301
|
Appleby MW, Ramsdell F. Scurfy, the Foxp3 locus, and the molecular basis of peripheral tolerance. Curr Top Microbiol Immunol 2008; 321:151-68. [PMID: 18727491 DOI: 10.1007/978-3-540-75203-5_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability to rapidly and efficiently recognize and eliminate pathogens while sparing normal self tissue is a hallmark of the mammalian immune system. When it fails, however, autoimmune disease results. The genetic and environmental factors that control the process of making such distinctions, not to mention the specific targeted tissues, are extraordinarily complex in the human population; only now are we characterizing the candidate genes responsible for these responses to pathogens. The examination of specific traits in murine models of disease has led to the identification of many of the candidate genes for human disease. The study of mouse mutations (both induced and spontaneous) has also greatly advanced our understanding of the immune responses and autoimmune disease. Here, we describe the use of classical mouse genetics to identify one gene centrally involved in the control of immune responses. Furthermore, although mutations in the orthologous human gene result in a virtually identical phenotype to that seen in the mouse, it is unlikely that studying the human disease populations alone would have successfully identified this gene. Thus, despite the complete sequencing of the human and mouse genomes, the examination of murine mutations remains a powerful and unbiased tool to connect genotype and phenotype.
Collapse
|
302
|
|
303
|
Kirk AD, Elster EA. Immunology of Transplantation. Surgery 2008. [DOI: 10.1007/978-0-387-68113-9_81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
304
|
Kahan BD. Concepts and challenges in transplantation: rejection, immunosuppression and tolerance. Clin Immunol 2008. [DOI: 10.1016/b978-0-323-04404-2.10080-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
305
|
Goodnow CC. Nossal and Pike 1975: a turning point in the effort to define self-tolerance mechanisms. THE JOURNAL OF IMMUNOLOGY 2007; 179:5617-8. [PMID: 17947630 DOI: 10.4049/jimmunol.179.9.5617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Christopher C Goodnow
- Australian National University, Immunology and Genetics Division, Australian Phenomics Facility, John Curtin School of Medical Research, Canberra, australian Capital Territory, Australia.
| |
Collapse
|
306
|
McLachlan SM, Nagayama Y, Pichurin PN, Mizutori Y, Chen CR, Misharin A, Aliesky HA, Rapoport B. The link between Graves' disease and Hashimoto's thyroiditis: a role for regulatory T cells. Endocrinology 2007; 148:5724-33. [PMID: 17823263 DOI: 10.1210/en.2007-1024] [Citation(s) in RCA: 84] [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: 01/04/2023]
Abstract
Hyperthyroidism in Graves' disease is caused by thyroid-stimulating autoantibodies to the TSH receptor (TSHR), whereas hypothyroidism in Hashimoto's thyroiditis is associated with thyroid peroxidase and thyroglobulin autoantibodies. In some Graves' patients, thyroiditis becomes sufficiently extensive to cure the hyperthyroidism with resultant hypothyroidism. Factors determining the balance between these two diseases, the commonest organ-specific autoimmune diseases affecting humans, are unknown. Serendipitous findings in transgenic BALB/c mice, with the human TSHR A-subunit targeted to the thyroid, shed light on this relationship. Of three transgenic lines, two expressed high levels and one expressed low intrathyroidal A-subunit levels (Hi- and Lo-transgenics, respectively). Transgenics and wild-type littermates were depleted of T regulatory cells (Treg) using antibodies to CD25 (CD4(+) T cells) or CD122 (CD8(+) T cells) before TSHR-adenovirus immunization. Regardless of Treg depletion, high-expressor transgenics remained tolerant to A-subunit-adenovirus immunization (no TSHR antibodies and no hyperthyroidism). Tolerance was broken in low-transgenics, although TSHR antibody levels were lower than in wild-type littermates and no mice became hyperthyroid. Treg depletion before immunization did not significantly alter the TSHR antibody response. However, Treg depletion (particularly CD25) induced thyroid lymphocytic infiltrates in Lo-transgenics with transient or permanent hypothyroidism (low T(4), elevated TSH). Neither thyroid lymphocytic infiltration nor hypothyroidism developed in similarly treated wild-type littermates. Remarkably, lymphocytic infiltration was associated with intermolecular spreading of the TSHR antibody response to other self thyroid antigens, murine thyroid peroxidase and thyroglobulin. These data suggest a role for Treg in the natural progression of hyperthyroid Graves' disease to Hashimoto's thyroiditis and hypothyroidism in humans.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | |
Collapse
|
307
|
Siggs OM, Miosge LA, Yates AL, Kucharska EM, Sheahan D, Brdicka T, Weiss A, Liston A, Goodnow CC. Opposing functions of the T cell receptor kinase ZAP-70 in immunity and tolerance differentially titrate in response to nucleotide substitutions. Immunity 2007; 27:912-26. [PMID: 18093540 PMCID: PMC3163119 DOI: 10.1016/j.immuni.2007.11.013] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 09/05/2007] [Accepted: 11/02/2007] [Indexed: 01/26/2023]
Abstract
Null mutations that cripple T cell receptor (TCR) signaling explain rare primary immunodeficiencies, but it is not understood why more common polymorphisms that lead to subtle TCR signaling defects are paradoxically associated with autoimmunity. Here we analyzed how a series of Zap70 variants with step-wise decreases in TCR signaling impacted upon opposing TCR functions of immunity and tolerance. One Zap70 variant, murdock, moderately decreased TCR signaling and thymic selection without compromising immunological tolerance, whereas a more severe Zap70 defect, mrtless, abolished thymic-positive selection and led to immunodeficiency. Signaling capacities between these two thresholds disproportionately compromised negative selection and Foxp3(+) regulatory T cell formation, creating a cellular imbalance between immunogenic and tolerogenic functions that resulted in the excessive production of autoantibodies and immunoglobulin E (IgE). The pleiotropic functions of ZAP-70 and their differential response to graded variation provide a paradigm for understanding the complex outcomes of human genetic variation.
Collapse
Affiliation(s)
- Owen M Siggs
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| | - Lisa A Miosge
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| | - Adèle L Yates
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| | - Edyta M Kucharska
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| | - Daniel Sheahan
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| | - Tomas Brdicka
- Department of Medicine and Howard Hughes Medical Institute, University of California, San Francisco, California 94143
| | - Arthur Weiss
- Department of Medicine and Howard Hughes Medical Institute, University of California, San Francisco, California 94143
| | - Adrian Liston
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| | - Christopher C Goodnow
- John Curtin School of Medical Research and Australian Phenomics Facility, The Australian National University, Canberra 2601, Australia
| |
Collapse
|
308
|
|
309
|
Deng L, Mariuzza RA. Recognition of self-peptide-MHC complexes by autoimmune T-cell receptors. Trends Biochem Sci 2007; 32:500-8. [PMID: 17950605 PMCID: PMC2739108 DOI: 10.1016/j.tibs.2007.08.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 08/08/2007] [Accepted: 08/13/2007] [Indexed: 10/22/2022]
Abstract
T cell receptors (TCR) recognize antigenic peptides displayed by MHC molecules. Whereas T-cell recognition of foreign peptides is essential for immune defense against microbial pathogens, recognition of self-peptides can cause autoimmune disease. Structural studies of anti-foreign TCR showed remarkable similarities in the topology of TCR binding to peptide-MHC, which maximize interactions with the ligand. However, recent structures involving autoimmune and tumor-specific TCR have revealed that they engage self-peptide-MHC with different topologies, which are suboptimal for TCR binding. These differences might reflect the distinct selection pressures exerted on anti-microbial versus autoreactive T cells. The structures also provide new insights into TCR cross-reactivity, which can contribute to autoimmunity by increasing the likelihood of self-peptide-MHC recognition.
Collapse
Affiliation(s)
- Lu Deng
- Center for Advanced Research in Biotechnology, W.M. Keck Laboratory for Structural Biology, University of Maryland Biotechnology Institute, Rockville, MD 20850, USA
| | | |
Collapse
|
310
|
Immunogenicity of protein therapeutics. Trends Immunol 2007; 28:482-90. [DOI: 10.1016/j.it.2007.07.011] [Citation(s) in RCA: 359] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 07/31/2007] [Accepted: 07/31/2007] [Indexed: 01/31/2023]
|
311
|
Naeher D, Daniels MA, Hausmann B, Guillaume P, Luescher I, Palmer E. A constant affinity threshold for T cell tolerance. ACTA ACUST UNITED AC 2007; 204:2553-9. [PMID: 17938233 PMCID: PMC2118488 DOI: 10.1084/jem.20070254] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
T cell tolerance depends on the T cell receptor's affinity for peptide/major histocompatibility complex (MHC) ligand; this critical parameter determines whether a thymocyte will be included (positive selection) or excluded (negative selection) from the T cell repertoire. A quantitative analysis of ligand binding was performed using an experimental system permitting receptor–coreceptor interactions on live cells under physiological conditions. Using three transgenic mouse strains expressing distinct class I MHC–restricted T cell receptors, we determined the affinity that defines the threshold for negative selection. The affinity threshold for self-tolerance appears to be a constant for cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Dieter Naeher
- Laboratory of Transplantation Immunology, Department of Research, University Hospital, 4031 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
312
|
Fowell D, Powrie F, Saoudi A, Seddon B, Heath V, Mason D. The role of subsets of CD4+ T cells in autoimmunity. CIBA FOUNDATION SYMPOSIUM 2007; 195:173-82; discussion 182-8. [PMID: 8724837 DOI: 10.1002/9780470514849.ch12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
It is generally considered that T cells which are reactive with self-antigens are effectively eliminated by two processes: clonal deletion and the induction of T cell anergy. More recently, it has been shown that some potentially autoreactive T cells remain unactivated because the self-antigens for which they are specific are not presented on competent antigen-presenting cells. All these mechanisms of self-tolerance may be regarded as passive in the sense that the autoreactive cells are either deleted or are intrinsically non-responsive. If this view of self-tolerance is adopted, then one would predict that rendering animals relatively lymphopoenic should not give rise to autoimmune disease. This prediction is not verified by experiment. Rats rendered relatively lymphopoenic by adult thymectomy followed by repeated low dose gamma-irradiation develop a high incidence of autoimmune diabetes. Furthermore, it has been shown that the reconstitution of these rats with a specific subset of CD4+ T cells from syngeneic donors prevents the development of this disease. The protective cells have the CD45RClow phenotype, they are resistant to adult thymectomy and the majority of them appear to be non-activated in the donor rats. In contrast, the CD45RChigh CD4+ subset does not provide protection from diabetes. Instead, on injection into athymic rats, it gives rise to pathological changes in a variety of organs: stomach, pancreas, liver, thyroid and lung. In addition, the CD45RClow CD4+ subset prevents these manifestations of autoimmunity in these circumstances. Recently, we have shown that CD4+ CD8- thymocytes are a highly potent source of cells that have the ability to control autoimmune diabetes in rats. It appears that the thymus has three distinct functions: positive selection; negative selection; and the generation of a population of cells that seem specialized for the control of autoimmunity.
Collapse
Affiliation(s)
- D Fowell
- MRC Cellular Immunology Unit, Sir William Dunn School of Pathology, University of Oxford, UK
| | | | | | | | | | | |
Collapse
|
313
|
Huber BT, Beutner U, Subramanyam M. The role of superantigens in the immunobiology of retroviruses. CIBA FOUNDATION SYMPOSIUM 2007; 187:132-40; discussion 140-3. [PMID: 7796668 DOI: 10.1002/9780470514672.ch9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Murine mammary tumour viruses (MMTVs) are retroviruses that encode superantigens capable of stimulating T cells via superantigen-reactive T cell receptor V beta chains. MMTVs are transmitted to the suckling offspring via the milk. We have established that class II and B cell-deficient mice that were foster nursed by virus-secreting mice do not transfer infectious MMTVs to their offspring. No MMTV proviruses could be detected in the spleen and mammary tissue of these mice and there was no deletion of MMTV superantigen-reactive T cells. These results confirm that superantigen expression in the context of MHC class II molecules is required for MMTV transmission. We conclude that B cells are essential for the completion of the viral life cycle in vivo. This indicates that B cells are infected first and that viral amplification takes place only if infected B cells present the MMTV superantigen on their surface which, in turn, results in activation of T cells expressing the appropriate T cell receptor V beta chains. These activated T cells stimulate B cells which enables viral replication. Human T cells carry all the structural features required for an efficient response to murine retrovirally encoded superantigens. Superantigen-like stimulation of human T cells has been demonstrated in both infectious and autoimmune diseases. Human immunodeficiency virus may encode a superantigen but this has not been proven.
Collapse
Affiliation(s)
- B T Huber
- Department of Pathology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | |
Collapse
|
314
|
Sundberg EJ, Deng L, Mariuzza RA. TCR recognition of peptide/MHC class II complexes and superantigens. Semin Immunol 2007; 19:262-71. [PMID: 17560120 PMCID: PMC2949352 DOI: 10.1016/j.smim.2007.04.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 04/23/2007] [Accepted: 04/23/2007] [Indexed: 11/21/2022]
Abstract
Major histocompatibility complex (MHC) class II molecules display peptides to the T cell receptor (TCR). The ability of the TCR to discriminate foreign from self-peptides presented by MHC molecules is a requirement of an effective adaptive immune response. Dysregulation of this molecular recognition event often leads to a disease state. Recently, a number of structural studies have provided significant insight into several such dysregulated interactions between peptide/MHC complexes and TCR molecules. These include TCR recognition of self-peptides, which results in autoimmune reactions, and of mutant self-peptides, common in the immunosurveillance of tumors, as well as the engagement of TCRs by superantigens, a family of bacterial toxins responsible for toxic shock syndrome.
Collapse
Affiliation(s)
- Eric J Sundberg
- Boston Biomedical Research Institute, Watertown, MA 02472, USA.
| | | | | |
Collapse
|
315
|
McMahan RH, Slansky JE. Mobilizing the low-avidity T cell repertoire to kill tumors. Semin Cancer Biol 2007; 17:317-29. [PMID: 17651986 PMCID: PMC2040124 DOI: 10.1016/j.semcancer.2007.06.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/08/2007] [Accepted: 06/07/2007] [Indexed: 01/09/2023]
Abstract
Optimally, T cells destroy infected and transformed cells of the host. To be effective the T cell repertoire must have a sufficiently diverse number of T cell receptors (TCRs) to recognize the abundance of foreign and tumor antigens presented by MHC molecules. The T cell repertoire must also not be reactive toward self-antigens on healthy cells to prevent autoimmunity. Unlike antigens derived from pathogens, most tumor-associated antigens (TAA) are also self-antigens. Therefore, central and peripheral tolerance mechanisms delete or inhibit tumor-reactive T cells. Although there are T cells within the peripheral repertoire that recognize TAA, these T cells are not sufficient to prevent growth of clinically relevant tumors. We will discuss how this dysfunction results, in part, from the low functional avidity of T cells for tumor, or antigen presenting cells (APC) displaying TAA. We discuss the limitations of these low-avidity tumor-reactive T cells and review current immunotherapies aimed at enhancing the avidity and antitumor activity of the tumor-specific T cell repertoire.
Collapse
Affiliation(s)
- Rachel H. McMahan
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, CO 80206, USA
| | - Jill E. Slansky
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, CO 80206, USA
| |
Collapse
|
316
|
Pillai A, Teo P, George T, Mukhopadhyay A, Dejbakhsh-Jones S, Strober S. Alloantigen recognition is critical for CD8 T cell-mediated graft anti-tumor activity against murine BCL1 lymphoma after myeloablative bone marrow transplantation. Bone Marrow Transplant 2007; 40:487-97. [PMID: 17603512 DOI: 10.1038/sj.bmt.1705759] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The goal of the current study was to determine whether whole bone marrow cells or splenic CD8(+) T cells from C57BL/6 (H-2(b)) donor mice, which are tolerant to BALB/c (H-2(d)) alloantigens, are capable of mediating graft anti-tumor activity against a BALB/c B-cell lymphoma after injection into irradiated BALB/c hosts. The experimental results show that high doses of splenic CD8(+) T cells mixed with T cell-depleted bone marrow cells from C57BL/6 non-tolerant (normal) donors eliminate the BCL(1) B-cell lymphoma cells and induce lethal graft-versus-host disease (GVHD). CD8(+) T cells from tolerant donors simultaneously lose both their ability to induce GVHD and their anti-tumor activity. Whole bone marrow cell transplants from normal donors eliminated BCL(1) tumor cells without inducing GVHD, and bone marrow cells from tolerant donors failed to eliminate the tumor cells. The infused BCL(1) tumor cells expressed an immunogenic tumor-specific idiotype antigen disparate from host alloantigens, indicating that recognition of the tumor-specific antigen alone was insufficient to elicit graft anti-tumor activity from unimmunized allotolerant donor splenic CD8(+) T cells or whole bone marrow cells. We conclude that CD8(+) T cells from unimmunized normal donor mice require alloantigen recognition to mediate their anti-tumor activity following allogeneic BMT.
Collapse
Affiliation(s)
- A Pillai
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5166, USA
| | | | | | | | | | | |
Collapse
|
317
|
Field EH, Kulhankova K, Nasr ME. Natural Tregs, CD4+CD25+ inhibitory hybridomas, and their cell contact dependent suppression. Immunol Res 2007; 39:62-78. [DOI: 10.1007/s12026-007-0064-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/30/2022]
|
318
|
Yunis EJ, Zuniga J, Romero V, Yunis EJ. Chimerism and tetragametic chimerism in humans: implications in autoimmunity, allorecognition and tolerance. Immunol Res 2007; 38:213-36. [DOI: 10.1007/s12026-007-0013-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 10/23/2022]
|
319
|
Lee KY, Choi YI, Kim J, Choi JW, Sohn DH, Lee C, Jeon SH, Seong RH. Down-Regulation of the SWI/SNF Chromatin Remodeling Activity by TCR Signaling Is Required for Proper Thymocyte Maturation. THE JOURNAL OF IMMUNOLOGY 2007; 178:7088-96. [PMID: 17513758 DOI: 10.4049/jimmunol.178.11.7088] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The process of thymocyte development requires an exquisite regulation of many genes via transcription factors and chromatin remodeling activities. Even though the SWI/SNF chromatin remodeling complex has been thought to play important roles during thymocyte development, its known function is very limited. In this study, we show that the SWI/SNF chromatin remodeling activity is finely regulated during thymocyte maturation process, especially during thymocyte selections. We found that TCR signaling directly down-regulates mBRG1 and SWI3-related gene, the core components of murine SWI/SNF complex, during thymocyte maturation. Constitutive expression of SWI3-related gene in developing thymocytes attenuated the down-regulation of the SWI/SNF complex and resulted in a change in the expression of genes such as linker for activation of T cells and casitas B lineage lymphoma, which affected the TCR-mediated intracellular signaling pathway. The defects in TCR signaling resulted in the disruption of both positive and negative selections in specific TCR transgenic mice systems. Our results state, for the first time, that the chromatin remodeling activity needs to be finely controlled for proper thymocyte selection and maturation processes.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Chromatin/genetics
- Chromatin/metabolism
- Chromosomal Proteins, Non-Histone/antagonists & inhibitors
- Chromosomal Proteins, Non-Histone/physiology
- Down-Regulation/genetics
- Down-Regulation/immunology
- Female
- Growth Inhibitors/antagonists & inhibitors
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/genetics
- Male
- Mice
- Mice, Inbred A
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/antagonists & inhibitors
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/physiology
- Repressor Proteins/antagonists & inhibitors
- Repressor Proteins/biosynthesis
- Repressor Proteins/genetics
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Trans-Activators/antagonists & inhibitors
- Trans-Activators/biosynthesis
- Trans-Activators/genetics
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Kyoo Y Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, and Research Center for Functional Cellulomics, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
320
|
Abstract
This review describes a body of work on computational immune systems that behave analogously to the natural immune system. These artificial immune systems (AIS) simulate the behavior of the natural immune system and in some cases have been used to solve practical engineering problems such as computer security. AIS have several strengths that can complement wet lab immunology. It is easier to conduct simulation experiments and to vary experimental conditions, for example, to rule out hypotheses; it is easier to isolate a single mechanism to test hypotheses about how it functions; agent-based models of the immune system can integrate data from several different experiments into a single in silico experimental system.
Collapse
Affiliation(s)
- Stephanie Forrest
- Department of Computer Science, University of New Mexico, Albuquerque, NM 87131, USA.
| | | |
Collapse
|
321
|
Abstract
We review recent advances toward a comprehensive mathematical theory of T-cell immunity. A key insight is that the efficacy of the T-cell response is best analyzed in terms of T-cell receptor (TCR) avidity and the distribution of this avidity across the TCR repertoire (the 'avidity spectrum'). Modification of this avidity spectrum by a wide range of tuning and tolerance mechanisms allows the system to adapt cross-reactivity and specificity to the challenge at hand while avoiding inappropriate responses against non-pathogenic cells and tissues. Theoretical models relate molecular kinetic parameters and cellular properties to systemic level statistics such as avidity spectra. Such bridge equations are crucial for rational clinical manipulation of T cells at the molecular level.
Collapse
Affiliation(s)
- Hugo A van den Berg
- Warwick Systems Biology Centre, Mathematics Institute, University of Warwick, Coventry, UK.
| | | |
Collapse
|
322
|
Cai G, Hafler DA. Multispecific responses by T cells expanded by endogenous self-peptide/MHC complexes. Eur J Immunol 2007; 37:602-12. [PMID: 17304631 DOI: 10.1002/eji.200636787] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The paradox of autoreactivity to self-peptides in physiological as opposed to pathological immune responses is not well understood. Here, we directly examined the human T cell response to endogenous self-peptides in a series of healthy subjects. CFSE-labeled T cells were stimulated with unmanipulated antigen-presenting cells containing endogenous self-antigen, and the resulting CD4+ populations entering into cell cycle (CFSE(low)) or non-proliferating CD4+ cells (CFSE(high)) were single-cell sorted, cloned and screened against a panel of self-antigens and microbial recall antigens to interrogate their antigen reactivity. The percentage of CD4+ T cells entering cell cycle in response to self-peptide/MHC was calculated to be 0.04%, and entry into cell cycle was dependent upon CD28 costimulation. Clones derived from CFSE(low) T cells exhibited significantly greater cross-reactivity to multiple antigens than CFSE(high) clones or other CD4+ clones generated after microbial antigen stimulation. Sequencing the TCRbeta chains indicated that CFSE(low) clones were indeed clonal. These data demonstrate that T cell clones generated on stimulation by endogenous self-peptides exhibit a high degree of multispecificity, and we speculate that their multispecificity is based upon recognition of shared-backbone MHC determinants.
Collapse
Affiliation(s)
- Guifang Cai
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
323
|
Liston A, Hardy K, Pittelkow Y, Wilson SR, Makaroff LE, Fahrer AM, Goodnow CC. Impairment of organ-specific T cell negative selection by diabetes susceptibility genes: genomic analysis by mRNA profiling. Genome Biol 2007; 8:R12. [PMID: 17239257 PMCID: PMC1839132 DOI: 10.1186/gb-2007-8-1-r12] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 10/23/2006] [Accepted: 01/21/2007] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND T cells in the thymus undergo opposing positive and negative selection processes so that the only T cells entering circulation are those bearing a T cell receptor (TCR) with a low affinity for self. The mechanism differentiating negative from positive selection is poorly understood, despite the fact that inherited defects in negative selection underlie organ-specific autoimmune disease in AIRE-deficient people and the non-obese diabetic (NOD) mouse strain RESULTS Here we use homogeneous populations of T cells undergoing either positive or negative selection in vivo together with genome-wide transcription profiling on microarrays to identify the gene expression differences underlying negative selection to an Aire-dependent organ-specific antigen, including the upregulation of a genomic cluster in the cytogenetic band 2F. Analysis of defective negative selection in the autoimmune-prone NOD strain demonstrates a global impairment in the induction of the negative selection response gene set, but little difference in positive selection response genes. Combining expression differences with genetic linkage data, we identify differentially expressed candidate genes, including Bim, Bnip3, Smox, Pdrg1, Id1, Pdcd1, Ly6c, Pdia3, Trim30 and Trim12. CONCLUSION The data provide a molecular map of the negative selection response in vivo and, by analysis of deviations from this pathway in the autoimmune susceptible NOD strain, suggest that susceptibility arises from small expression differences in genes acting at multiple points in the pathway between the TCR and cell death.
Collapse
Affiliation(s)
- Adrian Liston
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Kristine Hardy
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Yvonne Pittelkow
- Mathematical Sciences Institute, The Australian National University, Canberra, ACT 2601, Australia
| | - Susan R Wilson
- Mathematical Sciences Institute, The Australian National University, Canberra, ACT 2601, Australia
| | - Lydia E Makaroff
- Biochemistry and Molecular Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Aude M Fahrer
- Biochemistry and Molecular Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Christopher C Goodnow
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
- The Australian Phenomics Facility, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
324
|
Abstract
Transplantation is an acceptable therapy for failing organs, however, the balance between prevention of acute rejection and immunosuppressant-induced toxicity remains elusive. Organ transplantation from a genetically disparate donor induces an immune response toward donor antigens in the recipient. An uncontrolled cumulative effect of these responses may jeopardize the recipient's life and destroy the grafted tissue. The donor antigen in the form of passenger leukocytes from the allograft migrating to the organized lymphoid collection is a prerequisite for initiation of acute rejection. In the host lymphoid tissue donor-specific dendritic cells primed with donor peptide activate naïve CD4 helper T cells which in turn activate effector CD8 T-cell clones through the release of cytokines. Activated effector CD8 cells return to the graft and augment destructive activity with the help of adhesive molecules and perforin. This seems to be the mechanism of adaptive immunity to destroy viral pathogens; the pattern of allograft injury is not much different. Adaptation and tolerance are based on the principle of exhaustion of donor-specific immune responses by an activation-deletion-exhaustion pathway.
Collapse
Affiliation(s)
- H L Trivedi
- Institute of Kidney Diseases and Transplantation Sciences, Gulabben Rasiklal Doshi and Kamlaben Mafatlal Mehta Institute, Research Center and Institute of Civil Hospital Campus, Asarwa Ahmedabad, Gujarat, India.
| |
Collapse
|
325
|
O'Sullivan B, Thompson A, Thomas R. NF-kappa B as a therapeutic target in autoimmune disease. Expert Opin Ther Targets 2007; 11:111-22. [PMID: 17227228 DOI: 10.1517/14728222.11.2.111] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
NF-kappaB transmits signals from the cell surface to the nucleus. Signaling through cell surface receptors to activate NF-kappaB and mitogen-activated protein kinases through adaptor molecules is of critical importance to survival and activation of all cells in the body, including those regulating innate and adaptive immunity. As such, NF-kappaB is a key signaling component in autoimmunity and an attractive target for autoimmune disease therapy. However, given its global importance, targeting NF-kappaB tends to be immunosuppressive. In this review, the authors discuss the roles played by NF-kappaB in autoimmunity, drugs which target it, and complexities which need to be addressed to improve the use of NF-kappaB as a target. Finally, the authors highlight some novel approaches that are likely to be important in the next generation of NF-kappaB therapies.
Collapse
Affiliation(s)
- Brendan O'Sullivan
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland, 4102, Australia
| | | | | |
Collapse
|
326
|
Cortez-Gonzalez X, Zanetti M. Telomerase immunity from bench to bedside: round one. J Transl Med 2007; 5:12. [PMID: 17324292 PMCID: PMC1839079 DOI: 10.1186/1479-5876-5-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2006] [Accepted: 02/26/2007] [Indexed: 11/10/2022] Open
Abstract
Telomerase, a reverse transcriptase primarily devoted to the elongation of telomeres in mammalian cells, is also the first bona fide common tumor antigen. In fact, telomerase is over-expressed in > 85% of tumor cells irrespective of origin and histological type. In the past seven years, there has been considerable interest in assessing telomerase as substrate for vaccination in cancer patients to induce CD8 T cell responses. Because the activation of T cells is restricted by the MHC molecules on antigen presenting cells or tumor cells, the identification of telomerase peptides immunogenic for humans is tightly linked with HLA types. To date, a handful of peptides have been identified through a variety of screening procedures, including bioinformatics prediction, in vivo immunization of HLA transgenic mice, in vitro immunization of PBMC from normal donors and cancer patients, and processing in human tumor cells. Currently, there exist putative peptides for five major HLA types (A2, A1, A3, A24 and B7). Due to the complexity of the HLA system, trials have been performed focusing on the most prevalent HLA type, HLA-A2. Here, we summarize this collective effort and highlight results obtained in Phase 1 trials including a Phase 1 trial performed at the UCSD Cancer Center.
Collapse
Affiliation(s)
- Xochtil Cortez-Gonzalez
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0837, USA.
| | | |
Collapse
|
327
|
Bharat A, Fields RC, Trulock EP, Patterson GA, Mohanakumar T. Induction of IL-10 suppressors in lung transplant patients by CD4+25+ regulatory T cells through CTLA-4 signaling. THE JOURNAL OF IMMUNOLOGY 2007; 177:5631-8. [PMID: 17015751 DOI: 10.4049/jimmunol.177.8.5631] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell-mediated autoimmunity to collagen V (col-V), a sequestered yet immunogenic self-protein, can induce chronic lung allograft rejection in rodent models. In this study we characterized the role of CD4+ CD25+ regulatory T cells (Tregs) in regulating col-V autoimmunity in human lung transplant (LT) recipients. LT recipients revealed a high frequency of col-V-reactive, IL-10-producing CD4+ T cells (T IL-10 cells) with low IL-2-, IFN-gamma-, IL-5-, and no IL-4-producing T cells. These T(IL-10) cells were distinct from Tregs because they lacked constitutive expression of both CD25 and Foxp3. Expansion of T IL-10 cells during col-V stimulation in vitro involved CTLA-4 on Tregs, because both depleting and blocking Tregs with anti-CTLA4 F(ab')2 mAbs resulted in loss of T IL-10 cells with a concomitant increase in IFN-gamma producing Th1 cells (TIFN-gamma cells). A Transwell culture of col-V-specific T IL-10 cells with Th1 cells (those generated in absence of Tregs) from the same patient resulted in marked inhibition of IFN-gamma and proliferation of T(IFN-gamma) cells, which was reversed by neutralizing IL-10. Furthermore, the T IL-10 cells were HLA class II restricted because blocking HLA class II on APCs resulted in the loss of IL-10 production. Chronic lung allograft rejection was associated with the loss of Tregs with a concomitant decrease in T IL-10 cells and an increase in T IFN-gamma cells. We conclude that LT patients have col-V-specific T cells that can be detected in the peripheral blood. The predominant col-V-specific T cells produce IL-10 that suppresses autoreactive Th1 cells independently of direct cellular contact. Tregs are pivotal for the induction of these "suppressor" T IL-10 cells.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery, Washington University School of Medicine, St. Louis. MO 63110, USA
| | | | | | | | | |
Collapse
|
328
|
Valencia X, Yarboro C, Illei G, Lipsky PE. Deficient CD4+CD25high T Regulatory Cell Function in Patients with Active Systemic Lupus Erythematosus. THE JOURNAL OF IMMUNOLOGY 2007; 178:2579-88. [PMID: 17277168 DOI: 10.4049/jimmunol.178.4.2579] [Citation(s) in RCA: 449] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD4(+)CD25(+) T regulatory cells (Tregs) play an essential role in maintaining immunologic homeostasis and preventing autoimmunity. Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by a loss of tolerance to nuclear components. We hypothesized that altered function of CD4(+)CD25(high) Tregs might play a role in the breakdown of immunologic self-tolerance in patients with SLE. In this study, we report a significant decrease in the suppressive function of CD4(+)CD25(high) Tregs from peripheral blood of patients with active SLE as compared with normal donors and patients with inactive SLE. Notably, CD4(+)CD25(high) Tregs isolated from patients with active SLE expressed reduced levels of FoxP3 mRNA and protein and poorly suppressed the proliferation and cytokine secretion of CD4(+) effector T cells in vitro. In contrast, the expression of FoxP3 mRNA and protein and in vitro suppression of the proliferation of CD4(+) effector T cells by Tregs isolated from inactive SLE patients, was comparable to that of normal individuals. In vitro activation of CD4(+)CD25(high) Tregs from patients with active SLE increased FoxP3 mRNA and protein expression and restored their suppressive function. These data are the first to demonstrate a reversible defect in CD4(+)CD25(high) Treg function in patients with active SLE, and suggest that strategies to enhance the function of these cells might benefit patients with this autoimmune disease.
Collapse
Affiliation(s)
- Xavier Valencia
- Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases/National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
329
|
Abdulahad WH, Stegeman CA, van der Geld YM, Doornbos-van der Meer B, Limburg PC, Kallenberg CGM. Functional defect of circulating regulatory CD4+ T cells in patients with Wegener's granulomatosis in remission. ACTA ACUST UNITED AC 2007; 56:2080-91. [PMID: 17530650 DOI: 10.1002/art.22692] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Accumulating data support the role of regulatory T cells, a subset of CD4+ T cells that expresses CD25(high) and the transcription factor forkhead box P3 (FoxP3), in controlling and preventing autoimmunity. In Wegener's granulomatosis (WG), an autoimmune vasculitis, up-regulation of CD25 on circulating CD4+ T cells has been observed, even in patients in remission. The objective of this study was to test whether the frequency and/or function of Treg cells from WG patients in remission are disturbed. METHODS Peripheral blood mononuclear cells were freshly isolated from 52 WG patients in remission and from 27 age- and sex-matched healthy control subjects. The proportion of circulating Treg cells was assessed by flow cytometry using CD4, CD25, FoxP3, and CD45RO markers. Anergy and suppressive function of CD25(high),CD4+ T cells were determined using polyclonal stimulants and coculture assay in 10 WG patients in remission and in 10 age- and sex-matched healthy controls. RESULTS In WG patients, a significant increase was observed in the percentage of circulating CD25(high),CD4+ and CD25(low),CD4+ T cells, whereas CD25-,CD4+ T cells were decreased, as compared with healthy controls. Among circulating CD4+ T cells, an expanded percentage of Treg cells (CD25(high),FoxP3+) with memory phenotype was present in WG patients. However, when the suppressive function of CD25(high),CD4+ T cells was tested, CD25(high),CD4+ T cells from WG patients showed diminished or absent suppression of responder T cell proliferation. The impaired suppression was not due to responder cell resistance (as shown by crisscross experiments with T cells from healthy controls) or altered survival of Treg cells. CONCLUSION These data indicate that WG patients in remission have an expanded proportion of Treg cells that are functionally defective. This observation may be relevant to the development and relapsing course of this autoimmune vasculitis.
Collapse
|
330
|
Abstract
Cell therapy, pioneered for the treatment of malignancies in the form of bone marrow transplantation, has subsequently been tested and successfully employed in autoimmune diseases. Autologous haemopoietic stem cell transplantation (HSCT) has become a curative option for conditions with very poor prognosis such as severe forms of scleroderma, multiple sclerosis, and lupus, in which targeted therapies have little or no effect. The refinement of the conditioning regimens has virtually eliminated transplant-related mortality, thus making HSCT a relatively safe choice. Although HSCT remains a nonspecific approach, the knowledge gained in this field has led to the identification of new avenues. In fact, it has become evident that the therapeutic efficacy of HSCT cannot merely be the consequence of a high-dose immuno-suppression, but rather the result of a resetting of the abnormal immune regulation underlying autoimmune conditions. The identification of professional and nonprofessional immunosuppressive cells and their biological properties is generating a huge interest for their clinical exploitation. Regulatory T cells, found abnormal in several autoimmune diseases, have been proposed as central to achieve long-term remissions. Mesenchymal stem cells of bone marrow origin have more recently been shown not only to be able to differentiate into multiple tissues, but also to exert a potent antiproliferative effect that results in the inhibition of immune responses and prolonged survival of haemopoietic stem cells. All of these potential resources clearly need to be investigated at the preclinical level but support a great deal of enthusiasm for cell therapy of autoimmune diseases.
Collapse
Affiliation(s)
- Francesco Dazzi
- Stem Cell Biology Section, Kennedy Institute of Rheumatology, Imperial College Faculty of Medicine, London, UK.
| | | | | | | |
Collapse
|
331
|
Abstract
The thymic microenvironment provides essential support for the generation of a functional and diverse population of human T cells. In particular, the three-dimensional (3D) thymic architecture contributes to critical cell-cell interactions. We report that thymic stroma, arrayed on a synthetic 3D matrix, supports the development of functional human T cells from hematopoietic precursor cells. Newly generated T cells contain T-cell receptor excision circles and are both fully mature and functional. The coculture of T-cell progenitors with thymic stroma can thus be used to generate de novo functional and diverse T-cell populations. This novel tissue engineered thymic system has biological applications for the study of T-lymphopoiesis and self-tolerance as well as potential therapeutic applications including the immune reconstitution of immunocompromised patients and the induction of tolerance in individuals receiving tissue or organ transplants.
Collapse
Affiliation(s)
- Fabrizio Vianello
- Department of Hematology, University Medical School of Padova, Italy
| | | |
Collapse
|
332
|
Nobori S, Shimizu A, Okumi M, Samelson-Jones E, Griesemer A, Hirakata A, Sachs DH, Yamada K. Thymic rejuvenation and the induction of tolerance by adult thymic grafts. Proc Natl Acad Sci U S A 2006; 103:19081-6. [PMID: 17148614 PMCID: PMC1748180 DOI: 10.1073/pnas.0605159103] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The thymus, the site of origin of T cell immunity, shapes the repertoire of T cell reactivity through positive selection of developing T cells and prevents autoimmunity through negative selection of autoreactive T cells. Previous studies have demonstrated an important role for the thymus not only in central deletional tolerance, but also in the induction of peripheral tolerance by vascularized renal allografts in juvenile miniature swine recipients. The same protocol did not induce tolerance in thymectomized recipients nor in recipients beyond the age of thymic involution. We subsequently reported that vascularized thymic lobe grafts from juvenile donors were capable of inducing tolerance in thymectomized juvenile hosts. However, the important question remained whether aged, involuted thymus could also induce tolerance if transplanted into thymectomized hosts, which, if true, would imply that thymic involution is not an intrinsic property of thymic tissue but is rather determined by host factors extrinsic to the thymus. We report here that aged, involuted thymus transplanted as a vascularized graft into juvenile recipients leads to rejuvenation of both thymic structure and function, suggesting that factors extrinsic to the thymus are capable of restoring juvenile thymic function to aged recipients. We show furthermore that rejuvenated aged thymus has the ability to induce transplant tolerance across class I MHC barriers. These findings indicate that it may be possible to manipulate thymic function in adults to induce transplantation tolerance after the age of thymic involution.
Collapse
Affiliation(s)
- Shuji Nobori
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Akira Shimizu
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Masayoshi Okumi
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Emma Samelson-Jones
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Adam Griesemer
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Atsushi Hirakata
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - David H. Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Kazuhiko Yamada
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
333
|
DeVoss J, Hou Y, Johannes K, Lu W, Liou GI, Rinn J, Chang H, Caspi RR, Caspi R, Fong L, Anderson MS. Spontaneous autoimmunity prevented by thymic expression of a single self-antigen. J Exp Med 2006; 203:2727-35. [PMID: 17116738 PMCID: PMC2118158 DOI: 10.1084/jem.20061864] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 10/26/2006] [Indexed: 12/20/2022] Open
Abstract
The expression of self-antigen in the thymus is believed to be responsible for the deletion of autoreactive T lymphocytes, a critical process in the maintenance of unresponsiveness to self. The Autoimmune regulator (Aire) gene, which is defective in the disorder autoimmune polyglandular syndrome type 1, has been shown to promote the thymic expression of self-antigens. A clear link, however, between specific thymic self-antigens and a single autoimmune phenotype in this model has been lacking. We show that autoimmune eye disease in aire-deficient mice develops as a result of loss of thymic expression of a single eye antigen, interphotoreceptor retinoid-binding protein (IRBP). In addition, lack of IRBP expression solely in the thymus, even in the presence of aire expression, is sufficient to trigger spontaneous eye-specific autoimmunity. These results suggest that failure of thymic expression of selective single self-antigens can be sufficient to cause organ-specific autoimmune disease, even in otherwise self-tolerant individuals.
Collapse
Affiliation(s)
- Jason DeVoss
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Huseby ES, Crawford F, White J, Marrack P, Kappler JW. Interface-disrupting amino acids establish specificity between T cell receptors and complexes of major histocompatibility complex and peptide. Nat Immunol 2006; 7:1191-9. [PMID: 17041605 DOI: 10.1038/ni1401] [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: 06/13/2006] [Accepted: 09/20/2006] [Indexed: 12/21/2022]
Abstract
T cell receptors (TCRs) bind complexes of cognate major histocompatibility complex (MHC) and peptide at relatively low affinities (1-200 microM). Nevertheless, TCR-MHC-peptide interactions are usually specific for the peptide and the allele encoding the MHC. Here we show that to escape thymocyte negative selection, TCRs must interact with many of the side chains of MHC-peptide complexes as 'hot spots' for TCR binding. Moreover, even when the 'parental' side chain did not contribute binding affinity, some MHC-peptide residues contributed to TCR specificity, as amino acid substitutions substantially reduced binding affinity. The presence of such 'interface-disruptive' side chains helps to explain how TCRs generate specificity at low-affinity interfaces and why TCRs often 'accommodate' a subset of amino acids at a given MHC-peptide position.
Collapse
Affiliation(s)
- Eric S Huseby
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Medical and Research Center, Denver, Colorado 80206, USA
| | | | | | | | | |
Collapse
|
335
|
Reibke R, Garbi N, Ganss R, Hämmerling GJ, Arnold B, Oelert T. CD8+ regulatory T cells generated by neonatal recognition of peripheral self-antigen. Proc Natl Acad Sci U S A 2006; 103:15142-7. [PMID: 17008409 PMCID: PMC1622790 DOI: 10.1073/pnas.0602622103] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Indexed: 02/03/2023] Open
Abstract
In comparison with CD4+ regulatory T cells, the generation and function of immunomodulatory CD8+T cells is less well defined. Here we describe the existence of regulatory anti-Kb-specific CD8+ T cells that are rendered tolerant during neonatal life via antigen contact exclusively on keratinocytes. These regulatory T cells maintain tolerance during adulthood as they prevent Kb-specific graft rejection by naïve CD8+ T cells. Third-party immune responses remain unaffected. Up-regulation of TGF-beta1 and granzyme B in the regulatory CD8+ T cell population suggests the involvement of these molecules in common suppressive pathways shared with CD4+ regulatory T cells. In summary, CD8+ regulatory T cells can be induced extrathymically through antigen contact on neonatally accessible parenchymal cells and maintain tolerance throughout adult life.
Collapse
Affiliation(s)
- Roland Reibke
- *Division of Molecular Immunology (D050), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; and
| | - Natalio Garbi
- *Division of Molecular Immunology (D050), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; and
| | - Ruth Ganss
- *Division of Molecular Immunology (D050), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; and
- Laboratory for Cancer Medicine, Western Australian Institute for Medical Research, University of Western Australia, Perth WA 6000, Australia
| | - Günter J. Hämmerling
- *Division of Molecular Immunology (D050), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; and
| | - Bernd Arnold
- *Division of Molecular Immunology (D050), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; and
| | - Thilo Oelert
- *Division of Molecular Immunology (D050), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; and
| |
Collapse
|
336
|
Abstract
Because of the use of somewhat artificial models for the elucidation of negative selection [superantigen, T-cell receptor (TCR) transgenic mice], there is still considerable uncertainty at what stages of T-cell development negative selection can occur and whether it becomes manifest as developmental arrest, lineage diversion, or induction of apoptotic cell death. Here, experimental evidence is reviewed that excludes developmental arrest and lineage diversion as the sole mechanisms of negative selection. The data emphasize that both CD4+ CD8+ double-positive cortical as well as semi-mature, single-positive, medullary thymocytes are targets of deletion in experimental models employing superantigen and TCR transgenic mice with premature as well as 'timely' onset of TCR expression.
Collapse
Affiliation(s)
- Harald von Boehmer
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | | |
Collapse
|
337
|
Hirose S, Jiang Y, Nishimura H, Shirai T. Significance of MHC class II haplotypes and IgG Fc receptors in SLE. ACTA ACUST UNITED AC 2006; 28:163-74. [PMID: 16972051 DOI: 10.1007/s00281-006-0036-1] [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: 07/01/2006] [Accepted: 07/04/2006] [Indexed: 10/24/2022]
Abstract
Systemic lupus erythematosus (SLE) is a systemic antibody-mediated autoimmune disease that develops under the control of multiple susceptibility genes. Genetic studies in murine and human SLE have identified several chromosomal intervals that contain candidate susceptibility genes. However, the ultimate identification of the genes and their roles in disease process need much further investigation. Spontaneous murine SLE models provide useful tools in this respect. In this chapter, we show this line of investigation, particularly focusing on the roles of major histocompatibility complex (MHC) class II and immunoglobulin G Fc receptors (FcgammaRs). The existence of high-affinity autoantibodies is evidence that autoimmunity in SLE is antigen-driven. Thereby, MHC class II haplotypes have been implicated in SLE susceptibility; however, because of the linkage disequilibrium that exists among the class I, II and III genes within the MHC complex, it has been difficult to discriminate the relative contributions of individual loci. On the other hand, the extent of antibody synthesis upon antigen stimulation and associated inflammatory cascades are controlled in several ways by the balance of stimulatory and inhibitory signaling molecules on immune cells. Stimulatory/inhibitory FcgammaRs mediate one such mechanism, and there are reports indicating the association between polymorphic FcgammaRs and SLE. However, as stimulatory and inhibitory FcgammaRs cluster on the telomeric chromosome 1, the absolute contribution of individual genes has been difficult to dissect. In studies of genetic dissection using interval-congenic and intragenic recombinant mouse strains of SLE models, we show evidence and discuss how and to what extent MHC class II molecules and stimulatory/inhibitory FcgammaRs are involved in SLE susceptibility.
Collapse
Affiliation(s)
- Sachiko Hirose
- Department of Pathology, Juntendo University School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | | | | | | |
Collapse
|
338
|
Bund D, Mayr C, Kofler DM, Hallek M, Wendtner CM. Human Ly9 (CD229) as novel tumor-associated antigen (TAA) in chronic lymphocytic leukemia (B-CLL) recognized by autologous CD8+ T cells. Exp Hematol 2006; 34:860-9. [PMID: 16797413 DOI: 10.1016/j.exphem.2006.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 03/28/2006] [Accepted: 04/11/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE CD229, a cell-surface molecule being involved in cell adhesion, is overexpressed in B-CLL cells. In this study we wanted to explore whether CD229 might function as B-CLL-specific tumor-associated antigen (TAA). PATIENTS AND METHODS Autologous, CD229-specific HLA-A2-restricted T cells were identified using IFN-gamma-ELISPOT assays and HLA-A2/dimer-peptide staining after 4 weeks of in vitro culture. RESULTS We were able to expand autologous T cells from 9/11 B-CLL patients using native B-CLL cells as antigen presenting cells (APCs) in 5 cases, whereas for 4 samples an autologous T-cell response could only be evoked by use of CD40L-stimulated B-CLL cells as APCs. The number of CD8+ T cells could be expanded during 4 weeks of in vitro culture with native or CD40L-activated B-CLL cells while the amount of specific T cells recognizing CD229 peptides bound to HLA-A2 dimers increased on average 12-fold (native CLL) and 13-fold (CD40L-activated CLL), respectively. Using IFN-gamma-ELISPOT assays we could demonstrate that the expanded T cells were able to secrete IFN-gamma upon recognition of the antigen. These T cells not only recognized HLA-A0201-binding CD229-derived peptides presented by T2 cells, but also CD229-overexpressing autologous B-CLL cells in an MHC-I-restricted manner. CONCLUSION In summary, CD229 was shown to be naturally processed and presented as TAA in primary B-CLL cells, enabling the expansion of autologous tumor-specific T cells.
Collapse
Affiliation(s)
- Dagmar Bund
- KKG Gene Therapy, GSF-National Research Center for Environment and Health, Munich, Germany
| | | | | | | | | |
Collapse
|
339
|
Marrero I, Huffman D, Kalil J, Sercarz EE, Coelho V. TAP1-/- mice present oligoclonal BV-BJ expansions following the rejection of grafts bearing self antigens. Immunology 2006; 118:461-71. [PMID: 16895555 PMCID: PMC1782321 DOI: 10.1111/j.1365-2567.2006.02387.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Our previous work showed that transporter associated with antigen processing 1 (TAP1)-/- (H-2b) mice rejected grafts from H-2b mice which display a normal density of class I major histocompatibility complex (MHC) molecules at the cell surface. Our results indicated that H-2b molecules themselves may be a target in this kind of rejection and that CD4+ T cells play a major role in this autoreactive process. Our data also suggested that TAP1-/- mice, in addition to the well-recognized phenotype of class I and CD8+ T-cell deficiency, present a functional alteration in their autoreactive CD4+ T-cell repertoires. In this model of inflammatory autoreactivity to modified self, we have analysed T-cell receptor (TCR) V-beta-J-beta (BV-BJ) usage by complementarity determining region 3 (CDR3) length spectratyping in splenocytes from naïve TAP1-/- mice and transplanted TAP1-/- mice that rejected B6 heart grafts or responded to synthetic self H-2Kb peptides. Importantly, oligoclonal T-cell expansions shared by different animals were detected in the peripheral T-cell repertoire of transplanted TAP1-/- mice. Such public expansions were also induced in vitro by H-2Kb peptides, suggesting that dominant class I peptides can induce preferential expansions of restricted T-cell populations during rejection. Some of these public T-cell expansions were also detected in transplanted mice even before in vitro stimulation with peptides, indicating that post-transplantation expansion of these populations had occurred in vivo. The functional activity of these T-cell populations awaits elucidation, as do the underlying mechanisms involved in the inflammatory autoreactive process, in TAP1-/- mice.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP-Binding Cassette Transporters/genetics
- Animals
- Antigen Presentation
- Autoantigens
- Autoimmunity
- CD4-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Complementarity Determining Regions/immunology
- Cytotoxicity, Immunologic
- Graft Rejection/immunology
- H-2 Antigens/immunology
- Heart Transplantation
- In Situ Hybridization, Fluorescence
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Animal
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- Idania Marrero
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Donald Huffman
- Division of Immune Regulation, La Jolla Institute for Allergy and ImmunologySan Diego, CA, USA
| | - Jorge Kalil
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Eli E Sercarz
- Department of Immune Regulation, Torrey Pines Institute for Molecular StudiesSan Diego, CA, USA
| | - Verônica Coelho
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| |
Collapse
|
340
|
Gronski MA, Weinem M. Death pathways in T cell homeostasis and their role in autoimmune diabetes. Rev Diabet Stud 2006; 3:88-95. [PMID: 17487332 PMCID: PMC1783577 DOI: 10.1900/rds.2006.3.88] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
T cell apoptosis is a process necessary for central and peripheral tolerance. It ensures the proper removal of autoreactive T cells during thymic development as well as T cell homeostasis and the downregulation of immune responses against antigens in the periphery. Thus it is essential for the prevention of autoimmunity. Apoptotic pathways can be triggered by intrinsic (mitochondria-based) and extrinsic (receptor-based) stimuli. Both pathways involve a cascade of proteolytic enzymes called caspases whose activation commits the cell to death. In the periphery, autoreactive lymphocytes can be silenced by developmental arrest (anergy), or deleted by programmed cell death (apoptosis) through receptor-based activation-induced cell death (AICD). Central tolerance seems to rely more heavily on the mitochondria-based, T cell receptor (TCR)-stimulated apoptotic pathway, since thymocytes lacking the pro-apoptotic Bcl-2 family member Bim are resistant to TCR-induced apoptosis. Furthermore, defects in the intrinsic pathway of apoptosis may impair clonal deletion of autoreactive T cells. Several animal models exist in which impaired apoptosis results in autoimmunity. Here, we discuss data that suggest defects in T cell apoptosis in type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Matthew A Gronski
- Beirne Carter Center for Immunology Research, University of Virginia, Lane Road, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
341
|
Abstract
BACKGROUND Allergic disease has currently reached epidemic proportions, with a high percentage of individuals in the developed world exhibiting an allergic response after exposure to some common environmental factors. Although new strategies for the treatment and management of allergic diseases have decreased the mortality rate, a high percentage of affected persons still require frequent hospitalization and experience decreased quality of life. METHODS An internet-based literature search was performed for recent contributions on the underlying mechanisms provoking an allergic response and their potential for therapeutic approaches. RESULTS Novel concepts on allergic responses have emerged: allergic disease may result from an imbalance between allergen activation of regulatory T cells and effector T helper 2 cells (Th2), a process in which dendritic cells are key players. Cytokines such as interleukin (IL)-6, IL-21, IL-25, and human thymic stromal lymphopoietin (TSLP) seem to be important contributors in allergic processes. New data on IgE effector responses and on the IgE-independent mechanisms involved in allergic reactions have resolved some unanswered questions about these reactions. CONCLUSIONS These new findings on allergic diseases have important implications for diagnosis and management, with potential improvements in prevention and treatment, which could provide a cure in the future.
Collapse
Affiliation(s)
- M T Montero Vega
- Servicio de Bioquímica-Investigación, Hospital Ramón y Cajal, Madrid, Spain.
| |
Collapse
|
342
|
Franzke A, Hunger JK, Dittmar KEJ, Ganser A, Buer J. Regulatory T-cells in the control of immunological diseases. Ann Hematol 2006; 85:747-58. [PMID: 16871392 DOI: 10.1007/s00277-006-0117-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Accepted: 03/30/2006] [Indexed: 12/22/2022]
Abstract
The immune system is challenged by randomly generated immune receptors that by chance can recognize self-antigens. Immunological tolerance functions as a fundamental concept in the control of a broad spectrum of immune responses not only to autoantigens but also to foreign antigens. During the past decade, CD4+ CD25+ regulatory T-cells (Tregs) have emerged as key players in the development of immunological tolerance. This review will present an update on the current knowledge about the phenotype, function, and clinical relevance of this regulatory T-cell population. The therapeutical potential of Tregs to specifically suppress immune responses in autoimmunity and transplantation and their inhibitory effects in anti-tumor immune responses will be discussed.
Collapse
Affiliation(s)
- A Franzke
- Department of Hematology, Hemostaseology and Oncology, Hannover Medical School, Hannover, Germany,
| | | | | | | | | |
Collapse
|
343
|
Matsutani T, Ohmori T, Ogata M, Soga H, Yoshioka T, Suzuki R, Itoh T. Alteration of T-cell receptor repertoires during thymic T-cell development. Scand J Immunol 2006; 64:53-60. [PMID: 16784491 DOI: 10.1111/j.1365-3083.2006.01776.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The majority of thymocytes die in the thymus, whereas small populations of T cells that are able to specifically recognize an antigen are considered to survive. Although the thymic selection is thought to have a profound effect on T-cell receptor (TCR) repertoire, little is known how TCR repertoire is formed during the thymocyte developmental process. We examined TCRalpha- and beta-chain variable regions (TCRAV and TCRBV) repertoire in thymic T-cell subpopulations from mice bearing different major histocompatibility (MHC) haplotypes. In Balb/c mice, but not C57BL/6, remarkable alterations of the TCR repertoire were observed in mature T-cell subpopulations as previously reported. In contrast, there were no significant differences of TCRBV repertoire between DN3 (CD25(+)CD44(-)) and DN4 (CD25(-)CD44(-)), and between DN4 and DP. These results suggest that (1) TCR repertoire of mature T cells was formed mainly under the influence of endogenous superantigens, while MHC haplotypes played the least role; (2) the 'beta-selection' process during immature stages had little impact on TCRBV repertoire formation; and (3) TCR repertoire in immature T-cell subpopulations was extremely similar between different strains of mice. We thus consider that pre-selection TCR repertoire in immature T cells could be determined by some genetic factors conserved among different strains.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Differentiation
- Clonal Deletion
- Female
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Male
- Mammary Tumor Virus, Mouse/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Polymerase Chain Reaction/methods
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Species Specificity
- Spleen/cytology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- Thymus Gland/cytology
Collapse
Affiliation(s)
- T Matsutani
- Division of Immunology and Embryology, Department of Cell Biology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | |
Collapse
|
344
|
Chao DL, Davenport MP, Forrest S, Perelson AS. The effects of thymic selection on the range of T cell cross-reactivity. Eur J Immunol 2006; 35:3452-9. [PMID: 16285012 PMCID: PMC1857316 DOI: 10.1002/eji.200535098] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Based on the results of a computational model of thymic selection, we propose a mechanism that produces the observed wide range of T cell cross-reactivity. The model suggests that the cross-reactivity of a T cell that survives thymic selection is correlated with its affinity for self peptides. In order to survive thymic selection, a T cell with low affinity for all self peptides expressed in the thymus must have high affinity for major histocompatibility complex (MHC), which makes it highly cross-reactive. A T cell with high affinity for any self peptide must have low MHC affinity to survive selection, which makes it highly specific for its cognate peptide. Our model predicts that (1) positive selection reduces by only 17% the number of T cells that can detect any given foreign peptide, even though it eliminates over 95% of pre-selection cells; (2) negative selection decreases the average cross-reactivity of the pre-selection repertoire by fivefold; and (3) T cells responding to foreign peptides similar to self peptides will have a lower average cross-reactivity than cells responding to epitopes dissimilar to self.
Collapse
Affiliation(s)
- Dennis L Chao
- Fred Hutchinson Cancer Research Center, Seattle, USA
| | | | | | | |
Collapse
|
345
|
Siemionow M, Izycki D, Ozer K, Ozmen S, Klimczak A. Role of thymus in operational tolerance induction in limb allograft transplant model. Transplantation 2006; 81:1568-76. [PMID: 16770246 DOI: 10.1097/01.tp.0000209508.37345.82] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In this study, we evaluated the role of host thymus in tolerance induction in composite tissue allografts (CTA) across major histocompatibility complex (MHC) barrier during a 7-day alphabeta- T-cell receptor (TCR)/ cyclosporine A (CsA) protocol. MATERIALS AND METHODS A total of 62 limb allograft transplants were studied. Euthymic (group A) and thymectomized (group B) Lewis recipients (LEW, RT1(1)) received vascularized hind-limb allografts from hybrid Lewis x Brown-Norway (F1), (LBN, RT1(1+n)) donors. Mixed lymphocyte reaction (MLR) and skin grafting assessed donor-specific tolerance in vitro and in vivo, respectively. Flow cytometry determined the efficacy of immunosuppressive protocols and the presence of donor-specific chimerism. Immunocytochemistry revealed the presence of donor-specific cells in the lymphoid organs of recipients. RESULTS Isograft transplants survived indefinitely. For thymectomized rats, the median survival time (MST) of limb allograft in non-treated recipients was 7 days; monotherapy with alphabeta-TCR extended MST to 16 days, and CsA therapy extended it to 30 days. Using the alphabeta-TCR/CsA protocol, the MST of allografts was 51 days. For euthymic rats, the MST of limb allograft in non-treated recipients was 7 days; monotherapy with alphabeta-TCR or CsA extended MST to 13 or 22 days, respectively. Treatment with alphabeta-TCR/CsA resulted in indefinite allografts survival (MST=370 days). MLR and skin grafting confirmed donor-specific tolerance in euthymic recipients. Flow cytometry showed stable chimerism in the euthymic rats and transient chimerism in thymectomized limb recipients. Immunoperoxidase staining revealed the persistence of donor-derived cells in the lymphoid tissues of euthymic recipients. CONCLUSION We found that the presence of thymus was imperative for the induction of donor-specific tolerance in rat hind-limb composite tissue allografts using a alphabeta-TCR/CsA protocol.
Collapse
MESH Headings
- Animals
- Chimerism
- Cyclosporine/therapeutic use
- Flow Cytometry
- Hindlimb/pathology
- Hindlimb/physiopathology
- Hindlimb/transplantation
- Immunohistochemistry
- Immunosuppressive Agents/therapeutic use
- Lymphocyte Culture Test, Mixed
- Lymphoid Tissue/chemistry
- Lymphoid Tissue/pathology
- Lymphoid Tissue/physiopathology
- Major Histocompatibility Complex/immunology
- Male
- Rats
- Rats, Inbred BN
- Rats, Inbred Lew
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Antigen, T-Cell, alpha-beta/therapeutic use
- Survival Rate
- Thymectomy
- Thymus Gland/immunology
- Thymus Gland/surgery
- Time Factors
- Transplantation Tolerance/drug effects
- Transplantation Tolerance/immunology
- Transplantation, Homologous/immunology
- Transplantation, Homologous/pathology
Collapse
Affiliation(s)
- Maria Siemionow
- Department of Plastic Surgery, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | | | | | | | |
Collapse
|
346
|
Bharat A, Fields RC, Mohanakumar T. Regulatory T cell-mediated transplantation tolerance. Immunol Res 2006; 33:195-212. [PMID: 16461998 DOI: 10.1385/ir:33:3:195] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The existence of naturally occurring regulatory T cells in normal hosts and their pivotal role in maintaining both auto- and allo-tolerance have direct implications on the therapy of autoimmune disorders and for achieving immunosuppression-free allotransplantation. Among the various forms of regulatory T cells described, CD4(+)CD25(+) T cells have emerged as one of the most potent tolerogenic subsets. In this review, we discuss the molecular basis of development and function of these regulatory T cells and their potential role in the context of chronic lung allograft rejection.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
347
|
Li MO, Wan YY, Sanjabi S, Robertson AKL, Flavell RA. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol 2006; 24:99-146. [PMID: 16551245 DOI: 10.1146/annurev.immunol.24.021605.090737] [Citation(s) in RCA: 1717] [Impact Index Per Article: 90.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is a potent regulatory cytokine with diverse effects on hemopoietic cells. The pivotal function of TGF-beta in the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. In addition, TGF-beta controls the initiation and resolution of inflammatory responses through the regulation of chemotaxis, activation, and survival of lymphocytes, natural killer cells, dendritic cells, macrophages, mast cells, and granulocytes. The regulatory activity of TGF-beta is modulated by the cell differentiation state and by the presence of inflammatory cytokines and costimulatory molecules. Collectively, TGF-beta inhibits the development of immunopathology to self or nonharmful antigens without compromising immune responses to pathogens. This review highlights the findings that have advanced our understanding of TGF-beta in the immune system and in disease.
Collapse
Affiliation(s)
- Ming O Li
- Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | | | | | | | |
Collapse
|
348
|
Abstract
The 620W allelic variant of the intracellular tyrosine phosphatase, PTPN22, is associated with a number of different autoimmune disorders, and this provides direct evidence for common mechanisms underlying many of these diseases. The associated allele appears to influence thresholds for T cell receptor signaling, and a variety of disease models involving both central and peripheral tolerance can be proposed. However, given the fact that PTPN22 is expressed in a variety of immunologically relevant cell types, the precise mechanisms for these associations remain unclear. In general, the PTPN22 620W allele appears to play a role in autoimmune disorders that have a prominent humoral component, suggesting that further investigation of PTPN22 activity in B cells will be useful. From a genetic perspective, the data highlights the genetic heterogeneity underlying autoimmunity in different ethnic groups.
Collapse
Affiliation(s)
- Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, North Shore LIJ Health System, 350 Community Drive, Manhasset, NY 11030, United States.
| | | | | | | |
Collapse
|
349
|
Stahl D. Warm Autoimmune Hemolytic Anemia: A Clinical Model to Study Mechanisms of Immunoregulation*. Transfus Med Hemother 2006. [DOI: 10.1159/000091108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
350
|
Valencia X, Stephens G, Goldbach-Mansky R, Wilson M, Shevach EM, Lipsky PE. TNF downmodulates the function of human CD4+CD25hi T-regulatory cells. Blood 2006; 108:253-61. [PMID: 16537805 PMCID: PMC1895836 DOI: 10.1182/blood-2005-11-4567] [Citation(s) in RCA: 627] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD4+CD25+ T-regulatory cells (Tregs) play an essential role in maintaining immunologic homeostasis and preventing autoimmunity. However, little is known about the exogenous factors that regulate their differentiation and function. Here, we report that TNF inhibits the suppressive function of both naturally occurring CD4+CD25+ Tregs and TGFbeta1-induced CD4+CD25+ T-regulatory cells. The mechanism of this inhibition involves signaling through TNFRII that is constitutively expressed selectively on unstimulated Tregs and that is up-regulated by TNF. TNF-mediated inhibition of suppressive function is related to a decrease in FoxP3 mRNA and protein expression by the Tregs. Notably, CD4+CD25hi Tregs isolated from patients with active rheumatoid arthritis (RA) expressed reduced levels of FoxP3 mRNA and protein and poorly suppressed the proliferation and cytokine secretion of CD4+ effector T cells in vitro. Treatment with anti-TNF antibody (infliximab) increased FOXP3 mRNA and protein expression by CD4+CD25hi Tregs and restored their suppressive function. Thus, TNF has a novel action in modulating autoimmunity, by inhibiting CD4+CD25+ Treg activity.
Collapse
Affiliation(s)
- Xavier Valencia
- Autoimmunity Branch, the Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases/National Institute of Health (NIAMS/NIH), Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|