1
|
Pritzl CJ, Luera D, Knudson KM, Quaney MJ, Calcutt MJ, Daniels MA, Teixeiro E. IKK2/NFkB signaling controls lung resident CD8 + T cell memory during influenza infection. Nat Commun 2023; 14:4331. [PMID: 37468506 DOI: 10.1038/s41467-023-40107-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/13/2023] [Indexed: 07/21/2023] Open
Abstract
CD8+ T cell tissue resident memory (TRM) cells are especially suited to control pathogen spread at mucosal sites. However, their maintenance in lung is short-lived. TCR-dependent NFkB signaling is crucial for T cell memory but how and when NFkB signaling modulates tissue resident and circulating T cell memory during the immune response is unknown. Here, we find that enhancing NFkB signaling in T cells once memory to influenza is established, increases pro-survival Bcl-2 and CD122 levels thus boosting lung CD8+ TRM maintenance. By contrast, enhancing NFkB signals during the contraction phase of the response leads to a defect in CD8+ TRM differentiation without impairing recirculating memory subsets. Specifically, inducible activation of NFkB via constitutive active IKK2 or TNF interferes with TGFβ signaling, resulting in defects of lung CD8+ TRM imprinting molecules CD69, CD103, Runx3 and Eomes. Conversely, inhibiting NFkB signals not only recovers but improves the transcriptional signature and generation of lung CD8+ TRM. Thus, NFkB signaling is a critical regulator of tissue resident memory, whose levels can be tuned at specific times during infection to boost lung CD8+ TRM.
Collapse
Affiliation(s)
- Curtis J Pritzl
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Building, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Dezzarae Luera
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Building, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Karin M Knudson
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Michael J Quaney
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Michael J Calcutt
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Building, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Roy Blunt NextGen Precision Health Building, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
2
|
Tanaka A, Maeda S, Nomura T, Llamas-Covarrubias MA, Tanaka S, Jin L, Lim EL, Morikawa H, Kitagawa Y, Akizuki S, Ito Y, Fujimori C, Hirota K, Murase T, Hashimoto M, Higo J, Zamoyska R, Ueda R, Standley DM, Sakaguchi N, Sakaguchi S. Construction of a T cell receptor signaling range for spontaneous development of autoimmune disease. J Exp Med 2023; 220:213728. [PMID: 36454183 PMCID: PMC9718937 DOI: 10.1084/jem.20220386] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
Thymic selection and peripheral activation of conventional T (Tconv) and regulatory T (Treg) cells depend on TCR signaling, whose anomalies are causative of autoimmunity. Here, we expressed in normal mice mutated ZAP-70 molecules with different affinities for the CD3 chains, or wild type ZAP-70 at graded expression levels under tetracycline-inducible control. Both manipulations reduced TCR signaling intensity to various extents and thereby rendered those normally deleted self-reactive thymocytes to become positively selected and form a highly autoimmune TCR repertoire. The signal reduction more profoundly affected Treg development and function because their TCR signaling was further attenuated by Foxp3 that physiologically repressed the expression of TCR-proximal signaling molecules, including ZAP-70, upon TCR stimulation. Consequently, the TCR signaling intensity reduced to a critical range generated pathogenic autoimmune Tconv cells and concurrently impaired Treg development/function, leading to spontaneous occurrence of autoimmune/inflammatory diseases, such as autoimmune arthritis and inflammatory bowel disease. These results provide a general model of how altered TCR signaling evokes autoimmune disease.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Frontier Research in Tumor Immunology, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinji Maeda
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Nomura
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mara Anais Llamas-Covarrubias
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Institute of Research in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Satoshi Tanaka
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Lin Jin
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ee Lyn Lim
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Hiromasa Morikawa
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Yohko Kitagawa
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shuji Akizuki
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshinaga Ito
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Chihiro Fujimori
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Tosei Murase
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Motomu Hashimoto
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Junichi Higo
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Rose Zamoyska
- Institute for Immunology and Infection Research, The University of Edinburgh, Edinburgh, UK
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Aichi, Japan
| | - Daron M Standley
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Noriko Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
3
|
Kästle M, Merten C, Hartig R, Plaza-Sirvent C, Schmitz I, Bommhardt U, Schraven B, Simeoni L. Type of PaperY192 within the SH2 Domain of Lck Regulates TCR Signaling Downstream of PLC-γ1 and Thymic Selection. Int J Mol Sci 2022; 23:ijms23137271. [PMID: 35806279 PMCID: PMC9267008 DOI: 10.3390/ijms23137271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Signaling via the TCR, which is initiated by the Src-family tyrosine kinase Lck, is crucial for the determination of cell fates in the thymus. Because of its pivotal role, ablation of Lck results in a profound block of T-cell development. Here, we show that, in addition to its well-known function in the initiation of TCR signaling, Lck also acts at a more downstream level. This novel function of Lck is determined by the tyrosine residue (Y192) located in its SH2 domain. Thymocytes from knock-in mice expressing a phosphomimetic Y192E mutant of Lck initiate TCR signaling upon CD3 cross-linking up to the level of PLC-γ1 phosphorylation. However, the activation of downstream pathways including Ca2+ influx and phosphorylation of Erk1/2 are impaired. Accordingly, positive and negative selections are blocked in LckY192E knock-in mice. Collectively, our data indicate that Lck has a novel function downstream of PLCγ-1 in the regulation of thymocyte differentiation and selection.
Collapse
Affiliation(s)
- Matthias Kästle
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
| | - Camilla Merten
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
| | - Roland Hartig
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
| | - Carlos Plaza-Sirvent
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Department of Molecular Immunology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ingo Schmitz
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Department of Molecular Immunology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39120 Magdeburg, Germany
- Correspondence: (B.S.); (L.S.)
| | - Luca Simeoni
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39120 Magdeburg, Germany
- Correspondence: (B.S.); (L.S.)
| |
Collapse
|
4
|
Kästle M, Merten C, Hartig R, Kaehne T, Liaunardy-Jopeace A, Woessner NM, Schamel WW, James J, Minguet S, Simeoni L, Schraven B. Tyrosine 192 within the SH2 domain of the Src-protein tyrosine kinase p56 Lck regulates T-cell activation independently of Lck/CD45 interactions. Cell Commun Signal 2020; 18:183. [PMID: 33225946 PMCID: PMC7682018 DOI: 10.1186/s12964-020-00673-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/13/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Upon engagement of the T-cell receptor (TCR), the Src-family protein tyrosine kinase p56Lck phosphorylates components of the TCR (e.g. the TCRζ chains), thereby initiating T-cell activation. The enzymatic activity of Lck is primarily regulated via reversible and dynamic phosphorylation of two tyrosine residues, Y394 and Y505. Lck possesses an additional highly conserved tyrosine Y192, located within the SH2 domain, whose role in T-cell activation is not fully understood. METHODS Knock-in mice expressing a phospho-mimetic (Y192E) form of Lck were generated. Cellular and biochemical characterization was performed to elucidate the function of Y192 in primary T cells. HEK 293T and Jurkat T cells were used for in vitro studies. RESULTS Co-immunoprecipitation studies and biochemical analyses using T cells from LckY192E knock-in mice revealed a diminished binding of LckY192E to CD45 and a concomitant hyperphosphorylation of Y505, thus corroborating previous data obtained in Jurkat T cells. Surprisingly however, in vitro kinase assays showed that LckY192E possesses a normal enzymatic activity in human and murine T cells. FLIM/FRET measurements employing an LckY192E biosensor further indicated that the steady state conformation of the LckY192E mutant is similar to Lckwt. These data suggest that Y192 might regulate Lck functions also independently from the Lck/CD45-association. Indeed, when LckY192E was expressed in CD45-/-/Csk-/- non-T cells (HEK 293T cells), phosphorylation of Y505 was similar to Lckwt, but LckY192E still failed to optimally phosphorylate and activate the Lck downstream substrate ZAP70. Furthermore, LckY19E was recruited less to CD3 after TCR stimulation. CONCLUSIONS Taken together, phosphorylation of Y192 regulates Lck functions in T cells at least twofold, by preventing Lck association to CD45 and by modulating ligand-induced recruitment of Lck to the TCR. MAJOR FINDINGS Our data change the current view on the function of Y192 and suggest that Y192 also regulates Lck activity in a manner independent of Y505 phosphorylation. Video Abstract.
Collapse
Affiliation(s)
- Matthias Kästle
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Leipziger Str.44, Building 26, 39120, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Camilla Merten
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Leipziger Str.44, Building 26, 39120, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Roland Hartig
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Leipziger Str.44, Building 26, 39120, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Thilo Kaehne
- Institute of Experimental Internal Medicine, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Nadine M Woessner
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Faculty of Biology, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Wolfgang W Schamel
- Faculty of Biology, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - John James
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC-LMB, Cambridge, UK.,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry, UK
| | - Susana Minguet
- Faculty of Biology, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Luca Simeoni
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Leipziger Str.44, Building 26, 39120, Magdeburg, Germany. .,Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Leipziger Str.44, Building 26, 39120, Magdeburg, Germany. .,Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
5
|
Iqbal MA, Hong K, Kim JH, Choi Y. Severe combined immunodeficiency pig as an emerging animal model for human diseases and regenerative medicines. BMB Rep 2020. [PMID: 31722780 PMCID: PMC6889892 DOI: 10.5483/bmbrep.2019.52.11.267] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Severe combined immunodeficiency (SCID) is a group of inherited disorders characterized by compromised T lymphocyte differentiation related to abnormal development of other lymphocytes [i.e., B and/or natural killer (NK) cells], leading to death early in life unless treated immediately with hematopoietic stem cell transplant. Functional NK cells may impact engraftment success of life-saving procedures such as bone marrow transplantation in human SCID patients. Therefore, in animal models, a T cell−/B cell−/NK cell+ environment provides a valuable tool for understanding the function of the innate immune system and for developing targeted NK therapies against human immune diseases. In this review, we focus on underlying mechanisms of human SCID, recent progress in the development of SCID animal models, and utilization of SCID pig model in biomedical sciences. Numerous physiologies in pig are comparable to those in human such as immune system, X-linked heritability, typical T−B+NK− cellular phenotype, and anatomy. Due to analogous features of pig to those of human, studies have found that immunodeficient pig is the most appropriate model for human SCID.
Collapse
Affiliation(s)
- Muhammad Arsalan Iqbal
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Jin Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| |
Collapse
|
6
|
Odagiu L, May J, Boulet S, Baldwin TA, Labrecque N. Role of the Orphan Nuclear Receptor NR4A Family in T-Cell Biology. Front Endocrinol (Lausanne) 2020; 11:624122. [PMID: 33597928 PMCID: PMC7883379 DOI: 10.3389/fendo.2020.624122] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
The nuclear orphan receptors NR4A1, NR4A2, and NR4A3 are immediate early genes that are induced by various signals. They act as transcription factors and their activity is not regulated by ligand binding and are thus regulated via their expression levels. Their expression is transiently induced in T cells by triggering of the T cell receptor following antigen recognition during both thymic differentiation and peripheral T cell responses. In this review, we will discuss how NR4A family members impact different aspects of the life of a T cell from thymic differentiation to peripheral response against infections and cancer.
Collapse
Affiliation(s)
- Livia Odagiu
- Laboratory of Immunology, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Julia May
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Salix Boulet
- Laboratory of Immunology, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
| | - Troy A. Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Troy A. Baldwin, ; Nathalie Labrecque,
| | - Nathalie Labrecque
- Laboratory of Immunology, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
- Département de Médecine, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Troy A. Baldwin, ; Nathalie Labrecque,
| |
Collapse
|
7
|
Abstract
Generating and maintaining a diverse repertoire of naive T cells is essential for protection against pathogens, and developing a mechanistic and quantitative description of the processes involved lies at the heart of our understanding of vertebrate immunity. Here, we review the biology of naive T cells from birth to maturity and outline how the integration of mathematical models and experiments has helped us to develop a full picture of their life histories.
Collapse
Affiliation(s)
- Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, UK
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| |
Collapse
|
8
|
DNA-binding of the Tet-transactivator curtails antigen-induced lymphocyte activation in mice. Nat Commun 2017; 8:1028. [PMID: 29044097 PMCID: PMC5647323 DOI: 10.1038/s41467-017-01022-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 08/14/2017] [Indexed: 12/31/2022] Open
Abstract
The Tet-On/Off system for conditional transgene expression constitutes state-of-the-art technology to study gene function by facilitating inducible expression in a timed and reversible manner. Several studies documented the suitability and versatility of this system to trace lymphocyte fate and to conditionally express oncogenes or silence tumour suppressor genes in vivo. Here, we show that expression of the tetracycline/doxycycline-controlled Tet-transactivator, while tolerated well during development and in immunologically unchallenged animals, impairs the expansion of antigen-stimulated T and B cells and thereby curtails adaptive immune responses in vivo. Transactivator-mediated cytotoxicity depends on DNA binding, but can be overcome by BCL2 overexpression, suggesting that apoptosis induction upon lymphocyte activation limits cellular and humoral immune responses. Our findings suggest a possible system-intrinsic biological bias of the Tet-On/Off system in vivo that will favour the outgrowth of apoptosis resistant clones, thus possibly confounding data published using such systems. Tet-transactivators are used for direct regulation of gene expression, RNA interference and for CRISPR/Cas9-based systems. Here the authors show that DNA-bound Tet-transactivators can induce cell death in antigen-activated lymphocytes in vivo, putting into question the use of, and in vivo data generated with, these molecular tools.
Collapse
|
9
|
Tan TCJ, Knight J, Sbarrato T, Dudek K, Willis AE, Zamoyska R. Suboptimal T-cell receptor signaling compromises protein translation, ribosome biogenesis, and proliferation of mouse CD8 T cells. Proc Natl Acad Sci U S A 2017; 114:E6117-E6126. [PMID: 28696283 PMCID: PMC5544288 DOI: 10.1073/pnas.1700939114] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Global transcriptomic and proteomic analyses of T cells have been rich sources of unbiased data for understanding T-cell activation. Lack of full concordance of these datasets has illustrated that important facets of T-cell activation are controlled at the level of translation. We undertook translatome analysis of CD8 T-cell activation, combining polysome profiling and microarray analysis. We revealed that altering T-cell receptor stimulation influenced recruitment of mRNAs to heavy polysomes and translation of subsets of genes. A major pathway that was compromised, when TCR signaling was suboptimal, was linked to ribosome biogenesis, a rate-limiting factor in both cell growth and proliferation. Defective TCR signaling affected transcription and processing of ribosomal RNA precursors, as well as the translation of specific ribosomal proteins and translation factors. Mechanistically, IL-2 production was compromised in weakly stimulated T cells, affecting the abundance of Myc protein, a known regulator of ribosome biogenesis. Consequently, weakly activated T cells showed impaired production of ribosomes and a failure to maintain proliferative capacity after stimulation. We demonstrate that primary T cells respond to various environmental cues by regulating ribosome biogenesis and mRNA translation at multiple levels to sustain proliferation and differentiation.
Collapse
Affiliation(s)
- Thomas C J Tan
- Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
| | - John Knight
- Medical Research Council Toxicology Unit, Leicester LE1 9HN, United Kingdom
| | - Thomas Sbarrato
- Medical Research Council Toxicology Unit, Leicester LE1 9HN, United Kingdom
| | - Kate Dudek
- Medical Research Council Toxicology Unit, Leicester LE1 9HN, United Kingdom
| | - Anne E Willis
- Medical Research Council Toxicology Unit, Leicester LE1 9HN, United Kingdom
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom;
| |
Collapse
|
10
|
Chiang YJ, Hodes RJ. T-cell development is regulated by the coordinated function of proximal and distal Lck promoters active at different developmental stages. Eur J Immunol 2016; 46:2401-2408. [PMID: 27469439 DOI: 10.1002/eji.201646440] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/05/2016] [Accepted: 07/26/2016] [Indexed: 12/21/2022]
Abstract
Expression of Lck, a T-cell lineage-specific tyrosine kinase critical for T-cell development and activation, can be mediated by either proximal or distal lck promoter. We generated BAC transgenic mice in which BAC lck promoter was deleted and bred these transgenes to an Lck knockout background. Lck-PROX mice, in which only the proximal promoter is functional, have maximal Lck protein and normal thymic development through CD4- CD8- double negative (DN) and CD4+ CD8+ double positive (DP) stages, but undetectable Lck later in development and reduced mature single positive thymocytes. In contrast, Lck-DIST mice, in which only distal promoter was functional, are deficient in Lck protein in DN and DP thymocytes and severely defective in early T-cell development, with a block at the DN3-DN4 beta checkpoint equivalent to complete Lck knockouts. The ability of the proximal lck promoter to support thymic development is independent of Fyn; while, in contrast, the distal lck promoter alone is completely unable to support development in the absence of Fyn. Notably, normal thymocyte development is restored by presence of both proximal and distal promoters, even when independently expressed on different lck genes. These results define distinct and complementary requirements for proximal and distal lck promoters during T-cell development.
Collapse
Affiliation(s)
- Y Jeffrey Chiang
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Richard J Hodes
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, USA. .,National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Goplen NP, Saxena V, Knudson KM, Schrum AG, Gil D, Daniels MA, Zamoyska R, Teixeiro E. IL-12 Signals through the TCR To Support CD8 Innate Immune Responses. THE JOURNAL OF IMMUNOLOGY 2016; 197:2434-43. [PMID: 27521342 DOI: 10.4049/jimmunol.1600037] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 07/13/2016] [Indexed: 01/19/2023]
Abstract
CD8 T cells must integrate antigenic and inflammatory signals to differentiate into efficient effector and memory T cells able to protect us from infections. The mechanisms by which TCR signaling and proinflammatory cytokine receptor signaling cooperate in these processes are poorly defined. In this study, we show that IL-12 and other proinflammatory cytokines transduce signals through the TCR signalosome in a manner that requires Fyn activity and self-peptide-MHC (self-pMHC) interactions. This mechanism is crucial for CD8 innate T cell functions. Loss of Fyn activity or blockade of self-pMHC interactions severely impaired CD8 T cell IFN-γ and NKG2D expression, proliferation, and cytotoxicity upon cytokine-mediated bystander activation. Most importantly, in the absence of self-pMHC interactions, CD8 memory T cells fail to undergo bystander activation upon an unrelated infection. Thus, CD8 T cell bystander activation, although independent of cognate Ag, still requires self-pMHC and TCR signaling.
Collapse
Affiliation(s)
- Nicholas P Goplen
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Vikas Saxena
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Karin M Knudson
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Adam G Schrum
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| | - Diana Gil
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212;
| |
Collapse
|
12
|
Gorelik G, Sawalha AH, Patel D, Johnson K, Richardson B. T cell PKCδ kinase inactivation induces lupus-like autoimmunity in mice. Clin Immunol 2015; 158:193-203. [PMID: 25829232 DOI: 10.1016/j.clim.2015.03.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
Genetic and environmental factors contribute to the onset and progression of lupus. CD4+ T cells from patients with active lupus show a decreased ERK signaling pathway, which causes changes in gene expression. The defect points to its upstream regulator, PKCδ, which exhibits a deficient activity due to oxidative stress. Our aim was to investigate the effect of a defective PKCδ in the development of lupus. We generated a double transgenic C57BL6 × SJL mouse that expresses a doxycycline-induced dominant negative PKCδ (dnPKCδ) in T cells. The transgenic mice displayed decreased T cell ERK signaling, decreased DNMT1 expression and overexpression of methylation sensitive genes involved in the exaggerated immune response in the pathogenesis of lupus. The mice developed anti-dsDNA autoantibodies and glomerulonephritis with IgG deposition. The study indicates common pathogenic mechanisms with human lupus, suggesting that environmentally-mediated T cell PKCδ inactivation plays a causative role in lupus.
Collapse
Affiliation(s)
- Gabriela Gorelik
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Amr H Sawalha
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Dipak Patel
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kent Johnson
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Bruce Richardson
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA; Ann Arbor VA Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
13
|
A Zap70-dependent feedback circuit is essential for efficient selection of CD4 lineage thymocytes. Immunol Cell Biol 2015; 93:406-16. [PMID: 25601273 DOI: 10.1038/icb.2014.107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 02/05/2023]
Abstract
During positive selection of CD4(+), CD8(+) double positive (DP) thymocytes, expression of the tyrosine kinase Zap70 is subject to developmental regulation. Signalling downstream of T-cell receptor (TCR) induces Zap70 expression, forming a positive feedback circuit. Although previous studies show this circuit is required for generation of CD8 lineage cells, it is not known whether selection of CD4 T cells also depends on intact developmental regulation of Zap70. To address this, we analysed development of Class II-restricted thymocytes in mice lacking the Zap70 transcriptional circuitry. Rescue of Zap70 expression in Zap70(-/-) mice using a tetracycline-inducible Zap70 transgene, that is not subject to positive feedback by TCR signalling, restored positive selection of Class-II-restricted thymocytes. However, in conditions of static Zap70 expression, approximately half of selecting thymocytes failed to commit normally to the CD4 lineage. Instead, cells that failed to develop into CD4 T cells resembled CD8 lineage precursor DP thymocytes but failed to survive in vivo. Therefore, the Zap70 feedback circuit is essential to efficiently mediate the CD4 lineage differentiation programme in response to Class II selecting ligands.
Collapse
|
14
|
Ligand-engaged TCR is triggered by Lck not associated with CD8 coreceptor. Nat Commun 2014; 5:5624. [PMID: 25427562 PMCID: PMC4248239 DOI: 10.1038/ncomms6624] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/20/2014] [Indexed: 11/28/2022] Open
Abstract
The earliest molecular events in T cell recognition have not yet been fully described, and the initial T cell receptor (TCR) triggering mechanism remains a subject of controversy. Here, using TIRF/FRET microscopy, we observe a two-stage interaction between TCR, CD8, and MHCp. There is an early (within seconds) interaction between CD3ζ and the coreceptor CD8 that is independent of the binding of CD8 to MHC, but that requires CD8 association with Lck. Later (several minutes) CD3ζ-CD8 interactions require CD8-MHC binding. Lck can be found free or bound to the coreceptor. This work indicates that the initial TCR triggering event is induced by free Lck.
Collapse
|
15
|
Marshall D, Sinclair C, Tung S, Seddon B. Differential requirement for IL-2 and IL-15 during bifurcated development of thymic regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:5525-33. [PMID: 25348623 DOI: 10.4049/jimmunol.1402144] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The developmental pathways of regulatory T cells (T(reg)) generation in the thymus are not fully understood. In this study, we reconstituted thymic development of Zap70-deficient thymocytes with a tetracycline-inducible Zap70 transgene to allow temporal dissection of T(reg) development. We find that T(reg) develop with distinctive kinetics, first appearing by day 4 among CD4 single-positive (SP) thymocytes. Accepted models of CD25(+)Foxp3(+) T(reg) selection suggest development via CD25(+)Foxp3(-) CD4 SP precursors. In contrast, our kinetic analysis revealed the presence of abundant CD25(-)Foxp3(+) cells that are highly efficient at maturing to CD25(+)Foxp3(+) cells in response to IL-2. CD25(-)Foxp3(+) cells more closely resembled mature T(reg) both with respect to kinetics of development and avidity for self-peptide MHC. These population also exhibited distinct requirements for cytokines during their development. CD25(-)Foxp3(+) cells were IL-15 dependent, whereas generation of CD25(+)Foxp3(+) specifically required IL-2. Finally, we found that IL-2 and IL-15 arose from distinct sources in vivo. IL-15 was of stromal origin, whereas IL-2 was of exclusively from hemopoetic cells that depended on intact CD4 lineage development but not either Ag-experienced or NKT cells.
Collapse
Affiliation(s)
- Daniel Marshall
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London, NW7 1AA, United Kingdom
| | - Charles Sinclair
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London, NW7 1AA, United Kingdom
| | - Sim Tung
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London, NW7 1AA, United Kingdom
| | - Benedict Seddon
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London, NW7 1AA, United Kingdom
| |
Collapse
|
16
|
Schim van der Loeff I, Hsu LY, Saini M, Weiss A, Seddon B. Zap70 is essential for long-term survival of naive CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:2873-80. [PMID: 25092893 DOI: 10.4049/jimmunol.1400858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Survival of naive T cells requires engagement of TCR with self-peptide major histocompatibility Ags. The signaling pathways required to transmit this survival signal are poorly understood. In this study, we asked whether the tyrosine kinase Zap70 is required to transmit survival signals in naive CD8 T cells. In the absence of Zap70 expression, thymic development is completely blocked. Using a tetracycline-inducible Zap70 transgene (TetZap70), thymic development of Zap70-deficient TCR transgenic F5 mice was restored. Feeding mice doxycycline to induce Zap70 expression resulted in repopulation of the peripheral naive compartment. Zap70 transgene expression was then ablated by withdrawal of doxycycline. Survival of Zap70-deficient naive CD8 T cells depended on host environment. In hosts with a replete T cell compartment, naive T cells died rapidly in the absence of Zap70 expression. In lymphopenic hosts, Zap70-deficient T cells survived far longer, in an IL-7-dependent manner, but failed to undergo lymphopenia-induced proliferation. Analyzing mixed bone marrow chimeras revealed that intact Zap70-dependent signaling was important for integration of recent thymic emigrants into the mature naive compartment. Finally, we asked whether adaptor function conferred by Zap70 tyrosines 315 and 319 was necessary for transmission of homeostatic TCR signals. This was done by analyzing F5 mice expressing mutant Zap70 in which these residues had been mutated to alanines (Zap70(YYAA)). Inducible Zap70 expression rescued thymic development in F5 TetZap70 Zap70(YYAA) mice. However, in the absence of wild-type Zap70 expression, the Zap70(YYAA) mutant failed to transmit either survival or proliferative homeostatic signals.
Collapse
Affiliation(s)
- Ina Schim van der Loeff
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom; and
| | - Lih-Yun Hsu
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Manoj Saini
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom; and
| | - Art Weiss
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Benedict Seddon
- Division of Immune Cell Biology, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom; and
| |
Collapse
|
17
|
Ahmed Rahim MM, Chrobak P, Priceputu E, Hanna Z, Jolicoeur P. Normal development and function but impaired memory phenotype of CD8+ T cells in transgenic mice expressing HIV-1 Nef in its natural target cells. Virology 2013; 438:84-97. [DOI: 10.1016/j.virol.2013.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/08/2012] [Accepted: 01/16/2013] [Indexed: 10/27/2022]
|
18
|
Self-peptides in TCR repertoire selection and peripheral T cell function. Curr Top Microbiol Immunol 2013; 373:49-67. [PMID: 23612987 DOI: 10.1007/82_2013_319] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The vertebrate antigen receptors are anticipatory in their antigen recognition and display a vast diversity. Antigen receptors are assembled through V(D)J recombination, in which one of each Variable, (Diverse), and Joining gene segment are randomly utilized and recombined. Both gene rearrangement and mutational insertion are generated through randomness; therefore, the process of antigen receptors generation requires a rigorous testing system to select every receptor which is useful to recognize foreign antigens, but which would cause no harm to self cells. In the case of T cell receptors (TCR), such a quality control responsibility rests in thymic positive and negative selection. In this review, we focus on the critical involvement of self-peptides in the generation of a T cell repertoire, discuss the role of T cell thymic development in shaping the specificity of TCR repertoire, and directing function fitness of mature T cells in periphery. Here, we consider thymic positive selection to be not merely a one-time maturing experience for an individual T cell, but a life-long imprinting which influences the function of each individual T cell in periphery.
Collapse
|
19
|
Distinct mechanisms mediate naive and memory CD8 T-cell tolerance. Proc Natl Acad Sci U S A 2012; 109:21438-43. [PMID: 23236165 DOI: 10.1073/pnas.1217409110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Peripheral tolerance to developmentally regulated antigens is necessary to sustain tissue homeostasis. We have now devised an inducible and reversible system that allows interrogation of T-cell tolerance induction in endogenous naïve and memory CD8 T cells. Our data show that peripheral CD8 T-cell tolerance can be preserved through two distinct mechanisms, antigen addiction leading to anergy for naïve T cells and ignorance for memory T cells. Induction of antigen in dendritic cells resulted in substantial expansion and maintenance of endogenous antigen-specific CD8 T cells. The self-reactive cells initially exhibited effector activity but eventually became unresponsive. Upon antigen removal, the antigen-specific population waned, resulting in development of a self-specific memory subset that recalled to subsequent challenge. In striking contrast to naïve CD8 T cells, preexisting antigen-specific memory CD8 T cells failed to expand after antigen induction and essentially ignored the antigen despite widespread expression by dendritic cells. The inclusion of inflammatory signals partially overcame memory CD8 T-cell ignorance of self-antigen. Thus, peripheral CD8 T-cell tolerance for naïve CD8 T cells depended on the continuous presence of antigen, whereas memory CD8 T cells were prohibited from autoreactivity in the absence of inflammation.
Collapse
|
20
|
Berge T, Grønningsæter IHB, Lorvik KB, Abrahamsen G, Granum S, Sundvold-Gjerstad V, Corthay A, Bogen B, Spurkland A. SH2D2A modulates T cell mediated protection to a B cell derived tumor in transgenic mice. PLoS One 2012; 7:e48239. [PMID: 23144743 PMCID: PMC3483153 DOI: 10.1371/journal.pone.0048239] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 09/21/2012] [Indexed: 12/20/2022] Open
Abstract
Background T cell specific adapter protein (TSAd), encoded by the SH2D2A gene, modulates signaling downstream of the T cell receptor (TCR). Young, unchallenged SH2D2A-deficient C57BL/6 mice exhibit a relatively normal immune phenotype. To address whether SH2D2A regulates physiologic immune responses, SH2D2A-deficient TCR-transgenic BALB/c mice were generated. The transgenic TCR recognizes a myeloma-derived idiotypic (Id) peptide in the context of the major histocompatibility complex (MHC) class II molecule I-Ed, and confers T cell mediated resistance to transplanted multiple myeloma development in vivo. Principal Findings The immune phenotype of SH2D2A-deficient C57BL/6 and BALB/c mice did not reveal major differences compared to the corresponding wild type mice. When challenged with myeloma cells, Id-specific TCR-transgenic BALB/c mice lacking SH2D2A displayed increased resistance towards tumor development. Tumor free TCR-transgenic SH2D2A-deficient mice had higher numbers of Id-specific single positive CD4+ thymocytes compared to TCR-transgenic wild-type mice. Conclusion Our results suggest a modulatory role for SH2D2A in T cell mediated immune surveillance of cancer. However, it remains to be established whether its effect is T-cell intrinsic. Further studies are required to determine whether targeting SH2D2A function in T cells may be a potential adjuvant in cancer immunotherapy.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Flow Cytometry
- Histocompatibility Antigens Class II/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Multiple Myeloma/genetics
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Thymocytes/immunology
- Thymocytes/metabolism
Collapse
Affiliation(s)
- Tone Berge
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The development of T cell in the thymus and the activation of mature T cells in the secondary lymphoid tissues require T cell to make adaptive responses to signaling molecules of environment. The activation of T cell receptor (TCR) signaling pathway could be induced by the interaction of the TCR and its co-receptor CD4 and CD8 with MHC/peptide complex. This process involves co-stimulatory molecules and signals mediated by cytokine receptors, which eventually leads to the occurrence of T cell immune response. The Src-family kinases lymphocyte-specific protein tyrosine kinase (Lck) and proto-oncogene tyrosine-protein kinase (Fyn) are expressed in T cells and serve as the signaling molecules that are activated downstream of TCR. These signaling molecules play key roles in development, positive selection, and peripheral maintenance of naive T cells and lymphopenia-induced proliferation of peripheral T cells. Both Lck and Fyn are required for each of these TCR-based signaling pathways, and Lck seems to be the major contributor, while Fyn can only supplement some functions of Lck. In this review, we discussed the mechanisms by which these two proteins perform functions in T cell development based on our current understanding.
Collapse
|
22
|
Gascoigne NRJ, Casas J, Brzostek J, Rybakin V. Initiation of TCR phosphorylation and signal transduction. Front Immunol 2011; 2:72. [PMID: 22566861 PMCID: PMC3342367 DOI: 10.3389/fimmu.2011.00072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 11/21/2011] [Indexed: 01/14/2023] Open
Abstract
Recent data with CD8+ T cells show that the initial phase of T cell receptor (TCR) binding to MHC–peptide (MHCp) is quickly followed by a second, stronger, binding phase representing the binding of CD8 to the MHCp. This second phase requires signaling by a Src-family kinase such as Lck. These data point out two aspects of the initial stage of TCR signaling that have not yet been clearly resolved. Firstly, how and by which Src-family kinase, is the initial phosphorylation of CD3ζ accomplished, given that the Lck associated with the co-receptors (CD4 or CD8) is not yet available. Secondly, what is the mechanism by which the co-receptor is brought close to the bound TCR before the co-receptor binds to MHCp?
Collapse
Affiliation(s)
- Nicholas R J Gascoigne
- Department of Immunology and Microbial Science, The Scripps Research Institute La Jolla, CA, USA.
| | | | | | | |
Collapse
|
23
|
Wiede F, Shields BJ, Chew SH, Kyparissoudis K, van Vliet C, Galic S, Tremblay ML, Russell SM, Godfrey DI, Tiganis T. T cell protein tyrosine phosphatase attenuates T cell signaling to maintain tolerance in mice. J Clin Invest 2011; 121:4758-74. [PMID: 22080863 PMCID: PMC3226006 DOI: 10.1172/jci59492] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 10/07/2011] [Indexed: 12/14/2022] Open
Abstract
Many autoimmune diseases exhibit familial aggregation, indicating that they have genetic determinants. Single nucleotide polymorphisms in PTPN2, which encodes T cell protein tyrosine phosphatase (TCPTP), have been linked with the development of several autoimmune diseases, including type 1 diabetes and Crohn's disease. In this study, we have identified TCPTP as a key negative regulator of TCR signaling, which might explain the association of PTPN2 SNPs with autoimmune disease. We found that TCPTP dephosphorylates and inactivates Src family kinases to regulate T cell responses. Using T cell-specific TCPTP-deficient mice, we established that TCPTP attenuates T cell activation and proliferation in vitro and blunts antigen-induced responses in vivo. TCPTP deficiency lowered the in vivo threshold for TCR-dependent CD8(+) T cell proliferation. Consistent with this, T cell-specific TCPTP-deficient mice developed widespread inflammation and autoimmunity that was transferable to wild-type recipient mice by CD8(+) T cells alone. This autoimmunity was associated with increased serum levels of proinflammatory cytokines and anti-nuclear antibodies, T cell infiltrates in non-lymphoid tissues, and liver disease. These data indicate that TCPTP is a critical negative regulator of TCR signaling that sets the threshold for TCR-induced naive T cell responses to prevent autoimmune and inflammatory disorders arising.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/biosynthesis
- Autoimmune Diseases/enzymology
- Autoimmune Diseases/etiology
- Autoimmune Diseases/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- Immune Tolerance/immunology
- Inflammation/blood
- Inflammation/genetics
- Inflammation/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Phosphorylation
- Protein Processing, Post-Translational
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology
- Radiation Chimera
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction/immunology
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- Thymocytes/pathology
- ZAP-70 Protein-Tyrosine Kinase/physiology
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Florian Wiede
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Benjamin J. Shields
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Sock Hui Chew
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Konstantinos Kyparissoudis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Catherine van Vliet
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Sandra Galic
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Michel L. Tremblay
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Sarah M. Russell
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Dale I. Godfrey
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
24
|
Sinclair C, Saini M, Schim van der Loeff I, Sakaguchi S, Seddon B. The Long-Term Survival Potential of Mature T Lymphocytes Is Programmed During Development in the Thymus. Sci Signal 2011; 4:ra77. [DOI: 10.1126/scisignal.2002246] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Moran AE, Holzapfel KL, Xing Y, Cunningham NR, Maltzman JS, Punt J, Hogquist KA. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. ACTA ACUST UNITED AC 2011; 208:1279-89. [PMID: 21606508 PMCID: PMC3173240 DOI: 10.1084/jem.20110308] [Citation(s) in RCA: 785] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ability of antigen receptors to engage self-ligands with varying affinity is crucial for lymphocyte development. To further explore this concept, we generated transgenic mice expressing GFP from the immediate early gene Nr4a1 (Nur77) locus. GFP was up-regulated in lymphocytes by antigen receptor stimulation but not by inflammatory stimuli. In T cells, GFP was induced during positive selection, required major histocompatibility complex for maintenance, and directly correlated with the strength of T cell receptor (TCR) stimulus. Thus, our results define a novel tool for studying antigen receptor activation in vivo. Using this model, we show that regulatory T cells (T(reg) cells) and invariant NKT cells (iNKT cells) perceived stronger TCR signals than conventional T cells during development. However, although T(reg) cells continued to perceive strong TCR signals in the periphery, iNKT cells did not. Finally, we show that T(reg) cell progenitors compete for recognition of rare stimulatory TCR self-ligands.
Collapse
Affiliation(s)
- Amy E Moran
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Tsun A, Qureshi I, Stinchcombe JC, Jenkins MR, de la Roche M, Kleczkowska J, Zamoyska R, Griffiths GM. Centrosome docking at the immunological synapse is controlled by Lck signaling. J Cell Biol 2011; 192:663-74. [PMID: 21339332 PMCID: PMC3044125 DOI: 10.1083/jcb.201008140] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 01/25/2011] [Indexed: 11/22/2022] Open
Abstract
Docking of the centrosome at the plasma membrane directs lytic granules to the immunological synapse. To identify signals controlling centrosome docking at the synapse, we have studied cytotoxic T lymphocytes (CTLs) in which expression of the T cell receptor-activated tyrosine kinase Lck is ablated. In the absence of Lck, the centrosome is able to translocate around the nucleus toward the immunological synapse but is unable to dock at the plasma membrane. Lytic granules fail to polarize and release their contents, and target cells are not killed. In CTLs deficient in both Lck and the related tyrosine kinase Fyn, centrosome translocation is impaired, and the centrosome remains on the distal side of the nucleus relative to the synapse. These results show that repositioning of the centrosome in CTLs involves at least two distinct steps, with Lck signaling required for the centrosome to dock at the plasma membrane.
Collapse
Affiliation(s)
- Andy Tsun
- Cambridge Institute for Medical Research, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0XY, England, UK
| | - Ihjaaz Qureshi
- Division of Immune Cell Biology, National Institute for Medical Research, London NW7 1AA, England, UK
| | - Jane C. Stinchcombe
- Cambridge Institute for Medical Research, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0XY, England, UK
| | - Misty R. Jenkins
- Cambridge Institute for Medical Research, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0XY, England, UK
| | - Maike de la Roche
- Cambridge Institute for Medical Research, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0XY, England, UK
| | - Joanna Kleczkowska
- Division of Immune Cell Biology, National Institute for Medical Research, London NW7 1AA, England, UK
| | - Rose Zamoyska
- Division of Immune Cell Biology, National Institute for Medical Research, London NW7 1AA, England, UK
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, Scotland, UK
| | - Gillian M. Griffiths
- Cambridge Institute for Medical Research, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0XY, England, UK
| |
Collapse
|
27
|
Caserta S, Kleczkowska J, Mondino A, Zamoyska R. Reduced functional avidity promotes central and effector memory CD4 T cell responses to tumor-associated antigens. THE JOURNAL OF IMMUNOLOGY 2010; 185:6545-54. [PMID: 21048115 DOI: 10.4049/jimmunol.1001867] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effect of TCR signals on the differentiation of memory T cells is poorly defined. Conventional wisdom suggests that high-avidity interactions are best for the selection of vaccine Ag candidates or T cell specificities for adoptive T cell therapy to stimulate robust responses. However, in conditions of Ag persistence, high-avidity clones might exhaust and fail to form long-lived protective memory. We have manipulated the functional avidity of CD4 T cells by reducing expression of Lck, a key kinase involved in TCR triggering. Using a mouse model, we followed tetramer-positive T cells responding to a tumor Ag expressed by an adenocarcinoma. We show that reducing the functional avidity increased effector-effector memory responses and improved the generation of self-renewing, recirculating, tumor Ag-specific memory phenotype CD4 T cells. Moreover, such cells together with wild type CD8 T cells were better able to control tumor growth. Mechanistically, reducing Lck prolonged IL-2 production and cell turnover in the central memory population while reducing expression of exhaustion markers in the face of chronic Ag. Our data indicate that, in situations of persistent Ag challenge, generating T cells with reduced functional avidity may elicit more effective immune responses.
Collapse
Affiliation(s)
- Stefano Caserta
- Institute for Immunology and Infection Research, The University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | |
Collapse
|
28
|
Mislocalization of Lck impairs thymocyte differentiation and can promote development of thymomas. Blood 2010; 117:108-17. [PMID: 20876849 DOI: 10.1182/blood-2010-03-277160] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
T-cell development is critically dependent on the activities of the Src-family kinases p56(lck) and p59(fyn). While Lck plays a dominant role in the initiation of T-cell receptor (TCR) signaling and in thymocyte differentiation, Fyn plays a more subtle regulatory role. We sought to determine the role of intracellular localization in the differing functions of Lck and Fyn in T cells. By generating transgenic mice that express chimeric Lck-Fyn proteins, we showed that the N-terminal unique domain determines the intracellular localization and function of Lck in pre-TCR and mature αβTCR signaling in vivo. Furthermore, coexpression of a "domain-swap" Lck protein containing the Fyn unique domain with an inducible Lck transgene resulted in the development of thymomas. In contrast to previous reports of Lck-driven thymomas, tumor development was dependent on either pre-TCR or mature TCR signals, and was completely ablated when mice were crossed to a recombination activating gene 1 (Rag1)-deficient background. These data provide a mechanistic basis for the differing roles of Lck and Fyn in T-cell development, and show that intracellular localization as determined by the N-terminal unique domains is critical for Src-family kinase function in vivo.
Collapse
|
29
|
Chrobak P, Simard MC, Bouchard N, Ndolo TM, Guertin J, Hanna Z, Dave V, Jolicoeur P. HIV-1 Nef Disrupts Maturation of CD4+T Cells through CD4/Lck Modulation. THE JOURNAL OF IMMUNOLOGY 2010; 185:3948-59. [DOI: 10.4049/jimmunol.1001064] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
Hayes SM, Laird RM, Love PE. Beyond alphabeta/gammadelta lineage commitment: TCR signal strength regulates gammadelta T cell maturation and effector fate. Semin Immunol 2010; 22:247-51. [PMID: 20452783 PMCID: PMC3129014 DOI: 10.1016/j.smim.2010.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 04/02/2010] [Accepted: 04/14/2010] [Indexed: 12/25/2022]
Abstract
Signaling by the gammadelta T cell receptor (TCR) is required not only for alphabeta/gammadelta lineage commitment but also to activate and elicit effector functions in mature gammadelta T cells. Notably, at both of these stages, the signal delivered by the gammadeltaTCR is more robust than the one delivered by either the preTCR or the alphabetaTCR. Recent studies now provide evidence that signaling by the gammadeltaTCR is also required at other stages during gammadelta T cell development. Remarkably, the strength of the gammadeltaTCR signal also plays a role at these other stages, as evidenced by the findings that genetic manipulation of gammadeltaTCR signal strength affects gammadelta T cell maturation and effector fate. In this review, we discuss how a strong TCR signal is a recurring theme in gammadelta T cell development and activation.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Lineage
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Signal Transduction
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Sandra M Hayes
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| | | | | |
Collapse
|
31
|
Alexandropoulos K, Regelmann AG. Regulation of T-lymphocyte physiology by the Chat-H/CasL adapter complex. Immunol Rev 2010; 232:160-74. [PMID: 19909363 DOI: 10.1111/j.1600-065x.2009.00831.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The Cas family of proteins consists of at least four members implicated in the regulation of diverse cellular processes such as cell proliferation, adhesion, motility, and cancer cell metastasis. Cas family members have conserved C-termini that mediate constitutive heterotypic interactions with members of a different group of proteins, the NSP family. Both the Cas and NSP proteins have conserved domains that mediate protein-protein interactions with other cytoplasmic intermediates. Signaling modules assembled by these proteins in turn regulate signal transduction downstream of a variety of receptors including integrin, chemokine, and antigen receptors. T lymphocytes express the NSP protein NSP3/Chat-H and the Cas protein Hef1/CasL, which are found in a constitutive complex in naive T cells. We recently showed that Chat-H and Hef1/CasL regulate integrin-mediated adhesion and promote T-cell migration and trafficking downstream of activated chemokine receptors. It is currently unclear if the Chat-H/CasL module also plays a role in antigen receptor signaling. Here we review our current knowledge of how Chat-H and Hef1/CasL regulate T-cell physiology and whether this protein complex plays a functional role downstream of T-cell receptor activation.
Collapse
Affiliation(s)
- Konstantina Alexandropoulos
- Department of Medicine, Division of Clinical Immunology, The Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
32
|
Abstract
During alphabeta T cell development, cells diverge into alternate CD4 helper and CD8(+) cytotoxic T cell lineages. The precise correlation between a T cell's CD8 and CD4 choice and its TCR specificity to class I or class II MHC was noted more than 20 years ago, and establishing the underlying mechanism has remained a focus of intense study since then. This review deals with three formerly discrete topics that are gradually becoming interconnected: the role of TCR signaling in lineage commitment, the regulation of expression of the CD4 and CD8 genes, and transcriptional regulation of lineage commitment. It is widely accepted that TCR signaling exerts a decisive influence on lineage choice, although the underlying mechanism remains intensely debated. Current evidence suggests that both duration and intensity of TCR signaling may control lineage choice, as proposed by the kinetic signaling and quantitative instructive models, respectively. Alternate expression of the CD4 and CD8 genes is the most visible manifestation of lineage choice, and much progress has been made in defining the responsible cis elements and transcription factors. Finally, important clues to the molecular basis of lineage commitment have been provided by the recent identification of the transcription factor ThPOK as a key regulator of lineage choice. ThPOK is selectively expressed in class II-restricted cells at the CD4(+)8(lo) stage and is necessary and sufficient for development to the CD4 lineage. Given the central role of ThPOK in lineage commitment, understanding its upstream regulation and downstream gene targets is expected to reveal further important aspects of the molecular machinery underlying lineage commitment.
Collapse
Affiliation(s)
- Xi He
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | |
Collapse
|
33
|
Saini M, Sinclair C, Marshall D, Tolaini M, Sakaguchi S, Seddon B. Regulation of Zap70 expression during thymocyte development enables temporal separation of CD4 and CD8 repertoire selection at different signaling thresholds. Sci Signal 2010; 3:ra23. [PMID: 20332428 DOI: 10.1126/scisignal.2000702] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
To investigate the temporal regulation of the commitment of immature thymocytes to either the CD4(+) or the CD8(+) lineage in the thymus, we developed a transgenic mouse that expressed a tetracycline-inducible gene encoding the tyrosine kinase zeta chain-associated protein kinase of 70 kD (Zap70), which restored development in Zap70(-/-) thymocytes arrested at the preselection, CD4(+)CD8(+) double-positive (DP) stage. After induction of the expression of Zap70 and the production of Zap70 protein, CD4(+) single-positive (SP) cells that expressed Zbtb7b (which encodes the CD4(+) T cell-associated transcription factor ThPOK) became abundant within 30 hours, whereas CD8(+) SP cells were not detectable until day 4. We found that mature CD4(+) and CD8(+) cells arose from phenotypically distinct subsets of DP thymocytes that developed with different kinetics and contrasting sensitivities to stimulation of the T cell antigen receptor (TCR). In wild-type mice, expression of endogenous Zap70 progressively increased during maturation of the DP subsets, and the abundance of Zap70 protein determined the sensitivity of the cells to stimulation of the TCR. This temporal gradient in the amount of Zap70 protein enabled the selection of CD4(+) and CD8(+) repertoires in separate temporal windows and at different TCR signaling thresholds, thereby facilitating discrimination of distinct positive selection signals in these lineages.
Collapse
Affiliation(s)
- Manoj Saini
- Division of Immune Cell Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
The T cell antigen receptor (TCR) serves as a paradigm for how membrane receptors transmit signals to the cytoplasm because it controls many aspects of T cell differentiation and function by detecting atom-sized variations in the quality of the ligand that is recognized. The mechanisms that underlie the different signaling outcomes are unclear. Studies that suggest a ligand-tailored, qualitatively different signal are confronted with evidence that favors a quantitative model, and studies of TCR-dependent T cell differentiation in the thymus are no exception. Mature T cells with an alphabeta TCR are classified according to two major distinct subsets based on the mutually exclusive presence of the co-receptors CD4 and CD8, which play essential roles in recognition of the major histocompatibility complex (MHC) class II and I ligands, respectively, and in the recruitment of the tyrosine kinase Lck to the TCR complex. Mature CD4(+) and CD8(+) T cells derive from a common precursor in the thymus, a double-positive (DP) thymocyte, which has both co-receptors. Early signaling models suggested that the differential capacity of CD4 and CD8 to recruit Lck to the TCR underlay lineage decision. A study now shows that differentiation into the CD8(+) lineage requires the TCR-induced increased abundance of the tyrosine kinase zeta chain-associated protein kinase of 70 kD (Zap70). This finding, together with the known importance of Lck in the determination of CD4(+) and CD8(+) lineages, enables us to propose that a balance between the activation of these two kinases by the TCR determines lineage decisions.
Collapse
Affiliation(s)
- Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid 28049, Spain.
| | | |
Collapse
|
35
|
Laird RM, Hayes SM. Roles of the Src tyrosine kinases Lck and Fyn in regulating gammadeltaTCR signal strength. PLoS One 2010; 5:e8899. [PMID: 20126650 PMCID: PMC2811189 DOI: 10.1371/journal.pone.0008899] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 01/06/2010] [Indexed: 01/17/2023] Open
Abstract
Lck and Fyn, members of the Src family of tyrosine kinases, are key components of the αβTCR-coupled signaling pathway. While it is generally accepted that both Lck and Fyn positively regulate signal transduction by the αβTCR, recent studies have shown that Lck and Fyn have distinct functions in this signaling pathway, with Lck being a positive regulator and Fyn being a negative regulator of αβTCR signal transduction. To determine whether Lck and Fyn also differentially regulate γδTCR signal transduction, we analyzed γδ T cell development and function in mice with reduced Lck or Fyn expression levels. We found that reducing Lck or Fyn levels altered the strength of the γδTCR signaling response, with low levels of Lck weakening γδTCR signal strength and low levels of Fyn augmenting γδTCR signal strength. These alterations in γδTCR signal strength had profound effects not only on αβ/γδ lineage choice, but also on γδ thymocyte maturation and γδ T cell effector function. These results indicate that the cellular levels of Lck and Fyn play a role in regulating the strength of the γδTCR signaling response at different stages in the life of the γδ T cell.
Collapse
Affiliation(s)
- Renee M. Laird
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, United States of America
| | - Sandra M. Hayes
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, United States of America
- * E-mail:
| |
Collapse
|
36
|
Salmond RJ, Emery J, Okkenhaug K, Zamoyska R. MAPK, Phosphatidylinositol 3-Kinase, and Mammalian Target of Rapamycin Pathways Converge at the Level of Ribosomal Protein S6 Phosphorylation to Control Metabolic Signaling in CD8 T Cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:7388-97. [DOI: 10.4049/jimmunol.0902294] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
Boding L, Bonefeld CM, Nielsen BL, Lauritsen JPH, von Essen MR, Hansen AK, Larsen JM, Nielsen MM, Odum N, Geisler C. TCR down-regulation controls T cell homeostasis. THE JOURNAL OF IMMUNOLOGY 2009; 183:4994-5005. [PMID: 19801521 DOI: 10.4049/jimmunol.0901539] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR and cytokine receptor signaling play key roles in the complex homeostatic mechanisms that maintain a relative stable number of T cells throughout life. Despite the homeostatic mechanisms, a slow decline in naive T cells is typically observed with age. The CD3gamma di-leucine-based motif controls TCR down-regulation and plays a central role in fine-tuning TCR expression and signaling in T cells. In this study, we show that the age-associated decline of naive T cells is strongly accelerated in CD3gammaLLAA knock-in mice homozygous for a double leucine to alanine mutation in the CD3gamma di-leucine-based motif, whereas the number of memory T cells is unaffected by the mutation. This results in premature T cell population senescence with a severe dominance of memory T cells and very few naive T cells in middle-aged to old CD3gamma mutant mice. The reduced number of naive T cells in CD3gamma mutant mice was caused by the combination of reduced thymic output, decreased T cell apoptosis, and increased transition of naive T cells to memory T cells. Experiments with bone marrow chimeric mice confirmed that the CD3gammaLLAA mutation exerted a T cell intrinsic effect on T cell homeostasis that resulted in an increased transition of CD3gammaLLAA naive T cells to memory T cells and a survival advantage of CD3gammaLLAA T cells compared with wild-type T cells. The experimental observations were further supported by mathematical modeling of T cell homeostasis. Our study thus identifies an important role of CD3gamma-mediated TCR down-regulation in T cell homeostasis.
Collapse
Affiliation(s)
- Lasse Boding
- Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Salmond RJ, Filby A, Qureshi I, Caserta S, Zamoyska R. T-cell receptor proximal signaling via the Src-family kinases, Lck and Fyn, influences T-cell activation, differentiation, and tolerance. Immunol Rev 2009; 228:9-22. [PMID: 19290918 DOI: 10.1111/j.1600-065x.2008.00745.x] [Citation(s) in RCA: 270] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
T-cell development in the thymus and activation of mature T cells in secondary lymphoid organs requires the ability of cells to respond appropriately to environmental signals at multiple stages of their development. The process of thymocyte selection insures a functional T-cell repertoire, while activation of naive peripheral T cells induces proliferation, gain of effector function, and, ultimately, long-lived T-cell memory. The T-cell immune response is initiated upon engagement of the T-cell receptor (TCR) and coreceptor, CD4 or CD8, by cognate antigen/major histocompatibility complexes presented by antigen-presenting cells. TCR/coreceptor engagement induces the activation of biochemical signaling pathways that, in combination with signals from costimulator molecules and cytokine receptors, direct the outcome of the response. Activation of the src-family kinases p56(lck) (Lck) and p59(fyn) (Fyn) is central to the initiation of TCR signaling pathways. This review focuses on our current understanding of the mechanisms by which these two proteins orchestrate T-cell function.
Collapse
Affiliation(s)
- Robert J Salmond
- Molecular Immunology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, UK
| | | | | | | | | |
Collapse
|
39
|
Komaniwa S, Hayashi H, Kawamoto H, Sato SB, Ikawa T, Katsura Y, Udaka K. Lipid-mediated presentation of MHC class II molecules guides thymocytes to the CD4 lineage. Eur J Immunol 2008; 39:96-112. [DOI: 10.1002/eji.200838796] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
40
|
Restoration of T-box-containing protein expressed in T cells protects against allergen-induced asthma. J Allergy Clin Immunol 2008; 123:479-85. [PMID: 19081613 DOI: 10.1016/j.jaci.2008.10.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 10/21/2008] [Accepted: 10/22/2008] [Indexed: 12/22/2022]
Abstract
BACKGROUND A T(H)1-specific transcription factor, T-box-containing protein expressed in T cells (T-bet), controls the production of both T(H)1 and T(H)2 cytokines in T(H) cell differentiation by means of distinct mechanisms. T-bet-deficient mice overproduce T(H)2 cytokines and have spontaneous airway inflammation. OBJECTIVES We tested whether T-bet overexpression could protect against the development or progression of asthma. METHODS We generated a T cell-specific and inducible line of T-bet-transgenic mice on a T-bet-deficient genetic background and used it to study the function of T-bet in an ovalbumin (OVA)-induced asthma model. RESULTS Induction of T-bet in a T cell-specific manner in an OVA model of asthma concomitant with OVA injection prevented airway hyperresponsiveness, eosinophilic and lymphocytic inflammation, and IL-5 and IL-13 production in bronchoalveolar lavage fluid and also reduced serum IgE and T(H)2 cytokine production by peripheral T cells. Even when T-bet expression was induced during later stages of asthma progression, T-bet overexpression still attenuated airway hyperresponsiveness and goblet cell hyperplasia, as well as T(H)2 cytokine production. CONCLUSIONS Our results suggest that T-bet expression in T cells can prevent the initiation of airway inflammation and progression of chronic inflammation and might be extrapolated to human asthma.
Collapse
|
41
|
Werneck MBF, Lugo-Villarino G, Hwang ES, Cantor H, Glimcher LH. T-bet plays a key role in NK-mediated control of melanoma metastatic disease. THE JOURNAL OF IMMUNOLOGY 2008; 180:8004-10. [PMID: 18523263 DOI: 10.4049/jimmunol.180.12.8004] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Antitumor responses depend on type 1 immunity, which is severely impaired in mice deficient for the T-box expressed in T cells (T-bet) transcription factor. Both T-bet-deficient (T-bet(-/-)) NK and CTL show defective function, which can be overcome by strong stimuli due to the expression of eomesodermin, another member of the T-box family. The effective response from T-bet(-/-) mice to viral infection and tumor initiation corroborates with these findings. However, T-bet(-/-) animals fail to control cancer metastasis and are, therefore, highly susceptible to tumor spread. The mechanism of T-bet-dependent resistance to metastatic disease is not known. In this study, we show that T-bet plays a role in inhibiting cancer metastasis by regulating the longevity and function of NK cells. Our data demonstrate that the absence of a proper innate immune response driven by NK cells in T-bet(-/-) mice precludes the initiation of a potent adaptive response to tumors. Adoptive transfer of wild-type activated NK cells protects T-bet(-/-) animals after melanoma challenge showing that reconstitution of the NK compartment in these mice is sufficient to mediate a significant reduction in tumor burden. Transfer of T-bet(-/-) A-NK cells fails to do so, due to their reduced in vivo survival, inefficient lysis of cancer cells, and poor IFN-gamma production. Taken together, these results show for the first time an irreplaceable role for T-bet in the NK-mediated cross-talk between innate and adaptive immune responses to metastatic disease.
Collapse
Affiliation(s)
- Miriam B F Werneck
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
42
|
Defective T-cell ERK signaling induces interferon-regulated gene expression and overexpression of methylation-sensitive genes similar to lupus patients. Genes Immun 2008; 9:368-78. [PMID: 18523434 DOI: 10.1038/gene.2008.29] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies against a host of nuclear antigens. The pathogenesis of lupus is incompletely understood. Environmental factors may play a role via altering DNA methylation, a mechanism regulating gene expression. In lupus, genes including CD11a and CD70 are overexpressed in T cells as a result of promoter hypomethylation. T-cell DNA methyltransferase expression is regulated in part by the extracellular signal-regulated kinase (ERK) signaling pathway. In this study, we investigate the effects of decreased ERK pathway signaling in T cells using transgenic animals. We generated a transgenic mouse that inducibly expresses a dominant-negative MEK in T cells in the presence of doxycycline. We show that decreased ERK pathway signaling in T cells results in decreased expression of DNA methyltransferase 1 and overexpression of the methylation-sensitive genes CD11a and CD70, similar to T cells in human lupus. Our transgenic animal model also develops anti-dsDNA antibodies. Interestingly, microarray expression assays revealed overexpression of several interferon-regulated genes in the spleen similar to peripheral blood cells of lupus patients. This model supports the contention that ERK pathway signaling defects in T cells contribute to the development of autoimmunity.
Collapse
|
43
|
Turner MJ, Jellison ER, Lingenheld EG, Puddington L, Lefrançois L. Avidity maturation of memory CD8 T cells is limited by self-antigen expression. J Exp Med 2008; 205:1859-68. [PMID: 18625745 PMCID: PMC2525599 DOI: 10.1084/jem.20072390] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 05/28/2008] [Indexed: 12/22/2022] Open
Abstract
Immune tolerance to self-antigens is a complex process that utilizes multiple mechanisms working in concert to maintain homeostasis and prevent autoimmunity. We developed a system that revealed a population of self-specific CD8 T cells within the endogenous T cell repertoire. Immunization of ovalbumin (OVA)-expressing transgenic mice with recombinant viruses expressing OVA-peptide variants induced self-reactive T cells in vivo that matured into memory T cells able to respond to secondary infection. However, whereas the avidity of memory cells in normal mice increased dramatically with repeated immunizations, avidity maturation was limited for self-specific CD8 T cells. Despite decreased avidity, such memory cells afforded protection against infection, but did not induce overt autoimmunity. Further, up-regulation of self-antigen expression in dendritic cells using an inducible system promoted programmed death-1 expression, but not clonal expansion of preexisting memory cells. Thus, the self-reactive T cell repertoire is controlled by overlapping mechanisms influenced by antigen dose.
Collapse
Affiliation(s)
- Michael J Turner
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | |
Collapse
|
44
|
He X, Park K, Wang H, He X, Zhang Y, Hua X, Li Y, Kappes DJ. CD4-CD8 lineage commitment is regulated by a silencer element at the ThPOK transcription-factor locus. Immunity 2008; 28:346-58. [PMID: 18342007 DOI: 10.1016/j.immuni.2008.02.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2007] [Revised: 01/28/2008] [Accepted: 02/07/2008] [Indexed: 10/22/2022]
Abstract
The transcription factor ThPOK is necessary and sufficient to trigger adoption of the CD4 lymphocyte fate. Here we investigate the regulation of ThPOK expression and its subsequent control of CD4+ T cell commitment. Treatment of immature thymocytes with anti-TCR (T cell receptor) showed that TCR signals were important in ThPOK induction and that the CD4+8lo stage was the likely target of the inductive TCR signal. We identified at the ThPOK locus a key distal regulatory element (DRE) that mediated its differential expression in class I- versus II-restricted CD4+8lo thymocytes. The DRE was both necessary for suppression of ThPOK expression in class I-restricted thymocytes and sufficient for its induction in class II-restricted thymocytes. Mutagenesis analysis defined an essential 80bp core DRE sequence and its potential regulatory motifs. We propose a silencer-dependent model of lineage choice, whereby inactivation of the DRE silencer by a strong TCR signal leads to CD4 commitment, whereas continued silencer activity leads to CD8 commitment.
Collapse
Affiliation(s)
- Xi He
- Fox Chase Cancer Center, 7701 Burholme Ave., Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Furmanski AL, Ferreira C, Bartok I, Dimakou S, Rice J, Stevenson FK, Millrain MM, Simpson E, Dyson J. Public T Cell Receptor β-Chains Are Not Advantaged during Positive Selection. THE JOURNAL OF IMMUNOLOGY 2008; 180:1029-39. [DOI: 10.4049/jimmunol.180.2.1029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
46
|
Albu DI, Feng D, Bhattacharya D, Jenkins NA, Copeland NG, Liu P, Avram D. BCL11B is required for positive selection and survival of double-positive thymocytes. ACTA ACUST UNITED AC 2007; 204:3003-15. [PMID: 17998389 PMCID: PMC2118514 DOI: 10.1084/jem.20070863] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Transcriptional control of gene expression in double-positive (DP) thymocytes remains poorly understood. We show that the transcription factor BCL11B plays a critical role in DP thymocytes by controlling positive selection of both CD4 and CD8 lineages. BCL11B-deficient DP thymocytes rearrange T cell receptor (TCR) α; however, they display impaired proximal TCR signaling and attenuated extracellular signal-regulated kinase phosphorylation and calcium flux, which are all required for initiation of positive selection. Further, provision of transgenic TCRs did not improve positive selection of BCL11B-deficient DP thymocytes. BCL11B-deficient DP thymocytes have altered expression of genes with a role in positive selection, TCR signaling, and other signaling pathways intersecting the TCR, which may account for the defect. BCL11B-deficient DP thymocytes also presented increased susceptibility to spontaneous apoptosis associated with high levels of cleaved caspase-3 and an altered balance of proapoptotic/prosurvival factors. This latter susceptibility was manifested even in the absence of TCR signaling and was only partially rescued by provision of the BCL2 transgene, indicating that control of DP thymocyte survival by BCL11B is nonredundant and, at least in part, independent of BCL2 prosurvival factors.
Collapse
Affiliation(s)
- Diana I Albu
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Filby A, Seddon B, Kleczkowska J, Salmond R, Tomlinson P, Smida M, Lindquist JA, Schraven B, Zamoyska R. Fyn Regulates the Duration of TCR Engagement Needed for Commitment to Effector Function. THE JOURNAL OF IMMUNOLOGY 2007; 179:4635-44. [PMID: 17878361 DOI: 10.4049/jimmunol.179.7.4635] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In naive T cells, engagement of the TCR with agonist peptide:MHC molecules leads to phosphorylation of key intracellular signaling intermediates within seconds and this peaks within minutes. However, the cell does not commit to proliferation and IL-2 cytokine production unless receptor contact is sustained for several hours. The biochemical basis for this transition to full activation may underlie how T cells receive survival signals while maintaining tolerance, and is currently not well understood. We show here that for CD8 T cells commitment to proliferation and cytokine production requires sustained activation of the Src family kinase Lck and is opposed by the action of Fyn. Thus, in the absence of Fyn, commitment to activation occurs more rapidly, the cells produce more IL-2, and undergo more rounds of division. Our data demonstrate a role for Fyn in modulating the response to Ag in primary T cells.
Collapse
Affiliation(s)
- Andrew Filby
- Division of Molecular Immunology, Medical Research Council, National Institute for Medical Research, The Ridgeway, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The ability to generate genetically manipulated mice has revolutionized the study of development, cell biology, immunobiology and transplantation. Conventional gene targeting approaches lead to inactivation of the target gene in all tissues. This approach often has unintended consequences, such as embryonic lethality, which preclude studying the originally intended tissue. Newer approaches allowing conditional gene expression in a tissue-specific or temporally controlled fashion have the advantage of normal development with gene deletion only in the desired tissues. While nuances to these techniques continue to be developed, the underlying concepts remain consistent. This minireview focuses on the use of conditional gene targeting in mice using the Cre-loxP system and drug inducible gene expression using the tetracycline system.
Collapse
Affiliation(s)
- J S Maltzman
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| | | |
Collapse
|
49
|
Fragoso RC, Pyarajan S, Irie HY, Burakoff SJ. A CD8/Lck transgene is able to drive thymocyte differentiation. THE JOURNAL OF IMMUNOLOGY 2006; 177:6007-17. [PMID: 17056525 DOI: 10.4049/jimmunol.177.9.6007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Efficient development of thymocytes requires participation of a CD8 or CD4 coreceptor in the TCR:MHC interaction. Both CD8 and CD4 coreceptor cytoplasmic domains associate with Lck. In this study, we attempted to delineate the role of CD8alpha-associated Lck in driving CD8 single positive (SP) thymocyte development. We used a chimeric molecule encoding the extracellular and transmembrane domains of CD8alpha fused to full-length Lck. In mice deficient for CD8alpha and transgenic for 2C, a MHC class I-restricted TCR, robust reconstitution of CD8 SP thymocytes occurred both centrally and peripherally. The reconstituted CD8 SP population was phenotypically and functionally comparable to 2C wild-type counterparts expressing endogenous CD8alpha. A CD8alpha/Lck kinase-dead chimera also resulted in reconstitution of CD8 SP thymocytes. Our results suggest that CD8alpha-associated Lck is sufficient to drive CD8 SP thymocyte development. Furthermore, this CD8 SP development may not necessarily depend on Lck kinase activity.
Collapse
Affiliation(s)
- Ruben C Fragoso
- Dana-Farber Cancer Institute, Department of Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
50
|
Lovatt M, Filby A, Parravicini V, Werlen G, Palmer E, Zamoyska R. Lck regulates the threshold of activation in primary T cells, while both Lck and Fyn contribute to the magnitude of the extracellular signal-related kinase response. Mol Cell Biol 2006; 26:8655-65. [PMID: 16966372 PMCID: PMC1636771 DOI: 10.1128/mcb.00168-06] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 02/28/2006] [Accepted: 08/24/2006] [Indexed: 11/20/2022] Open
Abstract
The src family kinases p56lck (Lck) and p59fyn (Fyn) are the most proximal signaling molecules to be activated downstream of the T-cell receptor. Using an inducible transgenic model, we can regulate the expression of Lck in primary T cells and ask how the signaling cascade and differentiation potential are affected by the absence or the presence of reduced levels of Lck. We show that in naïve T cells, Lck controls the threshold of activation by preferentially regulating multiple signaling pathways that result in the mobilization of Ca2+ through activation of phospholipase C-gamma and protein kinase C as well as activation of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway. Fyn is also able to stimulate the ERK/MAPK pathway in primary T cells but has little influence on the mobilization of Ca2+. Only Lck efficiently stimulates production of diacylglycerol and therefore RasGRP1 recruitment to the plasma membrane and phosphorylation of Shc, suggesting that Fyn activates ERK via a different upstream signaling route. Finally, we show that signals through Lck are essential for the development of T-cell-effector potential, particularly for effective cytokine transcription.
Collapse
Affiliation(s)
- Matthew Lovatt
- Molecular Immunology, MRC National Institute for Medical Research, The Ridgeway, London NW71AA, United Kingdom
| | | | | | | | | | | |
Collapse
|