12, blocked viral attachment and uptake by epithelial cells in vitro. We used a cloned CHO cell line that expresses monomeric, dimeric and polymeric species of IgA2 b12 for large-scale production of dIgA2 b12. Subsequently, we generated a CHO cell line to express recombinant human secretory component (rhSC). Here, we combined dIgA2 b12 and CHO-expressed rhSC via column chromatography to produce SIgA2 b12 that remains fully intact upon elution with 0.1M citric acid, pH 3.0. We have performed biochemical analysis of the synthesized SIgA to confirm the species is of the expected size and retains the functional properties previously described for IgA2 b12. We show that SIgA2 b12 binds to the HIV-1 gp120 glycoprotein with similar apparent affinity to that of monomeric and dimeric forms of IgA2 b12 and neutralizes HIV-1 isolates with similar potency. An average yield of 6 mg of SIgA2 b12 was achieved from the combination of 20mg of purified dIgA2 b12 and 2L of rhSC-containing CHO cell supernatant. We conclude that synthesized production of stable SIgA can be generated by co-purification. This process introduces a simplified means of generating a variety of pathogen-specific SIgA antibodies for research and clinical applications.
Collapse
Affiliation(s)
- Brian Moldt
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Karen Saye-Francisco
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Niccole Schultz
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Boston, MA, USA
| | - Ann J Hessell
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
161
|
Abstract
Primary antibody deficiencies (PADs) are the most common inherited immunodeficiencies in humans. The use of novel approaches, such as whole-exome sequencing and mouse genetic engineering, has helped to identify new genes that are involved in the pathogenesis of PADs and has enabled the characterization of the molecular pathways that are involved in B cell development and function. Here, we review the different PADs in terms of their known or putative mechanisms, which can be B cell intrinsic, B cell extrinsic or not defined so far. We also describe the clinical manifestations (including susceptibility to infections, autoimmunity and cancer) that have been associated with the various PADs.
Collapse
Affiliation(s)
- Anne Durandy
- National Institute of Health and Medical Research, INSERM U768, Necker Children's Hospital, F-75015 Paris, France.
| | | | | |
Collapse
|
162
|
The pyloric caeca area is a major site for IgM(+) and IgT(+) B cell recruitment in response to oral vaccination in rainbow trout. PLoS One 2013; 8:e66118. [PMID: 23785475 PMCID: PMC3681912 DOI: 10.1371/journal.pone.0066118] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/01/2013] [Indexed: 11/26/2022] Open
Abstract
Although previous studies have characterized some aspects of the immune response of the teleost gut in response to diverse pathogens or stimuli, most studies have focused on the posterior segments exclusively. However, there are still many details of how teleost intestinal immunity is regulated that remain unsolved, including the location of IgM+ and IgT+ B cells along the digestive tract and their role during the course of a local stimulus. Thus, in the current work, we have studied the B cell response in five different segments of the rainbow trout (Oncorhynchus mykiss) digestive tract in both naïve fish and fish orally vaccinated with an alginate-encapsulated DNA vaccine against infectious pancreatic necrosis virus (IPNV). IgM+ and IgT+ cells were identified all along the tract with the exception of the stomach in naïve fish. While IgM+ cells were mostly located in the lamina propria (LP), IgT+ cells were primarily localized as intraepithelial lymphocytes (IELs). Scattered IgM+ IELs were only detected in the pyloric caeca. In response to oral vaccination, the pyloric caeca region was the area of the digestive tract in which a major recruitment of B cells was demonstrated through both real time PCR and immunohistochemistry, observing a significant increase in the number of both IgM+ and IgT+ IELs. Our findings demonstrate that both IgM+ and IgT+ respond to oral stimulation and challenge the paradigm that teleost IELs are exclusively T cells. Unexpectedly, we have also detected B cells in the fat tissue associated to the digestive tract that respond to vaccination, suggesting that these cells surrounded by adipocytes also play a role in mucosal defense.
Collapse
|
163
|
Davis CL, Wahid R, Toapanta FR, Simon JK, Sztein MB, Levy D. Applying mathematical tools to accelerate vaccine development: modeling Shigella immune dynamics. PLoS One 2013; 8:e59465. [PMID: 23589755 PMCID: PMC3614931 DOI: 10.1371/journal.pone.0059465] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 02/11/2013] [Indexed: 12/02/2022] Open
Abstract
We establish a mathematical framework for studying immune interactions with Shigella, a bacteria that kills over one million people worldwide every year. The long-term goal of this novel approach is to inform Shigella vaccine design by elucidating which immune components and bacterial targets are crucial for establishing Shigella immunity. Our delay differential equation model focuses on antibody and B cell responses directed against antigens like lipopolysaccharide in Shigella’s outer membrane. We find that antibody-based vaccines targeting only surface antigens cannot elicit sufficient immunity for protection. Additional boosting prior to infection would require a four-orders-of-magnitude increase in antibodies to sufficiently prevent epithelial invasion. However, boosting anti-LPS B memory can confer protection, which suggests these cells may correlate with immunity. We see that IgA antibodies are slightly more effective per molecule than IgG, but more total IgA is required due to spatial functionality. An extension of the model reveals that targeting both LPS and epithelial entry proteins is a promising avenue to advance vaccine development. This paper underscores the importance of multifaceted immune targeting in creating an effective Shigella vaccine. It introduces mathematical models to the Shigella vaccine development effort and lays a foundation for joint theoretical/experimental/clinical approaches to Shigella vaccine design.
Collapse
Affiliation(s)
- Courtney L Davis
- Natural Science Division, Pepperdine University, Malibu, California, United States of America.
| | | | | | | | | | | |
Collapse
|
164
|
Agnello D, Denimal D, Lavaux A, Blondeau-Germe L, Lu B, Gerard NP, Gerard C, Pothier P. Intrarectal immunization and IgA antibody-secreting cell homing to the small intestine. THE JOURNAL OF IMMUNOLOGY 2013; 190:4836-47. [PMID: 23547118 DOI: 10.4049/jimmunol.1202979] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
According to the current paradigm, lymphocyte homing to the small intestine requires the expression of two tissue-specific homing receptors, the integrin α4β7 and the CCL25 receptor CCR9. In this study, we investigated the organ distribution and the homing molecule expression of IgA Ab-secreting cells (ASCs) induced by intrarectal immunization with a particulate Ag, in comparison with other mucosal immunization routes. Intrarectal immunization induces gut-homing IgA ASCs that localize not only in the colon but also in the small intestine, although they are not responsive to CCL25, unlike IgA ASCs induced by oral immunization. The mucosal epithelial chemokine CCL28, known to attract all IgA ASCs, does not compensate for the lack of CCL25 responsiveness, because the number of Ag-specific cells is not decreased in the gut of CCR10-deficient mice immunized by the intrarectal route. However, Ag-specific IgA ASCs induced by intrarectal immunization express the integrin α4β7, and their number is considerably decreased in the gut of β7-deficient mice immunized by the intrarectal route, indicating that α4β7 enables these cells to migrate into the small intestine, even without CCL25 responsiveness. In contrast, IgA ASCs induced by intranasal immunization express low α4β7 levels and are usually excluded from the gut. Paradoxically, after intranasal immunization, Ag-specific IgA ASCs are significantly increased in the small intestine of β7-deficient mice, demonstrating that lymphocyte homing is a competitive process and that integrin α4β7 determines not only the intestinal tropism of IgA ASCs elicited in GALTs but also the intestinal exclusion of lymphocytes primed in other inductive sites.
Collapse
Affiliation(s)
- Davide Agnello
- Laboratoire de Virologie et Centre National de Référence des Virus Entériques, Centre Hospitalier Universitaire de Dijon, 21070 Dijon Cedex, France.
| | | | | | | | | | | | | | | |
Collapse
|
165
|
Brandtzaeg P. Secretory immunity with special reference to the oral cavity. J Oral Microbiol 2013; 5:20401. [PMID: 23487566 PMCID: PMC3595421 DOI: 10.3402/jom.v5i0.20401] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 11/14/2022] Open
Abstract
The two principal antibody classes present in saliva are secretory IgA (SIgA) and IgG; the former is produced as dimeric IgA by local plasma cells (PCs) in the stroma of salivary glands and is transported through secretory epithelia by the polymeric Ig receptor (pIgR), also named membrane secretory component (SC). Most IgG in saliva is derived from the blood circulation by passive leakage mainly via gingival crevicular epithelium, although some may be locally produced in the gingiva or salivary glands. Gut-associated lymphoid tissue (GALT) and nasopharynx-associated lymphoid tissue (NALT) do not contribute equally to the pool of memory/effector B cells differentiating to mucosal PCs throughout the body. Thus, enteric immunostimulation may not be the best way to activate the production of salivary IgA antibodies although the level of specific SIgA in saliva may still reflect an intestinal immune response after enteric immunization. It remains unknown whether 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 levels of IgA in submandibular secretions of AIDS patients, paralleling their highly upregulated intestinal IgA system, while the parotid IgA level is decreased. Parotid SIgA could more consistently be linked to immune induction in palatine tonsils/adenoids (human NALT) and cervical lymph nodes, as supported by the homing molecule profile observed after immune induction at these sites. Several other variables influence the levels of antibodies in salivary 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 these problems, saliva is an easily accessible biological fluid with interesting scientific and clinical potentials.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Per Brandtzaeg, Department of Pathology, Oslo University Hospital, Rikshospitalet, PO Box 4950 Oslo, NO-0424 Norway. Tel: +47-23072743, Fax: 47-23071511.
| |
Collapse
|
166
|
Clarke ET, Williams NA, Dull PM, Findlow J, Borrow R, Finn A, Heyderman RS. Polysaccharide-protein conjugate vaccination induces antibody production but not sustained B-cell memory in the human nasopharyngeal mucosa. Mucosal Immunol 2013; 6:288-96. [PMID: 22806100 DOI: 10.1038/mi.2012.70] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Colonization of the nasopharyngeal mucosa by meningococcus and other polysaccharide (PS)-encapsulated bacteria precedes invasion. PS-conjugate vaccines induce PS-specific B-cell memory (B(MEM)) and also prevent colonization, thus blocking person-to-person transmission, generating herd protection. However, in isolation the B(MEM) are unable to sustain immunity. Furthermore, the duration of herd protection the vaccines induce appears limited. We demonstrate that, despite the persistence of PS-specific B(MEM), the population is not maintained within the nasopharynx. Although booster immunization results in the transient appearance of PS-specific B(MEM) within the mucosa, this reflects the re-circulation of systemic B(MEM) through the site rather than the generation of resident mucosal B(MEM). The induction of sustained PS-specific B(MEM) in the nasopharynx would allow the population to be activated by colonization, thus inhibiting subsequent invasion. It would also be expected to boost local mucosal immunity, thus extending herd protection. Strategies to generate PS-specific B(MEM) in the mucosa warrant further investigation.
Collapse
Affiliation(s)
- E T Clarke
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| | | | | | | | | | | | | |
Collapse
|
167
|
Takata K, Sato Y, Nakamura N, Tokunaka M, Miki Y, Yukie Kikuti Y, Igarashi K, Ito E, Harigae H, Kato S, Hayashi E, Oka T, Hoshii Y, Tari A, Okada H, Al-Kader LA, Mohamad AAL, Maeda Y, Tanimoto M, Kinoshita T, Yoshino T. Duodenal follicular lymphoma lacks AID but expresses BACH2 and has memory B-cell characteristics. Mod Pathol 2013; 26:22-31. [PMID: 22899287 DOI: 10.1038/modpathol.2012.127] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We have reported previously that duodenal follicular lymphoma (FL) is distinct from nodal FL and showed more resemblance to mucosa-associated lymphoid tissue lymphoma, and that FL frequently involved the duodenal second portion. In the present study, we examined duodenal FLs and gastric/colonic FLs to clarify the clinicopathological and immunological differences between the tumor types. We analyzed 8 samples of gastric FL, 17 of duodenal ones, and 5 of colonic/rectal ones, and characterized them by immunohistochemistry, immunogenotyping, and histology. Gastric and colonic FLs presented in submucosal to subserosal areas, whereas duodenal ones presented in the mucosal to submucosal layers. Immunohistochemical analysis revealed that duodenal FLs exhibited the following phenotypes: CD10 (+), B-cell lymphoma 2 (BCL-2) (+), BCL-6 (+), activation-induced cytidine deaminase (AID) (-), BACH2 (+), CD27 (+), MUM-1 (-), Blimp-1 (-), and loose CD21 network (duodenal pattern). Gastric/colonic FLs exhibited the following phenotypes: CD10 (+), BCL-2 (+), BCL-6 (+), AID (+), BACH2 (+), CD27 (-), MUM-1 (-), Blimp-1 (-), and a dense CD21 network (nodal pattern). Expression of AID and CD27 in lymphoma cells and the CD21 network pattern were considerably different between duodenal FLs and gastric/colonic ones. Moreover, in situ hybridization revealed that, in the duodenal FLs, BACH2 was expressed at the periphery of the tumor follicle and tumor villi. The number of immunoglobulin heavy-chain variable domains VH4 and VH5 were higher in duodenal follicular lymphomoas than in gastric FLs. The lymphoma cells of duodenal FLs are different from those of gastric/colonic FLs, and duodenal FL is distinct even within the gastrointestinal tract. Somatic hypermutation in immunoglobulin genes and CD27 expression are hallmarks of memory B cells. We suggest that duodenal FL cells are in the memory B-cell stage, and require BACH2 instead of AID for ongoing mutation.
Collapse
Affiliation(s)
- Katsuyoshi Takata
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Soheili H, Abolhassani H, Arandi N, Khazaei HA, Shahinpour S, Hirbod-Mobarakeh A, Rezaei N, Aghamohammadi A. Evaluation of Natural Regulatory T Cells in Subjects with Selective IgA Deficiency: From Senior Idea to Novel Opportunities. Int Arch Allergy Immunol 2013; 160:208-14. [DOI: 10.1159/000339867] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 05/30/2012] [Indexed: 01/19/2023] Open
|
169
|
Buchanan RM, Tetland S, Wilson HL. Low dose antigen exposure for a finite period in newborn rats prevents induction of mucosal tolerance. PLoS One 2012; 7:e51437. [PMID: 23251533 PMCID: PMC3520849 DOI: 10.1371/journal.pone.0051437] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/31/2012] [Indexed: 11/18/2022] Open
Abstract
Background In adult rats, initial exposure to antigens by a mucosal route triggers tolerance such that any subsequent re-exposure, even by a systemic route, results in suppression of immunity. The newborn’s gut is semi-permeable for a finite period to allow maternal antibodies to enter the newborn’s circulation. We propose that antigens introduced in extreme early life can readily traverse the gut wall and therefore circumvent induction of mucosal tolerance. Methodology/Principle Findings Rat pups were gavaged with low-doses of ovalbumin (OVA; oral exposure group) or saline (parenteral control group) every second day for several weeks followed by an intraperitoneal (i.p.) injection at 1 month of age. When gavage was initiated the day after birth, newborn oral exposure pups responded with significantly higher anti-OVA IgA, IgM, IgG2a, and IgG1 titres in their serum and anti-OVA IgA, IgG2a and IgG1 titres in their lungs compared to negative control pups. Oral exposure alone failed to induce immunity. Pups exposed to the same treatment regimen starting at 14 days of age showed induction of mucosal tolerance after i.p. immunization. Newborn oral exposure groups subjected to secondary i.p. immunization responded with significantly increased humoral immunity in lung and sera suggesting that once antigen-specific mucosal tolerance if circumvented, it persists. Lymphocytes derived from mesenteric lymph node cells re-simulated with OVA ex vivo, from newborn oral exposure pups exposed to secondary immunization produced significantly higher IFN-γ expression and lymphocyte proliferation relative to control pups indicating prevention of tolerance in the cell-mediated immune system. Conclusions/Significance This work demonstrates that newborns may be uniquely qualified to prevent induction of mucosal tolerance to oral antigens. These results should be further explored to establish whether prevention of tolerance by early life oral vaccination can be exploited to prime for mucosal as well as systemic immunity and thus protect this susceptible population against infectious diseases.
Collapse
Affiliation(s)
- Rachelle M. Buchanan
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Sherry Tetland
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Heather L. Wilson
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
- * E-mail:
| |
Collapse
|
170
|
Gourbeyre P, Desbuards N, Grémy G, Le Gall S, Champ M, Denery-Papini S, Bodinier M. Exposure to a galactooligosaccharides/inulin prebiotic mix at different developmental time points differentially modulates immune responses in mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:11942-11951. [PMID: 23145871 DOI: 10.1021/jf3036403] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Prebiotics constitute emerging tools to alleviate immune pathologies. This study aimed to evaluate the effect of prebiotic exposure during perinatal and postweaning periods on immune and gut regulations. Mice were fed either a galactooligosaccharides/inulin prebiotic mix-enriched diet or a control diet during the perinatal and/or postweaning periods. Biomarkers related to gut barrier function (SCFA, heat shock proteins, zonula occludens protein-1, and mucin-2) and immune mechanisms (IgA, IgE, IgG1, IgG2a, IL-10, TGF-β, IL-4, IL-17A, and IFN-γ) were analyzed. The milk of dams fed the prebiotic diet was more concentrated in both IgA and TGF-β when prebiotics were introduced during both the perinatal and postweaning periods; IL-10, IgA, and IgG2a were increased in pups; and expression of intestinal markers was more pronounced. Postweaning exposure to prebiotics alone induced higher INF-γ and TGF-β levels, whereas IgA levels fell. Combined exposure periods (perinatal/postweaning) to prebiotics increased tolerance-related immunoglobulins in pups and reinforced gut barrier functions.
Collapse
Affiliation(s)
- Pascal Gourbeyre
- INRA, UR 1268 BIA, rue de la Géraudière, B.P. 71627, 44316 Nantes Cedex 03, France
| | | | | | | | | | | | | |
Collapse
|
171
|
Budec M, Markovic D, Vignjevic S, Mitrovic O, Dikic D, Koko V, Cokic VP. Neuronal nitric oxide synthase mediates the effect of ethanol on IgA. Alcohol Alcohol 2012; 48:53-8. [PMID: 23059423 DOI: 10.1093/alcalc/ags110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS We showed previously that the acute effect of ethanol on intestinal immunoglobulin A (IgA) expression might be mediated by endogenous nitric oxide (NO). To extend these findings, this study was designed to investigate a possible role of neuronal NO synthase (nNOS) in the observed phenomenon, using 7-nitroindazole (7-NI), a selective inhibitor of its activity. METHODS Adult male Wistar rats were treated with: (a) ethanol (4 g/kg, intraperitoneally, i.p.), (b) 7-NI (25 mg/kg, i.p.) followed by ethanol (4 g/kg, i.p.) 30 min later and (c) 7-NI (25 mg/kg, i.p.) followed by saline 30 min later. Untreated rats were used as controls. The concentrations of serum and intestinal IgA were measured by enzyme-linked immunosorbent assay, while the expression of nNOS was determined using western blot and immunohistochemistry. RESULTS Acute ethanol treatment significantly increased the concentration of IgA in the ileal extracts, whereas it decreased its serum level. Inhibition of nNOS activity by 7-NI abolished this action of alcohol on IgA. Additionally, western blot analysis revealed that the acute alcohol administration induced an increase in the expression of intestinal nNOS. Furthermore, nNOS-immunoreactive cells, observed within the lamina propria of small intestine, were numerous in ethanol-treated rats. CONCLUSION Taken together, these results extended our previous findings suggesting that nNOS mediates the acute effect of ethanol on IgA and supported an immunomodulatory role of this enzyme isoform.
Collapse
Affiliation(s)
- Mirela Budec
- University of Belgrade, Institute for Medical Research, Dr Subotića 4, Belgrade 102, Serbia.
| | | | | | | | | | | | | |
Collapse
|
172
|
Mesin L, Sollid LM, Di Niro R. The intestinal B-cell response in celiac disease. Front Immunol 2012; 3:313. [PMID: 23060888 PMCID: PMC3463893 DOI: 10.3389/fimmu.2012.00313] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 09/18/2012] [Indexed: 12/19/2022] Open
Abstract
The function of intestinal immunity is to provide protection toward pathogens while preserving the composition of the microflora and tolerance to orally fed nutrients. This is achieved via a number of tightly regulated mechanisms including production of IgA antibodies by intestinal plasma cells. Celiac disease is a common gut disorder caused by a dysfunctional immune regulation as signified, among other features, by a massive intestinal IgA autoantibody response. Here we review the current knowledge of this B-cell response and how it is induced, and we discuss key questions to be addressed in future research.
Collapse
Affiliation(s)
- Luka Mesin
- Centre for Immune Regulation, Department of Immunology, Oslo University Hospital-Rikshospitalet, University of Oslo, Oslo, Norway
| | | | | |
Collapse
|
173
|
Sotolongo J, Ruiz J, Fukata M. The role of innate immunity in the host defense against intestinal bacterial pathogens. Curr Infect Dis Rep 2012; 14:15-23. [PMID: 22139594 DOI: 10.1007/s11908-011-0234-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Eradication of infectious disease is our global health challenge. After encountering intestinal infection with a bacterial pathogen, the host defense program is initiated by local antigen-presenting cells (APCs) that eliminate invading pathogens by phagocytosis and establish localized inflammation by secreting cytokines and chemokines. These pathogen-experienced APCs migrate to the mesenteric lymph nodes, where host immune responses are precisely orchestrated. Initiation and regulation of this defense program appear to be largely dependent on innate immunity which is antigen non-specific and provides a rapid defense against broader targets. On the other hand, many bacterial enteropathogens have evoked abilities to modify the host defense program to their advantage. Therefore, better understanding of the host-pathogen interactions is essential to establish effective eradication strategies for enteric infectious diseases. In this review, we will discuss the current understanding of innate immune regulation of the host defense mechanisms against intestinal infection by bacterial pathogens.
Collapse
Affiliation(s)
- John Sotolongo
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Post Office Box 016960 (D-149), Miami, FL, 33101, USA
| | | | | |
Collapse
|
174
|
Reikvam DH, Derrien M, Islam R, Erofeev A, Grcic V, Sandvik A, Gaustad P, Meza-Zepeda LA, Jahnsen FL, Smidt H, Johansen FE. Epithelial-microbial crosstalk in polymeric Ig receptor deficient mice. Eur J Immunol 2012; 42:2959-70. [DOI: 10.1002/eji.201242543] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 06/19/2012] [Accepted: 07/10/2012] [Indexed: 12/14/2022]
Affiliation(s)
- Dag Henrik Reikvam
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
| | - Muriel Derrien
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Rejoanoul Islam
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Alexander Erofeev
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
| | - Vedrana Grcic
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
| | - Anders Sandvik
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
| | - Peter Gaustad
- Institute of Microbiology; University of Oslo; Oslo Norway
| | - Leonardo A. Meza-Zepeda
- Department of Tumor Biology; Oslo University Hospital - The Norwegian Radium Hospital; Oslo Norway
- Norwegian Microarray Consortium,; Department of Molecular Biosciences; University of Oslo; Oslo Norway
| | - Frode L. Jahnsen
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
| | - Hauke Smidt
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Finn-Eirik Johansen
- Department of Pathology and Centre for Immune Regulation; University of Oslo and Oslo University Hospital - Rikshospitalet; Oslo Norway
- Department of Molecular Biosciences; University of Oslo; Oslo Norway
| |
Collapse
|
175
|
Van de Perre P, Rubbo PA, Viljoen J, Nagot N, Tylleskar T, Lepage P, Vendrell JP, Tuaillon E. HIV-1 Reservoirs in Breast Milk and Challenges to Elimination of Breast-Feeding Transmission of HIV-1. Sci Transl Med 2012; 4:143sr3. [DOI: 10.1126/scitranslmed.3003327] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
176
|
Berin MC. Mucosal antibodies in the regulation of tolerance and allergy to foods. Semin Immunopathol 2012; 34:633-42. [PMID: 22777546 DOI: 10.1007/s00281-012-0325-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 06/20/2012] [Indexed: 01/01/2023]
Abstract
The intestinal mucosa is densely packed with antibody-secreting B cells, the majority of which produce IgA. Mucosal antibodies have traditionally been thought of as neutralizing antibodies that exclude antigens, but they also function in antigen sampling, allowing for selective transcytosis of antigens from the intestinal lumen. IgE-mediated antigen uptake can facilitate the development of allergic reactions to foods, but emerging evidence indicates that IgG-mediated antigen uptake may also play an important role in the development of immune tolerance to foods, particularly in the neonate. This review will focus on the role of intestinal immunoglobulins in the development of clinical tolerance and allergy to food antigens.
Collapse
Affiliation(s)
- M Cecilia Berin
- Division of Allergy and Immunology, Department of Pediatrics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
177
|
Rancez M, Couëdel-Courteille A, Cheynier R. Chemokines at mucosal barriers and their impact on HIV infection. Cytokine Growth Factor Rev 2012; 23:233-43. [PMID: 22728258 DOI: 10.1016/j.cytogfr.2012.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Aside from representing a physical barrier and providing an unfavorable chemical milieu to viral and bacterial infections, mucosae of gut and female genital tract also contain organized lymphoid structures that support the initiation of anti-microbial immune responses, and more diffuse lymphoid tissues that represent immune effector mucosal sites. Local expression of specific chemokines orchestrates lymphoid cell trafficking and positioning in the mucosa-associated lymphoid tissues, leading to their efficient priming during antigenic stimulations as well as their specific homing back where they were primed. This review examines productions and roles of mucosae-specific chemokines in healthy and pathological conditions, as well as their possible positive and deleterious effects during mucosal HIV infection.
Collapse
|
178
|
Genes involved in systemic and arterial bed dependent atherosclerosis--Tampere Vascular study. PLoS One 2012; 7:e33787. [PMID: 22509262 PMCID: PMC3324479 DOI: 10.1371/journal.pone.0033787] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 02/19/2012] [Indexed: 12/23/2022] Open
Abstract
Background Atherosclerosis is a complex disease with hundreds of genes influencing its progression. In addition, the phenotype of the disease varies significantly depending on the arterial bed. Methodology/Principal Findings We characterized the genes generally involved in human advanced atherosclerotic (AHA type V–VI) plaques in carotid and femoral arteries as well as aortas from 24 subjects of Tampere Vascular study and compared the results to non-atherosclerotic internal thoracic arteries (n=6) using genome-wide expression array and QRT-PCR. In addition we determined genes that were typical for each arterial plaque studied. To gain a comprehensive insight into the pathologic processes in the plaques we also analyzed pathways and gene sets dysregulated in this disease using gene set enrichment analysis (GSEA). According to the selection criteria used (>3.0 fold change and p-value <0.05), 235 genes were up-regulated and 68 genes down-regulated in the carotid plaques, 242 genes up-regulated and 116 down-regulated in the femoral plaques and 256 genes up-regulated and 49 genes down-regulated in the aortic plaques. Nine genes were found to be specifically induced predominantly in aortic plaques, e.g., lactoferrin, and three genes in femoral plaques, e.g., chondroadherin, whereas no gene was found to be specific for carotid plaques. In pathway analysis, a total of 28 pathways or gene sets were found to be significantly dysregulated in atherosclerotic plaques (false discovery rate [FDR] <0.25). Conclusions This study describes comprehensively the gene expression changes that generally prevail in human atherosclerotic plaques. In addition, site specific genes induced only in femoral or aortic plaques were found, reflecting that atherosclerotic process has unique features in different vascular beds.
Collapse
|
179
|
Live oral typhoid vaccine Ty21a induces cross-reactive humoral immune responses against Salmonella enterica serovar Paratyphi A and S. Paratyphi B in humans. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:825-34. [PMID: 22492745 DOI: 10.1128/cvi.00058-12] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Enteric fever caused by Salmonella enterica serovar Paratyphi A infection has emerged as an important public health problem. Recognizing that in randomized controlled field trials oral immunization with attenuated S. enterica serovar Typhi live vaccine Ty21a conferred significant cross-protection against S. Paratyphi B but not S. Paratyphi A disease, we undertook a clinical study to ascertain whether humoral immune responses could explain the field trial results. Ty21a immunization of adult residents of Maryland elicited predominantly IgA antibody-secreting cells (ASC) that recognize S. Typhi lipopolysaccharide (LPS). Cross-reactivity to S. Paratyphi A LPS was significantly lower than that to S. Paratyphi B LPS. ASC producing IgG and IgA that bind LPS from each of these Salmonella serovars expressed CD27 and integrin α4β7 (gut homing), with a significant proportion coexpressing CD62L (secondary lymphoid tissue homing). No significant differences were observed in serum antibody against LPS of the different serovars. Levels of IgA B memory (B(M)) cells to S. Typhi LPS were significantly higher than those against S. Paratyphi A or B LPS, with no differences observed between S. Paratyphi A and B. The response of IgA B(M) to outer membrane proteins (OMP) from S. Typhi was significantly stronger than that to OMP of S. Paratyphi A but similar to that to OMP of S. Paratyphi B. The percentages of IgG or IgA B(M) responders to LPS or OMP from these Salmonella strains were similar. Whereas cross-reactive humoral immune responses to S. Paratyphi A or B antigens are demonstrable following Ty21a immunization, they cannot explain the efficacy data gleaned from controlled field trials.
Collapse
|
180
|
Palkola NV, Pakkanen SH, Kantele JM, Rossi N, Puohiniemi R, Kantele A. Pathogen-specific circulating plasmablasts in patients with pneumonia. PLoS One 2012; 7:e34334. [PMID: 22479603 PMCID: PMC3314017 DOI: 10.1371/journal.pone.0034334] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 02/26/2012] [Indexed: 11/19/2022] Open
Abstract
Lower respiratory tract infections (LRTI) are the leading cause of death world-wide, with Streptococcus pneumoniae (Pnc) as the most prevalent pathogen. Local immune mechanisms appear central to protection against the disease, yet they are poorly characterized. Infections at other, non-respiratory mucosal sites are associated with a transient circulation of mucosa-originating lymphocytes from the mucosal site to blood and back to the mucosa. The present study explored whether pathogen-specific plasmablasts appear in the circulation also in patients with infection of the lower respiratory tract. 16 patients with bacteremic Pnc pneumonia and 14 healthy volunteers were explored for circulating plasmablasts secreting antibodies against their own pathogenic Pnc strain isolated in blood cultures (patients) or against several pathogenic strains from pneumonia patients (14 controls) or a mixture of nine different purified pneumococcal polysaccharides (8 controls). Both patients and volunteers were studied for all plasmablasts. The cells were identified with ELISPOT as Pnc-specific antibody-secreting cells (ASC) and as all immunoglobulin-secreting cells (ISC). High numbers of circulating Pnc-specific ASC were found in the acute phase of the disease in all patients with pneumonia (median 97 ASC/10(6) PBMC), but in none of the controls. IgG isotype predominated in 9/16 patients. The numbers of ISC were significantly higher in the patients than in the healthy controls, yet Pnc-specific ASC only accounted for 0.7% of all the patients' ISC.The present study is the first to show that antigen-specific plasmablasts appear in the circulation in pneumonia, suggesting that pulmonary lypmhocytes recirculate in humans. Assessing these cells provides a novel tool for studying immune response to antigens encountered at the LRT.
Collapse
Affiliation(s)
- Nina V. Palkola
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Medicine, Division of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
| | - Sari H. Pakkanen
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Medicine, Division of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
| | - Jussi M. Kantele
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Niina Rossi
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Medicine, Division of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
| | - Ritvaleena Puohiniemi
- Department of Bacteriology, Helsinki University Hospital Laboratory (HUSLAB), Helsinki, Finland
| | - Anu Kantele
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Medicine, Division of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
- Department of Medicine, Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
181
|
Frölich D, Blassfeld D, Reiter K, Giesecke C, Daridon C, Mei HE, Burmester GR, Goldenberg DM, Salama A, Dörner T. The anti-CD74 humanized monoclonal antibody, milatuzumab, which targets the invariant chain of MHC II complexes, alters B-cell proliferation, migration, and adhesion molecule expression. Arthritis Res Ther 2012; 14:R54. [PMID: 22404985 PMCID: PMC3446420 DOI: 10.1186/ar3767] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 02/25/2012] [Accepted: 03/09/2012] [Indexed: 01/21/2023] Open
Abstract
Introduction Targeting CD74 as the invariant chain of major histocompatibility complexes (MHC) became possible by the availability of a specific humanized monoclonal antibody, milatuzumab, which is under investigation in patients with hematological neoplasms. CD74 has been reported to regulate chemo-attractant migration of macrophages and dendritic cells, while the role of CD74 on peripheral naïve and memory B cells also expressing CD74 remains unknown. Therefore, the current study addressed the influence of milatuzumab on B-cell proliferation, chemo-attractant migration, and adhesion molecule expression. Methods Surface expression of CD74 on CD27- naïve and CD27+ memory B cells as well as other peripheral blood mononuclear cells (PBMCs) obtained from normals, including the co-expression of CD44, CXCR4, and the adhesion molecules CD62L, β7-integrin, β1-integrin and CD9 were studied after binding of milatuzumab using multicolor flow cytometry. The influence of the antibody on B-cell proliferation and migration was analyzed in vitro in detail. Results In addition to monocytes, milatuzumab also specifically bound to human peripheral B cells, with a higher intensity on CD27+ memory versus CD27- naïve B cells. The antibody reduced B-cell proliferation significantly but moderately, induced enhanced spontaneous and CXCL12-dependent migration together with changes in the expression of adhesion molecules, CD44, β7-integrin and CD62L, mainly of CD27- naïve B cells. This was independent of macrophage migration-inhibitory factor as a ligand of CD74/CD44 complexes. Conclusions Milatuzumab leads to modestly reduced proliferation, alterations in migration, and adhesion molecule expression preferentially of CD27- naïve B cells. It thus may be a candidate antibody for the autoimmune disease therapy by modifying B cell functions.
Collapse
Affiliation(s)
- Daniela Frölich
- Department of Medicine, Rheumatology and Clinical Immunology, Charité - University Medicine Berlin, Chariteplatz 1, Berlin 10117, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Hayden CA, Streatfield SJ, Lamphear BJ, Fake GM, Keener TK, Walker JH, Clements JD, Turner DD, Tizard IR, Howard JA. Bioencapsulation of the hepatitis B surface antigen and its use as an effective oral immunogen. Vaccine 2012; 30:2937-42. [PMID: 22406456 DOI: 10.1016/j.vaccine.2012.02.072] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 02/05/2012] [Accepted: 02/25/2012] [Indexed: 01/14/2023]
Abstract
Hepatitis B remains a major global health problem despite the availability of a safe and effective vaccine. Segments of the population lack access to or respond poorly to the parenteral vaccine, perpetuating the infection-transmission cycle. A low cost, orally delivered vaccine has the potential to alleviate many of these problems. Here we describe the expression of a bioencapsulated hepatitis B surface antigen (HBsAg) in maize and its immunogenicity, demonstrating for the first time a commercially feasible oral subunit vaccine production system for a major disease. This work surmounts previous barriers to plant-produced vaccines by expressing HBsAg at much higher levels and retaining antigen immunogenicity post-processing: factors which facilitated a robust immune response in mice without the need for an adjuvant. This method provides a practical solution to the delivery of a low-cost, stable oral vaccine.
Collapse
Affiliation(s)
- Celine A Hayden
- Applied Biotechnology Institute, Cal Poly Tech Park, San Luis Obispo, CA 93407, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Kiyono H, Okada K. [Mucosal immune systems--frontline of the biological defence mechanism]. ACTA ACUST UNITED AC 2012; 114:843-50. [PMID: 22295432 DOI: 10.3950/jibiinkoka.114.843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
184
|
Lindner C, Wahl B, Föhse L, Suerbaum S, Macpherson AJ, Prinz I, Pabst O. Age, microbiota, and T cells shape diverse individual IgA repertoires in the intestine. ACTA ACUST UNITED AC 2012; 209:365-77. [PMID: 22249449 PMCID: PMC3280880 DOI: 10.1084/jem.20111980] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Intestinal immunoglobulin A (IgA) ensures host defense and symbiosis with our commensal microbiota. Yet previous studies hint at a surprisingly low diversity of intestinal IgA, and it is unknown to what extent the diverse Ig arsenal generated by somatic recombination and diversification is actually used. In this study, we analyze more than one million mouse IgA sequences to describe the shaping of the intestinal IgA repertoire, its determinants, and stability over time. We show that expanded and infrequent clones combine to form highly diverse polyclonal IgA repertoires with very little overlap between individual mice. Selective homing allows expanded clones to evenly seed the small but not large intestine. Repertoire diversity increases during aging in a dual process. On the one hand, microbiota-, T cell-, and transcription factor RORγt-dependent but Peyer's patch-independent somatic mutations drive the diversification of expanded clones, and on the other hand, new clones are introduced into the repertoire of aged mice. An individual's IgA repertoire is stable and recalled after plasma cell depletion, which is indicative of functional memory. These data provide a conceptual framework to understand the dynamic changes in the IgA repertoires to match environmental and intrinsic stimuli.
Collapse
Affiliation(s)
- Cornelia Lindner
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
185
|
Lepage P, Van de Perre P. The Immune System of Breast Milk: Antimicrobial and Anti-inflammatory Properties. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 743:121-37. [DOI: 10.1007/978-1-4614-2251-8_9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
186
|
Ben Suleiman Y, Yoshida M, Nishiumi S, Tanaka H, Mimura T, Nobutani K, Yamamoto K, Takenaka M, Aoganghua A, Miki I, Ota H, Takahashi S, Matsui H, Nakamura M, Blumberg RS, Azuma T. Neonatal Fc receptor for IgG (FcRn) expressed in the gastric epithelium regulates bacterial infection in mice. Mucosal Immunol 2012; 5:87-98. [PMID: 22089027 PMCID: PMC3964614 DOI: 10.1038/mi.2011.53] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neonatal Fc receptors for immunoglobulin (Ig)G (FcRn) assume a central role in regulating host IgG levels and IgG transport across polarized epithelial barriers. We have attempted to elucidate the contribution of FcRn in controlling Helicobacter infection in the stomach. C57BL/6J wild-type or FcRn(-/-) mice were infected with Helicobacter heilmannii, and gastric lesions, bacterial load and the levels of antigen-specific IgG in serum and gastric juice were analyzed. The elevated levels of anti-H. heimannii IgG in gastric juice were observed exclusively in wild-type mice but not in FcRn(-/-) mice. In contrast, an increase in lymphoid follicles and bacterial loads along with deeper gastric epithelium invasion were noted in FcRn(-/-) mice. C57BL/6J wild-type or FcRn(-/-) mice were also infected with Helicobacter pylori SS1, and the results of the bacterial load in stomachs of these mice and the anti-H. pylori IgG levels in serum and gastric juice were similar to those from H. heilmannii infection. Our data suggest that FcRn can be functionally expressed in the stomach, which is involved in transcytosis of IgG, and prevent colonization by H. heilmannii and the associated pathological consequences of infection.
Collapse
Affiliation(s)
- Y Ben Suleiman
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - M Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
,The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan.
,Division of Metabolomics Research, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - S Nishiumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - H Tanaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - T Mimura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - K Nobutani
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - K Yamamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - M Takenaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - A Aoganghua
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - I Miki
- Department of Medical Pharmaceutics, Kobe Pharmaceutical University, Kobe, Japan.
| | - H Ota
- Department of Biomedical Laboratory Sciences, School of Health Sciences, Shinshu University School of Medicine, Nagano, Japan.
| | - S Takahashi
- Third Department of Internal Medicine, Kyorin University, Tokyo, Japan.
| | - H Matsui
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitsato University, Tokyo, Japan.
| | - M Nakamura
- Center for Clinical Pharmacy and Clinical Sciences, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan.
| | - RS Blumberg
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - T Azuma
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
187
|
B lymphocyte intestinal homing in inflammatory bowel disease. BMC Immunol 2011; 12:71. [PMID: 22208453 PMCID: PMC3261110 DOI: 10.1186/1471-2172-12-71] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 12/30/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is thought to be due to an abnormal interaction between the host immune system and commensal microflora. Within the intestinal immune system, B cells produce physiologically natural antibodies but pathologically atypical anti-neutrophil antibodies (xANCAs) are frequently observed in patients with IBD. The objective is to investigate the localisation of immunoglobulin-producing cells (IPCs) in samples of inflamed intestinal tissue taken from patients with IBD, and their possible relationship with clinical features. METHODS The IPCs in small intestinal, colonic and rectal biopsy specimens of patients with IBD were analysed by means of immunofluorescence using polyclonal rabbit anti-human Ig and goat anti-human IgM. The B cell phenotype of the IPC-positive samples was assessed using monoclonal antibodies specific for CD79, CD20, CD23, CD21, CD5, λ and κ chains. Statistical correlations were sought between the histological findings and clinical expression. RESULTS The study involved 96 patients (64 with ulcerative colitis and 32 with Crohn's disease). Two different patterns of B lymphocyte infiltrates were found in the intestinal tissue: one was characterised by a strong to moderate stromal localisation of small IgM+/CD79+/CD20-/CD21-/CD23-/CD5± IPCs (42.7% of cases); in the other (57.3%) no such small IPCs were detected in stromal or epithelial tissues. IPCs were significantly less frequent in the patients with Crohn's disease than in those with ulcerative colitis (p = 0.004). CONCLUSION Our findings suggest that different immunopathogenetic pathways underlie chronic intestinal inflammation with different clinical expressions. The presence of small B lymphocytes resembling B-1 cells also seemed to be negatively associated with Crohn's disease. It can therefore be inferred that the gut contains an alternative population of B cells that have a regulatory function.
Collapse
|
188
|
Micol R, Kayal S, Mahlaoui N, Beauté J, Brosselin P, Dudoit Y, Obenga G, Barlogis V, Aladjidi N, Kebaili K, Thomas C, Dulieu F, Monpoux F, Nové-Josserand R, Pellier I, Lambotte O, Salmon A, Masseau A, Galanaud P, Oksenhendler E, Tabone MD, Teira P, Coignard-Biehler H, Lanternier F, Join-Lambert O, Mouillot G, Theodorou I, Lecron JC, Alyanakian MA, Picard C, Blanche S, Hermine O, Suarez F, Debré M, Lecuit M, Lortholary O, Durandy A, Fischer A. Protective effect of IgM against colonization of the respiratory tract by nontypeable Haemophilus influenzae in patients with hypogammaglobulinemia. J Allergy Clin Immunol 2011; 129:770-7. [PMID: 22153772 DOI: 10.1016/j.jaci.2011.09.047] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 09/01/2011] [Accepted: 09/26/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Primary immunoglobulin deficiencies lead to recurrent bacterial infections of the respiratory tract and bronchiectasis, even with adequate immunoglobulin replacement therapy. It is not known whether patients able to secrete IgM (eg, those with hyper-IgM [HIgM] syndrome) are as susceptible to these infections as patients who lack IgM production (eg, those with panhypogammaglobulinemia [PHG]). OBJECTIVE This study is aimed at identifying specific microbiological and clinical (infections) characteristics that distinguish immunoglobulin-substituted patients with PHG from patients with HIgM syndrome. METHODS A cohort of patients with HIgM syndrome (n = 25) and a cohort of patients with PHG (n = 86) were monitored prospectively for 2 years while receiving similar polyvalent immunoglobulin replacement therapies. Regular bacterial analyses of nasal swabs and sputum were performed, and clinical events were recorded. In parallel, serum and saliva IgM antibody concentrations were measured. RESULTS When compared with patients with PHG, patients with HIgM syndrome were found to have a significantly lower risk of nontypeable Haemophilus influenzae carriage in particular (relative risk, 0.39; 95% CI, 0.21-0.63). Moreover, patients with HIgM syndrome (including those unable to generate somatic hypermutations of immunoglobulin genes) displayed anti-nontypeable H influenzae IgM antibodies in their serum and saliva. Also, patients with HIgM syndrome had a lower incidence of acute respiratory tract infections. CONCLUSIONS IgM antibodies appear to be microbiologically and clinically protective and might thus attenuate the infectious consequences of a lack of production of other immunoglobulin isotypes in patients with HIgM syndrome. Polyvalent IgG replacement therapy might not fully compensate for IgM deficiency. It might thus be worth adapting long-term antimicrobial prophylactic regimens according to the underlying B-cell immunodeficiency phenotype.
Collapse
Affiliation(s)
- Romain Micol
- CEREDIH Network (French National Reference Center for Primary Immunodeficiencies), Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Intranasal administration of a flagellin-adjuvanted inactivated influenza vaccine enhances mucosal immune responses to protect mice against lethal infection. Vaccine 2011; 30:466-74. [PMID: 22051136 DOI: 10.1016/j.vaccine.2011.10.058] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 09/16/2011] [Accepted: 10/22/2011] [Indexed: 11/21/2022]
Abstract
The influenza virus, a mucosal pathogen that infects the respiratory tract, is a major global health issue. There have been attempts to mucosally administer inactivated influenza vaccines to induce both mucosal and systemic immune responses. However, mucosally administered inactivated influenza vaccine has low immunogenicity, which is partially due to the lack of an effective mucosal adjuvant. The development of a safe and effective mucosal adjuvant is a prerequisite to the practical use of a mucosal inactivated influenza vaccine. We have previously demonstrated that a bacterial flagellin, Vibrio vulnificus FlaB, when mixed with antigen and administered intranasally, exerts a strong mucosal adjuvant activity by stimulating the Toll-like receptor 5 (TLR5). In this study, we tested whether the FlaB protein could serve as an effective mucosal adjuvant for an inactivated trivalent influenza vaccine (TIV) manufactured for humans; in a murine vaccination model, this vaccine consists of A/Brisbane/59/07 (H1N1 subtype), A/Uruguay/716/07 (H3N2 subtype), and B/Florida/4/06 (B type). Intranasal co-administration of the TIV with FlaB induced prominent humoral responses as demonstrated by high influenza-specific IgA levels in both the mucosal secretions and serum and significant specific IgG induction in the systemic compartment. The FlaB protein significantly potentiated influenza-specific cytokine production by draining lymph node cells and splenocytes. The FlaB mucosal adjuvant conferred excellent protection against a lethal challenge with a live virulent virus with high hemagglutination inhibition (HAI) antibody (Ab) titers. The FlaB did not accumulate in the olfactory nerve and epithelium, guaranteeing against a retrograde uptake into the central nervous system. These results suggest that FlaB can be used as a promising mucosal adjuvant for nasal inactivated influenza vaccine development.
Collapse
|
190
|
Analysis of plasma proteome from cases of the different traditional Chinese medicine syndromes in patients with chronic hepatitis B. J Pharm Biomed Anal 2011; 59:173-8. [PMID: 22030074 DOI: 10.1016/j.jpba.2011.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 09/30/2011] [Accepted: 10/04/2011] [Indexed: 12/12/2022]
Abstract
A proteomic analysis method, two dimensional gel electrophoresis (2-DE) followed by matrix-assisted laser desorption/ionization time-of-flight MS (MALDI-TOF-MS), was used to explore the link between plasma proteome and the different syndromes of traditional Chinese medicine (TCM) in patients with chronic hepatitis B (CHB). In compared with the plasma proteomes from health donors, the alterations in protein expression from cases of the five TCM syndromes, including damp heat stasis in the middle-Jiao syndrome, liver Qi stagnation and spleen deficiency syndrome, spleen and kidney Yang deficiency syndrome, liver and kidney Yin deficiency syndrome, and blood stasis into collateral syndrome with CHB were identified (P<0.05). In the cases of the five TCM syndromes with CHB, immunoglobulin J-chains (IGJ) and C-reactive protein (CRP) were up-regulated, while haptoglobin (HPT), retinol binding protein (RBP) and vitronectin were down-regulated. To further confirm these results, four proteins, including CRP, IGJ, HPT and RBP, from more plasma samples were quantified by ELISA. The results showed that the changes of protein levels were consistent with those from the 2-DE experiment. Importantly, the upregulation tendency of IGJ level in plasma is related with the different TCM syndromes with CHB (P<0.05). Our results show that IGJ may serve as a novel potential biomarker for diagnosis of the different TCM syndromes in patients with CHB.
Collapse
|
191
|
Ye J, Bromage E, Kaattari I, Kaattari S. Transduction of binding affinity by B lymphocytes: a new dimension in immunological regulation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:982-990. [PMID: 21300090 DOI: 10.1016/j.dci.2011.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 01/15/2011] [Accepted: 01/19/2011] [Indexed: 05/30/2023]
Abstract
To date, immunologists have operated with two primary paradigms governing the antibody response: (1) that affinity maturation is primarily dependent upon antigen-driven selection of both the germline and somatically amended repertoires, and (2) that antibody effector function is isotypically determined. The teleost model now suggests that these classical paradigms should be broadened to incorporate the ability of the B cell to transduce the strength of antigen recognition (affinity) into structural modifications of its antibody product, which, in turn, modulates the antibody's effector function. Although this relationship, thus far, has only been examined and demonstrated in the teleost, we find a number of the individual elements of this structural/functional relationship have been reported for mammalian IgM, which prompts future investigations into its universality. In sum, these findings suggest a heretofore unrecognized feature of B lymphocyte affinity discrimination, which transduces the affinity of antigen recognition into functionally modified antibodies.
Collapse
Affiliation(s)
- Jianmin Ye
- Virginia Institute of Marine Science, College of William and Mary, Gloucester Point, VA 23062, United States
| | | | | | | |
Collapse
|
192
|
Riddle MS, Kaminski RW, Williams C, Porter C, Baqar S, Kordis A, Gilliland T, Lapa J, Coughlin M, Soltis C, Jones E, Saunders J, Keiser PB, Ranallo RT, Gormley R, Nelson M, Turbyfill KR, Tribble D, Oaks EV. Safety and immunogenicity of an intranasal Shigella flexneri 2a Invaplex 50 vaccine. Vaccine 2011; 29:7009-19. [DOI: 10.1016/j.vaccine.2011.07.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/07/2011] [Accepted: 07/11/2011] [Indexed: 11/25/2022]
|
193
|
Mesin L, Di Niro R, Thompson KM, Lundin KEA, Sollid LM. Long-lived plasma cells from human small intestine biopsies secrete immunoglobulins for many weeks in vitro. THE JOURNAL OF IMMUNOLOGY 2011; 187:2867-74. [PMID: 21841131 DOI: 10.4049/jimmunol.1003181] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To understand the biology of Ab-secreting cells in the human small intestine, we examined Ab production of intestinal biopsies kept in culture. We found sustained IgA and IgM secretion as well as viable IgA- or IgM-secreting cells after >4 wk of culture. The Ab-secreting cells were nonproliferating and expressing CD27 and CD138, thus having a typical plasma cell phenotype. Culturing of biopsies without tissue disruption gave the highest Ab production and plasma cell survival suggesting that the environment regulates plasma cell longevity. Cytokine profiling of the biopsy cultures demonstrated a sustained presence of IL-6 and APRIL. Blocking of the activity of endogenous APRIL and IL-6 with BCMA-Fc and anti-human IL-6 Ab demonstrated that both these factors were essential for plasma cell survival and Ab secretion in the biopsy cultures. This study demonstrates that the human small intestine harbors a population of nonproliferating plasma cells that are instructed by the microenvironment for prolonged survival and Ab secretion.
Collapse
Affiliation(s)
- Luka Mesin
- Center for Immune Regulation, University of Oslo, Q3 N-0027 Oslo, Norway.
| | | | | | | | | |
Collapse
|
194
|
Fox RI. The importance of minor salivary gland biopsy in prediction of lymphoma in Sjögren's syndrome: should we be obtaining more information about prognosis from minor salivary gland samples? Ann Rheum Dis 2011; 70:1351-3. [PMID: 21715358 DOI: 10.1136/ard.2011.152751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
195
|
Brandtzaeg P. The gut as communicator between environment and host: immunological consequences. Eur J Pharmacol 2011; 668 Suppl 1:S16-32. [PMID: 21816150 DOI: 10.1016/j.ejphar.2011.07.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 07/01/2011] [Accepted: 07/07/2011] [Indexed: 12/18/2022]
Abstract
During human evolution, the mucosal immune system developed two anti-inflammatory mechanisms: immune exclusion by secretory antibodies (SIgA and SIgM) to control epithelial colonization of microorganisms and inhibit penetration of harmful substances; and immunosuppression to counteract local and peripheral hypersensitivity against innocuous antigens such as food proteins. The latter function is referred to as oral tolerance when induced via the gut. Similar mechanisms also control immunity to commensal bacteria. The development of immune homeostasis depends on "windows of opportunity" where adaptive and innate immunities are coordinated by antigen-presenting cells; their function is not only influenced by microbial products but also by dietary constituents, including vitamin A and lipids like polyunsaturated omega-3 fatty acids. These factors can in several ways exert beneficial effects on the immunophenotype of the infant. Also breast milk provides immune-modulating factors and SIgA antibodies - reinforcing the gut barrier. Mucosal immunity is most abundantly expressed in the gut, and the intestinal mucosa of an adult contains at least 80% of the body's activated B cells - terminally differentiated to plasmablasts and plasma cells (PCs). Most mucosal PCs produce dimeric IgA which is exported by secretory epithelia expressing the polymeric Ig receptor (pIgR), also called membrane secretory component (SC). Immune exclusion is therefore performed mainly by SIgA. Notably, pIgR knockout mice which lack SIgs show increased uptake of food and microbial antigens and they have a hyper-reactive immune system with disposition for anaphylaxis; but this untoward development is counteracted by cognate oral tolerance induction as a homeostatic back-up mechanism.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology, Centre for Immune Regulation, University of Oslo, and Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| |
Collapse
|
196
|
Brandtzaeg P. Potential of Nasopharynx-associated Lymphoid Tissue for Vaccine Responses in the Airways. Am J Respir Crit Care Med 2011; 183:1595-604. [DOI: 10.1164/rccm.201011-1783oc] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
197
|
Production of the plasma-cell survival factor a proliferation-inducing ligand (APRIL) peaks in myeloid precursor cells from human bone marrow. Blood 2011; 118:1838-44. [PMID: 21642598 DOI: 10.1182/blood-2011-01-332940] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The bone marrow (BM) is an organ extremely efficient in mediating long-term survival of plasma cells (PCs), ensuring an immune humoral memory. This implies that the BM must provide continuously key PC survival factors. Our results show that the BM is an organ constitutively rich in a proliferation-inducing ligand (APRIL), a member of the tumor necrosis factor superfamily implicated in PC survival. APRIL production is induced during hematopoiesis in myeloid cells by non-lineage-committing factors such as stem cell factor, thrombopoietin, IL-3, and FMS-like tyrosine kinase 3 ligand. Notably, APRIL production, both in the human and mouse systems, peaks in myeloid precursor cells, before dropping in fully mature granulocytes. Myeloid cells secrete APRIL that circulates freely in BM plasma to act on PCs, usually at distance from APRIL production sites. Selective APRIL in vivo antagonism and in vitro coculture experiments further demonstrated that myeloid precursor cells mediates PC survival in an APRIL-dependent manner Thus, APRIL production by myeloid precursor cells shows that the 2 main BM functions, hematopoiesis and long-term PC survival, are linked. Such constitutive and high APRIL production may explain why BM mediates long-term PC survival.
Collapse
|
198
|
Medrano M, Racedo SM, Rolny IS, Abraham AG, Pérez PF. Oral administration of kefiran induces changes in the balance of immune cells in a murine model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:5299-5304. [PMID: 21504180 DOI: 10.1021/jf1049968] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The aim of the present study was to evaluate the effect of the oral administration of kefiran on the balance of immune cells in a murine model. Six week old BALB/c mice were treated with kefiran (300 mg/L) for 0, 2 and 7 days. Kefiran treatment increased the number of IgA+ cells in lamina propria after 2 and 7 days. Percentage of B220+/MHCII(high) cells in mesenteric lymph nodes (2 days) and Peyer's patches (7 days) was higher compared to untreated control mice. An increase of macrophages (F4/80+ cells) was observed in lamina propria and peritoneal cavity (2 and 7 days). In contrast, at day 7, macrophage population decreased in Peyer's patches. These results show the ability of kefiran to modify the balance of immune cells in intestinal mucosa. This property could be highly relevant for the comprehension of the probiotic effect attributed to kefir.
Collapse
Affiliation(s)
- Micaela Medrano
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos, CCT La Plata, CONICET, UNLP, La Plata, Argentina
| | | | | | | | | |
Collapse
|
199
|
Stappenbeck TS, Rioux JD, Mizoguchi A, Saitoh T, Huett A, Darfeuille-Michaud A, Wileman T, Mizushima N, Carding S, Akira S, Parkes M, Xavier RJ. Crohn disease: a current perspective on genetics, autophagy and immunity. Autophagy 2011; 7:355-74. [PMID: 20729636 PMCID: PMC3842289 DOI: 10.4161/auto.7.2.13074] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 04/17/2010] [Indexed: 12/13/2022] Open
Abstract
Crohn disease (CD) is a chronic and debilitating inflammatory condition of the gastrointestinal tract. Prevalence in Western populations is 100-150/100,000 and somewhat higher in Ashkenazi Jews. Peak incidence is in early adult life, although any age can be affected and a majority of affected individuals progress to relapsing and chronic disease. Medical treatments rely significantly on empirical corticosteroid therapy and immunosuppression, and intestinal resectional surgery is frequently required. Thus, 80% of patients with CD come to surgery for refractory disease or complications. It is hoped that an improved understanding of pathogenic mechanisms, for example by studying the genetic basis of CD and other forms of inflammatory bowel diseases (IBD), will lead to improved therapies and possibly preventative strategies in individuals identified as being at risk.
Collapse
Affiliation(s)
- Thaddeus S. Stappenbeck
- Departments of Pathology and Immunology; Washington University School of Medicine; St. Louis, MO USA
| | - John D. Rioux
- Université de Montréal; Montréal, Québec Canada
- Montreal Heart Institute; Montréal, Québec Canada
| | - Atsushi Mizoguchi
- Center for the Study of Inflammatory Bowel Disease; Massachusetts General Hospital and Harvard Medical School; Boston, MA USA
- Department of Pathology; Massachusetts General Hospital and Harvard Medical School; Boston, MA USA
| | - Tatsuya Saitoh
- Laboratory of Host Defense; WPI Immunology Frontier Research Center; Osaka University; Suita, Osaka Japan
- Department of Host Defense Osaka; Japan
| | - Alan Huett
- Center for the Study of Inflammatory Bowel Disease; Massachusetts General Hospital and Harvard Medical School; Boston, MA USA
| | | | - Tom Wileman
- Infection and Immunity; School of Medicine; Faculty of Health; University of East Anglia; East Anglia, Norfolk UK
| | - Noboru Mizushima
- Department of Physiology and Cell Biology at Tokyo Medical and Dental University; Bunkyo-ku, Tokyo Japan
| | | | - Shizuo Akira
- Laboratory of Host Defense; WPI Immunology Frontier Research Center; Osaka University; Suita, Osaka Japan
- Department of Host Defense Osaka; Japan
| | - Miles Parkes
- IBD Research Group; Addenbrooke’s Hospital; University of Cambridge; Cambridge UK
| | - Ramnik J. Xavier
- Center for the Study of Inflammatory Bowel Disease; Massachusetts General Hospital and Harvard Medical School; Boston, MA USA
| |
Collapse
|
200
|
Stappenbeck TS, Rioux JD, Mizoguchi A, Saitoh T, Huett A, Darfeuille-Michaud A, Wileman T, Mizushima N, Carding S, Akira S, Parkes M, Xavier RJ. Crohn disease: a current perspective on genetics, autophagy and immunity. Autophagy 2011. [PMID: 20729636 DOI: 10.4161/auto.7.4.13074] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Crohn disease (CD) is a chronic and debilitating inflammatory condition of the gastrointestinal tract. Prevalence in Western populations is 100-150/100,000 and somewhat higher in Ashkenazi Jews. Peak incidence is in early adult life, although any age can be affected and a majority of affected individuals progress to relapsing and chronic disease. Medical treatments rely significantly on empirical corticosteroid therapy and immunosuppression, and intestinal resectional surgery is frequently required. Thus, 80% of patients with CD come to surgery for refractory disease or complications. It is hoped that an improved understanding of pathogenic mechanisms, for example by studying the genetic basis of CD and other forms of inflammatory bowel diseases (IBD), will lead to improved therapies and possibly preventative strategies in individuals identified as being at risk.
Collapse
Affiliation(s)
- Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|