1
|
Zhong T, Lei K, Lin X, Xie Z, Luo S, Zhou Z, Zhao B, Li X. Protein ubiquitination in T cell development. Front Immunol 2022; 13:941962. [PMID: 35990660 PMCID: PMC9386135 DOI: 10.3389/fimmu.2022.941962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022] Open
Abstract
As an important form of posttranslational modification, protein ubiquitination regulates a wide variety of biological processes, including different aspects of T cell development and differentiation. During T cell development, thymic seeding progenitor cells (TSPs) in the thymus undergo multistep maturation programs and checkpoints, which are critical to build a functional and tolerant immune system. Currently, a tremendous amount of research has focused on the transcriptional regulation of thymocyte development. However, in the past few years, compelling evidence has revealed that the ubiquitination system also plays a crucial role in the regulation of thymocyte developmental programs. In this review, we summarize recent findings on the molecular mechanisms and cellular pathways that regulate thymocyte ubiquitination and discuss the roles of E3 ligases and deubiquitinating enzymes (DUBs) involved in these processes. Understanding how T cell development is regulated by ubiquitination and deubiquitination will not only enhance our understanding of cell fate determination via gene regulatory networks but also provide potential novel therapeutic strategies for treating autoimmune diseases and cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bin Zhao
- *Correspondence: Bin Zhao, ; Xia Li,
| | - Xia Li
- *Correspondence: Bin Zhao, ; Xia Li,
| |
Collapse
|
2
|
Džafo E, Bianchi N, Monticelli S. Cell-intrinsic mechanisms to restrain inflammatory responses in T lymphocytes. Immunol Rev 2021; 300:181-193. [PMID: 33507562 DOI: 10.1111/imr.12932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/29/2020] [Accepted: 11/08/2020] [Indexed: 12/14/2022]
Abstract
A mechanistic understanding of the regulatory circuits that control the effector responses of memory T helper lymphocytes, and in particular their ability to produce pro-inflammatory cytokines, may lead to effective therapeutic interventions in all immune-related diseases. Activation of T lymphocytes induces robust immune responses that in most cases lead to the complete eradication of invading pathogens or tumor cells. At the same time, however, such responses must be both highly controlled in magnitude and limited in time to avoid unnecessary damage. To achieve such sophisticated level of control, T lymphocytes have at their disposal an array of transcriptional and post-transcriptional regulatory mechanisms that ensure the acquisition of a phenotype that is tailored to the incoming stimulus while restraining unwarranted activation, eventually leading to the resolution of the inflammatory response. Here, we will discuss some of these cell-intrinsic mechanisms that control T cell responses and involve transcription factors, microRNAs, and RNA-binding proteins. We will also explore how the same mechanisms can be involved both in anti-tumor responses and in autoimmunity.
Collapse
Affiliation(s)
- Emina Džafo
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Niccolò Bianchi
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Silvia Monticelli
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| |
Collapse
|
3
|
Petrou T, Olsen HL, Thrasivoulou C, Masters JR, Ashmore JF, Ahmed A. Intracellular Calcium Mobilization in Response to Ion Channel Regulators via a Calcium-Induced Calcium Release Mechanism. J Pharmacol Exp Ther 2016; 360:378-387. [PMID: 27980039 PMCID: PMC5267512 DOI: 10.1124/jpet.116.236695] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/14/2016] [Indexed: 01/19/2023] Open
Abstract
Free intracellular calcium ([Ca2+]i), in addition to being an important second messenger, is a key regulator of many cellular processes including cell membrane potential, proliferation, and apoptosis. In many cases, the mobilization of [Ca2+]i is controlled by intracellular store activation and calcium influx. We have investigated the effect of several ion channel modulators, which have been used to treat a range of human diseases, on [Ca2+]i release, by ratiometric calcium imaging. We show that six such modulators [amiodarone (Ami), dofetilide, furosemide (Fur), minoxidil (Min), loxapine (Lox), and Nicorandil] initiate release of [Ca2+]i in prostate and breast cancer cell lines, PC3 and MCF7, respectively. Whole-cell currents in PC3 cells were inhibited by the compounds tested in patch-clamp experiments in a concentration-dependent manner. In all cases [Ca2+]i was increased by modulator concentrations comparable to those used clinically. The increase in [Ca2+]i in response to Ami, Fur, Lox, and Min was reduced significantly (P < 0.01) when the external calcium was reduced to nM concentration by chelation with EGTA. The data suggest that many ion channel regulators mobilize [Ca2+]i. We suggest a mechanism whereby calcium-induced calcium release is implicated; such a mechanism may be important for understanding the action of these compounds.
Collapse
Affiliation(s)
- Terry Petrou
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, United Kingdom (T.P., A.A.); Sophion Bioscience A/S, Biolin Scientific, Ballerup, Denmark (H.L.O.); Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics (C.T.), Division of Surgery (J.R.M.), and Ear Institute, (J.F.A.), University College London, London, United Kingdom
| | - Hervør L Olsen
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, United Kingdom (T.P., A.A.); Sophion Bioscience A/S, Biolin Scientific, Ballerup, Denmark (H.L.O.); Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics (C.T.), Division of Surgery (J.R.M.), and Ear Institute, (J.F.A.), University College London, London, United Kingdom
| | - Christopher Thrasivoulou
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, United Kingdom (T.P., A.A.); Sophion Bioscience A/S, Biolin Scientific, Ballerup, Denmark (H.L.O.); Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics (C.T.), Division of Surgery (J.R.M.), and Ear Institute, (J.F.A.), University College London, London, United Kingdom
| | - John R Masters
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, United Kingdom (T.P., A.A.); Sophion Bioscience A/S, Biolin Scientific, Ballerup, Denmark (H.L.O.); Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics (C.T.), Division of Surgery (J.R.M.), and Ear Institute, (J.F.A.), University College London, London, United Kingdom
| | - Jonathan F Ashmore
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, United Kingdom (T.P., A.A.); Sophion Bioscience A/S, Biolin Scientific, Ballerup, Denmark (H.L.O.); Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics (C.T.), Division of Surgery (J.R.M.), and Ear Institute, (J.F.A.), University College London, London, United Kingdom
| | - Aamir Ahmed
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, United Kingdom (T.P., A.A.); Sophion Bioscience A/S, Biolin Scientific, Ballerup, Denmark (H.L.O.); Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics (C.T.), Division of Surgery (J.R.M.), and Ear Institute, (J.F.A.), University College London, London, United Kingdom
| |
Collapse
|
4
|
Monaco S, Jahraus B, Samstag Y, Bading H. Nuclear calcium is required for human T cell activation. J Cell Biol 2016; 215:231-243. [PMID: 27810914 PMCID: PMC5084645 DOI: 10.1083/jcb.201602001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/27/2016] [Indexed: 01/12/2023] Open
Abstract
Monaco et al. demonstrate that calcium signals in activated human T cells consist of a cytoplasmic and a nuclear component, which are both required for the immune response. Blockade of nuclear calcium signaling inhibits T cell activation and induces an anergy-like state. Calcium signals in stimulated T cells are generally considered single entities that merely trigger immune responses, whereas costimulatory events specify the type of reaction. Here we show that the “T cell calcium signal” is a composite signal harboring two distinct components that antagonistically control genomic programs underlying the immune response. Using human T cells from healthy individuals, we establish nuclear calcium as a key signal in human T cell adaptogenomics that drives T cell activation and is required for signaling to cyclic adenosine monophosphate response element–binding protein and the induction of CD25, CD69, interleukin-2, and γ-interferon. In the absence of nuclear calcium signaling, cytosolic calcium activating nuclear factor of activated T cells translocation directed the genomic response toward enhanced expression of genes that negatively modulate T cell activation and are associated with a hyporesponsive state. Thus, nuclear calcium controls the T cell fate decision between a proliferative immune response and tolerance. Modulators of nuclear calcium–driven transcription may be used to develop a new type of pro-tolerance immunosuppressive therapy.
Collapse
Affiliation(s)
- Sara Monaco
- Interdisciplinary Center for Neurosciences, Department of Neurobiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Beate Jahraus
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, 69120 Heidelberg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Interdisciplinary Center for Neurosciences, Department of Neurobiology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Effector, Memory, and Dysfunctional CD8(+) T Cell Fates in the Antitumor Immune Response. J Immunol Res 2016; 2016:8941260. [PMID: 27314056 PMCID: PMC4893440 DOI: 10.1155/2016/8941260] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/28/2016] [Indexed: 12/31/2022] Open
Abstract
The adaptive immune system plays a pivotal role in the host's ability to mount an effective, antigen-specific immune response against tumors. CD8(+) tumor-infiltrating lymphocytes (TILs) mediate tumor rejection through recognition of tumor antigens and direct killing of transformed cells. In growing tumors, TILs are often functionally impaired as a result of interaction with, or signals from, transformed cells and the tumor microenvironment. These interactions and signals can lead to transcriptional, functional, and phenotypic changes in TILs that diminish the host's ability to eradicate the tumor. In addition to effector and memory CD8(+) T cells, populations described as exhausted, anergic, senescent, and regulatory CD8(+) T cells have been observed in clinical and basic studies of antitumor immune responses. In the context of antitumor immunity, these CD8(+) T cell subsets remain poorly characterized in terms of fate-specific biomarkers and transcription factor profiles. Here we discuss the current characterization of CD8(+) T cell fates in antitumor immune responses and discuss recent insights into how signals in the tumor microenvironment influence TIL transcriptional networks to promote CD8(+) T cell dysfunction.
Collapse
|
6
|
Maggi J, Schafer C, Ubilla-Olguín G, Catalán D, Schinnerling K, Aguillón JC. Therapeutic Potential of Hyporesponsive CD4(+) T Cells in Autoimmunity. Front Immunol 2015; 6:488. [PMID: 26441992 PMCID: PMC4585084 DOI: 10.3389/fimmu.2015.00488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/07/2015] [Indexed: 01/31/2023] Open
Abstract
The interaction between dendritic cells (DCs) and T cells is crucial on immunity or tolerance induction. In an immature or semi-mature state, DCs induce tolerance through T-cell deletion, generation of regulatory T cells, and/or induction of T-cell anergy. Anergy is defined as an unresponsive state that retains T cells in an “off” mode under conditions in which immune activation is undesirable. This mechanism is crucial for the control of T-cell responses against self-antigens, thereby preventing autoimmunity. Tolerogenic DCs (tDCs), generated in vitro from peripheral blood monocytes of healthy donors or patients with autoimmune pathologies, were shown to modulate immune responses by inducing T-cell hyporesponsiveness. Animal models of autoimmune diseases confirmed the impact of T-cell anergy on disease development and progression in vivo. Thus, the induction of T-cell hyporesponsiveness by tDCs has become a promising immunotherapeutic strategy for the treatment of T-cell-mediated autoimmune disorders. Here, we review recent findings in the area and discuss the potential of anergy induction for clinical purposes.
Collapse
Affiliation(s)
- Jaxaira Maggi
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile , Santiago , Chile ; Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| | - Carolina Schafer
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile , Santiago , Chile ; Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| | - Gabriela Ubilla-Olguín
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile , Santiago , Chile ; Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| | - Diego Catalán
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile , Santiago , Chile ; Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| | - Katina Schinnerling
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile , Santiago , Chile ; Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| | - Juan C Aguillón
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile , Santiago , Chile ; Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| |
Collapse
|
7
|
Waugh KA, Leach SM, Slansky JE. Targeting Transcriptional Regulators of CD8+ T Cell Dysfunction to Boost Anti-Tumor Immunity. Vaccines (Basel) 2015; 3:771-802. [PMID: 26393659 PMCID: PMC4586477 DOI: 10.3390/vaccines3030771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Transcription is a dynamic process influenced by the cellular environment: healthy, transformed, and otherwise. Genome-wide mRNA expression profiles reflect the collective impact of pathways modulating cell function under different conditions. In this review we focus on the transcriptional pathways that control tumor infiltrating CD8+ T cell (TIL) function. Simultaneous restraint of overlapping inhibitory pathways may confer TIL resistance to multiple mechanisms of suppression traditionally referred to as exhaustion, tolerance, or anergy. Although decades of work have laid a solid foundation of altered transcriptional networks underlying various subsets of hypofunctional or “dysfunctional” CD8+ T cells, an understanding of the relevance in TIL has just begun. With recent technological advances, it is now feasible to further elucidate and utilize these pathways in immunotherapy platforms that seek to increase TIL function.
Collapse
Affiliation(s)
- Katherine A Waugh
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA.
| | - Jill E Slansky
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| |
Collapse
|
8
|
Shin DS, Jordan A, Basu S, Thomas RM, Bandyopadhyay S, de Zoeten EF, Wells AD, Macian F. Regulatory T cells suppress CD4+ T cells through NFAT-dependent transcriptional mechanisms. EMBO Rep 2014; 15:991-9. [PMID: 25074018 DOI: 10.15252/embr.201338233] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Regulatory T cells (Tregs) control autoreactive T cells by inhibiting activation-induced proliferation and cytokine expression. The molecular mechanisms responsible for the inactivation of effector T cells by Tregs remain yet to be fully characterized. We report that T-helper cells stimulated in the presence of Tregs quickly activate NFAT1 and have increased NFAT1-dependent expression of the transcription repressor Ikaros. NFAT1 deficiency or dominant-negative Ikaros compromises Treg-mediated inhibition of T-helper cells in vitro and in vivo. Thus, our results place NFAT-dependent mechanisms as general regulators of T-cell tolerance and show that Treg-mediated suppression of T-helper cells results from the activation of NFAT-regulated gene expression.
Collapse
Affiliation(s)
- Daniel S Shin
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ayana Jordan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Samik Basu
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rajan M Thomas
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine University of Pennsylvania and Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | | | - Edwin F de Zoeten
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine University of Pennsylvania and Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Andrew D Wells
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine University of Pennsylvania and Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
9
|
Tle4 regulates epigenetic silencing of gamma interferon expression during effector T helper cell tolerance. Mol Cell Biol 2013; 34:233-45. [PMID: 24190972 DOI: 10.1128/mcb.00902-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In response to suboptimal activation, T cells become hyporesponsive, with a severely reduced capacity to proliferate and produce cytokines upon reencounter with antigen. Chromatin analysis of T cells made tolerant by use of different in vitro and in vivo approaches reveals that the expression of gamma interferon (IFN-γ) is epigenetically silenced in anergic effector TH1 cells. In those T cells, calcium signaling triggers the expression of Tle4, a member of the Groucho family of corepressors, which is then recruited to a distal regulatory element in the Ifng locus and causes the establishment of repressive epigenetic marks at the Ifng gene regulatory elements. Consequently, impaired Tle4 activity results in a markedly reduced capacity to inhibit IFN-γ production in tolerized T cells. We propose that Blimp1-dependent recruitment of Tle4 to the Ifng locus causes epigenetic silencing of the expression of the Ifng gene in anergic TH1 cells. These results define a novel function of Groucho family corepressors in peripheral T cells and demonstrate that specific mechanisms are activated in tolerant T helper cells to directly repress expression of effector cytokines, supporting the hypothesis that stable epigenetic imprinting contributes to the maintenance of the tolerance-associated hyporesponsive phenotype in T cells.
Collapse
|
10
|
Palin AC, Ramachandran V, Acharya S, Lewis DB. Human neonatal naive CD4+ T cells have enhanced activation-dependent signaling regulated by the microRNA miR-181a. THE JOURNAL OF IMMUNOLOGY 2013; 190:2682-91. [PMID: 23408835 DOI: 10.4049/jimmunol.1202534] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Compared with older children and adults, human neonates have reduced and delayed CD4(+) T cell immunity to certain pathogens, but the mechanisms for these developmental differences in immune function remain poorly understood. We investigated the hypothesis that impaired human neonatal CD4(+) T cell immunity was due to reduced signaling by naive CD4(+) T cells following engagement of the αβ-TCR/CD3 complex and CD28. Surprisingly, calcium flux following engagement of CD3 was significantly higher in neonatal naive CD4(+) T cells from umbilical cord blood (CB) compared with naive CD4(+) T cells from adult peripheral blood. Enhanced calcium flux was also observed in adult CD4(+) recent thymic emigrants. Neonatal naive CD4(+) T cells also had higher activation-induced Erk phosphorylation. The microRNA miR-181a, which enhances activation-induced calcium flux in murine thymocytes, was expressed at significantly higher levels in CB naive CD4(+) T cells compared with adult cells. Overexpression of miR-181a in adult naive CD4(+) T cells increased activation-induced calcium flux, implying that the increased miR-181a levels of CB naive CD4(+) T cells contributed to their enhanced signaling. In contrast, AP-1-dependent transcription, which is downstream of Erk and required for full T cell activation, was decreased in CB naive CD4(+) T cells compared with adult cells. Thus, CB naive CD4(+) T cells have enhanced activation-dependent calcium flux, indicative of the retention of a thymocyte-like phenotype. Enhanced calcium signaling and Erk phosphorylation are decoupled from downstream AP-1-dependent transcription, which is reduced and likely contributes to limitations of human fetal and neonatal CD4(+) T cell immunity.
Collapse
Affiliation(s)
- Amy C Palin
- Department of Pediatrics, Program in Immunology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
11
|
Abstract
Diacylglycerol kinase α (DGKα) regulates diacylglycerol levels, catalyzing its conversion into phosphatidic acid. The α isoform is central to immune response regulation; it downmodulates Ras-dependent pathways and is necessary for establishment of the unresponsive state termed anergy. DGKα functions are regulated in part at the transcriptional level although the mechanisms involved remain poorly understood. Here, we analyzed the 5' end structure of the mouse DGKα gene and detected three binding sites for forkhead box O (FoxO) transcription factors, whose function was confirmed using luciferase reporter constructs. FoxO1 and FoxO3 bound to the 5' regulatory region of DGKα in quiescent T cells, as well as after interleukin-2 (IL-2) withdrawal in activated T cells. FoxO binding to this region was lost after complete T cell activation or IL-2 addition, events that correlated with FoxO phosphorylation and a sustained decrease in DGKα gene expression. These data strongly support a role for FoxO proteins in promoting high DGKα levels and indicate a mechanism by which DGKα function is downregulated during productive T cell responses. Our study establishes a basis for a causal relationship between DGKα downregulation, IL-2, and anergy avoidance.
Collapse
|
12
|
Bandyopadhyay S, Montagna C, Macian F. Silencing of the Il2 gene transcription is regulated by epigenetic changes in anergic T cells. Eur J Immunol 2012; 42:2471-83. [PMID: 22684523 DOI: 10.1002/eji.201142307] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 04/24/2012] [Accepted: 05/30/2012] [Indexed: 11/11/2022]
Abstract
Anergy is induced in T cells as a consequence of a partial or suboptimal stimulation. Anergic T cells become unresponsive and fail to proliferate and produce cytokines. We had previously shown that in anergic CD4(+) T cells, Ikaros participates in the transcriptional repression of the Il2 gene by recruiting histone deacetylases that cause core histone deacetylation at the Il2 promoter. Here we show that deacetylation at the Il2 promoter is the initial step in a process that leads to the stable silencing of the Il2 gene transcription in anergic T cells. We have found that anergy-induced deacetylation of the Il2 promoter permits binding of the histone methyl-transferase Suv39H1, which trimethylates lysine-9 of histone H3 (Me3H3-K9). Furthermore, the establishment of the Me3H3-K9 mark allows the recruitment of the heterochromatin protein HP1, allowing the silenced Il2 loci to reposition close to heterochromatin-rich regions. Our results indicate that silencing of Il2 transcription in anergic T cells is attained through a series of epigenetic changes that involve the establishment of repressive marks and the subsequent nuclear repositioning of the Il2 loci, which become juxtaposed to transcriptionally silent regions. This mechanism may account for the stable nature of the inhibition of IL-2 production in anergic cells.
Collapse
Affiliation(s)
- Sanmay Bandyopadhyay
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
13
|
Abstract
Orai1 is the key subunit of the Ca(2+)-release-activated Ca(2+) channel. Our previous report has demonstrated that Orai1 expression in the airway was upregulated in the ovalbumin (OVA)-induced allergic rhinitis (AR) mouse models. To observe whether inhibition of Orai1 expression in the airway could suppress symptoms in a murine model of AR and to assess the impacts of this inhibition on the responses of local and systemic immunocytes, we administered recombinant lentivirus vectors that encoded shRNA against ORAI1 (lenti-ORAI1) into the nostrils of OVA-sensitized mice before the challenges, and analyzed its effect on allergic responses, as compared with the unsensitized mice and untreated AR mice. Administration of lenti-ORAI1 into the nasal cavity successfully infected cells in the epithelial layer of the nasal mucosa, and significantly decreased the frequencies of sneezing and nasal rubbing of the mice. Protein levels of leukotriene C4, OVA-specific IgE, and IL-4 in the nasal lavage fluid and serum and eosinophil cation protein in the serum were also significantly reduced by lenti-ORAI1, as were the mRNA levels of these factors in the nasal mucosa and spleen. These data suggested that administration of lenti-ORAI1 into the nasal cavity effectively decreased Orai1 expression in the nasal mucosa, alleviated AR symptoms, and partially inhibited the hyperresponsiveness of the local and systemic immune cells including T cells, B cells, mast cells and eosinophils that are involved in the pathogenesis of AR.
Collapse
Affiliation(s)
- Yi Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | | | | |
Collapse
|
14
|
Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of treg-mediated T cell suppression. Front Immunol 2012; 3:51. [PMID: 22566933 PMCID: PMC3341960 DOI: 10.3389/fimmu.2012.00051] [Citation(s) in RCA: 499] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 03/01/2012] [Indexed: 12/22/2022] Open
Abstract
CD4(+)CD25(high)Foxp3(+) regulatory T cells (Tregs) can suppress other immune cells and, thus, are critical mediators of peripheral self-tolerance. On the one hand, Tregs avert autoimmune disease and allergies. On the other hand, Tregs can prevent immune reactions against tumors and pathogens. Despite the importance of Tregs, the molecular mechanisms of suppression remain incompletely understood and controversial. Proliferation and cytokine production of CD4(+)CD25(-) conventional T cells (Tcons) can be inhibited directly by Tregs. In addition, Tregs can indirectly suppress Tcon activation via inhibition of the stimulatory capacity of antigen presenting cells. Direct suppression of Tcons by Tregs can involve immunosuppressive soluble factors or cell contact. Different mechanisms of suppression have been described, so far with no consensus on one universal mechanism. Controversies might be explained by the fact that different mechanisms may operate depending on the site of the immune reaction, on the type and activation state of the suppressed target cell as well as on the Treg activation status. Further, inhibition of T cell effector function can occur independently of suppression of proliferation. In this review, we summarize the described molecular mechanisms of suppression with a particular focus on suppression of Tcons and rapid suppression of T cell receptor-induced calcium (Ca(2+)), NFAT, and NF-κB signaling in Tcons by Tregs.
Collapse
Affiliation(s)
- Angelika Schmidt
- Division of Immunogenetics, Tumorimmunology Program, German Cancer Research Center (DKFZ) Heidelberg, Germany
| | | | | |
Collapse
|
15
|
Huang Z, Moya C, Jayaraman A, Hahn J. Using the Tet-On system to develop a procedure for extracting transcription factor activation dynamics. MOLECULAR BIOSYSTEMS 2010; 6:1883-9. [PMID: 20552111 DOI: 10.1039/c003229h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The regulation of gene expression by transcription factors through different expression and activation dynamics is an important aspect of genomics and systems biology. Reporter systems using green fluorescent protein (GFP) or luciferase are often used to infer transcription factor dynamics. We recently used an inverse problem solution of GFP reporter profiles to demonstrate that the activation dynamics of a model transcription actor (NF-kappaB) can be reconstructed from GFP data. This approach assumes that the general nature of the transcription factor dynamics is known; however, it is non-trivial to determine the exact nature of the transcription factor dynamics as it often depends upon the cell type and the stimulus used to activate the transcription factor. This, in turn, limits the determination of accurate transcription factor dynamics from reporter data, especially since the model used for solution of an inverse problem needs to be verified. To address this point, we developed a reporter cell line for expressing GFP using an inducible, artificial transcription factor (tTA) and minimal promoter system. The artificial transcription factor can be activated independent of the cellular regulatory machinery through addition of doxycycline. This allows us to directly control the dynamics of the artificial transcription factor, and thereby, develop a model describing its activation dynamics from reporter data. Our experimental data and model predictions are in good agreement, and illustrate the utility of our approach. Future work will focus on using the developed approach, i.e. solution of an inverse problem involving the model describing expression of GFP, to extract the dynamics of transcription factors that are currently uncharacterized.
Collapse
Affiliation(s)
- Zuyi Huang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, 200 Jack E Brown, College Station, TX 77843-3122, USA
| | | | | | | |
Collapse
|
16
|
Abstract
CD8(+) T cells (also called cytotoxic T lymphocytes) play a major role in protective immunity against many infectious pathogens and can eradicate malignant cells. The path from naive precursor to effector and memory CD8(+) T-cell development begins with interactions between matured antigen-bearing dendritic cells (DCs) and antigen-specific naive T-cell clonal precursors. By integrating differences in antigenic, costimulatory, and inflammatory signals, a developmental program is established that governs many key parameters associated with the ensuing response, including the extent and magnitude of clonal expansion, the functional capacities of the effector cells, and the size of the memory pool that survives after the contraction phase. In this review, we discuss the multitude of signals that drive effector and memory CD8(+) T-cell differentiation and how the differences in the nature of these signals contribute to the diversity of CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Ramon Arens
- Laboratory of Cellular Immunology, The La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Stephen P. Schoenberger
- Laboratory of Cellular Immunology, The La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|
17
|
Arimilli S, Sharma SK, Yammani R, Reid SD, Parks GD, Alexander-Miller MA. Pivotal Advance: Nonfunctional lung effectors exhibit decreased calcium mobilization associated with reduced expression of ORAI1. J Leukoc Biol 2010; 87:977-88. [PMID: 20103768 DOI: 10.1189/jlb.0809575] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CD8(+) T cells play a critical role in the clearance of respiratory pathogens. Thus, it is surprising that functional inactivation of lung effectors has been observed in many models of viral infection. Currently, the molecular defect responsible for the shut-off of function in these cells is unknown. In the present study, we addressed this question using a model of respiratory infection with the paramyxovirus SV5. Nonfunctional cells were found to exhibit decreases in SOCE, resulting in reduced NFAT1 activation. Notably, function could be restored by the provision of increased levels of extracellular calcium. The reduced ability to mobilize calcium was associated with reduced expression of ORAI1, the CRAC channel subunit. These findings reveal a previously unknown mechanism for the negative regulation of function in effector T cells.
Collapse
Affiliation(s)
- Subhashini Arimilli
- 1. Room 5140 Gray Building, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
18
|
Feske S. ORAI1 and STIM1 deficiency in human and mice: roles of store-operated Ca2+ entry in the immune system and beyond. Immunol Rev 2009; 231:189-209. [PMID: 19754898 DOI: 10.1111/j.1600-065x.2009.00818.x] [Citation(s) in RCA: 252] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Store-operated Ca2+ entry (SOCE) is a mechanism used by many cells types including lymphocytes and other immune cells to increase intracellular Ca2+ concentrations to initiate signal transduction. Activation of immunoreceptors such as the T-cell receptor, B-cell receptor, or Fc receptors results in the release of Ca2+ ions from endoplasmic reticulum (ER) Ca2+ stores and subsequent activation of plasma membrane Ca2+ channels such as the well-characterized Ca2+ release-activated Ca2+ (CRAC) channel. Two genes have been identified that are essential for SOCE: ORAI1 as the pore-forming subunit of the CRAC channel in the plasma membrane and stromal interaction molecule-1 (STIM1) sensing the ER Ca2+ concentration and activating ORAI1-CRAC channels. Intense efforts in the past several years have focused on understanding the molecular mechanism of SOCE and the role it plays for cell functions in vitro and in vivo. A number of transgenic mouse models have been generated to investigate the role of ORAI1 and STIM1 in immunity. In addition, mutations in ORAI1 and STIM1 identified in immunodeficient patients provide valuable insight into the role of both genes and SOCE. This review focuses on the role of ORAI1 and STIM1 in vivo, discussing the phenotypes of ORAI1- and STIM1-deficient human patients and mice.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
19
|
Baine I, Abe BT, Macian F. Regulation of T-cell tolerance by calcium/NFAT signaling. Immunol Rev 2009; 231:225-40. [PMID: 19754900 DOI: 10.1111/j.1600-065x.2009.00817.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cells that escape negative selection in the thymus must be inactivated or eliminated in the periphery through a series of mechanisms that include the induction of anergy, dominant suppression by regulatory T cells, and peripheral deletion of self-reactive T cells. Calcium signaling plays a central role in the induction of anergy in T cells, which become functionally inactivated and incapable of proliferating and expressing cytokines following antigen re-encounter. Suboptimal stimulation of T cells results in the activation of a calcium/calcineurin/nuclear factor of activated T cells-dependent cell-intrinsic program of self-inactivation. The proteins encoded by those genes are required to impose a state of functional unresponsiveness through different mechanisms that include downregulation of T-cell receptor signaling and inhibition of cytokine transcription.
Collapse
Affiliation(s)
- Ian Baine
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
20
|
Abstract
This year marks the 25th anniversary of the first Annual Review of Immunology article to describe features of the T cell antigen receptor (TCR). In celebration of this anniversary, we begin with a brief introduction outlining the chronology of the earliest studies that established the basic paradigm for how the engaged TCR transduces its signals. This review continues with a description of the current state of our understanding of TCR signaling, as well as a summary of recent findings examining other key aspects of T cell activation, including cross talk between the TCR and integrins, the role of costimulatory molecules, and how signals may negatively regulate T cell function.Acronyms and DefinitionsAdapter protein: cellular protein that functions to bridge molecular interactions via characteristic domains able to mediate protein/protein or protein/lipid interactions Costimulation: signals delivered to T cells by cell surface receptors other than the TCR itself that potentiate T cell activation cSMAC: central supramolecular activation cluster Immunoreceptor tyrosine-based activation motif (ITAM): a short peptide sequence in the cytoplasmic tails of key surface receptors on hematopoietic cells that is characterized by tyrosine residues that are phosphorylated by Src family PTKs, enabling the ITAM to recruit activated Syk family kinases Inside-out signaling: signals initiated by engagement of immunoreceptors that lead to conformational changes and clustering of integrins, thereby increasing the affinity and avidity of the integrins for their ligands NFAT: nuclear factor of activated T cells PI3K: phosphoinositide 3-kinase PKC: protein kinase C PLC: phospholipase C pMHC: peptide major histocompatibility complex (MHC) complex pSMAC: peripheral supramolecular activation cluster PTK: protein tyrosine kinase Signal transduction: biochemical events linking surface receptor engagement to cellular responses TCR: T cell antigen receptor
Collapse
Affiliation(s)
- Jennifer E Smith-Garvin
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
21
|
|
22
|
|
23
|
Soto-Nieves N, Puga I, Abe BT, Bandyopadhyay S, Baine I, Rao A, Macian F. Transcriptional complexes formed by NFAT dimers regulate the induction of T cell tolerance. ACTA ACUST UNITED AC 2009; 206:867-76. [PMID: 19307325 PMCID: PMC2715123 DOI: 10.1084/jem.20082731] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In T cells, anergy can be induced after T cell receptor engagement in the absence of costimulation. Under these conditions, the expression of a specific set of anergy-associated genes is activated. Several lines of evidence suggest that nuclear factor of activated T cells (NFAT) proteins may regulate the expression of many of those genes; however, the nature of the complexes responsible for the induction of this new program of gene expression is unknown. Here, we show that transcriptional complexes formed by NFAT homodimers are directly responsible for the activation of at least two anergy-inducing genes, Grail and Caspase3. Our data shows that Grail expression is activated by direct binding of NFAT dimers to the Grail promoter at two different sites. Consequently, a mutant NFAT protein with impaired ability to dimerize is not able to induce an unresponsive state in T cells. Our results not only identify a new biological function for NFAT dimers but also reveal the different nature of NFAT-containing complexes that induce anergy versus those that are activated during a productive immune response. These data also establish a basis for the design of immunomodulatory strategies that specifically target each type of complex.
Collapse
Affiliation(s)
- Noemi Soto-Nieves
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Targeted cleavage of signaling proteins by caspase 3 inhibits T cell receptor signaling in anergic T cells. Immunity 2008; 29:193-204. [PMID: 18701083 DOI: 10.1016/j.immuni.2008.06.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2007] [Revised: 03/26/2008] [Accepted: 06/10/2008] [Indexed: 11/22/2022]
Abstract
T cell receptor (TCR) engagement in the absence of costimulation induces the calcium-dependent upregulation of a program of gene expression that leads to the establishment of T cell anergy. Casp3 is one of the genes activated during anergy induction. Here we show that caspase 3 is required for the induction of T cell unresponsiveness. Suboptimal T cell stimulation induced caspase 3 activation, which did not result in cell death. Furthermore, caspase 3-deficient T cells showed impaired responses to anergizing stimuli. In anergic T cells, activated caspase 3 associated to the plasma membrane, where it cleaved and inactivated proteins such as the Grb2-related adaptor downstream of shc (GADS) and the guanine-nucleotide exchange factor Vav1, causing a blockade in TCR signaling. Our results identify a role for caspase 3 in nonapoptotic T cells and support that caspase 3-dependent proteolytic inactivation of signaling proteins is essential to maintain T cell tolerance.
Collapse
|
25
|
Duré M, Macian F. IL-2 signaling prevents T cell anergy by inhibiting the expression of anergy-inducing genes. Mol Immunol 2008; 46:999-1006. [PMID: 18990450 DOI: 10.1016/j.molimm.2008.09.029] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 08/19/2008] [Accepted: 09/23/2008] [Indexed: 12/14/2022]
Abstract
T cell responses are determined by the environment in which antigen is encountered. In the absence of proper costimulation, anergizing stimuli induce the activation of a specific program of gene expression. Proteins encoded by these genes impose a state of functional unresponsiveness in anergic T cells through the activation of different mechanisms that include dampening of the T cell receptor signaling and direct inhibition of cytokine expression. Anergy can be reversed by stimulating T cells in the presence of interleukin (IL-)2. Signaling through the IL-2 receptor has been shown to activate mTOR, which plays an important role in the integration of signals that determine the fate of T cells. The mechanisms underlying the IL-2-dependent regulation of T cell tolerance are still not fully elucidated. In this study we show that IL-2 receptor signaling mediated through JAK3 and mTOR inhibits the expression of anergy-inducing genes independently of any effect on cell cycle progression. Interestingly, we also show that this effect is likely due to changes on the levels of AP-1 activation induced by IL-2 receptor signaling in T cells. Our data identifies a mechanism that can explain how IL-2 may prevent or reverse the establishment of anergy in T cells and, therefore, helps to understand how the cytokine environment can be determinant to shape the outcome of T cell responses - tolerance or activation - when antigen is encountered.
Collapse
Affiliation(s)
- Myrianne Duré
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | |
Collapse
|
26
|
Huang Z, Senocak F, Jayaraman A, Hahn J. Integrated modeling and experimental approach for determining transcription factor profiles from fluorescent reporter data. BMC SYSTEMS BIOLOGY 2008; 2:64. [PMID: 18637177 PMCID: PMC2491602 DOI: 10.1186/1752-0509-2-64] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 07/17/2008] [Indexed: 01/12/2023]
Abstract
Background The development of quantitative models of signal transduction, as well as parameter estimation to improve existing models, depends on the ability to obtain quantitative information about various proteins that are part of the signaling pathway. However, commonly-used measurement techniques such as Western blots and mobility shift assays provide only qualitative or semi-quantitative data which cannot be used for estimating parameters. Thus there is a clear need for techniques that enable quantitative determination of signal transduction intermediates. Results This paper presents an integrated modeling and experimental approach for quantitatively determining transcription factor profiles from green fluorescent protein (GFP) reporter data. The technique consists of three steps: (1) creating data sets for green fluorescent reporter systems upon stimulation, (2) analyzing the fluorescence images to determine fluorescence intensity profiles using principal component analysis (PCA) and K-means clustering, and (3) computing the transcription factor concentration from the fluorescence intensity profiles by inverting a model describing transcription, translation, and activation of green fluorescent proteins. We have used this technique to quantitatively characterize activation of the transcription factor NF-κB by the cytokine TNF-α. In addition, we have applied the quantitative NF-κB profiles obtained from our technique to develop a model for TNF-α signal transduction where the parameters were estimated from the obtained data. Conclusion The technique presented here for computing transcription factor profiles from fluorescence microscopy images of reporter cells generated quantitative data on the magnitude and dynamics of NF-κB activation by TNF-α. The obtained results are in good agreement with qualitative descriptions of NF-κB activation as well as semi-quantitative experimental data from the literature. The profiles computed from the experimental data have been used to re-estimate parameters for a NF-κB model and the results of additional experiments are predicted very well by the model with the new parameter values. While the presented approach has been applied to NF-κB and TNF-α signaling, it can be used to determine the profile of any transcription factor as long as GFP reporter fluorescent profiles are available.
Collapse
Affiliation(s)
- Zuyi Huang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843-3122, USA.
| | | | | | | |
Collapse
|
27
|
Gómez-Martín D, Díaz-Zamudio M, Alcocer-Varela J. Ubiquitination system and autoimmunity: The bridge towards the modulation of the immune response. Autoimmun Rev 2008; 7:284-90. [DOI: 10.1016/j.autrev.2007.11.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 11/01/2007] [Indexed: 11/25/2022]
|
28
|
Control of virus-specific CD8+ T-cell exhaustion and immune-mediated pathology by E3 ubiquitin ligase Cbl-b during chronic viral infection. J Virol 2008; 82:3353-68. [PMID: 18199651 DOI: 10.1128/jvi.01350-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A characteristic feature in the immune response to many persistent viral infections is the dysfunction or deletion of antigen-specific T cells (exhaustion). This down-regulation of virus-specific T-cell response represents a critical control mechanism that exists within T-cell activation pathways to prevent lethal disease by inappropriate responses against disseminating virus infections. However, the molecular mechanisms by which the immune system determines whether to mount a full response to such infections remain largely unexplored. Here, we have established that in the murine lymphocytic choriomeningitis virus (LCMV) model, induction of the T-cell receptor signaling inhibitor molecule E3 ligase Cbl-b is critically involved in this decision. In particular, our data revealed that Cbl-b controls the program responsible for T-cell tolerance (exhaustion) induction during a chronic viral infection. Thus, Cbl-b(-/-) mice infected with a low dose of LCMV Docile mount a strong CD8(+) T-cell response that rapidly clears the infection, and the animals remain healthy; in contrast, down-regulation of the epitope-specific CD8(+) T-cell population in persistently infected Cbl-b(-/-) mice, compared to that in chronically infected B6 mice, was significantly delayed, and this was associated with increased morbidity and eventual death in nearly 20% of the animals. Interestingly, infection of Cbl-b(-/-) mice with a moderate virus dose resulted in rapid death with 100% mortality by 7 to 8 days after infection, caused by a dysregulated antiviral T-cell response, whereas the infected B6 mice survived and remained healthy. In conclusion, our results suggest that Cbl-b is critically involved in T-cell exhaustion and prevention of lethal disease.
Collapse
|