1
|
Balagopalan L, Moreno T, Qin H, Angeles BC, Kondo T, Yi J, McIntire KM, Alvinez N, Pallikkuth S, Lee ME, Yamane H, Tran AD, Youkharibache P, Cachau RE, Taylor N, Samelson LE. Generation of antitumor chimeric antigen receptors incorporating T cell signaling motifs. Sci Signal 2024; 17:eadp8569. [PMID: 39042728 PMCID: PMC11389647 DOI: 10.1126/scisignal.adp8569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have been used to successfully treat various blood cancers, but adverse effects have limited their potential. Here, we developed chimeric adaptor proteins (CAPs) and CAR tyrosine kinases (CAR-TKs) in which the intracellular ζ T cell receptor (TCRζ) chain was replaced with intracellular protein domains to stimulate signaling downstream of the TCRζ chain. CAPs contain adaptor domains and the kinase domain of ZAP70, whereas CAR-TKs contain only ZAP70 domains. We hypothesized that CAPs and CAR-TKs would be more potent than CARs because they would bypass both the steps that define the signaling threshold of TCRζ and the inhibitory regulation of upstream molecules. CAPs were too potent and exhibited high tonic signaling in vitro. In contrast, CAR-TKs exhibited high antitumor efficacy and significantly enhanced long-term tumor clearance in leukemia-bearing NSG mice as compared with the conventional CD19-28ζ-CAR-T cells. CAR-TKs were activated in a manner independent of the kinase Lck and displayed slower phosphorylation kinetics and prolonged signaling compared with the 28ζ-CAR. Lck inhibition attenuated CAR-TK cell exhaustion and improved long-term function. The distinct signaling properties of CAR-TKs may therefore be harnessed to improve the in vivo efficacy of T cells engineered to express an antitumor chimeric receptor.
Collapse
MESH Headings
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Humans
- Signal Transduction/immunology
- Mice
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- ZAP-70 Protein-Tyrosine Kinase/metabolism
- ZAP-70 Protein-Tyrosine Kinase/genetics
- ZAP-70 Protein-Tyrosine Kinase/immunology
- Immunotherapy, Adoptive/methods
- Mice, Inbred NOD
- Cell Line, Tumor
- Phosphorylation
Collapse
Affiliation(s)
- Lakshmi Balagopalan
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Taylor Moreno
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Haiying Qin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD 20892, USA
| | - Benjamin C. Angeles
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Taisuke Kondo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD 20892, USA
| | - Jason Yi
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Katherine M. McIntire
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Neriah Alvinez
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Sandeep Pallikkuth
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Mariah E. Lee
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Hidehiro Yamane
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| | - Andy D. Tran
- Laboratory of Cancer Biology and Genetics (CCR Microscopy Core), National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philippe Youkharibache
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Raul E. Cachau
- Integrated Data Science Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD 20892, USA
| | - Lawrence E. Samelson
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD 20892 USA
| |
Collapse
|
2
|
Vögtle T, Baig AA, Volz J, Duchow TB, Pleines I, Dütting S, Nitschke L, Watson SP, Nieswandt B. Critical redundant functions of the adapters Grb2 and Gads in platelet (hem)ITAM signaling in mice. Platelets 2020; 31:801-811. [DOI: 10.1080/09537104.2019.1709633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Timo Vögtle
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Ayesha A. Baig
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Julia Volz
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Timothy B. Duchow
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Irina Pleines
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Sebastian Dütting
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Lars Nitschke
- Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany
| | - Stephen P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, UK
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
3
|
Gerth E, Mattner J. The Role of Adaptor Proteins in the Biology of Natural Killer T (NKT) Cells. Front Immunol 2019; 10:1449. [PMID: 31293596 PMCID: PMC6603179 DOI: 10.3389/fimmu.2019.01449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/10/2019] [Indexed: 12/31/2022] Open
Abstract
Adaptor proteins contribute to the selection, differentiation and activation of natural killer T (NKT) cells, an innate(-like) lymphocyte population endowed with powerful immunomodulatory properties. Distinct from conventional T lymphocytes NKT cells preferentially home to the liver, undergo a thymic maturation and differentiation process and recognize glycolipid antigens presented by the MHC class I-like molecule CD1d on antigen presenting cells. NKT cells express a semi-invariant T cell receptor (TCR), which combines the Vα14-Jα18 chain with a Vβ2, Vβ7, or Vβ8 chain in mice and the Vα24 chain with the Vβ11 chain in humans. The avidity of interactions between their TCR, the presented glycolipid antigen and CD1d govern the selection and differentiation of NKT cells. Compared to TCR ligation on conventional T cells engagement of the NKT cell TCR delivers substantially stronger signals, which trigger the unique NKT cell developmental program. Furthermore, NKT cells express a panoply of primarily inhibitory NK cell receptors (NKRs) that control their self-reactivity and avoid autoimmune activation. Adaptor proteins influence NKT cell biology through the integration of TCR, NKR and/or SLAM (signaling lymphocyte-activation molecule) receptor signals or the variation of CD1d-restricted antigen presentation. TCR and NKR ligation engage the SH2 domain-containing leukocyte protein of 76kDa slp-76 whereas the SLAM associated protein SAP serves as adaptor for the SLAM receptor family. Indeed, the selection and differentiation of NKT cells selectively requires co-stimulation via SLAM receptors. Furthermore, SAP deficiency causes X-linked lymphoproliferative disease with multiple immune defects including a lack of circulating NKT cells. While a deletion of slp-76 leads to a complete loss of all peripheral T cell populations, mutations in the SH2 domain of slp-76 selectively affect NKT cell biology. Furthermore, adaptor proteins influence the expression and trafficking of CD1d in antigen presenting cells and subsequently selection and activation of NKT cells. Adaptor protein complex 3 (AP-3), for example, is required for the efficient presentation of glycolipid antigens which require internalization and processing. Thus, our review will focus on the complex contribution of adaptor proteins to the delivery of TCR, NKR and SLAM receptor signals in the unique biology of NKT cells and CD1d-restricted antigen presentation.
Collapse
MESH Headings
- Adaptor Protein Complex 3/immunology
- Adaptor Protein Complex 3/metabolism
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antigen Presentation/immunology
- Antigens, CD1d/immunology
- Antigens, CD1d/metabolism
- Humans
- Lymphocyte Activation/immunology
- Mice
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Phosphoproteins/immunology
- Phosphoproteins/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Natural Killer Cell/immunology
- Receptors, Natural Killer Cell/metabolism
- Signaling Lymphocytic Activation Molecule Family/immunology
- Signaling Lymphocytic Activation Molecule Family/metabolism
Collapse
Affiliation(s)
| | - Jochen Mattner
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
Zhang T, de Waard AA, Wuhrer M, Spaapen RM. The Role of Glycosphingolipids in Immune Cell Functions. Front Immunol 2019; 10:90. [PMID: 30761148 PMCID: PMC6361815 DOI: 10.3389/fimmu.2019.00090] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/14/2019] [Indexed: 01/06/2023] Open
Abstract
Glycosphingolipids (GSLs) exhibit a variety of functions in cellular differentiation and interaction. Also, they are known to play a role as receptors in pathogen invasion. A less well-explored feature is the role of GSLs in immune cell function which is the subject of this review article. Here we summarize knowledge on GSL expression patterns in different immune cells. We review the changes in GSL expression during immune cell development and differentiation, maturation, and activation. Furthermore, we review how immune cell GSLs impact membrane organization, molecular signaling, and trans-interactions in cellular cross-talk. Another aspect covered is the role of GSLs as targets of antibody-based immunity in cancer. We expect that recent advances in analytical and genome editing technologies will help in the coming years to further our knowledge on the role of GSLs as modulators of immune cell function.
Collapse
Affiliation(s)
- Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Antonius A de Waard
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
5
|
Lewis JB, Scangarello FA, Murphy JM, Eidell KP, Sodipo MO, Ophir MJ, Sargeant R, Seminario MC, Bunnell SC. ADAP is an upstream regulator that precedes SLP-76 at sites of TCR engagement and stabilizes signaling microclusters. J Cell Sci 2018; 131:jcs215517. [PMID: 30305305 PMCID: PMC6240300 DOI: 10.1242/jcs.215517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 09/17/2018] [Indexed: 12/31/2022] Open
Abstract
Antigen recognition by the T cell receptor (TCR) directs the assembly of essential signaling complexes known as SLP-76 (also known as LCP2) microclusters. Here, we show that the interaction of the adhesion and degranulation-promoting adaptor protein (ADAP; also known as FYB1) with SLP-76 enables the formation of persistent microclusters and the stabilization of T cell contacts, promotes integrin-independent adhesion and enables the upregulation of CD69. By analyzing point mutants and using a novel phospho-specific antibody, we show that Y595 is essential for normal ADAP function, that virtually all tyrosine phosphorylation of ADAP is restricted to a Y595-phosphorylated (pY595) pool, and that multivalent interactions between the SLP-76 SH2 domain and its binding sites in ADAP are required to sustain ADAP phosphorylation. Although pY595 ADAP enters SLP-76 microclusters, non-phosphorylated ADAP is enriched in protrusive actin-rich structures. The pre-positioning of ADAP at the contact sites generated by these structures favors the retention of nascent SLP-76 oligomers and their assembly into persistent microclusters. Although ADAP is frequently depicted as an effector of SLP-76, our findings reveal that ADAP acts upstream of SLP-76 to convert labile, Ca2+-competent microclusters into stable adhesive junctions with enhanced signaling potential.
Collapse
Affiliation(s)
- Juliana B Lewis
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frank A Scangarello
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
- Medical Scientist Training Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Joanne M Murphy
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Keith P Eidell
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Michelle O Sodipo
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Michael J Ophir
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Ryan Sargeant
- Pacific Immunology Corporation, Ramona, CA 92065, USA
| | | | - Stephen C Bunnell
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
6
|
Breuning J, Philip B, Brown MH. Addition of the C-terminus of CD6 to a chimeric antigen receptor enhances cytotoxicity and does not compromise expression. Immunology 2018; 156:130-135. [PMID: 30300924 PMCID: PMC6328988 DOI: 10.1111/imm.13009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/23/2022] Open
Abstract
T cells expressing chimeric antigen receptors (CARs) are a promising new cancer immunotherapy that has now reached the clinic. CARs are synthetic receptors that redirect T cells towards a tumour‐associated antigen and activate them through various fused signalling regions, for example derived from CD3ζ, 4‐1BB or CD28. Analysis of the optimal combination of CAR components including signalling domains is not yet comprehensive and may vary with the particular application. The C‐terminus of the T‐cell surface receptor CD6 is critical for its co‐stimulatory effects and signals through two phospho‐tyrosine motifs that bind to the intracellular adaptor proteins GADS and SLP‐76. Addition of the C terminus of CD6 did not compromise CAR expression, showing it was a stable moiety that can be used independently of the native receptor. A third‐generation CAR containing 4‐1BB, CD3ζ and the C terminus of CD6 (4‐1BBz‐CD6) enhanced interferon‐γ release and cytotoxicity when compared with the second‐generation 4‐1BB CD3ζ (4‐1BBz) CAR. The CD6 C terminus is a valuable addition to potential components for modular design of CARs to improve effector function, particularly cytotoxicity.
Collapse
Affiliation(s)
| | - Brian Philip
- Cancer Institute, University College, London, UK
| | | |
Collapse
|
7
|
Dezorella N, Katz BZ, Shapiro M, Polliack A, Perry C, Herishanu Y. SLP76 integrates into the B-cell receptor signaling cascade in chronic lymphocytic leukemia cells and is associated with an aggressive disease course. Haematologica 2016; 101:1553-1562. [PMID: 27443285 DOI: 10.3324/haematol.2015.139154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 07/12/2016] [Indexed: 01/07/2023] Open
Abstract
I In the last decade, the B-cell receptor has emerged as a pivotal stimulus in the pathogenesis of chronic lymphocytic leukemia, and a very feasible therapeutic target in this disease. B-cell receptor responsiveness in chronic lymphocytic leukemia cells is heterogeneous among patients and correlates with aggressiveness of the disease. Here we show, for the first time, that SLP76, a key scaffold protein in T-cell receptor signaling, is ectopically expressed in chronic lymphocytic leukemia cells, with variable levels among patients, and correlates positively with unmutated immunoglobulin heavy chain variable gene status and ZAP-70 expression. We found that SLP76 was functionally active in chronic lymphocytic leukemia cells. A SYK-dependent basal level of phosphorylated SLP76 exists in the cells, and upon B-cell receptor engagement, SLP76 tyrosine phosphorylation is significantly enhanced concomitantly with increased physical association with BTK. B-cell receptor-induced SLP76 phosphorylation is mediated by upstream signaling events involving LCK and SYK. Knockdown of SLP76 in the cells resulted in decreased induction of BTK, PLCγ2 and IκB phosphorylation, as well as cell viability after B-cell receptor activation with anti-IgM. Consistent with our biochemical findings, high total SLP76 expression in chronic lymphocytic leukemia cells correlated with a more aggressive disease course. IN CONCLUSION SLP76 is ectopically expressed in chronic lymphocytic leukemia cells where it plays a role in B-cell receptor signaling.
Collapse
Affiliation(s)
- Nili Dezorella
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Jerusalem, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Jerusalem, Israel
| | - Ben-Zion Katz
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Jerusalem, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Jerusalem, Israel
| | - Mika Shapiro
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Jerusalem, Israel
| | - Aaron Polliack
- Department of Hematology, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Chava Perry
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Jerusalem, Israel
| | - Yair Herishanu
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Jerusalem, Israel .,Sackler Faculty of Medicine, Tel-Aviv University, Jerusalem, Israel
| |
Collapse
|
8
|
Li K, Xiang X, Sun J, He HT, Wu J, Wang Y, Zhu C. Imaging Spatiotemporal Activities of ZAP-70 in Live T Cells Using a FRET-Based Biosensor. Ann Biomed Eng 2016; 44:3510-3521. [PMID: 27384937 DOI: 10.1007/s10439-016-1683-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/21/2016] [Indexed: 01/20/2023]
Abstract
The zeta-chain-associated protein kinase 70 kDa (ZAP-70), a member of the spleen tyrosine kinase (Syk) family, plays an essential role in early T cell receptor (TCR) signaling. Defects in ZAP-70 lead to impaired thymocyte development and peripheral T cell activation. To better understand its activation dynamics and regulation, we visualized ZAP-70 activities in single live T cells with a Förster resonance energy transfer (FRET)-based biosensor, which was designed for probing kinase activities of the Syk family. We observed in Jurkat E6.1 T cells rapid and specific FRET changes following anti-CD3 stimulation and subsequent piceatannol inhibition. The initiation of ZAP-70 activation was prompt (within 10 s) and correlates with the accompanied intracellular calcium elevation, as revealed by simultaneous imaging of the biosensor and calcium. Different from the previously reported ZAP-70 activation in the immunological synapse and the opposite pole (anti-synapse), we have observed rapid and sustained ZAP-70 activation only at the synapse with superantigen-pulsed Raji B cells. Furthermore, ZAP-70 signaling was impaired by cholesterol depletion, further supporting the importance of membrane organization in TCR signaling. Together our results provide a direct characterization of the spatiotemporal features of ZAP-70 activity in real time at subcellular levels.
Collapse
Affiliation(s)
- Kaitao Li
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Xue Xiang
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,School of Life Sciences, SUN YAT-SEN University, Guangzhou, China.,UnionPay Smart Co., Ltd, Shanghai, China
| | - Jie Sun
- Department of Bioengineering and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Hai-Tao He
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288, Marseille, France
| | - Jianhua Wu
- School of Life Sciences, SUN YAT-SEN University, Guangzhou, China.,School of Bioscience, South China University of Technology, Guangzhou, China
| | - Yingxiao Wang
- Department of Bioengineering and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA.,Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
9
|
The serine/threonine kinase Ndr2 controls integrin trafficking and integrin-dependent neurite growth. J Neurosci 2014; 34:5342-54. [PMID: 24719112 DOI: 10.1523/jneurosci.2728-13.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Integrins have been implicated in various processes of nervous system development, including proliferation, migration, and differentiation of neuronal cells. In this study, we show that the serine/threonine kinase Ndr2 controls integrin-dependent dendritic and axonal growth in mouse hippocampal neurons. We further demonstrate that Ndr2 is able to induce phosphorylation at the activity- and trafficking-relevant site Thr(788/789) of β1-integrin to stimulate the PKC- and CaMKII-dependent activation of β1-integrins, as well as their exocytosis. Accordingly, Ndr2 associates with integrin-positive early and recycling endosomes in primary hippocampal neurons and the surface expression of activated β1-integrins is reduced on dendrites of Ndr2-deficient neurons. The role of Ndr2 in dendritic differentiation is also evident in vivo, because Ndr2-null mutant mice show arbor-specific alterations of dendritic complexity in the hippocampus. This indicates a role of Ndr2 in the fine regulation of dendritic growth; in fact, treatment of primary neurons with Semaphorin 3A rescues Ndr2 knock-down-induced dendritic growth deficits but fails to enhance growth beyond control level. Correspondingly, Ndr2-null mutant mice show a Semaphorin 3A(-/-)-like phenotype of premature dendritic branching in the hippocampus. The results of this study show that Ndr2-mediated integrin trafficking and activation are crucial for neurite growth and guidance signals during neuronal development.
Collapse
|
10
|
Tomar N, De RK. A model of an integrated immune system pathway in Homo sapiens and its interaction with superantigen producing expression regulatory pathway in Staphylococcus aureus: comparing behavior of pathogen perturbed and unperturbed pathway. PLoS One 2013; 8:e80918. [PMID: 24324645 PMCID: PMC3855681 DOI: 10.1371/journal.pone.0080918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/17/2013] [Indexed: 11/19/2022] Open
Abstract
Response of an immune system to a pathogen attack depends on the balance between the host immune defense and the virulence of the pathogen. Investigation of molecular interactions between the proteins of a host and a pathogen helps in identifying the pathogenic proteins. It is necessary to understand the dynamics of a normally behaved host system to evaluate the capacity of its immune system upon pathogen attack. In this study, we have compared the behavior of an unperturbed and pathogen perturbed host system. Moreover, we have developed a formalism under Flux Balance Analysis (FBA) for the optimization of conflicting objective functions. We have constructed an integrated pathway system, which includes Staphylococcal Superantigen (SAg) expression regulatory pathway and TCR signaling pathway of Homo sapiens. We have implemented the method on this pathway system and observed the behavior of host signaling molecules upon pathogen attack. The entire study has been divided into six different cases, based on the perturbed/unperturbed conditions. In other words, we have investigated unperturbed and pathogen perturbed human TCR signaling pathway, with different combinations of optimization of concentrations of regulatory and signaling molecules. One of these cases has aimed at finding out whether minimization of the toxin production in a pathogen leads to the change in the concentration levels of the proteins coded by TCR signaling pathway genes in the infected host. Based on the computed results, we have hypothesized that the balance between TCR signaling inhibitory and stimulatory molecules can keep TCR signaling system into resting/stimulating state, depending upon the perturbation. The proposed integrated host-pathogen interaction pathway model has accurately reflected the experimental evidences, which we have used for validation purpose. The significance of this kind of investigation lies in revealing the susceptible interaction points that can take back the Staphylococcal Enterotoxin (SE)-challenged system within the range of normal behavior.
Collapse
Affiliation(s)
- Namrata Tomar
- Machine Intelligence Unit, Indian Statistical Institute, Kolkata, India
| | - Rajat K. De
- Machine Intelligence Unit, Indian Statistical Institute, Kolkata, India
| |
Collapse
|
11
|
Shim EK, Jung SH, Lee JR. Role of two adaptor molecules SLP-76 and LAT in the PI3K signaling pathway in activated T cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:2926-35. [PMID: 21282515 DOI: 10.4049/jimmunol.1001785] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previously, we identified p85, a subunit of PI3K, as one of the molecules that interacts with the N-terminal region of Src homology 2 domain-containing leukocyte protein of 76 kDa (SLP-76). We also demonstrated that tyrosine phosphorylation either at the 113 and/or 128 position is sufficient for the association of SLP-76 with the Src homology 2 domain near the N terminus of p85. The present study further examines the role of the association of these two molecules on the activation of PI3K signaling cascade. Experiments were done to determine the role of SLP-76, either wild-type, tyrosine mutants, or membrane-targeted forms of various SLP-76 constructs, on the membrane localization and phosphorylation of Akt, which is an event downstream of PI3K activation. Reconstitution studies with these various SLP-76 constructs in a Jurkat variant cell line that lacks SLP-76 or linker for activation of T cells (LAT) show that the activation of PI3K pathway following TCR ligation requires both SLP-76 and LAT adaptor proteins. The results suggest that SLP-76 associates with p85 after T cell activation and that LAT recruits this complex to the membrane, leading to Akt activation.
Collapse
Affiliation(s)
- Eun Kyung Shim
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | | | | |
Collapse
|
12
|
Hořejší V, Otáhal P, Brdička T. LAT - an important raft-associated transmembrane adaptor protein. Delivered on 6 July 2009 at the 34th FEBS Congress in Prague, Czech Republic. FEBS J 2010; 277:4383-97. [DOI: 10.1111/j.1742-4658.2010.07831.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Independent and cooperative roles of adaptor molecules in proximal signaling during FcepsilonRI-mediated mast cell activation. Mol Cell Biol 2010; 30:4188-96. [PMID: 20606011 DOI: 10.1128/mcb.00305-10] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Activation through FcepsilonRI, a high-affinity IgE-binding receptor, is critical for mast cell function during allergy. The formation of a multimolecular proximal signaling complex nucleated by the adaptor molecules SLP-76 and LAT1 is required for activation through this receptor. Based on previous T-cell studies, current dogma dictates that LAT1 is required for plasma membrane recruitment and function of SLP-76. Unexpectedly, we found that the recruitment and phosphorylation of SLP-76 were preserved in LAT1(-/-) mast cells and that SLP-76(-/-) and LAT1(-/-) mast cells harbored distinct functional and biochemical defects. The LAT1-like molecule LAT2 was responsible for the preserved membrane localization and phosphorylation of SLP-76 in LAT1(-/-) mast cells. Although LAT2 supported SLP-76 phosphorylation and recruitment to the plasma membrane, LAT2 only partially compensated for LAT1-mediated cell signaling due to its decreased ability to stabilize interactions with phospholipase Cgamma (PLCgamma). Comparison of SLP-76(-/-) LAT1(-/-) and SLP-76(-/-) mast cells revealed that some functions of LAT1 could occur independently of SLP-76. We propose that while SLP-76 and LAT1 depend on each other for many of their functions, LAT2/SLP-76 interactions and SLP-76-independent LAT1 functions also mediate a positive signaling pathway downstream of FcepsilonRI in mast cells.
Collapse
|
14
|
Kambayashi T, Larosa DF, Silverman MA, Koretzky GA. Cooperation of adapter molecules in proximal signaling cascades during allergic inflammation. Immunol Rev 2010; 232:99-114. [PMID: 19909359 DOI: 10.1111/j.1600-065x.2009.00825.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Activation of mast cells through their high-affinity immunoglobulin E receptor (FcepsilonRI) plays an important role in allergic disorders. Other mast cell-activating stimuli, such as Toll-like receptor (TLR) ligands, synergize with FcepsilonRI to enhance allergic inflammation. Thus, there is much interest in understanding how signaling occurs downstream of these receptors. One key event for FcepsilonRI-mediated mast cell activation is the inducible formation of multimolecular proximal signaling complexes. These complexes are nucleated by adapter proteins, scaffolds that localize various signaling molecules through their multiple molecule-binding domains. Here we review recent findings in proximal signaling cascades with an emphasis on how adapter molecules cooperate with each other to generate an optimal signal in mast cells, and we discuss how signals crosstalk between FcepsilonRI and TLRs in enhancing mast cell activation. Deciphering the molecular mechanisms leading to mast cell activation will hopefully bring new ideas for the development of novel therapeutics to control allergic diseases.
Collapse
Affiliation(s)
- Taku Kambayashi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
15
|
Spatiotemporal control of cyclic AMP immunomodulation through the PKA-Csk inhibitory pathway is achieved by anchoring to an Ezrin-EBP50-PAG scaffold in effector T cells. FEBS Lett 2010; 584:2681-8. [PMID: 20420835 DOI: 10.1016/j.febslet.2010.04.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Revised: 04/16/2010] [Accepted: 04/20/2010] [Indexed: 11/23/2022]
Abstract
A variety of immunoregulatory signals to effector T cells from monocytes, macrophages and regulatory T cells act through cyclic adenosine monophosphate. In the effector T cell, the protein kinase A (PKA) type I isoenzyme localizes to lipid rafts during T cell activation and modulates directly the proximal events that take place after engagement of the T cell receptor. The most proximal target for PKA phosphorylation is C-terminal Src kinase (Csk), which initiates a negative signal pathway that fine-tunes the T cell activation process. The A kinase anchoring protein Ezrin colocalizes PKA and Csk by forming a supramolecular signaling complex consisting of PKA, Ezrin, Ezrin/radixin/moesin (ERM) binding protein of 50 kDa (EBP50), phosphoprotein associated with glycosphingolipid-enriched membrane microdomains (GEMs) (PAG) and Csk.
Collapse
|
16
|
Mislocalization of SLP-76 leads to aberrant inflammatory cytokine and autoantibody production. Blood 2009; 115:2186-95. [PMID: 20029045 DOI: 10.1182/blood-2009-08-237438] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Central and peripheral tolerance is required to prevent immune responses to self-antigens. We now present a mouse model in which wild-type (WT) SH2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) has been constitutively targeted to the membrane, where CD4+ T cells become spontaneously dysregulated and develop an inflammatory phenotype. Mice bearing membrane-targeted SLP-76 (MTS) have a partial T-cell lymphopenia and impaired signaling though the mature T-cell receptor. The CD4+ T cells that develop in these mice possess an activated-like phenotype and are skewed toward the inflammatory T(H)1 and T(H)17 lineages. MTS mice also spontaneously develop autoantibodies at an early age. To rule out abnormal thymic selection as the sole cause of the MTS phenotype, we expressed WT SLP-76 along with the MTS followed by deletion of the WT allele in peripheral T cells. The peripheral MTS-expressing T cells demonstrate skewed cytokine responses when transferred into lymphopenic hosts. Thus, the abnormal effector T-cell phenotype still occurs in the presence of preserved central and peripheral tolerance, suggesting that diminished T-cell receptor signaling can promote skewed T-cell responses.
Collapse
|
17
|
Koretzky GA. The Role of SH2 Domain-containing Leukocyte Phosphoprotein of 76 kDa in the Regulation of Immune Cell Development and Function. Immune Netw 2009; 9:75-83. [PMID: 20107536 PMCID: PMC2803302 DOI: 10.4110/in.2009.9.3.75] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 05/14/2009] [Indexed: 01/14/2023] Open
Abstract
Recent years have seen an explosion of new knowledge defining the molecular events that are critical for development and activation of immune cells. Much of this new information has come from a careful molecular dissection of key signal transduction pathways that are initiated when immune cell receptors are engaged. In addition to the receptors themselves and critical effector molecules, these signaling pathways depend on adapters, proteins that have no intrinsic effector function but serve instead as scaffolds to nucleate multimolecular complexes. This review summarizes some of what has been learned about one such adapter protein, SH2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76), and how it regulates and integrates signals after engagement of immunoreceptors and integrins on various immune cell lineages.
Collapse
Affiliation(s)
- Gary A Koretzky
- Department of Pathology and Laboratory Medicine and Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| |
Collapse
|
18
|
Bezman NA, Baker RG, Lenox LE, Jordan MS, Koretzky GA. Cutting edge: rescue of pre-TCR but not mature TCR signaling in mice expressing membrane-targeted SLP-76. THE JOURNAL OF IMMUNOLOGY 2009; 182:5183-7. [PMID: 19380761 DOI: 10.4049/jimmunol.0802176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
SLP-76 (Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa) organizes signaling from immunoreceptors, including the platelet collagen receptor, the pre-TCR, and the TCR, and is required for T cell development. In this study we examine a mouse in which wild-type SLP-76 is replaced with a mutant constitutively targeted to the cell membrane. Membrane-targeted SLP-76 (MTS) supports ITAM signaling in platelets and from the pre-TCR. Signaling from the mature TCR, however, is defective in MTS thymocytes, resulting in failed T cell differentiation. Defective thymic selection by MTS is not rescued by a SLP-76 mutant whose localization is restricted to the cytosol. Thus, fixed localization of SLP-76 reveals differential requirements for the subcellular localization of signaling complexes downstream of the pre-TCR vs mature TCR.
Collapse
Affiliation(s)
- Natalie A Bezman
- Abramson Family Cancer Research Institute, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
19
|
Park I, Yun Y. Transmembrane adaptor proteins positively regulating the activation of lymphocytes. Immune Netw 2009; 9:53-7. [PMID: 20107544 PMCID: PMC2803307 DOI: 10.4110/in.2009.9.2.53] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 04/03/2009] [Indexed: 12/25/2022] Open
Abstract
Engagement of the immunoreceptors initiates signaling cascades resulting in lymphocyte activation and differentiation to effector cells, which are essential for the elimination of pathogens from the body. For the transduction of these immunoreceptor-mediated signals, several linker proteins termed transmembrane adaptor proteins (TRAPs) were shown to be required. TRAPs serve as platforms for the assembly and membrane targeting of the specific signaling proteins. Among seven TRAPs identified so far, LAT and LIME were shown to act as a positive regulator in TCR-mediated signaling pathways. In this review, we will discuss the functions of LAT and LIME in modulating T cell development, activation and differentiation.
Collapse
Affiliation(s)
- Inyoung Park
- Department of Life Science, Ewha Womans' University, Seoul 120-750, Korea
| | | |
Collapse
|
20
|
Abstract
Tyrosine phosphorylation and dephosphorylation of proteins play a critical role for many T-cell functions. The opposing actions of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) determine the level of tyrosine phosphorylation at any time. It is well accepted that PTKs are essential during T-cell signaling; however, the role and importance of PTPs are much less known and appreciated. Both transmembrane and cytoplasmic tyrosine phosphatases have been identified in T cells and shown to regulate T-cell responses. This review focuses on the roles of the two cytoplasmic PTPs, the Src-homology 2 domain (SH2)-containing SHP-1 and SHP-2, in T-cell signaling, development, differentiation, and function.
Collapse
Affiliation(s)
- Ulrike Lorenz
- Department of Microbiology and The Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908-0734, USA.
| |
Collapse
|
21
|
Targeted cleavage of signaling proteins by caspase 3 inhibits T cell receptor signaling in anergic T cells. Immunity 2008; 29:193-204. [PMID: 18701083 DOI: 10.1016/j.immuni.2008.06.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2007] [Revised: 03/26/2008] [Accepted: 06/10/2008] [Indexed: 11/22/2022]
Abstract
T cell receptor (TCR) engagement in the absence of costimulation induces the calcium-dependent upregulation of a program of gene expression that leads to the establishment of T cell anergy. Casp3 is one of the genes activated during anergy induction. Here we show that caspase 3 is required for the induction of T cell unresponsiveness. Suboptimal T cell stimulation induced caspase 3 activation, which did not result in cell death. Furthermore, caspase 3-deficient T cells showed impaired responses to anergizing stimuli. In anergic T cells, activated caspase 3 associated to the plasma membrane, where it cleaved and inactivated proteins such as the Grb2-related adaptor downstream of shc (GADS) and the guanine-nucleotide exchange factor Vav1, causing a blockade in TCR signaling. Our results identify a role for caspase 3 in nonapoptotic T cells and support that caspase 3-dependent proteolytic inactivation of signaling proteins is essential to maintain T cell tolerance.
Collapse
|
22
|
Kabouridis PS, Jury EC. Lipid rafts and T-lymphocyte function: implications for autoimmunity. FEBS Lett 2008; 582:3711-8. [PMID: 18930053 PMCID: PMC2596348 DOI: 10.1016/j.febslet.2008.10.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 10/06/2008] [Accepted: 10/07/2008] [Indexed: 12/11/2022]
Abstract
Experimental evidence indicates that the mammalian cell membrane is compartmentalized. A structural feature that supports membrane segmentation implicates assemblies of selected lipids broadly referred to as lipid rafts. In T-lymphocytes, lipid rafts are implicated in signalling from the T-cell antigen receptor (TCR) and in localization and function of proteins residing proximal to the receptor. This review summarizes the current literature that deals with lipid raft involvement in T-cell activation and places particular emphasis in recent studies investigating lipid rafts in autoimmunity. The potential of lipid rafts as targets for the development of a new class of immune-modulating compounds is discussed.
Collapse
Affiliation(s)
- Panagiotis S Kabouridis
- William Harvey Research Institute, Queen Mary's School of Medicine & Dentistry, University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | | |
Collapse
|
23
|
Abstract
Although dynamic imaging technologies have provided important insights into the underlying processes responsible for T-cell activation, the processes that link antigen recognition to downstream signaling remain poorly defined. Converging lines of inquiry indicate that T-cell receptor (TCR) microclusters are the minimal structures capable of directing effective TCR signaling. Furthermore, imaging studies have determined that these structures trigger the assembly of oligomeric signaling scaffolds that contain the adapters and effectors required for T-cell activation. Existing models of T-cell activation accurately explain the sensitivity and selectivity of antigen recognition. However, these models do not account for important properties of microclusters, including their peripheral formation, size, and movement on the actin cytoskeleton. Here we examine how lipid rafts, galectin lattices, and protein scaffolds contribute to the assembly, function, and fate of TCR microclusters within immune synapses. Finally, we propose a 'mechanical segregation' model of signal initiation in which cytoskeletal forces contribute to the lateral segregation of molecules and cytoskeletal scaffolds provide a template for microclusters assembly.
Collapse
|
24
|
|
25
|
Jordan MS, Maltzman JS, Kliche S, Shabason J, Smith JE, Obstfeld A, Schraven B, Koretzky GA. In vivo disruption of T cell development by expression of a dominant-negative polypeptide designed to abolish the SLP-76/Gads interaction. Eur J Immunol 2007; 37:2961-72. [PMID: 17823979 DOI: 10.1002/eji.200636855] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Multi-molecular complexes nucleated by adaptor proteins play a central role in signal transduction. In T cells, one central axis consists of the assembly of several signaling proteins linked together by the adaptors linker of activated T cells (LAT), Src homology 2 domain-containing leukocyte-specific phosphoprotein of 76 kDa (SLP-76), and Grb2-related adaptor downstream of Shc (Gads). Each of these adaptors has been shown to be important for normal T cell development, and their proper sub-cellular localization is critical for optimal function in cell lines. We previously demonstrated in Jurkat T cells and a rat basophilic leukemic cell line that expression of a 50-amino acid polypeptide identical to the site on SLP-76 that binds to Gads blocks proper localization of SLP-76 and SLP-76-dependent signaling events. Here we extend these studies to investigate the ability of this polypeptide to inhibit TCR-induced integrin activity in Jurkat cells and to inhibit in vivo thymocyte development and primary T cell function. These data provide evidence for the in vivo function of a dominant-negative peptide based upon the biology of SLP-76 action and suggest the possibility of therapeutic potential of targeting the SLP-76/Gads interaction.
Collapse
Affiliation(s)
- Martha S Jordan
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Adapters are multidomain molecules that recruit effector proteins during signal transduction by immunoreceptors and integrins. The absence of these scaffolding molecules profoundly affects development and function of various hematopoietic lineages, underscoring their importance as regulators of signaling cascades. An emerging aspect of the mechanism by which engaged immunoreceptors and integrins transmit signals within the cell is by differential usage of various adapters that function to nucleate formation of distinct signaling complexes in a specific location within the cell. In this review, we discuss the mechanisms by which adapter proteins coordinate signal transduction with an emphasis on the role of subcellular compartmentalization in adapter function.
Collapse
Affiliation(s)
- Natalie Bezman
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
27
|
Abstract
Integrin adhesion receptors are critical for antigen recognition by T cells and for regulated recirculation and trafficking into and through various tissues in the body. T-cell receptor (TCR) signaling induces rapid increases in integrin function that facilitate T-cell activation by promoting stable contact with antigen-presenting cells and extracellular proteins in the environment. In this review, we outline the molecular mechanisms by which the TCR signals to integrins and present a model that highlights four key events: (i) initiation of proximal TCR signals nucleated by the linker for activated T cells (LAT) adapter protein and involving Itk, phospholipase C-gamma1, Vav1, and Src homology 2 domain-containing leukocyte-specific phosphoprotein of 76 kDa; (ii) transmission of integrin activation signals from the LAT signalosome to integrins by protein kinase (PK) C and the adapter protein, adhesion and degranulation-promoting adapter protein; (iii) assembly of integrin-associated signaling complexes that include PKD, the guanosine triphosphatase Rap1 and its effectors, and talin; and (iv) reorganization of the actin cytoskeleton by WAVE2 and other actin-remodeling proteins. These events coordinate changes in integrin conformation and clustering that result in enhanced integrin functional activity following TCR stimulation.
Collapse
Affiliation(s)
- Brandon J Burbach
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
28
|
Sankarshanan M, Ma Z, Iype T, Lorenz U. Identification of a novel lipid raft-targeting motif in Src homology 2-containing phosphatase 1. THE JOURNAL OF IMMUNOLOGY 2007; 179:483-90. [PMID: 17579069 DOI: 10.4049/jimmunol.179.1.483] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The tyrosine phosphatase Src homology 2-containing phosphatase 1 (SHP-1) is a key negative regulator of TCR-mediated signaling. Previous studies have shown that in T cells a fraction of SHP-1 constitutively localizes to membrane microdomains, commonly referred to as lipid rafts. Although this localization of SHP-1 is required for its functional regulation of T cell activation events, how SHP-1 is targeted to the lipid rafts was unclear. In this study, we identify a novel, six-amino acid, lipid raft-targeting motif within the C terminus of SHP-1 based on several biochemical and functional observations. First, mutations of this motif in the context of full-length SHP-1 result in the loss of lipid raft localization of SHP-1. Second, this motif alone restores raft localization when fused to a mutant of SHP-1 (SHP-1 DeltaC) that fails to localize to rafts. Third, a peptide encompassing the 6-mer motif directly binds to phospholipids whereas a mutation of this motif abolishes lipid binding. Fourth, whereas full-length SHP-1 potently inhibits TCR-induced tyrosine phosphorylation of specific proteins, expression of a SHP-1-carrying mutation within the 6-mer motif does not. Additionally, although SHP-1 DeltaC was functionally inactive, the addition of the 6-mer motif restored its functionality in inhibiting TCR-induced tyrosine phosphorylation. Finally, this 6-mer mediated targeting of SHP-1 lipid rafts was essential for the function of this phosphatase in regulating IL-2 production downstream of TCR. Taken together, these data define a novel 6-mer motif within SHP-1 that is necessary and sufficient for lipid raft localization and for the function of SHP-1 as a negative regulator of TCR signaling.
Collapse
Affiliation(s)
- Mohan Sankarshanan
- Department of Microbiology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
29
|
Beach D, Gonen R, Bogin Y, Reischl IG, Yablonski D. Dual role of SLP-76 in mediating T cell receptor-induced activation of phospholipase C-gamma1. J Biol Chem 2006; 282:2937-46. [PMID: 17148460 DOI: 10.1074/jbc.m606697200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Phospholipase C-gamma1 (PLC-gamma1) activation depends on a heterotrimeric complex of adaptor proteins composed of LAT, Gads, and SLP-76. Upon T cell receptor stimulation, a portion of PLC-gamma1 is recruited to a detergent-resistant membrane fraction known as the glycosphingolipid-enriched membrane microdomains (GEMs), or lipid rafts, to which LAT is constitutively localized. In addition to LAT, PLC-gamma1 GEM recruitment depended on SLP-76, and, in particular, required the Gads-binding domain of SLP-76. The N-terminal tyrosine phosphorylation sites and P-I region of SLP-76 were not required for PLC-gamma1 GEM recruitment, but were required for PLC-gamma1 phosphorylation at Tyr(783). Thus, GEM recruitment can be insufficient for full activation of PLC-gamma1 in the absence of a second SLP-76-mediated event. Indeed, a GEM-targeted derivative of PLC-gamma1 depended on SLP-76 for T cell receptor-induced phosphorylation at Tyr783 and subsequent NFAT activation. On a biochemical level, SLP-76 inducibly associated with both Vav and catalytically active ITK, which efficiently phosphorylated a PLC-gamma1 fragment at Tyr783 in vitro. Both associations were disrupted upon mutation of the N-terminal tyrosine phosphorylation sites of SLP-76. The P-I region deletion disrupted Vav association and reduced SLP-76-associated kinase activity. A smaller deletion within the P-I region, which does not impair PLC-gamma1 activation, did not impair the association with Vav, but reduced SLP-76-associated kinase activity. These results provide new insight into the multiple roles of SLP-76 and the functional importance of its interactions with other signaling proteins.
Collapse
Affiliation(s)
- Dvora Beach
- Rappaport Family Institute for Research in the Medical Sciences, Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | | | | | | | | |
Collapse
|
30
|
Bunnell SC, Singer AL, Hong DI, Jacque BH, Jordan MS, Seminario MC, Barr VA, Koretzky GA, Samelson LE. Persistence of cooperatively stabilized signaling clusters drives T-cell activation. Mol Cell Biol 2006; 26:7155-66. [PMID: 16980618 PMCID: PMC1592897 DOI: 10.1128/mcb.00507-06] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antigen recognition triggers the recruitment of the critical adaptor protein SLP-76 to small macromolecular clusters nucleated by the T-cell receptor (TCR). These structures develop rapidly, in parallel with TCR-induced increases in tyrosine phosphorylation and cytosolic calcium, and are likely to contribute to TCR-proximal signaling. Previously, we demonstrated that these SLP-76-containing clusters segregate from the TCR and move towards the center of the contact interface. Neither the function of these clusters nor the structural requirements governing their persistence have been examined extensively. Here we demonstrate that defects in cluster assembly and persistence are associated with defects in T-cell activation in the absence of Lck, ZAP-70, or LAT. Clusters persist normally in the absence of phospholipase C-gamma1, indicating that in the absence of a critical effector, these structures are insufficient to drive T-cell activation. Furthermore, we show that the critical adaptors LAT and Gads localize with SLP-76 in persistent clusters. Mutational analyses of LAT, Gads, and SLP-76 indicated that multiple domains within each of these proteins contribute to cluster persistence. These data indicate that multivalent cooperative interactions stabilize these persistent signaling clusters, which may correspond to the functional complexes predicted by kinetic proofreading models of T-cell activation.
Collapse
Affiliation(s)
- Stephen C Bunnell
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kliche S, Breitling D, Togni M, Pusch R, Heuer K, Wang X, Freund C, Kasirer-Friede A, Menasche G, Koretzky GA, Schraven B. The ADAP/SKAP55 signaling module regulates T-cell receptor-mediated integrin activation through plasma membrane targeting of Rap1. Mol Cell Biol 2006; 26:7130-44. [PMID: 16980616 PMCID: PMC1592884 DOI: 10.1128/mcb.00331-06] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Adhesion of T cells after stimulation of the T-cell receptor (TCR) is mediated via signaling processes that have collectively been termed inside-out signaling. The molecular basis for inside-out signaling is not yet completely understood. Here, we show that a signaling module comprising the cytosolic adapter proteins ADAP and SKAP55 is involved in TCR-mediated inside-out signaling and, moreover, that the interaction between ADAP and SKAP55 is mandatory for integrin activation. Disruption of the ADAP/SKAP55 module leads to displacement of the small GTPase Rap1 from the plasma membrane without influencing its GTPase activity. These findings suggest that the ADAP/SKAP55 complex serves to recruit activated Rap1 to the plasma membrane. In line with this hypothesis is the finding that membrane targeting of the ADAP/SKAP55 module induces T-cell adhesion in the absence of TCR-mediated stimuli. However, it appears as if the ADAP/SKAP55 module can exert its signaling function outside of the classical raft fraction of the cell membrane.
Collapse
Affiliation(s)
- Stefanie Kliche
- Institute of Immunology, Otto von Guericke University, 39120 Magdeburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abtahian F, Bezman N, Clemens R, Sebzda E, Cheng L, Shattil SJ, Kahn ML, Koretzky GA. Evidence for the requirement of ITAM domains but not SLP-76/Gads interaction for integrin signaling in hematopoietic cells. Mol Cell Biol 2006; 26:6936-49. [PMID: 16943434 PMCID: PMC1592869 DOI: 10.1128/mcb.01040-06] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Syk tyrosine kinase and Src homology 2 (SH2) domain-containing leukocyte-specific phosphoprotein of 76 kDa (SLP-76) are signaling mediators activated downstream of immunoreceptor tyrosine-based activation motif (ITAM)-containing immunoreceptors and integrins. While the signaling cascades descending from integrins are similar to immunoreceptors, the mechanism of Syk activation and SLP-76 recruitment remains unclear. We used an in vivo structure-function approach to study the requirements for the domains of Syk and SLP-76 in immunoreceptor and integrin signaling. We found that both SH2 domains and the kinase domain of Syk are required for immunoreceptor-dependent signaling and cellular response via integrins. While the Gads-binding domain of SLP-76 is needed for immunoreceptor signaling, it appears dispensable for integrin signaling. Syk and SLP-76 also are required for initiating and/or maintaining separation between the blood and lymphatic vasculature. Therefore, we correlated the signaling requirement of the various domains of Syk and SLP-76 to their requirement in regulating vascular separation. Our data suggest ITAMs are required in Syk-dependent integrin signaling, demonstrate the separation of the structural features of SLP-76 to selectively support immunoreceptor versus integrin signaling, and provide evidence that the essential domains of SLP-76 for ITAM signals are those which most efficiently support separation between lymphatic and blood vessels.
Collapse
Affiliation(s)
- Farhad Abtahian
- Signal Transduction Program, Abramson Family Cancer Research Institute, University of Pennsylvania, 421 Curie Blvd., Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Watanabe R, Harada Y, Takeda K, Takahashi J, Ohnuki K, Ogawa S, Ohgai D, Kaibara N, Koiwai O, Tanabe K, Toma H, Sugamura K, Abe R. Grb2 and Gads exhibit different interactions with CD28 and play distinct roles in CD28-mediated costimulation. THE JOURNAL OF IMMUNOLOGY 2006; 177:1085-91. [PMID: 16818765 DOI: 10.4049/jimmunol.177.2.1085] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although both CD28 and ICOS bind PI3K and provide stimulatory signal for T cell activation, unlike CD28, ICOS does not costimulate IL-2 secretion. CD28 binds both PI3K and Grb2, whereas ICOS binds only PI3K. We have generated an ICOS mutant, which can bind Grb2 by replacement of its PI3K binding motif YMFM with the CD28 YMNM motif, and shown that it induces significant activation of the IL-2 promoter. However, this mutant ICOS was insufficient to activate the NF-kappaB pathway. In this study, we show that Gads, but not Grb2, is essential for CD28-mediated NF-kappaB activation, and its binding to CD28 requires the whole CD28 cytoplasmic domain in addition to the YMNM motif. Mutagenesis experiments have indicated that mutations in the N-terminal and/or C-terminal PXXP motif(s) of CD28 significantly reduce their association with Gads, whereas their associations with Grb2 are maintained. They induced strong activity of the NFAT/AP-1 reporter comparable with the CD28 wild type, but weak activity of the NF-kappaB reporter. Grb2- and Gads-dominant-negative mutants had a strong effect on NFAT/AP-1 reporter, but only Gads-dominant-negative significantly inhibited NF-kappaB reporter. Our data suggest that, in addition to the PI3K binding motif, the PXXP motif in the CD28 cytoplasmic domain may also define a functional difference between the CD28- and ICOS-mediated costimulatory signals by binding to Gads.
Collapse
Affiliation(s)
- Ryosuke Watanabe
- Research Institute for Biological Sciences, Faculty of Science and Technology, Tokyo University of Science, 1669 Yamazaki, Noda, Chiba 278-0022, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Barr VA, Balagopalan L, Barda-Saad M, Polishchuk R, Boukari H, Bunnell SC, Bernot KM, Toda Y, Nossal R, Samelson LE. T-Cell Antigen Receptor-Induced Signaling Complexes: Internalization Via a Cholesterol-Dependent Endocytic Pathway. Traffic 2006; 7:1143-62. [PMID: 16919152 DOI: 10.1111/j.1600-0854.2006.00464.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
T-cell antigen receptor engagement causes the rapid assembly of signaling complexes. The adapter protein SLP-76, detected as SLP-yellow fluorescent protein, initially clustered with the TCR and other proteins, then translocated medially on microtubules. As shown by total internal reflection fluorescence microscopy and the inhibition of SLP-76 movement at 16 degrees C, this movement required endocytosis. Immunoelectron microscopy showed SLP-76 staining of smooth pits and tubules. Cholesterol depletion decreased the movement of SLP-76 clusters, as did coexpression of the ubiquitin-interacting motif domain from eps15. These data are consistent with the internalization of SLP-76 via a lipid raft-dependent pathway that requires interaction of the endocytic machinery with ubiquitinylated proteins. The endocytosed SLP-76 clusters contained phosphorylated SLP-76 and phosphorylated LAT. The raft-associated, transmembrane protein LAT likely targets SLP-76 to endocytic vesicles. The endocytosis of active SLP-76 and LAT complexes suggests a possible mechanism for downregulation of signaling complexes induced by TCR activation.
Collapse
Affiliation(s)
- Valarie A Barr
- Laboratory of Cellular and Molecular Biology, Department of Health and Human Services, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Silverman MA, Shoag J, Wu J, Koretzky GA. Disruption of SLP-76 interaction with Gads inhibits dynamic clustering of SLP-76 and FcepsilonRI signaling in mast cells. Mol Cell Biol 2006; 26:1826-38. [PMID: 16479002 PMCID: PMC1430252 DOI: 10.1128/mcb.26.5.1826-1838.2006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We developed a confocal real-time imaging approach that allows direct observation of the subcellular localization pattern of proteins involved in proximal FcepsilonRI signaling in RBL cells and primary bone marrow-derived mast cells. The adaptor protein Src homology 2 (SH2) domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) is critical for FcepsilonRI-induced calcium flux, degranulation, and cytokine secretion. In this study, we imaged SLP-76 and found it in the cytosol of unstimulated cells. Upon FcepsilonRI cross-linking, SLP-76 translocates to the cell membrane, forming clusters that colocalize with the FcepsilonRI, the tyrosine kinase Syk, the adaptor LAT, and phosphotyrosine. The disruption of the SLP-76 interaction with its constitutive binding partner, Gads, through the mutation of SLP-76 or the expression of the Gads-binding region of SLP-76, inhibits the translocation and clustering of SLP-76, suggesting that the interaction of SLP-76 with Gads is critical for appropriate subcellular localization of SLP-76. We further demonstrated that the expression of the Gads-binding region of SLP-76 in bone marrow-derived mast cells inhibits FcepsilonRI-induced calcium flux, degranulation, and cytokine secretion. These studies revealed, for the first time, that SLP-76 forms signaling clusters following FcepsilonRI stimulation and demonstrated that the Gads-binding region of SLP-76 regulates clustering of SLP-76 and FcepsilonRI-induced mast cell responses.
Collapse
Affiliation(s)
- Michael A Silverman
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
36
|
Koretzky GA, Abtahian F, Silverman MA. SLP76 and SLP65: complex regulation of signalling in lymphocytes and beyond. Nat Rev Immunol 2006; 6:67-78. [PMID: 16493428 DOI: 10.1038/nri1750] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SLP76 and SLP65 are adaptor proteins that lack intrinsic enzymatic activity but contain multiple protein-binding domains. These proteins are essential for signalling downstream of integrins and receptors that contain immunoreceptor tyrosine-based activation motifs. The absence of these adaptor proteins profoundly affects various lineages in the haematopoietic compartment and severely compromises vascular development, highlighting their importance as regulators of signalling cascades. In this Review, we discuss the role of SLP76 and SLP65 in several signalling pathways in haematopoietic cells, with an emphasis on recent studies that provide insight into their mechanisms of action.
Collapse
Affiliation(s)
- Gary A Koretzky
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania, 415 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
37
|
Karnell FG, Monroe JG. The Role of Membrane Lipids in the Regulation of Immune Cell Activity. Transfus Med Hemother 2006. [DOI: 10.1159/000090192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
38
|
Abstract
The molecular events and the protein components that are involved in signalling by the T cell receptor (TCR) for antigen have been extensively studied. Activation of signalling cascades following TCR stimulation depends on the phosphorylation of the receptor by the tyrosine kinase Lck, which localizes to the cytoplasmic face of the plasma membrane by virtue of its post-translational modification. However, the precise order of events during TCR phosphorylation at the plasma membrane, remains to be defined. A current theory that describes early signalling events incorporates the function of lipid rafts, microdomains at the plasma membrane with distinct lipid and protein composition. Lipid rafts have been implicated in diverse biological functions in mammalian cells. In T cells, molecules with a key role in TCR signalling, including Lck, localize to these domains. Importantly, mutant versions of these proteins which fail to localise to raft domains were unable to support signalling by the TCR. Biochemical studies using purified detergent-resistant membranes (DRM) and confocal microscopy have suggested that upon stimulation, the TCR and Lck-containing lipid rafts may come into proximity allowing phosphorylation of the receptor. Further, there are data suggesting that phosphorylation of the TCR could depend on a transient increase in Lck activity that takes place within lipid rafts to initiate signalling. Current results and a model of how lipid rafts may regulate TCR signalling are discussed.
Collapse
Affiliation(s)
- Panagiotis S Kabouridis
- Bone & Joint Research Unit, William Harvey Research Institute, Queen Mary's School of Medicine & Dentistry, University of London, UK.
| |
Collapse
|
39
|
Yamasaki S, Ishikawa E, Sakuma M, Ogata K, Sakata-Sogawa K, Hiroshima M, Wiest DL, Tokunaga M, Saito T. Mechanistic basis of pre–T cell receptor–mediated autonomous signaling critical for thymocyte development. Nat Immunol 2005; 7:67-75. [PMID: 16327787 DOI: 10.1038/ni1290] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Accepted: 10/13/2005] [Indexed: 01/08/2023]
Abstract
The pre-T cell receptor (TCR) is crucial for early T cell development and is proposed to function in a ligand-independent way. However, the molecular mechanism underlying the autonomous signals remains elusive. Here we show that the pre-TCR complex spontaneously formed oligomers. Specific charged residues in the extracellular domain of the pre-TCR alpha-chain mediated formation of the oligomers in vitro. Alteration of these residues eliminated the ability of the pre-TCR alpha-chain to support pre-TCR signaling in vivo. Dimerization but not raft localization of CD3epsilon was sufficient to simulate pre-TCR function and promote beta-selection. These results suggest that the pre-TCR complex can deliver its signal autonomously through oligomerization of the pre-TCR alpha-chain mediated by charged residues.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cell Differentiation/immunology
- Hematopoietic Stem Cells/cytology
- Humans
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Microscopy, Confocal
- Molecular Sequence Data
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- T-Lymphocytes/cytology
Collapse
Affiliation(s)
- Sho Yamasaki
- Laboratory for Cell Signaling, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ziemba SE, Mattingly RR, McCabe MJ, Rosenspire AJ. Inorganic mercury inhibits the activation of LAT in T-cell receptor-mediated signal transduction. Toxicol Sci 2005; 89:145-53. [PMID: 16251484 DOI: 10.1093/toxsci/kfj029] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Little is known as to the molecular mechanisms involved with mercury intoxication at very low levels. Although the mechanism is not known, animal studies have nevertheless shown that low levels of mercury may target the immune system. Inorganic mercury (Hg2+) at very low (but non-toxic) levels can disrupt immune system homeostasis, in that genetically susceptible rodents develop idiosyncratic autoimmune disease, which is associated with defective T-cell function. T lymphocyte function is intimately coupled to the T-cell receptor. We have previously reported that on a molecular level, low concentrations of Hg2+ disrupt signaling from the T-cell receptor by interfering with activation of Ras and ERK MAP kinase. In this report we expand upon those results by showing that in T lymphocytes exposed to low concentration of Hg2+, Ras fails to become properly activated because upstream of Ras in the T cell signal transduction pathway, the important scaffolding element Linker for Activation of T Cells (LAT) fails to become properly phosphorylated. Hypo-phosphorylation of LAT occurs, because upstream of LAT, the LAT reactive tyrosine kinase ZAP-70 is also not properly activated in Hg2+ treated cells.
Collapse
Affiliation(s)
- Stamatina E Ziemba
- Department of Immunology & Microbiology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | |
Collapse
|
41
|
Garrett-Sinha LA, Hou P, Wang D, Grabiner B, Araujo E, Rao S, Yun TJ, Clark EA, Simon MC, Clark MR. Spi-1 and Spi-B control the expression of the Grap2 gene in B cells. Gene 2005; 353:134-46. [PMID: 15936902 DOI: 10.1016/j.gene.2005.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2004] [Revised: 03/21/2005] [Accepted: 04/07/2005] [Indexed: 11/16/2022]
Abstract
The Ets family members Spi-1 and Spi-B have been implicated in the regulation of genes important for B cell antigen receptor (BCR) signaling. Mice deficient in Spi-B exhibit reduced B cell proliferation in response to BCR cross-linking and impaired T cell-dependent immune responses. This defect is exacerbated in the presence of Spi-1 haplo-insufficiency (Spi1+/- SpiB-/-). Tyrosine phosphorylation and calcium mobilization induced by BCR engagement is diminished in Spi1+/- SpiB-/- B lymphocytes, although many key BCR signaling proteins are expressed, suggesting that Spi-1 and Spi-B regulate expression of additional, unidentified signaling molecules. We now demonstrate that expression of the adaptor protein Grap2 is impaired in Spi1+/- SpiB+/- and Spi1+/- SpiB-/- B lymphocytes. Analysis of two alternate murine Grap2 promoters revealed a functionally important Spi-1 and Spi-B DNA binding element located in the downstream promoter. Ectopic expression of Grap2 in Grap2-deficient B cells reduced the recruitment of BLNK to Igalpha and the phosphorylation of specific substrates. Regulation of BLNK recruitment was dependent upon the Grap2 proline-rich domain, while modulation of phosphorylation was dependent upon both the proline-rich and SH2 domains. These data indicate that Spi-1 and Spi-B directly regulate the expression of Grap2 and that Grap2 functions to modulate BCR signaling, but that reduced Grap2 expression is unlikely to account for the BCR signaling defects observed in Spi1+/- SpiB-/- B cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- B-Lymphocytes/metabolism
- Base Sequence
- Binding Sites/genetics
- Blotting, Northern
- Blotting, Western
- Cell Line, Tumor
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Down-Regulation/genetics
- Electrophoretic Mobility Shift Assay
- Gene Expression/genetics
- Genotype
- Heterozygote
- Mice
- Mice, Knockout
- Molecular Sequence Data
- Mutation
- Oligonucleotides/genetics
- Oligonucleotides/metabolism
- Promoter Regions, Genetic/genetics
- Protein Binding
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Sequence Alignment
- Sequence Homology, Nucleic Acid
- Signal Transduction
- Spleen/cytology
- Spleen/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Lee Ann Garrett-Sinha
- Department of Biochemistry, State University of New York at Buffalo, 140 Farber Hall, 3435 Main Street, Buffalo, NY 14214, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tian W, Feng B, Liou HC. Silencing OCILRP2 leads to intrinsic defects in T cells in response to antigenic stimulation. Cell Immunol 2005; 235:72-84. [PMID: 16143319 DOI: 10.1016/j.cellimm.2005.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Revised: 07/01/2005] [Accepted: 07/14/2005] [Indexed: 10/25/2022]
Abstract
We have previously demonstrated that OCILRP2 interaction with its ligand NKRP1f provides a co-stimulatory signal for optimal T cell proliferation and IL-2 production. Here, using RNA interference technology, we will demonstrate that silencing OCILRP2 in vivo leads to intrinsic impairment in T cell response to CD3- and CD28-cross-linking as well as antigenic stimulation. OCILRP2-silenced T cells have reduced cell proliferation and IL-2 production, which can be bypassed by PMA and ionomycin treatment. OCILRP2-silenced T cells also failed to undergo TCR capping and had impaired cytoskeleton reorganization. Moreover, in OCILRP2-silenced T cells, tyrosine phosphorylation of Lck was diminished, while tyrosine phosphorylation of linkers for activation of T cells was unchanged. Interestingly, NF-kappaB activation was also impaired as the result of OCILRP2 silencing. Together, our data strongly support a novel role for OCILRP2 C-type lectin in TCR-mediated signal transduction. The observation that OCILRP2 is involved in TCR capping and cytoskeletal organization suggests that OCILRP2-NKRP1f may facilitate lipid rafts and immunological synapse formation during T cell interaction with antigen presenting cells.
Collapse
Affiliation(s)
- Wenzhi Tian
- Division of Immunology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | |
Collapse
|
43
|
Abstract
Adapter molecules are multidomain proteins lacking intrinsic catalytic activity, functioning instead by nucleating molecular complexes during signal transduction. The SLP-76 family of adapters includes SH2 domain-containing leukocyte phosphoprotein of 76kDa (SLP-76), B cell linker protein (BLNK), and cytokine-dependent hematopoietic cell linker (Clnk). These proteins are critical for integration of numerous signaling cascades downstream of immunotyrosine-based activation motif (ITAM)-bearing receptors and integrins in diverse hematopoietic cell types. Mutations in genes encoding SLP-76 family adapters result in severe phenotypes, underscoring the critical role these proteins play in cellular development and function by directing formation of signaling complexes in a temporally- and spatially-specific manner.
Collapse
Affiliation(s)
- Jennifer N Wu
- Department of Laboratory Medicine and Pathology, School of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania, 415 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | | |
Collapse
|
44
|
Abstract
Memory T cells exhibit low activation thresholds and mediate rapid effector responses when recalled by antigen; contrasting the higher activation threshold, slower responses and predominant IL-2 production by naive T cells. While the sequence of intracellular events coupling the T cell-receptor (TCR) to naive T cell activation is well characterized, biochemical control of memory T cell differentiation and function remains undefined. In this review, we will discuss recent developments in T cell-receptor signal transduction as they pertain to memory T cells, and will discuss how signal dampening may drive memory generation, and more efficient spatial organization of signaling molecules may promote rapid recall responses.
Collapse
Affiliation(s)
- Meena R Chandok
- Division of Transplantation, Department of Surgery, University of Maryland School of Medicine, MSTF Building, Room 400, 685 W. Baltimore St., Baltimore, MD 21201, USA
| | | |
Collapse
|
45
|
Fawcett VCJ, Lorenz U. Localization of Src homology 2 domain-containing phosphatase 1 (SHP-1) to lipid rafts in T lymphocytes: functional implications and a role for the SHP-1 carboxyl terminus. THE JOURNAL OF IMMUNOLOGY 2005; 174:2849-59. [PMID: 15728495 DOI: 10.4049/jimmunol.174.5.2849] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The protein tyrosine phosphatase Src homology 2 domain-containing phosphatase 1 (SHP-1) has previously been shown to be a negative regulator of signaling mediated via the TCR. A growing body of evidence indicates that the regulated localization of proteins within certain membrane subdomains, referred to as lipid rafts, is important for the successful transduction of signaling events downstream of the TCR. However, considerably less is known about the localization of negative regulators during these lipid raft-dependent signaling events. In this study we have investigated the subcellular localization of SHP-1 and its role in regulation of TCR-mediated signaling. Our studies demonstrate that in a murine T cell hybridoma as well as in primary murine thymocytes, a fraction of SHP-1 localizes to the lipid rafts, both basally and after TCR stimulation. Interestingly, although SHP-1 localized in the nonraft fractions is tyrosine phosphorylated, the SHP-1 isolated from the lipid rafts lacks the TCR-induced tyrosine phosphorylation, suggesting physical and/or functional differences between these two subpopulations. We identify a requirement for the C-terminal residues of SHP-1 in optimal localization to the lipid rafts. Although expression of SHP-1 that localizes to lipid rafts potently inhibits TCR-mediated early signaling events and IL-2 production, the expression of lipid raft-excluded SHP-1 mutants fails to elicit any of the inhibitory effects. Taken together these studies reveal a key role for lipid raft localization of SHP-1 in mediating the inhibitory effects on T cell signaling events.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Binding Sites, Antibody
- Cell Line
- Interleukin-2/antagonists & inhibitors
- Interleukin-2/biosynthesis
- Intracellular Signaling Peptides and Proteins
- Membrane Microdomains/enzymology
- Membrane Microdomains/genetics
- Membrane Microdomains/metabolism
- Mice
- Mice, Inbred C3H
- Mutagenesis, Site-Directed
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Peptide Fragments/physiology
- Phosphorylation
- Protein Phosphatase 1
- Protein Structure, Tertiary
- Protein Tyrosine Phosphatase, Non-Receptor Type 6
- Protein Tyrosine Phosphatases/biosynthesis
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/metabolism
- Protein Tyrosine Phosphatases/physiology
- Receptor-CD3 Complex, Antigen, T-Cell/antagonists & inhibitors
- Receptor-CD3 Complex, Antigen, T-Cell/physiology
- Sequence Deletion
- Signal Transduction/immunology
- Subcellular Fractions/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/enzymology
- Tyrosine/metabolism
- src Homology Domains/genetics
Collapse
Affiliation(s)
- Vicki C J Fawcett
- Department of Microbiology and The Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
46
|
Köhler F, Storch B, Kulathu Y, Herzog S, Kuppig S, Reth M, Jumaa H. A leucine zipper in the N terminus confers membrane association to SLP-65. Nat Immunol 2005; 6:204-10. [PMID: 15654340 DOI: 10.1038/ni1163] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Accepted: 12/22/2004] [Indexed: 11/09/2022]
Abstract
Membrane recruitment of adaptor proteins is crucial for coupling antigen receptors to downstream signaling events. Despite the essential function of the B cell adaptor SLP-65, the mechanism of its recruitment to the plasma membrane is not yet understood. Here we show that a highly conserved leucine zipper in the SLP-65 N terminus is responsible for membrane association. Alterations in the N terminus abolished SLP-65 membrane localization and activity, both of which were restored by replacement of the N terminus with a myristoylation signal. The N terminus is an autonomous domain that confers specific localization and function when transferred to green fluorescent protein or the adaptor protein SLP-76. Our data elucidate the mechanism of SLP-65 membrane recruitment and suggest that leucine zipper motifs are essential interaction domains of signaling proteins.
Collapse
Affiliation(s)
- Fabian Köhler
- Institute for Biology III, Albert-Ludwigs University of Freiburg and Max-Planck-Institute for Immunobiology, Stuebeweg 51, 79108 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Gonen R, Beach D, Ainey C, Yablonski D. T cell receptor-induced activation of phospholipase C-gamma1 depends on a sequence-independent function of the P-I region of SLP-76. J Biol Chem 2004; 280:8364-70. [PMID: 15623534 DOI: 10.1074/jbc.m409437200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SLP-76 forms part of a hematopoietic-specific adaptor protein complex, and is absolutely required for T cell development and activation. T cell receptor (TCR)-induced activation of phospholipase C-gamma1 (PLC-gamma1) depends on three features of SLP-76: the N-terminal tyrosine phosphorylation sites, the Gads-binding site, and an intervening sequence, denoted the P-I region, which binds to the SH3 domain of PLC-gamma1 (SH3(PLC)) via a low affinity interaction. Despite extensive research, the mechanism whereby SLP-76 regulates PLC-gamma1 remains uncertain. In this study, we uncover and explore an apparent paradox: whereas the P-I region as a whole is essential for TCR-induced activation of PLC-gamma1 and nuclear factor of activated T cells (NFAT), no particular part of this region is absolutely required. To better understand the contribution of the P-I region to PLC-gamma1 activation, we mapped the PLC-gamma1-binding site within the region, and created a SLP-76 mutant that fails to bind SH3(PLC), but is fully functional, mediating TCR-induced phosphorylation of PLC-gamma1 at tyrosine 783, calcium flux, and nuclear factor of activated T cells activation. Unexpectedly, full functionality of this mutant was maintained even under less than optimal stimulation conditions, such as a low concentration of the anti-TCR antibody. Another SLP-76 mutant, in which the P-I region was scrambled to abolish any sequence-dependent protein-binding motifs, also retained significant functionality. Our results demonstrate that SLP-76 need not interact with SH3(PLC) to activate PLC-gamma1, and further suggest that the P-I region of SLP-76 serves a structural role that is sequence-independent and is not directly related to protein-protein interactions.
Collapse
Affiliation(s)
- Ronnie Gonen
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, P. O. Box 9649, Bat Galim, Haifa 31096, Israel
| | | | | | | |
Collapse
|
48
|
Razzaq TM, Ozegbe P, Jury EC, Sembi P, Blackwell NM, Kabouridis PS. Regulation of T-cell receptor signalling by membrane microdomains. Immunology 2004; 113:413-26. [PMID: 15554919 PMCID: PMC1782593 DOI: 10.1111/j.1365-2567.2004.01998.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 09/09/2004] [Accepted: 09/15/2004] [Indexed: 01/04/2023] Open
Abstract
There is now considerable evidence suggesting that the plasma membrane of mammalian cells is compartmentalized by functional lipid raft microdomains. These structures are assemblies of specialized lipids and proteins and have been implicated in diverse biological functions. Analysis of their protein content using proteomics and other methods revealed enrichment of signalling proteins, suggesting a role for these domains in intracellular signalling. In T lymphocytes, structure/function experiments and complementary pharmacological studies have shown that raft microdomains control the localization and function of proteins which are components of signalling pathways regulated by the T-cell antigen receptor (TCR). Based on these studies, a model for TCR phosphorylation in lipid rafts is presented. However, despite substantial progress in the field, critical questions remain. For example, it is unclear if membrane rafts represent a homogeneous population and if their structure is modified upon TCR stimulation. In the future, proteomics and the parallel development of complementary analytical methods will undoubtedly contribute in further delineating the role of lipid rafts in signal transduction mechanisms.
Collapse
Affiliation(s)
- Tahir M Razzaq
- Bone and Joint Research Unit, William Harvey Research Institute, Queen Mary's School of Medicine and Dentistry, Queen Mary's College, London
| | | | | | | | | | | |
Collapse
|
49
|
Horejsí V, Zhang W, Schraven B. Transmembrane adaptor proteins: organizers of immunoreceptor signalling. Nat Rev Immunol 2004; 4:603-16. [PMID: 15286727 DOI: 10.1038/nri1414] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Václav Horejsí
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, 142 20 Prague 4, Czech Republic
| | | | | |
Collapse
|
50
|
Singer AL, Bunnell SC, Obstfeld AE, Jordan MS, Wu JN, Myung PS, Samelson LE, Koretzky GA. Roles of the Proline-rich Domain in SLP-76 Subcellular Localization and T Cell Function. J Biol Chem 2004; 279:15481-90. [PMID: 14722089 DOI: 10.1074/jbc.m313339200] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The adaptor protein Src homology (SH)2 domain-containing and leukocyte-specific phosphoprotein of 76 kDa (SLP-76) is critical for signal transduction in multiple hematopoietic lineages. It links proximal and distal T cell receptor signaling events through its function as a molecular scaffold in the assembly of multimolecular signaling complexes. Here we studied the functional roles of sub-domains within the SLP-76 proline-rich region, specifically the Gads binding domain and the recently defined P1 domain. To gain a further understanding of the functions mediated by this region, we used three complementary approaches as follows: reconstitution of SLP-76-deficient cells with functional domain deletion mutants, blocking molecular associations through the expression of a dominant negative protein fragment, and directed localization of SLP-76 to assess the role of the domains in SLP-76 recruitment. We find the Gads binding domain and the P1 domain are both necessary for optimal SLP-76 function, and in the absence of these two regions, SLP-76 is functionally inert. Furthermore, we provide direct evidence that SLP-76 localization and, in turn, function are dependent upon association with Gads.
Collapse
Affiliation(s)
- Andrew L Singer
- Signal Transduction Program, Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|