1
|
Allez M. A CEACAM5-derived Peptide Activating CD8 + Regulatory T Cells: A Future Option for Restoring Mucosal Homeostasis in Crohn's Disease? Gastroenterology 2022; 163:822-824. [PMID: 35931106 DOI: 10.1053/j.gastro.2022.07.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Matthieu Allez
- Département de Gastroentérologie, Hôpital Saint-Louis, APHP, INSERM U1160, Université Paris Cité, Paris, France.
| |
Collapse
|
2
|
Characterizing CEACAM5 interaction with CD8α and CD1d in intestinal homeostasis. Mucosal Immunol 2014; 7:615-24. [PMID: 24104458 PMCID: PMC3981948 DOI: 10.1038/mi.2013.80] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 09/06/2013] [Indexed: 02/04/2023]
Abstract
Normal intestinal epithelial cells (IECs) could act as non-professional antigen-presenting cells, selectively activating CD8(+)-suppressor T cells. An epithelial cell surface glycoprotein, gp180, recognized by monoclonal antibodies B9 and L12 was determined to be critical in this process. Purification and sequence analysis of mAb B9 reactive material revealed amino-acid sequence homology with CEACAM5. We demonstrate that CEACAM5 has properties attributed to gp180, such as CD8α binding and activation of CD8-associated Lck. CEACAM5 is the only CEACAM member interacting with CD1d through the B3 domain. Its N domain (recognized by B9) is required for CD8α binding. Removal of the N-domain glycosylated residues reduces B9 recognition, CD8α binding affinity, and activation of LcK. Therefore, conformational changes in CEACAM5 glycosylation site are critical for its interaction with CD8α. CEACAM5-activated CD8(+) T cells acquire the ability to suppress the proliferation of CD4(+) T cells in vitro in the presence of interleukin (IL)-15 or IL-7. We provide new insights into the role of CEACAM5 and define its specific immunoregulatory properties among the CEACAMs expressed on IECs. We suggest that unique set of interactions between CEACAM5, CD1d, and CD8 render CD1d more class I-like molecule, facilitating antigen presentation and activation of CD8(+)-suppressor regulatory T cells.
Collapse
|
3
|
The role of the gut mucosal immunity in the development of tolerance against allergy to food. Curr Opin Allergy Clin Immunol 2010; 10:220-5. [PMID: 20431370 DOI: 10.1097/aci.0b013e32833982ab] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To provide an overview on the role of gut mucosal immunity in the development of tolerance against allergy to food. RECENT FINDINGS The gastrointestinal tract, through innate and specific immunologic factors, acts as a defense against ingested antigens. In addition to the mucous membrane integrity and digestion, numerous specific immunologic cells and mediators orchestrate such defensive mechanisms. In case of food antigens, the outcome is usually in favor of tolerance. Defects in that barrier, however, can lead to the development of aberrant immunologic responses, including hypersensitivity reactions. SUMMARY The prevailing evidence is that healthy mucosal immunity plus appropriate feeding regimen during early infancy are in favor of food tolerance. However, in addition to genetic predisposition, development of allergy is facilitated by defects in the gut barrier (immune or nonimmune) and the food allergen load.
Collapse
|
4
|
Roda G, Sartini A, Zambon E, Calafiore A, Marocchi M, Caponi A, Belluzzi A, Roda E. Intestinal epithelial cells in inflammatory bowel diseases. World J Gastroenterol 2010; 16:4264-71. [PMID: 20818809 PMCID: PMC2937106 DOI: 10.3748/wjg.v16.i34.4264] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of inflammatory bowel diseases (IBDs) seems to involve a primary defect in one or more of the elements responsible for the maintenance of intestinal homeostasis and oral tolerance. The most important element is represented by the intestinal barrier, a complex system formed mostly by intestinal epithelial cells (IECs). IECs have an active role in producing mucus and regulating its composition; they provide a physical barrier capable of controlling antigen traffic through the intestinal mucosa. At the same time, they are able to play the role of non-professional antigen presenting cells, by processing and presenting antigens directly to the cells of the intestinal immune system. On the other hand, immune cells regulate epithelial growth and differentiation, producing a continuous bi-directional cross-talk within the barrier. Several alterations of the barrier function have been identified in IBD, starting from mucus features up to its components, from epithelial junctions up to the Toll-like receptors, and altered immune responses. It remains to be understood whether these defects are primary causes of epithelial damage or secondary effects. We review the possible role of the epithelial barrier and particularly describe the role of IECs in the pathogenesis of IBD.
Collapse
|
5
|
Roda G, Dahan S, Mezzanotte L, Caponi A, Roth-Walter F, Pinn D, Mayer L. Defect in CEACAM family member expression in Crohn's disease IECs is regulated by the transcription factor SOX9. Inflamm Bowel Dis 2009; 15:1775-83. [PMID: 19637360 PMCID: PMC3005567 DOI: 10.1002/ibd.21023] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 05/12/2009] [Indexed: 12/15/2022]
Abstract
BACKGROUND CEACAM1, CEACAM5, and CEACAM6 represent 3 of the CEACAM (carcinoembryonic antigen-related cell adhesion molecule) subfamily members expressed on intestinal epithelial cells (IECs). Deficiency in their expression, as seen in inflammatory bowel disease (IBD), results in the lack of activation of CD8+ regulatory T cells in the mucosa. Since CEACAM expression was shown to be regulated by the transcription factor SOX9, we sought to determine whether the defect in CEACAM expression in IBD was related to aberrant SOX9 expression. METHODS IECs and lamina propria lymphocytes (LPLs) were freshly isolated from colonic tissues. T84 and HT29 16E cells were cocultured with LPLs. SOX9 and CEACAM subfamily member expression was assessed by real-time polymerase chain reaction (PCR), Western blot, immunohistochemistry, and immunofluorescence. RESULTS In Crohn's disease (CD) but not in ulcerative colitis (UC), a significant reduction in mRNA and protein expression for CEACAM1 and 5 was noted; in contrast, no difference in SOX9 mRNA expression was seen. However, nuclear SOX9 immunostaining was increased in CD IECs. Furthermore, SOX9 protein was reduced in the cytoplasm of LPL-stimulated T84 and HT29 16E cells, while CEACAM5 expression was increased. CONCLUSIONS The defect in CEACAM family members in CD IECs appears to be related to the aberrant nuclear localization of SOX9. Changes in SOX9 expression in the CD mucosa relate to the local microenvironment and altered IEC:LPL crosstalk.
Collapse
Affiliation(s)
- Giulia Roda
- Department of Clinical medicine, Gastroenterology Unit, S. Orsola Hospital, Bologna, Italy
| | - Stephanie Dahan
- Immunology Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Laura Mezzanotte
- Departement of Pharmaceutical Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Caponi
- Department of Clinical medicine, Gastroenterology Unit, S. Orsola Hospital, Bologna, Italy
| | - Franziska Roth-Walter
- Center of Physiology and Pathophysiology, Medical University of Vienna, Vienna, Austria
| | - David Pinn
- Immunology Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Lloyd Mayer
- Immunology Institute, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
6
|
Konya C, Goronzy JJ, Weyand CM. Treating autoimmune disease by targeting CD8(+) T suppressor cells. Expert Opin Biol Ther 2009; 9:951-65. [PMID: 19522557 DOI: 10.1517/14712590903020759] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Current treatments for autoimmune disease are hampered by the non-specificity of immunomodulatory interventions, having to accept broad suppression of immunoresponsiveness with potentially serious side effects, such as infection or malignancy. The development of antigen-specific approaches, downregulating pathogenic immune responses while maintaining protective immunity, would be a major step forward. One possible approach involves the targeting of physiological regulatory mechanisms, such as inhibitory CD8 T cells that are now recognized to fine-tune many aspects of immune responses. CD8 T suppressor (Ts) cells may directly inhibit other T cells or condition antigen-presenting cells in such a way that immune amplification steps are dampened. The promise of CD8 Ts cells lies in their potential to disrupt host-injurious immune responses in a targeted fashion. For therapeutic purposes, such CD8 Ts cells could either be generated in vitro and transferred into the host or their numbers and activity could be modulated by treating the patient with established or novel immunomodulators. Emerging evidence shows that several subsets of CD8 Ts cells exist. While there is still considerable uncertainty about the molecular mechanisms through which CD8 Ts cells can reset immune responses to protect the host, their potential diagnostic and therapeutic use is intriguing and has generated renewed interest.
Collapse
Affiliation(s)
- Christine Konya
- Emory University School of Medicine, Kathleen B. and Mason I. Lowance Center for Human Immunology and Rheumatology, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
7
|
French JD, Roark CL, Born WK, O'Brien RL. Gammadelta T lymphocyte homeostasis is negatively regulated by beta2-microglobulin. THE JOURNAL OF IMMUNOLOGY 2009; 182:1892-900. [PMID: 19201842 DOI: 10.4049/jimmunol.0803165] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Successful application of gammadelta T cells in adoptive cell therapies depends upon our ability to maintain these cells in vivo. Using an adoptive transfer model to study lymphopenia-induced homeostatic expansion, we show that CD8(+) and NK1.1(+) gammadelta T cell subsets are differentially regulated. While CD8(+) gammadelta T cells have an early and sustained advantage following transfer into TCRbeta(-/-)/delta(-/-) mice, NK1.1(+) gammadelta T cells proliferate slowly and are maintained at low numbers. The advantage of the CD8(+) subset could not be explained by increased bcl-2 or cytokine receptor expression but did correlate with Vgamma4(+) and Vdelta5(+) expression. Despite the role of CD8 in MHC class I recognition by alphabeta T cells, beta(2)-microglobulin (beta(2)m)-associated MHC class I molecules were not required for CD8(+) gammadelta T cell homeostatic expansion. Surprisingly, all gammadelta T cells, including the CD8(+) subset, exhibited enhanced proliferation following adoptive transfer into Rag1(-/-)/beta(2)m(-/-) compared with Rag1(-/-) recipients. This effect was most notable for the NK1.1(+) subset, which expresses high levels of NKG2A/CD94 and Ly49. Although expression of these inhibitory receptors correlated with poor homeostatic expansion in the presence of beta(2)m, gammadelta T cell homeostatic proliferation in TCRbeta(-/-)/delta(-/-) mice was not altered in the presence of Ly49C/I- and NKG2-blocking Abs. While the mechanism by which beta(2)m negatively regulates gammadelta T cell homeostasis remains to be determined, this observation is unique to gammadelta T cells and confirms that multiple mechanisms are in place to maintain strict regulation of both the size and the composition of the gammadelta T cell pool.
Collapse
Affiliation(s)
- Jena D French
- National Jewish Health, Integrated Department of Immunology, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
8
|
Nanni P, Mezzanotte L, Roda G, Caponi A, Levander F, James P, Roda A. Differential proteomic analysis of HT29 Cl.16E and intestinal epithelial cells by LC ESI/QTOF mass spectrometry. J Proteomics 2009; 72:865-73. [PMID: 19168159 DOI: 10.1016/j.jprot.2008.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 11/27/2008] [Accepted: 12/31/2008] [Indexed: 12/15/2022]
Abstract
Intestinal epithelial cells (IECs) play a key role in Crohn's disease, a chronic inflammatory bowel disease which requires invasive examinations to be diagnosed. The comparison of the cellular protein expression profiles of Crohn's disease patients and healthy subjects is fundamental for the identification of proteins clinically relevant as new biomarkers or as drug targets. For this purpose a differential label-free nano-LC ESI/QTOF mass spectrometry (MS) approach combined with targeted MS/MS analysis has been developed and applied to isolated IECs. We report here a study of the protein variations in IECs from healthy subjects (H) and Crohn's disease patients (CD). The method was previously validated using HT29 Cl.16E cell line, normal or treated with interferon-gamma as a model of inflammation. Subcellular fractions proteins were extracted from HT29 and IECs and for each fraction monodimensional gel-electrophoresis was performed and the proteins subjected to tryptic digestion. The resulting peptides were analysed by LC ESI/QTOF MS and the obtained chromatographic runs were aligned with msInspect software. The peptides differently expressed were statistically evaluated using the Proteios Software Environment (ProSE) and identified by LC ESI/QTOF MS/MS analysis and database search. The preliminary results obtained allowed the identification of many proteins involved in the inflammation processes.
Collapse
Affiliation(s)
- Paolo Nanni
- Department of Pharmaceutical Sciences, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | | | | | | | | | | | | |
Collapse
|
9
|
Dotan I, Allez M, Nakazawa A, Brimnes J, Schulder-Katz M, Mayer L. Intestinal epithelial cells from inflammatory bowel disease patients preferentially stimulate CD4+ T cells to proliferate and secrete interferon-gamma. Am J Physiol Gastrointest Liver Physiol 2007; 292:G1630-40. [PMID: 17347451 DOI: 10.1152/ajpgi.00294.2006] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies have suggested that intestinal epithelial cells (IECs) have the capacity to function as nonprofessional antigen presenting cells that in the normal state preferentially activate CD8+ T cells. However, under pathological conditions, such as those found in inflammatory bowel disease (IBD), persistent activation of CD4+ T cells is seen. The aim of this study was to determine whether the IBD IECs contribute to CD4+ T cell activation. Freshly isolated human IECs were obtained from surgical specimens of patients with or without IBD and cocultured with autologous or allogeneic peripheral blood T lymphocytes. Cocultures of normal T cells and IECs derived from IBD patients resulted in the preferential activation of CD4+ T cell proliferation that was associated with significant IFN-gamma, but not IL-2, secretion. Cytokine secretion and CD4+ T cell proliferation was inhibited by pretreatment of the IBD IECs with the anti-DR MAb L243. In contrast, normal IECs stimulated the proliferation and cytokine secretion by CD4+ T cells to a significantly lesser degree than IBD IECs. Furthermore, blockade of human leukocyte antigen-DR had a lesser effect in the normal IEC-CD4+ T cell cocultures. We conclude that IECs can contribute to the ongoing CD4+ T cell activation seen in IBD. We suggest that the apparent differences between the secreted levels of IFN-gamma indicate that it may play a dual role in intestinal homeostasis, in which low levels contribute to physiological inflammation whereas higher levels are associated with an uncontrolled inflammatory state.
Collapse
Affiliation(s)
- Iris Dotan
- IBD Service, Dept. of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel.
| | | | | | | | | | | |
Collapse
|
10
|
Perera L, Shao L, Patel A, Evans K, Meresse B, Blumberg R, Geraghty D, Groh V, Spies T, Jabri B, Mayer L. Expression of nonclassical class I molecules by intestinal epithelial cells. Inflamm Bowel Dis 2007; 13:298-307. [PMID: 17238179 DOI: 10.1002/ibd.20026] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well recognized that the nature of the immune response is different in the intestinal tract than in peripheral lymphoid organs. The immunologic tone of the gut-associated lymphoid tissue is one of suppression rather than active immunity, distinguishing pathogens from normal flora. Failure to control mucosal immune responses may lead to inflammatory diseases such as Crohn's disease (CD) and ulcerative colitis (UC) and celiac disease. It has been suggested that this normally immunosuppressed state may relate to unique antigen-presenting cells and unique T-cell populations. The intestinal epithelial cell (IEC) has been proposed to act as a nonprofessional antigen-presenting cell (APC). Previous studies have suggested that antigens presented by IECs result in the activation a CD8(+) regulatory T-cell subset in a nonclassical MHC I molecule restricted manner. We therefore analyzed the expression of nonclassical MHC I molecules by normal IECs and compared this to those expressed by inflammatory bowel disease (IBD) IECs. Normal surface IEC from the colon and, to a much lesser extent, the small bowel express nonclassical MHC I molecules on their surface. In contrast, mRNA is expressed in all intestinal epithelial cells. Surface IEC express CD1d, MICA/B, and HLA-E protein. In contrast, crypt IECs express less or no nonclassical MHC I molecules but do express mRNA for these molecules. Furthermore, the regulation of expression of distinct nonclassical class I molecules is different depending on the molecule analyzed. Interestingly, IECs derived from patients with UC fail to express any nonclassical MHC I molecules (protein and HLA-E mRNA). IECs from CD patients express HLA-E and MICA/B comparable to that seen in normal controls but fail to express CD1d. Thus, in UC there may be a failure to activate any nonclassical MHC I molecule restricted regulatory T cells that may result in unopposed active inflammatory responses. In CD only the CD1d-regulated T cells would be affected.
Collapse
Affiliation(s)
- Lilani Perera
- Immunobiology Center, Mount Sinai Medical Center, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Safadi R, Alvarez CE, Ohta M, Brimnes J, Kraus T, Mehal W, Bromberg J, Mayer L, Friedman SL. Enhanced oral tolerance in transgenic mice with hepatocyte secretion of IL-10. THE JOURNAL OF IMMUNOLOGY 2005; 175:3577-83. [PMID: 16148101 DOI: 10.4049/jimmunol.175.6.3577] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Several cytokines derived from Th3 and Tr1 cells, including IL-10, are believed to regulate oral tolerance, but direct evidence is lacking. We have explored the potential role of IL-10 by generating transgenic (TG) mice with sustained hepatocyte-specific expression of rat IL-10. TG mice expressed rat IL-10 downstream of a transthyretin promoter, which led to serum levels that were increased 10- to 100-fold compared with normal animals. Animals were orally administered 1 mg of whole OVA for 5 consecutive days, with control animals receiving PBS. There were six animal groups: Either OVA or PBS were fed orally to rat IL-10 TG mice, non-TG wild-type mice without IL-10 administration, and non-TG wild-type mice administered rat IL-10 systemically. On day 8, all mice were immunized with two injections of OVA, and then analyzed on day 18. T cell proliferation responses were reduced by 65.8 +/- 14.3% after feeding of OVA in rIL-10 TG animals, compared with 39.4 +/- 15.6% in the non-TG mice (p = 0.02). Anti-OVA titers were expressed as fold increase over naive non-TG mice. After feeding, titers decreased by approximately 33% (from 3- to 2-fold) in TG animals and, to a lesser extent, in non-TG animals. IFN-gamma secretion by cultured popliteal lymphocytes decreased in TG animals by 83% after feeding and by 69% in non-TG animals. IL-4 secretion increased 4-fold in TG-fed mice, but did not significantly change in non-TG OVA-fed animals. In contrast to hepatic TG expression of rIL-10, systemic administration of rIL-10 had only a modest effect on tolerance. IL-10, when transgenically expressed in the liver enhances mucosal tolerance to an oral Ag.
Collapse
Affiliation(s)
- Rifaat Safadi
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Shao L, Jacobs AR, Johnson VV, Mayer L. Activation of CD8+ regulatory T cells by human placental trophoblasts. THE JOURNAL OF IMMUNOLOGY 2005; 174:7539-47. [PMID: 15944253 DOI: 10.4049/jimmunol.174.12.7539] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immunological basis by which a mother tolerates her semi-allogeneic fetus remains poorly understood. Several mechanisms are likely to contribute to this phenomenon including active immune regulation by regulatory T cells. In this article, we report that human placental trophoblasts activate a clonal population of CD8(+) T cells with regulatory function. These cells are not MHC class I restricted, but require costimulation through a member of the carcinoembryonic Ag family present on early gestation trophoblasts. These regulatory T cells express the mucosal markers CD101 and CD103 and display selective usage of the TCR gene Vbeta9. CD8(+) T cells isolated from the peripheral blood of pregnant mothers (16-28 wk) also demonstrate expansions in the same Vbeta family (Vbeta9), signaling a possible role for these cells in preventing fetal rejection in vivo. We have previously characterized a subset of CD8(+) regulatory T cells activated by the combination of the nonclassical class I molecule CD1d and a costimulatory molecule of the carcinoembryonic Ag family present on the intestinal epithelium. These data support the concept that distinct regulatory T cell populations exist at different sites and may be regulated locally by unique restriction elements, costimulatory signals, and Ags.
Collapse
Affiliation(s)
- Ling Shao
- Immunobiology Center and Department of Obstetrics and Gynecology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
13
|
Shao L, Kamalu O, Mayer L. Non-classical MHC class I molecules on intestinal epithelial cells: mediators of mucosal crosstalk. Immunol Rev 2005; 206:160-76. [PMID: 16048548 DOI: 10.1111/j.0105-2896.2005.00295.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The mucosal immune environment consists of a complex combination of lymphoid cells, non-lymphoid cells, and lumenal bacteria. Signals from lumenal bacteria are constantly transmitted to the underlying tissues across the intestinal epithelial barrier. Intestinal epithelial cells (IECs) can sense these signals, integrate them, and interpret them for lamina propria lymphoid populations. One mechanism by which these signals are communicated is by the expression of non-classical major histocompatibility complex (MHC) class I molecules by IECs. Epithelial cells can express a surprising variety of non-classical MHC class I molecules. In some cases, IECs can act as non-professional antigen-presenting cells utilizing the expression of such non-classical MHC class I molecules to directly present bacterial antigens. In other cases, the expression of non-classical MHC class I molecules may act as a co-stimulatory molecule or adhesion molecule that can modify the mucosal immune response. Finally, the expression of these molecules on IECs can lead to a broad array of responses ranging from tolerance to inflammation. Overall, the IEC, via the expression of non-classical MHC class I molecules, is a central mediator of the constant crosstalk between the intestinal lumen and the mucosal immune system.
Collapse
Affiliation(s)
- Ling Shao
- The Center for Immunobiology, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
14
|
Kraus TA, Brimnes J, Muong C, Liu JH, Moran TM, Tappenden KA, Boros P, Mayer L. Induction of mucosal tolerance in Peyer's patch-deficient, ligated small bowel loops. J Clin Invest 2005; 115:2234-43. [PMID: 16041410 PMCID: PMC1177996 DOI: 10.1172/jci19102] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2003] [Accepted: 05/24/2005] [Indexed: 11/17/2022] Open
Abstract
To explore the requirement for M cells and the Peyer's patch (PP) in induction of oral tolerance and address the potential in vivo role of intestinal epithelial cells as nonprofessional APCs, we have attempted to induce tolerance in mice with ligated small bowel loops without M cells and Peyer's patches. A 2-centimeter section of vascularized small bowel was spliced away from the gut without disruption of the mesenteric attachments. We introduced OVA directly into the lumen of the loop prior to footpad immunization. By excising segments of bowel that contain PPs in some mice and segments without patches in others, we could study the necessity of the M cell and the underlying patch versus epithelial cells in induction of mucosal tolerance. We show that OVA-specific T cell proliferation and serum antibody responses are reduced in mice that have previously been given OVA both in PP-containing loops and in loops without patches. Furthermore, both high- and low-dose tolerance could be induced in the absence of PPs. Low-dose tolerance is associated with bystander suppression and requires IL-10, which indicates active suppression and the induction of regulatory cells. These data suggest that there is a critical role for components of the mucosal immune system other than PPs in antigen sampling and induction of oral tolerance.
Collapse
Affiliation(s)
- Thomas A Kraus
- Immunobiology Center, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Brimnes J, Allez M, Dotan I, Shao L, Nakazawa A, Mayer L. Defects in CD8+ regulatory T cells in the lamina propria of patients with inflammatory bowel disease. THE JOURNAL OF IMMUNOLOGY 2005; 174:5814-22. [PMID: 15843585 DOI: 10.4049/jimmunol.174.9.5814] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mucosal tolerance is believed to be partly mediated by regulatory T cells. Intestinal epithelial cells (IECs) may play an important role in the generation of such regulatory cells, because they are able to process and present Ag to T cells. Furthermore, we have previously demonstrated that IECs are able to generate regulatory CD8(+) T cells in vitro. In the present study, we have analyzed lamina propria (LP) lymphocytes for the presence of such regulatory CD8(+) T cells in normal individuals as well as in patients with inflammatory bowel disease (IBD). The results of the present study show that LP CD8(+) T cells derived from normal controls possess regulatory activity, whereas both unfractionated LP lymphocytes and purified LP CD4(+) T cells do not. The LP CD8(+) T cells suppress Ig production by pokeweed mitogen-stimulated PBMCs by 31-80%, in a cell contact-dependent manner. No significant difference in suppression between CD28(+) and CD28(-)CD8(+) LP T cells was observed. In contrast to CD8(+) T cells from normal LP, CD8(+) T cells isolated from LP of IBD patients, did not suppress Ig production by pokeweed mitogen-stimulated PBMC (five of six ulcerative colitis specimens; six of six Crohn's disease specimens). Furthermore, we demonstrate that the frequency of TCR Vbeta5.1-positive CD8(+) T cells, which we previously have demonstrated to be regulatory and to be expanded by IECs in vitro, is decreased in IBD LP compared with normal LP. In conclusion, this study demonstrates that CD8(+) T cells with regulatory activity are present in the LP of normal healthy individuals, but not in patients with IBD, suggesting that these cells might play an active role in mucosal tolerance.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Communication/immunology
- Cells, Cultured
- Coculture Techniques
- Cytokines/pharmacology
- Female
- Humans
- Immunoglobulins/biosynthesis
- Inflammatory Bowel Diseases/immunology
- Inflammatory Bowel Diseases/metabolism
- Inflammatory Bowel Diseases/pathology
- Intestinal Mucosa/cytology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/pathology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Lymphocyte Culture Test, Mixed
- Male
- Middle Aged
- Receptors, Antigen, T-Cell/biosynthesis
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Jens Brimnes
- Immunobiology Center, Mount Sinai Medical Center, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
16
|
|
17
|
|
18
|
Carton J, Byrne B, Madrigal-Estebas L, O'Donoghue DP, O'Farrelly C. CD4+CD8+ human small intestinal T cells are decreased in coeliac patients, with CD8 expression downregulated on intra-epithelial T cells in the active disease. Eur J Gastroenterol Hepatol 2004; 16:961-8. [PMID: 15371918 DOI: 10.1097/00042737-200410000-00003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND/OBJECTIVE The intestinal lesion of coeliac disease is thought to be initiated and exacerbated by dysregulation of local T-lymphocyte sub-populations. This study examines changes in intestinal T cells from coeliac patients, with a particular focus on CD4CD8 T cells, immunoregulatory cells normally found in relatively high proportions in the small intestine. METHODS Cells were obtained from duodenal biopsies from active and treated coeliac patients using chelating and reducing agents (epithelial layer) followed by collagenase treatment (lamina propria). Cell yield and viability were assessed and flow cytometric analysis was used to examine CD4CD8 T cells and to quantify CD8 expression. RESULTS Surprisingly, total T-cell yields in the epithelial layer did not increase in active coeliac disease although enterocyte counts decreased significantly, giving an appearance of infiltration. In active coeliac patients, CD4CD8 T cell percentages were significantly decreased in both the epithelial layer and lamina propria. Levels of CD8 expression by CD4CD8 T cells in the epithelial layer were decreased significantly in patients with active coeliac disease. CD4CD8 T cell proportions did not return to normal in treated coeliac patients whose villous architecture had responded to gluten withdrawal. CONCLUSIONS No increase of intra-epithelial lymphocytes in the coeliac lesion may require us to reconsider the definition of coeliac disease as an inflammatory condition. Low CD4CD8 populations in treated as well as untreated coeliac patients indicate that these T cells are inherently absent in individuals genetically predisposed to coeliac disease.
Collapse
Affiliation(s)
- Janet Carton
- Education and Research Centre, St Vincent's University Hospital, Dublin, Ireland
| | | | | | | | | |
Collapse
|
19
|
Abstract
The mucosa-associated lymphoid tissue (MALT) has the task of protecting the host from pathogens while maintaining the integrity of the gut. Immune responses are tightly regulated such that there is tolerance of nonpathogenic bacteria as well as dietary antigens present in the intestinal lumen. The failure to control these responses leads to a disruption in tolerance, which has been proposed as one mechanism involved in the development of inflammatory bowel disease (IBD). Different mechanisms are involved in the control of immune responses in the intestinal tract, including active suppression by regulatory T cells. Distinct subsets of regulatory T cells coexist in the intestinal mucosa, which is a fertile environment for their growth. Most of these are defined by their phenotype and/or their ability to produce regulatory cytokines such as interleukin-10 and transforming growth factor-beta A lack of activation and/or expansion of regulatory cells could play a role in the uncontrolled inflammation seen in IBD. Regulatory T cells may be activated by cytokines, and their inductive phase may be antigen-driven. There are limited data relating to the true surface interactions regulating the activation of these cells. Most of the CD4 regulatory T cells (Tr1, Th3, and CD4 CD25+) are thought to interact with dendritic cells. Subsets of regulatory T cells (such as CD8 TrE cells) may recognize antigens presented by intestinal epithelial cells. A better understanding of the mechanisms by which these regulatory T cells are expanded and/or activated in the intestinal mucosa may provide clues as how to use them as a novel therapeutic tool in the treatment of patients with IBD.
Collapse
Affiliation(s)
- Matthieu Allez
- Service de Gastroentérologic and INSERM U396, Hôpital Saint-Louis, Paris, France.
| | | |
Collapse
|
20
|
Nakazawa A, Dotan I, Brimnes J, Allez M, Shao L, Tsushima F, Azuma M, Mayer L. The expression and function of costimulatory molecules B7H and B7-H1 on colonic epithelial cells. Gastroenterology 2004; 126:1347-57. [PMID: 15131796 DOI: 10.1053/j.gastro.2004.02.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Previous studies have suggested that intestinal epithelial cells (IECs) may function as antigen-presenting cells for CD4+ and CD8+ T cells. However, these cells fail to express conventional costimulatory molecules (CD80, CD86), leading to the possibility that antigen presented by normal IECs could result in anergy. Other members of the B7 family have recently been identified. B7h interacts with inducible costimulator (ICOS) on T cells and provides a positive signal, whereas B7-H1 and B7-DC interact with PD-1 and transmit an inhibitory signal. Our aim was to determine whether IECs express novel B7 family members and whether these molecules play a role in IEC:T-cell interactions. METHODS B7h and B7-H1 expression was assessed in isolated IECs and IEC lines. The functional role of B7h and B7-H1 in the interaction between IECs and T cells was assessed in coculture experiments using purified anti-B7h or B7-H1 monoclonal antibodies (mAbs), B7h immunoglobulin (Ig), or B7-H1 fusion proteins. RESULTS B7h and B7-H1 messenger RNA was detected in IEC lines and IECs from healthy controls and patients with inflammatory bowel disease (IBD). IECs from patients with IBD but not healthy controls expressed B7h and B7-H1 protein on their surface. Proliferation of IEC-stimulated T cells was inhibited only by B7h immunoglobulin treatment, whereas interferon gamma secretion in these cocultures was inhibited by both anti-B7h mAb and B7h Ig. No difference was seen between IBD or normal IEC populations. CONCLUSIONS These data suggest that the B7h-ICOS costimulatory pathway may be important in IEC:T-cell interactions.
Collapse
Affiliation(s)
- Atsushi Nakazawa
- Immunobiology Center, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Allez M, Brimnes J, Dotan I, Mayer L. Expansion of CD8+ T cells with regulatory function after interaction with intestinal epithelial cells. Gastroenterology 2002; 123:1516-26. [PMID: 12404227 DOI: 10.1053/gast.2002.36588] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Regulatory T cells play a role in the control of immune responses in the intestinal mucosa and their absence may predispose to inflammatory bowel disease (IBD). We have previously shown that T cells activated by intestinal epithelial cells (IECs) are suppressive in function. Our goal was to characterize the phenotype and function of T cells proliferating after interaction with IECs. METHODS Irradiated human IECs, isolated from normal resection specimens, were cultured with carboxy fluorescein succinimidyl ester (CFSE) labeled T cells. Flow cytometric analysis of T cells was performed at days 5-10. CD8+ T cells proliferating in culture with IECs were sorted and added to suppressive assays. RESULTS The precursor frequency of T cells proliferating in response to IECs ranged from 0.3%-0.9%. Several subpopulations were shown to proliferate (CD8+CD28-/CD8+CD28+/CD4+CD25+), but one population (CD8+CD28-CD101+CD103+) appeared to be dependent on contact with the CD8 ligand gp180. After sorting, culture in the presence of interleukin (IL)-7 and IL-15 allowed for the generation of cell lines. IEC-activated CD8+ T cells, but not nonactivated CD8+ T cells, were suppressive in function. Suppression belonged to the CD101+CD103+ subset of IEC-activated CD8+ T cells and appeared to require cell contact. CD8+ lamina propria T cells also showed suppressive function, suggesting the presence of CD8+ regulatory T cells in the mucosa. CONCLUSIONS IECs are able to induce the proliferation of a small fraction of CD8+ peripheral T cells. The CD8+CD28- subset of IEC-activated CD8+ T cells, which express CD101 and CD103, interacts with IECs through gp180 and has regulatory function.
Collapse
Affiliation(s)
- Matthieu Allez
- Immunobiology Center, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | | | | | |
Collapse
|
22
|
Devine L, Rogozinski L, Naidenko OV, Cheroutre H, Kavathas PB. The complementarity-determining region-like loops of CD8 alpha interact differently with beta 2-microglobulin of the class I molecules H-2Kb and thymic leukemia antigen, while similarly with their alpha 3 domains. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3881-6. [PMID: 11937542 DOI: 10.4049/jimmunol.168.8.3881] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The murine CD8 glycoprotein interacts with both classical MHC class I molecules and some nonclassical molecules, including the thymic leukemia Ag (TL). TL binds preferentially to CD8alphaalpha homodimers with a 10-fold higher affinity than H-2K(b) class I molecules. To understand the molecular basis for this difference, we created a panel of CD8alpha mutants and tested the ability of the CD8alphaalpha homodimers to bind to H-2K(b) tetramers and TL tetramers. Mutations in three CD8 residues located on the complementarity-determining region-like loops contacting the negatively charged loop in the alpha3 domain of MHC class I greatly reduced binding to both tetramers. Because TL and H-2K(b) class I sequences are highly conserved in the alpha3 domain of MHC class I, this suggests that CD8 contacts the alpha3 domain of TL and H-2K(b) in a similar manner. In contrast, mutations in residues on the A and B beta strands of CD8 that are involved in contact with beta(2)-microglobulin affected interaction with the H-2K(b) tetramer, but not the TL tetramer. Therefore, the orientation of interaction of TL with CD8 appears to be different from that of H-2K(b). The unique high affinity binding of TL with CD8alphaalpha is most likely a result of amino acid differences in the alpha3 domain between TL and H-2K(b), particularly at positions 198 (K to D) and 228 (M to T), which are contact residues in the CD8alphaalpha-H-2K(b) cocrystal.
Collapse
Affiliation(s)
- Lesley Devine
- Department of Laboratory Medicine and Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
23
|
Campbell NA, Park MS, Toy LS, Yio XY, Devine L, Kavathas P, Mayer L. A non-class I MHC intestinal epithelial surface glycoprotein, gp180, binds to CD8. Clin Immunol 2002; 102:267-74. [PMID: 11890713 DOI: 10.1006/clim.2001.5170] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The activation of CD8(+) T cells by normal intestinal epithelial cells in antigen-specific or allogeneic mixed cell culture systems has significant implications for the modulation of mucosal immune responses due to the fact that these T cells appear to have regulatory rather than cytolytic activity. A 180-kDa glycoprotein (gp180) has been identified and shown to be important in CD8(+) T cell activation by intestinal epithelial cells. In this study, we examine, in further detail, the role that the CD8 molecule plays in this interaction. It has been previously shown that monoclonal antibodies against gp180 inhibited the activation of CD8-associated p56(lck) in T cells. Although indirectly suggested by these data, there was no evidence that the activation of this protein tyrosine kinase was a direct result of gp180 interacting with the CD8 molecule. In this study, we document that soluble gp180 is able to bind to CD8-Fc fusion proteins and is absorbed by human CD8 alpha but not CD4 transfected murine T cells and that this interaction is dependent upon carbohydrate on the gp180 molecule. Furthermore, the sites used for binding by gp180 are distinct from those used by the conventional CD8 ligand, class I MHC. Thus, gp180 appears to be a novel CD8 ligand that plays an important role in the activation of CD8-associated kinases and of CD8(+) T cells.
Collapse
Affiliation(s)
- N A Campbell
- Division of Clinical Immunology, Mount Sinai Medical Center, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Nakajima A, Iijima H, Neurath MF, Nagaishi T, Nieuwenhuis EES, Raychowdhury R, Glickman J, Blau DM, Russell S, Holmes KV, Blumberg RS. Activation-induced expression of carcinoembryonic antigen-cell adhesion molecule 1 regulates mouse T lymphocyte function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1028-35. [PMID: 11801635 DOI: 10.4049/jimmunol.168.3.1028] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) consists of highly related homologs in humans and rodents that are characterized by significant alternate splicing generating isoforms capable of negative intracellular signaling by virtue of two immunoreceptor tyrosine-based inhibition motifs in its cytoplasmic (cyt) tail. Although human T cells have been recently observed to express CEACAM1, the expression and function of CEACAM1 in mouse T cells have not been defined. Although resting mouse spleen T cells exhibited no evidence of CEACAM1 on the cell surface, CEACAM1 was rapidly up-regulated on CD4+ and CD8+ T cells after activation with either Con A or anti-CD3 without a requirement for either de novo transcription or translation due to the fact that CEACAM1 was present intracellularly before activation. Using a GST-CEACAM1-cytoplasmic tail fusion protein, it was shown that the cytoplasmic tail of CEACAM1 bound the src homology domain-containing phosphatase 1 and adaptor protein 1 complex in its phosphorylated and nonphosphorylated states, respectively. CEACAM1 ligation with an anti-CEACAM1 mAb resulted in inhibition of an allogeneic MLR and anti-CD3 plus anti-CD28 Ab-induced proliferation of spleen T cells in vitro and inhibition of a delayed-type hypersensitivity response to oxazolone in vivo. Inhibition of the delayed-type hypersensitivity response required that the anti-CEACAM1-specific mAb be present at the time of T cell sensitization. These studies support a role for CEACAM1 as a novel class of immunoreceptor tyrosine-based inhibition motif-bearing regulatory molecules on T cells that are active during early phases of the immune response in mice.
Collapse
MESH Headings
- Adaptor Proteins, Vesicular Transport
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD/biosynthesis
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, CD/physiology
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Antigens, Differentiation/physiology
- Carcinoembryonic Antigen/biosynthesis
- Carcinoembryonic Antigen/immunology
- Carcinoembryonic Antigen/metabolism
- Carcinoembryonic Antigen/physiology
- Carrier Proteins/metabolism
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Cell Adhesion Molecules/physiology
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cells, Cultured
- Injections, Intraperitoneal
- Interphase/immunology
- Intracellular Signaling Peptides and Proteins
- Ligands
- Lymphocyte Activation
- Male
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Protein Phosphatase 1
- Protein Tyrosine Phosphatase, Non-Receptor Type 11
- Protein Tyrosine Phosphatase, Non-Receptor Type 6
- Protein Tyrosine Phosphatases/metabolism
- SH2 Domain-Containing Protein Tyrosine Phosphatases
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
- src Homology Domains/immunology
Collapse
Affiliation(s)
- Atsushi Nakajima
- Gastroenterology Division, Departments of Medicine and Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Human peripheral blood CD8+ T cells comprise cells that are in different states of differentiation and under the control of complex homeostatic processes. In a number of situations ranging from chronic inflammatory conditions and infectious diseases to ageing, immunodeficiency, iron overload and heavy alcohol intake, major phenotypic changes, usually associated with an increase in CD8+ T cells lacking CD28 expression, take place. CD8+CD28- T cells are characterized by a low proliferative capacity to conventional stimulation in vitro and by morphological and functional features of activated/memory T cells. Although the nature of the signals that give origin to this T-cell subset is uncertain, growing evidence argues for the existence of an interplay between epithelial cells, molecules with the MHC-class I fold and CD8+ T cells. The possibility that the generation of CD8+CD28- T cells is the combination of TCR/CD3zeta- and regulatory factor-mediated signals as a result of the sensing of modifications of the internal environment is discussed.
Collapse
Affiliation(s)
- Fernando A Arosa
- Laboratory of Molecular Immunology, Institute for Molecular and Cell Biology (IBMC) and Molecular Immunology and Pathology, Abel Salazar Institute for the Biomedical Sciences(ICBAS), Porto, Portugal.
| |
Collapse
|
26
|
Abstract
For decades intestinal epithelial cells were thought of as passive barriers to luminal contents, cells involved in nutrient absorption and electrolyte secretion. Studies during the past 10 to 15 years have changed that concept as our understanding of mucosal immunity has evolved and as we have come to understand that the regulation of this system is unique in terms of cellular interactions and factors produced. The intestinal epithelial cell has moved to the forefront of these studies where it has been shown to be an active participant in mucosal immunoregulation and inflammation. Results of the earliest studies suggested that epithelial cells might be involved in immune regulation because they expressed a series of cell surface molecules that correlate with classical antigen presentation (class I and II MHC molecules). Later on, the expression of nonclassical class I molecules was identified on these cells as well, raising the possibility of novel forms of interactions with unique cell populations. This was followed by the observation that epithelial cells secrete cytokines and chemokines, which not only regulate mucosal immune responses but also regulate inflammatory responses. By such processes, the IEC has been proposed as being a bridge between innate and adaptive immunity. The recent description of Toll-like receptors on IECs adds further support to this concept. Clearly there is growing appreciation of the multifaceted role that the IEC plays in the gut. The work on IECs in the past year has helped to refine this role.
Collapse
Affiliation(s)
- L Mayer
- Immunology Center, Mount Sinai Medical Center, New York, New York 10029, USA.
| |
Collapse
|
27
|
Hoffman RA. Intraepithelial lymphocytes coinduce nitric oxide synthase in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2000; 278:G886-94. [PMID: 10859218 DOI: 10.1152/ajpgi.2000.278.6.g886] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The study of mucosal immunity has revealed that complex reciprocal interactions occur between intestinal intraepithelial lymphocytes (IEL) and intestinal epithelial cells (IEC). The present study focuses on the induction of inducible nitric oxide (NO) synthase in cocultures of freshly isolated rat IEL and the rat epithelial cell line IEC-18 after the addition of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha, or lipopolysaccharide. When IEL and IEC were separated using Transwell chambers, NO synthesis was not induced, indicating that cell-cell contact was required. Culture of IEC-18 with IEL, even in the absence of inflammatory stimuli such as IL-1beta, resulted in upregulation of class I and II antigens on IEC-18, due to the interferon-gamma (IFN-gamma) that is constitutively produced by IEL. Addition of anti-IFN-gamma antibody to the NO-producing cocultures resulted in inhibition of NO synthesis as well as the upregulation of class I and II antigen expression. These data indicate that IFN-gamma production by IEL conditions IEC for the expression of other components of the inflammatory process.
Collapse
Affiliation(s)
- R A Hoffman
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.
| |
Collapse
|
28
|
Hershberg RM, Mayer LF. Antigen processing and presentation by intestinal epithelial cells - polarity and complexity. IMMUNOLOGY TODAY 2000; 21:123-8. [PMID: 10689299 DOI: 10.1016/s0167-5699(99)01575-3] [Citation(s) in RCA: 189] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The mechanisms by which gut-associated lymphoid tissue (GALT) maintains a balance between oral tolerance and active immune response in the face of exposure to high antigen concentrations remains a central question in mucosal immunity. Here, Robert Hershberg and colleagues discuss the evidence that human intestinal epithelial cells function as antigen-presenting cells (APCs) capable of regulating T-cell responses in the intestinal mucosa
Collapse
Affiliation(s)
- R M Hershberg
- Divisions of Gastroenterology and Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
29
|
Yun CH, Lillehoj HS, Lillehoj EP. Intestinal immune responses to coccidiosis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2000; 24:303-324. [PMID: 10717295 DOI: 10.1016/s0145-305x(99)00080-4] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Intestinal parasitism is a major stress factor leading to malnutrition and lowered performance and production efficiency of livestock and poultry. Coccidiosis is an intestinal infection caused by intracellular protozoan parasites belonging to several different species of Eimeria. Infection with coccidia parasites seriously impairs the growth and feed utilization of chickens and costs the US poultry industry more than $1.5 billion in annual losses. Although acquired immunity to Eimeria develops following natural infection, due to the complex life cycle and intricate host immune response to Eimeria, vaccine development has been difficult and a better understanding of the basic immunobiology of pertinent host-parasite interactions is necessary for developing effective immunological control strategies against coccidiosis. Chickens infected with Eimeria produce parasite specific antibodies in both the circulation and mucosal secretions but humoral immunity plays only a minor role in protection against this disease. Rather, recent evidence implicates cell-mediated immunity as the major factor conferring resistance to coccidiosis. This review will summarize current understanding of the avian intestinal immune system and its response to Eimeria as well as provide a conceptual overview of the complex molecular and cellular events involved in intestinal immunity to coccidiosis. It is anticipated that increased knowledge of the interaction between parasites and host immunity will stimulate the birth of novel immunological and molecular biological concepts in the control of intestinal parasitism.
Collapse
Affiliation(s)
- C H Yun
- USDA, Agricultural Research Service, Immunology and Disease Resistance Laboratory, Livestock and Poultry Sciences Institute, Beltsville, MD 20705, USA
| | | | | |
Collapse
|
30
|
Abstract
The intestine is the largest lymphoid organ in the body by virtue of lymphocyte numbers and quantity of immunoglobulin produced. This is largely related to the enormous antigen load to which these cells are exposed on a daily basis. However, despite this, the mucosa-associated lymphoid tissue appears to be regulated by unique mechanisms, and this is reflected in specific phenomena (oral tolerance, controlled or physiologic inflammation) as well as unusual lymphoid populations (intraepithelial lymphocytes) that respond to alternative pathways of activation. This, coupled with the existence of novel antigen-presenting cells (intestinal epithelial cells) sets the scene for distinct immune responses. It is these distinct regulatory factors that support immunosuppression or tolerance rather than active immunity at a site juxtaposed to the external environment. This review defines these novel interactions and suggests how alteration in normal function may result in allergic or inflammatory responses. A clearer understanding of mucosal immunoregulation may lead to new therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- L Mayer
- Division of Clinical Immunology, Mt. Sinai Medical Center, New York, New York 10029-6574, USA
| |
Collapse
|
31
|
Affiliation(s)
- L Mayer
- Mount Sinai Medical Center, 1425 Madison Avenue, New York, New York 10029, USA.
| |
Collapse
|
32
|
Campbell N, Yio XY, So LP, Li Y, Mayer L. The intestinal epithelial cell: processing and presentation of antigen to the mucosal immune system. Immunol Rev 1999; 172:315-24. [PMID: 10631956 DOI: 10.1111/j.1600-065x.1999.tb01375.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The immunologic tone of the intestinal tract is one of suppressed or highly regulated responses. While there are several components (intrinsic and extrinsic to the gut-associated lymphoid tissue) responsible for this immunologically suppressed tone, the intestinal epithelial call (IEC) has been proposed as a key player in this process. IECs can take up and process antigen but distinct surface molecules and restriction elements allow them to present these antigens to unique regulatory T cells. These include the expression of the class Ib molecule CD1d as well as a novel CD8 ligand, gp180. These molecules come together to activate a subpopulation of CD8+ regulatory cells whose function is to suppress immune responses in an antigen non-specific fashion most likely through cognate interactions. This form of regulation may be unique to the gut-associated lymphoid tissue which is consistent with the unusual demands upon this part of the immune system.
Collapse
Affiliation(s)
- N Campbell
- Mount Sinai Medical Center, Immunobiology Center, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
33
|
Colgan SP, Hershberg RM, Furuta GT, Blumberg RS. Ligation of intestinal epithelial CD1d induces bioactive IL-10: critical role of the cytoplasmic tail in autocrine signaling. Proc Natl Acad Sci U S A 1999; 96:13938-43. [PMID: 10570177 PMCID: PMC24169 DOI: 10.1073/pnas.96.24.13938] [Citation(s) in RCA: 136] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/1999] [Indexed: 11/18/2022] Open
Abstract
The intestinal epithelium is anatomically positioned to serve as the critical interface between the lumen and the mucosal immune system. In addition to MHC class I and II antigens, intestinal epithelia constitutively express the nonclassical MHC molecule CD1d, a transmembrane molecule with a short cytoplasmic tail expressed as a beta(2)-microglobulin-associated 48-kDa glycoprotein and novel beta(2)-microglobulin-independent 37-kDa nonglycosylated protein on intestinal epithelia. At present, it is not known whether extracellular ligands can signal intestinal epithelial CD1d. To define signaling of CD1d cytoplasmic tail, retrovirus-mediated gene transfer was used to generate stable cell lines expressing wild-type CD1d or a chimeric molecule (extracellular CD1d and cytoplasmic CD1a), and surface CD1d was triggered by antibody crosslinking. Although wild-type CD1d was readily activated (tyrosine phosphorylation), no demonstrable signal was evident in cell lines expressing the chimeric molecule. Subsequent studies revealed that anti-CD1d crosslinking specifically induces epithelial IL-10 mRNA and protein and is blocked by the tyrosine kinase inhibitor genistein. Further studies addressing epithelial-derived IL-10 revealed that anti-CD1d crosslinking attenuates IFN-gamma signaling and that such attenuation is reversed by addition of functionally inhibitory IL-10 antibodies. These results define signaling through surface CD1d, and, importantly, they demonstrate that this pathway may serve to dampen epithelial proinflammatory signals.
Collapse
Affiliation(s)
- S P Colgan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
34
|
Devine L, Kavathas PB. Molecular analysis of protein interactions mediating the function of the cell surface protein CD8. Immunol Res 1999; 19:201-10. [PMID: 10493174 DOI: 10.1007/bf02786488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The T cell coreceptor CD8 is a cell-surface glycoprotein expressed either as a disulfide-linked homodimer of two CD8alpha monomers, or a heterodimer of CD8alpha and CD8beta. These receptors interact with ligands, such as major histocompatibility complex (MHC) class I, on the outside of the cell, with proteins inside the cell, such as the tyrosine kinase p56lck, and possibly with proteins on the same cell-surface. The molecular details describing such protein interactions can shed light on how the proteins function and the functional differences between the two forms of CD8. Crystal structures, mutational analysis, affinity measurements, and other approaches are providing those details.
Collapse
Affiliation(s)
- L Devine
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520-8035, USA
| | | |
Collapse
|
35
|
Campbell NA, Kim HS, Blumberg RS, Mayer L. The nonclassical class I molecule CD1d associates with the novel CD8 ligand gp180 on intestinal epithelial cells. J Biol Chem 1999; 274:26259-65. [PMID: 10473580 DOI: 10.1074/jbc.274.37.26259] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies have shown that normal intestinal epithelial cells (IECs) are able to selectively activate CD8(+) T cells with suppressor activity, inducing proliferation associated with the activation of both the CD8-associated kinase p56(lck) and the T cell receptor (TCR)-associated kinase p59(fyn). This process appears to relate in part to a 180-kDa IEC surface glycoprotein, gp180, which binds to CD8 and activates CD8-associated p56(lck). However, purified gp180 alone is unable to induce T cell proliferation and does not activate p59(fyn). Because the class Ib molecule CD1d is expressed by IECs and monoclonal antibodies (mAbs) against CD1d inhibit IEC-induced proliferation of CD8(+) T cells, co-immunoprecipitation and enzyme-linked immunosorbent assay studies were performed, which demonstrated an association of gp180 and CD1d on the IEC surface. Interestingly, the activation of p59(fyn) in IEC-T cell co-cultures was blocked by the anti-CD1d mAb D5 but not by the anti-gp180 mAb B9. Conversely, treatment of IECs with mAb B9 inhibited IEC-induced activation of p56(lck) but not p59(fyn). More directly, a human CD1d cDNA (FO-1 D5) transfectant was able to activate p59(fyn) but not p56(lck). These data suggest that the CD1d-gp180 complex on the surface of IECs can be recognized by the TCR-CD8 co-receptor, resulting in the activation of CD8(+) T cells.
Collapse
Affiliation(s)
- N A Campbell
- Division of Clinical Immunology, Mount Sinai Medical Center, New York, New York 10029, USA.
| | | | | | | |
Collapse
|
36
|
|
37
|
Morales VM, Christ A, Watt SM, Kim HS, Johnson KW, Utku N, Texieira AM, Mizoguchi A, Mizoguchi E, Russell GJ, Russell SE, Bhan AK, Freeman GJ, Blumberg RS. Regulation of Human Intestinal Intraepithelial Lymphocyte Cytolytic Function by Biliary Glycoprotein (CD66a). THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.3.1363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Human small intestinal intraepithelial lymphocytes (iIEL) are a unique population of CD8αβ+ TCR-αβ+ but CD28− T lymphocytes that may function in intestinal epithelial cell immunosurveillance. In an attempt to define novel cell surface molecules involved in iIEL function, we raised several mAbs against activated iIELs derived from the small intestine that recognized an Ag on activated, but not resting, iIELs. Using expression cloning and binding studies with Fc fusion proteins and transfectants, the cognate Ag of these mAbs was identified as the N domain of biliary glycoprotein (CD66a), a carcinoembryonic Ag-related molecule that contains an immune receptor tyrosine-based inhibitory motif. Functionally, these mAbs inhibited the anti-CD3-directed and lymphokine-activated killer activity of the P815 cell line by iIELs derived from the human small intestine. These studies indicate that the expression of biliary glycoprotein on activated human iIELs and, potentially, other mucosal T lymphocytes is involved in the down-regulation of cytolytic function.
Collapse
Affiliation(s)
- Victor M. Morales
- *Gastroenterology Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Andreas Christ
- *Gastroenterology Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Suzanne M. Watt
- †Medical Research Council, Molecular Haematology Unit, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hyun S. Kim
- *Gastroenterology Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Kevin W. Johnson
- *Gastroenterology Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Nalan Utku
- ‡Institut Fuer Medizinische Immunologie, Charité-Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ana M. Texieira
- §Imperial Cancer Research Fund-Medical Oncology Unit, St. Bartholomew’s Hospital Medical College, London, United Kingdom
| | - Atsushi Mizoguchi
- ¶Immunopathology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Emiko Mizoguchi
- ¶Immunopathology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Gary J. Russell
- ∥Combined Program in Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital and Harvard Medical School, and
| | - Sara E. Russell
- *Gastroenterology Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Atul K. Bhan
- ¶Immunopathology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Gordon J. Freeman
- #Department of Adult Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115
| | - Richard S. Blumberg
- *Gastroenterology Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
38
|
Abstract
Up to 8% of children less than 3 years of age and approximately 2% of the adult population experience food-induced allergic disorders. A limited number of foods are responsible for the vast majority of food-induced allergic reactions: milk, egg, peanuts, fish, and tree nuts in children and peanuts, tree nuts, fish, and shellfish in adults. Food-induced allergic reactions are responsible for a variety of symptoms involving the skin, gastrointestinal tract, and respiratory tract and may be caused by IgE-mediated and non-IgE-mediated mechanisms. In part 1 of this series, immunopathogenic mechanisms and clinical disorders of food allergy are described.
Collapse
Affiliation(s)
- H A Sampson
- Jaffe Food Allergy Institute, The Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| |
Collapse
|
39
|
Abstract
Advances have been made in understanding specialized aspects of acquired mucosal immunity and the importance of cross-talk between host innate and acquired immune responses in host mucosal defense. These advances include elucidation of a molecular basis for the coordinated trafficking and retention of lymphocytes in intestinal sites, new insights into the possible extrathymic origin of mucosal T cells in the intestine, and the description of ligands that may be the targets of intraepithelial T-cell recognition. The past year has witnessed the characterization of an array of regulated epithelial cell mediators and responses important for host mucosal defense against enteric microbial pathogens and firmly establishes epithelial cells as an integral component of the mucosal immune network. Animal models continued to further understanding of intestinal inflammatory disease, and studies on tissue transglutaminase have generated significant new insights into the immunopathogenesis of human celiac disease.
Collapse
Affiliation(s)
- M B Dwinell
- Department of Medicine (0623D), Laboratory of Mucosal Immunology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0623, USA
| | | |
Collapse
|
40
|
Mayer L. Current concepts in mucosal immunity. I. Antigen presentation in the intestine: new rules and regulations. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:G7-9. [PMID: 9458767 DOI: 10.1152/ajpgi.1998.274.1.g7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The recognition that immune responses in the intestine differ from those seen systemically has led to a search for novel pathways involved in mucosal immunoregulation. One cell type that has surfaced as a prime candidate for such a regulatory role is the intestinal epithelial cell. A number of laboratories have documented that intestinal epithelial cells sample luminal antigens and process and present these to primed T cells. However, several unique features have emerged, making their potential role as antigen-presenting cells a critical part of mucosal homeostasis.
Collapse
Affiliation(s)
- L Mayer
- Immunobiology Center, Mount Sinai Medical Center, New York, New York 10029, USA
| |
Collapse
|
41
|
Toy LS, Yio XY, Lin A, Honig S, Mayer L. Defective expression of gp180, a novel CD8 ligand on intestinal epithelial cells, in inflammatory bowel disease. J Clin Invest 1997; 100:2062-71. [PMID: 9329971 PMCID: PMC508397 DOI: 10.1172/jci119739] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Previous studies support a role for intestinal epithelial cells (IEC) as antigen-presenting cells in mucosal immune responses. T cells activated by IEC are CD8+, suppressor in function, and dependent upon CD8-associated p56lck activation. A 180-kD glycoprotein (gp180) recognized by mAbs B9 and L12 has been identified and shown to be important in CD8+ T cell activation by IEC. Since IEC derived from patients with inflammatory bowel disease (IBD) are incapable of activating CD8+ T cells, we asked whether this correlated with gp180 expression. While frozen sections of normal bowel revealed bright gp180 staining on all IEC, both inflamed and uninflamed ulcerative colitis (UC) specimens showed patchy staining. In Crohn's disease (CD), staining was faint to absent. Flow cytometry confirmed immunohistochemical data. The staining patterns correlated with the ability of IEC to activate CD8-associated p56lck. Normal IEC induced phosphorylation of p56lck in CD8alpha but not CD4+ transfectants. In contrast, both UC and CD IEC activated CD4 and, to a much lesser extent, CD8-associated p56lck. Thus, gp180 expression by IBD IEC appears to be altered, and correlates with a functional alteration of lck activation. This defect may reflect a more proximal event in the pathogenesis of IBD.
Collapse
Affiliation(s)
- L S Toy
- Division of Clinical Immunology, Mount Sinai Medical Center, New York 10029, USA
| | | | | | | | | |
Collapse
|
42
|
Affiliation(s)
- L Mayer
- Immunobiology Center, Mount Sinai Medical Center, New York, NY 10029, USA
| |
Collapse
|
43
|
Mayer L. Regulation of mucosal immune responses: distinct antigens and antigen presenting cells. J Clin Immunol 1997; 17:349-53. [PMID: 9327333 DOI: 10.1023/a:1027382805727] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- L Mayer
- Mount Sinai School of Medicine, Division of Clinical Immunology, New York, New York 10029, USA
| |
Collapse
|
44
|
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
|