251
|
Young JA, Becker AM, Medeiros JJ, Shapiro VS, Wang A, Farrar JD, Quill TA, van Huijsduijnen RH, van Oers NS. The protein tyrosine phosphatase PTPN4/PTP-MEG1, an enzyme capable of dephosphorylating the TCR ITAMs and regulating NF-kappaB, is dispensable for T cell development and/or T cell effector functions. Mol Immunol 2008; 45:3756-66. [PMID: 18614237 PMCID: PMC2596642 DOI: 10.1016/j.molimm.2008.05.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 05/27/2008] [Accepted: 05/28/2008] [Indexed: 11/16/2022]
Abstract
T cell receptor signaling processes are controlled by the integrated actions of families of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPases). Several distinct cytosolic protein tyrosine phosphatases have been described that are able to negatively regulate TCR signaling pathways, including SHP-1, SHP-2, PTPH1, and PEP. Using PTPase substrate-trapping mutants and wild type enzymes, we determined that PTPN4/PTP-MEG1, a PTPH1-family member, could complex and dephosphorylate the ITAMs of the TCR zeta subunit. In addition, the substrate-trapping derivative augmented basal and TCR-induced activation of NF-kappaB in T cells. To characterize the contribution of this PTPase in T cells, we developed PTPN4-deficient mice. T cell development and TCR signaling events were comparable between wild type and PTPN4-deficient animals. The magnitude and duration of TCR-regulated ITAM phosphorylation, as well as overall protein phosphorylation, was unaltered in the absence of PTPN4. Finally, Th1- and Th2-derived cytokines and in vivo immune responses to Listeria monocytogenes were equivalent between wild type and PTPN4-deficient mice. These findings suggest that additional PTPases are involved in controlling ITAM phosphorylations.
Collapse
Affiliation(s)
- Jennifer A. Young
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amy M. Becker
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jennifer J. Medeiros
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Virginia S. Shapiro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Andrew Wang
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - J. David Farrar
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Timothy A. Quill
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Nicolai S.C. van Oers
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
252
|
Tailor P, Tsai S, Shameli A, Serra P, Wang J, Robbins S, Nagata M, Szymczak-Workman AL, Vignali DAA, Santamaria P. The proline-rich sequence of CD3epsilon as an amplifier of low-avidity TCR signaling. THE JOURNAL OF IMMUNOLOGY 2008; 181:243-55. [PMID: 18566390 DOI: 10.4049/jimmunol.181.1.243] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Engagement of peptide-MHC by the TCR induces a conformational change in CD3epsilon that exposes a proline-rich sequence (PRS) and recruits the cytoskeletal adaptor Nck. This event, which precedes phosphorylation of the CD3epsilon ITAM, has been implicated in synapse formation and T cell function. However, there is compelling evidence that responsiveness to TCR ligation is CD3epsilon PRS independent. In this study, we show that the CD3epsilon PRS is necessary for peptide-MHC-induced phosphorylation of CD3epsilon and for recruitment of protein kinase Ctheta to the immune synapse in differentiated CD8+ T lymphocytes. However, whereas these two events are dispensable for functional T cell responsiveness to high-avidity ligands, they are required for responsiveness to low-avidity ones. Thus, in at least certain T cell clonotypes, the CD3epsilon PRS amplifies weak TCR signals by promoting synapse formation and CD3epsilon phosphorylation.
Collapse
Affiliation(s)
- Pankaj Tailor
- Julia McFarlane Diabetes Research Centre and Department of Microbiology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
253
|
Peripheral CD8+ T cell tolerance to self-proteins is regulated proximally at the T cell receptor. Immunity 2008; 28:662-74. [PMID: 18424189 DOI: 10.1016/j.immuni.2008.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 02/12/2008] [Accepted: 03/04/2008] [Indexed: 11/20/2022]
Abstract
CD8(+) T cell tolerance, although essential for preventing autoimmunity, poses substantial obstacles to eliciting immune responses to tumor antigens, which are generally overexpressed normal proteins. Development of effective strategies to overcome tolerance for clinical applications would benefit from elucidation of the immunologic mechanism(s) regulating T cell tolerance to self. To examine how tolerance is maintained in vivo, we engineered dual-T cell receptor (TCR) transgenic mice in which CD8(+) T cells recognize two distinct antigens: a foreign viral-protein and a tolerizing self-tumor protein. Encounter with peripheral self-antigen rendered dual-TCR T cells tolerant to self, but these cells responded normally through the virus-specific TCR. Moreover, proliferation induced by virus rescued function of tolerized self-tumor-reactive TCR, restoring anti-tumor activity. These studies demonstrate that peripheral CD8(+) T cell tolerance to self-proteins can be regulated at the level of the self-reactive TCR complex rather than by central cellular inactivation and suggest an alternate strategy to enhance adoptive T cell immunotherapy.
Collapse
|
254
|
Gil D, Schrum AG, Daniels MA, Palmer E. A role for CD8 in the developmental tuning of antigen recognition and CD3 conformational change. THE JOURNAL OF IMMUNOLOGY 2008; 180:3900-9. [PMID: 18322198 DOI: 10.4049/jimmunol.180.6.3900] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR engagement by peptide-MHC class I (pMHC) ligands induces a conformational change (Deltac) in CD3 (CD3Deltac) that contributes to T cell signaling. We found that when this interaction took place between primary T lineage cells and APCs, the CD8 coreceptor was required to generate CD3Deltac. Interestingly, neither enhancement of Ag binding strength nor Src kinase signaling explained this coreceptor activity. Furthermore, Ag-induced CD3Deltac was developmentally attenuated by the increase in sialylation that accompanies T cell maturation and limits CD8 activity. Thus, both weak and strong ligands induced CD3Deltac in preselection thymocytes, but only strong ligands were effective in mature T cells. We propose that CD8 participation in the TCR/pMHC interaction can physically regulate CD3Deltac induction by "translating" productive Ag encounter from the TCR to the CD3 complex. This suggests one mechanism by which the developmentally regulated variation in CD8 sialylation may contribute to the developmental tuning of T cell sensitivity.
Collapse
Affiliation(s)
- Diana Gil
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
255
|
Takeuchi K, Yang H, Ng E, Park SY, Sun ZYJ, Reinherz EL, Wagner G. Structural and functional evidence that Nck interaction with CD3epsilon regulates T-cell receptor activity. J Mol Biol 2008; 380:704-16. [PMID: 18555270 DOI: 10.1016/j.jmb.2008.05.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 05/14/2008] [Accepted: 05/15/2008] [Indexed: 01/28/2023]
Abstract
Recruitment of signaling molecules to the cytoplasmic domains of the CD3 subunits of the T-cell receptor (TCR) is crucial for early T-cell activation. These transient associations either do or do not require tyrosine phosphorylation of CD3 immune tyrosine activation motifs (ITAMs). Here we show that the non-ITAM-requiring adaptor protein Nck forms a complex with an atypical PxxDY motif of the CD3epsilon tail, which encompasses Tyr166 within the ITAM and a TCR endocytosis signal. As suggested by the structure of the complex, we find that Nck binding inhibits phosphorylation of the CD3epsilon ITAM by Fyn and Lck kinases in vitro. Moreover, the CD3epsilon-Nck interaction downregulates TCR surface expression upon physiological stimulation in mouse primary lymph node cells. This indicates that Nck performs an important regulatory function in T lymphocytes by inhibiting ITAM phosphorylation and/or removing cell surface TCR via CD3epsilon interaction.
Collapse
Affiliation(s)
- Koh Takeuchi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
256
|
Sebestyén Z, Schooten E, Sals T, Zaldivar I, San José E, Alarcón B, Bobisse S, Rosato A, Szöllősi J, Gratama JW, Willemsen RA, Debets R. Human TCR That Incorporate CD3ζ Induce Highly Preferred Pairing between TCRα and β Chains following Gene Transfer. THE JOURNAL OF IMMUNOLOGY 2008; 180:7736-46. [DOI: 10.4049/jimmunol.180.11.7736] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
257
|
|
258
|
Abstract
The interaction between the T cell receptor (TCR) and a peptide-loaded major histocompatibility complex (pMHC) is one of the most-studied interactions in immunology, and yet the precise mechanism by which this system operates is still not fully understood. One key issue is whether TCR triggering minimally requires monomeric pMHC complexes or higher-order multimers (two or more pMHCs). Any model of TCR triggering must explain the high sensitivity, specificity, and dynamic range of ligand responsiveness that this receptor system exhibits. Most models of TCR triggering have not fully appreciated the dynamic aspects of TCR triggering. TCR triggering happens very quickly, and the properties of sensitivity and specificity can be explained by a model that accounts for the interaction dynamics of such a receptor system. In this paper, it is proposed that the important parameter in TCR triggering is the immobilization of the TCR-pMHC complex in the plasma membrane. Whether this involves monomeric or multimeric pMHCs may depend on the affinity of the TCR for the pMHC.
Collapse
Affiliation(s)
- Rajat Varma
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
259
|
Abstract
Although dynamic imaging technologies have provided important insights into the underlying processes responsible for T-cell activation, the processes that link antigen recognition to downstream signaling remain poorly defined. Converging lines of inquiry indicate that T-cell receptor (TCR) microclusters are the minimal structures capable of directing effective TCR signaling. Furthermore, imaging studies have determined that these structures trigger the assembly of oligomeric signaling scaffolds that contain the adapters and effectors required for T-cell activation. Existing models of T-cell activation accurately explain the sensitivity and selectivity of antigen recognition. However, these models do not account for important properties of microclusters, including their peripheral formation, size, and movement on the actin cytoskeleton. Here we examine how lipid rafts, galectin lattices, and protein scaffolds contribute to the assembly, function, and fate of TCR microclusters within immune synapses. Finally, we propose a 'mechanical segregation' model of signal initiation in which cytoskeletal forces contribute to the lateral segregation of molecules and cytoskeletal scaffolds provide a template for microclusters assembly.
Collapse
|
260
|
Mingueneau M, Sansoni A, Grégoire C, Roncagalli R, Aguado E, Weiss A, Malissen M, Malissen B. The proline-rich sequence of CD3epsilon controls T cell antigen receptor expression on and signaling potency in preselection CD4+CD8+ thymocytes. Nat Immunol 2008; 9:522-32. [PMID: 18408722 DOI: 10.1038/ni.1608] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Accepted: 03/13/2008] [Indexed: 12/11/2022]
Abstract
Antigen recognition by T cell antigen receptors (TCRs) is thought to 'unmask' a proline-rich sequence (PRS) present in the CD3epsilon cytosolic segment, which allows it to trigger T cell activation. Using 'knock-in' mice with deletion of the PRS, we demonstrate here that elimination of the CD3epsilon PRS had no effect on mature T cell responsiveness. In contrast, in preselection CD4+CD8+ thymocytes, the CD3epsilon PRS acted together with the adaptor protein SLAP to promote CD3zeta degradation, thereby contributing to downregulation of TCR expression on the cell surface. In addition, analysis of CD4+CD8+ thymocytes of TCR-transgenic mice showed that the CD3epsilon PRS enhanced TCR sensitivity to weak ligands. Our results identify previously unknown functions for the evolutionarily conserved CD3epsilon PRS at the CD4+CD8+ developmental stage and suggest a rather limited function in mature T cells.
Collapse
Affiliation(s)
- Michaël Mingueneau
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerrannée, Case 906, Institut National de la Santé et de la Recherche Médicale U631, and Centre National de la Recherche Scientifique UMR6102, 13288 Marseille Cedex 9, France
| | | | | | | | | | | | | | | |
Collapse
|
261
|
Risueño RM, Schamel WWA, Alarcón B. T cell receptor engagement triggers its CD3epsilon and CD3zeta subunits to adopt a compact, locked conformation. PLoS One 2008; 3:e1747. [PMID: 18320063 PMCID: PMC2254190 DOI: 10.1371/journal.pone.0001747] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 10/08/2007] [Indexed: 11/24/2022] Open
Abstract
How the T cell antigen receptor (TCR) discriminates between molecularly related peptide/Major Histocompatibility Complex (pMHC) ligands and converts this information into different possible signaling outcomes is still not understood. One current model proposes that strong pMHC ligands, but not weak ones, induce a conformational change in the TCR. Evidence supporting this comes from a pull-down assay that detects ligand-induced binding of the TCR to the N-terminal SH3 domain of the adapter protein Nck, and also from studies with a neoepitope-specific antibody. Both methods rely on the exposure of a polyproline sequence in the CD3ε subunit of the TCR, and neither indicates whether the conformational change is transmitted to other CD3 subunits. Using a protease-sensitivity assay, we now show that the cytoplasmic tails of CD3ε and CD3ζ subunits become fully protected from degradation upon TCR triggering. These results suggest that the TCR conformational change is transmitted to the tails of CD3ε and CD3ζ, and perhaps all CD3 subunits. Furthermore, the resistance to protease digestion suggests that CD3 cytoplasmic tails adopt a compact structure in the triggered TCR. These results are consistent with a model in which transduction of the conformational change induced upon TCR triggering promotes condensation and shielding of the CD3 cytoplasmic tails.
Collapse
Affiliation(s)
- Ruth M. Risueño
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
262
|
The extracellular part of ζ is buried in the T cell antigen receptor complex. Immunol Lett 2008; 116:203-10. [DOI: 10.1016/j.imlet.2007.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 11/27/2007] [Accepted: 11/30/2007] [Indexed: 11/22/2022]
|
263
|
A permissive geometry model for TCR–CD3 activation. Trends Biochem Sci 2008; 33:51-7. [DOI: 10.1016/j.tibs.2007.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 10/09/2007] [Accepted: 10/19/2007] [Indexed: 01/23/2023]
|
264
|
|
265
|
Abstract
More than a quarter of a century has passed since the observation that T cells rapidly polarize their actin and microtubule cytoskeletal systems toward antigen-presenting cells during activation. Since this initial discovery, several receptors on T cells (e.g., T cell receptor [TCR], co-receptors, integrins, and chemokine receptors) have been identified to regulate these two cytoskeletal networks through complex signaling pathways, which are still being elucidated. There is now an undeniable body of biochemical, pharmacological, and genetic evidence indicating that regulators of actin and microtubule dynamics are crucial for T cell activation and effector functions. In fact, the actin cytoskeleton participates in the initial clustering of TCR-major histocompatibility complex or peptide complexes, formation and stabilization of the immune synapse, integrin-mediated adhesion, and receptor sequestration, whereas both the actin and microtubule cytoskeletons regulate the establishment of cell polarity, cell migration, and directed secretion of cytokines and cytolytic granules. Over the past several years, we have begun to more thoroughly understand the contributions of specific actin-regulatory and actin-nucleating proteins that govern these processes. Herein, we discuss our current understanding of how activating receptors on T lymphocytes regulate the actin and microtubule cytoskeletons, and how in turn, these distinct but integrated cytoskeletal networks coordinate T cell immune responses.
Collapse
|
266
|
Minguet S, Swamy M, Schamel WWA. The short length of the extracellular domain of zeta is crucial for T cell antigen receptor function. Immunol Lett 2007; 116:195-202. [PMID: 18207249 DOI: 10.1016/j.imlet.2007.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 11/27/2007] [Accepted: 11/30/2007] [Indexed: 11/15/2022]
Abstract
The T cell antigen receptor (TCR-CD3) consists of the pMHC-binding TCRalphabeta heterodimer and the signalling dimers CD3deltaepsilon, CD3gammaepsilon and zetazeta. The very short length of the extracellular domain (EC) of the zeta chain is preserved through evolution, however a rational explanation for this observation has not been elucidated. Here, we show that TCR-CD3 assembly is clearly defective when the murine zeta EC domain is artificially enlarged. Under these conditions, the TCR-CD3 complex is super-competent in transducing activation signals upon engagement. Furthermore, the TCR-CD3 complexes containing enlarged zeta EC domains underwent ligand-induced conformation changes with higher efficiency than TCR-CD3 complexes with an unmodified zeta EC domain. Together these data suggest that a short zeta EC domain is needed to correctly assemble the TCR-CD3 complex. When this domain is enlarged, the resulting TCR-CD3 complex is distorted leading to a hyperactive phenotype and enhanced T cell activation.
Collapse
Affiliation(s)
- Susana Minguet
- Max Planck Institute of Immunbiology and Faculty of Biology III, University of Freiburg, Stübeweg 51, 79108 Freiburg, Germany
| | | | | |
Collapse
|
267
|
|
268
|
Amon MA, Manolios N. Hypothesis: TCR signal transduction--A novel tri-modular signaling system. Mol Immunol 2007; 45:876-80. [PMID: 17915329 DOI: 10.1016/j.molimm.2007.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 08/10/2007] [Indexed: 11/21/2022]
Abstract
Antigenic peptides initiate an immune response in T cells via the T cell receptor (TCR). The TCR itself is widely regarded as one of the most complex receptors in nature, as it is comprised of at least six different subunits, the antigen recognizing TCRalpha and beta chains, and the signal transmitting CD3deltavarepsilon, gammaepsilon, and zeta2 dimers. In order for a signal to be transmitted from the TCR to the cytoplasm, the CD3 chains must "sense" that an antigenic peptide has been presented to the TCRalpha and beta subunits. After accomplishing this, there are a total of 10 different immunoreceptor tyrosine activation motifs (ITAMs) present within the CD3 chains which effectively activate the T cell and hence the immune response. The importance of each CD3 chain and subsequently each ITAM has been the focus of intense research. However, the precise role(s) played by each CD3 chain has remained elusive. Using the immunomodulatory peptide termed core peptide (CP), which is proposed to inhibit TCR activation by disrupting TCR-CD3 interactions, a tri-modular signaling system for T cell activation is proposed. By contrast to the existing two distinct signaling model (zeta2, CD3epsilongamma/epsilondelta), in this model each of the three dimers, CD3gammaepsilon, deltaepsilon, and zeta2, are proposed to act as three separate and distinct signaling modules, performing both specific and redundant functions.
Collapse
Affiliation(s)
- Michael A Amon
- Rheumatology Department, Westmead Hospital, Westmead, NSW 2145, Australia
| | | |
Collapse
|
269
|
Chen IJ, Chen HL, Demetriou M. Lateral compartmentalization of T cell receptor versus CD45 by galectin-N-glycan binding and microfilaments coordinate basal and activation signaling. J Biol Chem 2007; 282:35361-72. [PMID: 17897956 DOI: 10.1074/jbc.m706923200] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Lateral compartmentalization of membrane proteins into microdomains regulates signal transduction; however, structural determinants are incompletely understood. Membrane glycoproteins bind galectins in proportion to the number (i.e. NX(S/T) sites) and degree of GlcNAc branching within attached N-glycans, forming a molecular lattice that negatively regulates T cell function and autoimmunity. We find that in resting T cells, partition of CD45 inside and T cell receptor (TCR)/CD4-Lck/Zap-70 outside microdomains is positively and negatively regulated by the galectin lattice and actin cytoskeleton, respectively. In the absence of TCR ligands, the galectin lattice counteracts F-actin to retain CD45 in microdomains while concurrently blocking TCR/CD4-Lck/Zap-70 partition to microdomains by preventing a conformational change in the TCR that recruits Nck/Wiscott Aldrich Syndrome (WASp)/SLP76/F-actin/CD4 to TCR. The counterbalancing activities of the galectin lattice and actin cytoskeleton negatively and positively regulate Lck activity in resting cells and CD45 versus TCR clustering and signaling at the early immune synapse, respectively. Microdomain-localized CD45 inactivates Lck and inhibits TCR signaling at the early immune synapse. Thus, the galectin lattice and actin cytoskeleton interact on opposing sides of the plasma membrane to control microdomain structure and function, coupling basal growth signaling with thresholds to activation.
Collapse
Affiliation(s)
- I-Ju Chen
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California 92697, USA
| | | | | |
Collapse
|
270
|
Kesti T, Ruppelt A, Wang JH, Liss M, Wagner R, Taskén K, Saksela K. Reciprocal regulation of SH3 and SH2 domain binding via tyrosine phosphorylation of a common site in CD3epsilon. THE JOURNAL OF IMMUNOLOGY 2007; 179:878-85. [PMID: 17617578 DOI: 10.4049/jimmunol.179.2.878] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recruitment of cellular signaling proteins by the CD3 polypeptides of the TCR complex mediates T cell activation. We have screened a human Src homology 3 (SH3) domain phage display library for proteins that can bind to the proline-rich region of CD3epsilon. This screening identified Eps8L1 (epidermal growth factor receptor pathway substrate 8-like 1) together with the N-terminal SH3 domain of Nck1 and Nck2 as its preferred SH3 partners. Studies with recombinant proteins confirmed strong binding of CD3epsilon to Eps8L1 and Nck SH3 domains. CD3epsilon bound well also to Eps8 and Eps8L3, and modestly to Eps8L2, but not detectably to other SH3 domains tested. Interestingly, binding of Nck and Eps8L1 SH3 domains was mapped to a PxxDY motif that shared its tyrosine residue (Y166) with the ITAM of CD3epsilon. Phosphorylation of this residue abolished binding of Eps/Nck SH3 domains in peptide spot filter assays, as well as in cells cotransfected with a dominantly active Lck kinase. TCR ligation-induced binding and phosphorylation-dependent loss of binding were also demonstrated between Eps8L1 and endogenous CD3epsilon in Jurkat T cells. Thus, phosphorylation of Y166 serves as a molecular switch during T cell activation that determines the capacity of CD3epsilon to interact with either SH3 or SH2 domain-containing proteins.
Collapse
Affiliation(s)
- Tapio Kesti
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
271
|
Feinerman O, Germain RN, Altan-Bonnet G. Quantitative challenges in understanding ligand discrimination by alphabeta T cells. Mol Immunol 2007; 45:619-31. [PMID: 17825415 PMCID: PMC2131735 DOI: 10.1016/j.molimm.2007.03.028] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2007] [Accepted: 03/02/2007] [Indexed: 11/30/2022]
Affiliation(s)
- Ofer Feinerman
- ImmunoDynamics Group – Program in Computational Biology & Immunology – Memorial Sloan-Kettering Cancer Center – New York NY – USA
| | - Ronald N. Germain
- Lymphocyte Biology Section – Laboratory of Immunology – National Institute of Allergy and Infectious Disease – National Institute of Health – Bethesda MD - USA
| | - Grégoire Altan-Bonnet
- ImmunoDynamics Group – Program in Computational Biology & Immunology – Memorial Sloan-Kettering Cancer Center – New York NY – USA
- Corresponding author:
| |
Collapse
|
272
|
Rubin B, Knibiehler M, Gairin JE. Allosteric Changes in the TCR/CD3 Structure Upon Interaction With Extra- or Intra-cellular Ligands. Scand J Immunol 2007; 66:228-37. [PMID: 17635800 DOI: 10.1111/j.1365-3083.2007.01979.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T lymphocytes are activated by the interaction between the T-cell antigen receptor (TCR) and peptides presented by major histocompatibility complex (MHC) molecules. The avidity of this TCR-pMHC interaction is very low. Therefore, several hypotheses have been put forward to explain how T cells become specifically activated despite this handicap: conformational change model, aggregation model, kinetic segregation model, sequential interaction model and permissive geometry model. In the present paper, we conducted experiments to distinguish between the TCR-aggregation model and the TCR-conformational change model. The results obtained using a TCR capture ELISA with Brij 98-solubilized TCR molecules from normal or activated T cells showed that the ligand-TCR interaction causes structural changes in the CD3 epsilon cytoplasmic tail as well as in the extracellular TCR beta FG loop region. Size-fractionation experiments with Brij 98-solubilized TCR/CD3/co-receptor complexes from naïve or activated CD4(+) or CD8(+) T cells demonstrated that such complexes are found as either dimers or tetramers. No monomers or multimers were detected. We propose that: (1) ligand-TCR interaction results in conformational changes in the CD3 epsilon cytoplasmic tail leading to T-cell activation; (2) CD3 epsilon cytoplasmic tail interaction with intracellular proteins may dissociate pMHC and co-receptors (CD4 or CD8) from TCR/CD3 complexes, thus leading to the arrest of T-cell activation; and (3) T-cell activation appears to occur among dimers or tetramers of TCR/CD3/co-receptor complexes interacting with self and non-self (foreign) peptide-MHC complexes.
Collapse
MESH Headings
- Allosteric Regulation/immunology
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/metabolism
- CD3 Complex/chemistry
- CD3 Complex/genetics
- CD3 Complex/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Enzyme-Linked Immunosorbent Assay
- Extracellular Fluid/metabolism
- Intracellular Fluid/metabolism
- Ligands
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Molecular Sequence Data
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
Collapse
Affiliation(s)
- B Rubin
- Institut de Sciences et Technologies du Médicament de Toulouse (ISTMT), Toulouse, France.
| | | | | |
Collapse
|
273
|
Duchardt E, Sigalov AB, Aivazian D, Stern LJ, Schwalbe H. Structure induction of the T-cell receptor zeta-chain upon lipid binding investigated by NMR spectroscopy. Chembiochem 2007; 8:820-7. [PMID: 17410622 DOI: 10.1002/cbic.200600413] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The conformation of the cytoplasmic part of the zeta-chain of the T-cell receptor (TCR) in its free form and bound to detergent micelles has been investigated by heteronuclear NMR spectroscopy. The zeta-chain is considered to be a mediator between the extracellular antigen and the intracellular signal-transduction cascade leading to T-cell activation. Earlier studies suggested a T-cell activation mechanism that involved a TCR-state-dependent lipid incorporation propensity of the zeta-chain accompanied by a helical folding transition. In order to support this proposed mechanism, standard protein NMR assignment and secondary-structure-elucidation techniques have been applied to the free TCR zeta-chain and to the zeta-chain bound to the detergent LMPG, which forms a micelle, in order to obtain the structural characteristics of this folding transition in a residue-resolved manner. We could assign the resonances of the free zeta-chain at 278 K, and this formed the basis for chemical-shift-perturbation studies to identify lipid binding sites. Our NMR results show that the free TCR zeta-chain is indeed intrinsically unstructured. Regions around the ITAM2 and ITAM3 sequences are involved in a highly dynamic binding of the free zeta-chain to a detergent micelle formed by the acidic lipid LMPG.
Collapse
Affiliation(s)
- Elke Duchardt
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University Frankfurt, 60439 Frankfurt, Germany
| | | | | | | | | |
Collapse
|
274
|
Haverson K, Rehakova Z, Sinkora J, Sver L, Bailey M. Immune development in jejunal mucosa after colonization with selected commensal gut bacteria: a study in germ-free pigs. Vet Immunol Immunopathol 2007; 119:243-53. [PMID: 17643495 DOI: 10.1016/j.vetimm.2007.05.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 03/17/2007] [Accepted: 05/31/2007] [Indexed: 12/27/2022]
Abstract
The immunological structure of the porcine jejunal lamina propria in germ-free piglets was compared with that of their counterparts associated with two strains of commensal Escherichia coli, A0 34/86 serotype O83:K24:H31 and the O86 E. coli strain, up to 20 days post-colonization. In the antigen-presenting compartment, both dendritic cells (DC) and cells expressing CD163, probably macrophages were investigated. In addition we also assessed the number of CD2+/CD3+ (T) cells. In contrast to some previous reports, we show a total lack of both DC and T cells for germ-free animals in the diffuse lymphoid tissue of villi and crypts of the jejunum. Association with either strain of commensal E. coli had a profound effect on the immune structure and resulted in extensive recruitment of DC to the lamina propria and of T cells to epithelium and lamina propria. The data suggest that the earliest immigrant cells were monocytes, which soon acquired the phenotype of mucosal DC. T cells migrated in at a slightly slower rate. Nevertheless, the response could be extremely rapid: within 3 days of colonization with O83, the magnitude of this response was comparable to that observed 20 days post-colonization.
Collapse
Affiliation(s)
- Karin Haverson
- Department of Clinical Veterinary Science, University of Bristol, Langford BS40 5DU, UK.
| | | | | | | | | |
Collapse
|
275
|
Bello R, Feito MJ, Ojeda G, Portolés P, Rojo JM. Loss of N-terminal charged residues of mouse CD3 epsilon chains generates isoforms modulating antigen T cell receptor-mediated signals and T cell receptor-CD3 interactions. J Biol Chem 2007; 282:22324-34. [PMID: 17561508 DOI: 10.1074/jbc.m701875200] [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] [Indexed: 11/06/2022] Open
Abstract
The antigen T cell receptor (TCR)-CD3 complexes present on the cell surface of CD4(+) T lymphocytes and T cell lines express CD3 epsilon chain isoforms with different isoelectric points (pI), with important structural and functional consequences. The pI values of the isoforms fit the predicted pI values of CD3 epsilon chains lacking one, two, and three negatively charged amino acid residues present in the N-terminal region. Different T cells have different ratios of CD3 epsilon chain isoforms. At a high pI, degraded CD3 epsilon isoforms can be better recognized by certain anti-CD3 monoclonal antibodies such as YCD3-1, the ability of which to bind to the TCR-CD3 complex is directly correlated with the pI of CD3 epsilon. The abundance of CD3 epsilon isoforms can be modified by treatment of T cells with the proteinase inhibitor phenanthroline. In addition, these CD3 epsilon isoforms have functional importance. This is shown, first, by the different structure of TCR-CD3 complexes in cells possessing different amounts of isoforms (as observed in surface biotinylation experiments), by their different antigen responses, and by the stronger interaction between low pI CD3 epsilon isoforms and the TCR. Second, incubation of cells with phenanthroline diminished the proportion of degraded high pI CD3 epsilon isoforms, but also the ability of the cells to deliver early TCR activation signals. Third, cells expressing mutant CD3 epsilon chains lacking N-terminal acid residues showed facilitated recognition by antibody YCD3-1 and enhanced TCR-mediated activation. Furthermore, the binding avidity of antibody YCD3-1 was different in distinct thymus populations. These results suggest that changes in CD3 epsilon N-terminal chains might help to fine-tune the response of the TCR to its ligands in distinct activation situations or in thymus selection.
Collapse
Affiliation(s)
- Raquel Bello
- Departamento de Fisiopatología Celular y Molecular, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, E-28040 Madrid, Spain
| | | | | | | | | |
Collapse
|
276
|
Choudhuri K, van der Merwe PA. Molecular mechanisms involved in T cell receptor triggering. Semin Immunol 2007; 19:255-61. [PMID: 17560121 DOI: 10.1016/j.smim.2007.04.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 04/14/2007] [Indexed: 12/19/2022]
Abstract
Despite intensive investigation we still do not understand how the T cell antigen receptor (TCR) tranduces signals across the plasma membrane, a process referred to as TCR triggering. Three basic mechanisms have been proposed, involving aggregation, conformational change, or segregation of the TCR upon binding pMHC ligand. Given the low density of pMHC ligand it remains doubtful that TCR aggregation initiates triggering, although it is likely to enhance subsequent signalling. Structural studies to date have not provided definitive evidence for or against a conformational change mechanism, but they have ruled out certain types of conformational change. Size-induced segregation of the bound TCR from inhibitory membrane tyrosine phosphatases seems to be required, but is probably not the only mechanism. Current evidence suggests that TCR triggering is initiated by a combination of segregation and conformational change, with subsequent aggregation contributing to amplification of the signal.
Collapse
Affiliation(s)
- Kaushik Choudhuri
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, United Kingdom
| | | |
Collapse
|
277
|
Krogsgaard M, Juang J, Davis MM. A role for "self" in T-cell activation. Semin Immunol 2007; 19:236-44. [PMID: 17548210 PMCID: PMC2731063 DOI: 10.1016/j.smim.2007.04.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 04/16/2007] [Accepted: 04/17/2007] [Indexed: 11/24/2022]
Abstract
The mechanisms by which alphabeta T-cells are selected in the thymus and then recognize peptide MHC (pMHC) complexes in the periphery remain an enigma. Recent work particularly with respect to quantification of T-cell sensitivity and the role of self-ligands in T-cell activation has provided some important clues to the details of how TCR signaling might be initiated. Here, we highlight recent experimental data that provides insights into the initiation of T-cell activation and also discuss the main controversies and uncertainties in this area.
Collapse
Affiliation(s)
- Michelle Krogsgaard
- Department of Pathology and NYU Cancer Institute, NYU School of Medicine, New York, NY 10016, USA.
| | | | | |
Collapse
|
278
|
Swamy M, Minguet S, Siegers GM, Alarcón B, Schamel WWA. A native antibody-based mobility-shift technique (NAMOS-assay) to determine the stoichiometry of multiprotein complexes. J Immunol Methods 2007; 324:74-83. [PMID: 17568608 DOI: 10.1016/j.jim.2007.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 05/03/2007] [Accepted: 05/07/2007] [Indexed: 02/09/2023]
Abstract
Characterization of multiprotein complexes (MPCs) is an important step toward an integrative view of protein interaction networks and prerequisite for a molecular understanding of how a certain MPC functions. Here, we present a technique utilizing monoclonal subunit-specific antibodies for an electrophoretic immunoshift assay in Blue Native-gels (NAMOS-assay), which allows the determination of the stoichiometry of MPCs. First, we use the B cell antigen receptor as a model MPC whose stoichiometry is known, confirming the HC(2)LC(2)Igalpha/beta(1) stoichiometry. Second, we demonstrate that the digitonin-extracted T cell antigen receptor (TCR) extracted from T cells has a stoichiometry of alphabetaepsilon(2)gammadeltazeta(2). We then show that the NAMOS-assay does not require purified MPCs, since it can determine the stoichiometry of an MPC in cell lysates. The NAMOS-assay is also compatible with use of epitope tags appended to the protein of interest, as e.g. the widely used HA-tag, and anti-epitope antibodies for the assay. Given its general applicability, this method has a wide potential for MPC research.
Collapse
MESH Headings
- Animals
- Antibodies/metabolism
- Cell Line
- Cell Line, Tumor
- Electrophoresis, Polyacrylamide Gel
- Electrophoretic Mobility Shift Assay
- Humans
- Immunoglobulin Heavy Chains/metabolism
- Immunoglobulin Light Chains/metabolism
- Mice
- Multiprotein Complexes/chemistry
- Multiprotein Complexes/immunology
- Multiprotein Complexes/metabolism
- Receptors, Antigen, B-Cell/chemistry
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Mahima Swamy
- Max Planck-Institut für Immunbiologie and Universität Freiburg, Biologie III, Stübeweg 51, 79108 Freiburg, Germany
| | | | | | | | | |
Collapse
|
279
|
Abstract
Stochastic and spatial aspects are becoming increasingly recognized as an important factor in T-cell activation. Activation occurs in an intrinsically noisy environment, requiring only a handful of agonist peptide-major histocompatibility complex molecules, thus making consideration of signal to noise of prime importance in understanding sensitivity and specificity. Furthermore, it is widely established that surface-bound ligands are more effective at activation than soluble forms, while surface patternation has highlighted the role of spatial relocation in activation. Here we consider the results of a number of models of T-cell activation, from a realistic model of kinetic segregation-induced T-cell receptor (TCR) triggering through to simple queuing theory models. These studies highlight the constraints on cell activation by a surface receptor that recruits kinases. Our analysis shows that TCR triggering based on trapping of bound TCRs in regions of close proximity that exclude large ectodomain-containing molecules, such as the phosphatases CD45 and CD148, can effectively reproduce known signaling characteristics and is a viable 'signal transduction' mechanism distinct from oligomerization and conformation-based mechanisms. A queuing theory analysis shows the interrelation between sensitivity and specificity, emphasizing that these are properties of individual cell functions and need not be, nor are likely to be, uniform across different functions. In fact, threshold-based mechanisms of detection are shown to be poor at ligand discrimination because, although they can be highly specific, that specificity is limited to a small range of peptide densities. Time integration mechanisms however are able to control noise effectively, while kinetic proofreading mechanisms endow them with good specificity properties. Thus, threshold mechanisms are likely to be important for rapidly detecting minimal signaling requirements, thus achieving efficient scanning of antigen-presenting cells. However, for good specificity, time integration on a scale of hours is required.
Collapse
Affiliation(s)
- Nigel J Burroughs
- Mathematics Institute and Warwick Systems Biology, University of Warwick, Coventry, UK.
| | | |
Collapse
|
280
|
DeFord-Watts LM, Young JA, Pitcher LA, van Oers NSC. The membrane-proximal portion of CD3 epsilon associates with the serine/threonine kinase GRK2. J Biol Chem 2007; 282:16126-34. [PMID: 17420248 DOI: 10.1074/jbc.m609418200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The activation of protein kinases is one of the primary mechanisms whereby T cell receptors (TCR) propagate intracellular signals. To date, the majority of kinases known to be involved in the early stages of TCR signaling are protein-tyrosine kinases such as Lck, Fyn, and ZAP-70. Here we report a constitutive association between the TCR and a serine/threonine kinase, which was mediated through the membrane-proximal portion of CD3 epsilon. Mass spectrometry analysis of CD3 epsilon-associated proteins identified G protein-coupled receptor kinase 2 (GRK2) as a candidate Ser/Thr kinase. Transient transfection assays and Western blot analysis verified the ability of GRK2 to interact with the cytoplasmic domain of CD3 epsilon within a cell. These findings are consistent with recent reports demonstrating the ability of certain G protein-coupled receptors (GPCR) and G proteins to physically associate with the alpha/beta TCR. Because GRK2 is primarily involved in arresting GPCR signals, its interaction with CD3 epsilon may provide a novel means whereby the TCR can negatively regulate signals generated through GPCRs.
Collapse
Affiliation(s)
- Laura M DeFord-Watts
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9093, USA
| | | | | | | |
Collapse
|
281
|
Choi S, Schwartz RH. Molecular mechanisms for adaptive tolerance and other T cell anergy models. Semin Immunol 2007; 19:140-52. [PMID: 17400472 PMCID: PMC2045643 DOI: 10.1016/j.smim.2007.02.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 02/16/2007] [Indexed: 01/09/2023]
Abstract
Since the original description of T cell anergy in CD4 clones from mice and humans, a number of different unresponsive states have been described, both in vivo and in vitro, that have been called anergic. While initial attempts were made to understand the similarities between the different models, it has now become clear from biochemical experiments that many of them have different molecular mechanisms underlying their unresponsiveness. In this review we will detail our own work on the in vivo model referred to as adaptive tolerance and then attempt to compare this biochemical state to the multitude of other states that have been described in the literature.
Collapse
Affiliation(s)
- Seeyoung Choi
- National Institutes of Health, LCMI, NIAID, Bethesda, MD 20892-0420, USA
| | | |
Collapse
|
282
|
Uhlin M, Masucci M, Levitsky V. Is the activity of partially agonistic MHC:peptide ligands dependent on the quality of immunological help? Scand J Immunol 2007; 64:581-7. [PMID: 17083613 DOI: 10.1111/j.1365-3083.2006.01850.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
CD8(+) cytotoxic T lymphocytes (CTL) are important for the immunological control of infections and tumours. Engagement of the T-cell receptor (TCR) with major histocompatibility complex (MHC) class I/peptide complexes on antigen-presenting cells (APC) is the key interaction, which initiates the process of T-cell activation. Depending on the affinity of this interaction, different arrays of signalling pathways and functional outcomes can be activated in the specific T cells. Molecular alterations in the peptide bound to the MHC class I can lead to a lower affinity of the MHC:TCR interaction resulting in incomplete or qualitatively different T-cell responses. Altered peptide ligands (APL) exhibiting such activity are referred to as partial agonists and often occur naturally through genetic instability, which affects T-cell epitopes derived from rapidly mutating viruses or tumour-associated cellular antigens. Partial agonists are usually viewed as peptide variants, which escape efficient CTL recognition. Our recent data suggest that APL can not only trigger incomplete activation but also induce and modulate intrinsic T-cell programmes leading to the shut-off of specific CTL responses. This APL-induced suppression appears to be more prominent in the absence of immunological help, suggesting that under conditions of immune deregulation APL may actively inhibit CTL responses against infectious agents or tumours. In this review, we discuss experimental data supporting this model and possible role of APL-induced immunosuppression in different pathological conditions.
Collapse
Affiliation(s)
- M Uhlin
- Department of Microbiology, Tumor and Cell Biology Center and, Karolinska Institutet, Stockholm, Sweden.
| | | | | |
Collapse
|
283
|
Minguet S, Swamy M, Alarcón B, Luescher IF, Schamel WWA. Full Activation of the T Cell Receptor Requires Both Clustering and Conformational Changes at CD3. Immunity 2007; 26:43-54. [PMID: 17188005 DOI: 10.1016/j.immuni.2006.10.019] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 10/10/2006] [Accepted: 10/27/2006] [Indexed: 11/26/2022]
Abstract
T cell receptor (TCR-CD3) triggering involves both receptor clustering and conformational changes at the cytoplasmic tails of the CD3 subunits. The mechanism by which TCRalphabeta ligand binding confers conformational changes to CD3 is unknown. By using well-defined ligands, we showed that induction of the conformational change requires both multivalent engagement and the mobility restriction of the TCR-CD3 imposed by the plasma membrane. The conformational change is elicited by cooperative rearrangements of two TCR-CD3 complexes and does not require accompanying changes in the structure of the TCRalphabeta ectodomains. This conformational change at CD3 reverts upon ligand dissociation and is required for T cell activation. Thus, our permissive geometry model provides a molecular mechanism that rationalizes how the information of ligand binding to TCRalphabeta is transmitted to the CD3 subunits and to the intracellular signaling machinery.
Collapse
Affiliation(s)
- Susana Minguet
- Max Planck-Institut für Immunbiologie and Faculty of Biology, University of Freiburg, Stübeweg 51, 79108 Freiburg, Germany
| | | | | | | | | |
Collapse
|
284
|
Clements CS, Dunstone MA, Macdonald WA, McCluskey J, Rossjohn J. Specificity on a knife-edge: the alphabeta T cell receptor. Curr Opin Struct Biol 2006; 16:787-95. [PMID: 17011774 DOI: 10.1016/j.sbi.2006.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 08/31/2006] [Accepted: 09/15/2006] [Indexed: 11/27/2022]
Abstract
The interaction between the alphabeta T cell receptor (TCR) and the peptide bound to the major histocompatibility complex class I molecule (pMHC-I) constitutes a central interaction in adaptive immunity. How these receptors interact with such low affinity while maintaining exquisite specificity for peptide antigen and host MHC (MHC-I restriction) remains a challenge to be explained by structural immunologists. Moreover, how this extracellular interaction is transmitted as an intracellular signal via the CD3 complex remains unresolved. Nevertheless, several structures of TCRs, non-liganded and ligated to a defined pMHC-I, combined with detailed biophysical analyses, have provided insight of the structural basis of MHC-I restriction. In addition, structures of isolated CD3 components have enabled T cell signalling mechanisms to be postulated. Recent findings in this area, which include seven distinct TCR/pMHC-I complexes, have fundamental implications in adaptive immunity as well as therapeutic applications to modulate the adaptive immune response.
Collapse
Affiliation(s)
- Craig S Clements
- The Protein Crystallography Unit, Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Victoria 3800, Australia
| | | | | | | | | |
Collapse
|
285
|
Yachi PP, Ampudia J, Zal T, Gascoigne NRJ. Altered Peptide Ligands Induce Delayed CD8-T Cell Receptor Interaction—a Role for CD8 in Distinguishing Antigen Quality. Immunity 2006; 25:203-11. [PMID: 16872849 DOI: 10.1016/j.immuni.2006.05.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Revised: 04/21/2006] [Accepted: 05/11/2006] [Indexed: 10/24/2022]
Abstract
How T cells translate T cell receptor (TCR) recognition of almost identical pMHC ligands into distinct biological responses has remained enigmatic. Although differences in affinity or off rate are important, they offer at best an incomplete explanation. By using Förster resonance energy transfer (FRET), we have visualized the ligand-induced interaction between OT-I TCR and CD8. We found that both recruitment of TCR to the immunological synapse and the TCR-CD8 interaction induced by weak agonists (positive-selecting ligands) was delayed but not necessarily weaker than strong agonists (negative selectors). A delayed and perhaps longer lasting CD8-TCR interaction results in delayed phospho-ERK recruitment to the synapse. The kinetics of the TCR-CD8 interaction can reconcile previously anomalous data, where biological activity did not correlate with TCR-pMHC binding kinetics for certain ligands. Our findings indicate that the T cell translates antigen recognition into T cell responses by differential recruitment of CD8 to the TCR.
Collapse
Affiliation(s)
- Pia P Yachi
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
286
|
Pitcher LA, Mathis MA, Young JA, DeFord LM, Purtic B, Wulfing C, van Oers NSC. The CD3 gamma epsilon/delta epsilon signaling module provides normal T cell functions in the absence of the TCR zeta immunoreceptor tyrosine-based activation motifs. Eur J Immunol 2006; 35:3643-54. [PMID: 16259006 DOI: 10.1002/eji.200535136] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
T cell receptor (TCR) signal transduction is mediated by the immunoreceptor tyrosine-based activation motifs (ITAM). The ten ITAM in the TCR complex are distributed in two distinct signaling modules termed TCR zetazeta and CD3 gammaepsilon/deltaepsilon. To delineate the specific role of the zeta ITAM in T cell development and TCR signal transmission, we compared the properties of T cells from different TCR zeta-transgenic lines wherein tyrosine-to-phenylalanine substitutions had been introduced in the zeta subunit. These lines lack selected phosphorylated forms of TCR zeta including just p23, both p21 and p23, or all phospho-zeta derivatives. We report herein that the efficiency of positive selection in HY TCR-transgenic female mice was directly related to the number of zeta ITAM in the TCR. In contrast, TCR-mediated signal transmission and T cell proliferative responses following agonist peptide stimulation were similar and independent of the zeta ITAM. Only the duration of MAPK activation was affected by multiple zeta ITAM substitutions. These results strongly suggest that the ITAM in the CD3 gammaepsilon/deltaepsilon module can provide normal TCR signal transmission, with zeta ITAM providing a secondary function facilitating MAPK activation and positive selection.
Collapse
Affiliation(s)
- Lisa A Pitcher
- Center for Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | |
Collapse
|
287
|
Risueño RM, van Santen HM, Alarcón B. A conformational change senses the strength of T cell receptor-ligand interaction during thymic selection. Proc Natl Acad Sci U S A 2006; 103:9625-30. [PMID: 16766661 PMCID: PMC1480457 DOI: 10.1073/pnas.0601785103] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
T cell antigen receptor (TCR) triggering determines the fate of immature thymocytes. The affinity of the TCR for its endogenous peptide/MHC ligands serves as a signal for positive or negative selection through mechanisms that are still little understood. We have used a conformation-specific antibody to demonstrate that recognition of TCR ligands that lead to negative selection induces a conformational change in the TCR in situ. In contrast, this conformational change is elicited in only a small percentage of immature thymocytes during positive selection. Using a TUNEL assay, we demonstrate that the conformational change in the TCR is strongly linked to activation of programmed cell death in conditions leading to negative selection. Furthermore, the few conformational change-positive thymocytes detected in conditions that preferably lead to positive selection are also TUNEL-positive. Thus, the conformational change in the TCR may underlie the discrimination of ligands leading to positive and negative selection.
Collapse
Affiliation(s)
- Ruth M. Risueño
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Hisse M. van Santen
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | |
Collapse
|
288
|
Burroughs NJ, Lazic Z, van der Merwe PA. Ligand detection and discrimination by spatial relocalization: A kinase-phosphatase segregation model of TCR activation. Biophys J 2006; 91:1619-29. [PMID: 16751250 PMCID: PMC1544308 DOI: 10.1529/biophysj.105.080044] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We develop a model of tyrosine phosphorylation and activation of the T-cell receptor (TCR) by localization to regions of close membrane-membrane proximity (close contact) that physically exclude tyrosine phosphatases such as CD45. Phosphatase exclusion generates regions of low phosphatase and high kinase activity and thus our model provides a framework to examine the kinetic segregation model of TCR activation. We incorporate a sequence of activation steps modeling the construction of the signalosome with a final sequestered, or high-stability, signaling state. The residence time of unbound TCRs in tyrosine kinase-rich domains is shown to be too short for accumulation of activation steps, whereas binding to an agonist lengthens the localization time and leads to generation of fully active TCRs. Agonist detection depends only on this localization, and therefore kinetic segregation represents a viable ligand detection mechanism, or signal transduction mechanism across membranes, distinct from receptor oligomerization and conformational change. We examine the degree of discrimination of agonists from a background of null (self) peptides, and from weak agonists achievable by this mechanism.
Collapse
Affiliation(s)
- Nigel J Burroughs
- Mathematics Institute, University of Warwick, Coventry, United Kingdom.
| | | | | |
Collapse
|
289
|
Kölsch U, Arndt B, Reinhold D, Lindquist JA, Jüling N, Kliche S, Pfeffer K, Bruyns E, Schraven B, Simeoni L. Normal T-cell development and immune functions in TRIM-deficient mice. Mol Cell Biol 2006; 26:3639-48. [PMID: 16612002 PMCID: PMC1447406 DOI: 10.1128/mcb.26.9.3639-3648.2006] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The transmembrane adaptor molecule TRIM is strongly expressed within thymus and in peripheral CD4(+) T cells. Previous studies suggested that TRIM is an integral component of the T-cell receptor (TCR)/CD3 complex and might be involved in regulating TCR cycling. To elucidate the in vivo function of TRIM, we generated TRIM-deficient mice by homologous recombination. TRIM(-/-) mice develop normally and are healthy and fertile. However, the animals show a mild reduction in body weight that appears to be due to a decrease in the size and/or cellularity of many organs. The morphology and anatomy of nonlymphoid as well as primary and secondary lymphoid organs is normal. The frequency of thymocyte and peripheral T-cell subsets does not differ from control littermates. In addition, a detailed analysis of lymphocyte development revealed that TRIM is not required for either positive or negative selection. Although TRIM(-/-) CD4(+) T cells showed an augmented phosphorylation of the serine/threonine kinase Akt, the in vitro characterization of peripheral T cells indicated that proliferation, survival, activation-induced cell death, migration, adhesion, TCR internalization and recycling, TCR-mediated calcium fluxes, tyrosine phosphorylation, and mitogen-activated protein family kinase activation are not affected in the absence of TRIM. Similarly, the in vivo immune response to T-dependent and T-independent antigens as well as the clinical course of experimental autoimmune encephalomyelitis, a complex Th1-mediated autoimmune model, is comparable to that of wild-type animals. Collectively, these results demonstrate that TRIM is dispensable for T-cell development and peripheral immune functions. The lack of an evident phenotype could indicate that TRIM shares redundant functions with other transmembrane adaptors involved in regulating the immune response.
Collapse
Affiliation(s)
- Uwe Kölsch
- Institute of Immunology, Otto von Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
Lettau M, Qian J, Linkermann A, Latreille M, Larose L, Kabelitz D, Janssen O. The adaptor protein Nck interacts with Fas ligand: Guiding the death factor to the cytotoxic immunological synapse. Proc Natl Acad Sci U S A 2006; 103:5911-6. [PMID: 16595635 PMCID: PMC1458672 DOI: 10.1073/pnas.0508562103] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Fas ligand (FasL) is a key death factor of cytotoxic T lymphocytes and natural killer cells. It is stored intracellularly as a transmembrane protein of secretory lysosomes. Upon activation, these vesicles are transported to the cytotoxic immunological synapse (IS), and FasL becomes exposed to the cell surface to trigger cell death through ligation of its receptor Fas (CD95) on the target cell. We propose that the FasL-associated adaptor protein Nck is involved in the actin-dependent transport of FasL-bearing secretory lysosomes to the IS. Nck binds to the proline-rich portion of FasL and alters its subcellular distribution when coexpressed in 293T cells. In T lymphocytes, endogenous Nck partially colocalizes with lysosome-associated FasL. When T cell clones or lines are exposed to target cells, both proteins and other components of secretory lysosomes (i.e., granzyme B or cathepsin D) are transported to the cell-cell interface. The present data suggest that T cell receptor engagement provokes a rapid, tyrosine kinase- and actin-dependent transport of Nck-associated FasL-carrying lysosomes to the contact area. Our observations support the previous notion that the unique cytoplasmic tail of FasL is crucial for its directed transport to the cell surface and into the assembling cytotoxic IS.
Collapse
Affiliation(s)
- Marcus Lettau
- *Institute for Immunology, University Hospital Schleswig–Holstein Campus Kiel, 24105 Kiel, Germany; and
| | - Jing Qian
- *Institute for Immunology, University Hospital Schleswig–Holstein Campus Kiel, 24105 Kiel, Germany; and
| | - Andreas Linkermann
- *Institute for Immunology, University Hospital Schleswig–Holstein Campus Kiel, 24105 Kiel, Germany; and
| | - Mathieu Latreille
- Polypeptide Laboratory, McGill University, Montreal, QC, Canada H3A 2B2
| | - Louise Larose
- Polypeptide Laboratory, McGill University, Montreal, QC, Canada H3A 2B2
| | - Dieter Kabelitz
- *Institute for Immunology, University Hospital Schleswig–Holstein Campus Kiel, 24105 Kiel, Germany; and
| | - Ottmar Janssen
- *Institute for Immunology, University Hospital Schleswig–Holstein Campus Kiel, 24105 Kiel, Germany; and
- To whom correspondence should be addressed at: Institute for Immunology, University Hospital Schleswig–Holstein Campus Kiel, Michaelisstrasse 5, 24105 Kiel, Germany. E-mail:
| |
Collapse
|
291
|
Schamel WWA, Risueño RM, Minguet S, Ortíz AR, Alarcón B. A conformation- and avidity-based proofreading mechanism for the TCR–CD3 complex. Trends Immunol 2006; 27:176-82. [PMID: 16527543 DOI: 10.1016/j.it.2006.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Revised: 01/24/2006] [Accepted: 02/16/2006] [Indexed: 10/24/2022]
Abstract
During antigen recognition, T cells show high sensitivity and specificity, and a wide dynamic range. Paradoxically, these characteristics are based on low-affinity receptor-ligand interactions [between the T-cell antigen receptor (TCR-CD3) complex and the antigen peptide bound to MHC]. Recent evidence indicates that the TCR-CD3 is expressed as multivalent complexes in the membrane of non-stimulated T cells and that conformational changes in the TCR-CD3 can be induced by strong but not weak agonists. Here, we propose a thermodynamic model whereby the specificity of the TCR-CD3-pMHC interaction is explained by its multivalent nature. We also propose that the free energy barriers involved in the change in conformation of the receptor impose a response threshold and determine the kinetic properties of recognition. Finally, we suggest that multivalent TCR-CD3s can amplify signals by spreading them from pMHC-engaged TCR-CD3s to unengaged complexes as a consequence of the cooperativity in the system.
Collapse
Affiliation(s)
- Wolfgang W A Schamel
- Max Planck-Institut für Immunbiologie and University of Freiburg, Stübeweg 51, 79108 Freiburg, Germany
| | | | | | | | | |
Collapse
|
292
|
Stone JD, Stern LJ. CD8 T cells, like CD4 T cells, are triggered by multivalent engagement of TCRs by MHC-peptide ligands but not by monovalent engagement. THE JOURNAL OF IMMUNOLOGY 2006; 176:1498-505. [PMID: 16424178 DOI: 10.4049/jimmunol.176.3.1498] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell activation is initiated by recognition of antigenic peptide presented in complex with MHC molecules on the surface of APCs. The mechanism by which this recognition occurs is still unclear, and many models exist in the literature. CD4 T cells have been shown to respond to soluble oligomers of activating class II MHC-peptide complexes, but not to soluble monomers. In determining the reactivity of CD8 T cells to soluble activating class I MHC-peptide complexes, a complicating phenomenon had been observed whereby peptide from soluble complexes was loaded onto cell surface MHCs on the T cells and re-presented to other T cells, clouding the true valency requirement for activation. This study uses soluble allogeneic class I MHC-peptide monomers and oligomers to stimulate murine CD8 T cells without the possible complication of peptide re-presentation. The results show that MHC class I monomers bind to, but do not activate, CD8 T cells whether the cells are in solution or adhered to a surface. Monomeric MHC class I binding can antagonize the stimulation triggered by soluble oligomers, a phenomenon also observed for CD4 T cells. Dimeric engagement is necessary and sufficient to stimulate downstream activation processes including TCR down-regulation, Zap70 phosphorylation, and CD25 and CD69 up-regulation, even in T cells that do not express the MHC coreceptor CD8. Thus, the valency dependence of the response of CD8 T cells to soluble MHC-peptide reagents is the same as previously observed for CD4 T cells.
Collapse
Affiliation(s)
- Jennifer D Stone
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
293
|
Abstract
When T cells encounter antigens via the T cell antigen receptor (TCR), information about the quantity and quality of antigen engagement is relayed to the intracellular signal transduction machinery. This process is poorly understood. The TCR itself lacks a significant intracellular domain. Instead, it is associated with CD3 molecules that contain intracellular signaling domains that couple the TCR/CD3 complex to the downstream signaling machinery. The earliest events in TCR signaling must involve the transfer of information from the antigen binding TCR subunit to the CD3 signaling subunits of the TCR/CD3 complex. Elucidating the structural organization of the TCR with the associated CD3 signaling molecules is necessary for understanding the mechanism by which TCR engagement is coupled to activation. Here, we review the current state of our understanding of the structure and organization of the TCR/CD3 complex.
Collapse
Affiliation(s)
- Michael S Kuhns
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | |
Collapse
|
294
|
Abstract
Since the first crystal structure determinations of alphabeta T cell receptors (TCRs) bound to class I MHC-peptide (pMHC) antigens in 1996, a sizable database of 24 class I and class II TCR/pMHC complexes has been accumulated that now defines a substantial degree of structural variability in TCR/pMHC recognition. Recent determination of free and bound gammadelta TCR structures has enabled comparisons of the modes of antigen recognition by alphabeta and gammadelta T cells and antibodies. Crystal structures of TCR accessory (CD4, CD8) and coreceptor molecules (CD3epsilondelta, CD3epsilongamma) have further advanced our structural understanding of most of the components that constitute the TCR signaling complex. Despite all these efforts, the structural basis for MHC restriction and signaling remains elusive as no structural features that define a common binding mode or signaling mechanism have yet been gleaned from the current set of TCR/pMHC complexes. Notwithstanding, the impressive array of self, foreign (microbial), and autoimmune TCR complexes have uncovered the diverse ways in which antigens can be specifically recognized by TCRs.
Collapse
Affiliation(s)
- Markus G Rudolph
- Department of Molecular Structural Biology, University of Göttingen, 37077 Göttingen, Germany.
| | | | | |
Collapse
|
295
|
Park S, Takeuchi K, Wagner G. Solution structure of the first SRC homology 3 domain of human Nck2. JOURNAL OF BIOMOLECULAR NMR 2006; 34:203-8. [PMID: 16604428 DOI: 10.1007/s10858-006-0019-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Accepted: 01/23/2006] [Indexed: 05/08/2023]
Affiliation(s)
- Sunghyouk Park
- Department of Biological Chemistry and Molecular Pharmacology, 240 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|
296
|
McCurdy RD, Féron F, Perry C, Chant DC, McLean D, Matigian N, Hayward NK, McGrath JJ, Mackay-Sim A. Cell cycle alterations in biopsied olfactory neuroepithelium in schizophrenia and bipolar I disorder using cell culture and gene expression analyses. Schizophr Res 2006; 82:163-73. [PMID: 16406496 DOI: 10.1016/j.schres.2005.10.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 10/18/2005] [Accepted: 10/22/2005] [Indexed: 10/25/2022]
Abstract
We previously demonstrated that olfactory cultures from individuals with schizophrenia had increased cell proliferation compared to cultures from healthy controls. The aims of this study were to (a) replicate this observation in a new group of individuals with schizophrenia, (b) examine the specificity of these findings by including individuals with bipolar I disorder and (c) explore gene expression differences that may underlie cell cycle differences in these diseases. Compared to controls (n = 10), there was significantly more mitosis in schizophrenia patient cultures (n = 8) and significantly more cell death in the bipolar I disorder patient cultures (n = 8). Microarray data showed alterations to the cell cycle and phosphatidylinositol signalling pathways in schizophrenia and bipolar I disorder, respectively. Whilst caution is required in the interpretation of the array results, the study provides evidence indicating that cell proliferation and cell death in olfactory neuroepithelial cultures is differentially altered in schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Richard D McCurdy
- Eskitis Institute for Cell and Molecular Therapies, Griffith University, Brisbane, QLD 4111, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Canté-Barrett K, Gallo EM, Winslow MM, Crabtree GR. Thymocyte Negative Selection Is Mediated by Protein Kinase C- and Ca2+-Dependent Transcriptional Induction of Bim. THE JOURNAL OF IMMUNOLOGY 2006; 176:2299-306. [PMID: 16455986 DOI: 10.4049/jimmunol.176.4.2299] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The processes of positive and negative selection in the thymus both determine the population of T cells that will enter the peripheral immune system and eliminate self-reactive T cells by apoptosis. Substantial evidence indicates that TCR signal intensity mediates this cell fate choice: low-intensity signals lead to survival and differentiation, whereas high-intensity signals generated by self-Ag lead to cell death. The molecular mechanism by which these graded signals are converted to discrete outcomes is not understood. Positive selection requires the Ca(2+)-dependent phosphatase calcineurin, whereas negative selection requires the proapoptotic Bcl-2 family member Bcl-2-interacting mediator of cell death (Bim). In this study, we investigated the regulation of Bim expression and the role of Ca(2+) in mediating negative selection. Our results show that transcription is necessary for both negative selection and Bim induction. Surprisingly, we also found that Ca(2+) is necessary for Bim induction. Induction of bim transcription appears to involve protein kinase C, but not calcineurin, JNK, p38 MAPK, or MEK. These results localize the decision point in positive vs negative selection to a step downstream of Ca(2+) signaling and suggest that negative selection signals induce Ca(2+)-dependent bim transcription through PKC.
Collapse
Affiliation(s)
- Kirsten Canté-Barrett
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University, CA 94305, USA
| | | | | | | |
Collapse
|
298
|
Nitta T, Takahama Y. [Central tolerance and autoimmune diseases]. NIHON RINSHO MEN'EKI GAKKAI KAISHI = JAPANESE JOURNAL OF CLINICAL IMMUNOLOGY 2006; 29:8-15. [PMID: 16505598 DOI: 10.2177/jsci.29.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Central tolerance is established by the repertoire selection of immature T lymphocytes in the thymus, avoiding autoimmune responses to self-antigens. Differential ligand-TCR interactions that result in positive and negative selection initiate differential intracellular signals that, in turn, lead to the survival-or-death decision of immature thymocytes. TCR signal dysregulation due to the mutation of ZAP-70 or defective apoptosis of autoreactive thymocytes due to the deficiency of pro-apoptotic protein Bim impair tolerance and cause autoimmunity. Thymic repertoire selection also induces the development of CD25(+)CD4(+) regulatory T cells, which play important roles for maintaining peripheral tolerance. Furthermore, the establishment of central tolerance requires the development of thymic medulla that is mediated by the activation of NF-kappaB signaling pathway, promiscuous expression of tissue-specific self-antigens by medullary epithelial cells that is regulated by AIRE, and cortex-to-medulla migration of developing thymocytes that is regulated by CCR7-mediated chemokine signals.
Collapse
Affiliation(s)
- Takeshi Nitta
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima
| | | |
Collapse
|
299
|
Yamasaki S, Ishikawa E, Sakuma M, Ogata K, Sakata-Sogawa K, Hiroshima M, Wiest DL, Tokunaga M, Saito T. Mechanistic basis of pre–T cell receptor–mediated autonomous signaling critical for thymocyte development. Nat Immunol 2005; 7:67-75. [PMID: 16327787 DOI: 10.1038/ni1290] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Accepted: 10/13/2005] [Indexed: 01/08/2023]
Abstract
The pre-T cell receptor (TCR) is crucial for early T cell development and is proposed to function in a ligand-independent way. However, the molecular mechanism underlying the autonomous signals remains elusive. Here we show that the pre-TCR complex spontaneously formed oligomers. Specific charged residues in the extracellular domain of the pre-TCR alpha-chain mediated formation of the oligomers in vitro. Alteration of these residues eliminated the ability of the pre-TCR alpha-chain to support pre-TCR signaling in vivo. Dimerization but not raft localization of CD3epsilon was sufficient to simulate pre-TCR function and promote beta-selection. These results suggest that the pre-TCR complex can deliver its signal autonomously through oligomerization of the pre-TCR alpha-chain mediated by charged residues.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cell Differentiation/immunology
- Hematopoietic Stem Cells/cytology
- Humans
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Microscopy, Confocal
- Molecular Sequence Data
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- T-Lymphocytes/cytology
Collapse
Affiliation(s)
- Sho Yamasaki
- Laboratory for Cell Signaling, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
300
|
Hogquist KA, Baldwin TA, Jameson SC. Central tolerance: learning self-control in the thymus. Nat Rev Immunol 2005; 5:772-82. [PMID: 16200080 DOI: 10.1038/nri1707] [Citation(s) in RCA: 435] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the past few years, there has been a flurry of discoveries and advancements in our understanding of how the thymus prepares T cells to exist at peace in normal healthy tissue: that is, to be self-tolerant. In the thymus, one of the main mechanisms of T-cell central tolerance is clonal deletion, although the selection of regulatory T cells is also important and is gaining enormous interest. In this Review, we discuss the emerging consensus about which models of clonal deletion are most physiological, and we review recent data that define the molecular mechanisms of central tolerance.
Collapse
Affiliation(s)
- Kristin A Hogquist
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, 312 Church Street South East, Minneapolis, Minnesota 55455, USA.
| | | | | |
Collapse
|