101
|
Denoeud J, Moser M. Role of CD27/CD70 pathway of activation in immunity and tolerance. J Leukoc Biol 2010; 89:195-203. [PMID: 20699361 DOI: 10.1189/jlb.0610351] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The CD70/CD27 axis has gained increasing interest among the immunologists, because of its capacity to regulate immunity versus tolerance. Recent studies clearly show that expression of CD70 may prevent tolerance induced by antigen presentation in the steady-state, i.e., by nonactivated DCs. In addition, CD27 signaling appears critical for T cell expansion and survival and therefore, induction of long-term memory. It contributes to germinal center formation, B cell activation, and production of neutralizing antibodies but can also be subverted by viruses, in particular, during chronic infections. The potential role of the CD27/CD70 pathway in the course of inflammatory diseases, as in EAE, arthritis, and inflammatory bowel disease models, suggests that CD70 may be a target for immune intervention. Conversely, the potency of costimulation through CD27 suggests that the CD27/CD70 axis could be exploited for the design of anti-cancer vaccines.
Collapse
Affiliation(s)
- Julie Denoeud
- Departement de Biologie Moleculaire,Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | | |
Collapse
|
102
|
Kohler ME, Hallett WH, Chen QR, Khan J, Johnson BD, Orentas RJ. Early expression of stem cell-associated genes within the CD8 compartment after treatment with a tumor vaccine. Cell Immunol 2010; 265:65-73. [PMID: 20692654 PMCID: PMC2935508 DOI: 10.1016/j.cellimm.2010.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 05/26/2010] [Accepted: 07/14/2010] [Indexed: 10/19/2022]
Abstract
Using a mouse neuroblastoma cell line, we have demonstrated that vaccination of tumor-free mice with a cell-based vaccine leads to productive immunity and resistance to tumor challenge, while vaccination of tumor-bearing mice does not. The T cell immunity induced by this vaccine, as measured by in vitro assays, is amplified by the depletion of Treg. Our goal is to understand this barrier to the development of protective cellular immunity. mRNA microarray analyses of CD8(+) T cells from naïve or tumor-bearing mice undergoing vaccination were carried out with or without administering anti-CD25 antibody. Gene-expression pathway analysis revealed the presence of CD8(+) T cells expressing stem cell-associated genes early after induction of productive anti-tumor immunity in tumor-free mice, prior to any phenotypic changes, but not in tumor-bearing mice. These data demonstrate that early after the induction of productive immune response, cells within the CD8(+) T cell compartment adopt a stem cell-related genetic phenotype that correlates with increased anti-tumor function.
Collapse
Affiliation(s)
- M. Eric Kohler
- Dept. Pediatrics, Section of Hematology Oncology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226
| | - William H.D. Hallett
- Dept. Pediatrics, Section of Hematology Oncology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226
| | - Qing-Rong Chen
- Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Advanced Technology Center, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Javed Khan
- Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Advanced Technology Center, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Bryon D. Johnson
- Dept. Pediatrics, Section of Hematology Oncology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226
| | - Rimas J. Orentas
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., 1W3840, Bethesda, MD 20892, T: 414-4565897, F: 414-456-7010
| |
Collapse
|
103
|
Abstract
CD28 costimulation regulates a wide range of cellular processes, from proliferation and survival to promoting the differentiation of specialized T-cell subsets. Since first being identified over 20 years ago, CD28 has remained a subject of intense study because of its profound consequences on T cell function and its potential for therapeutic manipulation. In this review we highlight the signaling cascades initiated by the major signaling motifs in CD28, focusing on PI-3 kinase-dependent and -independent pathways and how these are linked to specific cellular outcomes. Recent studies using gene targeted knockin mice have clarified the relative importance of these motifs on in vivo immune responses; however, much remains to be elucidated. Understanding the mechanism behind costimulation holds great potential for development of new clinically relevant reagents, a fact beginning to be realized with the advent of drugs that prevent CD28 ligation and signaling.
Collapse
Affiliation(s)
- Jonathan S Boomer
- Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | |
Collapse
|
104
|
Spatiotemporal control of cyclic AMP immunomodulation through the PKA-Csk inhibitory pathway is achieved by anchoring to an Ezrin-EBP50-PAG scaffold in effector T cells. FEBS Lett 2010; 584:2681-8. [PMID: 20420835 DOI: 10.1016/j.febslet.2010.04.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Revised: 04/16/2010] [Accepted: 04/20/2010] [Indexed: 11/23/2022]
Abstract
A variety of immunoregulatory signals to effector T cells from monocytes, macrophages and regulatory T cells act through cyclic adenosine monophosphate. In the effector T cell, the protein kinase A (PKA) type I isoenzyme localizes to lipid rafts during T cell activation and modulates directly the proximal events that take place after engagement of the T cell receptor. The most proximal target for PKA phosphorylation is C-terminal Src kinase (Csk), which initiates a negative signal pathway that fine-tunes the T cell activation process. The A kinase anchoring protein Ezrin colocalizes PKA and Csk by forming a supramolecular signaling complex consisting of PKA, Ezrin, Ezrin/radixin/moesin (ERM) binding protein of 50 kDa (EBP50), phosphoprotein associated with glycosphingolipid-enriched membrane microdomains (GEMs) (PAG) and Csk.
Collapse
|
105
|
Pharmacodynamic monitoring of calcineurin inhibition therapy: principles, performance, and perspectives. Ther Drug Monit 2010; 32:3-10. [PMID: 20009796 DOI: 10.1097/ftd.0b013e3181c0eecb] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The calcineurin inhibitors (CNIs) cyclosporin A and tacrolimus are immunosuppressive drugs used extensively in allograft recipients. These drugs show large interindividual pharmacokinetic variation and are associated with severe adverse affects, including nephrotoxicity and cardiovascular disease. In current practice, CNIs are combined with other immunosuppressive drugs such as steroids and mycophenolate mofetil. Dosage is titrated based on blood concentration measurement. For further optimization of calcineurin (CN) inhibition therapy, new monitoring strategies are required. Pharmacodynamic-monitoring strategies constitute novel approaches for optimization of CNIs therapy. This review focuses on the general aspects of immunosuppressive drug pharmacodynamic monitoring and describes the methodologies used for monitoring CN inhibition therapy. Two different types of pharmacodynamic-monitoring strategies can be distinguished: (1) enzymatic strategies, which monitor inhibition of drug-target enzyme activity, and (2) immunologic strategies, which measure cellular responsiveness after in vitro simulated immunologic responses. Enzymatic tests are drug type-specific markers in which CN activity is directly determined. Immunologic strategies measure immune responsiveness at several levels, such as mRNA transcripts (intracellular) concentrations/excretion of cytokines, expression of surface activation markers, and cell proliferation. This review also discusses analytical issues and clinical experience with these techniques. The call for new methodologies to evaluate immunosuppressive therapy has led to the development of a large variety of pharmacodynamic-monitoring strategies. The first reports of their clinical relevance are available, but further understanding of the analytical and clinical variables involved are required for the development of accurate, reproducible, and clinically relevant markers.
Collapse
|
106
|
Falvo JV, Tsytsykova AV, Goldfeld AE. Transcriptional control of the TNF gene. ACTA ACUST UNITED AC 2010; 11:27-60. [PMID: 20173386 DOI: 10.1159/000289196] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The cytokine TNF is a critical mediator of immune and inflammatory responses. The TNF gene is an immediate early gene, rapidly transcribed in a variety of cell types following exposure to a broad range of pathogens and signals of inflammation and stress. Regulation of TNF gene expression at the transcriptional level is cell type- and stimulus-specific, involving the recruitment of distinct sets of transcription factors to a compact and modular promoter region. In this review, we describe our current understanding of the mechanisms through which TNF transcription is specifically activated by a variety of extracellular stimuli in multiple cell types, including T cells, B cells, macrophages, mast cells, dendritic cells, and fibroblasts. We discuss the role of nuclear factor of activated T cells and other transcription factors and coactivators in enhanceosome formation, as well as the contradictory evidence for a role for nuclear factor kappaB as a classical activator of the TNF gene. We describe the impact of evolutionarily conserved cis-regulatory DNA motifs in the TNF locus upon TNF gene transcription, in contrast to the neutral effect of single nucleotide polymorphisms. We also assess the regulatory role of chromatin organization, epigenetic modifications, and long-range chromosomal interactions at the TNF locus.
Collapse
Affiliation(s)
- James V Falvo
- Immune Disease Institute and Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | | | | |
Collapse
|
107
|
Holz LE, Warren A, Le Couteur DG, Bowen DG, Bertolino P. CD8+ T cell tolerance following antigen recognition on hepatocytes. J Autoimmun 2010; 34:15-22. [DOI: 10.1016/j.jaut.2009.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 08/13/2009] [Indexed: 02/02/2023]
|
108
|
Bjørgo E, Taskén K. Novel mechanism of signaling by CD28. Immunol Lett 2010; 129:1-6. [PMID: 20123110 DOI: 10.1016/j.imlet.2010.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
Ligation of both the T cell receptor (TCR) and the CD28 receptor is required for full T cell activation to occur. Engagement of the TCR in primary T cells is followed by rapid cAMP production in lipid rafts and activation of the cAMP-protein kinase A (PKA)-Csk pathway inhibiting proximal T cell signaling. However, CD28 stimulation leads to recruitment of a beta-arrestin/phosphodiesterase-4 (PDE4) complex to rafts, resulting in down-regulation of cAMP levels. Thus, the activities of both PKA and PDE4 seem to be important for regulation of TCR-induced signaling and T cell function. This review will focus on the novel mechanism whereby CD28 through PI3K regulates recruitment of a PKB/beta-arrestin/PDE4 complex thereby allowing a complete T cell activation to proceed.
Collapse
Affiliation(s)
- Elisa Bjørgo
- The Biotechnology Centre of Oslo and Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, P.O. Box 1125, Blindern, N-0317 Oslo, Norway
| | | |
Collapse
|
109
|
Cross talk between phosphatidylinositol 3-kinase and cyclic AMP (cAMP)-protein kinase a signaling pathways at the level of a protein kinase B/beta-arrestin/cAMP phosphodiesterase 4 complex. Mol Cell Biol 2010; 30:1660-72. [PMID: 20086095 DOI: 10.1128/mcb.00696-09] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Engagement of the T-cell receptor (TCR) in human primary T cells activates a cyclic AMP (cAMP)-protein kinase A (PKA)-Csk inhibitory pathway that prevents full T-cell activation in the absence of a coreceptor stimulus. Here, we demonstrate that stimulation of CD28 leads to recruitment to lipid rafts of a beta-arrestin/phosphodiesterase 4 (PDE4) complex that serves to degrade cAMP locally. Redistribution of the complex from the cytosol depends on Lck and phosphatidylinositol 3-kinase (PI3K) activity. Protein kinase B (PKB) interacts directly with beta-arrestin to form part of the supramolecular complex together with sequestered PDE4. Translocation is mediated by the PKB plextrin homology (PH) domain, thus revealing a new role for PKB as an adaptor coupling PI3K and cAMP signaling. Functionally, PI3K activation and phosphatidylinositol-(3,4,5)-triphosphate (PIP3) production, leading to recruitment of the supramolecular PKB/beta-arrestin/PDE4 complex to the membrane via the PKB PH domain, results in degradation of the TCR-induced cAMP pool located in lipid rafts, thereby allowing full T-cell activation to proceed.
Collapse
|
110
|
Abstract
Lymphocyte development and function are regulated by tyrosine kinase and G-protein coupled receptors. Each of these classes of receptors activates phosphoinositide 3-kinase (PI3K). In this chapter, we summarize current understanding of how PI3K contributes to key aspects of the adaptive immune system.
Collapse
Affiliation(s)
- Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, the Babraham Institute, Cambridge, UK Phone: 44-1223-49-6573 Fax: 44-1223-49-6023
| | - David A. Fruman
- Department of Molecular Biology and Biochemistry, and Institute for Immunology, University of California, Irvine, Irvine, CA 92697-3900, USA. Phone: 1-949-824-1947 Fax: 1-949-824-8551
| |
Collapse
|
111
|
Beurel E, Michalek SM, Jope RS. Innate and adaptive immune responses regulated by glycogen synthase kinase-3 (GSK3). Trends Immunol 2009; 31:24-31. [PMID: 19836308 DOI: 10.1016/j.it.2009.09.007] [Citation(s) in RCA: 317] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 09/21/2009] [Accepted: 09/23/2009] [Indexed: 11/30/2022]
Abstract
In just a few years, the view of glycogen synthase kinase-3 (GSK3) has been transformed from an obscure enzyme seldom encountered in the immune literature to one implicated in an improbably large number of roles. GSK3 is a crucial regulator of the balance between pro- and anti-inflammatory cytokine production in both the periphery and the central nervous system, so that GSK3 inhibitors such as lithium can diminish inflammation. GSK3 influences T-cell proliferation, differentiation and survival. Many effects stem from GSK3 regulation of critical transcription factors, such as NF-kappaB, NFAT and STATs. These discoveries led to the rapid application of GSK3 inhibitors to animal models of sepsis, arthritis, colitis, multiple sclerosis and others, demonstrating their potential for therapeutic intervention.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry, University of Alabama at Birmingham, Birmingham, AL 35294-0017, USA
| | | | | |
Collapse
|
112
|
Garcia CA, Wang H, Benakanakere MR, Barrett E, Kinane DF, Martin M. c-jun controls the ability of IL-12 to induce IL-10 production from human memory CD4+ T cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:4475-82. [PMID: 19734233 DOI: 10.4049/jimmunol.0901283] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
IL-12p70 is an immunoregulatory cytokine that has been shown to induce IL-10 production from CD4+ T cells, yet the underlying cellular mechanisms controlling this process are poorly understood. In the present study, we demonstrate that IL-12p70 induces IL-10 production from human memory CD4+ T cells via a PI3K-dependent signaling mechanism. Specifically, stimulation of human memory CD4+ T cells in the presence of IL-12p70 lead to increased PI3K activity and the subsequent phosphorylation and inactivation of the downstream constitutively active serine/threonine kinase, glycogen synthase kinase-3beta (GSK3beta). Inhibition of PI3K prevented the inactivation of GSK3beta by IL-12p70, as well as the subsequent ability of IL-12p70 to augment IL-10 levels by memory CD4+ T cells. Moreover, ectopic expression of a constitutively active form of GSK3beta abrogated the ability of IL-12p70 to increase IL-10 production by TCR-stimulated CD4+ T cells. In contrast, direct inhibition of GSK3 mimicked the effect of IL-12p70 on IL-10 production by memory CD4+ T cells. Analysis of downstream transcription factors identified that the ability of IL-12p70 to inactivate GSK3beta lead to increased levels of c-jun. The ability of IL-12p70 to inactivate GSK3beta and induce c-jun levels was required for IL-12 to augment IL-10 production by human memory CD4+ T cells, since small interfering RNA-mediated gene silencing of c-jun abrogated this process. These studies identify the cellular mechanism by which IL-12 induces IL-10 production from human memory CD4+ T cells.
Collapse
Affiliation(s)
- Carlos A Garcia
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | | | | | | | | | | |
Collapse
|
113
|
Li T, Wong VKW, Yi XQ, Wong YF, Zhou H, Liu L. Pseudolaric acid B suppresses T lymphocyte activation through inhibition of NF-κB signaling pathway and p38 phosphorylation. J Cell Biochem 2009; 108:87-95. [DOI: 10.1002/jcb.22230] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
114
|
Abstract
The healthy immune system makes use of a variety of surveillance mechanisms at different stages of lymphoid development to prevent the occurrence and expansion of potentially harmful autoreactive T cell clones. Disruption of these mechanisms may lead to inappropriate activation of T cells and the development of autoimmune and lymphoproliferative diseases [such as multiple sclerosis, rheumatoid arthritis, lupus erythematosus, diabetes and autoimmune lymphoproliferative syndrome (ALPS)]. Clonal deletion of T cells with high affinities for self-peptide-MHC via programmed cell death (apoptosis) is an essential mechanism leading to self-tolerance. Referred to as negative selection, central tolerance in the thymus serves as the first checkpoint for the developing T cell repertoire and involves the apoptotic elimination of potentially autoreactive T cells clones bearing high affinity T cell receptors (TCR) that recognize autoantigens presented by thymic epithelial cells. Autoreactive T cells that escape negative selection are held in check in the periphery by either functional inactivation ("anergy") or extrathymic clonal deletion, both of which are dependent on the strength and frequency of the TCR signal and the costimulatory context, or by regulatory T cells. This review provides an overview of the different molecular executioners of cell death programs that are vital to intrathymic or extrathymic clonal deletion of T cells. Further, the potential involvement of various apoptotic signaling paradigms are discussed with respect to the genesis and pathophysiology of autoimmune disease.
Collapse
Affiliation(s)
- Martina Gatzka
- Department of Molecular Biology and Biochemistry, Center for Immunology, University of California, Irvine, CA, USA.
| | | |
Collapse
|
115
|
Smith AE, Chronis C, Christodoulakis M, Orr SJ, Lea NC, Twine NA, Bhinge A, Mufti GJ, Thomas NSB. Epigenetics of human T cells during the G0-->G1 transition. Genome Res 2009; 19:1325-37. [PMID: 19546172 DOI: 10.1101/gr.085530.108] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We investigated functional epigenetic changes that occur in primary human T lymphocytes during entry into the cell cycle and mapped these at the single-nucleosome level by ChIP-chip on tiling arrays for chromosomes 1 and 6. We show that nucleosome loss and flanking active histone marks define active transcriptional start sites (TSSs). Moreover, these signatures are already set at many inducible genes in quiescent cells prior to cell stimulation. In contrast, there is a dearth of the inactive histone mark H3K9me3 at the TSS, and under-representation of H3K9me2 and H3K9me3 defines the body of active genes. At the DNA level, cytosine methylation (meC) is enriched for nucleosomes that remain at the TSS, whereas in general there is a dearth of meC at TSSs. Furthermore, a drop in meC also marks 3' transcription termination, and a peak of meC occurs at stop codons. This mimics the 3' nucleosomal distribution in yeast, which we show does not occur in human T cells.
Collapse
Affiliation(s)
- Alexander E Smith
- King's College London, Department of Haematological Medicine, Leukaemia Sciences Laboratories, Rayne Institute, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Blomberg KEM, Boucheron N, Lindvall JM, Yu L, Raberger J, Berglöf A, Ellmeier W, Smith CIE. Transcriptional signatures of Itk-deficient CD3+, CD4+ and CD8+ T-cells. BMC Genomics 2009; 10:233. [PMID: 19450280 PMCID: PMC2689280 DOI: 10.1186/1471-2164-10-233] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 05/18/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Tec-family kinase Itk plays an important role during T-cell activation and function, and controls also conventional versus innate-like T-cell development. We have characterized the transcriptome of Itk-deficient CD3+ T-cells, including CD4+ and CD8+ subsets, using Affymetrix microarrays. RESULTS The largest difference between Itk-/- and Wt CD3+ T-cells was found in unstimulated cells, e.g. for killer cell lectin-like receptors. Compared to anti-CD3-stimulation, anti-CD3/CD28 significantly decreased the number of transcripts suggesting that the CD28 co-stimulatory pathway is mainly independent of Itk. The signatures of CD4+ and CD8+ T-cell subsets identified a greater differential expression than in total CD3+ cells. Cyclosporin A (CsA)-treatment had a stronger effect on transcriptional regulation than Itk-deficiency, suggesting that only a fraction of TCR-mediated calcineurin/NFAT-activation is dependent on Itk. Bioinformatic analysis of NFAT-sites of the group of transcripts similarly regulated by Itk-deficiency and CsA-treatment, followed by chromatin-immunoprecipitation, revealed NFATc1-binding to the Bub1, IL7R, Ctla2a, Ctla2b, and Schlafen1 genes. Finally, to identify transcripts that are regulated by Tec-family kinases in general, we compared the expression profile of Itk-deficient T-cells with that of Btk-deficient B-cells and a common set of transcripts was found. CONCLUSION Taken together, our study provides a general overview about the global transcriptional changes in the absence of Itk.
Collapse
Affiliation(s)
- K Emelie M Blomberg
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Huddinge, Sweden
| | - Nicole Boucheron
- Division of Immunobiology, Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Jessica M Lindvall
- Department for Informatics, Center for Bioinformatics, Post box 1080 Blindern NO-0316, Oslo, Norway
| | - Liang Yu
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Huddinge, Sweden
| | - Julia Raberger
- Division of Immunobiology, Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Anna Berglöf
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Huddinge, Sweden
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - CI Edvard Smith
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Huddinge, Sweden
| |
Collapse
|
117
|
Beyersdorf N, Braun A, Vögtle T, Varga-Szabo D, Galdos RR, Kissler S, Kerkau T, Nieswandt B. STIM1-independent T cell development and effector function in vivo. THE JOURNAL OF IMMUNOLOGY 2009; 182:3390-7. [PMID: 19265116 DOI: 10.4049/jimmunol.0802888] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) is believed to be of pivotal importance in T cell physiology. To test this hypothesis, we generated mice constitutively lacking the SOCE-regulating Ca(2+) sensor stromal interaction molecule 1 (STIM1). In vitro analyses showed that SOCE and Ag receptor complex-triggered Ca(2+) flux into STIM1-deficient T cells is virtually abolished. In vivo, STIM1-deficient mice developed a lymphoproliferative disease despite normal thymic T cell maturation and normal frequencies of CD4(+)Foxp3(+) regulatory T cells. Unexpectedly, STIM1-deficient bone marrow chimeric mice mounted humoral immune responses after vaccination and STIM1-deficient T cells were capable of inducing acute graft-versus-host disease following adoptive transfer into allogeneic hosts. These results demonstrate that STIM1-dependent SOCE is crucial for homeostatic T cell proliferation, but of much lesser importance for thymic T cell differentiation or T cell effector functions.
Collapse
Affiliation(s)
- Niklas Beyersdorf
- Institute for Virology and Immunobiology, Deutsche Forschungsgemeinschaft Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
118
|
Abstract
SUMMARY Programmed death-1 (PD-1) is a cell surface molecule that regulates the adaptive immune response. Engagement of PD-1 by its ligands PD-L1 or PD-L2 transduces a signal that inhibits T-cell proliferation, cytokine production, and cytolytic function. While a great deal is known concerning the biologic roles PD-1 plays in regulating the primary immune response and in T-cell exhaustion, comparatively little is known regarding how PD-1 ligation alters signaling pathways. PD-1 ligation is known to inhibit membrane-proximal T-cell signaling events, while ligation of the related inhibitory molecule cytotoxic T-lymphocyte antigen-4 appears to target more downstream signaling pathways. A major obstacle to an in-depth understanding of PD-1 signaling is the lack of physiologic models in which to study signal transduction. This review focuses on: (i) signaling pathways altered by PD-1 ligation, (ii) factors recruited upon PD-1 phosphorylation, and (iii) exploring the hypothesis that PD-1 ligation induces distinct signals during various stages of immune-cell differentiation. Lastly, we describe models to dissect the function of the PD-1 cytoplasmic tail using primary cells in the absence of agonist antibodies.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/immunology
- Apoptosis Regulatory Proteins/metabolism
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- CTLA-4 Antigen
- Humans
- Mice
- Programmed Cell Death 1 Receptor
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- James L Riley
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, The University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
119
|
Podojil JR, Miller SD. Molecular mechanisms of T-cell receptor and costimulatory molecule ligation/blockade in autoimmune disease therapy. Immunol Rev 2009; 229:337-55. [PMID: 19426232 PMCID: PMC2845642 DOI: 10.1111/j.1600-065x.2009.00773.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
SUMMARY Pro-inflammatory CD4(+) T-cell-mediated autoimmune diseases, such as multiple sclerosis and type 1 diabetes, are hypothesized to be initiated and maintained by activated antigen-presenting cells presenting self antigen to self-reactive interferon-gamma and interleukin-17-producing CD4(+) T-helper (Th) type 1/Th17 cells. To date, the majority of Food and Drug Administration-approved therapies for autoimmune disease primarily focus on the global inhibition of immune inflammatory activity. The goal of ongoing research in this field is to develop both therapies that inhibit/eliminate activated autoreactive cells as well as antigen-specific treatments, which allow for the directed blockade of the deleterious effects of self-reactive immune cell function. According to the two-signal hypothesis, activation of a naive antigen-specific CD4(+) T cell requires both stimulation of the T-cell receptor (TCR) (signal 1) and stimulation of costimulatory molecules (signal 2). There also exists a balance between pro-inflammatory and anti-inflammatory immune cell activity, which is regulated by the type and strength of the activating signal as well as the local cytokine milieu in which the naive CD4(+) T cell is activated. To this end, the majority of ongoing research is focused on the delivery of suboptimal TCR stimulation in the absence of costimulatory molecule stimulation, or potential blockade of stimulatory accessory molecules. Therefore, the signaling pathways involved in the induction of CD4(+) T-cell anergy, as apposed to activation, are topics of intense interest.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
120
|
Abstract
SUMMARY OX40 (CD134) and its binding partner, OX40L (CD252), are members of the tumor necrosis factor receptor/tumor necrosis factor superfamily and are expressed on activated CD4(+) and CD8(+) T cells as well as on a number of other lymphoid and non-lymphoid cells. Costimulatory signals from OX40 to a conventional T cell promote division and survival, augmenting the clonal expansion of effector and memory populations as they are being generated to antigen. OX40 additionally suppresses the differentiation and activity of T-regulatory cells, further amplifying this process. OX40 and OX40L also regulate cytokine production from T cells, antigen-presenting cells, natural killer cells, and natural killer T cells, and modulate cytokine receptor signaling. In line with these important modulatory functions, OX40-OX40L interactions have been found to play a central role in the development of multiple inflammatory and autoimmune diseases, making them attractive candidates for intervention in the clinic. Conversely, stimulating OX40 has shown it to be a candidate for therapeutic immunization strategies for cancer and infectious disease. This review provides a broad overview of the biology of OX40 including the intracellular signals from OX40 that impact many aspects of immune function and have promoted OX40 as one of the most prominent costimulatory molecules known to control T cells.
Collapse
Affiliation(s)
- Michael Croft
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
121
|
Targeted knock-in mice expressing mutations of CD28 reveal an essential pathway for costimulation. Mol Cell Biol 2009; 29:3710-21. [PMID: 19398586 DOI: 10.1128/mcb.01869-08] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Despite extensive study, the role of phosphatidylinositol 3-kinase (PI3-kinase) activation in CD28 function has been highly contentious. To definitively address this question, we generated knock-in mice expressing mutations in two critical domains of the cytoplasmic tail of CD28. Mutation of the proximal tyrosine motif interrupted PI3-kinase binding and prevented CD28-dependent phosphorylation of protein kinase B (PKB)/Akt; however, there was no detectable effect on interleukin-2 (IL-2) secretion, expression of Bcl-X(L), or on T-cell function in vivo. Furthermore, we demonstrate that signaling initiated by the C-terminal proline motif is directly responsible for tyrosine phosphorylation of phosphoinosotide-dependent kinase 1, protein kinase C theta, and glycogen synthase kinase 3beta, as well as contributing to threonine phosphorylation of PKB. T cells mutated in this domain were profoundly impaired in IL-2 secretion, and the mice had marked impairment of humoral responses as well as less severe disease manifestations in experimental allergic encephalomyelitis. These data demonstrate that the distal proline motif initiates a critical nonredundant signaling pathway, whereas direct activation of PI3-kinase by the proximal tyrosine motif of CD28 is not required for normal T-cell function.
Collapse
|
122
|
Abstract
CD28 is recognized as the primary costimulatory molecule involved in the activation of naïve T cells. However, the biochemical signaling pathways that are activated by CD28 and how these pathways are integrated with TCR signaling are still not understood. We have recently shown that there are at least two independent activation pathways induced by CD28 costimulation. One is integrated with TCR signaling in the context of the immunological synapse and is mediated through transcriptional enhancement and the second is mediated through the induction of mRNA stability. Here, we review the immunological consequences and biochemical mechanisms associated with CD28 costimulation and discuss the major questions that need to be resolved to understand the molecular mechanisms that transduce CD28 costimulation.
Collapse
|
123
|
Park SG, Schulze-Luehrman J, Hayden MS, Hashimoto N, Ogawa W, Kasuga M, Ghosh S. The kinase PDK1 integrates T cell antigen receptor and CD28 coreceptor signaling to induce NF-kappaB and activate T cells. Nat Immunol 2009; 10:158-66. [PMID: 19122654 PMCID: PMC2768497 DOI: 10.1038/ni.1687] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Accepted: 10/31/2008] [Indexed: 11/08/2022]
Abstract
In addition to ligation of the T cell antigen receptor (TCR), activation of the CD28 coreceptor by the costimulatory molecule B7 is required for induction of the transcription factor NF-kappaB and robust T cell activation, although the contribution of CD28 to this process remains incompletely understood. We show here that phosphoinositide-dependent kinase 1 (PDK1) is essential for integrating the TCR and CD28 signals. After we deleted PDK1 from T cells, TCR-CD28 signals were unable to induce activation of NF-kappaB or phosphorylation of protein kinase C-theta, although T cell survival and pathways dependent on the kinases p38 and Jnk or the transcription factor NFAT were unaffected. CD28 facilitated NF-kappaB activation by regulating recruitment and phosphorylation of PDK1, which are necessary for efficient binding of PDK1 to protein kinase C-theta and the adaptor CARMA1 and thus for NF-kappaB induction.
Collapse
Affiliation(s)
- Sung-Gyoo Park
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jan Schulze-Luehrman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Matthew S. Hayden
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Naoko Hashimoto
- Department of Clinical Molecular Medicine, Division of Diabetes and Digestive and Kidney Diseases, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Wataru Ogawa
- Department of Clinical Molecular Medicine, Division of Diabetes and Digestive and Kidney Diseases, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Masato Kasuga
- Department of Clinical Molecular Medicine, Division of Diabetes and Digestive and Kidney Diseases, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Sankar Ghosh
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Molecular Biophysics & Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
124
|
Garcia CA, Benakanakere MR, Alard P, Kosiewicz MM, Kinane DF, Martin M. Antigenic experience dictates functional role of glycogen synthase kinase-3 in human CD4+ T cell responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:8363-71. [PMID: 19050253 PMCID: PMC2849970 DOI: 10.4049/jimmunol.181.12.8363] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Signals induced by the TCR and CD28 costimulatory pathway have been shown to lead to the inactivation of the constitutively active enzyme, glycogen synthase kinase-3 (GSK3), which has been implicated in the regulation of IL-2 and T cell proliferation. However, it is unknown whether GSK3 plays a similar role in naive and memory CD4(+) T cell responses. Here we demonstrate a divergence in the dependency on the inactivation of GSK3 in the proliferative responses of human naive and memory CD4(+) T cells. We find that although CD28 costimulation increases the frequency of phospho-GSK3 inactivation in TCR-stimulated naive and memory CD4(+) T cells, memory cells are less reliant on GSK3 inactivation for their proliferative responses. Rather we find that GSK3beta plays a previously unrecognized role in the selective regulation of the IL-10 recall response by human memory CD4(+) T cells. Furthermore, GSK3beta-inactivated memory CD4(+) T cells acquired the capacity to suppress the bystander proliferation of CD4(+) T cells in an IL-10-dependent, cell contact-independent manner. Our findings reveal a dichotomy present in the function of GSK3 in distinct human CD4(+) T cell populations.
Collapse
Affiliation(s)
- Carlos A. Garcia
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Manjunatha R. Benakanakere
- Oral Health and Systemic Disease Research Group, University of Louisville School of Dentistry, Louisville, KY 40292
| | - Pascale Alard
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Michelle M. Kosiewicz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Denis F. Kinane
- Oral Health and Systemic Disease Research Group, University of Louisville School of Dentistry, Louisville, KY 40292
| | - Michael Martin
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
- Oral Health and Systemic Disease Research Group, University of Louisville School of Dentistry, Louisville, KY 40292
| |
Collapse
|
125
|
Sanchez-Lockhart M, Graf B, Miller J. Signals and sequences that control CD28 localization to the central region of the immunological synapse. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7639-48. [PMID: 19017952 PMCID: PMC3993010 DOI: 10.4049/jimmunol.181.11.7639] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
During T cell interaction with APC, CD28 is recruited to the central region (cSMAC) of the immunological synapse. CD28-mediated signaling through PI3K results in the recruitment of protein kinase C-theta (PKCtheta) to the cSMAC, activation of NF-kappaB, and up-regulation of IL-2 transcription. However, the mechanism that mediates CD28 localization to the cSMAC and the functional consequences of CD28 localization to the cSMAC are not understood. In this report, we show that CD28 recruitment and persistence at the immunological synapse requires TCR signals and CD80 engagement. Addition of mAb to either MHC class II or CD80 results in the rapid displacement of CD28 from the immunological synapse. Ligand binding is not sufficient for CD28 localization to the immunological synapse, as truncation of the cytosolic tail of CD28 disrupts synapse localization without effecting the ability of CD28 to bind CD80. Furthermore, a single point mutation in the CD28 cytosolic tail (tyrosine 188) interferes with the ability of CD28 to preferentially accumulate at the cSMAC. PKCtheta distribution at the immunological synapse mirrors the distribution of tyrosine 188-mutated CD28, indicating that CD28 drives the localization of PKCtheta even when CD28 is not localized to the cSMAC. Mutation of tyrosine 188 also results in diminished activation of NF-kappaB, suggesting that CD28-mediated localization of PKCtheta to the cSMAC is important for efficient signal transduction. These data reinforce the importance of the interplay of signals between TCR and CD28 and suggest that CD28 signaling through PCKtheta may be mediated through localization to the cSMAC region of the immunological synapse.
Collapse
MESH Headings
- Amino Acid Substitution
- Animals
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- Cell Line
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Immunological Synapses/enzymology
- Immunological Synapses/genetics
- Immunological Synapses/immunology
- Interleukin-2/biosynthesis
- Interleukin-2/genetics
- Interleukin-2/immunology
- Isoenzymes/genetics
- Isoenzymes/immunology
- Isoenzymes/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Mutation, Missense
- NF-kappa B/genetics
- NF-kappa B/immunology
- NF-kappa B/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/immunology
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Kinase C/genetics
- Protein Kinase C/immunology
- Protein Kinase C/metabolism
- Protein Kinase C-theta
- Protein Structure, Tertiary/genetics
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/enzymology
- T-Lymphocytes/immunology
- Transcription, Genetic/genetics
- Transcription, Genetic/immunology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
| | - Beth Graf
- The David H. Smith Center for Vaccine Biology and Immunology and the
| | - Jim Miller
- The David H. Smith Center for Vaccine Biology and Immunology and the
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642
| |
Collapse
|
126
|
Abstract
Homer proteins are best known as scaffold proteins at the post-synaptic density where they facilitate synaptic signalling and are thought to be required for learning and memory. Evidence implicating Homer proteins in the development of the nervous system is also steadily accumulating. Homer is highly conserved and is expressed at key developmental time points in the nervous system of several species. Homer regulates intracellular calcium homeostasis, clustering and trafficking of receptors and proteins at the cytosolic surface of the plasma membrane, transcription and translation, and cytoskeletal organization. Each of these functions has obvious potential to regulate neuronal development, and indeed Homer is implicated in several pathologies associated with the developing nervous system. Current data justify more critical experimental approaches to the role of Homer in the developing nervous system and related neurological disorders.
Collapse
Affiliation(s)
- Lisa Foa
- Laboratory of Molecular Neurobiology, School of Medicine, University of Tasmania, Hobart, Australia.
| | | |
Collapse
|
127
|
Ebert PJ, Richie Ehrlich LI, Davis MM. Low ligand requirement for deletion and lack of synapses in positive selection enforce the gauntlet of thymic T cell maturation. Immunity 2008; 29:734-45. [PMID: 18993085 PMCID: PMC3762485 DOI: 10.1016/j.immuni.2008.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Revised: 07/18/2008] [Accepted: 09/09/2008] [Indexed: 11/16/2022]
Abstract
Immature double-positive (CD4(+)CD8(+)) thymocytes respond to negatively selecting peptide-MHC ligands by forming an immune synapse that sustains contact with the antigen-presenting cell (APC). Using fluorescently labeled peptides, we showed that as few as two agonist ligands could promote APC contact and subsequent apoptosis in reactive thymocytes. Furthermore, we showed that productive signaling for positive selection, as gauged by nuclear translocation of a green fluorescent protein (GFP)-labeled NFATc construct, did not involve formation of a synapse between thymocytes and selecting epithelial cells in reaggregate thymus cultures. Antibody blockade of endogenous positively selecting ligands prevented NFAT nuclear accumulation in such cultures and reversed NFAT accumulation in previously stimulated thymocytes. Together, these data suggest a "gauntlet" model in which thymocytes mature by continually acquiring and reacquiring positively selecting signals without sustained contact with epithelial cells, thereby allowing them to sample many cell surfaces for potentially negatively selecting ligands.
Collapse
Affiliation(s)
- Peter J.R. Ebert
- Howard Hughes Medical Institute and The Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren I. Richie Ehrlich
- Howard Hughes Medical Institute and The Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark M. Davis
- Howard Hughes Medical Institute and The Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
128
|
Wang M, Windgassen D, Papoutsakis ET. A global transcriptional view of apoptosis in human T-cell activation. BMC Med Genomics 2008; 1:53. [PMID: 18947405 PMCID: PMC2600644 DOI: 10.1186/1755-8794-1-53] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 10/23/2008] [Indexed: 12/22/2022] Open
Abstract
Background T-cell activation is an essential step of immune response. The process of proper T-cell activation is strictly monitored and regulated by apoptosis signaling. Yet, regulation of apoptosis, an integral and crucial facet during the process of T-cell activation, is not well understood. Methods In this study, a Gene-Ontology driven global gene expression analysis coupled with protein abundance and activity assays identified genes and pathways associated with regulation of apoptosis in primary human CD3+ T cells and separately CD4+ and CD8+ T cells. Results We identified significantly regulated apoptotic genes in several protein families, such as BCL2 proteins, CASPASE proteins, and TNF receptors, and detailed their transcriptional kinetics during the T-cell activation process. Transcriptional patterns of a few select genes (BCL2A1, BBC3 and CASP3) were validated at the protein level. Many of these apoptotic genes are involved in NF-κB signaling pathway, including TNFRSF10A, TNFRSF10B, TRAF4, TRAF1, TRAF3, and TRAF6. Upregulation of NF-κB and IκB family genes (REL, RELA, and RELB, NFKBIA, NFKBIE and NFKB1) at 48 to 96 hours, supported by the increase of phosphorylated RELA (p65), suggests that the involvement of the NF-κB complex in the process of T-cell proliferation is not only regulated at the protein level but also at the transcriptional level. Examination of genes involved in MAP kinase signalling pathway, important in apoptosis, suggests an induction of p38 and ERK1 cascades in T-cell proliferation (at 48 to 96 hours), which was explored using phosphorylation assays for p38 (MAPK14) and ERK1 (MAPK3). An immediate and short-lived increase of AP-1 activity measured by DNA-binding activity suggests a rapid and transient activation of p38 and/or JNK cascades upon T-cell activation. Conclusion This comparative genome-scale, transcriptional analysis of T-cell activation in the CD4+ and CD8+ subsets and the mixed CD3+ population identified many apoptosis genes not previously identified in the context of T-cell activation. Furthermore, it provided a comprehensive temporal analysis of the transcriptional program of apoptosis associated with T-cell activation.
Collapse
Affiliation(s)
- Min Wang
- Interdepartmental Biological Sciences Program, Northwestern University, Evanston, IL, USA.
| | | | | |
Collapse
|
129
|
Holz LE, Benseler V, Bowen DG, Bouillet P, Strasser A, O'Reilly L, d'Avigdor W, Bishop AG, McCaughan GW, Bertolino P. Intrahepatic murine CD8 T-cell activation associates with a distinct phenotype leading to Bim-dependent death. Gastroenterology 2008; 135:989-97. [PMID: 18619445 PMCID: PMC2956118 DOI: 10.1053/j.gastro.2008.05.078] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 05/19/2008] [Accepted: 05/29/2008] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Chronic infections by hepatotropic viruses such as hepatitis B and C are generally associated with an impaired CD8 T-cell immune response that is unable to clear the virus. The liver is increasingly recognized as an alternative site in which primary activation of CD8 T cells takes place, a property that might explain its role in inducing tolerance. However, the molecular mechanism by which intrahepatically activated T cells become tolerant is unknown. Here, we investigated the phenotype and fate of naïve CD8 T cells activated by hepatocytes in vivo. METHODS Transgenic mouse models in which the antigen is expressed in lymph nodes and/or in the liver were adoptively transferred with naïve CD8 T cells specific for the hepatic antigen. RESULTS Liver-activated CD8 T cells displayed poor effector functions and a unique CD25(low) CD54(low) phenotype. This phenotype was associated with increased expression of the proapoptotic protein Bim and caspase-3, demonstrating that these cells are programmed to die following intrahepatic activation. Importantly, we show that T cells deficient for Bim survived following intrahepatic activation. CONCLUSIONS This study identifies Bim for the first time as a critical initiator of T-cell death in the liver. Thus, strategies inhibiting the up-regulation of this molecule could potentially be used to rescue CD8 T cells, clear the virus, and reverse the outcome of viral chronic infections affecting the liver.
Collapse
Affiliation(s)
- Lauren E Holz
- Centenary Institute, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital and Faculty of Medicine, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Volker Benseler
- Centenary Institute, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital and Faculty of Medicine, University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Surgery, University of Regensburg, Bavaria, 93053, Germany
| | - David G Bowen
- Centenary Institute, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital and Faculty of Medicine, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3050, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3050, Australia
| | - Lorraine O'Reilly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3050, Australia
| | - William d'Avigdor
- Centenary Institute, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital and Faculty of Medicine, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Alex G Bishop
- Collaborative Transplant Laboratory, Blackburn Building, University of Sydney, NSW, 2006, Australia
| | - Geoffrey W McCaughan
- Centenary Institute, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital and Faculty of Medicine, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Patrick Bertolino
- Centenary Institute, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital and Faculty of Medicine, University of Sydney, Camperdown, NSW, 2050, Australia
| |
Collapse
|
130
|
Scottà C, Soligo M, Camperio C, Piccolella E. FOXP3 induced by CD28/B7 interaction regulates CD25 and anergic phenotype in human CD4+CD25- T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2008; 181:1025-33. [PMID: 18606654 DOI: 10.4049/jimmunol.181.2.1025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Among the signals necessary to generate CD4(+)CD25(+)FOXP3(+) T cells from CD4(+)CD25(-)FOXP3(-) T cells, a pivotal role is played by CD28. However, in humans, it is not known whether CD28 signaling independently of TCR promotes forkhead box protein 3 (FOXP3) expression and regulates CD4(+)CD25(+)FOXP3(+) T cell functions. To address this issue, starting from our previous experience, we analyzed the unique signals delivered by CD28 following stimulation by its natural ligand B7. Our results show that, in primary CD4(+)CD25(-) T cells, CD28 signals independent of TCR-mediated stimulatory pathways are sufficient to induce the transcription of FOXP3 in a small number of CD4(+)CD25(-) T cells committed to express FOXP3. These signals are dependent on CD28-derived PI3K/Akt pathways and resistant to cyclosporin A. In addition, we demonstrated that translated FOXP3 was recruited to CD25, Il-2, and Ctla4 target promoters. CD28-mediated FOXP3 expression was transient and correlated with CD25 expression. The presence of FOXP3 in CD28-activated CD4(+)CD25(-) T cells correlated with a transient unresponsiveness to antigenic stimuli. The addition of exogenous IL-2 did not influence either FOXP3 or CD25 expression but rescued CD28-activated T cells from apoptosis. Our results, demonstrating that FOXP3 expression driven solely by the CD28/B7 interaction inhibited T cell activation, support the role of CD28 in the regulation of peripheral tolerance and suggest a new mechanism through which it could occur.
Collapse
Affiliation(s)
- Cristiano Scottà
- Department of Cellular and Developmental Biology, University Sapienza of Rome, Rome, Italy
| | | | | | | |
Collapse
|
131
|
Genomic expression profiling of TNF-alpha-treated BDC2.5 diabetogenic CD4+ T cells. Proc Natl Acad Sci U S A 2008; 105:10107-12. [PMID: 18632574 DOI: 10.1073/pnas.0803336105] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
TNF-alpha plays an important role in immune regulation, inflammation, and autoimmunity. Chronic TNF exposure has been shown to down-modulate T cell responses. In a mouse T cell hybridoma model, TNF attenuated T cell receptor (TCR) signaling. We have confirmed that chronic TNF and anti-TNF exposure suppressed and increased T cell responses, respectively. In adult TCR (BDC2.5) transgenic nonobese diabetic mice, DNA microarray analysis of global gene expression in BDC2.5 CD4(+) T cells in response to chronic TNF or anti-TNF exposure showed that genes involved in functional categories including T cell signaling, cell cycle, proliferation, ubiquitination, cytokine synthesis, calcium signaling, and apoptosis were modulated. Genes such as ubiquitin family genes, cytokine inducible Src homology 2-containing genes, cyclin-dependent kinase inhibitors p21, p57, calmodulin family genes (calmodulin-1, -2, and -3) and calcium channel voltage-dependent, N type alpha1B subunit (CaV2.2) were induced by TNF, whereas Vav2, Rho GTPase-activating protein, calcium channel voltage-dependent, L type alpha1C subunit (CaV1.2), IL-1 receptor-associated kinase-1 and -2, and IL enhancer binding factor 3 were reduced by TNF. Genes such as CaV1.2 and proliferating cell nuclear antigen, repressed by TNF, were induced by anti-TNF treatment. Further, we showed that chronic TNF exposure impaired NF-kappaB and adaptor protein 1 transactivation activity, leading to T cell unresponsiveness. Thus, our results present a detailed picture of transcriptional programs affected by chronic TNF exposure and provide candidate target genes that may function to mediate TNF-induced T cell unresponsiveness.
Collapse
|
132
|
King C, Tangye SG, Mackay CR. T follicular helper (TFH) cells in normal and dysregulated immune responses. Annu Rev Immunol 2008; 26:741-66. [PMID: 18173374 DOI: 10.1146/annurev.immunol.26.021607.090344] [Citation(s) in RCA: 497] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
T cell help for antibody production is a fundamental aspect of immune responses. Only recently has a better understanding of the cellular and molecular mechanisms for T cell help emerged. A subset of T cells, termed T follicular helper cells (T(FH) cells), provides a helper function to B cells and represents one of the most numerous and important subsets of effector T cells in lymphoid tissues. T(FH) cells are distinguishable from Th1 and Th2 cells by several criteria, including chemokine receptor expression (CXCR5), location/migration (B cell follicles), and function (B cell help). Central to the function of CD4(+) T cells is IL-21, a "helper" cytokine produced by T(FH) cells that potently stimulates the differentiation of B cells into Ab-forming cells through IL-21R. Consequently, dysregulation of T(FH) cell function, and over- or under-expression of T(FH) cell-associated molecules such as ICOS or IL-21, most likely contributes to the pathogenesis of certain autoimmune diseases or immunodeficiencies.
Collapse
Affiliation(s)
- Cecile King
- Immunology and Inflammation Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia.
| | | | | |
Collapse
|
133
|
Merino E, Avila-Flores A, Shirai Y, Moraga I, Saito N, Mérida I. Lck-dependent tyrosine phosphorylation of diacylglycerol kinase alpha regulates its membrane association in T cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:5805-15. [PMID: 18424699 DOI: 10.4049/jimmunol.180.9.5805] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR engagement triggers phospholipase Cgamma1 activation through the Lck-ZAP70-linker of activated T cell adaptor protein pathway. This leads to generation of diacylglycerol (DAG) and mobilization of intracellular Ca(2+), both essential for TCR-dependent transcriptional responses. TCR ligation also elicits transient recruitment of DAG kinase alpha (DGKalpha) to the lymphocyte plasma membrane to phosphorylate DAG, facilitating termination of DAG-regulated signals. The precise mechanisms governing dynamic recruitment of DGKalpha to the membrane have not been fully elucidated, although Ca(2+) influx and tyrosine kinase activation were proposed to be required. We show that DGKalpha is tyrosine phosphorylated, and identify tyrosine 335 (Y335), at the hinge between the atypical C1 domains and the catalytic region, as essential for membrane localization. Generation of an Ab that recognizes phosphorylated Y335 demonstrates Lck-dependent phosphorylation of endogenous DGKalpha during TCR activation and shows that pY335DGKalpha is a minor pool located exclusively at the plasma membrane. Our results identify Y335 as a residue critical for DGKalpha function and suggest a mechanism by which Lck-dependent phosphorylation and Ca(2+) elevation regulate DGKalpha membrane localization. The concerted action of these two signals results in transient, receptor-regulated DGKalpha relocalization to the site at which it exerts its function as a negative modulator of DAG-dependent signals.
Collapse
Affiliation(s)
- Ernesto Merino
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
134
|
Abidi SHI, Dong T, Vuong MT, Sreenu VB, Rowland-Jones SL, Evans EJ, Davis SJ. Differential remodeling of a T-cell transcriptome following CD8- versus CD3-induced signaling. Cell Res 2008; 18:641-8. [PMID: 18475290 PMCID: PMC2731849 DOI: 10.1038/cr.2008.56] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
CD8 engagement with class I major histocompatibility antigens greatly enhances T-cell activation, but it is not clear how this is achieved. We address the question of whether or not the antibody-mediated ligation of CD8 alone induces transcriptional remodeling in a T-cell clone, using serial analysis of gene expression. Even though it fails to induce overt phenotypic changes, we find that CD8 ligation profoundly alters transcription in the T-cell clone, at a scale comparable to that induced by antibody-mediated ligation of CD3. The character of the resulting changes is distinct, however, with the net effect of CD8 ligation being substantially inhibitory. We speculate that ligating CD8 induces weak, T-cell receptor (TCR)-mediated inhibitory signals reminiscent of the effects of TCR antagonists. Our results imply that CD8 ligation alone is incapable of activating the T-cell clone because it fails to fully induce NFAT-dependent transcription.
Collapse
Affiliation(s)
| | - Tao Dong
- Nuffield Department of Clinical Medicine and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Mai T. Vuong
- Nuffield Department of Clinical Medicine and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Vattipally B. Sreenu
- Nuffield Department of Clinical Medicine and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Sarah L. Rowland-Jones
- Nuffield Department of Clinical Medicine and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Edward J. Evans
- Nuffield Department of Clinical Medicine and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Simon J. Davis
- Nuffield Department of Clinical Medicine and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| |
Collapse
|
135
|
Wang M, Windgassen D, Papoutsakis ET. Comparative analysis of transcriptional profiling of CD3+, CD4+ and CD8+ T cells identifies novel immune response players in T-cell activation. BMC Genomics 2008; 9:225. [PMID: 18485203 PMCID: PMC2396644 DOI: 10.1186/1471-2164-9-225] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Accepted: 05/16/2008] [Indexed: 11/29/2022] Open
Abstract
Background T-cell activation is an essential step of the immune response and relies on the tightly controlled orchestration of hundreds of genes/proteins, yet the cellular and molecular events underlying this complex process are not fully understood, especially at the genome-scale. Significantly, a comparative genome-scale transcriptional analysis of two T-cell subsets (CD4+ and CD8+) against each other and against the naturally mixed population (CD3+ cells) remains unexplored. Results Comparison of the microarray-based gene expression patterns between CD3+ T cells, and the CD4+ and CD8+ subsets revealed largely conserved, but not identical, transcriptional patterns. We employed a Gene-Ontology-driven transcriptional analysis coupled with protein abundance assays in order to identify novel T-cell activation genes and cell-type-specific genes associated with the immune response. We identified potential genes involved in the communication between the two subsets (including IL23A, NR4A2, CD83, PSMB2, -8, MIF, IFI16, TNFAIP1, POU2AF1, and OTUB1) and would-be effector-function-specific genes (XCL2, SLAMF7, TNFSF4, -5, -9, CSF3, CD48 and CD244). Chemokines induced during T-cell activation, but not previously identified in T cells, include CCL20, CXCL9, -10, -11 (in all three populations), and XCL2 (preferentially in CD8+ T cells). Increased expression of other unexpected cytokines (GPI, OSM and MIF) suggests their involvement in T-cell activation with their functions yet to be examined. Differential expression of many receptors, not previously reported in the context of T-cell activation, including CCR5, CCR7, IL1R2, IL1RAP, IL6R, TNFRSF25 and TNFRSF1A, suggests their role in this immune process. Several receptors involved in TCR activation (CD3D, CD3G, TRAT1, ITGAL, ITGB1, ITGB2, CD8A and B (CD8+ T-cell specific) along with LCK, ZAP70 and TYROBP were synchronously downregulated. Members of cell-surface receptors (HLA-Ds and KLRs), none previously identified in the context of T-cell activation, were also downregulated. Conclusion This comparative genome-scale, transcriptional analysis of T-cell activation in the CD4+ and CD8+ subsets and the mixed CD3+ populations made possible the identification of many immune-response genes not previously identified in the context of T-cell activation. Significantly, it made possible to identify the temporal patterns of many previously known T-cell activation genes, and also identify genes implicated in effector functions of and communication between CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Min Wang
- Interdepartmental Biological Sciences Program, Northwestern University, Evanston, IL, USA.
| | | | | |
Collapse
|
136
|
Saenz L, Lozano JJ, Valdor R, Baroja-Mazo A, Ramirez P, Parrilla P, Aparicio P, Sumoy L, Yélamos J. Transcriptional regulation by poly(ADP-ribose) polymerase-1 during T cell activation. BMC Genomics 2008; 9:171. [PMID: 18412984 PMCID: PMC2375913 DOI: 10.1186/1471-2164-9-171] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Accepted: 04/16/2008] [Indexed: 12/23/2022] Open
Abstract
Background Accumulating evidence suggests an important role for the enzyme poly(ADP-ribose) polymerase-1 (PARP-1) as an integral part of the gene expression regulatory machinery during development and in response to specific cellular signals. PARP-1 might modulate gene expression through its catalytic activity leading to poly(ADP-ribosyl)ation of nuclear proteins or by its physical association with relevant proteins. Recently, we have shown that PARP-1 is activated during T cell activation. However, the proposed role of PARP-1 in reprogramming T cell gene expression upon activation remains largely unexplored. Results In the present study we use oligonucleotide microarray analysis to gain more insight into the role played by PARP-1 during the gene expression reprogramming that takes place in T cells upon activation with anti-CD3 stimulation alone, or in combination with anti-CD28 co-stimulation. We have identified several groups of genes with expression modulated by PARP-1. The expression of 129 early-response genes to anti-CD3 seems to be regulated by PARP-1 either in a positive (45 genes) or in a negative manner (84 genes). Likewise, in the presence of co-stimulation (anti-CD3 + anti-CD28 stimulation), the expression of 203 genes is also regulated by PARP-1 either up (173 genes) or down (30 genes). Interestingly, PARP-1 deficiency significantly alters expression of genes associated with the immune response such as chemokines and genes involved in the Th1/Th2 balance. Conclusion This study provides new insights into changes in gene expression mediated by PARP-1 upon T cell activation. Pathway analysis of PARP-1 as a nuclear signalling molecule in T cells would be of relevance for the future development of new therapeutic approaches targeting PARP-1 in the acquired immune response.
Collapse
Affiliation(s)
- Luis Saenz
- Transplant Unit, Department of Surgery, University Hospital Virgen de Arrixaca, University of Murcia, Ciberehd, Murcia, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Wang L, Montano M, Rarick M, Sebastiani P. Conditional clustering of temporal expression profiles. BMC Bioinformatics 2008; 9:147. [PMID: 18334028 PMCID: PMC2335301 DOI: 10.1186/1471-2105-9-147] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Accepted: 03/11/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Many microarray experiments produce temporal profiles in different biological conditions but common cluster techniques are not able to analyze the data conditional on the biological conditions. RESULTS This article presents a novel technique to cluster data from time course microarray experiments performed across several experimental conditions. Our algorithm uses polynomial models to describe the gene expression patterns over time, a full Bayesian approach with proper conjugate priors to make the algorithm invariant to linear transformations, and an iterative procedure to identify genes that have a common temporal expression profile across two or more experimental conditions, and genes that have a unique temporal profile in a specific condition. CONCLUSION We use simulated data to evaluate the effectiveness of this new algorithm in finding the correct number of clusters and in identifying genes with common and unique profiles. We also use the algorithm to characterize the response of human T cells to stimulations of antigen-receptor signaling gene expression temporal profiles measured in six different biological conditions and we identify common and unique genes. These studies suggest that the methodology proposed here is useful in identifying and distinguishing uniquely stimulated genes from commonly stimulated genes in response to variable stimuli. Software for using this clustering method is available from the project home page.
Collapse
Affiliation(s)
- Ling Wang
- Novartis Vaccines and Diagnostics, Emeryville, CA 94608, USA.
| | | | | | | |
Collapse
|
138
|
Pei Y, Zhu P, Dang Y, Wu J, Yang X, Wan B, Liu JO, Yi Q, Yu L. Nuclear export of NF90 to stabilize IL-2 mRNA is mediated by AKT-dependent phosphorylation at Ser647 in response to CD28 costimulation. THE JOURNAL OF IMMUNOLOGY 2008; 180:222-9. [PMID: 18097023 DOI: 10.4049/jimmunol.180.1.222] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-2 is one of the most important cytokines required for T cell-mediated immune responses. Costimulation of CD28 in T cells up-regulates IL-2 mRNA levels via transcription activation and mRNA stabilization. Upon T cell activation, NF90, an AU-rich element (ARE)-binding protein, translocates from the nucleus into the cytoplasm, where it binds to the ARE-containing 3' untranslated regions of IL-2 mRNA and slows down degradation of IL-2 mRNA. The translocation of NF90 is mediated through a nuclear export signal at its N terminus, but how it is triggered is still unclear. Phosphorylation of ARE-binding proteins has been reported as a signal transduction pathway to stabilize ARE-containing transcripts. In this study, we demonstrate that AKT phosphorylates NF90 on Ser647 upon CD28 costimulation. This phosphorylation is necessary for nuclear export of NF90 and IL-2 mRNA stabilization by this protein, because a mutation at Ser647 abolished both functions. We observed that treatment of cells with CD28 costimulation induced distinct increase in phosphorylation of AKT and NF90 at Ser647 concomitantly. Phosphorylation at Ser647 of NF90 up-regulated IL-2 production in response to CD28 costimulation. In vivo and in vitro data support a model in which CD28 costimulation activates AKT to phosphorylate NF90 at Ser647 and phosphorylation triggers NF90 to relocate to the cytoplasm and stabilize IL-2 mRNA.
Collapse
Affiliation(s)
- Yuan Pei
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
|
140
|
Dynamic covariation between gene expression and genome characteristics. Gene 2008; 410:53-66. [PMID: 18191345 DOI: 10.1016/j.gene.2007.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Revised: 11/13/2007] [Accepted: 11/29/2007] [Indexed: 11/21/2022]
Abstract
Gene and protein expression is controlled so that cells can react to changing intra- and extracellular signals by modulating biochemical networks and pathways. We have previously shown that gene expression and the properties of expressed proteins are dynamically correlated. Here we investigated correlations between gene related parameters and gene expression patterns, and found statistically significant correlations in microarray datasets for different cell types, organisms and processes, including human B and T cell stimulation, cell cycle in HeLa cells, infection in intestinal epithelial cells, Drosophila melanogaster life span, and Saccharomyces cerevisiae cell cycle. Our method was applied to time course datasets individually for each time point. We derived from sequence information numerous parameters for nucleotide composition, two-base composition, codon usage, skew parameters, and codon bias. In addition to coding regions, we also investigated correlations for complete genes and introns. Significant dynamic correlations were identified for each of the analyses. Our method also proved useful for detecting dynamic shifts in gene expression profiles, such as in the D. melanogaster dataset. Detection of changes in the properties of expressed genes and proteins might be useful for predicting or following biological processes, responses, growth, differentiation and possibly in related disorders.
Collapse
|
141
|
Bluestone JA, Hebrok M. Safer, longer-lasting regulatory T cells with beta-catenin. Nat Med 2008; 14:118-9. [PMID: 18256611 DOI: 10.1038/nm0208-118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
142
|
Huang GN, Huso DL, Bouyain S, Tu J, McCorkell KA, May MJ, Zhu Y, Lutz M, Collins S, Dehoff M, Kang S, Whartenby K, Powell J, Leahy D, Worley PF. NFAT binding and regulation of T cell activation by the cytoplasmic scaffolding Homer proteins. Science 2008; 319:476-81. [PMID: 18218901 DOI: 10.1126/science.1151227] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
T cell receptor (TCR) and costimulatory receptor (CD28) signals cooperate in activating T cells, although understanding of how these pathways are themselves regulated is incomplete. We found that Homer2 and Homer3, members of the Homer family of cytoplasmic scaffolding proteins, are negative regulators of T cell activation. This is achieved through binding of nuclear factor of activated T cells (NFAT) and by competing with calcineurin. Homer-NFAT binding was also antagonized by active serine-threonine kinase AKT, thereby enhancing TCR signaling via calcineurin-dependent dephosphorylation of NFAT. This corresponded with changes in cytokine expression and an increase in effector-memory T cell populations in Homer-deficient mice, which also developed autoimmune-like pathology. These results demonstrate a further means by which costimulatory signals are regulated to control self-reactivity.
Collapse
Affiliation(s)
- Guo N Huang
- Program in Biochemistry, Cellular and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Deschamps M, Robinet E, Certoux JM, Mercier P, Sauce D, De Vos J, Montcuquet N, Bonyhadi M, Rème T, Tiberghien P, Ferrand C. Transcriptome of retrovirally transduced CD8+ lymphocytes: Influence of cell activation, transgene integration, and selection process. Mol Immunol 2008; 45:1112-25. [PMID: 17825913 DOI: 10.1016/j.molimm.2007.07.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 07/16/2007] [Accepted: 07/18/2007] [Indexed: 11/24/2022]
Abstract
A suicide gene introduced by retroviral means can allow in vivo control of alloreactivity mediated by donor gene-modified T cells (GMTC) after allogeneic hematopoietic stem cell transplantation. The present study establishes the transcriptomic profile of GMTC prepared according to the GMTC production process used in our clinical trial (activation/selection methods, CD3/NeoR), which was previously demonstrated to induce phenotypical and functional alterations. This transcriptomic profile was compared with that of GMTC prepared by a novel process (CD3-CD28/DeltaNGFR-MACS) that limits alterations. Using a human pan-genomic microarray and GeneSpring software, we determined the gene expression profiles of CD8+ T cells from four healthy donors before and after the different steps required for gene modification. This analysis revealed that the gene expression pattern of GMTC is affected mainly by the activation step. Specific analysis of GMTC production processes showed that DeltaNGFR-MACS selection combined with CD3-CD28 activation limits the aberrant expression of genes involved in immunological functions and apoptotic pathways. Furthermore, our results indicate a limited risk of oncogenesis associated with retroviral-mediated gene transfer in CD8+ cells, a lower perturbation of the cell cycle regulation pathway after CD3-CD28 activation than after CD3 activation, and no significant involvement of the DeltaNGFR transduction signaling pathway when DeltaNGFR is used for selection. Moreover, genes that might be targeted to limit T cell functional alterations after ex vivo manipulation and culture were identified. These findings should be relevant to further adoptive T cell immunotherapy trials using ex vivo-expanded, gene-modified or unmodified T cells.
Collapse
|
144
|
Morancho B, Minguillón J, Molkentin JD, López-Rodríguez C, Aramburu J. Analysis of the transcriptional activity of endogenous NFAT5 in primary cells using transgenic NFAT-luciferase reporter mice. BMC Mol Biol 2008; 9:13. [PMID: 18221508 PMCID: PMC2262899 DOI: 10.1186/1471-2199-9-13] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 01/25/2008] [Indexed: 12/13/2022] Open
Abstract
Background The transcription factor NFAT5/TonEBP regulates the response of mammalian cells to hypertonicity. However, little is known about the physiopathologic tonicity thresholds that trigger its transcriptional activity in primary cells. Wilkins et al. recently developed a transgenic mouse carrying a luciferase reporter (9xNFAT-Luc) driven by a cluster of NFAT sites, that was activated by calcineurin-dependent NFATc proteins. Since the NFAT site of this reporter was very similar to an optimal NFAT5 site, we tested whether this reporter could detect the activation of NFAT5 in transgenic cells. Results The 9xNFAT-Luc reporter was activated by hypertonicity in an NFAT5-dependent manner in different types of non-transformed transgenic cells: lymphocytes, macrophages and fibroblasts. Activation of this reporter by the phorbol ester PMA plus ionomycin was independent of NFAT5 and mediated by NFATc proteins. Transcriptional activation of NFAT5 in T lymphocytes was detected at hypertonic conditions of 360–380 mOsm/kg (isotonic conditions being 300 mOsm/kg) and strongly induced at 400 mOsm/kg. Such levels have been recorded in plasma in patients with osmoregulatory disorders and in mice deficient in aquaporins and vasopressin receptor. The hypertonicity threshold required to activate NFAT5 was higher in bone marrow-derived macrophages (430 mOsm/kg) and embryonic fibroblasts (480 mOsm/kg). Activation of the 9xNFAT-Luc reporter by hypertonicity in lymphocytes was insensitive to the ERK inhibitor PD98059, partially inhibited by the PI3-kinase inhibitor wortmannin (0.5 μM) and the PKA inhibitor H89, and substantially downregulated by p38 inhibitors (SB203580 and SB202190) and by inhibition of PI3-kinase-related kinases with 25 μM LY294002. Sensitivity of the reporter to FK506 varied among cell types and was greater in primary T cells than in fibroblasts and macrophages. Conclusion Our results indicate that NFAT5 is a sensitive responder to pathologic increases in extracellular tonicity in T lymphocytes. Activation of NFAT5 by hypertonicity in lymphocytes was mediated by a combination of signaling pathways that differed from those required in other cell types. We propose that the 9xNFAT-Luc transgenic mouse model might be useful to study the physiopathological regulation of both NFAT5 and NFATc factors in primary cells.
Collapse
Affiliation(s)
- Beatriz Morancho
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
145
|
Podojil JR, Turley DM, Miller SD. Therapeutic blockade of T-cell antigen receptor signal transduction and costimulation in autoimmune disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 640:234-51. [PMID: 19065796 PMCID: PMC2853772 DOI: 10.1007/978-0-387-09789-3_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
CD4+ T-cell-mediated autoimmune diseases are initiated and maintained by the presentation of self-antigen by antigen-presenting cells (APCs) to self-reactive CD4+ T-cells. According to the two-signal hypothesis, activation of a naive antigen-specific CD4+ T-cell requires stimulation of both the T-cell antigen receptor (signal 1) and costimulatory molecules such as CD28 (signal 2). To date, the majority of therapies for autoimmune diseases approved by the Food and Drug Administration primarily focus on the global inhibition of immune inflammatory activity. The goal of ongoing research in this field is to develop antigen-specific treatments which block the deleterious effects of self-reactive immune cell function while maintaining the ability of the immune system to clear nonself antigens. To this end, the signaling pathways involved in the induction of CD4+ T-cell anergy, as apposed to activation, are a topic of intense interest. This chapter discusses components of the CD4+ T-cell activation pathway that may serve as therapeutic targets for the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Joseph R. Podojil
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Danielle M. Turley
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Stephen D. Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
146
|
Hu L, Pennington M, Jiang Q, Whartenby KA, Calabresi PA. Characterization of the functional properties of the voltage-gated potassium channel Kv1.3 in human CD4+ T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2007; 179:4563-70. [PMID: 17878353 DOI: 10.4049/jimmunol.179.7.4563] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies have shown that central memory T (T(CM)) cells predominantly use the calcium-dependent potassium channel KCa3.1 during acute activation, whereas effector memory T (T(EM)) cells use the voltage-gated potassium channel Kv1.3. Because Kv1.3-specific pharmacological blockade selectively inhibited anti-CD3-mediated proliferation, whereas naive T cells and T(CM) cells escaped inhibition due to up-regulation of KCa3.1, this difference indicated a potential for selective targeting of the T(EM) population. We examined the effects of pharmacological Kv1.3 blockers and a dominant-negative Kv1.x construct on T cell subsets to assess the specific effects of Kv1.3 blockade. Our studies indicated both T(CM) and T(EM) CD4+ T cells stimulated with anti-CD3 were inhibited by charybdotoxin, which can block both KCa3.1 and Kv1.3, whereas margatoxin and Stichodactyla helianthus toxin, which are more selective Kv1.3 inhibitors, inhibited proliferation and IFN-gamma production only in the T(EM) subset. The addition of anti-CD28 enhanced proliferation of freshly isolated cells and rendered them refractory to S. helianthus, whereas chronically activated T(EM) cell lines appeared to be costimulation independent because Kv1.3 blockers effectively inhibited proliferation and IFN-gamma regardless of second signal. Transduction of CD4+ T cells with dominant-negative Kv1.x led to a higher expression of CCR7+ T(CM) phenotype and a corresponding depletion of T(EM). These data provide further support for Kv1.3 as a selective target of chronically activated T(EM) without compromising naive or T(CM) immune functions. Specific Kv1.3 blockers may be beneficial in autoimmune diseases such as multiple sclerosis in which T(EM) are found in the target organ.
Collapse
Affiliation(s)
- Lina Hu
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
147
|
van der Velden JLJ, Schols AMWJ, Willems J, Kelders MCJM, Langen RCJ. Glycogen synthase kinase 3 suppresses myogenic differentiation through negative regulation of NFATc3. J Biol Chem 2007; 283:358-366. [PMID: 17977834 DOI: 10.1074/jbc.m707812200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle atrophy is a prominent and disabling feature in many chronic diseases. Prevention or reversal of muscle atrophy by stimulation of skeletal muscle growth could be an important therapeutic strategy. Glycogen synthase kinase 3beta (GSK-3beta) has been implicated in the negative regulation of skeletal muscle growth. Since myogenic differentiation is an essential part of muscle growth, we investigated if inhibition of GSK-3beta is sufficient to stimulate myogenic differentiation and whether this depended on regulation of the transcription factor nuclear factor of activated T-cells (NFAT). In both myogenically converted mouse embryonic fibroblasts and C2C12 myoblasts, deficiency of GSK-3beta protein (activity) resulted in enhanced myotube formation and muscle-specific gene expression during differentiation, which was reversed by reintroduction of wild type but not kinase-inactive (K85R) GSK-3beta. In addition, GSK-3beta inhibition restored myogenic differentiation following calcineurin blockade, which suggested the involvement of NFAT. GSK-3beta-deficient mouse embryonic fibroblasts or myoblasts displayed enhanced nuclear translocation of NFATc3 and elevated NFAT-sensitive promoter transactivation, which was reduced by reintroducing wild type, but not K85R GSK-3beta. Overexpression of NFATc3 increased muscle gene promoter transactivation, which was abolished by co-expression of wild type GSK-3beta. Finally, stimulation of muscle gene expression observed following GSK-3beta inhibition was strongly attenuated in NFATc3-deficient myoblasts, indicating that this response requires NFATc3. Collectively, our data demonstrate negative regulation of myogenic differentiation by GSK-3beta through a transcriptional mechanism that depends on NFATc3. Inhibition of GSK-3beta may be a potential strategy in prevention or treatment of muscle atrophy.
Collapse
Affiliation(s)
- Jos L J van der Velden
- Department of Respiratory Medicine, Nutrition and Toxicology Research Institute Maastricht, Maastricht University, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, Nutrition and Toxicology Research Institute Maastricht, Maastricht University, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Jodil Willems
- Department of Respiratory Medicine, Nutrition and Toxicology Research Institute Maastricht, Maastricht University, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Marco C J M Kelders
- Department of Respiratory Medicine, Nutrition and Toxicology Research Institute Maastricht, Maastricht University, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Ramon C J Langen
- Department of Respiratory Medicine, Nutrition and Toxicology Research Institute Maastricht, Maastricht University, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| |
Collapse
|
148
|
Filby A, Seddon B, Kleczkowska J, Salmond R, Tomlinson P, Smida M, Lindquist JA, Schraven B, Zamoyska R. Fyn regulates the duration of TCR engagement needed for commitment to effector function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:4635-44. [PMID: 17878361 DOI: 10.4049/jimmunol.179.7.4635] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In naive T cells, engagement of the TCR with agonist peptide:MHC molecules leads to phosphorylation of key intracellular signaling intermediates within seconds and this peaks within minutes. However, the cell does not commit to proliferation and IL-2 cytokine production unless receptor contact is sustained for several hours. The biochemical basis for this transition to full activation may underlie how T cells receive survival signals while maintaining tolerance, and is currently not well understood. We show here that for CD8 T cells commitment to proliferation and cytokine production requires sustained activation of the Src family kinase Lck and is opposed by the action of Fyn. Thus, in the absence of Fyn, commitment to activation occurs more rapidly, the cells produce more IL-2, and undergo more rounds of division. Our data demonstrate a role for Fyn in modulating the response to Ag in primary T cells.
Collapse
Affiliation(s)
- Andrew Filby
- Division of Molecular Immunology, Medical Research Council, National Institute for Medical Research, The Ridgeway, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Podtschaske M, Benary U, Zwinger S, Höfer T, Radbruch A, Baumgrass R. Digital NFATc2 activation per cell transforms graded T cell receptor activation into an all-or-none IL-2 expression. PLoS One 2007; 2:e935. [PMID: 17895976 PMCID: PMC1978524 DOI: 10.1371/journal.pone.0000935] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 08/30/2007] [Indexed: 11/19/2022] Open
Abstract
The expression of interleukin-2 (IL-2) is a key event in T helper (Th) lymphocyte activation, controlling both, the expansion and differentiation of effector Th cells as well as the activation of regulatory T cells. We demonstrate that the strength of TCR stimulation is translated into the frequency of memory Th cells expressing IL-2 but not into the amount of IL-2 per cell. This molecular switch decision for IL-2 expression per cell is located downstream of the cytosolic Ca2+ level. Here we show that in a single activated Th cell, NFATc2 activation is digital but NF-κB activation is graded after graded T cell receptor (TCR) signaling. Subsequently, NFATc2 translocates into the nucleus in an all-or-none fashion per cell, transforming the strength of TCR-stimulation into the number of nuclei positive for NFATc2 and IL-2 transcription. Thus, the described NFATc2 switch regulates the number of Th cells actively participating in an immune response.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/metabolism
- Calcineurin/pharmacology
- Calcium/metabolism
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Flow Cytometry
- Humans
- Interferon-gamma/metabolism
- Interleukin-2/genetics
- Interleukin-2/metabolism
- Ionomycin/pharmacology
- Lectins, C-Type
- Lymphocyte Activation/drug effects
- Models, Theoretical
- NF-kappa B/metabolism
- NFATC Transcription Factors/genetics
- NFATC Transcription Factors/metabolism
- Phosphorylation
- Receptors, Antigen, T-Cell/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
| | - Uwe Benary
- German Rheumatism Research Centre, Berlin, Germany
| | - Sandra Zwinger
- Institute of Medical Immunology, Charité, Humboldt-University Berlin, Berlin, Germany
| | - Thomas Höfer
- Department of Theoretical Biophysics, Humboldt-University Berlin, Berlin, Germany
| | | | - Ria Baumgrass
- German Rheumatism Research Centre, Berlin, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
150
|
Brouard S, Mansfield E, Braud C, Li L, Giral M, Hsieh SC, Baeten D, Zhang M, Ashton-Chess J, Braudeau C, Hsieh F, Dupont A, Pallier A, Moreau A, Louis S, Ruiz C, Salvatierra O, Soulillou JP, Sarwal M. Identification of a peripheral blood transcriptional biomarker panel associated with operational renal allograft tolerance. Proc Natl Acad Sci U S A 2007; 104:15448-53. [PMID: 17873064 PMCID: PMC2000539 DOI: 10.1073/pnas.0705834104] [Citation(s) in RCA: 293] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Indexed: 12/13/2022] Open
Abstract
Long-term allograft survival generally requires lifelong immunosuppression (IS). Rarely, recipients display spontaneous "operational tolerance" with stable graft function in the absence of IS. The lack of biological markers of this phenomenon precludes identification of potentially tolerant patients in which IS could be tapered and hinders the development of new tolerance-inducing strategies. The objective of this study was to identify minimally invasive blood biomarkers for operational tolerance and use these biomarkers to determine the frequency of this state in immunosuppressed patients with stable graft function. Blood gene expression profiles from 75 renal-transplant patient cohorts (operational tolerance/acute and chronic rejection/stable graft function on IS) and 16 healthy individuals were analyzed. A subset of samples was used for microarray analysis where three-class comparison of the different groups of patients identified a "tolerant footprint" of 49 genes. These biomarkers were applied for prediction of operational tolerance by microarray and real-time PCR in independent test groups. Thirty-three of 49 genes correctly segregated tolerance and chronic rejection phenotypes with 99% and 86% specificity. The signature is shared with 1 of 12 and 5 of 10 stable patients on triple IS and low-dose steroid monotherapy, respectively. The gene signature suggests a pattern of reduced costimulatory signaling, immune quiescence, apoptosis, and memory T cell responses. This study identifies in the blood of kidney recipients a set of genes associated with operational tolerance that may have utility as a minimally invasive monitoring tool for guiding IS titration. Further validation of this tool for safe IS minimization in prospective clinical trials is warranted.
Collapse
Affiliation(s)
- Sophie Brouard
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Elaine Mansfield
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Stanford, CA 94304
- Affymetrix, Inc., 3380 Central Expressway, Santa Clara, CA 95051
| | - Christophe Braud
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Li Li
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Stanford, CA 94304
| | - Magali Giral
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Szu-chuan Hsieh
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Stanford, CA 94304
| | - Dominique Baeten
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
- Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; and
| | - Meixia Zhang
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Stanford, CA 94304
- Department of Clinical Pharmacology, China Medical University, Shenyang 110001, China
| | - Joanna Ashton-Chess
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Cécile Braudeau
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Frank Hsieh
- Department of Veterans Affairs Palo Alto Health Care System (151-K), Palo Alto, CA 94304
| | - Alexandre Dupont
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Annaik Pallier
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Anne Moreau
- Service d'Anatomie Pathologique, CHU Hôtel-Dieu, 30 Bd Jean Monnet, 44093 Nantes Cedex 01, France
| | - Stéphanie Louis
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Catherine Ruiz
- TcLand, Halle 13 Bio-Ouest Ile de Nantes, 21 Rue de la Noue Bras de Fer, 44200 Nantes, France
| | - Oscar Salvatierra
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Stanford, CA 94304
| | - Jean-Paul Soulillou
- Institut National de la Santé et de la Recherche Médicale, U643, Centre Hospitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation, and Université de Nantes, Faculte de Medicine, F-44000 Nantes, France
| | - Minnie Sarwal
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Stanford, CA 94304
| |
Collapse
|