451
|
Abstract
The adaptive immune response in multiple sclerosis (MS) targets various myelin proteins and even some inducible heat shock proteins. A few attempts have been made to tolerize relapsing-remitting patients with MS to either full-length myelin basic protein or to a key peptide epitope between residues 83-99. These trials have demonstrated that this approach may potentially provide benefit to patients with relapsing- remitting MS. However, manipulation of responses to myelin proteins can have deleterious effects. The immune response to myelin components is positioned at a key tipping point in the pathophysiology of the disease. Clarification of the key target antigens in MS, and better understanding of practical methods to attain tolerance to a wide variety of myelin and neuronal molecules will provide the basis for the ultimately successful antigen specific therapy.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
452
|
Nussenblatt RB, Ferris F. Age-related macular degeneration and the immune response: implications for therapy. Am J Ophthalmol 2007; 144:618-26. [PMID: 17698021 PMCID: PMC2744410 DOI: 10.1016/j.ajo.2007.06.025] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 06/10/2007] [Accepted: 06/12/2007] [Indexed: 01/26/2023]
Abstract
PURPOSE To review the available information concerning the immune mediation of age-related macular degeneration (AMD) and to speculate on proposed mechanisms and immunotherapy. DESIGN Interpretative essay. METHODS Literature review and interpretation. RESULTS An ever-growing body of evidence is gathering concerning the role of the immune system in AMD. Evidence to date suggests that the underlying mechanism leading to AMD is the decline of the ocular downregulatory immune environment. The subsequent activation of the immune system would lead to T-cell sensitization. When combined with local antiangiogenic therapy, several existing immunotherapies may be used to downregulate the immune response, potentially leading to a more efficient inhibition of choroidal neovascularization. CONCLUSIONS The loss of the downregulatory immune environment is central to the development of AMD, permitting activation of the immune system. If so, immunotherapy could positively alter the course of the disease.
Collapse
Affiliation(s)
- Robert B Nussenblatt
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
453
|
Orsini B, Vivas JR, Ottanelli B, Amedei A, Surrenti E, Galli A, Milani S, Pinzani P, Del Prete G, Surrenti C, Baldari CT, Touati E, D' Elios MM. Human gastric epithelium produces IL-4 and IL-4delta2 isoform only upon Helicobacter pylori infection. Int J Immunopathol Pharmacol 2007; 20:809-818. [PMID: 18179754 DOI: 10.1177/039463200702000417] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Recent evidence suggests that interleukin-4 (IL-4) is related to mucosal tolerance by which an injurious immune response is prevented, suppressed or shifted to a non-injurious response. We investigated the expression of IL-4 and its splice variant isoform IL-4delta2 in gastric epithelial cells of healthy subjects and gastritis patients infected with Helicobacter pylori (H. pylori) with or without the cag pathogenicity island (cag-PAI). IL-4 and IL-4delta2 mRNAs were evaluated in microdissected gastric epithelium and in AGS cell lines co-cultured with H. pylori B128 or SS1 strains. IL-4 mRNA was consistently detected in microdissected gastric epithelial cells from healthy subjects. The IL-4 mRNA expression was low in H. pylori?infected patients, and markedly reduced in cag-PAI-positive ones. IL-4delta2 mRNA was expressed on gastric epithelium of H. pylori-infected patients, but not in healthy subjects. The IL-4delta2 expression was lower in cag-PAI-positive than in cag-PAI-negative H. pylori infected patients. AGS cells also produced IL-4 mRNA upon SS1 strain stimulation, whereas IL-4delta2 mRNA expression was detected in AGS co-cultured with either SS1 or B128 strains. An inverse correlation was documented between IL-4 and IL-4delta2 mRNA expression by microdissected gastric epithelial cells and the score of gastritis. IL-4, but not IL-4delta2, is expressed by gastric epithelium of healthy subjects, whereas IL-4delta2 and lesser IL-4 mRNA are detectable in the gastric epithelium of H. pylori-infected patients. Data suggest that gastric epithelial cells might regulate the balance between tolerance and immune response by the fine tuning of IL-4 and IL-4delta2 expression.
Collapse
Affiliation(s)
- B Orsini
- Department of Clinical Pathophysiology, Gastroenterology Unit, University of Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
454
|
Lu LF, Gavin MA, Rasmussen JP, Rudensky AY. G protein-coupled receptor 83 is dispensable for the development and function of regulatory T cells. Mol Cell Biol 2007; 27:8065-72. [PMID: 17893329 PMCID: PMC2169173 DOI: 10.1128/mcb.01075-07] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Global analyses of gene expression in regulatory T (Treg) cells, whose development is critically dependent upon the transcription factor Foxp3, have provided many clues as to the molecular mechanisms these cells employ to control immune responses and establish immune tolerance. Through these studies, G protein-coupled receptor 83 (GPR83) was found to be expressed at high levels in Treg-cell populations. However, its function remained unclear. Recently, it has been suggested that GPR83 is involved in the induction of Foxp3 expression in the peripheral nonregulatory Foxp3- CD4 T cells. To examine a role for GPR83 in Treg-cell biology, we generated and characterized GPR83-deficient mice. We have shown that GPR83 abolition does not result in measurable pathology or changes in the numbers or function of Foxp3+ Treg cells. Furthermore, while in vitro analysis suggested a potential involvement of GPR83 in transforming growth factor beta-dependent Foxp3 induction, there was no difference in the ability of nonregulatory GPR83-deficient and nondeficient Foxp3- T cells to acquire Foxp3 expression in vivo. Collectively, our results demonstrate that GPR83 is dispensable for Treg-cell development and function.
Collapse
Affiliation(s)
- Li-Fan Lu
- Department of Immunology, University of Washington, Seattle, WA 98195-7650, USA
| | | | | | | |
Collapse
|
455
|
van der Marel APJ, Samsom JN, Greuter M, van Berkel LA, O'Toole T, Kraal G, Mebius RE. Blockade of IDO inhibits nasal tolerance induction. THE JOURNAL OF IMMUNOLOGY 2007; 179:894-900. [PMID: 17617580 DOI: 10.4049/jimmunol.179.2.894] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The amino acid tryptophan is essential for the proliferation and survival of cells. Modulation of tryptophan metabolism has been described as an important regulatory mechanism for the control of immune responses. The enzyme IDO degrades the indole moiety of tryptophan, not only depleting tryptophan but also producing immunomodulatory metabolites called kynurenines, which have apoptosis-inducing capabilities. In this study, we show that IDO is more highly expressed in nonplasmacytoid dendritic cells of the nose draining lymph nodes (LNs), which form a unique environment to induce tolerance to inhaled Ags, when compared with other peripheral LNs. Upon blockade of IDO during intranasal OVA administration, Ag-specific immune tolerance was abrogated. Analysis of Ag-specific T cells in the LNs revealed that inhibition of IDO resulted in enhanced survival at 48 h after antigenic stimulation, although this result was not mediated through alterations in apoptosis or cell proliferation. Furthermore, no differences were found in CD4(+) T cells expressing FoxP3. Our data suggest that the level of IDO expression in dendritic cells, present in nose draining LNs, allows for the generation of a sufficient number of regulatory T cells to control and balance effector T cells in such a way that immune tolerance is induced, whereas upon IDO blockade, effector T cells will outnumber regulatory T cells, leading to immunity.
Collapse
Affiliation(s)
- Arnold P J van der Marel
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
456
|
Abstract
Regulatory T (T(reg)) cells show promise for treating autoimmune diseases, but their induction to elevated potency has been problematic when the most optimally derived cells are from diseased animals. To circumvent reliance on auto-antigen reactive T(reg) cells, stimulation to vaccine antigens (Ags) may offer a viable alternative while maintaining potency to protect against proinflammatory diseases. Our Salmonella vaccine expressing colonization factor Ag I (CFA/I) possesses anti-inflammatory properties, evident by elevated Th2 cell responses, reduced inflammatory cell infiltrates in the Peyer's patches, and an absence of proinflammatory cytokine production by infected macrophages. Given these findings, we hypothesized whether this vaccine would be protective against experimental autoimmune encephalomyelitis (EAE). As such, Salmonella-CFA/I protected in both prophylactic and therapeutic paradigms against proteolipid protein (PLP(139-151))-mediated EAE in SJL mice. The protected mice showed significantly reduced clinical disease and subsequent resolution when compared to PBS-treated controls. Histopathological studies showed reduced demyelination and no inflammation of spinal cords when compared to PBS- or Salmonella vector-treated mice. To ascertain whether the observed immune deviation was in part supported by T(reg) cells, analysis revealed involvement of FoxP3(+) CD25(+) CD4(+) T cells. Adoptive transfer of induced TGF-beta (+) T(reg) cells from vaccinated mice showed complete protection against PLP(139-151) challenge, but not by naive T(reg) cells. Partial protection to EAE was also achieved by the adoptive transfer of CD25(-) CD4(+) T cells, suggesting that Th2 cells also contributed. Thus, these data show that T(reg) cells are induced by oral vaccination with Salmonella-CFA/I contributing to the efficacious treatment of autoimmune disease.
Collapse
Affiliation(s)
- D W Pascual
- Veterinary Molecular Biology, Montana State University, Bozeman, Montana 59717-3610, USA.
| | | | | | | |
Collapse
|
457
|
Singh RP, La Cava A, Wong M, Ebling F, Hahn BH. CD8+ T cell-mediated suppression of autoimmunity in a murine lupus model of peptide-induced immune tolerance depends on Foxp3 expression. THE JOURNAL OF IMMUNOLOGY 2007; 178:7649-57. [PMID: 17548601 DOI: 10.4049/jimmunol.178.12.7649] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Systemic lupus erythematosus is an autoimmune disease caused by autoantibodies, including IgG anti-DNA. New Zealand Black/New Zealand White F(1) female mice, a model of spontaneous polygenic systemic lupus erythematosus, tolerized with an artificial peptide (pConsensus) based on anti-DNA IgG sequences containing MHC class I and class II T cell determinants, develop regulatory CD4+CD25+ T cells and CD8+ inhibitory T cells (CD8+ Ti), both of which suppress autoantibody production. CD8+ Ti inhibit primarily via secretion of TGF-beta. In the present study, we show that the inhibitory function of CD8+ T cells from tolerized mice is sustained for up to 8 wk and at all times depends on expression of Foxp3. Both CD28-positive and CD28-negative CD8+ T cells contain inhibitory cells, but the expression of mRNA for Foxp3 and for TGF-beta is higher and lasts longer in the CD28- subset. In vitro addition of TGF-beta (in the presence of IL-2) induces Foxp3 expression in a dose-response manner. Gene inhibition or blockade with small interfering RNA of Foxp3 abrogates the ability of the CD8+ Ti to inhibit anti-DNA production and the proliferation of CD4+ Th cells. Moreover, a significant correlation between expression of Foxp3 and ability of CD8+ Ti to secrete TGF-beta is observed. Therefore, CD8+ Ti in this system of tolerance are similar to CD4+CD25+ regulatory T cells in their dependence on expression of Foxp3, and there may be a bidirectional Foxp3/TGF-beta autocrine loop that determines the ability of the CD8+ T cells to control autoimmunity.
Collapse
Affiliation(s)
- Ram Pyare Singh
- Division of Rheumatology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, 1001 Veteran Avenue, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
458
|
Ishikawa H, Ochi H, Chen ML, Frenkel D, Maron R, Weiner HL. Inhibition of autoimmune diabetes by oral administration of anti-CD3 monoclonal antibody. Diabetes 2007; 56:2103-9. [PMID: 17456848 DOI: 10.2337/db06-1632] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anti-CD3 monoclonal antibody (mAb) has been shown to induce tolerance and to be an effective treatment for diabetes both in animal models and in human trials. We have shown that anti-CD3 mAb given orally is biologically active in the gut and suppresses experimental autoimmune encephalitis by the induction of a regulatory T-cell that expresses latency-associated peptide (LAP) on its surface. In the present study, we investigated the effect of oral anti-CD3 mAb on the prevention of autoimmune diabetes in AKR mice in which the low-dose streptozocin (STZ) model induces autoimmunity to the beta-cells of the islets. We found that oral anti-CD3 mAb given at doses of 50 and 250 microg/feeding suppressed the incidence of diabetes in this model with the best effects seen at the 50 microg/dose. Associated with suppression, we observed decreased cell proliferation in the spleen and conversion of T-helper (Th)1 responses into Th2/Th3 responses in the periphery, including the pancreatic lymph nodes. Oral anti-CD3 mAb increased the expression of LAP on CD4(+) T-cells, and these cells could adoptively transfer protection. Protection by oral anti-CD3 was transforming growth factor-beta dependent. Our results demonstrate that oral anti-CD3 is effective in the model of STZ-induced diabetes and may be a useful form of therapy for type 1 diabetes in humans.
Collapse
MESH Headings
- Administration, Oral
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- CD3 Complex/immunology
- CD4 Antigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytokines/biosynthesis
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Immunotherapy
- Insulin/metabolism
- Intestinal Absorption
- Intestines/drug effects
- Intestines/immunology
- Male
- Mice
- Mice, Inbred AKR
- Pancreas/metabolism
- Spleen/drug effects
- Spleen/metabolism
- Streptozocin/pharmacology
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Hiroki Ishikawa
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
459
|
Huibregtse IL, Snoeck V, de Creus A, Braat H, De Jong EC, Van Deventer SJH, Rottiers P. Induction of ovalbumin-specific tolerance by oral administration of Lactococcus lactis secreting ovalbumin. Gastroenterology 2007; 133:517-28. [PMID: 17681173 DOI: 10.1053/j.gastro.2007.04.073] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Accepted: 04/19/2007] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Obtaining antigen-specific immune suppression is an important goal in developing treatments of autoimmune, inflammatory, and allergic gastrointestinal diseases. Oral tolerance is a powerful means for inducing tolerance to a particular antigen, but implementing this strategy in humans has been difficult. Active delivery of recombinant autoantigens or allergens at the intestinal mucosa by genetically modified Lactococcus lactis (L lactis) provides a novel therapeutic approach for inducing tolerance. METHODS We engineered the food grade bacterium L lactis to secrete ovalbumin (OVA) and evaluated its ability to induce OVA-specific tolerance in OVA T-cell receptor (TCR) transgenic mice (DO11.10). Tolerance induction was assessed by analysis of delayed-type hypersensitivity responses, measurement of cytokines and OVA-specific proliferation, phenotypic analysis, and adoptive transfer experiments. RESULTS Intragastric administration of OVA-secreting L lactis led to active delivery of OVA at the mucosa and suppression of local and systemic OVA-specific T-cell responses in DO11.10 mice. This suppression was mediated by induction of CD4(+)CD25(-) regulatory T cells that function through a transforming growth factor beta-dependent mechanism. Restimulation of splenocytes and gut-associated lymph node tissue from these mice resulted in a significant OVA-specific decrease in interferon gamma and a significant increase in interleukin-10 production. Furthermore, Foxp3 and CTLA-4 were significantly up-regulated in the CD4(+)CD25(-) population. CONCLUSIONS Mucosal antigen delivery by oral administration of genetically engineered L lactis leads to antigen-specific tolerance. This approach can be used to develop effective therapeutics for systemic and intestinal immune-mediated inflammatory diseases.
Collapse
MESH Headings
- Administration, Oral
- Adoptive Transfer
- Animals
- Antigen-Presenting Cells/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation/metabolism
- CTLA-4 Antigen
- Cell Proliferation
- Dose-Response Relationship, Immunologic
- Female
- Forkhead Transcription Factors/metabolism
- Hypersensitivity, Delayed/immunology
- Hypersensitivity, Delayed/metabolism
- Immune Tolerance
- Immunity, Mucosal
- Interferon-gamma/metabolism
- Interleukin-10/metabolism
- Interleukin-2 Receptor alpha Subunit/analysis
- Intestinal Mucosa/metabolism
- Intestines/cytology
- Intestines/immunology
- Lactococcus lactis/genetics
- Lactococcus lactis/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Ovalbumin/biosynthesis
- Ovalbumin/genetics
- Ovalbumin/immunology
- Peyer's Patches/cytology
- Peyer's Patches/immunology
- Peyer's Patches/metabolism
- Probiotics/administration & dosage
- Probiotics/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Proteins/immunology
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Inge L Huibregtse
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
460
|
MESH Headings
- Administration, Oral
- Adoptive Transfer
- Animals
- Antigen-Presenting Cells/immunology
- Cell Proliferation
- Hypersensitivity, Delayed/immunology
- Hypersensitivity, Delayed/metabolism
- Immune Tolerance
- Immunity, Mucosal
- Interferon-gamma/metabolism
- Interleukin-10/metabolism
- Intestinal Mucosa/metabolism
- Intestines/cytology
- Intestines/immunology
- Lactococcus lactis/genetics
- Lactococcus lactis/metabolism
- Mice
- Ovalbumin/biosynthesis
- Ovalbumin/genetics
- Ovalbumin/immunology
- Probiotics/administration & dosage
- Probiotics/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Proteins/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transforming Growth Factor beta/metabolism
Collapse
|
461
|
|
462
|
Kurashima Y, Kunisawa J, Higuchi M, Gohda M, Ishikawa I, Takayama N, Shimizu M, Kiyono H. Sphingosine 1-Phosphate-Mediated Trafficking of Pathogenic Th2 and Mast Cells for the Control of Food Allergy. THE JOURNAL OF IMMUNOLOGY 2007; 179:1577-85. [PMID: 17641024 DOI: 10.4049/jimmunol.179.3.1577] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Sphingosine 1-phosphate (S1P) has been proposed as a regulator of lymphocyte trafficking, but its role in mucosa-associated diseases, such as in food allergies, remains to be elucidated. To examine the role of S1P in allergic diseases in the intestine, we used a Th2 cell-mediated Ag-specific allergic diarrhea model and demonstrated that type 1 S1P receptor (S1P(1)) expression was preferentially associated with pathogenic CD4(+) T cells for the development of allergic reactions. Consistent with this demonstration, treatment with FTY720, a modulator of the S1P(1), prevented allergic diarrhea by inhibiting the migration of systemically primed pathogenic CD4(+) T cells induced by oral challenge with allergen into the large intestine. In addition, FTY720 hampered mast cell infiltration into the large intestine, whereas eosinophil infiltration into the large intestine and total and allergen-specific serum IgE production were comparable between mock- and FTY720-treated groups. These results suggest that modulation of the S1P-mediated pathway to inhibit the migration of pathogenic CD4(+) T cells and mast cells into the large intestine could be a novel strategy for preventing allergic diarrhea.
Collapse
Affiliation(s)
- Yosuke Kurashima
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
463
|
Rubtsov YP, Rudensky AY. TGFbeta signalling in control of T-cell-mediated self-reactivity. Nat Rev Immunol 2007; 7:443-53. [PMID: 17525753 DOI: 10.1038/nri2095] [Citation(s) in RCA: 250] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the immune system, transforming growth factor-beta (TGFbeta) affects multiple cell lineages by either promoting or opposing their differentiation, survival and proliferation. Understanding the cellular mechanisms of TGFbeta-mediated regulation is complicated due to a broad distribution of TGFbeta receptors on the surface of different immune-cell types. Recent studies using in vivo genetic approaches revealed a critical role for TGFbeta signalling in T cells in restraining fatal autoimmune lesions. Here, we review recent advances in our understanding of a role for TGFbeta signalling in the regulation of T-cell differentiation in the thymus and in the periphery, with a particular emphasis on TGFbeta-mediated control of self-reactive T cells.
Collapse
Affiliation(s)
- Yuri P Rubtsov
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | | |
Collapse
|
464
|
Gryglewski A, Majcher P, Bryniarski K, Konturek S, Ptak M, Ptak W, Szczepanik M. Mesenteric lymph node Tgammadelta cells induced by gastrectomy in mice suppress cell-mediated immune response in vitro via released TGF-beta. J Surg Res 2007; 142:66-71. [PMID: 17612560 DOI: 10.1016/j.jss.2006.10.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 10/16/2006] [Accepted: 10/31/2006] [Indexed: 01/22/2023]
Abstract
We have shown previously that gastrectomy, but not laparotomy alone, severely impairs contact sensitivity responses in vivo and selectively alters cell trafficking in gut associated lymphatic tissue. Here, we investigate the immunological role of different subpopulations of mesenteric lymph node cells (MLNCs) in the inhibition of contact sensitivity as well as their suppressive mechanisms. Suppressive cells were isolated from the mesenteric lymph nodes of gastrectomized mice and were added to cultures of lymphocytes from mice immunized with trinitrophenyl-chloride. These MLNCs inhibited the proliferation of sensitized lymphocytes in response to antigen. Depletion experiments revealed that the suppressive MLNCs are Tgammadelta+ cells, but not Talphabeta+ cells. Neutralizing antibodies to IL-4, IL-10, and tissue growth factor-beta (TGF-beta) revealed that suppression was dependent on TGF-beta, but not the other cytokines. We conclude that surgical stress induced by gastrectomy causes accumulation of Tgammadelta+ lymphocytes in gut associated lymphatic tissue and that these cells suppress the cell-mediated response in vitro in an antigen-non specific manner via TGF-beta. This cytokine can possibly prevent in vivo the development of autoimmune responses following severe tissue trauma in the gastrointestinal tract.
Collapse
Affiliation(s)
- Andrzej Gryglewski
- Department of Anatomy, Jagiellonian University College of Medicine, Kraków, Poland
| | | | | | | | | | | | | |
Collapse
|
465
|
Ando T, Hatsushika K, Wako M, Ohba T, Koyama K, Ohnuma Y, Katoh R, Ogawa H, Okumura K, Luo J, Wyss-Coray T, Nakao A. Orally administered TGF-beta is biologically active in the intestinal mucosa and enhances oral tolerance. J Allergy Clin Immunol 2007; 120:916-23. [PMID: 17606291 DOI: 10.1016/j.jaci.2007.05.023] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2007] [Revised: 05/15/2007] [Accepted: 05/18/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Epidemiologic studies suggest that TGF-beta in breast milk provides protection against allergic disease during infancy. However, it is unclear whether orally administered TGF-beta, such as TGF-beta in human milk, retains and exerts its activity in the intestinal mucosa and can affect immune response (tolerance) to dietary antigens. OBJECTIVE We sought to determine whether orally administered TGF-beta is biologically active in intestinal mucosa and affects oral tolerance. METHODS Activity of orally administered TGF-beta in the intestinal mucosa was evaluated by means of in vivo imaging with transgenic mice expressing a Smad-responsive reporter construct (SBE-luc mice), by means of immunohistochemical staining with anti-phosphorylated Smad2 antibody, and by means of real-time RT-PCR analysis of TGF-beta and Smad7 mRNA expression. The effects of orally administered TGF-beta on oral tolerance induction were assessed in mice tolerized by means of high-dose ovalbumin (OVA) feeding. RESULTS The oral administration of TGF-beta increased Smad-responsive reporter activity in the intestines of SBE-luc mice and induced Smad2 phosphorylation and TGF-beta and Smad7 mRNA expression in the intestines of BALB/c mice. Serum TGF-beta levels were also increased after oral administration of TGF-beta. BALB/c mice treated orally with OVA and TGF-beta showed augmented reduction of OVA-specific IgE and IgG1 antibodies, T-cell reactivity, and immediate-type skin reactions when compared with the mice treated orally with OVA alone. CONCLUSIONS Orally administered TGF-beta retains sufficient biologic activity in intestinal mucosa and enhances oral tolerance. CLINICAL IMPLICATIONS Oral administration of TGF-beta might become a potential strategy to prevent allergic diseases, such as food allergy.
Collapse
Affiliation(s)
- Takashi Ando
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
466
|
Ruhlman T, Ahangari R, Devine A, Samsam M, Daniell H. Expression of cholera toxin B-proinsulin fusion protein in lettuce and tobacco chloroplasts--oral administration protects against development of insulitis in non-obese diabetic mice. PLANT BIOTECHNOLOGY JOURNAL 2007; 5:495-510. [PMID: 17490448 PMCID: PMC2590789 DOI: 10.1111/j.1467-7652.2007.00259.x] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Lettuce and tobacco chloroplast transgenic lines expressing the cholera toxin B subunit-human proinsulin (CTB-Pins) fusion protein were generated. CTB-Pins accumulated up to ~16% of total soluble protein (TSP) in tobacco and up to ~2.5% of TSP in lettuce. Eight milligrams of powdered tobacco leaf material expressing CTB-Pins or, as negative controls, CTB-green fluorescent protein (CTB-GFP) or interferon-GFP (IFN-GFP), or untransformed leaf, were administered orally, each week for 7 weeks, to 5-week-old female non-obese diabetic (NOD) mice. The pancreas of CTB-Pins-treated mice showed decreased infiltration of cells characteristic of lymphocytes (insulitis); insulin-producing beta-cells in the pancreatic islets of CTB-Pins-treated mice were significantly preserved, with lower blood or urine glucose levels, by contrast with the few beta-cells remaining in the pancreatic islets of the negative controls. Increased expression of immunosuppressive cytokines, such as interleukin-4 and interleukin-10 (IL-4 and IL-10), was observed in the pancreas of CTB-Pins-treated NOD mice. Serum levels of immunoglobulin G1 (IgG1), but not IgG2a, were elevated in CTB-Pins-treated mice. Taken together, T-helper 2 (Th2) lymphocyte-mediated oral tolerance is a likely mechanism for the prevention of pancreatic insulitis and the preservation of insulin-producing beta-cells. This is the first report of expression of a therapeutic protein in transgenic chloroplasts of an edible crop. Transplastomic lettuce plants expressing CTB-Pins grew normally and transgenes were maternally inherited in T(1) progeny. This opens up the possibility for the low-cost production and delivery of human therapeutic proteins, and a strategy for the treatment of various other autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Andrew Devine
- University of Central Florida, Department of Molecular Biology and Microbiology, Biomolecular Science, Building #20, Orlando, FL 32816-2364, USA
| | - Mohtahsem Samsam
- University of Central Florida, Department of Molecular Biology and Microbiology, Biomolecular Science, Building #20, Orlando, FL 32816-2364, USA
| | - Henry Daniell
- University of Central Florida, Department of Molecular Biology and Microbiology, Biomolecular Science, Building #20, Orlando, FL 32816-2364, USA
| |
Collapse
|
467
|
Sun X, Yamada H, Yoshihara K, Awaya A, Yoshikai Y. In vivo treatment with a nonapeptide thymic hormone, facteur thymique serique (FTS), ameliorates chronic colitis induced by dextran sulphate sodium in mice. Int Immunopharmacol 2007; 7:928-36. [PMID: 17499195 DOI: 10.1016/j.intimp.2007.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 02/26/2007] [Accepted: 02/27/2007] [Indexed: 01/12/2023]
Abstract
Facteur thymique serique (FTS), a thymic hormone with nonapeptide is involved in T cell differentiation in intestine. Here we investigated the effect of FTS on dextran sulphate sodium (DSS)-induced colitis. BALB/c mice were subcutaneously treated with 1 mug/mouse/day of FTS daily. FTS did not affect the course of acute colitis induced by DSS as assessed by survival rate, clinical activity of diseases, extent of tissue damage of colons. On the other hand, FTS significantly ameliorated chronic colitis induced by multiple cycles of DSS as reflected by lower lethality, weight loss, clinical scores and histological scores. The levels of interferon (IFN)-gamma, interleukin 1(IL-1)-beta, and IL-12p40 in the culture supernatants of lamina propria (LP) cells of colon without any stimulation and IFN-gamma by T cells in the LP T cells under T cell receptor (TCR) triggering were reduced in FTS-treated mice, whereas the levels of IL-10 by LP cells and LPT cells were higher in FTS-treated mice. Thus, FTS may serve to suppress inflammation in DSS-induced chronic colitis accompanied by increased IL-10 production.
Collapse
Affiliation(s)
- Xun Sun
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | | | | | | | |
Collapse
|
468
|
Abstract
The internal surfaces of the human body are covered by distinct types of epithelial cells and mucus-secreting cells. The mucosal surfaces serve many vital functions, such as respiration (nasal passage and lung), absorption (gastrointestinal tract), excretion (lung, urinary tract, large intestine), and reproduction (reproductive tract). In performing these functions, the host is inevitably exposed to environmental antigens, food particles, commensal flora, and pathogens. Mucosal surfaces contain specialized dendritic cells (DCs) capable of sensing these external stimuli and mounting appropriate local responses depending on the nature of the elements they encounter. In the absence of pathogens, mucosal DCs either ignore the antigen or induce regulatory responses. Upon recognition of microorganisms that invade the mucosal barrier, mucosal DCs mount robust protective immunity. This review highlights progress in our understanding of how mucosal DCs process external information and direct appropriate responses by mobilizing various cells of the innate and adaptive immune systems to achieve homeostasis and protection.
Collapse
Affiliation(s)
- Akiko Iwasaki
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| |
Collapse
|
469
|
Pabst O, Bernhardt G, Förster R. The impact of cell-bound antigen transport on mucosal tolerance induction. J Leukoc Biol 2007; 82:795-800. [PMID: 17565048 DOI: 10.1189/jlb.0307144] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mucosal surfaces are exposed continuously to a flood of foreign antigens demanding a tightly controlled balance between immunity and tolerance induction. Tolerance toward food and inhaled antigens, known as oral and respiratory tolerance, respectively, evokes a body-wide nonresponsiveness against the plethora of environmental antigens. Key issues in understanding the induction of mucosal tolerance relate to the site of antigen entrance, the mechanisms of antigen transport, and the exact anatomical location where lymphocytes meet their cognate antigens. In this regard, opposing ideas have been put forward: In one scenario, antigens taken up at mucosal surfaces are considered to spread throughout the body, thus potentially evoking tolerogenic immune responses in all secondary lymphoid organs. Alternatively, tolerance induction might be confined to the draining regional lymph nodes (LN). Recent observations strongly supported the latter scenario, emphasizing the importance of regional LN and their network of afferent lymphatics in this process. In this model, air-borne and intestinal antigens are captured at mucosal sites by dendritic cells, which then migrate exclusively in a CCR7-dependent way to draining regional LN. Tolerance is then induced actively by the activation of antigen-specific T cells, which are subsequently deleted, become anergic, or alternatively, differentiate into regulatory T cells. Thus, the concept of local induction of immune responses seems to hold true for the majority of immune reactions, regardless of whether they are tolerogenic or defensive in their outcome.
Collapse
Affiliation(s)
- Oliver Pabst
- Institute of Immunology, Hannover Medical School, Carl-Neuberg Strasse 1, 30625 Hannover, Germany.
| | | | | |
Collapse
|
470
|
Abstract
PURPOSE OF REVIEW The aim of this article is to review the potential use of T regulatory cells in pathologic immune responses, focusing on their clinical application and the challenges associated with these therapies. RECENT FINDINGS Numerous T regulatory (TR) cell based therapies have been proposed to be clinically beneficial in patients with autoimmunity based on extensive studies in experimental models. Cell based therapies with CD4+CD25+Foxp3+ T regulatory cells isolated from peripheral blood hold promise, but difficulties exist in obtaining large enough numbers of these cells or expanding these cells in vitro. Generation of suppressive lymphocyte populations, such as cytokine secreting Tr1 and Th3 cells, is also promising. Therapies with Foxp3+ expressing lymphocytes generated from naïve CD4 lymphocytes in vitro is a novel mechanism of T regulatory cell generation, although questions regarding the role of these cells in vivo remain. Finally, therapies designed to restore the suppressive properties of T regulatory cells may be an alternative to cell-based therapies. SUMMARY T regulatory cells hold considerable promise in the treatment of autoimmunity. There are many important questions, however, regarding the biology of these cells that need to be addressed before their broad implementation in human disease.
Collapse
Affiliation(s)
- James W Verbsky
- Department of Pediatrics, Division of Rheumatology, Medical College of Wisconsin, Milwaukee, Wisconsin 53201-1997, USA.
| |
Collapse
|
471
|
Bliss SK, Bliss SP, Beiting DP, Alcaraz A, Appleton JA. IL-10 Regulates Movement of Intestinally Derived CD4+T Cells to the Liver. THE JOURNAL OF IMMUNOLOGY 2007; 178:7974-83. [PMID: 17548634 DOI: 10.4049/jimmunol.178.12.7974] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Diseases that affect the intestine may have hepatic manifestations, but the mechanisms involved in establishing hepatic disease secondarily remain poorly understood. We previously reported that IL-10 knockout (KO) mice developed severe necrotizing hepatitis following oral infection with Trichinella spiralis. In this study, we used this model of intestinal inflammation to further examine the role of IL-10 in regulating hepatic injury. Hepatic damage was induced by migrating newborn larvae. By delivering the parasite directly into the portal vein, we demonstrated that an ongoing intestinal immune response was necessary for the development of hepatitis. Intestinally derived CD4+ cells increased in the livers of IL-10 KO mice, and Ab-mediated blockade of MAdCAM-1 inhibited the accumulation of CD4+alpha(4)beta(7)+ cells in the liver. Moreover, adoptive transfer of intestinally primed CD4+ T cells from IL-10 KO mice caused hepatitis in infected immunodeficient animals. Conversely, transfer of wild-type donor cells reduced the severity of hepatic inflammation in IL-10 KO recipients, demonstrating regulatory activity. Our results revealed that IL-10 prevented migration of intestinal T cells to the liver and inhibited the development of hepatitis.
Collapse
Affiliation(s)
- Susan K Bliss
- James A. Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | | | | | | | |
Collapse
|
472
|
Seabrook TJ, Thomas K, Jiang L, Bloom J, Spooner E, Maier M, Bitan G, Lemere CA. Dendrimeric Aβ1–15 is an effective immunogen in wildtype and APP-tg mice. Neurobiol Aging 2007; 28:813-23. [PMID: 16725229 DOI: 10.1016/j.neurobiolaging.2006.04.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2006] [Revised: 04/03/2006] [Accepted: 04/09/2006] [Indexed: 11/24/2022]
Abstract
Immunization of humans and APP-tg mice with full-length beta-amyloid (Abeta) results in reduced cerebral Abeta levels. However, due to adverse events in the AN1792 trial, alternative vaccines are required. We investigated dendrimeric Abeta1-15 (dAbeta1-15), which is composed of 16 copies of Abeta1-15 peptide on a branched lysine core and thus, includes an Abeta-specific B cell epitope but lacks the reported T cell epitope. Immunization by subcutaneous, transcutaneous, and intranasal routes of B6D2F1 wildtype mice led to anti-Abeta antibody production. Antibody isotypes were mainly IgG1 for subcutaneous or transcutaneous immunization and IgG2b for intranasal immunization, suggestive of a Th2-biased response. All Abeta antibodies preferentially recognized an epitope in Abeta1-7. Intranasal immunization of J20 APP-tg mice resulted in a robust humoral immune response with a corresponding significant reduction in cerebral plaque burden. Splenocyte proliferation against Abeta peptide was minimal indicating the lack of an Abeta-specific cellular immune response. Anti-Abeta antibodies bound monomeric, oligomeric, and fibrillar Abeta. Our data suggest that dAbeta1-15 may be an effective and potentially safer immunogen for Alzheimer's disease (AD) vaccination.
Collapse
Affiliation(s)
- Timothy J Seabrook
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | | | | | | | | | | | | | | |
Collapse
|
473
|
Kang SM, Tang Q, Bluestone JA. CD4+CD25+ regulatory T cells in transplantation: progress, challenges and prospects. Am J Transplant 2007; 7:1457-63. [PMID: 17511675 DOI: 10.1111/j.1600-6143.2007.01829.x] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The involvement of CD4(+)CD25(+) regulatory T cells (Treg) in general immune homeostasis and protection from autoimmune syndromes is now well established. Similarly, there has been increasing evidence for Treg involvement in allograft rejection and current immunotherapies. However, despite significant advances in understanding the development, function, and therapeutic efficacy of Treg in certain well-defined rodent models, the relevance of Treg to clinical transplantation remains unclear. In this review, we summarize our current understanding of the role of Treg in immunity and organ transplantation in experimental and clinical settings. In addition, we review advances in using Treg as a form of immune therapy. The goal is to highlight the complexities and opportunities in the field and to provide evidence to support the use of antigen-specific Tregs in the context of transplantation to facilitate a robust and selective state of immune tolerance.
Collapse
Affiliation(s)
- S M Kang
- Transplantation Division, Department of Surgery, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
474
|
Abstract
The status of autoimmune disease therapies is not satisfactory. Antigen-specific immunotherapy has potential as a future therapy that could deliver maximal efficacy with minimal adverse effects. Several trials of antigen-specific immunotherapy have been performed, but so far no clear directions have been established. With regard to antigen-specificity in the immune system, T cells are essential components. However, at present, we do not have a sufficient range of strategies for manipulating antigen-specific T cells. In this review, the authors propose that T cell receptor gene transfer could be used for antigen-specific immunotherapy. In the proposed technique, important disease-related and, thus, antigen-specific T cells in patients would first be identified, and then a pair of cDNAs encoding alpha and beta T cell receptors would be isolated from these single T cells. These genes would then be transferred into self lymphocytes. These engineered antigen-specific cells can also be manipulated to express appropriate functional genes that could then be applied to specific immunotherapy.
Collapse
Affiliation(s)
- Kazuhiko Yamamoto
- University of Tokyo, Department of Allergy and Rheumatology, Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113, Japan.
| | | | | |
Collapse
|
475
|
da Silva MFS, Nóbrega A, Ribeiro RC, Levy MS, Ribeiro OG, Tambourgi DV, Sant'Anna DA, da Silva AC. Genetic selection for resistance or susceptibility to oral tolerance imparts correlation to both Immunoglobulin E level and mast cell number phenotypes with a profound impact on the atopic potential of the individual. Clin Exp Allergy 2007; 36:1399-407. [PMID: 17083350 DOI: 10.1111/j.1365-2222.2006.02588.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Immunological oral tolerance is being studied with great interest due to its therapeutic potential in allergy and autoimmunity processes, although the cellular and molecular mechanisms linking these different phenomena remain elusive. In the present study, two mouse lines with extreme phenotypes for susceptibility [TS Line] or resistance [TR Line] to oral tolerance and their [TS x TR]F2 segregants were used in order to evaluate the impact of these traits on the atopic potential of the individuals. OBJECTIVE Demonstrate whether the tr and ts genes, cumulated during 18 generations of bidirectional genetic selection, influence expression of two important immunobiological traits (IgE and mast cell) critical to allergic response. METHODS Mice with extreme phenotypes for oral tolerance to ovalbumin (OVA), produced by assortative mating (TS and TR Line), and their (TS x TR)F2 segregating were used. Serum IgE levels assayed by ELISA, and mastocytes counted with toluidine blue staining were evaluated in naïve mice. Anaphylaxis was induced by intravenous injection of OVA, intestinal inflammation by oral administration of OVA 7 days after immunization, and pulmonary inflammation by intranasal and nebulization OVA challenges. Specific IgE was dosed by passive cutaneous anaphylaxis. RESULTS The naïve TS mice have a 20-fold lower serum IgE level and two- to threefold diminished mast cell numbers in mucosal sites, when compared with TR-mice, which were highly susceptible to allergic inflammation and anaphylactic shock. The associations of oral tolerance, serum IgE levels and mast cell numbers in naïve animals were confirmed analysing the simultaneous presence of these traits in individuals of a [TS x TR]F2 -segregating population. CONCLUSION The results suggest that the complex of genes controlling TS and TR phenotypes play a main role in the regulation of the atopic potential of the individual. The studies of these traits in interline F2 segregants demonstrated a co-segregation of TS and TR phenotypes with IgE responsiveness and mast cell numbers. Thus, the opposite capacity of the genetically modified mice may be involved in co-adaptative mechanisms reflecting a dynamic relation between gene frequencies in a natural population. These correlations give circumstantial evidence to support clinical applications of oral tolerance in allergic and autoimmune diseases.
Collapse
Affiliation(s)
- M F S da Silva
- Departamento de Imunologia, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
476
|
White JML, Goon ATJ, Jowsey IR, Basketter DA, Mak RKH, Kimber I, McFadden JP. Oral tolerance to contact allergens: a common occurrence? A review. Contact Dermatitis 2007; 56:247-54. [PMID: 17441845 DOI: 10.1111/j.1600-0536.2006.01066.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Experimental and clinical oral tolerance to contact allergens has been reported sporadically, most notably in respect of nickel, and is generally assumed to be an uncommon phenomenon. There has recently been increased understanding of the immunological mechanisms inducing and maintaining oral tolerance. There are several contact allergens, including fragrance, antioxidant, and preservative chemicals, to which subjects are exposed through both cutaneous and oral routes. We examine the possibility that oral tolerance to contact allergens may be more common than previously thought. Animal models of oral tolerance to contact allergens indicate that cutaneous exposure to small, subsensitizing doses of contact allergens might negate any subsequent attempts to induce tolerance by oral administration. Extrapolating these observations to common human practises raises the possibility that application of contact allergens (fragrances, preservatives and antioxidants) in consumer products used by children could prevent or inhibit the later acquisition of specific tolerance resulting from 'natural' dietary exposure after weaning. Existing data on formaldehyde may conflict with this theory, though this could be explained by allergen specificity. We propose that further work in this area is needed.
Collapse
Affiliation(s)
- Jonathan M L White
- Department of Cutaneous Allergy, St. John's Institute of Dermatology, St. Thomas' Hospital, London SE1 7EH, UK.
| | | | | | | | | | | | | |
Collapse
|
477
|
Verbeek R, van der Mark K, Wawrousek EF, Plomp AC, van Noort JM. Tolerization of an established alphaB-crystallin-reactive T-cell response by intravenous antigen. Immunology 2007; 121:416-26. [PMID: 17386078 PMCID: PMC2265960 DOI: 10.1111/j.1365-2567.2007.02592.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Tolerance induction to prevent activation of a naïve T-cell repertoire has been well documented in rodents and can be readily achieved by intravenous, oral or intranasal administration of antigen in the absence of adjuvants. In autoimmune diseases such as multiple sclerosis (MS) the presence of an established memory/effector T-cell repertoire against self-antigens is likely to be more relevant than the potential reactivity of naive T cells. Methods to eliminate such an established T-cell response are less well understood. In this study, we explored the effectiveness of intravenous soluble antigen to eliminate a pre-existing T-cell response against alphaB-crystallin, a candidate autoantigen in MS. We used mice that are deficient for the target antigen. This condition allowed for a vigourous T-cell and antibody response to develop upon immunization, and eliminated all possible endogenous mechanisms of tolerance for alphaB-crystallin that are found in normal rodents. When applied 3 weeks after priming with alphaB-crystallin, intravenous administration of soluble antigen almost completely abrogated the established T-cell response in a dose-dependent manner as evidenced by T-cell non-responsiveness in tolerized animals to a re-challenge with antigen in complete Freund's adjuvant. Evaluating delayed-type hypersensitivity responses after tolerance induction revealed that the tolerizing effect was achieved within 24 hr. Furthermore, the tolerizing effect was found to be antigen-specific and long lasting. In contrast, serum antibody levels were markedly increased. Our data clarify that in the absence of any natural form of immune regulation, antigen-specific memory/effector T cells can be effectively silenced by intravenous antigen.
Collapse
Affiliation(s)
- Richard Verbeek
- Department of Biosciences, TNO Quality of Life, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
478
|
Watanabe H, Gaide O, Pétrilli V, Martinon F, Contassot E, Roques S, Kummer JA, Tschopp J, French LE. Activation of the IL-1beta-processing inflammasome is involved in contact hypersensitivity. J Invest Dermatol 2007; 127:1956-63. [PMID: 17429439 DOI: 10.1038/sj.jid.5700819] [Citation(s) in RCA: 313] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The inflammasome is a cytosolic protein complex regulating the activation of caspase-1, which cleaves the pro-inflammatory cytokines IL-1beta and IL-18 into their active form. The inflammasome is composed of a NACHT-, LRR- and pyrin (NALP) family member that acts as a sensor for danger signals and the adaptor protein apoptosis-associated speck-like protein containing a CARD domain (ASC), which allows the recruitment of caspase-1 in the complex. In the skin, exposure to contact sensitizers (CS) such as trinitro-chlorobenzene causes an immune response called contact hypersensitivity (CHS) or eczema. In this delayed-type hypersensitivity response, efficient priming of the adaptive immunity depends on the concomitant activation of the innate immune system, including IL-1beta/IL-18 activation in the skin. To determine if the inflammasome contributes to CHS, we have analyzed its capacity to react to CS in vitro and in vivo. We show here that key components of the inflammasome are present in human keratinocytes and that CS like trinitro-chlorobenzene induce caspase-1/ASC dependent IL-1beta and IL-18 processing and secretion. We also show that ASC- and NALP3-deficient mice display an impaired response to CS. These findings suggest that CS act as danger signals that activate the inflammasome in the skin, and reveal a new role of NALP3 and ASC as regulators of innate immunity in CHS.
Collapse
Affiliation(s)
- Hideki Watanabe
- Department of Dermatology, Geneva University Medical School, Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
479
|
Abstract
Antigen-nonspecific inflammation appears to contribute to postischemic brain injury. Because there is a breach in the integrity of the blood-brain barrier after stroke, the immune system encounters novel central nervous system (CNS) antigens that allow for the development of a CNS antigen-specific autoimmune response. The nature of the immune response generated on antigen encounter is determined by the microenvironment at the site of antigen encounter. For instance, a systemic inflammatory response, such as that which would accompany an infection, could alter the microenvironment in such a way as to promote the initiation of deleterious autoimmunity. If patients who develop an infection in the immediate poststroke period are predisposed toward a CNS autoimmune response, it might help to explain why infection after stroke is associated with increased disability. We present data to support this hypothesis and to show that the breach in the blood-brain barrier can also be capitalized on to modulate the immune response to create a neuroprotective environment after stroke.
Collapse
Affiliation(s)
- J Michael Gee
- Harborview Medical Center, University of Washington School of Medicine, Seattle, WA 98104-2499, USA
| | | | | | | |
Collapse
|
480
|
Choi S, Schwartz RH. Molecular mechanisms for adaptive tolerance and other T cell anergy models. Semin Immunol 2007; 19:140-52. [PMID: 17400472 PMCID: PMC2045643 DOI: 10.1016/j.smim.2007.02.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 02/16/2007] [Indexed: 01/09/2023]
Abstract
Since the original description of T cell anergy in CD4 clones from mice and humans, a number of different unresponsive states have been described, both in vivo and in vitro, that have been called anergic. While initial attempts were made to understand the similarities between the different models, it has now become clear from biochemical experiments that many of them have different molecular mechanisms underlying their unresponsiveness. In this review we will detail our own work on the in vivo model referred to as adaptive tolerance and then attempt to compare this biochemical state to the multitude of other states that have been described in the literature.
Collapse
Affiliation(s)
- Seeyoung Choi
- National Institutes of Health, LCMI, NIAID, Bethesda, MD 20892-0420, USA
| | | |
Collapse
|
481
|
Abstract
The human gut offers more than 200 m2 of mucosal surface, where direct interactions between the immune system and foreign antigens take place to eliminate pathogens or induce immune tolerance toward food antigens or normal gut flora. Therefore, mucosally administered antigens can induce tolerance under certain circumstances. In autoimmune diabetes, mucosal vaccination with autoantigens elicits some efficacy in restoring tolerance in mice, but it never succeeded in humans. Furthermore, in some instances autoimmunity can be precipitated upon oral or intranasal autoantigen administration. Therefore, it is difficult to predict the effect of mucosal vaccination on autoimmunity and much effort should be put into establishing better assays to reduce the risk for possible adverse events in humans and enable a rapid and smooth translation.
Collapse
Affiliation(s)
- Georgia Fousteri
- La Jolla Institute for Allergy and Immunology, Department of Developmental Immunology 3, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
482
|
Abstract
The mucosal immune system is governed by a unique set of rules and regulations. The local microenvironment dictates the necessity for these differences. The intestinal epithelial cell (IEC) sits at the interface between an antigen-rich lumen and a lymphocyte-rich lamina propria (LP). The cross talk that occurs between these compartments serves to maintain intestinal homeostasis. IECs have the capacity to talk to LP lymphocytes, activating populations of unique regulatory T cells. These cells have the capacity to talk back to the epithelium, influencing epithelial cell growth and differentiation. This review looks at this cross talk and places it in the context of mucosal immunoregulation.
Collapse
Affiliation(s)
- Stephanie Dahan
- Immunobiology Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
483
|
Ribeiro LC, Dickman R, Bernardes AT, Vaz NM. Dynamic stability in random and scale-free B-lymphocyte networks. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2007; 75:031911. [PMID: 17500730 DOI: 10.1103/physreve.75.031911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2006] [Indexed: 05/15/2023]
Abstract
One of the most intriguing features of the immune system is regulation: a limited response when perturbed repeatedly. We propose a minimal network model for immune regulation in a lymphocyte network containing two types of elements: B lymphocytes and ligands that bind to their receptors. Effective interactions between B cells, mediated by other components of the immune system can be excitatory or inhibitory. In our model, B cell clones and ligand species are represented by nodes, and interactions by links. We expect that, as in many complex systems, the connectivity distribution is broad, motivating study of the model on a scale-free network; for comparison we study the same dynamics on a random graph. We characterize the dynamics of the model and its response to perturbations. Our model reproduces several key features of immune system dynamics: regulation (saturation of response), and more rapid response upon repeated perturbation with the same agents. Our results suggest that a scale-free network of interactions contributes to the regulation and dynamics of the immune system.
Collapse
Affiliation(s)
- Leonardo C Ribeiro
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, 30161-970 Belo Horizonte-MG, Brazil
| | | | | | | |
Collapse
|
484
|
|
485
|
Frossard CP, Steidler L, Eigenmann PA. Oral administration of an IL-10-secreting Lactococcus lactis strain prevents food-induced IgE sensitization. J Allergy Clin Immunol 2007; 119:952-9. [PMID: 17316776 DOI: 10.1016/j.jaci.2006.12.615] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 12/04/2006] [Accepted: 12/11/2006] [Indexed: 01/11/2023]
Abstract
BACKGROUND Because tolerance to food is potentially modulated by IL-10, strategies to prevent food allergy should favor an increased delivery of IL-10 to the gut. OBJECTIVES We hypothesized that administration of a Lactococcus lactis transfected to secrete murine IL-10 could prevent sensitization in a mouse model of food allergy. METHODS Before each oral sensitization with beta-lactoglobulin in the presence of cholera toxin, young mice were administered the transfected Lactococcus lactis. Antigen-induced anaphylaxis after oral challenge assessed clinical protection achieved by the pretreatment. Serum and feces antigen-specific antibody concentrations were sequentially measured. Antibody titers were correlated with antibody and IL-10-secreting cell numbers in the spleen and in Peyer patches. RESULTS Pretreatment with transfected Lactococcus lactis contributed to diminish anaphylaxis significantly, and inhibit antigen-specific serum IgE and IgG(1) production strongly. In addition, transfected Lactococcus lactis increased the production of antigen-specific IgA in the gut. Variations of antibody levels in the serum and the gut correlated with the numbers of antibody-producing cells. In addition, the presence of exogenous IL-10 in the gut by transfected Lactococcus lactis induced IL-10 secretion by Peyer patches cells. Increased IL-10 titers were also measured in the plasma. CONCLUSION These results suggest that a microorganism bioengineered to deliver IL-10 in the gut can decrease food-induced anaphylaxis and provide an option to prevent IgE-type sensitization to common food allergens. CLINICAL IMPLICATIONS Nonpathogenic IL-10-producing microorganisms in the gut could have a potential to prevent systemic food-induced anaphylaxis.
Collapse
|
486
|
Oliveira FM, Dos Santos EM, Alves AC, Campana-Pereira MA, Ramaldes GA, Cardoso VN, Ruiz-de-Souza V, Gontijo CM. Digestion, Absorption and Tissue Distribution of Ovalbumin and Palmitoyl-ovalbumin: Impact on Immune Responses Triggered by Orally Administered Antigens. Scand J Immunol 2007; 65:139-47. [PMID: 17257218 DOI: 10.1111/j.1365-3083.2006.01884.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Previous work in this laboratory has demonstrated that ovalbumin coupled to palmitoyl residues (palmitoyl-Ova) does not induce oral tolerance. The present study sought to determine whether this coupling affects digestion, absorption and transfer of antigen. Ova and palmitoyl-Ova were shown to be digested differently in vitro by proteolytic enzymes and presented different tissue distribution kinetics after being labelled with (99m)technetium and orally administered to animals. Palmitoyl-Ova remained longer in the stomach, while native Ova was quickly transferred to the gut and other organs. After 3 h, higher levels of palmitoyl-Ova were found in the blood, Peyer's patches, mesenteric lymph nodes, liver and, especially, the spleen, which appears to be essential for immunization with palmitoyl-Ova. In fact, splenectomized mice treated orally with palmitoyl-Ova became tolerant, while tolerance to Ova was not affected. Thus, palmitoyl coupling was demonstrated to affect antigen digestion, absorption and transport. This is the first time that the spleen has been shown to be required for oral immunization with palmitoyl-Ova.
Collapse
Affiliation(s)
- F M Oliveira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, UFMG, Belo Horizonte, MG, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
487
|
Takaiwa F. A Rice-Based Edible Vaccine Expressing Multiple T-Cell Epitopes to Induce Oral Tolerance and Inhibit Allergy. Immunol Allergy Clin North Am 2007; 27:129-39. [PMID: 17276883 DOI: 10.1016/j.iac.2006.11.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Plant pollens are the most common cause of seasonal allergic disease. The number of patients undergoing treatment for allergies to the pollen of Japanese cedar (major antigens, Cry j 1 and Cry j 2) has increased steadily each year. A rice seed-based edible vaccine has been shown to be effective for treating Japanese cedar pollinosis. Rice seeds containing the major T-cell epitopes derived from cedar pollen allergens were orally administrated to mice before systemic challenge with total pollen protein. Mucosal immune tolerance leading to a reduction of allergen-specific IgE, T-cell proliferative reactions, and histamine were induced, resulting in suppression of allergy-specific symptoms such as sneezing. Oral seed-based peptide immunotherapy offers a safe, simple, and cost-effective alternative to conventional allergen-specific immunotherapy using crude allergen extracts for treating allergic disease. A human version of rice seed-based edible vaccine containing seven T-cell epitopes from the Cry j 1 and Cry j 2 allergens was recently developed and is undergoing safety assessments.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Disease Models, Animal
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/therapeutic use
- Humans
- Mice
- Oryza/genetics
- Oryza/immunology
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/immunology
- Rhinitis, Allergic, Seasonal/immunology
- Rhinitis, Allergic, Seasonal/prevention & control
- Vaccination/methods
- Vaccines, Edible/immunology
Collapse
Affiliation(s)
- Fumio Takaiwa
- Transgenic Crop Research and Development Center, National Institute of Agrobiological Sciences, Kannondai 2-1-2, Tsukuba, Ibaraki 305-8602, Japan.
| |
Collapse
|
488
|
Ochoa-Repáraz J, Riccardi C, Rynda A, Jun S, Callis G, Pascual DW. Regulatory T cell vaccination without autoantigen protects against experimental autoimmune encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:1791-9. [PMID: 17237429 PMCID: PMC9811398 DOI: 10.4049/jimmunol.178.3.1791] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Regulatory T (T(reg)) cells show promise for treating autoimmune diseases, but their induction to elevated potency has been problematic when the most optimally derived cells are from diseased animals. To circumvent reliance on autoantigen-reactive T(reg) cells, stimulation to myelin-independent Ags may offer a viable alternative while maintaining potency to treat experimental autoimmune encephalomyelitis (EAE). The experimental Salmonella vaccine expressing colonization factor Ag I possesses anti-inflammatory properties and, when applied therapeutically, reduces further development of EAE in SJL mice. To ascertain T(reg) cell dependency, a kinetic analysis was performed showing increased levels of FoxP3(+)CD25(+)CD4(+) T cells. Inactivation of these T(reg) cells resulted in loss of protection. Adoptive transfer of the vaccine-induced T(reg) cells protected mice against EAE with greater potency than naive or Salmonella vector-induced T(reg) cells, and cytokine analysis revealed enhanced production of TGF-beta, not IL-10. The development of these T(reg) cells in conjunction with immune deviation by Th2 cells optimally induced protective T(reg) cells when compared those induced in the absence of Th2 cells. These data show that T(reg) cells can be induced to high potency to non-disease-inducing Ags using a bacterial vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | - David W. Pascual
- Address correspondence and reprint requests to Dr. David W. Pascual, Veterinary Molecular Biology, P.O. Box 173610, Montana State University, Bozeman, MT 59717-3610.
| |
Collapse
|
489
|
Pieters R. Detection of autoimmunity by pharmaceuticals. Methods 2007; 41:112-7. [PMID: 17161307 DOI: 10.1016/j.ymeth.2006.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Accepted: 09/22/2006] [Indexed: 02/02/2023] Open
Abstract
Despite the important health and economic impact of autoimmunogenicity or allergenicity by pharmaceuticals models to detect such adverse effects are not available yet. The most important reason for this is the related complex interplay of multiple factors, for which reason these adverse effects are also referred to as idiosyncratic in nature. Moreover, clinical effects are quite diverse, and involve both organ-specific and systemic effects, including a diversity of skin diseases. Because of its complexity on the one hand and the fundamental knowledge on certain particular mechanistic effects it may be more relevant to design a rationalistic toolbox of test models from which a predictive strategy can be composed. Since one mechanistic aspect centers around T cell sensitization a straightforward lymph node assay such as the reporter antigen-popliteal lymph node assay (RA-PLNA) would fit in such a toolbox. This RA-PLNA holds a strong promise to distinguish sensitizing and/or neoantigen-forming capacity of low molecular weight pharmaceuticals. In addition, from the pharmacokinetic point of view a rationalistic toolbox should also contain oral exposure models with immunological read out parameters in normal or in genetically predisposed animal strains. This review focuses on these two categories of candidate test methods, PLNA and oral exposure models, and proposes to use these in tandem in order to predict the hazard of induction of allergy or autoimmune phenomena by new pharmaceutical candidates.
Collapse
Affiliation(s)
- Raymond Pieters
- Institute for Risk Assessment Sciences, Immunotoxicology, Utrecht University, The Netherlands.
| |
Collapse
|
490
|
Shklovskaya E, Fazekas de St Groth B. Balancing tolerance and immunity: the role of dendritic cell and T cell subsets. Methods Mol Biol 2007; 380:25-46. [PMID: 17876085 DOI: 10.1007/978-1-59745-395-0_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The interaction between dendritic cells and T cells is crucial for the regulation of immunological tolerance and immunity. Although our understanding of the mechanisms responsible for these phenomena has advanced significantly in recent years, we are still lacking a fully integrated model of how dendritic cell phenotype correlates with function, and how complex interactions with multiple dendritic and T cell subpopulations shape the course of the immune response in vivo. In this review, we summarize the current state of knowledge in the field, highlighting the areas where further investigation is likely to advance our understanding of this fundamental immunological interaction.
Collapse
Affiliation(s)
- Elena Shklovskaya
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Australia
| | | |
Collapse
|
491
|
Fousteri G, Bresson D, von Herrath M. Rational development of antigen-specific therapies for type 1 diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 601:313-9. [PMID: 17713020 DOI: 10.1007/978-0-387-72005-0_34] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Administration of autoantigens, especially via the mucosal route, can induce tolerance under certain circumstances. In autoimmune diabetes, mucosal vaccination with autoantigens was frequently effective in restoring tolerance in mice but has not yet succeeded in humans. Furthermore, in some instances, autoimmunity can be precipitated upon autoantigen administration. We will here briefly discuss the underlying reasons and delineate which efforts should be made in the future to rationally translate antigen-specific immunotherapy, for example, by establishing better assays to reduce the risk for possible adverse events in humans.
Collapse
Affiliation(s)
- Georgia Fousteri
- La Jolla Institute for Allergy and Immunology, Department of Developmental Immunology, La Jolla, CA, USA
| | | | | |
Collapse
|
492
|
Nakayama T, Illoh K, Ruetzler C, Auh S, Sokoloff L, Hallenbeck J. Intranasal administration of E-selectin to induce immunological tolerization can suppress subarachnoid hemorrhage-induced vasospasm implicating immune and inflammatory mechanisms in its genesis. Brain Res 2006; 1132:177-84. [PMID: 17188657 PMCID: PMC1852471 DOI: 10.1016/j.brainres.2006.09.116] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2006] [Revised: 09/28/2006] [Accepted: 09/30/2006] [Indexed: 12/20/2022]
Abstract
Evidence that inflammatory and immune mechanisms may have a critical role in the development of vasospasm after subarachnoid hemorrhage is accumulating. We examined, therefore, whether induction of immunological tolerance to the adhesion molecule that is uniquely expressed on activated endothelium, E-selectin, could inhibit the vasospasm provoked by subarachnoid blood in a rat subarachnoid hemorrhage model. We found that intranasal instillation of E-selectin every other day for 10 days on a mucosal tolerization schedule suppressed delayed type hypersensitivity to E-selectin confirming tolerance to that molecule and markedly suppressed basilar artery spasm after subarachnoid hemorrhage. The results of this proof-of-concept study suggest that agents that can mimic the local effects of the mediators of mucosal tolerance could have therapeutic potential for the management of post-subarachnoid hemorrhage vasospasm.
Collapse
Affiliation(s)
- Toshiyuki Nakayama
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 49/Room 2A10, 49 Convent Drive, MSC 4476, Bethesda, MD 20892-4476, USA
- Laboratory of Cerebral Metabolism, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Kachikwu Illoh
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Christl Ruetzler
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 49/Room 2A10, 49 Convent Drive, MSC 4476, Bethesda, MD 20892-4476, USA
| | - Sungyoung Auh
- Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Louis Sokoloff
- Laboratory of Cerebral Metabolism, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - John Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 49/Room 2A10, 49 Convent Drive, MSC 4476, Bethesda, MD 20892-4476, USA
- *Corresponding author. Fax: +1 301 402 2769. E-mail address: (J. Hallenbeck)
| |
Collapse
|
493
|
Abstract
The immune system is organized as a number of distinct lymphoid organs interconnected by recirculating lymphocytes. These organs, such as lymph nodes, spleen, and gut-associated Peyer's patches, are compartmentalized, providing separate niches for T and B cells. In addition, regional compartmentalization of lymphoid organs themselves exists, leading to the distinction between the mucosal and the systemic immune systems. This distinction not only reflects the anatomical localization but also is based on functional differences, with predominant tolerance induction via mucosal routes and immunity seen after systemic antigen exposure. These differences are associated with regional differences in the lymphoid organs and with environmental conditions of the tissues in which the immune system functions. Recirculation patterns of lymphocytes differ between mucosal and systemic lymphoid organs, and more insight into the mechanisms that imprint this behavior has been generated recently. Differences in dendritic cells have been observed between mucosal and systemic sites, and knowledge on how local factors contribute to the immune system is emerging. From our studies on mucosal tolerance in mouse models, it has become evident that regional lymph nodes draining the mucosa are important sites to direct immune responses. Here, we discuss the way regional lymph nodes contribute to the direction of immune responses and what is known about the local factors and cell behavior that form the basis for these differences.
Collapse
Affiliation(s)
- Georg Kraal
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
494
|
Iweala OI, Nagler CR. Immune privilege in the gut: the establishment and maintenance of non-responsiveness to dietary antigens and commensal flora. Immunol Rev 2006; 213:82-100. [PMID: 16972898 DOI: 10.1111/j.1600-065x.2006.00431.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Immune privilege in the gut is the result of a complex interplay between the gut microbiome, gut luminal antigens, and the intestinal epithelial barrier. Composed of both physical and immunochemical components, the intestinal barrier secretes immunoregulatory mediators that promote the generation of tolerogenic antigen-presenting cells, phagocytic innate immune cells characterized by 'inflammatory anergy', and regulatory cells of the adaptive immune system. Innate immune cells mediate controlled transepithelial transport of luminal antigens as far as the mesenteric lymph nodes, where the intestinal and peripheral immune systems intersect. This promotes the generation of adaptive regulatory lymphocytes that actively suppress effector cell responses against gut luminal antigens and flora. The net result is the generation of tolerance to dietary antigens and the maintenance of gut homeostasis. Dysregulation of this complex immunoregulatory network leads to diseases such as food allergy and inflammatory bowel disease. Future therapies for these diseases will likely involve the functional restoration of the barrier and regulatory cell functions at the epithelial/luminal interface.
Collapse
Affiliation(s)
- Onyinye I Iweala
- Mucosal Immunology Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-4404, USA
| | | |
Collapse
|
495
|
Carrier Y, Yuan J, Kuchroo VK, Weiner HL. Th3 Cells in Peripheral Tolerance. I. Induction of Foxp3-Positive Regulatory T Cells by Th3 Cells Derived from TGF-β T Cell-Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 2006; 178:179-85. [PMID: 17182553 DOI: 10.4049/jimmunol.178.1.179] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
TGF-beta has been shown to be critical in the generation of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Because Th3 cells produce large amounts of TGF-beta, we asked whether induction of Th3 cells in the periphery was a mechanism by which CD4(+)CD25(+) Tregs were induced in the peripheral immune compartment. To address this issue, we generated a TGF-beta1-transgenic (Tg) mouse in which TGF-beta is linked to the IL-2 promoter and T cells transiently overexpress TGF-beta upon TCR stimulation but produce little or no IL-2, IL-4, IL-10, IL-13, or IFN-gamma. Naive TGF-beta-Tg mice are phenotypically normal with comparable numbers of lymphocytes and thymic-derived Tregs. We found that repeated antigenic stimulation of pathogenic myelin oligodendrocyte glycoprotein (MOG)-specific CD4(+)CD25(-) T cells from TGF-beta Tg mice crossed to MOG TCR-Tg mice induced Foxp3 expression in both CD25(+) and CD25(-) populations. Both CD25 subsets were anergic and had potent suppressive properties in vitro and in vivo. Furthermore, adoptive transfer of these induced regulatory CD25(+/-) T cells suppressed experimental autoimmune encephalomyelitis when administrated before disease induction or during ongoing experimental autoimmune encephalomyelitis. The suppressive effect of TGF-beta on T cell responses was due to the induction of Tregs and not to the direct inhibition of cell proliferation. The differentiation of Th3 cells in vitro was TGF-beta dependent as anti-TGF-beta abrogated their development. Thus, Ag-specific TGF-beta-producing Th3 cells play a crucial role in inducing and maintaining peripheral tolerance by driving the differentiation of Ag-specific Foxp3(+) regulatory cells in the periphery.
Collapse
Affiliation(s)
- Yijun Carrier
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
496
|
van Wijk F, Knippels L. Initiating mechanisms of food allergy: Oral tolerance versus allergic sensitization. Biomed Pharmacother 2006; 61:8-20. [PMID: 17218079 DOI: 10.1016/j.biopha.2006.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 08/21/2006] [Indexed: 12/12/2022] Open
Abstract
Immediately after birth the mucosa of the gastrointestinal tract, which represents the greatest body surface area exposed to the outside environment, is confronted with a large variety of foreign antigens. The immune system of the intestine now has to meet the task of discriminating between pathogens and harmless antigens, such as food proteins and commensal bacteria, and to respond accordingly. This important job is fulfilled by cells of the gut-associated lymphoid tissue, the largest immunologic organ in the body. Despite the large extent of food antigen exposure, only a small percentage of individuals experience adverse immunologic reactions to food. This is due to the fact that the normal immune response to dietary proteins is associated with the induction of oral tolerance, which refers to a state of active inhibition of immune responses to an antigen by means of prior exposure to that antigen via the oral route. Abrogation of oral tolerance or failure to induce oral tolerance may result in the development of food hypersensitivity. In the present review, factors that may play a role in the outcome of oral tolerance versus sensitization to food proteins are discussed.
Collapse
Affiliation(s)
- Femke van Wijk
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Department of Pediatric Immunology, Utrecht, The Netherlands
| | | |
Collapse
|
497
|
|
498
|
|
499
|
Ménager-Marcq I, Pomié C, Romagnoli P, van Meerwijk JP. CD8+CD28- regulatory T lymphocytes prevent experimental inflammatory bowel disease in mice. Gastroenterology 2006; 131:1775-85. [PMID: 17087950 PMCID: PMC1950262 DOI: 10.1053/j.gastro.2006.09.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 08/31/2006] [Indexed: 01/23/2023]
Abstract
BACKGROUND & AIMS Immune responses to innocuous intestinal antigens appear tightly controlled by regulatory T lymphocytes. While CD4+ T lymphocytes have recently attracted the most attention, CD8+ regulatory T-cell populations are also believed to play an important role in control of mucosal immunity. However, CD8+ regulatory T-cell function has mainly been studied in vitro and no direct in vivo evidence exists that they can control mucosal immune responses. We investigated the capacity of CD8+CD28- T cells to prevent experimental inflammatory bowel disease (IBD) in mice. METHODS CD8+CD28- regulatory T cells were isolated from unmanipulated mice and tested for their capacity to inhibit T-cell activation in allogeneic mixed lymphocyte cultures in vitro and to prevent IBD induced by injection of CD4+CD45RB(high) cells into syngeneic immunodeficient RAG-2 mutant mice. RESULTS CD8+CD28- T lymphocytes inhibited proliferation and interferon gamma production by CD4+ responder T cells in vitro. CD8+CD28- regulatory T cells freshly isolated from spleen or gut efficiently prevented IBD induced by transfer of colitogenic T cells into immunodeficient hosts. Regulatory CD8+CD28- T cells incapable of producing interleukin-10 did not prevent colitis. Moreover, IBD induced with colitogenic T cells incapable of responding to transforming growth factor beta could not be prevented with CD8+CD28- regulatory T cells. CD8+CD28+ T cells did not inhibit in vitro or in vivo immune responses. CONCLUSIONS Our findings show that naturally occurring CD8+CD28- regulatory T lymphocytes can prevent experimental IBD in mice and suggest that these cells may play an important role in control of mucosal immunity.
Collapse
Affiliation(s)
- Ingrid Ménager-Marcq
- Centre de Physiopathologie Toulouse Purpan
INSERM : U563 IFR30Université Paul Sabatier - Toulouse IIIHopital de Purpan Place du Docteur Baylac
31024 Toulouse,FR
| | - Céline Pomié
- Centre de Physiopathologie Toulouse Purpan
INSERM : U563 IFR30Université Paul Sabatier - Toulouse IIIHopital de Purpan Place du Docteur Baylac
31024 Toulouse,FR
| | - Paola Romagnoli
- Centre de Physiopathologie Toulouse Purpan
INSERM : U563 IFR30Université Paul Sabatier - Toulouse IIIHopital de Purpan Place du Docteur Baylac
31024 Toulouse,FR
| | - Joost P.M. van Meerwijk
- Centre de Physiopathologie Toulouse Purpan
INSERM : U563 IFR30Université Paul Sabatier - Toulouse IIIHopital de Purpan Place du Docteur Baylac
31024 Toulouse,FR
- Institut universitaire de France
103, bd Saint-Michel
75005 Paris,FR
- Faculté des sciences
Université Paul Sabatier - Toulouse IIIFR
| |
Collapse
|
500
|
Astier AL, Meiffren G, Freeman S, Hafler DA. Alterations in CD46-mediated Tr1 regulatory T cells in patients with multiple sclerosis. J Clin Invest 2006; 116:3252-7. [PMID: 17099776 PMCID: PMC1635165 DOI: 10.1172/jci29251] [Citation(s) in RCA: 223] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Accepted: 09/19/2006] [Indexed: 12/20/2022] Open
Abstract
Loss of Treg function appears to be a critical factor in the pathogenesis of human autoimmune diseases. Attention has focused on defects of CD4(+)CD25(high) Tregs, and techniques have been developed to determine their function. In contrast, the role of Tr1 regulatory T cells, which secrete the antiinflammatory cytokine IL-10, in autoimmune disease has not been well assessed. CD46 is a newly defined costimulatory molecule for T cell activation, and CD46-costimulated human T cells induce a Tr1 Treg phenotype with considerable amounts of IL-10 secretion. Here, we examined the role of Tr1 cells in patients with multiple sclerosis (MS) by stimulating CD4(+) T cells with anti-CD3 and -CD46 mAbs and measuring IL-10 secretion. There were striking defects in the induction of Tr1 cells with CD46 costimulation as measured by IL-10 but not IFN-gamma secretion in patients with MS compared with healthy subjects. This loss of Tr1 cell-associated IL-10 secretion was specific to CD46 and not CD28 costimulation and was associated with an altered regulation of the CD46-Cy2 isoform that differentially regulates T cell function in a CD46-transgenic murine model. These data demonstrate a second major Treg defect in human autoimmune disease associated with the CD46 pathway.
Collapse
Affiliation(s)
- Anne L. Astier
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| | - Gregory Meiffren
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| | - Samuel Freeman
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| | - David A. Hafler
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| |
Collapse
|