1
|
Beller A, Kruglov A, Durek P, von Goetze V, Werner K, Heinz GA, Ninnemann J, Lehmann K, Maier R, Hoffmann U, Riedel R, Heiking K, Zimmermann J, Siegmund B, Mashreghi MF, Radbruch A, Chang HD. Specific microbiota enhances intestinal IgA levels by inducing TGF-β in T follicular helper cells of Peyer's patches in mice. Eur J Immunol 2020; 50:783-794. [PMID: 32065660 DOI: 10.1002/eji.201948474] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 11/15/2019] [Accepted: 02/14/2020] [Indexed: 12/31/2022]
Abstract
In humans and mice, mucosal immune responses are dominated by IgA antibodies and the cytokine TGF-β, suppressing unwanted immune reactions but also targeting Ig class switching to IgA. It had been suggested that eosinophils promote the generation and maintenance of mucosal IgA-expressing plasma cells. Here, we demonstrate that not eosinophils, but specific bacteria determine mucosal IgA production. Co-housing of eosinophil-deficient mice with mice having high intestinal IgA levels, as well as the intentional microbiota transfer induces TGF-β expression in intestinal T follicular helper cells, thereby promoting IgA class switching in Peyer's patches, enhancing IgA+ plasma cell numbers in the small intestinal lamina propria and levels of mucosal IgA. We show that bacteria highly enriched for the genus Anaeroplasma are sufficient to induce these changes and enhance IgA levels when adoptively transferred. Thus, specific members of the intestinal microbiota and not the microbiota as such regulate gut homeostasis, by promoting the expression of immune-regulatory TGF-β and of mucosal IgA.
Collapse
Affiliation(s)
- Alexander Beller
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Andrey Kruglov
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany.,Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Victoria von Goetze
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Katharina Werner
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Gitta Anne Heinz
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Justus Ninnemann
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Katrin Lehmann
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - René Maier
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Ute Hoffmann
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - René Riedel
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany.,Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Kevin Heiking
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Jakob Zimmermann
- Maurice Müller Laboratories, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Britta Siegmund
- Medical Department I (Gastroenterology, Infectiology, and Rheumatology), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| | - Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Berlin, Germany
| |
Collapse
|
2
|
Wang Y, Zhao H, Liu J, Shao Y, Li J, Luo L, Xing M. Copper and arsenic-induced oxidative stress and immune imbalance are associated with activation of heat shock proteins in chicken intestines. Int Immunopharmacol 2018; 60:64-75. [DOI: 10.1016/j.intimp.2018.04.038] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 12/31/2022]
|
3
|
Efficient induction of comprehensive immune responses to control pathogenic E. coli by clay nano-adjuvant with the moderate size and surface charge. Sci Rep 2017; 7:13367. [PMID: 29042573 PMCID: PMC5645426 DOI: 10.1038/s41598-017-13570-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023] Open
Abstract
In recent decades, diseases caused by pathogenic Escherichia coli (E. coli), enterohaemorrhagic E. coli (EHEC) O26 have been increasingly reported worldwide, which are as severe as those caused by EHEC strain O157:H7 and require effective intervention strategies. Herein, we report the application of clay nanoparticles, i.e. hectorites as effective nano-adjuvants for vaccination against EHEC O26 colonization. We show that medium size HEC (hectorite, around 73~77 nm diameter) is able to induce efficient humoral and cellular immune responses against EHEC antigen - intimin β (IB), which are significantly higher than those triggered by commercially used adjuvants - QuilA and Alum. We also demonstrate that mice immunized with IB adjuvanted with HEC nanoparticles elicit sufficient secretion of mucosal IgA, capable of providing effective protection against EHEC O26 binding to ruminant and human cells. In addition, we demonstrate for the first time that hectorites are able to initiate maturation of RAW 264.7 macrophages, inducing expression of co-stimulatory cytokines at a low nanoparticle concentration (10 μg/mL). Together these data strongly suggest that hectorite with optimized size is a highly efficient vaccine nano-adjuvant.
Collapse
|
4
|
|
5
|
Guo X, Jiang X, Xiao Y, Zhou T, Guo Y, Wang R, Zhao Z, Xiao H, Hou C, Ma L, Lin Y, Lang X, Feng J, Chen G, Shen B, Han G, Li Y. IL-17A signaling in colonic epithelial cells inhibits pro-inflammatory cytokine production by enhancing the activity of ERK and PI3K. PLoS One 2014; 9:e89714. [PMID: 24586980 PMCID: PMC3934915 DOI: 10.1371/journal.pone.0089714] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 01/22/2014] [Indexed: 12/14/2022] Open
Abstract
Our previous data suggested that IL-17A contributes to the inhibition of Th1 cell function in the gut. However, the underlying mechanisms remain unclear. Here we demonstrate that IL-17A signaling in colonic epithelial cells (CECs) increases TNF-α-induced PI3K-AKT and ERK phosphorylation and inhibits TNF-α induced expression of IL-12P35 and of a Th1 cell chemokine, CXCL11 at mRNA level. In a co-culture system using HT-29 cells and PBMCs, IL-17A inhibited TNF-α-induced IL-12P35 expression by HT-29 cells and led to decreased expression of IFN-γ and T-bet by PBMCs. Finally, adoptive transfer of CECs from mice with Crohn's Disease (CD) led to an enhanced Th1 cell response and exacerbated colitis in CD mouse recipients. The pathogenic effect of CECs derived from CD mice was reversed by co-administration of recombinant IL-17A. Our data demonstrate a new IL-17A-mediated regulatory mechanism in CD. A better understanding of this pathway might shed new light on the pathogenesis of CD.
Collapse
Affiliation(s)
- Xiaoqin Guo
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
- Department of Immunology, Logistics College of Chinese People’s Armed Police Forces, Tianjin, China
| | - Xingwei Jiang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yan Xiao
- Department of Respiratory Medicine, First Affiliated Hospital of the Chinese PLA General Hospital, Beijing, China
| | - Tingting Zhou
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yueling Guo
- Department of Medical Engineering, Military General Hospital of Beijing PLA, Beijing, China
| | - Renxi Wang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhi Zhao
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - He Xiao
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chunmei Hou
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lingyun Ma
- Department of Respiratory Medicine, First Affiliated Hospital of the Chinese PLA General Hospital, Beijing, China
| | - Yanhua Lin
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiaoling Lang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jiannan Feng
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Guojiang Chen
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Beifen Shen
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Gencheng Han
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yan Li
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Ryan SO, Leal SM, Abbott DW, Pearlman E, Cobb BA. Mgat2 ablation in the myeloid lineage leads to defective glycoantigen T cell responses. Glycobiology 2013; 24:262-71. [PMID: 24310166 DOI: 10.1093/glycob/cwt107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
N-linked glycosylation is a central regulatory factor that influences the immune system in varied and profound ways, including leukocyte homing, T cell receptor signaling and others. Moreover, N-glycan branching has been demonstrated to change as a function of infection and inflammation. Our previous findings suggest that complex-type N-glycans on the class II major histocompatibility complex play an important role in antigen selection within antigen presenting cells (APCs) such that highly branched N-glycans promote polysaccharide (glycoantigen, GlyAg) presentation following Toll-like receptor 2 (TLR2)-dependent antigen processing. In order to explore the impact of N-glycan branching on the myeloid-derived APC population without the confounding problems of altering the branching of lymphocytes and non-hematopoietic cells, we created a novel myeloid-specific knockout of the β-1,2-N-acetylglucosaminyltransferase II (Mgat2) enzyme. Using this novel mouse, we found that the reduction in multi-antennary N-glycans characteristic of Mgat2 ablation had no impact on GlyAg-mediated TLR2 signaling. Likewise, no deficits in antigen uptake or cellular homing to lymph nodes were found. However, we discovered that Mgat2 ablation prevented GlyAg presentation and T cell activation in vitro and in vivo without apparent alterations in protein antigen response or myeloid-mediated protection from infection. These findings demonstrate that GlyAg presentation can be regulated by the N-glycan branching pattern of APCs, thereby establishing an in vivo model where the T cell-dependent activity of GlyAgs can be experimentally distinguished from GlyAg-mediated stimulation of the innate response through TLR2.
Collapse
|
7
|
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: 213] [Impact Index Per Article: 19.4] [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
|
8
|
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: 187] [Impact Index Per Article: 17.0] [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
|
9
|
Abstract
There is currently a major focus on the role of the gut barrier function in balancing mucosal immune responses. Increased epithelial permeability for exogenous antigens is a crucial primary or secondary event in the pathogenesis of several disorders affecting body surfaces and beyond. The epithelial gate-keeper function is determined by the individual's age (e.g. preterm vs. term infant), diet, genetics, mucus composition, interactions between mast cells, nerves and neuropeptides, concurrent infection, the commensal microbiota and the epithelium-shielding effect of secretory IgA (SIgA) antibodies provided by breast milk or produced in the individual's gut. The integrity of the epithelial barrier furthermore depends on homeostatic regulatory mechanisms, including mucosal induction of regulatory T cells, where commensal microbiota-host interactions apparently play decisive roles. Thus, both extrinsic and intrinsic factors have been identified that may have an impact on the dynamics of the epithelial cell-cell junctions in the gut and thereby increase or reduce paracellular permeability. Experiments have shown that SIgA normally cooperates with innate defence factors to protect the epithelium and reinforce its barrier function. In the absence of SIgA commensal gut bacteria overstimulate innate epithelial immunity at the expense of expression of genes that regulate fat and carbohydrate metabolism, resulting in an epithelial gene signature that correlates with the development of lipid malabsorption. This shows that the intestinal epithelial barrier is a cross-road between defence and nutrition, and that SIgA is essential to keep the balance between these two functions.
Collapse
Affiliation(s)
- P. Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo
- Department of Pathology, Oslo University Hospital, Rikshospitalet, P.O. Box 4950, 0424 Oslo, Norway
| |
Collapse
|
10
|
Introduction: Anti-inflammatory Treatment of Childhood Asthma: Cromoglycate and Nedocromil as Non-steroidal Alternatives? Mediators Inflamm 2012; 3:S5. [PMID: 18475606 PMCID: PMC2365601 DOI: 10.1155/s0962935194000670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
11
|
Abstract
PURPOSE OF REVIEW To review recent findings dealing with the involvement of mucosal immunoglobulin A (IgA) in the gut barrier function and various gastrointestinal diseases. New information will be discussed in the context of previous knowledge in this field. RECENT FINDINGS The epithelial barrier function seems to be central in many mucosal disorders because it is decisive for host-microbial interactions and penetration of soluble antigens into the lamina propria. Secretory IgA contributes to the barrier function and recent evidence strongly supports the notion that such antibodies are involved in immunological homeostasis. SUMMARY Inflammatory bowel disease involves a break of tolerance to the commensal microbiota. Aberrations in the mucosal IgA system may, therefore, be part of the inflammatory bowel disease pathogenesis. In gluten-induced enteropathy, however, it has been suggested that a mucosal IgA response may promote the progression of celiac disease and dermatitis herpetiformis by enhancing the uptake of gluten peptides and inhibiting the enzyme activity of tissue transglutaminase. A mucosal IgA response may also promote gastritis by protecting Helicobacter pylori from complement attack. In food allergy, several facets of the epithelial barrier function may show deficiency, including secretory IgA.
Collapse
|
12
|
Forsythe P, Bienenstock J. Immunomodulation by commensal and probiotic bacteria. Immunol Invest 2010; 39:429-48. [PMID: 20450286 DOI: 10.3109/08820131003667978] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over the past decade there has been an increasing awareness of the role played by commensal bacteria in modulating mucosal immune responses and as a consequence there is now great interest in the therapeutic potential of probiotics and other bacteria based strategies for a range of immune disorders. Here we review current understanding of the mechanisms underlying the immunomodulatory actions of commensal and probiotic bacteria and probiotic organisms. We discuss prominent cell types involved in transducing signals from these bacteria, including epithelial cells, dendritic cells and T regulatory cells. We also draw attention to emerging data indicating interplay between the gut microbiota, enteric neurons and the immune system. There is a focus on the specific aspects of bacteria-host interactions that may influence the ability of a specific organism to confer potentially beneficial changes in immune responses. It is clear that there is still much to learn regarding the determinants of the diverse immune responses elicited by different bacterial strains by building on our current knowledge in these areas it may be possible to design clinically effective, bacteria based strategies to maintain and promote health.
Collapse
Affiliation(s)
- Paul Forsythe
- The Brain Body Institute, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
13
|
Gallagher S, Phillips AC, Evans P, Der G, Hunt K, Carroll D. Caregiving is associated with low secretion rates of immunoglobulin A in saliva. Brain Behav Immun 2008; 22:565-72. [PMID: 18178376 DOI: 10.1016/j.bbi.2007.11.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 11/13/2007] [Accepted: 11/22/2007] [Indexed: 11/30/2022] Open
Abstract
Although the chronic stress of caring for a sick/disabled relative has been associated with poorer immunity using a range of outcomes, its impact on secretory immunoglobulin A (S-IgA) in saliva has yet to be examined. Three hypotheses were tested in analyses of data from a large community sample: first, caregivers would have lower S-IgA secretion rates than non-caregivers; second, the impact of caregiving on S-IgA would be particularly apparent in older participants; third, for caregivers, caregiving burden would be negatively associated with S-IgA. The sample comprised three distinct age cohorts, one young (N=623), one middle aged (N=639), and the other elderly (N=582). Participants were classified as caregivers if they regularly cared for somebody other than routine childcare. Caregiving strain was measured and a caregiving burden index was then derived as the composite of the number of people being cared for, the type of care provided, and the residential status of the person being cared for. From 2-min saliva samples, S-IgA secretion rate was measured. There was a significant caregiver status by age cohort interaction; caregivers in the eldest cohort had lower S-IgA secretion rates than their non-caregiving counterparts. Caregiving strain and burden and S-IgA were related, such that caregivers who experienced greater strain and burden had lower S-IgA secretion rates. These findings resonate with those from other studies using different immune outcomes. Considered together, it is clear that that the chronic stress of caregiving has widespread effects on immunity.
Collapse
Affiliation(s)
- Stephen Gallagher
- School of Sport and Exercise Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, England, United Kingdom
| | | | | | | | | | | |
Collapse
|
14
|
Takizawa R, Pawankar R, Yamagishi S, Takenaka H, Yagi T. Increased expression of HLA-DR and CD86 in nasal epithelial cells in allergic rhinitics: antigen presentation to T cells and up-regulation by diesel exhaust particles. Clin Exp Allergy 2007; 37:420-33. [PMID: 17359392 PMCID: PMC7164828 DOI: 10.1111/j.1365-2222.2007.02672.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND A proportion of nasal epithelial cells (NEC) in patients with allergic rhinitis (AR) are known to express the major histocompatibility complex Class II molecule (HLA-DR). OBJECTIVE We hypothesized that NEC may play a role in antigen presentation to T cells. To elucidate the possible role of NEC in antigen presentation, we examined the expression of HLA-DR, CD80 and CD86 in NEC, their regulation by cytokines and the capacity of NEC to induce antigen-specific proliferation of T cells. METHODS We examined the expression of HLA-DR, CD80 and CD86 in nasal epithelial scrapings of patients with seasonal allergic rhinitis (SAR) to Japanese cedar pollen pre-season and in-season, by immunohistochemistry. Next, we examined the effect of IL-1beta, TNF-alpha, (IFN-gamma), IL-4 alpha, IL-13 and diesel exhaust particles (DEP) on the HLA-DR, CD80 and CD86 expression in cultured nasal epithelial cells (CNEC), by flow cytometry. Further, we analysed the capacity of mite antigen (Der f II)-pulsed mitomycin-C-treated CNEC to induce proliferation of autologous T cells from patients with perennial allergic rhinitis. RESULTS NEC constitutively expressed HLA-DR and CD86, but not CD80. The expression of HLA-DR and CD86 in NEC was significantly increased in-season, in patients with SAR as compared with that of pre-season. While IFN-gamma up-regulated the expression of HLA-DR, IL-1beta and TNF-alpha up-regulated the expression of CD86 in CNEC. Furthermore, in the presence of mite antigen, CNEC induced the proliferation of autologous peripheral blood T lymphocytes. Anti-CD86 and anti-HLA-DR monoclonal antibody but not anti-CD80 inhibited the epithelial cell-induced T cell proliferation. Stimulation with a combination of DEP and mite antigen significantly up-regulated HLA-DR and CD86 expression in CNEC. CONCLUSIONS These studies suggest that NEC in patients with AR may play a role in antigen presentation through the enhanced expression of HLA-DR and CD86. Furthermore, these results suggest the possibility that DEP may enhance the antigen-presenting function of CNEC.
Collapse
Affiliation(s)
- R Takizawa
- Department of Otolaryngology, Nippon Medical School, Tokyo, Japan
| | | | | | | | | |
Collapse
|
15
|
Brandtzaeg P. Induction of secretory immunity and memory at mucosal surfaces. Vaccine 2007; 25:5467-84. [PMID: 17227687 DOI: 10.1016/j.vaccine.2006.12.001] [Citation(s) in RCA: 340] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 11/08/2006] [Accepted: 12/01/2006] [Indexed: 11/27/2022]
Abstract
Mucosal epithelia comprise an extensive vulnerable barrier which is reinforced by numerous innate defence mechanisms cooperating intimately with adaptive immunity. Local generation of secretory IgA (SIgA) constitutes the largest humoral immune system of the body. Secretory antibodies function both by performing antigen exclusion at mucosal surfaces and by virus and endotoxin neutralization within epithelial cells without causing tissue damage. SIgA is thus persistently containing commensal bacteria outside the epithelial barrier but can also target invasion of pathogens and penetration of harmful antigens. Resistance to toxin-producing bacteria such as Vibrio cholerae and enterotoxigenic Escherichia coli appears to depend largely on SIgA, and so does herd protection against horizontal faecal-oral spread of enteric pathogens under naïve or immunized conditions--with a substantial innate impact both on cross-reactivity and memory. Like natural infections, live mucosal vaccines or adequate combinations of non-replicating vaccines and mucosal adjuvants, give rise not only to SIgA antibodies but also to longstanding serum IgG and IgA responses. However, there is considerably disparity with regard to migration of memory/effector cells from mucosal inductive sites to secretory effector sites and systemic immune organs. Also, although immunological memory is generated after mucosal priming, this may be masked by a self-limiting response protecting the inductive lymphoid tissue in the gut. The intranasal route of vaccine application targeting nasopharynx-associated lymphoid tissue may be more advantageous for certain infections, but only if successful stimulation is achieved without the use of toxic adjuvants that might reach the central nervous system. The degree of protection obtained after mucosal vaccination ranges from reduction of symptoms to complete inhibition of re-infection. In this scenario, it is often difficult to determine the relative importance of SIgA versus serum antibodies, but infection models in knockout mice strongly support the notion that SIgA exerts a decisive role in protection and cross-protection against a variety of infectious agents. Nevertheless, relatively few mucosal vaccines have been approved for human use, and more basic work is needed in vaccine and adjuvant design, including particulate or live-vectored combinations.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology, Institute and Department of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Centre, N-0027 Oslo, Norway.
| |
Collapse
|
16
|
Abstract
The intestinal epithelium is a selective barrier where incompletely-digested food antigens are transmitted to the immune system. Food antigens are often the starting point of intestinal diseases such as food allergy or coeliac disease. The intestinal epithelial cells (IEC) take up and process food antigens mainly by fluid-phase transcytosis involving two functional pathways, one minor direct pathway without degradation and another major lysosomal degradative pathway. Among the peptidic metabolites generated during transepithelial transport of luminal antigens, some have a molecular mass compatible with a binding to restriction (major histocompatibility complex; MHC) molecules; the latter can be up regulated on enterocytes, especially in inflammatory conditions. Indeed, interferon-γ not only increases the paracellular absorption of antigens, but also their transcytosis across epithelial cells. It has been reported that enterocytes may even directly present peptidic epitopes to underlying T-cells. As a new potential way of transmitting peptidic information to the local or systemic immune system, the secretion by IEC of antigen-presenting vesicles called exosomes and bearing MHC–peptide complexes has recently been proposed. Many other factors such as nutritional or environmental factors can also influence the properties of the epithelial barrier and the outcome of the immune response to lumen antigens.
Collapse
|
17
|
Brandtzaeg P. The changing immunological paradigm in coeliac disease. Immunol Lett 2006; 105:127-39. [PMID: 16647763 DOI: 10.1016/j.imlet.2006.03.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Revised: 03/13/2006] [Accepted: 03/15/2006] [Indexed: 02/06/2023]
Abstract
When coeliac disease is referred to as an inflammatory disorder, this may detract from its true nature. Activation of innate and adaptive immunity takes place in the mucosal lesion, but the tissue reaction is not that of classical inflammation. In fact, coeliac disease contrasts strikingly with typical inflammatory bowel disorders such as ulcerative colitis and Crohn's disease. The coeliac lesion apparently reflects, in the main, immune-driven remodelling of mucosal architecture with only a minor inflammatory component - initially most likely resulting from innate signals. Complement split products might be one of several potential initial hits that lead to activation of lamina propria and epithelial cells with release of mediators such as interleukin-15. This cytokine appears to stimulate potentially pathogenic intraepithelial lymphocytes. In genetically susceptible individuals, such early innate events could turn into persistent pathogenic signalling with subsequent adaptive cellular and humoral immunopathology resulting in a chronic lesion. Nevertheless, mucosal homeostasis is surprisingly well preserved as signified by the remarkable dominance of plasma cells that produce dimeric immunoglobulin A as a basis for enhanced secretory immunity. This shows that the microvascular endothelium in the lesion largely maintains its 'gatekeeper' function for mucosal immune cells - in striking contrast to the 'promiscuous' situation in inflammatory bowel disease. Altogether, a two-signal model is emerging for the pathogenesis of coeliac disease - signal 1 generated by innate immunity and signal 2 by adaptive immunity. Hence, there is currently an increased focus on immune activation in the epithelial compartment rather than on changes in the microvasculature as a basis for classical inflammation.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Institute and Department of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Centre, N-0027 Oslo, Norway.
| |
Collapse
|
18
|
Stoicov C, Cai X, Li H, Klucevsek K, Carlson J, Saffari R, Houghton J. Major histocompatibility complex class II inhibits fas antigen-mediated gastric mucosal cell apoptosis through actin-dependent inhibition of receptor aggregation. Infect Immun 2005; 73:6311-21. [PMID: 16177302 PMCID: PMC1230908 DOI: 10.1128/iai.73.10.6311-6321.2005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Escape from normal apoptotic controls is thought to be essential for the development of cancer. During Helicobacter pylori infection, the leading cause of gastric cancer, activation of the Fas antigen (Fas Ag) apoptotic pathway is responsible for early atrophy and tissue loss. As disease progresses, metaplastic and dysplastic glands arise which express Fas Ag but are resistant to apoptosis and are believed to be the precursor cells for adenocarcinoma. In this report, we show that one mechanism of acquired Fas resistance is inhibition of receptor aggregation via a major histocompatibility complex class II (MHCII)-mediated, actin-dependent mechanism. For these studies we used the well-described C57BL/6 mouse model of Helicobacter pylori and Helicobacter felis infection. Under normal conditions, Fas Ag is expressed at low levels, and MHCII expression on gastric mucosal cells is negligible. With infection and inflammation, both receptors are upregulated, and 6.1% of gastric mucosal cells express MHCII in combination with Fas Ag. Using the rat gastric mucosal cell line RGM-1 transfected with murine Fas Ag and MHCIIalphabeta chains, we demonstrate that MHCII prevents Fas receptor aggregation and inhibits Fas-mediated signaling through its effects on the actin cytoskeleton. Depolymerization of actin with cytochalasin D allows receptors to aggregate and restores Fas sensitivity. These findings offer one mechanism by which gastric mucosal cells acquire Fas resistance.
Collapse
Affiliation(s)
- Calin Stoicov
- Department of Medicine, Division of Gastroenterology, University of Massachusetts Medical School, Worcester, 01615, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Johansen FE, Brandtzaeg P. Transcriptional regulation of the mucosal IgA system. Trends Immunol 2004; 25:150-7. [PMID: 15036043 DOI: 10.1016/j.it.2004.01.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Finn-Eirik Johansen
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Institute and Department of Pathology, Rikshospitalet University Hospital, N-0027 Oslo, Norway.
| | | |
Collapse
|
21
|
Gurevich P, Elhayany A, Ben-Hur H, Moldavsky M, Szvalb S, Zandbank J, Shperling I, Zusman I. An immunohistochemical study of the secretory immune system in human fetal membranes and decidua of the first trimester of pregnancy. Am J Reprod Immunol 2004; 50:13-9. [PMID: 14506924 DOI: 10.1034/j.1600-0897.2003.01201.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PROBLEM We analyzed the presence and distribution of components of the secretory immune system (SIS) in human fetal membranes (amnion, yolk sac, chorion) and decidua from the first trimester of pregnancy. MATERIALS AND METHODS Specimens from 17 embryos (4-8 weeks of pregnancy) and nine fetuses (9-12 weeks) were divided into those that had not been exposed to massive foreign antigenic effects (Group I, n = 18) and those that had suffered acute chorioamnionitis (Group II, n = 8). RESULTS Positive immunostaining for secretory component (SC), joining (J) chain, IgG, IgA, and macrophages was seen from 4 to 5 weeks of development and then during the whole first trimester of pregnancy in the syncytio- and cytotrophoblasts, the amniotic epithelium, the yolk sac endoderm and decidual cells. Macrophages with J chain, IgG and IgA were found in embryonic tissues on week 4, whereas lymphocytes, including those synthesizing IgA and IgM, appeared only at the end of the first trimester of pregnancy. In the decidua, lymphocytes and macrophages were recognized during the whole period of study. In cases with chorioamnionitis (Group II), reactivity of IgG and IgA in the mentioned cells of fetuses decreased sharply while the rate of immunoreactivity of SC and J chain as well as the number of T and B lymphocytes did not change. In the decidua, the number of immune reactive cells sharply increased with the appearance of plasma cells. CONCLUSIONS In the fetal membranes and decidua all the SIS components are present. We suggest that two SIS, maternal and fetal, participate in the formation of the barrier between a mother and the fetus. Both these systems have different origin and cellular content as well as different immune reactions.
Collapse
|
22
|
Fernandez MI, Pedron T, Tournebize R, Olivo-Marin JC, Sansonetti PJ, Phalipon A. Anti-inflammatory role for intracellular dimeric immunoglobulin a by neutralization of lipopolysaccharide in epithelial cells. Immunity 2003; 18:739-49. [PMID: 12818156 DOI: 10.1016/s1074-7613(03)00122-5] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal epithelial cells (IEC) play a central role in innate and acquired mucosal immunity. They ensure early signaling to trigger an inflammatory response against pathogens. Moreover, IEC mediate transcytosis of dimeric IgA (dIgA), through the polymeric-immunoglobulin receptor (pIgR), to provide secretory IgA, the major protective Ig in mucosal secretions. Using an in vitro model of polarized IEC, we describe an additional anti-inflammatory mechanism of dIgA-mediated protection against intracellular bacterial components involved in the proinflammatory activation of IEC. Specific dIgA colocalizes to lipopolysaccharide (LPS) in the apical recycling endosome compartment, preventing LPS-induced NF-kappaB translocation and subsequent proinflammatory response. Thus, intracellular neutralization by dIgA limits the acute local inflammation induced by proinflammatory pathogen-associated molecular patterns such as LPS.
Collapse
Affiliation(s)
- M Isabel Fernandez
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 rue du Dr. Roux, 75015, Paris, France.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Adaptive immunity mediated by secretory antibodies is important in the defence against mucosal infections. Specific secretory immunoglobulin A (SIgA) can inhibit initial pathogen colonization by performing immune exclusion both on the mucosal surface and within virus-infected secretory epithelial cells without causing tissue damage. Resistance against toxin-producing bacteria such as Vibrio cholerae appears to be particularly dependent on SIgA antibodies. Like natural infections, live topical vaccines or adequate combinations of inactivated vaccines and mucosal adjuvants give rise not only to SIgA antibodies, but also to long-standing serum IgG and IgA responses. The intranasal route of vaccine application could be particularly attractive to achieve this result, but only if successful stimulation is obtained without the use of toxic adjuvants. The degree of protection after vaccination may range from complete inhibition of reinfection to reduction of symptoms. In this scenario it is generally difficult to determine unequivocally the relative importance of SIgA versus serum antibodies. However, infection models in knockout mice strongly support the notion that SIgA exerts a decisive role in protection and cross-protection against a variety of infectious agents.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology LIIPAT, Institute of Pathology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway.
| |
Collapse
|
24
|
Abstract
In the last few decades, the comprehension of epidemiological, pathogenic and clinical aspects of coeliac disease has increasingly improved. Serological screening studies on the general population have shown that the true coeliac disease prevalence in Europe is higher than previously reported. It has become clear that tissue transglutaminase has a crucial role in the pathogenesis of coeliac disease pathogenesis, and there is evidence that substitution of deamidated amino acidic residues at a critical position along the gliadin sequence dramatically increases immunological activation. The toxicity of many gluten epitopes has been investigated, so far, but recent studies have indicated the region 57-75 of alpha gliadin as a possible candidate epitope in the pathogenesis of coeliac disease. However, the wide heterogeneity of gliadin and glutenin molecules complicate any attempts to identify the toxic epitope, and the fascinating idea to produce detoxified grains will represent a great challenge in the near future. From a clinical point of view, there is now evidence of a broad spectrum of gluten conditions. Extra-intestinal signs, i.e., alopecia, unexplained neurological disorders, cryptic hypertransaminasaemia, increased red cell width, frequently constitute the only clinical manifestation at the diagnosis.
Collapse
Affiliation(s)
- S Martucci
- Gastroenterology Unit, IRCCS Policlinico San Matteo, University of Pavia, Italy
| | | | | | | |
Collapse
|
25
|
Gurevich P, Ben-Hur H, Szvalb S, Moldavsky M, Zusman I. Lymphoid-epithelial secretory immune system in human fetuses in the second trimester of gestation. Pediatr Dev Pathol 2002; 5:22-8. [PMID: 11815865 DOI: 10.1007/s10024-001-0088-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2000] [Accepted: 05/19/2001] [Indexed: 10/26/2022]
Abstract
The development of the secretory immune system (SIS) in the respiratory, digestive, and urogenital tracts and other organs of fetuses in the second trimester of gestation is described. Tissues of all internal organs of human fetuses (n = 36) that had died between 13 and 25 weeks of gestation were studied immunohistochemically for the presence of secretory component (SC), J chain, IgA, IgM, IgG, macrophages, and different subsets of lymphocytes. We found protein elements of the SIS in fetuses during the entire second trimester in the epithelium of the digestive, respiratory, and urinary tracts; in hepatocytes; in the epithelium of the bile duct, renal tubules, and all the urinary tract; in the salivary glands, pancreas, and thyroid; in the epithelium of the Fallopian tubes and uterus; in the epididymis and the rete testes; in the skin; and in other organs. Immunocompetent cells, including IgA- and IgM-secreting cells, were located in these organs under the epithelium and sometimes between epithelial cells. In fetuses with acute infection, the number of immunocompetent cells was higher, reflecting a whole-immune system reaction, including the SIS. We conclude that the fetal SIS is a ramified, defensive immune system that is distributed throughout most organs of epithelial origin in second-trimester fetuses, and that it reacts against intrafetal infiltration by foreign antigens.
Collapse
Affiliation(s)
- Pavel Gurevich
- Department of Obstetrics and Gynecology, Kaplan Medical Center, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
26
|
Schjerven H, Brandtzaeg P, Johansen FE. A novel NF-kappa B/Rel site in intron 1 cooperates with proximal promoter elements to mediate TNF-alpha-induced transcription of the human polymeric Ig receptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6412-20. [PMID: 11714807 DOI: 10.4049/jimmunol.167.11.6412] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Secretory Abs constitute the first line of specific immune defense at mucosal surfaces. Such Abs are generated by the active transport of polymeric Ig (pIg) across secretory epithelia mediated by the pIgR, also known as transmembrane secretory component (SC). The proinflammatory cytokine TNF-alpha is a key mediator of host responses to infections, and it can stimulate protein synthesis-dependent transcriptional up-regulation of pIgR/SC in the HT-29 intestinal adenocarcinoma cell line. By reporter gene assay we identified a novel TNF-alpha-responsive region located within a 748-bp fragment in intron 1 of the human pIgR/SC gene which depended on an NF-kappaB/Rel site for full responsiveness. EMSAs demonstrated preferential binding of the NF-kappaB/Rel family member p65 (RelA) to this DNA element after TNF-alpha stimulation, with weaker and more delayed binding of p50. Furthermore, the TNF-alpha-responsive region in intron 1 required cooperation with DNA elements located in the proximal promoter region of the gene. Mutational analysis demonstrated that an IFN-stimulated response element near the transcriptional start site in exon 1 was involved in the TNF-alpha responsiveness. Thus, DNA elements located >4 kb apart were found to cooperate in TNF-alpha-induced pIgR/SC up-regulation. The intronic TNF-alpha-responsive enhancer overlapped with a recently identified IL-4-responsive enhancer. Several intronic DNA elements found to be functionally important in the human gene are highly conserved between the human and mouse pIgR/SC genes, suggesting the presence of a conserved cytokine-responsive enhancer region.
Collapse
Affiliation(s)
- H Schjerven
- Laboratory for Immunohistochemistry and Immunopathology, Institute of Pathology, University of Oslo, Rikshospitalet, Oslo, Norway.
| | | | | |
Collapse
|
27
|
van Niel G, Raposo G, Candalh C, Boussac M, Hershberg R, Cerf-Bensussan N, Heyman M. Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterology 2001; 121:337-49. [PMID: 11487543 DOI: 10.1053/gast.2001.26263] [Citation(s) in RCA: 541] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Given the observations that intestinal epithelial cells (IECs) can present antigens to CD4(+) T lymphocytes and that professional antigen-presenting cells secrete exosomes (antigen-presenting vesicles), we hypothesized that IECs may secrete exosomes carrying molecules implicated in antigen presentation, which may be able to cross the basement membrane and convey immune information to noncontiguous immune cells. METHODS Human IEC lines HT29-19A and T84-DRB1*0401/CIITA were grown on microporous filters. Release of exosomes under basal or inflammatory conditions was evaluated in conditioned apical and basolateral media after differential ultracentrifugations. Morphologic and biochemical characterization of exosomes was performed using immunoelectron microscopy, Western blotting, and matrix-assisted laser desorption ionization-time of flight mass spectrometry. RESULTS The intestinal cell lines released 30-90-nm-diameter vesicles from the apical and basolateral sides, and this release was significantly increased in the presence of interferon gamma. MHC class I, MHC class II, CD63, CD26/dipeptidyl-peptidase IV, and A33 antigen were present in epithelial-derived exosomes. CONCLUSIONS; Human IEC lines secrete exosomes bearing accessory molecules that may be involved in antigen presentation. These data are consistent with a model in which IECs may influence antigen presentation in the mucosal or systemic immune system independent of direct cellular contact with effector cells.
Collapse
Affiliation(s)
- G van Niel
- INSERM E9925, Faculté Necker-Enfants Malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France.
| | | | | | | | | | | | | |
Collapse
|
28
|
Kaneko M, Kawakita T, Yamaoka Y, Nomoto K. Development of the susceptibility to oral tolerance induction in infant mice administered a herbal drug, Hochu-ekki-to (Bu-Zhong-Yi-Qi-Tang). Int Immunopharmacol 2001; 1:219-27. [PMID: 11360923 DOI: 10.1016/s1567-5769(00)00022-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The susceptibility to oral tolerance in post-neonatal infant mice and the effect of a herbal drug, Hochu-ekki-to (HOT), on the susceptibility were investigated. To induce oral tolerance induction, infant and adult mice at 4 and 8 weeks of age, respectively, were orally administered a single high dose of OVA before an intraperitoneal immunization with OVA adsorbed on aluminum hydroxide. HOT (1000 mg/kg) was administered orally for 7 days before the induction. HOT significantly decreased the serum levels of OVA-specific IgE and IgG1 and the antigen-specific proliferation of spleen cells in infant mice, both of which were greatly enhanced compared to in adult mice. HOT increased the number of both CD4+ T cells and antigen-presenting cells expressing MHC class II as well as costimulatory molecules (CD40, CD80 and/or CD86) in the Peyer's patch (PP) of infant mice, which had fewer cells than adult mice. In the PP, moreover, HOT augmented the IL-12p40 mRNA expression and spontaneous or CD40-stimulated IL-12 production, and increased the number of CD4+ cells expressing CD40 ligand, which is up regulated by IL-12. These results suggest that HOT increases the number and improves the function of PP cells that are fully susceptible to the induction of oral tolerance.
Collapse
Affiliation(s)
- M Kaneko
- Kampo (Traditional Japanese Medicine) and Healthcare Research Laboratories, Kanebo Co. Ltd., Osaka, Japan
| | | | | | | |
Collapse
|
29
|
Abstract
Celiac disease is a genetically-based intolerance to gluten. In the past, celiac disease has been considered a rare disease of infancy characterized by chronic diarrhea and delayed growth. Besides the overt enteropathy, there are many other forms which appear later in life; target organs are not limited to the gut, but include liver, thyroid, skin and reproductive tract. It is now recognized that celiac disease is a relatively frequent disorder; the overall prevalence is at least 1:300 in Western Europe. Celiac disease may impair the reproductive life of affected women, eliciting delayed puberty, infertility, amenorrhea and precocious menopause. Clinical and epidemiological studies show that female patients with celiac disease are at higher risk of spontaneous abortions, low birth weight of the newborn and reduced duration of lactation. No adequate studies are available on the rate of birth defects in the progeny of affected women; however, celiac disease induces malabsorption and deficiency of factors essential for organogenesis, e.g. iron, folic acid and vitamin K. The overall evidence suggests that celiac disease patients can be a group particularly susceptible to reproductive toxicants; however, the pathogenesis of celiac disease-related reproductive disorders still awaits clarification. At present, like the other pathologies associated with celiac disease, the possible prevention or treatment of reproductive effects can only be achieved through a life-long maintenance of a gluten-free diet.
Collapse
Affiliation(s)
- A V Stazi
- Laboratory of Comparative Toxicology and Ecotoxicology, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
30
|
Schjerven H, Brandtzaeg P, Johansen FE. Mechanism of IL-4-mediated up-regulation of the polymeric Ig receptor: role of STAT6 in cell type-specific delayed transcriptional response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:3898-906. [PMID: 11034397 DOI: 10.4049/jimmunol.165.7.3898] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The polymeric IgR (pIgR) mediates transport of dimeric IgA and pentameric IgM across mucosal epithelia, thereby generating secretory Abs. Its expression is up-regulated at the transcriptional level by IL-4 in HT-29 cells. In this study, we demonstrate that IL-4 mediates up-regulation of human pIgR through a 554-bp IL-4-responsive enhancer in intron 1. Mutation of a binding site for STAT-6 within this region abolished IL-4-induced enhancement, while an adjacent putative C/EBP site was dispensable. IL-4 treatment induced binding of STAT6 to the intronic STAT6 site, but cooperation with nearby upstream and downstream DNA elements was required for IL-4 responsiveness. Furthermore, IL-4-mediated increased transcription of the pIgR-derived enhancer, like the endogenous pIgR gene, required de novo protein synthesis. Interestingly, a conditionally active form of STAT6 sufficed to activate a pIgR-derived enhancer in HT-29 cells, but not in Cos-1 cells, suggesting a requirement for cell type-specific factors. Thus, STAT6 activation mediates a delayed transcriptional enhancement of pIgR by induction of a de novo synthesized protein that cooperates with STAT6 itself bound to its cognate DNA element in intron 1. This mechanism may represent a general strategy for how pleiotropic cytokines elicit cell type-specific transcriptional responses.
Collapse
Affiliation(s)
- H Schjerven
- Laboratory for Immunohistochemistry and Immunopathology, Institute of Pathology, University of Oslo, Rikshospitalet, Norway
| | | | | |
Collapse
|
31
|
North RA, Taylor R, Li Zhou R, Schellenberg JC. The relationship of smoking, preeclampsia, and secretory component. Am J Obstet Gynecol 2000; 183:136-9. [PMID: 10920321 DOI: 10.1067/mob.2000.105745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We sought to determine whether total secretory component in serum is increased in women in whom preeclampsia subsequently develops. STUDY DESIGN Serum samples were collected serially throughout pregnancy and post partum from nulliparous women (N = 1496). Serum concentrations of total secretory component were measured by an enzyme-linked immunosorbent assay in all women in whom preeclampsia developed (n = 71) and a randomly selected group of normotensive women (n = 83). RESULTS Secretory component increased with smoking (P =.0003) and with gestation (P =.0001). In the whole group secretory component was not different in women with preeclampsia (P =.10), but there was a significant interaction of smoking, gravidity, and preeclampsia (P =.04). Among the women who smoked, secretory component was lower in women in whom preeclampsia subsequently developed compared with those who remained normotensive (P =.02). This difference was significant from 15 to 19 weeks' gestation. CONCLUSION Very high serum concentrations of secretory component in smokers may protect against the development of preeclampsia and may indicate the involvement of mucosal tolerance.
Collapse
Affiliation(s)
- R A North
- Department of Obstetrics and Gynaecology, National Women's Hospital, School of Medicine, University of Auckland, New Zealand
| | | | | | | |
Collapse
|
32
|
Corthésy B, Spertini F. Secretory immunoglobulin A: from mucosal protection to vaccine development. Biol Chem 1999; 380:1251-62. [PMID: 10614817 DOI: 10.1515/bc.1999.160] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Immune responses taking place in mucosal tissues are typified by secretory immunoglobulin A (S-IgA) molecules, which are assembled from proteins expressed in two cell lineages. The heavy and light chains as well as the J chain are produced in plasma cells, whereas the secretory component (SC) is associated to the immunoglobulin complex during transcytosis across the epithelial layer. S-IgA antibodies represent the predominant immunoglobulin class in external secretions, and the best defined entity providing specific immune protection for mucosal surfaces by blocking attachment of bacteria and viruses. S-IgA constitutes greater than 80% of all antibodies produced in mucosa-associated lymphoid tissues in humans. The existence of a common mucosal immune system permits immunization on one mucosal surface to induce secretion of antigen-specific S-IgA at distant sites. In addition, S-IgA antibodies not only function in external secretions, but also exert their antimicrobial properties within the epithelial cell during transport across the epithelium. Passive mucosal delivery of monoclonal IgA molecules neutralizes pathogens responsible for gastrointestinal and respiratory infections. Mucosal and systemic immunity can be achieved by orally administered recombinant S-IgA molecules carrying a protective bacterial epitope within the SC polypeptide primary sequence.
Collapse
Affiliation(s)
- B Corthésy
- Division d'Immunologie et d'allergie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | |
Collapse
|
33
|
Brandtzaeg P, Farstad IN, Johansen FE, Morton HC, Norderhaug IN, Yamanaka T. The B-cell system of human mucosae and exocrine glands. Immunol Rev 1999; 171:45-87. [PMID: 10582165 PMCID: PMC7159139 DOI: 10.1111/j.1600-065x.1999.tb01342.x] [Citation(s) in RCA: 200] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mucosae and exocrine glands harbour the largest activated B-cell system of the body, amounting to some 80-90% of all immunoglobulin (Ig)-producing cells. The major product of these immunocytes is polymeric (p)IgA (mainly dimers) with associated J chain. Both pIgA and pentameric IgM contain a binding site for the polymeric Ig receptor (pIgR), or secretory component (SC), which is a requirement for their active external transport through secretory epithelia. The pIgR/SC binding site depends on covalent incorporation of the J chain into the quaternary structure of the polymers when they are produced by the local immunocytes. This important differentiation characteristic appears to be sufficient functional justification for the J chain to be expressed also by most B cells terminating at secretory effector sites with IgD or IgG production; they probably represent a "spin-off" from sequential downstream CH switching on its way to pIgA expression, thus apparently reflecting a maturational stage of effector B-cell clones compatible with homing to these sites. Observations in IgA-deficient individuals suggest that the magnitude of this homing is fairly well maintained even when the differentiation pathway to IgA is blocked. Certain microenvironmental elements such as specific cytokines and dendritic cells appear to be required for induction of IgA synthesis, but it remains virtually unknown why this isotype normally is such a dominating product of local immunocytes and why they have such a high level of J chain expression. Also, despite the recent identification of some important requirements in terms of adhesion molecules (e.g. integrin alpha 4 beta 7 and MAdCAM-1) that explain the "gut-seeking" properties of enterically induced B cells, the origin of regionalized homing of B cells to secretory effector sites outside the gut remains elusive. Moreover, little is known about immune regulation underlying the striking disparity of both the class (IgD, IgM) and subclass (IgA1, IgA2, IgG1, IgG2) production patterns shown by local immunocytes in various regions of the body, although the topical microbiota and other environmental stimuli might be important. Rational design of local vaccines will depend on better knowledge of both inductive and migratory properties of human mucosal B cells.
Collapse
Affiliation(s)
- P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), University of Oslo, National Hospital, Norway.
| | | | | | | | | | | |
Collapse
|
34
|
Ahlstedt I, Lindholm C, Lönroth H, Hamlet A, Svennerholm AM, Quiding-Järbrink M. Role of local cytokines in increased gastric expression of the secretory component in Helicobacter pylori infection. Infect Immun 1999; 67:4921-5. [PMID: 10456951 PMCID: PMC96829 DOI: 10.1128/iai.67.9.4921-4925.1999] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Using immunohistochemical staining, we examined the presence of secretory component (SC) on epithelial cells in gastric and duodenal biopsy specimens collected from Helicobacter pylori-infected individuals and healthy controls. Gastric epithelial cells from healthy volunteers expressed low, but detectable, levels of SC. In contrast, significantly higher level of expression of SC (P < 0.001) was observed on epithelial cells in the antra of H. pylori-infected individuals. The antral SC expression correlated with staining for gamma interferon of intraepithelial and lamina propria lymphocytes (r(s) = 0.76 and 0.69, respectively, P < 0.001) and correlated weakly with production of tumor necrosis factor alpha (r(s) = 0.43, P < 0.05), but it did not correlate at all with interleukin-4 production.
Collapse
Affiliation(s)
- I Ahlstedt
- Departments of Medical Microbiology and Immunology, Göteborg University, Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
35
|
Bourreille A, Segain JP, Raingeard de la Blétière D, Siavoshian S, Vallette G, Galmiche JP, Blottière HM. Lack of interleukin 10 regulation of antigen presentation-associated molecules expressed on colonic epithelial cells. Eur J Clin Invest 1999; 29:48-55. [PMID: 10092989 DOI: 10.1046/j.1365-2362.1999.00410.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Colonic epithelial cells may behave as antigen-presenting cells. Interleukin 10 (IL-10) is known to play a major role in the intestinal immune system; however, it remains to be determined whether human intestinal epithelial cells express IL-10 receptor, and whether this cytokine modulates their expression of antigen presentation-associated molecules. METHODS The binding of biotinylated IL-10 was studied in SW 1116, HT-29 and T84 human colonic epithelial cell lines and freshly isolated normal colonic epithelial cells. Reverse transcription-polymerase chain reaction was also performed to detect IL-10 receptor mRNA. The effect of IL-10 on antigen presentation associated molecules was assessed by flow cytometry. RESULTS Biotinylated IL-10 bound to SW 1116, HT-29, T84, and normal colonic epithelial cells. IL-10 receptor mRNA was detected in SW 1116 and normal epithelial cells. SW 1116 and HT-29 cells expressed MHC class I and ICAM-1, but not CD80, and SW 1116 constitutively expressed HLA-DR. Interferon-gamma up-regulated HLA-DR and ICAM-1 expression on both cells, whereas lipopolysaccharide increased ICAM-1 expression only on SW 1116. IL-10 failed to modulate these antigens, even after stimulation by lipopolysaccharide or interferon-gamma. Moreover, these molecules decreased IL-10 binding in both lines. CONCLUSION The presence of IL-10 receptor on intestinal epithelial cells suggest that IL-10 may play a role in mucosal physiology, however its effect on the immune response remains to be determined.
Collapse
Affiliation(s)
- A Bourreille
- Centre de Recherche en Nutrition Humaine, Nantes, France
| | | | | | | | | | | | | |
Collapse
|
36
|
Nilsen EM, Johansen FE, Kvale D, Krajci P, Brandtzaeg P. Different regulatory pathways employed in cytokine-enhanced expression of secretory component and epithelial HLA class I genes. Eur J Immunol 1999; 29:168-79. [PMID: 9933098 DOI: 10.1002/(sici)1521-4141(199901)29:01<168::aid-immu168>3.0.co;2-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The transmembrane secretory component (SC, or pIg receptor) plays a crucial role in mucosal immunity by translocating dimeric IgA and pentameric IgM through exocrine epithelia. This receptor is up-regulated by cytokines in parallel with increased epithelial HLA expression. By use of the human epithelial cell line HT-29m3, we show that IFN-gamma, TNF-alpha and IL-4 activate transcription of the SC gene. This activation was slow, suggesting mediation via newly synthesized protein factors. IFN-gamma and TNF-alpha, but not IL-4, also up-regulated expression of HLA class I genes. However, this gene induction was rapid and did not depend on new protein synthesis. Nuclear run-on experiments showed that the transcription rate of HLA class I genes nearly peaked after only 30 min of IFN-gamma or TNF-alpha stimulation, whereas the SC transcription rate did not peak until after 20-36 h of IFN-gamma, TNF-alpha or IL-4 stimulation. Gel electrophoresis mobility shift assays demonstrated binding of nuclear proteins from cytokine-stimulated HT-29 cells to consensus elements in the promoter of the SC gene, involving the binding site for the nuclear factor-kappaB p50 subunit after TNF-alpha stimulation, and IFN-stimulated response element after IFN-gamma stimulation (and weakly after TNF-alpha. Our observations in vitro likely parallel events in vivo by which activated mucosal T cells and macrophages enhance pIg receptor-mediated external transport of secretory IgA and IgM and up-regulate epithelial HLA expression.
Collapse
Affiliation(s)
- E M Nilsen
- Laboratory for Immunohistochemistry and Immunopathology, Institute of Pathology, University of Oslo, The National Hospital, Norway.
| | | | | | | | | |
Collapse
|
37
|
Martín MG, Wang J, Li TW, Lam JT, Gutierrez EM, Solorzano-Vargas RS, Tsai AH. Characterization of the 5'-flanking region of the murine polymeric IgA receptor gene. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:G778-88. [PMID: 9756509 DOI: 10.1152/ajpgi.1998.275.4.g778] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The regulatory elements that control basal and activated transcriptional expression of the polymeric IgA receptor gene (pIgR) have not been defined. In this study, we performed functional analysis of the murine pIgR 5'-upstream region. Transient transfection studies identified the gene's minimal promoter to reside within 110 nucleotides upstream from the start of transcription. Substitution mutations of this region identified both a putative activator (-78 to -70) and a repressor (-66 to -52) element. DNase I footprint analysis confirmed an area of protection that spans from nucleotides -85 to -62. Mobility shift assays of the putative region confirmed binding of upstream stimulatory factor 1 (USF1) to an E box element at positions -75 and -70, representing the putative enhancer. Overexpression studies using various forms of USF suggest that both USF1 and USF2 enhance activity of the pIgR minimal promoter. We report the identification and characterization of the murine pIgR minimal promoter, as well as the critical role of USF in enhancing its basal level of transcription in Caco-2 cells.
Collapse
MESH Headings
- Aging
- Animals
- Base Sequence
- Binding Sites
- Cell Line
- DNA-Binding Proteins/metabolism
- Enhancer Elements, Genetic
- Exons
- Genomic Library
- Humans
- Immunoglobulin A/metabolism
- Intestine, Small/growth & development
- Intestine, Small/metabolism
- Mice/genetics
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Oligodeoxyribonucleotides
- Polymerase Chain Reaction
- Promoter Regions, Genetic
- Rats/genetics
- Receptors, Fc/biosynthesis
- Receptors, Fc/genetics
- Recombinant Proteins/biosynthesis
- Regulatory Sequences, Nucleic Acid
- Sequence Alignment
- Sequence Homology, Amino Acid
- TATA Box
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- M G Martín
- Division of Gastroenterology and Nutrition, Department of Pediatrics, University of California School of Medicine, Los Angeles, California 90095-1752, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Johansen FE, Bosløven BA, Krajci P, Brandtzaeg P. A composite DNA element in the promoter of the polymeric immunoglobulin receptor regulates its constitutive expression. Eur J Immunol 1998; 28:1161-71. [PMID: 9565355 DOI: 10.1002/(sici)1521-4141(199804)28:04<1161::aid-immu1161>3.0.co;2-s] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The polymeric immunoglobulin receptor (pIgR), which is constitutively expressed on the basolateral surface of secretory epithelial cells, mediates external translocation of polymeric IgA and pentameric IgM (collectively called pIg) to exocrine secretions. A high level of synthesis must be maintained because the receptor is continuously cleaved to release bound secretory component (SC) in secretory IgA and secretory IgM, as well as free SC from unoccupied receptor. We have isolated the promoter of the pIgR gene and identified a short activating region that is required for the expression of pIgR promoter-driven reporter genes. This region contained an E-box and an inverted repeat sequence (IRS). Gel electrophoresis mobility shift assays with nuclear extracts from different pIgR-expressing epithelial cell lines demonstrated proteins that bind independently to both the E-box and the IRS sequence of the pIgR promoter. In addition, a DNA probe that contained both the E-box and the IRS gave rise to a larger complex that could not be competed by either element on its own. Binding was confirmed by DNase I footprinting of the E-box and IRS sequences with nuclear extracts, and by dimethyl sulfide footprinting in living HT-29 epithelial cells. Finally, a mutation in the pIgR promoter that inhibited protein binding to the E-box and the formation of the larger complex, abolished activated transcription from the reporter gene.
Collapse
Affiliation(s)
- F E Johansen
- Laboratory of Immunohistochemistry and Immunopathology, Institute of Pathology, University of Oslo, The National Hospital, Rikshospitalet, Norway.
| | | | | | | |
Collapse
|
39
|
Jahnsen FL, Farstad IN, Aanesen JP, Brandtzaeg P. Phenotypic distribution of T cells in human nasal mucosa differs from that in the gut. Am J Respir Cell Mol Biol 1998; 18:392-401. [PMID: 9490657 DOI: 10.1165/ajrcmb.18.3.2995] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Phenotypic and functional studies are required to understand the immunoregulatory role of mucosal T cells. Information about T cells in the human upper respiratory tract is limited and conflicting. Therefore, we phenotyped T cells in nasal mucosa by means of multicolor in situ immunofluorescence. In normal mucosa, most CD3+ intraepithelial lymphocytes (IELs) and lamina propria lymphocytes (LPLs) (> 90%) expressed T-cell receptor (TCR)alpha/beta, and only approximately 5% expressed TCRgamma/delta. Although most IELs in the surface epithelium were CD8+ (64%), many expressed CD4 (30%) and the CD4 phenotype dominated (55%) only slightly in the lamina propria. This result was strikingly different from that obtained for comparable compartments in histologically normal jejunal mucosa, where IELs consisted of 83% CD8+ and LPLs of 73% CD4(+) T cells. Nasal CD3+ IELs and LPLs were mainly CD45RO+CD45RA- and usually expressed CD7. The integrin alphaEbeta7 was, as expected, more common on IELs than on LPLs (78 versus 20%). In conclusion, nasal T cells show several similarities to those of the normal jejunum but some notable differences exist, especially a relative increase in CD4+ T cells in the epithelium and a decrease in the lamina propria. It should be explored whether this disparity, together with an increased expression of epithelial adhesion molecules, might contribute to local immunological overstimulation and partly explain the relatively high frequency of airway allergy.
Collapse
Affiliation(s)
- F L Jahnsen
- Laboratory for Immunohistochemistry and Immunopathology, Institute of Pathology, Rikshospitalet, Oslo, Norway.
| | | | | | | |
Collapse
|
40
|
Welsh FK, Ramsden CW, MacLennan K, Sheridan MB, Barclay GR, Guillou PJ, Reynolds JV. Increased intestinal permeability and altered mucosal immunity in cholestatic jaundice. Ann Surg 1998; 227:205-12. [PMID: 9488518 PMCID: PMC1191237 DOI: 10.1097/00000658-199802000-00009] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To examine the effects of cholestatic jaundice on gut barrier function. SUMMARY BACKGROUND DATA Gut barrier failure occurs in animal models of jaundice. In humans, the presence of endotoxemia indirectly implicates failure of this host defense, but this has not previously been investigated in jaundiced patients. METHODS Twenty-seven patients with extrahepatic obstructive jaundice and 27 nonicteric subjects were studied. Intestinal permeability was measured using the lactulose-mannitol test. Small intestinal morphology and the presence of mucosal immunologic activation were examined in endoscopic biopsies of the second part of the duodenum. Systemic antiendotoxin core IgG antibodies and serum interleukin-6 and C-reactive protein were also quantified. Intestinal permeability was remeasured in 9 patients 5 weeks after internal biliary drainage. RESULTS The median lactulose-mannitol ratio was significantly increased in the jaundiced patients. This was accompanied by upregulation of HLA-DR expression on enterocytes and gut-associated lymphoid tissue, suggesting immune activation. A significant increase in the acute phase response and circulating antiendotoxin core antibodies was also observed in the jaundiced patients. After internal biliary drainage, intestinal permeability returned toward normal levels. CONCLUSIONS A reversible impairment in gut barrier function occurs in patients with cholestatic jaundice. Increased intestinal permeability is associated with local immune cell and enterocyte activation. In view of the role of gut defenses in the modern paradigm of sepsis, these data may directly identify an important underlying mechanism contributing to the high risk of sepsis in jaundiced patients.
Collapse
Affiliation(s)
- F K Welsh
- Department of Pathology, St. James's University Hospital, Leeds, England
| | | | | | | | | | | | | |
Collapse
|
41
|
Ernst PB, Crowe SE, Reyes VE. How does Helicobacter pylori cause mucosal damage? The inflammatory response. Gastroenterology 1997; 113:S35-42; discussion S50. [PMID: 9394758 DOI: 10.1016/s0016-5085(97)80009-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The role for Helicobacter pylori in the pathogenesis of disease provides the conundrum that only a subset of subjects infected with H. pylori will ever develop peptic ulcer or gastric cancer. Thus, variation in strain as well as environmental or host factors converge in the gastroduodenal milieu and control the final outcome of infection. The host immune and inflammatory response is emerging as an important element in the pathogenesis of these gastric diseases. The ideal host response provides protection to clear an infection without causing excessive amounts of inflammation that could compromise the integrity and function of host cells. This review will cover four main questions: (1) What are the mucosal immune/inflammatory responses that confer protection without damaging the host? (2) How do the gastric immune responses during infection with H. pylori differ from this ideal scenario? (3) Do these responses contribute to autoimmune-mediated damage to gastric tissue? (4) Can immunomodulation through vaccination enhance protective, nondestructive responses that prevent or treat infection or, at least, attenuate inflammation?
Collapse
Affiliation(s)
- P B Ernst
- Department of Pediatrics (Child Health Research Center), University of Texas Medical Branch, Galveston, USA
| | | | | |
Collapse
|
42
|
McKay DM, Philpott DJ, Perdue MH. Review article: In vitro models in inflammatory bowel disease research--a critical review. Aliment Pharmacol Ther 1997; 11 Suppl 3:70-80. [PMID: 9467981 DOI: 10.1111/j.1365-2036.1997.tb00811.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Research efforts in inflammatory bowel disease (IBD) have been directed towards the epithelium as it has become clear that epithelial cells play a critical role in inflammatory response. Most research involving IBD employs in vitro techniques. In vitro epithelial cell studies have played and are continuing to play a major role in providing specific information relevant to IBD. Thus, such studies have provided irrefutable evidence that epithelial responses can be induced by microbes/microbial products and by immune activation. Culture experiments have provided insights into the effects of individual cytokines and other inflammatory mediators on epithelial pathophysiology, injury and repair, apoptosis, necrosis, and other processes that may be involved in IBD. Activated epithelial cells can participate in and even orchestrate immune responses, by stimulating T cells (and possibly others) and by producing cytokines that recruit specific inflammatory cells. Physiological regulation of epithelial tight junctions has been demonstrated by in vitro studies; the implication of this information for treating IBD is just beginning to be explored. It is becoming increasingly clear that epithelial processing and presentation of antigens is critical to the outcome of the immune response.
Collapse
Affiliation(s)
- D M McKay
- Intestinal Disease Research Programme, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
43
|
Abstract
Immunoglobulin (Ig)-producing cells in mucosal tissues represent quantitatively the most important humoral immune system of the body. All exocrine tissue sites contain immunocytes (B-cell blasts and plasma cells) that mainly synthesize dimers and larger polymers of IgA (collectively called pIgA) with incorporated J chain. Such pIgA is actively transported to external secretions as secretory IgA (SIgA) by the polymeric Ig receptor (pIgR), a transmembrane epithelial glycoprotein also called the secretory component (SC). The same transport mechanism includes pentameric IgM to generate SIgM. Although the most active SIgA system occurs in the gut, secretory immunity also operates in the female genital tract, with considerable pIgA production in the cervical mucosa and fallopian tubes. The origin of these local IgA immunocytes remains undefined. In mice, both lymphoid tissue in the large bowel (GALT) and nasopharynx (NALT) have been suggested as inductive sites for B cells homing to the urogenital tract. It is well established that integrin alpha 4 beta 7 is used by primed lymphoid cells to enter the intestinal lamina propria through interactions with mucosal addressin cell adhesion molecule (MAdCAM)-1 expressed on venule endothelium. However, alpha 4 beta 7 does not appear to be an important homing molecule in the airways, and the same might be true for the urogenital tract; this could explain that high levels of IgA antibodies occur in cervicovaginal secretions of mice after nasal immunization. The endometrium can likewise perform pIgR-mediated external translocation of pIgA that in this tissue appears to be mainly derived from serum, partly under hormonal regulation. In addition, paracellular diffusion of serum-derived and locally produced IgG through epithelia is an important part of humoral immunity in the female genital tract.
Collapse
Affiliation(s)
- P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), University of Oslo, National Hospital, Norway.
| |
Collapse
|
44
|
Islam D, Veress B, Bardhan PK, Lindberg AA, Christensson B. Quantitative assessment of IgG and IgA subclass producing cells in rectal mucosa during shigellosis. J Clin Pathol 1997; 50:513-20. [PMID: 9378821 PMCID: PMC499993 DOI: 10.1136/jcp.50.6.513] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS To assess quantitatively both the morphological changes in the rectal mucosa and the changes in the relative frequency of IgA and IgG subclass producing cells found in the rectal mucosa during the acute phase of shigellosis and at convalescence. METHODS Rectal biopsies from 25 Shigella dysenteriae 1 infected patients, 10 Shigella flexneri infected patients, and 40 uninfected controls were studied. Morphological changes in the mucosa were graded. The frequency of IgA and IgG subclass producing cells was assessed. In addition, immunostaining for secretory component in epithelial cells was analysed. RESULTS Using morphological grading, 20% of the 35 patients studied had advanced inflammation (grade 3) in the acute phase of the disease. At convalescence, grade 1 inflammation was seen in 37% of the patients and in 10% of the controls. In the acute phase, as well as at convalescence, the number of IgA1, IgA2, and IgG2 positive cells was significantly higher than in the controls. The results were related to the histopathological degree of inflammation. CONCLUSIONS In shigellosis, there is evidence for a prolonged humoral response residing in the mucosa long after the clinical symptoms have resolved, suggesting that shigellosis induces persisting mucosal humoral immune and inflammatory responses, remaining at least until 30 days after the infection.
Collapse
Affiliation(s)
- D Islam
- Laboratory Sciences Division, International Centre for Diarrhoeal Disease Research, Bangladesh.
| | | | | | | | | |
Collapse
|
45
|
Sütas Y, Autio S, Rantala I, Isolauri E. IFN-gamma enhances macromolecular transport across Peyer's patches in suckling rats: implications for natural immune responses to dietary antigens early in life. J Pediatr Gastroenterol Nutr 1997; 24:162-9. [PMID: 9106102 DOI: 10.1097/00005176-199702000-00009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The capacity to generate (interferon-gamma) IFN-gamma, a potent immunoregulatory and inflammatory cytokine, is low in neonates and deficient in patients with food allergy. METHODS We investigated the effect of IFN-gamma on antigen transport in the gut. In experiment I rat pups were randomized into two groups at the age of 14 days i.e., before gut maturation: Group IFN was given intraperitoneally recombinant rat IFN-gamma on days 14, 16, 18, 20. In experiment II, rats were randomized into two groups at the age of 26 days, i.e., after gut maturation: Group IFN received the IFN-gamma treatment on days 26, 28, 30, 32. Controls in both experiments received sterile saline. The absorption of horseradish peroxidase (HRP) across jejunal segments with and without Peyer's patches was studied in Ussing chambers on days 21 and 33 for experiments I and II, respectively. RESULTS In experiment I, the absorption of intact HRP across both types of segments was significantly increased in Group IFN compared to controls. The mean (95% confidence interval) rate of degraded HRP absorption across patch-containing segments in Group IFN was significantly greater than in controls, 4420 (3162-6179) ng.h-1.cm-2 in comparison to 1550 (633-3790) ng.h-1.cm-2; F = 8.96, p = 0.009. CONCLUSION IFN-gamma increases macromolecular transport before gut maturation particularly across Peyer's patches. This Peyer's patch-targeted effect can be important eliciting mucosal immune responses against dietary antigens early in life and aiding their immune exclusion.
Collapse
Affiliation(s)
- Y Sütas
- Department of Pediatrics, Medical School, University of Tampere, Finland
| | | | | | | |
Collapse
|
46
|
Christ AD, Blumberg RS. The intestinal epithelial cell: immunological aspects. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1997; 18:449-61. [PMID: 9144864 DOI: 10.1007/bf00824052] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
IECs likely play an important role in immunological defense mechanism. Apart from being a passive barrier against luminal bacteria, IECs secrete protective and microbiocidal products such as ITF, complement components and cryptdins into the lumen. Moreover, IECs produce secretory component that is essential for the transport of IgA from the lamina propria into the lumen. IECs also have regulatory functions. They express adhesion molecules important in the homing of T cells and other leukocytes, and likely modulate T cell functions in a paracrine way. Furthermore, IECs secrete cytokines, either constitutively or after bacterial challenge, and they express cytokine receptors. Lastly, IECs may play an important role as non-professional antigen-presenting cells by expressing classical MHC class I and class II and nonclassical MHC class I molecules on the cell surface. This aspect is particularly intriguing in that IECs also express a FcR that may have a function in luminal antigen sampling.
Collapse
Affiliation(s)
- A D Christ
- Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
47
|
Brandtzaeg P, Haraldsen G, Rugtveit J. Immunopathology of human inflammatory bowel disease. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1997; 18:555-89. [PMID: 9144870 DOI: 10.1007/bf00824058] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), University of Oslo, National Hospital, Rikshospitalet, Norway
| | | | | |
Collapse
|
48
|
Murch SH, Costeloe K, Klein NJ, MacDonald TT. Early production of macrophage inflammatory protein-1 alpha occurs in respiratory distress syndrome and is associated with poor outcome. Pediatr Res 1996; 40:490-7. [PMID: 8865289 DOI: 10.1203/00006450-199609000-00020] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although progression to pulmonary fibrosis in preterm infants with respiratory distress syndrome (RDS) is related to the inflammatory response, the nature of this response remains controversial. We have therefore performed sequential bronchoalveolar lavages in 30 infants with RDS (13 of whom developed bronchopulmonary dysplasia) and 7 ventilated control infants, characterizing the cells obtained by immunohistochemical analysis of lineage-specific markers and assaying macrophage-associated chemokines and cytokines in supernatant fluid. At all ages from birth, lavage supernatants demonstrated highly significant increase over controls of the beta-chemokine macrophage inflammatory protein (MIP)-1 alpha, although not of regulated upon activation, normal T cell expressed and secreted (RANTES), of the cytokines tumor necrosis factor (TNF)-alpha and IL-1 beta, and of elastase/alpha-1 antitrypsin. Significantly higher concentrations of MIP-1 alpha in particular were associated with the later development of fibrosis. Increased numbers of macrophages expressing the activation marker RM/3-1 were found at all ages in bronchopulmonary dysplasic infants, whereas neutrophil numbers were increased from d 3. Dexamethasone administered to 10 infants induced rapid decrease in inflammatory cell numbers and concentrations of MIP-1 alpha, tumor necrosis factor-alpha, IL-1 beta, and elastase/alpha-1 antitrypsin. The inflammatory response in neonatal RDS begins within the first day of life. Long-term outcome is associated with the magnitude of this early response, in particular production of MIP-1 alpha. The early introduction of specific therapy is thus likely to be beneficial.
Collapse
Affiliation(s)
- S H Murch
- Department of Neonatal Medicine, Homerton Hospital, London, England
| | | | | | | |
Collapse
|
49
|
Huang GT, Eckmann L, Savidge TC, Kagnoff MF. Infection of human intestinal epithelial cells with invasive bacteria upregulates apical intercellular adhesion molecule-1 (ICAM)-1) expression and neutrophil adhesion. J Clin Invest 1996; 98:572-83. [PMID: 8755670 PMCID: PMC507463 DOI: 10.1172/jci118825] [Citation(s) in RCA: 147] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The acute host response to gastrointestinal infection with invasive bacteria is characterized by an accumulation of neutrophils in the lamina propria, and neutrophil transmigration to the luminal side of the crypts. Intestinal epithelial cells play an important role in the recruitment of inflammatory cells to the site of infection through the secretion of chemokines. However, little is known regarding the expression, by epithelial cells, of molecules that are involved in interactions between the epithelium and neutrophils following bacterial invasion. We report herein that expression of ICAM-1 on human colon epithelial cell lines, and on human enterocytes in an in vivo model system, is upregulated following infection with invasive bacteria. Increased ICAM-1 expression in the early period (4-9 h) after infection appeared to result mainly from a direct interaction between invaded bacteria and host epithelial cells since it co-localized to cells invaded by bacteria, and the release of soluble factors by epithelial cells played only a minor role in mediating increased ICAM-1 expression. Furthermore, ICAM-1 was expressed on the apical side of polarized intestinal epithelial cells, and increased expression was accompanied by increased neutrophil adhesion to these cells. ICAM-1 expression by intestinal epithelial cells following infection with invasive bacteria may function to maintain neutrophils that have transmigrated through the epithelium in close contact with the intestinal epithelium, thereby reducing further invasion of the mucosa by invading pathogens.
Collapse
Affiliation(s)
- G T Huang
- Laboratory of Mucosal Immunology, Department of Medicine, University of California, San Diego, La Jolla, California, 92093-0623, USA
| | | | | | | |
Collapse
|
50
|
Nihei Y, Maruyama K, Endo Y, Sato T, Kobayashi K, Kaneko F. Secretory component (polymeric immunoglobulin receptor) expression on human keratinocytes by stimulation with interferon-gamma and differences in response. J Dermatol Sci 1996; 11:214-22. [PMID: 8785173 DOI: 10.1016/0923-1811(95)00444-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Secretory IgA (sIgA) is a major protective factor in the mucosal immune system because of its great ability to form complexes with bacteria. Secretory component (SC) is an 80-kDa glycoprotein, a component of sIgA, which functions as a polymeric immunoglobulin receptor for IgA and aids the secretion of sIgA from the epithelial surface. We studied SC production by keratinocytes which were involved in the inflammatory process using interferon-gamma (IFN-gamma) as one of the major inflammatory promoters produced by helper T cells. Using two human squamous cell carcinoma cell lines (HSCs) and normal human keratinocytes (NHKs), results from flow cytometric analysis, enzyme-linked immunosorbent assay (ELISA), and Northern blotting revealed that HSCs produced SC when stimulated with IFN-gamma, although their responses differed; one line exhibited enhanced SC production whereas the production in the other line was suppressed. NHKs also exhibited SC expression on the cell surface by means of immunocytochemical analysis, flow cytometry and ELISA, however the responses were also different in each strain. Although the reason for the diversity of SC expression on keratinocytes is not clear, these differences may influence epidermal sIgA secretion level.
Collapse
Affiliation(s)
- Y Nihei
- Department of Dermatology, Fukushima Medical College, Japan
| | | | | | | | | | | |
Collapse
|