Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.536
Filter
1.
Annu Rev Immunol ; 36: 127-156, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29237129

ABSTRACT

T cells possess an array of functional capabilities important for host defense against pathogens and tumors. T cell effector functions require the T cell antigen receptor (TCR). The TCR has no intrinsic enzymatic activity, and thus signal transduction from the receptor relies on additional signaling molecules. One such molecule is the cytoplasmic tyrosine kinase ZAP-70, which associates with the TCR complex and is required for initiating the canonical biochemical signal pathways downstream of the TCR. In this article, we describe recent structure-based insights into the regulation and substrate specificity of ZAP-70, and then we review novel methods for determining the role of ZAP-70 catalytic activity-dependent and -independent signals in developing and mature T cells. Lastly, we discuss the disease states in mouse models and humans, which range from immunodeficiency to autoimmunity, that are caused by mutations in ZAP-70.


Subject(s)
Disease Susceptibility , Signal Transduction , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Autoimmunity , Biomarkers , Catalysis , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation , Humans , Immunity , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Phosphorylation , Protein Transport , Structure-Activity Relationship , Substrate Specificity , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/antagonists & inhibitors , ZAP-70 Protein-Tyrosine Kinase/chemistry , ZAP-70 Protein-Tyrosine Kinase/genetics
2.
Cell ; 187(9): 2305-2323.e33, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38614099

ABSTRACT

Cancer immunotherapy has transformed treatment possibilities, but its effectiveness differs significantly among patients, indicating the presence of alternative pathways for immune evasion. Here, we show that ITPRIPL1 functions as an inhibitory ligand of CD3ε, and its expression inhibits T cells in the tumor microenvironment. The binding of ITPRIPL1 extracellular domain to CD3ε on T cells significantly decreased calcium influx and ZAP70 phosphorylation, impeding initial T cell activation. Treatment with a neutralizing antibody against ITPRIPL1 restrained tumor growth and promoted T cell infiltration in mouse models across various solid tumor types. The antibody targeting canine ITPRIPL1 exhibited notable therapeutic efficacy against naturally occurring tumors in pet clinics. These findings highlight the role of ITPRIPL1 (or CD3L1, CD3ε ligand 1) in impeding T cell activation during the critical "signal one" phase. This discovery positions ITPRIPL1 as a promising therapeutic target against multiple tumor types.


Subject(s)
CD3 Complex , Lymphocyte Activation , T-Lymphocytes , Tumor Escape , Tumor Microenvironment , Animals , CD3 Complex/metabolism , CD3 Complex/immunology , Humans , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology , Dogs , Neoplasms/immunology , Cell Line, Tumor , Female , Protein Binding , ZAP-70 Protein-Tyrosine Kinase/metabolism , Antibodies, Neutralizing/immunology , Mice, Inbred C57BL
3.
Nat Immunol ; 23(9): 1355-1364, 2022 09.
Article in English | MEDLINE | ID: mdl-36045187

ABSTRACT

T cells recognize a few high-affinity antigens among a vast array of lower affinity antigens. According to the kinetic proofreading model, antigen discrimination properties could be explained by the gradual amplification of small differences in binding affinities as the signal is transduced downstream of the T cell receptor. Which early molecular events are affected by ligand affinity, and how, has not been fully resolved. Here, we used time-resolved high-throughput proteomic analyses to identify and quantify the phosphorylation events and protein-protein interactions encoding T cell ligand discrimination in antigen-experienced T cells. Although low-affinity ligands induced phosphorylation of the Cd3 chains of the T cell receptor and the interaction of Cd3 with the Zap70 kinase as strongly as high-affinity ligands, they failed to activate Zap70 to the same extent. As a result, formation of the signalosome of the Lat adaptor was severely impaired with low- compared with high-affinity ligands, whereas formation of the signalosome of the Cd6 receptor was affected only partially. Overall, this study provides a comprehensive map of molecular events associated with T cell ligand discrimination.


Subject(s)
Proteomics , T-Lymphocytes , Antigens/metabolism , Kinetics , Ligands , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism
4.
Cell ; 169(1): 108-119.e20, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340336

ABSTRACT

A T cell mounts an immune response by measuring the binding strength of its T cell receptor (TCR) for peptide-loaded MHCs (pMHC) on an antigen-presenting cell. How T cells convert the lifetime of the extracellular TCR-pMHC interaction into an intracellular signal remains unknown. Here, we developed a synthetic signaling system in which the extracellular domains of the TCR and pMHC were replaced with short hybridizing strands of DNA. Remarkably, T cells can discriminate between DNA ligands differing by a single base pair. Single-molecule imaging reveals that signaling is initiated when single ligand-bound receptors are converted into clusters, a time-dependent process requiring ligands with longer bound times. A computation model reveals that receptor clustering serves a kinetic proofreading function, enabling ligands with longer bound times to have disproportionally greater signaling outputs. These results suggest that spatial reorganization of receptors plays an important role in ligand discrimination in T cell signaling.


Subject(s)
Ligands , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism , DNA/metabolism , Humans , Jurkat Cells , Phosphorylation , Single Molecule Imaging , ZAP-70 Protein-Tyrosine Kinase/analysis
5.
Nat Immunol ; 20(11): 1481-1493, 2019 11.
Article in English | MEDLINE | ID: mdl-31611699

ABSTRACT

Self-non-self discrimination is central to T cell-mediated immunity. The kinetic proofreading model can explain T cell antigen receptor (TCR) ligand discrimination; however, the rate-limiting steps have not been identified. Here, we show that tyrosine phosphorylation of the T cell adapter protein LAT at position Y132 is a critical kinetic bottleneck for ligand discrimination. LAT phosphorylation at Y132, mediated by the kinase ZAP-70, leads to the recruitment and activation of phospholipase C-γ1 (PLC-γ1), an important effector molecule for T cell activation. The slow phosphorylation of Y132, relative to other phosphosites on LAT, is governed by a preceding glycine residue (G131) but can be accelerated by substituting this glycine with aspartate or glutamate. Acceleration of Y132 phosphorylation increases the speed and magnitude of PLC-γ1 activation and enhances T cell sensitivity to weaker stimuli, including weak agonists and self-peptides. These observations suggest that the slow phosphorylation of Y132 acts as a proofreading step to facilitate T cell ligand discrimination.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Lymphocyte Activation , Membrane Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Female , Ligands , Male , Membrane Proteins/immunology , Mice , Phospholipase C gamma/metabolism , Phosphorylation/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/metabolism , Tyrosine/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism
6.
Nat Immunol ; 19(7): 733-741, 2018 07.
Article in English | MEDLINE | ID: mdl-29915297

ABSTRACT

T cell-antigen receptor (TCR) signaling requires the sequential activities of the kinases Lck and Zap70. Upon TCR stimulation, Lck phosphorylates the TCR, thus leading to the recruitment, phosphorylation, and activation of Zap70. Lck binds and stabilizes phosho-Zap70 by using its SH2 domain, and Zap70 phosphorylates the critical adaptors LAT and SLP76, which coordinate downstream signaling. It is unclear whether phosphorylation of these adaptors occurs through passive diffusion or active recruitment. We report the discovery of a conserved proline-rich motif in LAT that mediates efficient LAT phosphorylation. Lck associates with this motif via its SH3 domain, and with phospho-Zap70 via its SH2 domain, thereby acting as a molecular bridge that facilitates the colocalization of Zap70 and LAT. Elimination of this proline-rich motif compromises TCR signaling and T cell development. These results demonstrate the remarkable multifunctionality of Lck, wherein each of its domains has evolved to orchestrate a distinct step in TCR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Proteins/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Motifs , Animals , HEK293 Cells , Humans , Jurkat Cells , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Phosphorylation , Proline/analysis , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/immunology
8.
Nat Immunol ; 18(1): 86-95, 2017 01.
Article in English | MEDLINE | ID: mdl-27869819

ABSTRACT

Cell-surface-receptor pathways amplify weak, rare and local stimuli to induce cellular responses. This task is accomplished despite signaling components that segregate into nanometer-scale membrane domains. Here we describe a 'catch-and-release' mechanism that amplified and dispersed stimuli by releasing activated kinases from receptors lacking intrinsic catalytic activity. Specifically, we discovered a cycle of recruitment, activation and release for Zap70 kinases at phosphorylated T cell antigen receptors (TCRs). This turned the TCR into a 'catalytic unit' that amplified antigenic stimuli. Zap70 released from the TCR remained at the membrane, translocated, and phosphorylated spatially distinct substrates. The mechanisms described here are based on widely used protein domains and post-translational modifications; therefore, many membrane-associated pathways might employ similar mechanisms for signal amplification and dispersion.


Subject(s)
Activity Cycles , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens/immunology , HEK293 Cells , Humans , Jurkat Cells , Lymphocyte Activation , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Phosphoproteins/metabolism , Receptor Cross-Talk , Receptors, Antigen, T-Cell/genetics
9.
Nat Immunol ; 18(2): 196-204, 2017 02.
Article in English | MEDLINE | ID: mdl-27941787

ABSTRACT

Calcineurin is a phosphatase whose primary targets in T cells are NFAT transcription factors, and inhibition of calcineurin activity by treatment with cyclosporin A (CsA) or FK506 is a cornerstone of immunosuppressive therapies. Here we found that calcineurin was recruited to the T cell antigen receptor (TCR) signaling complex, where it reversed inhibitory phosphorylation of the tyrosine kinase Lck on Ser59 (LckS59). Loss of calcineurin activity impaired phosphorylation of Tyr493 of the tyrosine kinase ZAP-70 (ZAP-70Y493), as well as some downstream pathways in a manner consistent with signaling in cells expressing LckS59A (Lck that cannot be phosphorylated) or LckS59E (a phosphomimetic mutant). Notably, CsA inhibited integrin-LFA-1-dependent and NFAT-independent adhesion of T cells to the intercellular adhesion molecule ICAM-1, with little effect on cells expressing mutant Lck. These results provide new understanding of how widely used immunosuppressive drugs interfere with essential processes in the immune response.


Subject(s)
Calcineurin/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Cell Adhesion/drug effects , Cyclosporine/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Lymphocyte Activation/drug effects , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Binding , Signal Transduction , T-Lymphocytes/drug effects , Tacrolimus/pharmacology
10.
Mol Cell ; 81(10): 2094-2111.e9, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33878293

ABSTRACT

Even though SYK and ZAP70 kinases share high sequence homology and serve analogous functions, their expression in B and T cells is strictly segregated throughout evolution. Here, we identified aberrant ZAP70 expression as a common feature in a broad range of B cell malignancies. We validated SYK as the kinase that sets the thresholds for negative selection of autoreactive and premalignant clones. When aberrantly expressed in B cells, ZAP70 competes with SYK at the BCR signalosome and redirects SYK from negative selection to tonic PI3K signaling, thereby promoting B cell survival. In genetic mouse models for B-ALL and B-CLL, conditional expression of Zap70 accelerated disease onset, while genetic deletion impaired malignant transformation. Inducible activation of Zap70 during B cell development compromised negative selection of autoreactive B cells, resulting in pervasive autoantibody production. Strict segregation of the two kinases is critical for normal B cell selection and represents a central safeguard against the development of autoimmune disease and B cell malignancies.


Subject(s)
Autoimmunity , Neoplasms/enzymology , Neoplasms/prevention & control , Syk Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Antigens, CD19/metabolism , B-Lymphocytes , Calcium/metabolism , Cell Differentiation , Cell Transformation, Neoplastic , Enzyme Activation , Humans , Immune Tolerance , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/pathology , Mice , Models, Genetic , NFATC Transcription Factors/metabolism , Neoplasm Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
11.
Nat Immunol ; 17(5): 574-582, 2016 May.
Article in English | MEDLINE | ID: mdl-26998761

ABSTRACT

It has been proposed that the local segregation of kinases and the tyrosine phosphatase CD45 underpins T cell antigen receptor (TCR) triggering, but how such segregation occurs and whether it can initiate signaling is unclear. Using structural and biophysical analysis, we show that the extracellular region of CD45 is rigid and extends beyond the distance spanned by TCR-ligand complexes, implying that sites of TCR-ligand engagement would sterically exclude CD45. We also show that the formation of 'close contacts', new structures characterized by spontaneous CD45 and kinase segregation at the submicron-scale, initiates signaling even when TCR ligands are absent. Our work reveals the structural basis for, and the potent signaling effects of, local CD45 and kinase segregation. TCR ligands have the potential to heighten signaling simply by holding receptors in close contacts.


Subject(s)
Leukocyte Common Antigens/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Crystallography, X-Ray , HEK293 Cells , Humans , Jurkat Cells , Leukocyte Common Antigens/chemistry , Leukocyte Common Antigens/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Microscopy, Electron , Microscopy, Fluorescence/methods , Models, Molecular , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Time Factors , ZAP-70 Protein-Tyrosine Kinase/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
12.
Nat Immunol ; 17(3): 286-96, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829767

ABSTRACT

SHARPIN forms a linear-ubiquitin-chain-assembly complex that promotes signaling via the transcription factor NF-κB. SHARPIN deficiency leads to progressive multi-organ inflammation and immune system malfunction, but how SHARPIN regulates T cell responses is unclear. Here we found that SHARPIN deficiency resulted in a substantial reduction in the number of and defective function of regulatory T cells (Treg cells). Transfer of SHARPIN-sufficient Treg cells into SHARPIN-deficient mice considerably alleviated their systemic inflammation. SHARPIN-deficient T cells displayed enhanced proximal signaling via the T cell antigen receptor (TCR) without an effect on the activation of NF-κB. SHARPIN conjugated with Lys63 (K63)-linked ubiquitin chains, which led to inhibition of the association of TCRζ with the signaling kinase Zap70; this affected the generation of Treg cells. Our study therefore identifies a role for SHARPIN in TCR signaling whereby it maintains immunological homeostasis and tolerance by regulating Treg cells.


Subject(s)
Carrier Proteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Colitis/immunology , Cytokines/immunology , Female , Flow Cytometry , Humans , Immune Tolerance/immunology , Immunoblotting , Immunoprecipitation , In Vitro Techniques , Inflammation , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/immunology , Signal Transduction , Ubiquitination , ZAP-70 Protein-Tyrosine Kinase/immunology
13.
Nat Immunol ; 16(9): 961-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26237552

ABSTRACT

Kinase recruitment to membrane receptors is essential for signal transduction. However, the underlying regulatory mechanisms are poorly understood. We investigated how conformational changes control T cell receptor (TCR) association and activity of the kinase Zap70. Structural analysis showed that TCR binding or phosphorylation of Zap70 triggers a transition from a closed, autoinhibited conformation to an open conformation. Using Zap70 mutants with defined conformations, we found that TCR dwell times controlled Zap70 activity. The closed conformation minimized TCR dwell times and thereby prevented activation by membrane-associated kinases. Parallel recruitment of coreceptor-associated Lck kinase to the TCR ensured Zap70 phosphorylation and stabilized Zap70 TCR binding. Our study suggests that the dynamics of cytosolic enzyme recruitment to the plasma membrane regulate the activity and function of receptors lacking intrinsic catalytic activity.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , CD3 Complex/metabolism , Cell Membrane/metabolism , Deuterium Exchange Measurement , Humans , Mass Spectrometry , Molecular Dynamics Simulation , Mutation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Time Factors , ZAP-70 Protein-Tyrosine Kinase/genetics
14.
Nat Immunol ; 16(12): 1253-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26390156

ABSTRACT

The key molecular mechanisms that control signaling via T cell antigen receptors (TCRs) remain to be fully elucidated. Here we found that Nrdp1, a ring finger-type E3 ligase, mediated Lys33 (K33)-linked polyubiquitination of the signaling kinase Zap70 and promoted the dephosphorylation of Zap70 by the acidic phosphatase-like proteins Sts1 and Sts2 and thereby terminated early TCR signaling in CD8(+) T cells. Nrdp1 deficiency significantly promoted the activation of naive CD8(+) T cells but not that of naive CD4(+) T cells after engagement of the TCR. Nrdp1 interacted with Zap70 and with Sts1 and Sts2 and connected K33 linkage of Zap70 to Sts1- and Sts2-mediated dephosphorylation. Our study suggests that Nrdp1 terminates early TCR signaling by inactivating Zap70 and provides new mechanistic insights into the non-proteolytic regulation of TCR signaling by E3 ligases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/immunology , Lymphocyte Activation/immunology , Lysine/immunology , ZAP-70 Protein-Tyrosine Kinase/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Lymphocyte Activation/genetics , Lysine/genetics , Lysine/metabolism , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phosphorylation/immunology , Polyubiquitin/immunology , Polyubiquitin/metabolism , Protein Binding/immunology , RNA Interference , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptome/genetics , Transcriptome/immunology , Ubiquitin-Protein Ligases , Ubiquitination/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
15.
Nat Immunol ; 15(7): 687-94, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24908390

ABSTRACT

The catalytic activity of Zap70 is crucial for T cell antigen receptor (TCR) signaling, but the quantitative and temporal requirements for its function in thymocyte development are not known. Using a chemical-genetic system to selectively and reversibly inhibit Zap70 catalytic activity in a model of synchronized thymic selection, we showed that CD4(+)CD8(+) thymocytes integrate multiple, transient, Zap70-dependent signals over more than 36 h to reach a cumulative threshold for positive selection, whereas 1 h of signaling was sufficient for negative selection. Titration of Zap70 activity resulted in graded reductions in positive and negative selection but did not decrease the cumulative TCR signals integrated by positively selected OT-I cells, which revealed heterogeneity, even among CD4(+)CD8(+) thymocytes expressing identical TCRs undergoing positive selection.


Subject(s)
T-Lymphocytes/physiology , ZAP-70 Protein-Tyrosine Kinase/physiology , Animals , Calcium/metabolism , Catalysis , Cell Differentiation , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred C57BL , Protein-Tyrosine Kinases/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction , Syk Kinase
16.
Nat Immunol ; 15(1): 80-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24241693

ABSTRACT

The subclassification of immunology into innate and adaptive immunity is challenged by innate-like T lymphocytes that use innate receptors to respond rapidly to stress despite expressing T cell antigen receptors (TCRs), a hallmark of adaptive immunity. In studies that explain how such cells can straddle innate and adaptive immunity, we found that signaling via antigen receptors, whose conventional role is to facilitate clonal T cell activation, was critical for the development of innate-like T cells but then was rapidly attenuated, which accommodated the cells' innate responsiveness. These findings permitted the identification of a previously unknown innate-like T cell subset and indicate that T cell hyporesponsiveness, a state traditionally linked to tolerance, may be fundamental to T cells entering the innate compartment and thereby providing lymphoid stress surveillance.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Animals , Animals, Newborn , Cells, Cultured , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction/immunology , T-Lymphocyte Subsets/metabolism , ZAP-70 Protein-Tyrosine Kinase/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
17.
Nat Immunol ; 15(4): 384-392, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24584089

ABSTRACT

T cell antigen receptor (TCR)-mediated activation of T cells requires the interaction of dozens of proteins. Here we used quantitative mass spectrometry and activated primary CD4(+) T cells from mice in which a tag for affinity purification was knocked into several genes to determine the composition and dynamics of multiprotein complexes that formed around the kinase Zap70 and the adaptors Lat and SLP-76. Most of the 112 high-confidence time-resolved protein interactions we observed were previously unknown. The surface receptor CD6 was able to initiate its own signaling pathway by recruiting SLP-76 and the guanine nucleotide-exchange factor Vav1 regardless of the presence of Lat. Our findings provide a more complete model of TCR signaling in which CD6 constitutes a signaling hub that contributes to the diversification of TCR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/immunology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , T-Lymphocyte Subsets/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Calcium Signaling/genetics , Cells, Cultured , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/metabolism , Phosphoproteins/genetics , Protein Binding/genetics , Proteomics , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Antigen, T-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism
18.
Nat Immunol ; 14(7): 741-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685786

ABSTRACT

Functionally diverse T cell populations interact to maintain homeostasis of the immune system. We found that human and mouse antigen-activated T cells with high expression of the lymphocyte surface marker CD52 suppressed other T cells. CD52(hi)CD4(+) T cells were distinct from CD4(+)CD25(+)Foxp3(+) regulatory T cells. Their suppression was mediated by soluble CD52 released by phospholipase C. Soluble CD52 bound to the inhibitory receptor Siglec-10 and impaired phosphorylation of the T cell receptor-associated kinases Lck and Zap70 and T cell activation. Humans with type 1 diabetes had a lower frequency and diminished function of CD52(hi)CD4(+) T cells responsive to the autoantigen GAD65. In diabetes-prone mice of the nonobese diabetic (NOD) strain, transfer of lymphocyte populations depleted of CD52(hi) cells resulted in a substantially accelerated onset of diabetes. Our studies identify a ligand-receptor mechanism of T cell regulation that may protect humans and mice from autoimmune disease.


Subject(s)
Antigens, CD/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Glycoproteins/immunology , Lymphocyte Activation/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Autoantigens/immunology , CD52 Antigen , Female , Flow Cytometry , Glycoproteins/genetics , Homeostasis/immunology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation/immunology , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , ZAP-70 Protein-Tyrosine Kinase/immunology
19.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35197288

ABSTRACT

Protein-protein binding domains are critical in signaling networks. Src homology 2 (SH2) domains are binding domains that interact with sequences containing phosphorylated tyrosines. A subset of SH2 domain-containing proteins has tandem domains, which are thought to enhance binding affinity and specificity. However, a trade-off exists between long-lived binding and the ability to rapidly reverse signaling, which is a critical requirement of noise-filtering mechanisms such as kinetic proofreading. Here, we use modeling to show that the unbinding rate of tandem, but not single, SH2 domains can be accelerated by phosphatases. Using surface plasmon resonance, we show that the phosphatase CD45 can accelerate the unbinding rate of zeta chain-associated protein kinase 70 (ZAP70), a tandem SH2 domain-containing kinase, from biphosphorylated peptides from the T cell receptor (TCR). An important functional prediction of accelerated unbinding is that the intracellular ZAP70-TCR half-life in T cells will not be fixed but rather, dependent on the extracellular TCR-antigen half-life, and we show that this is the case in both cell lines and primary T cells. The work highlights that tandem SH2 domains can break the trade-off between signal fidelity (requiring long half-life) and signal reversibility (requiring short half-life), which is a key requirement for T cell antigen discrimination mediated by kinetic proofreading.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Allosteric Regulation , Half-Life , Humans , Kinetics , Phosphorylation , Protein Binding
20.
J Clin Immunol ; 44(6): 131, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775840

ABSTRACT

RHOH, an atypical small GTPase predominantly expressed in hematopoietic cells, plays a vital role in immune function. A deficiency in RHOH has been linked to epidermodysplasia verruciformis, lung disease, Burkitt lymphoma and T cell defects. Here, we report a novel germline homozygous RHOH c.245G > A (p.Cys82Tyr) variant in a 21-year-old male suffering from recurrent, invasive, opportunistic infections affecting the lungs, eyes, and brain. His sister also succumbed to a lung infection during early adulthood. The patient exhibited a persistent decrease in CD4+ T, B, and NK cell counts, and hypoimmunoglobulinemia. The patient's T cell showed impaired activation upon in vitro TCR stimulation. In Jurkat T cells transduced with RHOHC82Y, a similar reduction in activation marker CD69 up-regulation was observed. Furthermore, the C82Y variant showed reduced RHOH protein expression and impaired interaction with the TCR signaling molecule ZAP70. Together, these data suggest that the newly identified autosomal-recessive RHOH variant is associated with T cell dysfunction and recurrent opportunistic infections, functioning as a hypomorph by disrupting ZAP70-mediated TCR signaling.


Subject(s)
Homozygote , Opportunistic Infections , Humans , Male , Young Adult , Jurkat Cells , Lymphocyte Activation/genetics , Opportunistic Infections/genetics , Opportunistic Infections/immunology , Pedigree , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recurrence , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL