1
|
Omar I, Alakhras A, Mutwali S, Bakhiet M. Molecular insights into T cell development, activation and signal transduction (Review). Biomed Rep 2025; 22:94. [PMID: 40247929 PMCID: PMC12001230 DOI: 10.3892/br.2025.1972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
T cell modulation plays a fundamental role to adaptive and innate immunity, which aids the recognition and defense against pathogens while also maintaining self-tolerance. Numerous molecular pathways participate in this process including thymic selection, T cell receptor and antigen-presenting cells cross linkage, along with co-stimulatory signaling cascades. The present review demonstrates a holistic analysis of various classic and novel mechanisms that govern T cell regulation and emerging therapeutic applications. Recent advancements have introduced novel roles in the journey of T cell modulation that can have a pivotal impact on the understanding of this process; for example, phase separation of the linker for activation of T cells, and the newer application of chimeric antigen receptor (CAR) T cell therapy in autoimmune diseases. While discoveries of proximal and distal signal transduction pathways have contributed to the comprehension of T cell anergy, cytokine-mediated differentiation and the delicate balance between immune activation and tolerance, there are still unresolved debates about further molecular mechanisms. There are also still questions about the long-term side effects of CAR-T cell therapy. Deeper research and analysis are required to further aid the understanding and use of this novel therapeutic approach.
Collapse
Affiliation(s)
- Isra Omar
- Department of Clinical Medicine, College of Medicine, Almaarefa University, 11597 Riyadh, Kingdom of Saudi Arabia
- Department of Clinical Medicine, College of Medicine, University of Medical Sciences and Technology, 3523 Kigali, Rwanda
- Royal College of Physicians of Ireland, Dublin D02 E434, Ireland
| | - Ahmed Alakhras
- Department of Clinical Medicine, College of Medicine, Almaarefa University, 11597 Riyadh, Kingdom of Saudi Arabia
| | - Samahir Mutwali
- TeleGeriatric Research Fellowship Program, Michigan State University, MI 48824, USA
| | - Moiz Bakhiet
- Department of Molecular Medicine and Medical Science, Arabian Gulf University, Manama 328, Kingdom of Bahrain
| |
Collapse
|
2
|
Kim JH, Jung SH, Park C, Lee JR. T cells in ARAP-deficient mice present defective T cell receptor signaling and reduced severity in an experimentally-induced autoimmune disease. Front Immunol 2025; 16:1556616. [PMID: 40264755 PMCID: PMC12011753 DOI: 10.3389/fimmu.2025.1556616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
We previously reported a novel adaptor protein, ARAP, required for T cell receptor signaling and integrin-mediated adhesion. The present study investigates further the role of ARAP in T cell biology using mice with an ARAP gene deficiency. Similar to wild-type mice, ARAP-deficient mice participate in normal breeding and immune cell development. Similar defects were observed in the T cell receptor signaling and adhesion of ARAP-deficient mice, as shown in previous studies investigating ARAP-suppressed Jurkat T cells. ARAP deficiencies analyzed in vivo presented a less severe clinical course of experimental autoimmune encephalomyelitis (EAE) following immunization of mice with the myelin oligodendrocyte glycoprotein (MOG). Serum levels of MOG-specific antibodies and IFN-γ were also reduced in ARAP-deficient EAE mice compared to wild-type EAE mice. Moreover, adoptive transfer of ARAP-deficient T cells induced less severe EAE in recombination-activating gene 1-deficient mice than wild-type T cell transfer. These results strongly suggest that ARAP positively regulates T cell function, while ARAP deficiency in T cells reduces the severity and incidence of EAE.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Mice
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Mice, Knockout
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/deficiency
- Female
- Mice, Inbred C57BL
- Disease Models, Animal
- Adoptive Transfer
- Humans
- Severity of Illness Index
Collapse
Affiliation(s)
| | | | | | - Jong Ran Lee
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Moroi AJ, Newman PJ. The LAT Rheostat as a Regulator of Megakaryocyte Activation. Thromb Haemost 2024; 124:937-947. [PMID: 38788774 DOI: 10.1055/a-2332-6321] [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] [Indexed: 05/26/2024]
Abstract
BACKGROUND Specifically positioned negatively charged residues within the cytoplasmic domain of the adaptor protein, linker for the activation of T cells (LAT), have been shown to be important for efficient phosphorylation of tyrosine residues that function to recruit cytosolic proteins downstream of immunoreceptor tyrosine-based activation motif (ITAM) receptor signaling. LAT tyrosine 132-the binding site for PLC-γ2-is a notable exception, preceded instead by a glycine, making it a relatively poor substrate for phosphorylation. Mutating Gly131 to an acidic residue has been shown in T cells to enhance ITAM-linked receptor-mediated signaling. Whether this is generally true in other cell types is not known. METHODS To examine whether LAT Gly131 restricts ITAM signaling in cells of the megakaryocyte lineage, we introduced an aspartic acid at this position in human induced pluripotent stem cells (iPSCs), differentiated them into megakaryocytes, and examined its functional consequences. RESULTS iPSCs expressing G131D LAT differentiated and matured into megakaryocytes normally, but exhibited markedly enhanced reactivity to glycoprotein VI (GPVI)-agonist stimulation. The rate and extent of LAT Tyr132 and PLC-γ2 phosphorylation, and proplatelet formation on GPVI-reactive substrates, were also enhanced. CONCLUSION These data demonstrate that a glycine residue at the -1 position of LAT Tyr132 functions as a kinetic bottleneck to restrain Tyr132 phosphorylation and signaling downstream of ITAM receptor engagement in the megakaryocyte lineage. These findings may have translational applications in the burgeoning field of in vitro platelet bioengineering.
Collapse
Affiliation(s)
- Alyssa J Moroi
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin, United States
| | - Peter J Newman
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology and, Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Neurath MF, Berg LJ. VAV1 as a putative therapeutic target in autoimmune and chronic inflammatory diseases. Trends Immunol 2024; 45:580-596. [PMID: 39060140 DOI: 10.1016/j.it.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
The guanine nucleotide exchange factor (GEF) VAV1, a previously 'undruggable' protein integral to T/B lymphocyte antigen-receptor signaling, promotes actin polymerization, immunological synapse formation, T cell activation and differentiation, and cytokine production. With the development of novel modalities for targeting proteins, we hypothesize that interventions targeting VAV1 will have therapeutic potential in T and T/B cell-mediated autoimmune and chronic inflammatory diseases. This opinion is supported by recent CRISPR-Cas9 studies showing VAV1 as a key positive regulator of T cell receptor (TCR) activation and cytokine production in primary human CD4+ and CD8+ T cells; data demonstrating that loss/suppression of VAV1 regulates autoimmunity and inflammation; and promising preclinical data from T and T/B cell-mediated disease models of arthritis and colitis showing the effectiveness of selective VAV1 targeting via protein degradation.
Collapse
Affiliation(s)
- Markus F Neurath
- Department of Medicine, 1 & Deutsches Zentrum Immuntherapie, University of Erlangen-Nürnberg, Kussmaul Campus for Medical Research, 91054 Erlangen, Germany
| | - Leslie J Berg
- Department of Immunology & Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
5
|
Weiss A. Peeking Into the Black Box of T Cell Receptor Signaling. Annu Rev Immunol 2024; 42:1-20. [PMID: 37788477 DOI: 10.1146/annurev-immunol-090222-112028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
I have spent more than the last 40 years at the University of California, San Francisco (UCSF), studying T cell receptor (TCR) signaling. I was blessed with supportive mentors, an exceptionally talented group of trainees, and wonderful collaborators and colleagues during my journey who have enabled me to make significant contributions to our understanding of how the TCR initiates signaling. TCR signaling events contribute to T cell development as well as to mature T cell activation and differentiation.
Collapse
Affiliation(s)
- Arthur Weiss
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, California, USA;
| |
Collapse
|
6
|
Mansour R, El-Hassan R, El-Orfali Y, Saidu A, Al-Kalamouni H, Chen Q, Benamar M, Dbaibo G, Hanna-Wakim R, Chatila TA, Massaad MJ. The opposing effects of two gene defects in STX11 and SLP76 on the disease in a patient with an inborn error of immunity. J Allergy Clin Immunol 2023; 152:1597-1606. [PMID: 37595757 DOI: 10.1016/j.jaci.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Inborn errors of immunity are mostly monogenic. However, disease phenotype and outcome may be modified by the coexistence of a second gene defect. OBJECTIVE We sought to identify the genetic basis of the disease in a patient who experienced bleeding episodes, pancytopenia, hepatosplenomegaly, and recurrent pneumonia that resulted in death. METHODS Genetic analysis was done using next-generation sequencing. Protein expression and phosphorylation were determined by immunoblotting. T-cell proliferation and F-actin levels were studied by flow cytometry. RESULTS The patient harbored 2 homozygous deletions in STX11 (c.369_370del, c.374_376del; p.V124fs60∗) previously associated with familial hemophagocytic lymphohistiocytosis and a novel homozygous missense variant in SLP76 (c.767C>T; p.T256I) that resulted in an approximately 85% decrease in SLP76 levels and absent T-cell proliferation. The patient's heterozygous family members showed an approximately 50% decrease in SLP76 levels but normal immune function. SLP76-deficient J14 Jurkat cells did not express SLP76 and had decreased extracellular signal-regulated kinase signaling, basal F-actin levels, and polymerization following T-cell receptor stimulation. Reconstitution of J14 cells with T256I mutant SLP76 resulted in low protein expression and abnormal extracellular signal-regulated kinase phosphorylation and F-actin polymerization after T-cell receptor activation compared with normal expression and J14 function when wild-type SLP76 was introduced. CONCLUSIONS The hypomorphic mutation in SLP76 tones down the hyperinflammation due to STX11 deletion, resulting in a combined immunodeficiency that overshadows the hemophagocytic lymphohistiocytosis phenotype. To our knowledge, this study represents the first report of the opposing effects of 2 gene defects on the disease in a patient with an inborn error of immunity.
Collapse
Affiliation(s)
- Rana Mansour
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rana El-Hassan
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Youmna El-Orfali
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Adam Saidu
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Habib Al-Kalamouni
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Ghassan Dbaibo
- Department of Biochemistry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; Research Center of Excellence in Immunity and Infections, American University of Beirut, Beirut, Lebanon
| | - Rima Hanna-Wakim
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Michel J Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; Research Center of Excellence in Immunity and Infections, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
7
|
Edwards ESJ, Ojaimi S, Ngui J, Seo GH, Kim J, Chunilal S, Yablonski D, O'Hehir RE, van Zelm MC. Combined immunodeficiency and impaired PI3K signaling in a patient with biallelic LCP2 variants. J Allergy Clin Immunol 2023; 152:807-813.e7. [PMID: 37211057 DOI: 10.1016/j.jaci.2023.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Inborn errors affecting components of the T-cell receptor signaling cascade cause combined immunodeficiency with various degrees of severity. Recently, homozygous variants in LCP2 were reported to cause pediatric onset of severe combined immunodeficiency with neutrophil, platelet, and T- and B-cell defects. OBJECTIVE We sought to unravel the genetic cause of combined immunodeficiency and early-onset immune dysregulation in a 26-year-old man who presented with specific antibody deficiency, autoimmunity, and inflammatory bowel disease since early childhood. METHODS The patient was subjected to whole-exome sequencing of genomic DNA and examination of blood neutrophils, platelets, and T and B cells. Expression levels of the Src homology domain 2-containing leukocyte protein of 76 kDa (SLP76) and tonic and ligand-induced PI3K signaling were evaluated by flow-cytometric detection of phosphorylated ribosomal protein S6 in B and T cells. RESULTS Compound heterozygous missense variants were identified in LCP2, affecting the proline-rich repeat domain of SLP76 (p.P190R and p.R204W). The patient's total B- and T-cell numbers were within the normal range, as was platelet function. However, neutrophil function, numbers of unswitched and class-switched memory B cells, and serum IgA were decreased. Moreover, intracellular SLP76 protein levels were reduced in the patient's B cells, CD4+ and CD8+ T cells, and natural killer cells. Tonic and ligand-induced levels of phosphorylated ribosomal protein S6 and ligand-induced phosphorylated PLCγ1 were decreased in the patient's B cells and CD4+ and CD8+ T cells. CONCLUSIONS Biallelic variants in LCP2 impair neutrophil function and T-cell and B-cell antigen-receptor signaling and can cause combined immunodeficiency with early-onset immune dysregulation, even in the absence of platelet defects.
Collapse
Affiliation(s)
- Emily S J Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, Australia; Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia
| | - Samar Ojaimi
- Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Monash Pathology, Monash Health, Melbourne, Australia; Monash Infectious Diseases, Monash Health, Melbourne, Australia; Monash Lung Sleep Allergy Immunology, Monash Health, Melbourne, Australia; Department of Medicine, Southern Clinical School, Monash Health and Monash University, Melbourne, Australia.
| | - James Ngui
- Monash Pathology, Monash Health, Melbourne, Australia
| | - Go Hun Seo
- Division of Medical Genetics, 3billion Inc, Seoul, Korea
| | - JiHye Kim
- Division of Medical Genetics, 3billion Inc, Seoul, Korea
| | - Sanjeev Chunilal
- Department of Pathology and Radiology, Monash Health, Melbourne, Australia
| | - Deborah Yablonski
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Robyn E O'Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, Australia; Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, Australia
| | - Menno C van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, Australia; Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, Australia.
| |
Collapse
|
8
|
Barr VA, Piao J, Balagopalan L, McIntire KM, Schoenberg FP, Samelson LE. Heterogeneity of Signaling Complex Nanostructure in T Cells Activated Via the T Cell Antigen Receptor. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:1503-1522. [PMID: 37488826 PMCID: PMC11230849 DOI: 10.1093/micmic/ozad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/08/2023] [Accepted: 06/18/2023] [Indexed: 07/26/2023]
Abstract
Activation of the T cell antigen receptor (TCR) is a key step in initiating the adaptive immune response. Single-molecule localization techniques have been used to investigate the arrangement of proteins within the signaling complexes formed around activated TCRs, but a clear picture of nanoscale organization in stimulated T cells has not emerged. Here, we have improved the examination of T cell nanostructure by visualizing individual molecules of six different proteins in a single sample of activated Jurkat T cells using the multiplexed antibody-size limited direct stochastic optical reconstruction microscopy (madSTORM) technique. We formally define irregularly shaped regions of interest, compare areas where signaling complexes are concentrated with other areas, and improve the statistical analyses of the locations of molecules. We show that nanoscale organization of proteins is mainly confined to the areas with dense concentrations of TCR-based signaling complexes. However, randomly distributed molecules are also found in some areas containing concentrated signaling complexes. These results are consistent with the view that the proteins within signaling complexes are connected by numerous weak interactions, leading to flexible, dynamic, and mutable structures which produce large variations in the nanostructure found in activated T cells.
Collapse
Affiliation(s)
- Valarie A Barr
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| | - Juan Piao
- Department of Statistics, University of California at Los Angeles, 8965 Math Sciences Building, Los Angeles, CA 90095-1554, USA
| | - Lakshmi Balagopalan
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| | - Katherine M McIntire
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| | - Frederic P Schoenberg
- Department of Statistics, University of California at Los Angeles, 8965 Math Sciences Building, Los Angeles, CA 90095-1554, USA
| | - Lawrence E Samelson
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| |
Collapse
|
9
|
Koretzky GA. Building on the Past, Meeting the Moment. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:849-854. [PMID: 36947823 DOI: 10.4049/jimmunol.2390003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Affiliation(s)
- Gary A Koretzky
- Department of Internal Medicine, Weill Cornell Medicine, New York, NY
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY
| |
Collapse
|
10
|
Lev A, Asleh M, Levy S, Lee YN, Simon AJ, Stepensky P, Nalbandyan K, Nahum A, Ben-Harosh M, Yablonski D, Broides A, Somech R. SLP76 Mutation Associated with Combined Immunodeficiency and EBV-Related Lymphoma. J Clin Immunol 2023; 43:625-635. [PMID: 36474126 DOI: 10.1007/s10875-022-01412-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Increased susceptibility to develop severe forms of Epstein-Barr virus (EBV) infection in early age is a significant hallmark of an underlying primary immunodeficiency (PID). Here, we present immunologic and genetic evaluations of a 3-year-old child who was born to first-cousins parents and presented with recurrent infections, failure to thrive, and severe EBV-related infection and proliferation. A diagnosis of diffuse large B cell lymphoma was made and the immunological workup was suggestive of T cell immunodeficiency. Unfortunately, the patient succumbed to EBV-related lymphoma. Whole-exome sequencing revealed a novel homozygous mutation, c.991del.C; p. Q331Sfs*6 in the SLP76 gene. The SLP76 protein, a TCR signaling molecule, was recently linked to a human disease of the immune system. In order to examine the effect of this new SLP76 mutation on T cell signaling, a SLP76-deficient Jurkat-derived T cell line was transduced either with wild-type (WT), or with the specific SLP76 mutant, or with a mock vector. Downstream TCR signaling events, including ERK1/2 phosphorylation, CD69 expression, and Ca2 + mobilization, were reduced in cells harboring the reported mutation, linking this novel mutation to the expected immunological outcome. SLP76 deficiency should be added to the growing list of monogenetic diseases that predispose affected individuals to acquire severe and uncontrolled EBV infections and to develop substantial complications. This case further links mutations in the SLP76 gene to a significant human immunodeficiency and extends its clinical phenotype.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Mahdi Asleh
- Pediatric Hemato-Oncology Department, Soroka University Medical Center, Beer Sheva, Israel.,Joyce & Irving Goldman Medical School, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Shiran Levy
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Yu Nee Lee
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Amos J Simon
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.,Division of Haematology and Bone Marrow Transplantation, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Polina Stepensky
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Karen Nalbandyan
- Department of Pathology, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Amit Nahum
- Pediatrics Department A and the Primary Immunodeficiency Research Laboratory, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Miriam Ben-Harosh
- Pediatric Hemato-Oncology Department, Soroka University Medical Center, Beer Sheva, Israel
| | - Deborah Yablonski
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Arnon Broides
- Pediatric Immunology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Raz Somech
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
11
|
Ruminski K, Celis-Gutierrez J, Jarmuzynski N, Maturin E, Audebert S, Malissen M, Camoin L, Voisinne G, Malissen B, Roncagalli R. Mapping the SLP76 interactome in T cells lacking each of the GRB2-family adaptors reveals molecular plasticity of the TCR signaling pathway. Front Immunol 2023; 14:1139123. [PMID: 37006259 PMCID: PMC10057548 DOI: 10.3389/fimmu.2023.1139123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
The propagation and diversification of signals downstream of the T cell receptor (TCR) involve several adaptor proteins that control the assembly of multimolecular signaling complexes (signalosomes). The global characterization of changes in protein-protein interactions (PPI) following genetic perturbations is critical to understand the resulting phenotypes. Here, by combining genome editing techniques in T cells and interactomics studies based on affinity purification coupled to mass spectrometry (AP-MS) analysis, we determined and quantified the molecular reorganization of the SLP76 interactome resulting from the ablation of each of the three GRB2-family adaptors. Our data showed that the absence of GADS or GRB2 induces a major remodeling of the PPI network associated with SLP76 following TCR engagement. Unexpectedly, this PPI network rewiring minimally affects proximal molecular events of the TCR signaling pathway. Nevertheless, during prolonged TCR stimulation, GRB2- and GADS-deficient cells displayed a reduced level of activation and cytokine secretion capacity. Using the canonical SLP76 signalosome, this analysis highlights the plasticity of PPI networks and their reorganization following specific genetic perturbations.
Collapse
Affiliation(s)
- Kilian Ruminski
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Javier Celis-Gutierrez
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
- Centre d’Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
| | - Nicolas Jarmuzynski
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
- Centre d’Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
| | - Emilie Maturin
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Stephane Audebert
- Institut Paoli-Calmettes, CRCM, Aix Marseille Université, CNRS, INSERM, Marseille Protóomique, Marseille, France
| | - Marie Malissen
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
- Centre d’Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
| | - Luc Camoin
- Institut Paoli-Calmettes, CRCM, Aix Marseille Université, CNRS, INSERM, Marseille Protóomique, Marseille, France
| | - Guillaume Voisinne
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Bernard Malissen
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
- Centre d’Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
- *Correspondence: Romain Roncagalli, ; Bernard Malissen,
| | - Romain Roncagalli
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
- *Correspondence: Romain Roncagalli, ; Bernard Malissen,
| |
Collapse
|
12
|
Kashiwakura JI, Oritani K, Matsuda T. The Functional Properties and Physiological Roles of Signal-Transducing Adaptor Protein-2 in the Pathogenesis of Inflammatory and Immune Disorders. Biomedicines 2022; 10:biomedicines10123079. [PMID: 36551835 PMCID: PMC9776019 DOI: 10.3390/biomedicines10123079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022] Open
Abstract
Adaptor molecules play a crucial role in signal transduction in immune cells. Several adaptor molecules, such as the linker for the activation of T cells (LAT) and SH2 domain-containing leukocyte protein of 76 kDa (SLP-76), are essential for T cell development and activation following T cell receptor (TCR) aggregation, suggesting that adaptor molecules are good therapeutic targets for T cell-mediated immune disorders, such as autoimmune diseases and allergies. Signal-transducing adaptor protein (STAP)-2 is a member of the STAP family of adaptor proteins. STAP-2 functions as a scaffold for various intracellular proteins, including BRK, signal transducer, and activator of transcription (STAT)3, STAT5, and myeloid differentiation primary response protein (MyD88). In T cells, STAP-2 is involved in stromal cell-derived factor (SDF)-1α-induced migration, integrin-dependent cell adhesion, and Fas-mediated apoptosis. We previously reported the critical function of STAP-2 in TCR-mediated T cell activation and T cell-mediated autoimmune diseases. Here, we review how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases in order to develop novel STAP-2-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Jun-ichi Kashiwakura
- Department of Life Science, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo 0068585, Hokkaido, Japan
- Correspondence: (J.-i.K.); (T.M.); Tel.: +81-11-676-8738 (J.-i.K.); +81-11-706-3243 (T.M.); Fax: +81-11-676-8666 (J.-i.K.); +81-11-706-4990 (T.M.)
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare, Narita 2868686, Chiba, Japan
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 0600812, Hokkaido, Japan
- Correspondence: (J.-i.K.); (T.M.); Tel.: +81-11-676-8738 (J.-i.K.); +81-11-706-3243 (T.M.); Fax: +81-11-676-8666 (J.-i.K.); +81-11-706-4990 (T.M.)
| |
Collapse
|
13
|
Li S, Zhao F, Ye J, Li K, Wang Q, Du Z, Yue Q, Wang S, Wu Q, Chen H. Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19. Med Microbiol Immunol 2022; 211:49-69. [PMID: 35022857 PMCID: PMC8755516 DOI: 10.1007/s00430-021-00727-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023]
Abstract
Metabolic pathways drive cellular behavior. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes lung tissue damage directly by targeting cells or indirectly by producing inflammatory cytokines. However, whether functional alterations are related to metabolic changes in lung cells after SARS-CoV-2 infection remains unknown. Here, we analyzed the lung single-nucleus RNA-sequencing (snRNA-seq) data of several deceased COVID-19 patients and focused on changes in transcripts associated with cellular metabolism. We observed upregulated glycolysis and oxidative phosphorylation in alveolar type 2 progenitor cells, which may block alveolar epithelial differentiation and surfactant secretion. Elevated inositol phosphate metabolism in airway progenitor cells may promote neutrophil infiltration and damage the lung barrier. Further, multiple metabolic alterations in the airway goblet cells are associated with impaired muco-ciliary clearance. Increased glycolysis, oxidative phosphorylation, and inositol phosphate metabolism not only enhance macrophage activation but also contribute to SARS-CoV-2 induced lung injury. The cytotoxicity of natural killer cells and CD8+ T cells may be enhanced by glycerolipid and inositol phosphate metabolism. Glycolytic activation in fibroblasts is related to myofibroblast differentiation and fibrogenesis. Glycolysis, oxidative phosphorylation, and glutathione metabolism may also boost the aging, apoptosis and proliferation of vascular smooth muscle cells, resulting in pulmonary arterial hypertension. In conclusion, this preliminary study revealed a possible cellular metabolic basis for the altered innate immunity, adaptive immunity, and niche cell function in the lung after SARS-CoV-2 infection. Therefore, patients with COVID-19 may benefit from therapeutic strategies targeting cellular metabolism in future.
Collapse
Affiliation(s)
- Shuangyan Li
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Fuxiaonan Zhao
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Jing Ye
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Kuan Li
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China
| | - Qi Wang
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China
| | - Zhongchao Du
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China
| | - Qing Yue
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Sisi Wang
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Qi Wu
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China.
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, 890 Jingu Road, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China.
| |
Collapse
|
14
|
Dinur-Schejter Y, Zaidman I, Mor-Shaked H, Stepensky P. The Clinical Aspect of Adaptor Molecules in T Cell Signaling: Lessons Learnt From Inborn Errors of Immunity. Front Immunol 2021; 12:701704. [PMID: 34456914 PMCID: PMC8397411 DOI: 10.3389/fimmu.2021.701704] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/26/2021] [Indexed: 12/22/2022] Open
Abstract
Adaptor molecules lack enzymatic and transcriptional activities. Instead, they exert their function by linking multiple proteins into intricate complexes, allowing for transmitting and fine-tuning of signals. Many adaptor molecules play a crucial role in T-cell signaling, following engagement of the T-cell receptor (TCR). In this review, we focus on Linker of Activation of T cells (LAT) and SH2 domain-containing leukocyte protein of 76 KDa (SLP-76). Monogenic defects in these adaptor proteins, with known roles in T-cell signaling, have been described as the cause of human inborn errors of immunity (IEI). We describe the current knowledge based on defects in cell lines, murine models and human patients. Germline mutations in Adhesion and degranulation adaptor protein (ADAP), have not resulted in a T-cell defect.
Collapse
Affiliation(s)
- Yael Dinur-Schejter
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Bone Marrow Transplantation and Cancer Immunotherapy Department, Hadassah Ein Kerem Medical Center, Jerusalem, Israel.,Allergy and Clinical Immunology Unit, Hadassah Ein-Kerem Medical Center, Jerusalem, Israel
| | - Irina Zaidman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Bone Marrow Transplantation and Cancer Immunotherapy Department, Hadassah Ein Kerem Medical Center, Jerusalem, Israel
| | - Hagar Mor-Shaked
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Monique and Jacques Roboh Department of Genetic Research, Hadassah Ein Kerem Medical Center, Jerusalem, Israel
| | - Polina Stepensky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,The Bone Marrow Transplantation and Cancer Immunotherapy Department, Hadassah Ein Kerem Medical Center, Jerusalem, Israel
| |
Collapse
|
15
|
Dine E, Reed EH, Toettcher JE. Positive feedback between the T cell kinase Zap70 and its substrate LAT acts as a clustering-dependent signaling switch. Cell Rep 2021; 35:109280. [PMID: 34161759 PMCID: PMC8292983 DOI: 10.1016/j.celrep.2021.109280] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/24/2021] [Accepted: 05/28/2021] [Indexed: 11/29/2022] Open
Abstract
Protein clustering is pervasive in cell signaling, yet how signaling from higher-order assemblies differs from simpler forms of molecular organization is still poorly understood. We present an optogenetic approach to switch between oligomers and heterodimers with a single point mutation. We apply this system to study signaling from the kinase Zap70 and its substrate linker for activation of T cells (LAT), proteins that normally form membrane-localized condensates during T cell activation. We find that fibroblasts expressing synthetic Zap70:LAT clusters activate downstream signaling, whereas one-to-one heterodimers do not. We provide evidence that clusters harbor a positive feedback loop among Zap70, LAT, and Src-family kinases that binds phosphorylated LAT and further activates Zap70. Finally, we extend our optogenetic approach to the native T cell signaling context, where light-induced LAT clustering is sufficient to drive a calcium response. Our study reveals a specific signaling function for protein clusters and identifies a biochemical circuit that robustly senses protein oligomerization state. Dine et al. study how different modes of molecular organization contribute to cell signaling using the kinase Zap70 and its substrate LAT as a model system. Optogenetic manipulation reveals that LAT:Zap70 clusters—but not dimers—trigger potent signaling via localized positive feedback among LAT, Zap70, and Src-family kinases.
Collapse
Affiliation(s)
- Elliot Dine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Ellen H Reed
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; IRCC International Research Collaboration Center, National Institutes of Natural Sciences, 4-3-13 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| | - Jared E Toettcher
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; IRCC International Research Collaboration Center, National Institutes of Natural Sciences, 4-3-13 Toranomon, Minato-ku, Tokyo 105-0001, Japan.
| |
Collapse
|
16
|
Hallumi E, Shalah R, Lo WL, Corso J, Oz I, Beach D, Wittman S, Isenberg A, Sela M, Urlaub H, Weiss A, Yablonski D. Itk Promotes the Integration of TCR and CD28 Costimulation through Its Direct Substrates SLP-76 and Gads. THE JOURNAL OF IMMUNOLOGY 2021; 206:2322-2337. [PMID: 33931484 DOI: 10.4049/jimmunol.2001053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/15/2021] [Indexed: 11/19/2022]
Abstract
The costimulatory receptor CD28 synergizes with the TCR to promote IL-2 production, cell survival, and proliferation; yet the obligatory interdependence of TCR and CD28 signaling is not well understood. Upon TCR stimulation, Gads, a Grb2-family adaptor, bridges the interaction of two additional adaptors, LAT and SLP-76, to form a TCR-induced effector signaling complex. SLP-76 binds the Tec-family tyrosine kinase, Itk, which phosphorylates SLP-76 Y173 and PLC-γ1 Y783. In this study, we identified TCR-inducible, Itk-mediated phosphorylation of Gads Y45 in a human T cell line and in mouse primary T cells. Y45 is found within the N-terminal SH3 domain of Gads, an evolutionarily conserved domain with no known signaling function. Gads Y45 phosphorylation depended on the interaction of Gads with SLP-76 and on the dimerization-dependent binding of Gads to phospho-LAT. We provide evidence that Itk acts through SLP-76 and Gads to promote the TCR/CD28-induced activation of the RE/AP transcriptional element from the IL-2 promoter. Two Itk-related features of SLP-76, Y173 and a proline-rich Itk SH3 binding motif on SLP-76, were dispensable for activation of NFAT but selectively required for the TCR/CD28-induced increase in cytoplasmic and nuclear c-Rel and consequent RE/AP activation. We provide evidence that unphosphorylated, monomeric Gads mediates an RE/AP-directed inhibitory activity that is mitigated upon Gads dimerization and Y45 phosphorylation. This study illuminates a new, to our knowledge, regulatory module, in which TCR-induced, Itk-mediated phosphorylation sites on SLP-76 and Gads control the transcriptional response to TCR/CD28 costimulation, thus enforcing the obligatory interdependence of the TCR and CD28 signaling pathways.
Collapse
Affiliation(s)
- Enas Hallumi
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Rose Shalah
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Wan-Lin Lo
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Jasmin Corso
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ilana Oz
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Dvora Beach
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Samuel Wittman
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Amy Isenberg
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Meirav Sela
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Bioanalytics Research Group, Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Arthur Weiss
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA
| | - Deborah Yablonski
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
17
|
Rudd CE. How the Discovery of the CD4/CD8-p56 lck Complexes Changed Immunology and Immunotherapy. Front Cell Dev Biol 2021; 9:626095. [PMID: 33791292 PMCID: PMC8005572 DOI: 10.3389/fcell.2021.626095] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
The past 25 years have seen enormous progress in uncovering the receptors and signaling mechanisms on T-cells that activate their various effecter functions. Until the late 1980s, most studies on T-cells had focused on the influx of calcium and the levels of cAMP/GMP in T-cells. My laboratory then uncovered the interaction of CD4 and CD8 co-receptors with the protein-tyrosine kinase p56lck which are now widely accepted as the initiators of the tyrosine phosphorylation cascade leading to T-cell activation. The finding explained how immune recognition receptors expressed by many immune cells, which lack intrinsic catalytic activity, can transduce activation signals via non-covalent association with non-receptor tyrosine kinases. The discovery also established the concept that a protein tyrosine phosphorylation cascade operated in T-cells. In this vein, we and others then showed that the CD4- and CD8-p56lck complexes phosphorylate the TCR complexes which led to the identification of other protein-tyrosine kinases such as ZAP-70 and an array of substrates that are now central to studies in T-cell immunity. Other receptors such as B-cell receptor, Fc receptors and others were also subsequently found to use src kinases to control cell growth. In T-cells, p56lck driven phosphorylation targets include co-receptors such as CD28 and CTLA-4 and immune cell-specific adaptor proteins such as LAT and SLP-76 which act to integrate signals proximal to surface receptors. CD4/CD8-p56lck regulated events in T-cells include intracellular calcium mobilization, integrin activation and the induction of transcription factors for gene expression. Lastly, the identification of the targets of p56lck in the TCR and CD28 provided the framework for the development of chimeric antigen receptor (CAR) therapy in the treatment of cancer. In this review, I outline a history of the development of events that led to the development of the "TCR signaling paradigm" and its implications to immunology and immunotherapy.
Collapse
Affiliation(s)
- Christopher E. Rudd
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Faculty of Medicine, Universite de Montreal, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Lev A, Lee YN, Sun G, Hallumi E, Simon AJ, Zrihen KS, Levy S, Beit Halevi T, Papazian M, Shwartz N, Somekh I, Levy-Mendelovich S, Wolach B, Gavrieli R, Vernitsky H, Barel O, Javasky E, Stauber T, Ma CA, Zhang Y, Amariglio N, Rechavi G, Hendel A, Yablonski D, Milner JD, Somech R. Inherited SLP76 deficiency in humans causes severe combined immunodeficiency, neutrophil and platelet defects. J Exp Med 2020; 218:211562. [PMID: 33231617 PMCID: PMC7690938 DOI: 10.1084/jem.20201062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/06/2020] [Accepted: 10/08/2020] [Indexed: 12/30/2022] Open
Abstract
The T cell receptor (TCR) signaling pathway is an ensemble of numerous proteins that are crucial for an adequate immune response. Disruption of any protein involved in this pathway leads to severe immunodeficiency and unfavorable clinical outcomes. Here, we describe an infant with severe immunodeficiency who was found to have novel biallelic mutations in SLP76. SLP76 is a key protein involved in TCR signaling and in other hematopoietic pathways. Previous studies of this protein were performed using Jurkat-derived human leukemic T cell lines and SLP76-deficient mice. Our current study links this gene, for the first time, to a human immunodeficiency characterized by early-onset life-threatening infections, combined T and B cell immunodeficiency, severe neutrophil defects, and impaired platelet aggregation. Hereby, we characterized aspects of the patient's immune phenotype, modeled them with an SLP76-deficient Jurkat-derived T cell line, and rescued some consequences using ectopic expression of wild-type SLP76. Understanding human diseases due to SLP76 deficiency is helpful in explaining the mixed T cell and neutrophil defects, providing a guide for exploring human SLP76 biology.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.,The Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Yu Nee Lee
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Guangping Sun
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Enas Hallumi
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amos J Simon
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.,Division of Haematology and Bone Marrow Transplantation, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren S Zrihen
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Shiran Levy
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Tal Beit Halevi
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Maria Papazian
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Neta Shwartz
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Ido Somekh
- Department of Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarina Levy-Mendelovich
- The Israeli National Hemophilia Center and Thrombosis Unit, The Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Baruch Wolach
- Department of Pediatrics and Laboratory for Leukocyte Function, Meir Medical Center, Kfar Saba, Israel
| | - Ronit Gavrieli
- Department of Pediatrics and Laboratory for Leukocyte Function, Meir Medical Center, Kfar Saba, Israel
| | - Helly Vernitsky
- Division of Haematology and Bone Marrow Transplantation, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ortal Barel
- The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel.,Cancer Research Center, Wohl Institute for Translational Medicine, Sheba Medical Center, Tel Hashomer, Israel
| | - Elisheva Javasky
- The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel.,Cancer Research Center, Wohl Institute for Translational Medicine, Sheba Medical Center, Tel Hashomer, Israel
| | - Tali Stauber
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Chi A Ma
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Yuan Zhang
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Ninette Amariglio
- The Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel.,Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Gideon Rechavi
- Cancer Research Center, Wohl Institute for Translational Medicine, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ayal Hendel
- The Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Deborah Yablonski
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Raz Somech
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
19
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
20
|
Andreotti AH, Joseph RE, Conley JM, Iwasa J, Berg LJ. Multidomain Control Over TEC Kinase Activation State Tunes the T Cell Response. Annu Rev Immunol 2019; 36:549-578. [PMID: 29677469 DOI: 10.1146/annurev-immunol-042617-053344] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Signaling through the T cell antigen receptor (TCR) activates a series of tyrosine kinases. Directly associated with the TCR, the SRC family kinase LCK and the SYK family kinase ZAP-70 are essential for all downstream responses to TCR stimulation. In contrast, the TEC family kinase ITK is not an obligate component of the TCR cascade. Instead, ITK functions as a tuning dial, to translate variations in TCR signal strength into differential programs of gene expression. Recent insights into TEC kinase structure have provided a view into the molecular mechanisms that generate different states of kinase activation. In resting lymphocytes, TEC kinases are autoinhibited, and multiple interactions between the regulatory and kinase domains maintain low activity. Following TCR stimulation, newly generated signaling modules compete with the autoinhibited core and shift the conformational ensemble to the fully active kinase. This multidomain control over kinase activation state provides a structural mechanism to account for ITK's ability to tune the TCR signal.
Collapse
Affiliation(s)
- Amy H Andreotti
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa 50011, USA; ,
| | - Raji E Joseph
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa 50011, USA; ,
| | - James M Conley
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA; ,
| | - Janet Iwasa
- Department of Biochemistry, University of Utah, Salt Lake City, Utah 84112, USA;
| | - Leslie J Berg
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA; ,
| |
Collapse
|
21
|
Yablonski D. Bridging the Gap: Modulatory Roles of the Grb2-Family Adaptor, Gads, in Cellular and Allergic Immune Responses. Front Immunol 2019; 10:1704. [PMID: 31402911 PMCID: PMC6669380 DOI: 10.3389/fimmu.2019.01704] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/08/2019] [Indexed: 01/07/2023] Open
Abstract
Antigen receptor signaling pathways are organized by adaptor proteins. Three adaptors, LAT, Gads, and SLP-76, form a heterotrimeric complex that mediates signaling by the T cell antigen receptor (TCR) and by the mast cell high affinity receptor for IgE (FcεRI). In both pathways, antigen recognition triggers tyrosine phosphorylation of LAT and SLP-76. The recruitment of SLP-76 to phospho-LAT is bridged by Gads, a Grb2 family adaptor composed of two SH3 domains flanking a central SH2 domain and an unstructured linker region. The LAT-Gads-SLP-76 complex is further incorporated into larger microclusters that mediate antigen receptor signaling. Gads is positively regulated by dimerization, which promotes its cooperative binding to LAT. Negative regulation occurs via phosphorylation or caspase-mediated cleavage of the linker region of Gads. FcεRI-mediated mast cell activation is profoundly impaired in LAT- Gads- or SLP-76-deficient mice. Unexpectedly, the thymic developmental phenotype of Gads-deficient mice is much milder than the phenotype of LAT- or SLP-76-deficient mice. This distinction suggests that Gads is not absolutely required for TCR signaling, but may modulate its sensitivity, or regulate a particular branch of the TCR signaling pathway; indeed, the phenotypic similarity of Gads- and Itk-deficient mice suggests a functional connection between Gads and Itk. Additional Gads binding partners include costimulatory proteins such as CD28 and CD6, adaptors such as Shc, ubiquitin regulatory proteins such as USP8 and AMSH, and kinases such as HPK1 and BCR-ABL, but the functional implications of these interactions are not yet fully understood. No interacting proteins or function have been ascribed to the evolutionarily conserved N-terminal SH3 of Gads. Here we explore the biochemical and functional properties of Gads, and its role in regulating allergy, T cell development and T-cell mediated immunity.
Collapse
Affiliation(s)
- Deborah Yablonski
- The Immune Cell Signaling Lab, Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
22
|
Raab M, Strebhardt K, Rudd CE. Immune adaptor SKAP1 acts a scaffold for Polo-like kinase 1 (PLK1) for the optimal cell cycling of T-cells. Sci Rep 2019; 9:10462. [PMID: 31320682 PMCID: PMC6639320 DOI: 10.1038/s41598-019-45627-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/06/2019] [Indexed: 02/06/2023] Open
Abstract
While the immune cell adaptor protein SKAP1 mediates LFA-1 activation induced by antigen-receptor (TCR/CD3) ligation on T-cells, it is unclear whether the adaptor interacts with other mediators of T-cell function. In this context, the serine/threonine kinase, polo-like kinase (PLK1) regulates multiple steps in the mitotic and cell cycle progression of mammalian cells. Here, we show that SKAP1 is phosphorylated by and binds to PLK1 for the optimal cycling of T-cells. PLK1 binds to the N-terminal residue serine 31 (S31) of SKAP1 and the interaction is needed for optimal PLK1 kinase activity. Further, siRNA knock-down of SKAP1 reduced the rate of T-cell division concurrent with a delay in the expression of PLK1, Cyclin A and pH3. Reconstitution of these KD cells with WT SKAP1, but not the SKAP1 S31 mutant, restored normal cell division. SKAP1-PLK1 binding is dynamically regulated during the cell cycle of T-cells. Our findings identify a novel role for SKAP1 in the regulation of PLK1 and optimal cell cycling needed for T-cell clonal expansion in response to antigenic activation.
Collapse
Affiliation(s)
- Monika Raab
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- Cell Signaling Section, Department of Pathology, Tennis Court Road, University of Cambridge, CB2 1Q, Cambridge, UK.
| | - Klaus Strebhardt
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Christopher E Rudd
- Cell Signaling Section, Department of Pathology, Tennis Court Road, University of Cambridge, CB2 1Q, Cambridge, UK.
- Centre de Recherch-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, Quebec, H1T 2M4, Canada.
- Département de Medicine, Université de Montréal, Montreal, Quebec, H3C 3J7, Canada.
| |
Collapse
|
23
|
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
|
24
|
Wei X, Zhao T, Ai K, Zhang Y, Li H, Yang J. c-Raf participates in adaptive immune response of Nile tilapia via regulating lymphocyte activation. FISH & SHELLFISH IMMUNOLOGY 2019; 86:507-515. [PMID: 30513386 DOI: 10.1016/j.fsi.2018.11.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 06/09/2023]
Abstract
RAF proto-oncogene serine/threonine-protein kinase (c-Raf) is a MAP kinase kinase kinase (MAPKKK) that participates in the Erk1/2 pathway and plays an important role in lymphocyte activation. However, the study on how c-Raf regulates adaptive immunity in non-mammal is still limited. In present study, based on analysis of sequence characteristics of c-Raf from Oreochromis niloticus (On-c-Raf), we investigated its regulation roles on teleost lymphocyte activation. The On-c-Raf was highly conserved during evolution, which was composed of a Raf-like Ras-binding domain (RBD), a protein kinase C conserved region 1 (C1) domain and a serine/threonine protein kinase catalytic (S_TKc) domain. Its mRNA showed a wide distribution in tissues of O. niloticus and with the highest expression in gill. After Aeromonas hydrophila infection, during the adaptive immune stage transcription level of On-c-Raf was significantly upregulated on day 8, but came back to original level on day 16 and 30, suggesting the potential involvement of On-c-Raf in primary response but not memory formation. Furthermore, On-c-Raf mRNA in leukocytes of Nile tilapias was obviously induced by in vitro stimulation of T cell mitogen PHA. More importantly, in vitro stimulation of lymphocytes agonist PMA augmented phosphorylation level of On-c-Raf in leukocytes detected by western-blot and immunofluorescent. Thus, c-Raf regulated lymphocyte activation of Nile tilapia on both mRNA and phosphorylation level. Together, our results revealed that the c-Raf from teleost Nile tilapia engaged in adaptive immune response by regulating lymphocytes activation. Since the regulatory mechanism of lymphocyte-mediated adaptive immunity is largely unknown in teleost, our study provided important evidences to understand teleost adaptive immunity, and also shed a novel perspective for the evolution of adaptive immune system.
Collapse
Affiliation(s)
- Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Tianyu Zhao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kete Ai
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yu Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Huiying Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
25
|
Wang QL, Liang JQ, Gong BN, Xie JJ, Yi YT, Lan X, Li Y. T Cell Receptor (TCR)-Induced PLC-γ1 Sumoylation via PIASxβ and PIAS3 SUMO E3 Ligases Regulates the Microcluster Assembly and Physiological Function of PLC-γ1. Front Immunol 2019; 10:314. [PMID: 30873169 PMCID: PMC6403162 DOI: 10.3389/fimmu.2019.00314] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/06/2019] [Indexed: 11/16/2022] Open
Abstract
The SUMO modification system plays an important role in T cell activation, yet how sumoylation regulates TCR-proximal signaling remains largely unknown. We show here that Phospholipase C-γ1 (PLC-γ1) is conjugated by SUMO1 at K54 and K987 upon TCR stimulation and that K54 sumoylation is pivotal for PLC-γ1-mediated T cell activation. We further demonstrate that TCR-induced K54 sumoylation of PLC-γ1 significantly promotes the formation of PLC-γ1 microclusters and the association of PLC-γ1 with the adaptor proteins SLP76 and Gads, but only slightly affects the phosphorylation of PLC-γ1 on Y783, which determines the enzyme catalytic activity. Moreover, upon TCR stimulation, the SUMO E3 ligases PIASxβ and PIAS3 both interact with PLC-γ1 and cooperate to sumoylate PLC-γ1, facilitating the assembly of PLC-γ1 microclusters. Together, our findings reveal a critical role of PLC-γ1 K54 sumoylation in PLC-γ1 microcluster assembly that controls PLC-γ1-mediated T cell activation, suggesting that sumoylation may have an important role in the microcluster assembly of TCR-proximal signaling proteins.
Collapse
Affiliation(s)
- Qi-Long Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jia-Qi Liang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Bei-Ni Gong
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ji-Ji Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yu-Ting Yi
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xin Lan
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yingqiu Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Yablonski D. Bridging the Gap: Modulatory Roles of the Grb2-Family Adaptor, Gads, in Cellular and Allergic Immune Responses. Front Immunol 2019; 10:1704. [PMID: 31402911 DOI: 10.3389/fimmu.2019.01704/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/08/2019] [Indexed: 05/22/2023] Open
Abstract
Antigen receptor signaling pathways are organized by adaptor proteins. Three adaptors, LAT, Gads, and SLP-76, form a heterotrimeric complex that mediates signaling by the T cell antigen receptor (TCR) and by the mast cell high affinity receptor for IgE (FcεRI). In both pathways, antigen recognition triggers tyrosine phosphorylation of LAT and SLP-76. The recruitment of SLP-76 to phospho-LAT is bridged by Gads, a Grb2 family adaptor composed of two SH3 domains flanking a central SH2 domain and an unstructured linker region. The LAT-Gads-SLP-76 complex is further incorporated into larger microclusters that mediate antigen receptor signaling. Gads is positively regulated by dimerization, which promotes its cooperative binding to LAT. Negative regulation occurs via phosphorylation or caspase-mediated cleavage of the linker region of Gads. FcεRI-mediated mast cell activation is profoundly impaired in LAT- Gads- or SLP-76-deficient mice. Unexpectedly, the thymic developmental phenotype of Gads-deficient mice is much milder than the phenotype of LAT- or SLP-76-deficient mice. This distinction suggests that Gads is not absolutely required for TCR signaling, but may modulate its sensitivity, or regulate a particular branch of the TCR signaling pathway; indeed, the phenotypic similarity of Gads- and Itk-deficient mice suggests a functional connection between Gads and Itk. Additional Gads binding partners include costimulatory proteins such as CD28 and CD6, adaptors such as Shc, ubiquitin regulatory proteins such as USP8 and AMSH, and kinases such as HPK1 and BCR-ABL, but the functional implications of these interactions are not yet fully understood. No interacting proteins or function have been ascribed to the evolutionarily conserved N-terminal SH3 of Gads. Here we explore the biochemical and functional properties of Gads, and its role in regulating allergy, T cell development and T-cell mediated immunity.
Collapse
Affiliation(s)
- Deborah Yablonski
- The Immune Cell Signaling Lab, Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
27
|
Raab M, Strebhardt K, Rudd CE. Immune adaptor protein SKAP1 (SKAP-55) forms homodimers as mediated by the N-terminal region. BMC Res Notes 2018; 11:869. [PMID: 30522503 PMCID: PMC6282339 DOI: 10.1186/s13104-018-3976-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 12/04/2018] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Immune cell adaptor protein SKAP1 couples the antigen-receptor (TCR/CD3) with the activation of LFA-1 adhesion in T-cells. Previous work by ourselves and others have shown that SKAP1 can directly bind to other adaptors such as ADAP and RapL. However, it has been unclear whether SKAP1 can form homodimers with itself and the regions within SKAP1 that mediated homodimer formation. RESULTS Here, we show that SKAP1 and SKAP2 form homodimers in cells. Homodimer formation of immune adaptor protein SKAP1 (SKAP-55) are mediated by residues A17 to L21 in the SKAP1 N-terminal region. SKAP1 dimer formation was not needed for its binding to RapL. These data indicate that the pathway linking SKAP1 to RapL is not dependent on the homo-dimerization of SKAP1.
Collapse
Affiliation(s)
- Monika Raab
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.,German Cancer Consortium (DKTK)/German Cancer Research Center, Heidelberg, Germany
| | - Christopher E Rudd
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany. .,Research Center-Maisonneuve-Rosemont Hospital (CRHMR), Montreal, QC, H1T 2M4, Canada. .,Cell Signalling Section, Department of Pathology, Cambridge University, Cambridge, CB2 1QP, UK.
| |
Collapse
|
28
|
Poissonnier A, Guégan JP, Nguyen HT, Best D, Levoin N, Kozlov G, Gehring K, Pineau R, Jouan F, Morere L, Martin S, Thomas M, Lazaro E, Douchet I, Ducret T, van de Weghe P, Blanco P, Jean M, Vacher P, Legembre P. Disrupting the CD95–PLCγ1 interaction prevents Th17-driven inflammation. Nat Chem Biol 2018; 14:1079-1089. [DOI: 10.1038/s41589-018-0162-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 09/25/2018] [Indexed: 11/09/2022]
|
29
|
Barreira M, Rodríguez-Fdez S, Bustelo XR. New insights into the Vav1 activation cycle in lymphocytes. Cell Signal 2018; 45:132-144. [PMID: 29410283 PMCID: PMC7615736 DOI: 10.1016/j.cellsig.2018.01.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 10/18/2022]
Abstract
Vav1 is a hematopoietic-specific Rho GDP/GTP exchange factor and signaling adaptor. Although these activities are known to be stimulated by direct Vav1 phosphorylation, little information still exists regarding the regulatory layers that influence the overall Vav1 activation cycle. Using a collection of cell models and activation-mimetic Vav1 mutants, we show here that the dephosphorylated state of Vav1 in nonstimulated T cells requires the presence of a noncatalytic, phospholipase Cγ1-Slp76-mediated inhibitory pathway. Upon T cell stimulation, Vav1 becomes rapidly phosphorylated via the engagement of Lck and, to a much lesser extent, other Src family kinases and Zap70. In this process, Lck, Zap70 and the adaptor protein Lat contribute differently to the dynamics and amplitude of the Vav1 phosphorylated pool. Consistent with a multiphosphosite activation mechanism, the optimal stimulation of Vav1 can only be recapitulated by the combination of several activation-mimetic phosphosite mutants. The analysis of these mutants has also unveiled the presence of different Vav1 signaling competent states that are influenced by phosphosites present in the N- and C-terminal domains of the protein.
Collapse
Affiliation(s)
- María Barreira
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
30
|
Courtney AH, Lo WL, Weiss A. TCR Signaling: Mechanisms of Initiation and Propagation. Trends Biochem Sci 2017; 43:108-123. [PMID: 29269020 DOI: 10.1016/j.tibs.2017.11.008] [Citation(s) in RCA: 367] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/21/2017] [Accepted: 11/21/2017] [Indexed: 10/18/2022]
Abstract
The mechanisms by which a T cell detects antigen using its T cell antigen receptor (TCR) are crucial to our understanding of immunity and the harnessing of T cells therapeutically. A hallmark of the T cell response is the ability of T cells to quantitatively respond to antigenic ligands derived from pathogens while remaining inert to similar ligands derived from host tissues. Recent studies have revealed exciting properties of the TCR and the behaviors of its signaling effectors that are used to detect and discriminate between antigens. Here we highlight these recent findings, focusing on the proximal TCR signaling molecules Zap70, Lck, and LAT, to provide mechanistic models and insights into the exquisite sensitivity and specificity of the TCR.
Collapse
Affiliation(s)
- Adam H Courtney
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| | - Wan-Lin Lo
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| | - Arthur Weiss
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA.
| |
Collapse
|
31
|
Sukenik S, Frushicheva MP, Waknin-Lellouche C, Hallumi E, Ifrach T, Shalah R, Beach D, Avidan R, Oz I, Libman E, Aronheim A, Lewinson O, Yablonski D. Dimerization of the adaptor Gads facilitates antigen receptor signaling by promoting the cooperative binding of Gads to the adaptor LAT. Sci Signal 2017; 10:10/498/eaal1482. [PMID: 28951535 DOI: 10.1126/scisignal.aal1482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The accurate assembly of signalosomes centered on the adaptor protein LAT (linker of activated T cells) is required for antigen receptor signaling in T cells and mast cells. During signalosome assembly, members of the growth factor receptor-bound protein 2 (Grb2) family of cytosolic adaptor proteins bind cooperatively to LAT through interactions with its phosphorylated tyrosine (pTyr) residues. We demonstrated the Src homology 2 (SH2) domain-mediated dimerization of the Grb2 family member, Grb2-related adaptor downstream of Shc (Gads). Gads dimerization was mediated by an SH2 domain interface, which is distinct from the pTyr binding pocket and which promoted cooperative, preferential binding of paired Gads to LAT. This SH2 domain-intrinsic mechanism of cooperativity, which we quantified by mathematical modeling, enabled Gads to discriminate between dually and singly phosphorylated LAT molecules. Mutational inactivation of the dimerization interface reduced cooperativity and abrogated Gads signaling in T cells and mast cells. The dimerization-dependent, cooperative binding of Gads to LAT may increase antigen receptor sensitivity by reducing signalosome formation at incompletely phosphorylated LAT molecules, thereby prioritizing the formation of complete signalosomes.
Collapse
Affiliation(s)
- Sigalit Sukenik
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Maria P Frushicheva
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Cecilia Waknin-Lellouche
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Enas Hallumi
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Talia Ifrach
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Rose Shalah
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Dvora Beach
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Reuven Avidan
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Ilana Oz
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Evgeny Libman
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Ami Aronheim
- Department of Cell Biology and Cancer Science, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Oded Lewinson
- Department of Biochemistry, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Deborah Yablonski
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525433, Israel.
| |
Collapse
|
32
|
Kruglova NA, Meshkova TD, Kopylov AT, Mazurov DV, Filatov AV. Constitutive and activation-dependent phosphorylation of lymphocyte phosphatase-associated phosphoprotein (LPAP). PLoS One 2017; 12:e0182468. [PMID: 28827793 PMCID: PMC5565103 DOI: 10.1371/journal.pone.0182468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 07/19/2017] [Indexed: 12/15/2022] Open
Abstract
Lymphocyte phosphatase-associated phosphoprotein (LPAP) is a small transmembrane protein expressed exclusively in lymphocytes. LPAP is a component of a supramolecular complex composed of the phosphatase CD45, the co-receptor CD4, and the kinase Lck. In contrast to its immunologically important partners, the function of LPAP is unknown. We hypothesized that the biological role of LPAP may be determined by analyzing LPAP phosphorylation. In the present study, we identified LPAP phosphorylation sites by site-directed mutagenesis, phospho-specific antibodies, and protein immunoprecipitation coupled to mass spectrometry analysis. Our results confirmed previous reports that Ser-99, Ser-153, and Ser-163 are phosphorylated, as well as provided evidence for the phosphorylation of Ser-172. Using various SDS-PAGE techniques, we detected and quantified a set of LPAP phosphoforms that were assigned to a combination of particular phosphorylation events. The phosphorylation of LPAP appears to be a tightly regulated process. Our results support the model: following phorbol 12-myristate 13-acetate (PMA) or TCR/CD3 activation of T cells, LPAP is rapidly dephosphorylated at Ser-99 and Ser-172 and slowly phosphorylated at Ser-163. Ser-153 exhibited a high basal level of phosphorylation in both resting and activated cells. Therefore, we suggest that LPAP may function as a co-regulator of T-cell signaling.
Collapse
|
33
|
Dieckmann NMG, Frazer GL, Asano Y, Stinchcombe JC, Griffiths GM. The cytotoxic T lymphocyte immune synapse at a glance. J Cell Sci 2017; 129:2881-6. [PMID: 27505426 DOI: 10.1242/jcs.186205] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The immune synapse provides an important structure for communication with immune cells. Studies on immune synapses formed by cytotoxic T lymphocytes (CTLs) highlight the dynamic changes and specialised mechanisms required to facilitate focal signalling and polarised secretion in immune cells. In this Cell Science at a Glance article and the accompanying poster, we illustrate the different steps that reveal the specialised mechanisms used to focus secretion at the CTL immune synapse and allow CTLs to be such efficient and precise serial killers.
Collapse
Affiliation(s)
- Nele M G Dieckmann
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge CB2 0XY, UK
| | - Gordon L Frazer
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge CB2 0XY, UK
| | - Yukako Asano
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge CB2 0XY, UK
| | - Jane C Stinchcombe
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge CB2 0XY, UK
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge CB2 0XY, UK
| |
Collapse
|
34
|
LFA-1 activates focal adhesion kinases FAK1/PYK2 to generate LAT-GRB2-SKAP1 complexes that terminate T-cell conjugate formation. Nat Commun 2017; 8:16001. [PMID: 28699640 PMCID: PMC5510181 DOI: 10.1038/ncomms16001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 05/23/2017] [Indexed: 01/27/2023] Open
Abstract
Lymphocyte function-associated antigen 1 (LFA-1) affinity and avidity changes have been assumed to mediate adhesion to intercellular adhesion molecule-1 for T-cell conjugation to dendritic cells (DC). Although the T-cell receptor (TCR) and LFA-1 can generate intracellular signals, the immune cell adaptor protein linker for the activation of T cells (LAT) couples the TCR to downstream events. Here, we show that LFA-1 can mediate both adhesion and de-adhesion, dependent on receptor clustering. Although increased affinity mediates adhesion, LFA-1 cross-linking induced the association and activation of the protein-tyrosine kinases FAK1/PYK1 that phosphorylated LAT selectively on a single Y-171 site for the binding to adaptor complex GRB-2-SKAP1. LAT-GRB2-SKAP1 complexes were distinct from canonical LAT-GADs-SLP-76 complexes. LFA-1 cross-linking increased the presence of LAT-GRB2-SKAP1 complexes relative to LAT-GADs-SLP-76 complexes. LFA-1-FAK1 decreased T-cell-dendritic cell (DC) dwell times dependent on LAT-Y171, leading to reduced DO11.10 T cell binding to DCs and proliferation to OVA peptide. Overall, our findings outline a new model for LFA-1 in which the integrin can mediate both adhesion and de-adhesion events dependent on receptor cross-linking. The T-cell integrin LFA-1 binds ICAM-1 on antigen presenting cells to affect TCR-MHC interactions. Here the authors show detailed mechanics of how LFA-1 ligation affects T-cell conjugation to dendritic cells to regulate adhesion and de-adhesion of these cells in the context of antigen presentation.
Collapse
|
35
|
Belmont J, Gu T, Mudd A, Salomon AR. A PLC-γ1 Feedback Pathway Regulates Lck Substrate Phosphorylation at the T-Cell Receptor and SLP-76 Complex. J Proteome Res 2017. [PMID: 28644030 DOI: 10.1021/acs.jproteome.6b01026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Phospholipase C gamma 1 (PLC-γ1) occupies a critically important position in the T-cell signaling pathway. While its functions as a regulator of both Ca2+ signaling and PKC-family kinases are well characterized, PLC-γ1's role in the regulation of early T-cell receptor signaling events is incompletely understood. Activation of the T-cell receptor leads to the formation of a signalosome complex between SLP-76, LAT, PLC-γ1, Itk, and Vav1. Recent studies have revealed the existence of both positive and negative feedback pathways from SLP-76 to the apical kinase in the pathway, Lck. To determine if PLC-γ1 contributes to the regulation of these feedback networks, we performed a quantitative phosphoproteomic analysis of PLC-γ1-deficient T cells. These data revealed a previously unappreciated role for PLC-γ1 in the positive regulation of Zap-70 and T-cell receptor tyrosine phosphorylation. Conversely, PLC-γ1 negatively regulated the phosphorylation of SLP-76-associated proteins, including previously established Lck substrate phosphorylation sites within this complex. While the positive and negative regulatory phosphorylation sites on Lck were largely unchanged, Tyr192 phosphorylation was elevated in Jgamma1. The data supports a model wherein Lck's targeting, but not its kinase activity, is altered by PLC-γ1, possibly through Lck Tyr192 phosphorylation and increased association of the kinase with protein scaffolds SLP-76 and TSAd.
Collapse
Affiliation(s)
- Judson Belmont
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States
| | - Tao Gu
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States
| | - Ashley Mudd
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States
| | - Arthur R Salomon
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States.,Department of Chemistry, Brown University , Providence, Rhode Island 02912, United States
| |
Collapse
|
36
|
Thaker YR, Recino A, Raab M, Jabeen A, Wallberg M, Fernandez N, Rudd CE. Activated Cdc42-associated kinase 1 (ACK1) binds the sterile α motif (SAM) domain of the adaptor SLP-76 and phosphorylates proximal tyrosines. J Biol Chem 2017; 292:6281-6290. [PMID: 28188290 PMCID: PMC5391757 DOI: 10.1074/jbc.m116.759555] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/09/2017] [Indexed: 01/24/2023] Open
Abstract
The adaptor protein Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) plays a crucial role in T cell activation by linking antigen receptor (T cell receptor, TCR) signals to downstream pathways. At its N terminus, SLP-76 has three key tyrosines (Tyr-113, Tyr-128, and Tyr-145, "3Y") as well as a sterile α motif (SAM) domain whose function is unclear. We showed previously that the SAM domain has two binding regions that mediate dimer and oligomer formation. In this study, we have identified SAM domain-carrying non-receptor tyrosine kinase, activated Cdc42-associated tyrosine kinase 1 (ACK1; also known as Tnk2, tyrosine kinase non-receptor 2) as a novel binding partner of SLP-76. Co-precipitation, laser-scanning confocal microscopy, and in situ proximity analysis confirmed the binding of ACK1 to SLP-76. Further, the interaction was induced in response to the anti-TCR ligation and abrogated by the deletion of SLP-76 SAM domain (ΔSAM) or mutation of Tyr-113, Tyr-128, and Tyr-145 to phenylalanine (3Y3F). ACK1 induced phosphorylation of the SLP-76 N-terminal tyrosines (3Y) dependent on the SAM domain. Further, ACK1 promoted calcium flux and NFAT-AP1 promoter activity and decreased the motility of murine CD4+ primary T cells on ICAM-1-coated plates, an event reversed by a small molecule inhibitor of ACK1 (AIM-100). These findings identify ACK1 as a novel SLP-76-associated protein-tyrosine kinase that modulates early activation events in T cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Amino Acid Motifs
- Amino Acid Substitution
- Animals
- Humans
- Jurkat Cells
- Lymphocyte Activation/physiology
- Mice
- Mutation, Missense
- Phosphoproteins/genetics
- Phosphoproteins/immunology
- Phosphoproteins/metabolism
- Phosphorylation/physiology
- Protein Domains
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/immunology
- Protein-Tyrosine Kinases/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tyrosine
Collapse
Affiliation(s)
- Youg R Thaker
- From the Cell Signaling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom,
| | - Asha Recino
- From the Cell Signaling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Monika Raab
- the Department of Obstetrics and Gynecology, School of Medicine, J. W. Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Asma Jabeen
- the School of Biological Science, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Maja Wallberg
- From the Cell Signaling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Nelson Fernandez
- the School of Biological Science, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Christopher E Rudd
- From the Cell Signaling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
- the Division of Immunology-Oncology Research Center Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada, and
- the Département de Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
37
|
Navas VH, Cuche C, Alcover A, Di Bartolo V. Serine Phosphorylation of SLP76 Is Dispensable for T Cell Development but Modulates Helper T Cell Function. PLoS One 2017; 12:e0170396. [PMID: 28107427 PMCID: PMC5249077 DOI: 10.1371/journal.pone.0170396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/04/2017] [Indexed: 12/26/2022] Open
Abstract
The adapter protein SLP76 is a key orchestrator of T cell receptor (TCR) signal transduction. We previously identified a negative feedback loop that modulates T cell activation, involving phosphorylation of Ser376 of SLP76 by the hematopoietic progenitor kinase 1 (HPK1). However, the physiological relevance of this regulatory mechanism was still unknown. To address this question, we generated a SLP76-S376A-expressing knock-in mouse strain and investigated the effects of Ser376 mutation on T cell development and function. We report here that SLP76-S376A-expressing mice exhibit normal thymocyte development and no detectable phenotypic alterations in mature T cell subsets or other lymphoid and myeloid cell lineages. Biochemical analyses revealed that mutant T cells were hypersensitive to TCR stimulation. Indeed, phosphorylation of several signaling proteins, including SLP76 itself, phospholipase Cγ1 and the protein kinases AKT and ERK1/2, was increased. These modifications correlated with increased Th1-type and decreased Th2-type cytokine production by SLP76-S376A T cells, but did not result in significant changes of proliferative capacity nor activation-induced cell death susceptibility. Hence, our results reveal that SLP76-Ser376 phosphorylation does not mediate all HPK1-dependent regulatory effects in T cells but it fine-tunes helper T cell responses.
Collapse
Affiliation(s)
- Victor H. Navas
- Lymphocyte Cell Biology Unit, Institut Pasteur, Paris, France
- CNRS URA 1961, Paris, France
- Université "Pierre et Marie Curie", Paris, France
| | - Céline Cuche
- Lymphocyte Cell Biology Unit, Institut Pasteur, Paris, France
- CNRS URA 1961, Paris, France
- INSERM U1221, Paris, France
| | - Andres Alcover
- Lymphocyte Cell Biology Unit, Institut Pasteur, Paris, France
- CNRS URA 1961, Paris, France
- INSERM U1221, Paris, France
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, Institut Pasteur, Paris, France
- CNRS URA 1961, Paris, France
- INSERM U1221, Paris, France
- * E-mail:
| |
Collapse
|
38
|
Barr VA, Sherman E, Yi J, Akpan I, Rouquette-Jazdanian AK, Samelson LE. Development of nanoscale structure in LAT-based signaling complexes. J Cell Sci 2016; 129:4548-4562. [PMID: 27875277 PMCID: PMC5201021 DOI: 10.1242/jcs.194886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/04/2016] [Indexed: 12/30/2022] Open
Abstract
The adapter molecule linker for activation of T cells (LAT) plays a crucial role in forming signaling complexes induced by stimulation of the T cell receptor (TCR). These multi-molecular complexes are dynamic structures that activate highly regulated signaling pathways. Previously, we have demonstrated nanoscale structure in LAT-based complexes where the adapter SLP-76 (also known as LCP2) localizes to the periphery of LAT clusters. In this study, we show that initially LAT and SLP-76 are randomly dispersed throughout the clusters that form upon TCR engagement. The segregation of LAT and SLP-76 develops near the end of the spreading process. The local concentration of LAT also increases at the same time. Both changes require TCR activation and an intact actin cytoskeleton. These results demonstrate that the nanoscale organization of LAT-based signaling complexes is dynamic and indicates that different kinds of LAT-based complexes appear at different times during T cell activation.
Collapse
Affiliation(s)
- Valarie A Barr
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Jason Yi
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Itoro Akpan
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | | | - Lawrence E Samelson
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
39
|
Fu G, Yu M, Chen Y, Zheng Y, Zhu W, Newman DK, Wang D, Wen R. Phospholipase Cγ1 is required for pre-TCR signal transduction and pre-T cell development. Eur J Immunol 2016; 47:74-83. [PMID: 27759161 DOI: 10.1002/eji.201646522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/14/2016] [Accepted: 10/17/2016] [Indexed: 12/23/2022]
Abstract
Pre-T cell receptor (TCR) signaling is required for pre-T cell survival, proliferation, and differentiation from the CD4 and CD8 double negative (DN) to the double positive (DP) stage. However, the pre-TCR signal transduction pathway is not fully understood and the signaling molecules involved have not been completely identified. Phospholipase Cγ (PLCγ) 1 is an important signaling molecule that generates two second messengers, diacylglycerol and inositol 1,4,5-trisphosphate, that are important to mediate PKC activation and intracellular Ca2+ flux in many signaling pathways. Previously, we have shown that PLCγ1 is important for TCR-mediated signaling, development and T-cell activation, but the role of PLCγ1 in pre-TCR signal transduction and pre-T cell development is not known. In this study, we demonstrated that PLCγ1 expression level in pre-T cells was comparable to that in mature T cells. Deletion of PLCγ1 prior to the pre-TCR signaling stage partially blocked the DN3 to DN4 transition and reduced thymic cellularity. We also demonstrated that deletion of PLCγ1 impaired pre-T cell proliferation without affecting cell survival. Further study showed that deficiency of PLCγ1 impaired pre-TCR mediated Ca2+ flux and Erk activation. Thus our studies demonstrate that PLCγ1 is important for pre-TCR mediated signal transduction and pre-T cell development.
Collapse
Affiliation(s)
- Guoping Fu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Mei Yu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Yuhong Chen
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Yongwei Zheng
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Wen Zhu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Interdisciplinary Program in Biomedical Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Debra K Newman
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Demin Wang
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Renren Wen
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
40
|
Starling S, Jolly C. LFA-1 Engagement Triggers T Cell Polarization at the HIV-1 Virological Synapse. J Virol 2016; 90:9841-9854. [PMID: 27558417 PMCID: PMC5068534 DOI: 10.1128/jvi.01152-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/15/2016] [Indexed: 12/01/2022] Open
Abstract
HIV-1 efficiently disseminates by cell-cell spread at intercellular contacts called virological synapses (VS), where the virus preferentially assembles and buds. Cell-cell contact triggers active polarization of organelles and viral proteins within infected cells to the contact site to support efficient VS formation and HIV-1 spread; critically, however, which cell surface protein triggers contact-induced polarization at the VS remains unclear. Additionally, the mechanism by which the HIV-1 envelope glycoprotein (Env) is recruited to the VS remains ill defined. Here, we use a reductionist bead-coupled antibody assay as a model of the VS and show that cross-linking the integrin LFA-1 alone is sufficient to induce active T cell polarization and recruitment of the microtubule organizing center (MTOC) in HIV-1-infected cells. Mutant cell lines coupled with inhibitors demonstrated that LFA-1-induced polarization was dependent on the T cell kinase ZAP70. Notably, immunofluorescent staining of viral proteins revealed an accumulation of surface Env at sites of LFA-1 engagement, with intracellular Env localized to a Golgi compartment proximal to the polarized MTOC. Furthermore, blocking LFA-1-induced MTOC polarization through ZAP70 inhibition prevented intracellular Env polarization. Taken together, these data reveal that LFA-1 is a key determinant in inducing dynamic T cell remodeling to the VS and suggest a model in which LFA-1 engagement triggers active polarization of the MTOC and the associated Env-containing secretory apparatus to sites of cell-cell contact to support polarized viral assembly and egress for efficient cell-cell spread. IMPORTANCE HIV-1 causes AIDS by spreading within immune cells and depletion of CD4 T lymphocytes. Rapid spread between these cells occurs by highly efficient cell-cell transmission that takes place at virological synapses (VS). VS are characterized by striking T cell remodeling that is spatially associated with polarized virus assembly and budding at sites of cell contact. Here, we show that the integrin LFA-1 triggers organelle polarization and viral protein recruitment, facilitating formation of the VS, and that this requires the T cell kinase ZAP70. Taken together, these data suggest a mechanism by which HIV-1-infected T cells sense and respond to cell contact to polarize viral egress and promote cell-cell spread. Understanding how cell-cell spread is regulated may help reveal therapeutic targets to specifically block this mode of HIV-1 dissemination.
Collapse
Affiliation(s)
- Shimona Starling
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
41
|
Burdeos GC, Ito J, Eitsuka T, Nakagawa K, Kimura F, Miyazawa T. δ and γ tocotrienols suppress human hepatocellular carcinoma cell proliferation via regulation of Ras-Raf-MEK-ERK pathway-associated upstream signaling. Food Funct 2016; 7:4170-4174. [DOI: 10.1039/c6fo00826g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
42
|
The Immune Adaptor SLP-76 Binds to SUMO-RANGAP1 at Nuclear Pore Complex Filaments to Regulate Nuclear Import of Transcription Factors in T Cells. Mol Cell 2015; 59:840-9. [PMID: 26321253 PMCID: PMC4576164 DOI: 10.1016/j.molcel.2015.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 06/01/2015] [Accepted: 07/17/2015] [Indexed: 12/03/2022]
Abstract
While immune cell adaptors regulate proximal T cell signaling, direct regulation of the nuclear pore complex (NPC) has not been reported. NPC has cytoplasmic filaments composed of RanGAP1 and RanBP2 with the potential to interact with cytoplasmic mediators. Here, we show that the immune cell adaptor SLP-76 binds directly to SUMO-RanGAP1 of cytoplasmic fibrils of the NPC, and that this interaction is needed for optimal NFATc1 and NF-κB p65 nuclear entry in T cells. Transmission electron microscopy showed anti-SLP-76 cytoplasmic labeling of the majority of NPCs in anti-CD3 activated T cells. Further, SUMO-RanGAP1 bound to the N-terminal lysine 56 of SLP-76 where the interaction was needed for optimal RanGAP1-NPC localization and GAP exchange activity. While the SLP-76-RanGAP1 (K56E) mutant had no effect on proximal signaling, it impaired NF-ATc1 and p65/RelA nuclear entry and in vivo responses to OVA peptide. Overall, we have identified SLP-76 as a direct regulator of nuclear pore function in T cells. Immune adaptor SLP-76 binds to SUMO-RanGAP1 of cytoplasmic fibrils of the NPC SLP-76 K-56 binding needed for optimal RanGAP1 localization and exchange activity SLP-76 K56E mutant impaired NF-ATc1 and NFκB p65 (RelA) nuclear entry Immune adaptors directly regulate nuclear entry of transcription factors in T cells
Collapse
|
43
|
Schmidt AM, Lu W, Sindhava VJ, Huang Y, Burkhardt JK, Yang E, Riese MJ, Maltzman JS, Jordan MS, Kambayashi T. Regulatory T cells require TCR signaling for their suppressive function. THE JOURNAL OF IMMUNOLOGY 2015; 194:4362-70. [PMID: 25821220 DOI: 10.4049/jimmunol.1402384] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 03/02/2015] [Indexed: 01/23/2023]
Abstract
Regulatory T cells (Tregs) are a subset of CD4(+) T cells that maintain immune tolerance in part by their ability to inhibit the proliferation of conventional CD4(+) T cells (Tconvs). The role of the TCR and the downstream signaling pathways required for this suppressive function of Tregs are not fully understood. To yield insight into how TCR-mediated signals influence Treg suppressive function, we assessed the ability of Tregs with altered TCR-mediated signaling capacity to inhibit Tconv proliferation. Mature Tregs deficient in Src homology 2 domain containing leukocyte protein of 76 kDa (SLP-76), an adaptor protein that nucleates the proximal signaling complex downstream of the TCR, were unable to inhibit Tconv proliferation, suggesting that TCR signaling is required for Treg suppressive function. Moreover, Tregs with defective phospholipase C γ (PLCγ) activation due to a Y145F mutation of SLP-76 were also defective in their suppressive function. Conversely, enhancement of diacylglycerol-mediated signaling downstream of PLCγ by genetic ablation of a negative regulator of diacylglycerol kinase ζ increased the suppressive ability of Tregs. Because SLP-76 is also important for integrin activation and signaling, we tested the role of integrin activation in Treg-mediated suppression. Tregs lacking the adaptor proteins adhesion and degranulation promoting adapter protein or CT10 regulator of kinase/CT10 regulator of kinase-like, which are required for TCR-mediated integrin activation, inhibited Tconv proliferation to a similar extent as wild-type Tregs. Together, these data suggest that TCR-mediated PLCγ activation, but not integrin activation, is required for Tregs to inhibit Tconv proliferation.
Collapse
Affiliation(s)
- Amanda M Schmidt
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Wen Lu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Vishal J Sindhava
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Yanping Huang
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104; and
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104; and
| | - Enjun Yang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew J Riese
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jonathan S Maltzman
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Martha S Jordan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
44
|
Modulation of TCR responsiveness by the Grb2-family adaptor, Gads. Cell Signal 2014; 27:125-34. [PMID: 25452106 DOI: 10.1016/j.cellsig.2014.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 10/16/2014] [Indexed: 12/11/2022]
Abstract
T cell antigen receptor (TCR) signaling depends on three interacting adaptor proteins: SLP-76, Gads, and LAT. Their mechanisms of signaling have been extensively explored, with the aid of fortuitously isolated LAT- and SLP-76-deficient T cell lines, but no such tools were available for Gads, a Grb2-family adaptor that bridges the TCR-inducible interaction between SLP-76 and LAT. TALEN-directed genome editing was applied to disrupt the first coding exon of human Gads in the Jurkat T cell line. Gads was dispensable for TCR-induced phosphorylation of SLP-76, but was a dose-dependent amplifier of TCR-induced CD69 expression. Gads conferred responsiveness to weak TCR stimuli, leading to PLC-γ1 phosphorylation and calcium flux. TALEN-derived, Gads-deficient T cell lines provide a uniquely tractable genetic platform for exploring its regulatory features, such as Gads phosphorylation at T262, which we observed by mass spectrometry. Upon mutation of this site, TCR responsiveness and sensitivity to weak TCR stimuli were increased. This study demonstrates the feasibility of TALEN-based reverse genetics in Jurkat T cells, while enriching our understanding of Gads as a regulated modulator of TCR sensitivity.
Collapse
|
45
|
Ji Q, Ding Y, Salomon AR. SRC homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) N-terminal tyrosine residues regulate a dynamic signaling equilibrium involving feedback of proximal T-cell receptor (TCR) signaling. Mol Cell Proteomics 2014; 14:30-40. [PMID: 25316710 DOI: 10.1074/mcp.m114.037861] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
SRC homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) is a cytosolic adaptor protein that plays an important role in the T-cell receptor-mediated T-cell signaling pathway. SLP-76 links proximal receptor stimulation to downstream effectors through interaction with many signaling proteins. Previous studies showed that mutation of three tyrosine residues, Tyr(112), Tyr(128), and Tyr(145), in the N terminus of SLP-76 results in severely impaired phosphorylation and activation of Itk and PLCγ1, which leads to defective calcium mobilization, Erk activation, and NFAT activation. To expand our knowledge of the role of N-terminal phosphorylation of SLP-76 from these three tyrosine sites, we characterized nearly 1000 tyrosine phosphorylation sites via mass spectrometry in SLP-76 reconstituted wild-type cells and SLP-76 mutant cells in which three tyrosine residues were replaced with phenylalanines (Y3F mutant). Mutation of the three N-terminal tyrosine residues of SLP-76 phenocopied SLP-76-deficient cells for the majority of tyrosine phosphorylation sites observed, including feedback on proximal T-cell receptor signaling proteins. Meanwhile, reversed phosphorylation changes were observed on Tyr(192) of Lck when we compared mutants to the complete removal of SLP-76. In addition, N-terminal tyrosine sites of SLP-76 also perturbed phosphorylation of Tyr(440) of Fyn, Tyr(702) of PLCγ1, Tyr(204), Tyr(397), and Tyr(69) of ZAP-70, revealing new modes of regulation on these sites. All these findings confirmed the central role of N-terminal tyrosine sites of SLP-76 in the pathway and also shed light on novel signaling events that are uniquely regulated by SLP-76 N-terminal tyrosine residues.
Collapse
Affiliation(s)
- Qinqin Ji
- From the ‡Department of Chemistry, Brown University Providence, RI 02903
| | - Yiyuan Ding
- From the ‡Department of Chemistry, Brown University Providence, RI 02903
| | - Arthur R Salomon
- From the ‡Department of Chemistry, Brown University Providence, RI 02903; §Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University Providence, RI 02903
| |
Collapse
|
46
|
Abstract
Rapid progress has recently been made regarding how phospholipase C (PLC)-β functions downstream of G protein-coupled receptors and how PLC-β functions in the nucleus. PLC-β has also been shown to interplay with tyrosine kinase-based signaling pathways, specifically to inhibit Stat5 activation by recruiting the protein-tyrosine phosphatase SHP-1. In this regard, a new multimolecular signaling platform, named SPS complex, has been identified. The SPS complex has important regulatory roles in tumorigenesis and immune cell activation. Furthermore, a growing body of work suggests that PLC-β also participates in the differentiation and activation of immune cells that control both the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Wenbin Xiao
- Department of Pathology, University Hospital Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
47
|
Multipoint binding of the SLP-76 SH2 domain to ADAP is critical for oligomerization of SLP-76 signaling complexes in stimulated T cells. Mol Cell Biol 2013; 33:4140-51. [PMID: 23979596 DOI: 10.1128/mcb.00410-13] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The adapter molecules SLP-76 and LAT play central roles in T cell activation by recruiting enzymes and other adapters into multiprotein complexes that coordinate highly regulated signal transduction pathways. While many of the associated proteins have been characterized, less is known concerning the mechanisms of assembly for these dynamic and potentially heterogeneous signaling complexes. Following T cell receptor (TCR) stimulation, SLP-76 is found in structures called microclusters, which contain many signaling complexes. Previous studies showed that a mutation to the SLP-76 C-terminal SH2 domain nearly abolished SLP-76 microclusters, suggesting that the SH2 domain facilitates incorporation of signaling complexes into microclusters. S. C. Bunnell, A. L. Singer, D. I. Hong, B. H. Jacque, M. S. Jordan, M. C. Seminario, V. A. Barr, G. A. Koretzky, and L. E. Samelson, Mol. Cell. Biol., 26:7155-7166, 2006). Using biophysical methods, we demonstrate that the adapter, ADAP, contains three binding sites for SLP-76, and that multipoint binding to ADAP fragments oligomerizes the SLP-76 SH2 domain in vitro. These results were complemented with confocal imaging and functional studies of cells expressing ADAP with various mutations. Our results demonstrate that all three binding sites are critical for SLP-76 microcluster assembly, but any combination of two sites will partially induce microclusters. These data support a model whereby multipoint binding of SLP-76 to ADAP facilitates the assembly of SLP-76 microclusters. This model has implications for the regulation of SLP-76 and LAT microclusters and, as a result, T cell signaling.
Collapse
|
48
|
Kawakami T, Xiao W. Phospholipase C-β in immune cells. Adv Biol Regul 2013; 53:249-57. [PMID: 23981313 DOI: 10.1016/j.jbior.2013.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 07/29/2013] [Accepted: 08/02/2013] [Indexed: 12/22/2022]
Abstract
Great progress has recently been made in structural and functional research of phospholipase C (PLC)-β. We now understand how PLC-β isoforms (β1-β4) are activated by GTP-bound Gαq downstream of G protein-coupled receptors. Numerous studies indicate that PLC-βs participate in the differentiation and activation of immune cells that control both the innate and adaptive immune systems. The PLC-β3 isoform also interplays with tyrosine kinase-based signaling pathways, to inhibit Stat5 activation by recruiting the protein-tyrosine phosphatase SHP-1, with which PLC-β3 and Stat5 form a multi-molecular signaling platform, named SPS complex. The SPS complex has important regulatory roles in tumorigenesis and immune cell activation.
Collapse
Affiliation(s)
- Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA; Laboratory of Allergic Disease, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan.
| | | |
Collapse
|
49
|
Liu H, Thaker YR, Stagg L, Schneider H, Ladbury JE, Rudd CE. SLP-76 sterile α motif (SAM) and individual H5 α helix mediate oligomer formation for microclusters and T-cell activation. J Biol Chem 2013; 288:29539-49. [PMID: 23935094 PMCID: PMC3795252 DOI: 10.1074/jbc.m112.424846] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the importance of the immune adaptor SLP-76 in T-cell immunity, it has been unclear whether SLP-76 directly self-associates to form higher order oligomers for T-cell activation. In this study, we show that SLP-76 self-associates in response to T-cell receptor ligation as mediated by the N-terminal sterile α motif (SAM) domain. SLP-76 co-precipitated alternately tagged SLP-76 in response to anti-CD3 ligation. Dynamic light scattering and fluorescent microscale thermophoresis of the isolated SAM domain (residues 1–78) revealed evidence of dimers and tetramers. Consistently, deletion of the SAM region eliminated SLP-76 co-precipitation of itself, concurrent with a loss of microcluster formation, nuclear factor of activated T-cells (NFAT) transcription, and interleukin-2 production in Jurkat or primary T-cells. Furthermore, the H5 α helix within the SAM domain contributed to self-association. Retention of H5 in the absence of H1–4 sufficed to support SLP-76 self-association with smaller microclusters that nevertheless enhanced anti-CD3-driven AP1/NFAT transcription and IL-2 production. By contrast, deletion of the H5 α helix impaired self-association and anti-CD3 induced AP1/NFAT transcription. Our data identified for the first time a role for the SAM domain in mediating SLP-76 self-association for T-cell function.
Collapse
Affiliation(s)
- Hebin Liu
- From the Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | | | | | | | | | | |
Collapse
|
50
|
Joseph N, Reicher B, Barda-Saad M. The calcium feedback loop and T cell activation: how cytoskeleton networks control intracellular calcium flux. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:557-68. [PMID: 23860253 DOI: 10.1016/j.bbamem.2013.07.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/30/2013] [Accepted: 07/08/2013] [Indexed: 12/31/2022]
Abstract
During T cell activation, the engagement of a T cell with an antigen-presenting cell (APC) results in rapid cytoskeletal rearrangements and a dramatic increase of intracellular calcium (Ca(2+)) concentration, downstream to T cell antigen receptor (TCR) ligation. These events facilitate the organization of an immunological synapse (IS), which supports the redistribution of receptors, signaling molecules and organelles towards the T cell-APC interface to induce downstream signaling events, ultimately supporting T cell effector functions. Thus, Ca(2+) signaling and cytoskeleton rearrangements are essential for T cell activation and T cell-dependent immune response. Rapid release of Ca(2+) from intracellular stores, e.g. the endoplasmic reticulum (ER), triggers the opening of Ca(2+) release-activated Ca(2+) (CRAC) channels, residing in the plasma membrane. These channels facilitate a sustained influx of extracellular Ca(2+) across the plasma membrane in a process termed store-operated Ca(2+) entry (SOCE). Because CRAC channels are themselves inhibited by Ca(2+) ions, additional factors are suggested to enable the sustained Ca(2+) influx required for T cell function. Among these factors, we focus here on the contribution of the actin and microtubule cytoskeleton. The TCR-mediated increase in intracellular Ca(2+) evokes a rapid cytoskeleton-dependent polarization, which involves actin cytoskeleton rearrangements and microtubule-organizing center (MTOC) reorientation. Here, we review the molecular mechanisms of Ca(2+) flux and cytoskeletal rearrangements, and further describe the way by which the cytoskeletal networks feedback to Ca(2+) signaling by controlling the spatial and temporal distribution of Ca(2+) sources and sinks, modulating TCR-dependent Ca(2+) signals, which are required for an appropriate T cell response. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
Affiliation(s)
- Noah Joseph
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Barak Reicher
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Mira Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|