Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Commun ; 15(1): 1583, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383515

ABSTRACT

Peripheral T cell lymphomas are typically aggressive with a poor prognosis. Unlike other hematologic malignancies, the lack of target antigens to discriminate healthy from malignant cells limits the efficacy of immunotherapeutic approaches. The T cell receptor expresses one of two highly homologous chains [T cell receptor ß-chain constant (TRBC) domains 1 and 2] in a mutually exclusive manner, making it a promising target. Here we demonstrate specificity redirection by rational design using structure-guided computational biology to generate a TRBC2-specific antibody (KFN), complementing the antibody previously described by our laboratory with unique TRBC1 specificity (Jovi-1) in targeting broader spectrum of T cell malignancies clonally expressing either of the two chains. This permits generation of paired reagents (chimeric antigen receptor-T cells) specific for TRBC1 and TRBC2, with preclinical evidence to support their efficacy in T cell malignancies.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , Immunotherapy , Receptors, Antigen, T-Cell
2.
ACS Chem Biol ; 19(2): 308-324, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38243811

ABSTRACT

A versatile, safe, and effective small-molecule control system is highly desirable for clinical cell therapy applications. Therefore, we developed a two-component small-molecule control system based on the disruption of protein-protein interactions using minocycline, an FDA-approved antibiotic with wide availability, excellent biodistribution, and low toxicity. The system comprises an anti-minocycline single-domain antibody (sdAb) and a minocycline-displaceable cyclic peptide. Here, we show how this versatile system can be applied to OFF-switch split CAR systems (MinoCAR) and universal CAR adaptors (MinoUniCAR) with reversible, transient, and dose-dependent suppression; to a tunable T cell activation module based on MyD88/CD40 signaling; to a controllable cellular payload secretion system based on IL12 KDEL retention; and as a cell/cell inducible junction. This work represents an important step forward in the development of a remote-controlled system to precisely control the timing, intensity, and safety of therapeutic interventions.


Subject(s)
Cell Communication , Minocycline , Minocycline/pharmacology , Tissue Distribution , Anti-Bacterial Agents/pharmacology , Signal Transduction
4.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37399355

ABSTRACT

BACKGROUND: We used a proliferating ligand (APRIL) to construct a ligand-based third generation chimeric antigen receptor (CAR) able to target two myeloma antigens, B-cell maturation antigen (BCMA) and transmembrane activator and CAML interactor. METHODS: The APRIL CAR was evaluated in a Phase 1 clinical trial (NCT03287804, AUTO2) in patients with relapsed, refractory multiple myeloma. Eleven patients received 13 doses, the first 15×106 CARs, and subsequent patients received 75,225,600 and 900×106 CARs in a 3+3 escalation design. RESULTS: The APRIL CAR was well tolerated. Five (45.5%) patients developed Grade 1 cytokine release syndrome and there was no neurotoxicity. However, responses were only observed in 45.5% patients (1×very good partial response, 3×partial response, 1×minimal response). Exploring the mechanistic basis for poor responses, we then compared the APRIL CAR to two other BCMA CARs in a series of in vitro assays, observing reduced interleukin-2 secretion and lack of sustained tumor control by APRIL CAR regardless of transduction method or co-stimulatory domain. There was also impaired interferon signaling of APRIL CAR and no evidence of autoactivation. Thus focusing on APRIL itself, we confirmed similar affinity to BCMA and protein stability in comparison to BCMA CAR binders but reduced binding by cell-expressed APRIL to soluble BCMA and reduced avidity to tumor cells. This indicated either suboptimal folding or stability of membrane-bound APRIL attenuating CAR activation. CONCLUSIONS: The APRIL CAR was well tolerated, but the clinical responses observed in AUTO2 were disappointing. Subsequently, when comparing the APRIL CAR to other BCMA CARs, we observed in vitro functional deficiencies due to reduced target binding by cell-expressed ligand.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Multiple Myeloma/drug therapy , Ligands , B-Cell Maturation Antigen/metabolism , B-Cell Maturation Antigen/therapeutic use , T-Lymphocytes
5.
Cancer Immunol Res ; 11(9): 1203-1221, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37352396

ABSTRACT

Adoptive T-cell therapy aims to achieve lasting tumor clearance, requiring enhanced engraftment and survival of the immune cells. Cytokines are paramount modulators of T-cell survival and proliferation. Cytokine receptors signal via ligand-induced dimerization, and this principle has been hijacked utilizing nonnative dimerization domains. A major limitation of current technologies resides in the absence of a module that recapitulates the natural cytokine receptor heterodimeric pairing. To circumvent this, we created a new engineered cytokine receptor able to constitutively recreate receptor-heterodimer utilizing the heterodimerization domain derived from the IgG1 antibody (dFab_CCR). We found that the signal delivered by the dFab_CCR-IL2 proficiently mimicked the cytokine receptor heterodimerization, with transcriptomic signatures like those obtained by activation of the native IL2 receptor. Moreover, we found that this dimerization structure was agnostic, efficiently activating signaling through four cytokine receptor families. Using a combination of in vivo and in vitro screening approaches, we characterized a library of 18 dFab_CCRs coexpressed with a clinically relevant solid tumor-specific GD2-specific chimeric antigen receptor (CAR). Based on this characterization, we suggest that the coexpression of either the common ß-chain GMCSF or the IL18 dFab_CCRs is optimal to improve CAR T-cell expansion, engraftment, and efficacy. Our results demonstrate how Fab dimerization is efficient and versatile in recapitulating a cytokine receptor heterodimerization signal. This module could be applied for the enhancement of adoptive T-cell therapies, as well as therapies based on other immune cell types. Furthermore, these results provide a choice of cytokine signal to incorporate with adoptive T-cell therapies.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Cytokine , Neoplasms/pathology , Cytokines
6.
Mol Ther Nucleic Acids ; 32: 603-621, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37200859

ABSTRACT

The hostile tumor microenvironment limits the efficacy of adoptive cell therapies. Activation of the Fas death receptor initiates apoptosis and disrupting these receptors could be key to increasing CAR T cell efficacy. We screened a library of Fas-TNFR proteins identifying several novel chimeras that not only prevented Fas ligand-mediated kill, but also enhanced CAR T cell efficacy by signaling synergistically with the CAR. Upon binding Fas ligand, Fas-CD40 activated the NF-κB pathway, inducing greatest proliferation and IFN-γ release out of all Fas-TNFRs tested. Fas-CD40 induced profound transcriptional modifications, particularly genes relating to the cell cycle, metabolism, and chemokine signaling. Co-expression of Fas-CD40 with either 4-1BB- or CD28-containing CARs increased in vitro efficacy by augmenting CAR T cell proliferation and cancer target cytotoxicity, and enhanced tumor killing and overall mouse survival in vivo. Functional activity of the Fas-TNFRs were dependent on the co-stimulatory domain within the CAR, highlighting crosstalk between signaling pathways. Furthermore, we show that a major source for Fas-TNFR activation derives from CAR T cells themselves via activation-induced Fas ligand upregulation, highlighting a universal role of Fas-TNFRs in augmenting CAR T cell responses. We have identified Fas-CD40 as the optimal chimera for overcoming Fas ligand-mediated kill and enhancing CAR T cell efficacy.

7.
Mol Ther ; 31(7): 2089-2104, 2023 07 05.
Article in English | MEDLINE | ID: mdl-36945773

ABSTRACT

CAR T cells recognizing CD19 effectively treat relapsed and refractory B-ALL and DLBCL. However, CD19 loss is a frequent cause of relapse. Simultaneously targeting a second antigen, CD22, may decrease antigen escape, but is challenging: its density is approximately 10-fold less than CD19, and its large structure may hamper immune synapse formation. The characteristics of the optimal CD22 CAR are underexplored. We generated 12 distinct CD22 antibodies and tested CARs derived from them to identify a CAR based on the novel 9A8 antibody, which was sensitive to low CD22 density and lacked tonic signaling. We found no correlation between affinity or membrane proximity of recognition epitope within Ig domains 3-6 of CD22 with CART function. The optimal strategy for CD19/CD22 CART co-targeting is undetermined. Co-administration of CD19 and CD22 CARs is costly; single CARs targeting CD19 and CD22 are challenging to construct. The co-expression of two CARs has previously been achieved using bicistronic vectors. Here, we generated a dual CART product by co-transduction with 9A8-41BBζ and CAT-41BBζ (obe-cel), the previously described CD19 CAR. CAT/9A8 CART eliminated single- and double-positive target cells in vitro and eliminated CD19- tumors in vivo. CAT/9A8 CART is being tested in a phase I clinical study (NCT02443831).


Subject(s)
Burkitt Lymphoma , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes , Neoplasm Recurrence, Local , Immunotherapy, Adoptive , Adaptor Proteins, Signal Transducing , Antigens, CD19 , Antibodies , Sialic Acid Binding Ig-like Lectin 2
8.
Biotechniques ; 72(4): 143-154, 2022 04.
Article in English | MEDLINE | ID: mdl-35234525

ABSTRACT

The development of multicistronic vectors enabling differential transgene expression is a goal of gene therapy and poses a significant engineering challenge. Current approaches rely on the insertion of long regulatory sequences that occupy valuable space in vectors, which have a finite and limited packaging capacity. Here we describe a simple method of achieving differential transgene expression by inserting stop codons and translational readthrough motifs (TRMs) to suppress stop codon termination. TRMs reduced downstream transgene expression ∼sixfold to ∼140-fold, depending on the combination of stop codon and TRM used. We show that a TRM can facilitate the controlled secretion of the highly potent cytokine IL-12 at therapeutically beneficial levels in an aggressive immunocompetent mouse melanoma model to prevent tumor growth. Given their compact size (6 bp) and ease of introduction, we envisage that TRMs will be widely adopted in recombinant DNA engineering to facilitate differential transgene expression.


Subject(s)
Protein Biosynthesis , Animals , Codon, Terminator , Mice , Protein Biosynthesis/genetics , Transgenes
9.
Sci Rep ; 11(1): 21902, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34754016

ABSTRACT

Chimeric antigen receptor (CAR) T cells are a promising form of cancer immunotherapy, although they are often associated with severe toxicities. Here, we present a split-CAR design incorporating separate antigen recognition and intracellular signaling domains. These exploit the binding between the tetracycline repressor protein and a small peptide sequence (TIP) to spontaneously assemble as a functional CAR. Addition of the FDA-approved, small molecule antibiotic minocycline, acts as an "off-switch" by displacing the signaling domain and down-tuning CAR T activity. Here we describe the optimization of this split-CAR approach to generate a CAR in which cytotoxicity, cytokine secretion and proliferation can be inhibited in a dose-dependent and reversible manner. Inhibition is effective during on-going CAR T cell activation and inhibits activation and tumor control in vivo. This work shows how optimization of split-CAR structure affects function and adds a novel design allowing easy CAR inhibition through an FDA-approved small molecule.


Subject(s)
Minocycline/pharmacology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Animals , Coculture Techniques , Female , HEK293 Cells , Humans , Mice , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
10.
Nat Med ; 27(10): 1797-1805, 2021 10.
Article in English | MEDLINE | ID: mdl-34642489

ABSTRACT

Chimeric antigen receptor (CAR) T cells targeting CD19 or CD22 have shown remarkable activity in B cell acute lymphoblastic leukemia (B-ALL). The major cause of treatment failure is antigen downregulation or loss. Dual antigen targeting could potentially prevent this, but the clinical safety and efficacy of CAR T cells targeting both CD19 and CD22 remain unclear. We conducted a phase 1 trial in pediatric and young adult patients with relapsed or refractory B-ALL (n = 15) to test AUTO3, autologous transduced T cells expressing both anti-CD19 and anti-CD22 CARs (AMELIA trial, EUDRA CT 2016-004680-39). The primary endpoints were the incidence of grade 3-5 toxicity in the dose-limiting toxicity period and the frequency of dose-limiting toxicities. Secondary endpoints included the rate of morphological remission (complete response or complete response with incomplete bone marrow recovery) with minimal residual disease-negative response, as well as the frequency and severity of adverse events, expansion and persistence of AUTO3, duration of B cell aplasia, and overall and event-free survival. The study endpoints were met. AUTO3 showed a favorable safety profile, with no dose-limiting toxicities or cases of AUTO3-related severe cytokine release syndrome or neurotoxicity reported. At 1 month after treatment the remission rate (that is, complete response or complete response with incomplete bone marrow recovery) was 86% (13 of 15 patients). The 1 year overall and event-free survival rates were 60% and 32%, respectively. Relapses were probably due to limited long-term AUTO3 persistence. Strategies to improve CAR T cell persistence are needed to fully realize the potential of dual targeting CAR T cell therapy in B-ALL.


Subject(s)
Antigens, CD19/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/administration & dosage , Sialic Acid Binding Ig-like Lectin 2/genetics , Adolescent , Adult , Antigens, CD19/immunology , Child , Child, Preschool , Female , Humans , Immunotherapy/adverse effects , Immunotherapy/trends , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/trends , Infant , Male , Pediatrics , Progression-Free Survival , Receptors, Chimeric Antigen/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Young Adult
11.
Mol Ther ; 26(5): 1266-1276, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29661681

ABSTRACT

Engineered T cell therapies show considerable promise in the treatment of refractory malignancies. Given the ability of engineered T cells to engraft and persist for prolonged periods along with unpredicted toxicities, incorporation of a suicide gene to allow selective depletion after administration is desirable. Rapamycin is a safe and widely available immunosuppressive pharmaceutical that acts by heterodimerization of FKBP12 with the FRB fragment of mTOR. The apical caspase caspase 9 is activated by homodimerization through its CARD domain. We developed a rapamycin-induced caspase 9 suicide gene. First, we showed that caspase 9 could be activated by a two-protein format with replacement of the CARD domain with both FRB and FKBP12. We next identified an optimal compact single-protein rapamycin caspase 9 (rapaCasp9) by fusing both FRB and FKBP12 with the catalytic domain of caspase 9. Functionality of rapaCasp9 when co-expressed with a CD19 CAR was demonstrated in vitro and in vivo.


Subject(s)
Caspase 9/genetics , Gene Expression Regulation/drug effects , Gene Expression , Genes, Transgenic, Suicide , Sirolimus/pharmacology , Animals , Biomarkers , Caspase 9/chemistry , Caspase 9/metabolism , Cells, Cultured , Cytotoxicity, Immunologic , Genetic Vectors/genetics , Humans , Immunophenotyping , Mice , Protein Interaction Domains and Motifs , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Immunol Cell Biol ; 95(5): 443-453, 2017 05.
Article in English | MEDLINE | ID: mdl-27899813

ABSTRACT

Liver fibrosis is a progressive pathological process involving inflammation and extracellular matrix deposition. Dipeptidyl peptidase 4 (DPP4), also known as CD26, is a cell surface glycoprotein and serine protease. DPP4 binds to fibronectin, can inactivate specific chemokines, incretin hormone and neuropeptides, and influences cell adhesion and migration. Such properties suggest a pro-fibrotic role for this peptidase but this hypothesis needs in vivo examination. Experimental liver injury was induced with carbon tetrachloride (CCl4) in DPP4 gene knockout (gko) mice. DPP4 gko had less liver fibrosis and inflammation and fewer B cell clusters than wild type mice in the fibrosis model. DPP4 inhibitor-treated mice also developed less liver fibrosis. DNA microarray and PCR showed that many immunoglobulin (Ig) genes and some metabolism-associated transcripts were differentially expressed in the gko strain compared with wild type. CCl4-treated DPP4 gko livers had more IgM+ and IgG+ intrahepatic lymphocytes, and fewer CD4+, IgD+ and CD21+ intrahepatic lymphocytes. These data suggest that DPP4 is pro-fibrotic in CCl4-induced liver fibrosis and that the mechanisms of DPP4 pro-fibrotic action include energy metabolism, B cells, NK cells and CD4+ cells.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Liver/enzymology , Liver/injuries , Animals , Carbon Tetrachloride , Cell Line , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Liver/pathology , Liver Cirrhosis/genetics , Mice , Mice, Knockout , Phenotype , Spleen/pathology , Up-Regulation
13.
Cell Rep ; 15(9): 1957-72, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27210755

ABSTRACT

Super-resolution microscopy has revealed that immune cell receptors are organized in nanoscale clusters at cell surfaces and immune synapses. However, mechanisms and functions for this nanoscale organization remain unclear. Here, we used super-resolution microscopy to compare the surface organization of paired killer Ig-like receptors (KIR), KIR2DL1 and KIR2DS1, on human primary natural killer cells and cell lines. Activating KIR2DS1 assembled in clusters two-fold larger than its inhibitory counterpart KIR2DL1. Site-directed mutagenesis established that the size of nanoclusters is controlled by transmembrane amino acid 233, a lysine in KIR2DS1. Super-resolution microscopy also revealed two ways in which the nanoscale clustering of KIR affects signaling. First, KIR2DS1 and DAP12 nanoclusters are juxtaposed in the resting cell state but coalesce upon receptor ligation. Second, quantitative super-resolution microscopy revealed that phosphorylation of the kinase ZAP-70 or phosphatase SHP-1 is favored in larger KIR nanoclusters. Thus, the size of KIR nanoclusters depends on the transmembrane sequence and affects downstream signaling.


Subject(s)
Cell Membrane/metabolism , Nanoparticles/chemistry , Receptors, KIR2DL1/metabolism , Receptors, KIR/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Cell Line , Clone Cells , Humans , Killer Cells, Natural/metabolism , Membrane Proteins/metabolism , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism
14.
J Immunol ; 195(11): 5432-9, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26500347

ABSTRACT

Optimal T cell activation typically requires engagement of both the TCR and costimulatory receptors, such as CD28. Engagement of CD28 leads to tyrosine phosphorylation of its cytoplasmic region and recruitment of cytoplasmic signaling proteins. Although the exact mechanism of CD28 signal transduction is unknown, CD28 triggering has similarities to the TCR, which was proposed to use the kinetic-segregation (KS) mechanism. The KS model postulates that, when small receptors engage their ligands within areas of close (∼15 nm) contact in the T cell/APC interface, this facilitates phosphorylation by segregating the engaged receptor/ligand complex from receptor protein tyrosine phosphatases with large ectodomains, such as CD45. To test this hypothesis, we examined the effect of elongating the extracellular region of the CD28 ligand, CD80, on its ability to costimulate IL-2 production by primary T cells. CD80 elongation reduced its costimulatory effect without abrogating CD28 binding. Confocal microscopy revealed that elongated CD80 molecules were less well segregated from CD45 at the T cell/APC interface. T cells expressing CD28 harboring a key tyrosine-170 mutation were less sensitive to CD80 elongation. In summary, the effectiveness of CD28 costimulation is inversely proportional to the dimensions of the CD28-CD80 complex. Small CD28-CD80 complex dimensions are required for optimal costimulation by segregation from large inhibitory tyrosine phosphatases. These results demonstrate the importance of ligand dimensions for optimal costimulation of IL-2 production by T cells and suggest that the KS mechanism contributes to CD28 signaling.


Subject(s)
B7-1 Antigen/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-2/biosynthesis , Animals , B7-1 Antigen/genetics , CD28 Antigens/genetics , CHO Cells , Cell Line , Cricetinae , Cricetulus , Leukocyte Common Antigens/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/immunology , Phosphorylation/immunology , Protein Binding , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology
15.
EMBO J ; 34(3): 393-409, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25535246

ABSTRACT

THEMIS is critical for conventional T-cell development, but its precise molecular function remains elusive. Here, we show that THEMIS constitutively associates with the phosphatases SHP1 and SHP2. This complex requires the adapter GRB2, which bridges SHP to THEMIS in a Tyr-phosphorylation-independent fashion. Rather, SHP1 and THEMIS engage with the N-SH3 and C-SH3 domains of GRB2, respectively, a configuration that allows GRB2-SH2 to recruit the complex onto LAT. Consistent with THEMIS-mediated recruitment of SHP to the TCR signalosome, THEMIS knock-down increased TCR-induced CD3-ζ phosphorylation, Erk activation and CD69 expression, but not LCK phosphorylation. This generalized TCR signalling increase led to augmented apoptosis, a phenotype mirrored by SHP1 knock-down. Remarkably, a KI mutation of LCK Ser59, previously suggested to be key in ERK-mediated resistance towards SHP1 negative feedback, did not affect TCR signalling nor ligand discrimination in vivo. Thus, the THEMIS:SHP complex dampens early TCR signalling by a previously unknown molecular mechanism that favours T-cell survival. We discuss possible implications of this mechanism in modulating TCR output signals towards conventional T-cell development and differentiation.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , CD3 Complex/genetics , CD3 Complex/metabolism , Cell Differentiation/genetics , Cell Survival/genetics , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , src Homology Domains
16.
Sci Signal ; 6(285): ra62, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23882121

ABSTRACT

Natural killer (NK) cell responses are regulated by a dynamic equilibrium between activating and inhibitory receptor signals at the immune synapse (or interface) with target cells. Although the organization of receptors at the immune synapse is important for appropriate integration of these signals, there is little understanding of this in detail, because research has been hampered by the limited resolution of light microscopy. Through the use of superresolution single-molecule fluorescence microscopy to reveal the organization of the NK cell surface at the single-protein level, we report that the inhibitory receptor KIR2DL1 is organized in nanometer-scale clusters at the surface of human resting NK cells. Nanoclusters of KIR2DL1 became smaller and denser upon engagement of the activating receptor NKG2D, establishing an unexpected crosstalk between activating receptor signals and the positioning of inhibitory receptors. These rearrangements in the nanoscale organization of surface NK cell receptors were dependent on the actin cytoskeleton. Together, these data establish that NK cell activation involves a nanometer-scale reorganization of surface receptors, which in turn affects models for signal integration and thresholds that control NK cell effector functions and NK cell development.


Subject(s)
Killer Cells, Natural/cytology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, KIR2DL1/metabolism , Actins/chemistry , Antibodies, Monoclonal/chemistry , CD28 Antigens/chemistry , Cell Membrane/metabolism , Cytoskeleton/metabolism , HLA Antigens/chemistry , Humans , Immune System , Killer Cells, Natural/ultrastructure , Microscopy, Confocal , Microscopy, Fluorescence , Plasmids/metabolism , Signal Transduction
17.
Blood ; 121(21): 4295-302, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23580664

ABSTRACT

T-cell receptor (TCR) triggering results in a cascade of intracellular tyrosine phosphorylation events that ultimately leads to T-cell activation. It is dependent on changes in the relative activities of membrane-associated tyrosine kinases and phosphatases near the engaged TCR. CD45 and CD148 are transmembrane tyrosine phosphatases with large ectodomains that have activatory and inhibitory effects on TCR triggering. This study investigates whether and how the ectodomains of CD45 and CD148 modulate their inhibitory effect on TCR signaling. Expression in T cells of forms of these phosphatases with truncated ectodomains inhibited TCR triggering. In contrast, when these phosphatases were expressed with large ectodomains, they had no inhibitory effect. Imaging studies revealed that truncation of the ectodomains enhanced colocalization of these phosphatases with ligated TCR at the immunological synapse. Our results suggest that the large ectodomains of CD45 and CD148 modulate their inhibitory effect by enabling their passive, size-based segregation from ligated TCR, supporting the kinetic-segregation model of TCR triggering.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Interleukin-2/metabolism , Leukocyte Common Antigens/chemistry , Leukocyte Common Antigens/genetics , Lymphocyte Activation/genetics , Mice , Models, Chemical , Mutagenesis/immunology , Protein Binding/immunology , Protein Structure, Tertiary/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Structure-Activity Relationship
18.
PLoS Comput Biol ; 9(3): e1003004, 2013.
Article in English | MEDLINE | ID: mdl-23555234

ABSTRACT

Receptor phosphorylation is thought to be tightly regulated because phosphorylated receptors initiate signaling cascades leading to cellular activation. The T cell antigen receptor (TCR) on the surface of T cells is phosphorylated by the kinase Lck and dephosphorylated by the phosphatase CD45 on multiple immunoreceptor tyrosine-based activation motifs (ITAMs). Intriguingly, Lck sequentially phosphorylates ITAMs and ZAP-70, a cytosolic kinase, binds to phosphorylated ITAMs with differential affinities. The purpose of multiple ITAMs, their sequential phosphorylation, and the differential ZAP-70 affinities are unknown. Here, we use a systems model to show that this signaling architecture produces emergent ultrasensitivity resulting in switch-like responses at the scale of individual TCRs. Importantly, this switch-like response is an emergent property, so that removal of multiple ITAMs, sequential phosphorylation, or differential affinities abolishes the switch. We propose that highly regulated TCR phosphorylation is achieved by an emergent switch-like response and use the systems model to design novel chimeric antigen receptors for therapy.


Subject(s)
Models, Immunological , Receptors, Antigen, T-Cell/metabolism , Systems Biology/methods , Animals , Humans , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction
19.
Front Immunol ; 3: 29, 2012.
Article in English | MEDLINE | ID: mdl-22566913

ABSTRACT

Based on studies in model systems it has been proposed that the cytoplasmic domains of T cell receptor signaling subunits that have polybasic motifs associate with the plasma membrane, and that this regulates their phosphorylation. Recent experiments in more physiological systems have confirmed membrane association but raised questions as to its function.

20.
Proc Natl Acad Sci U S A ; 108(48): 19323-8, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22084078

ABSTRACT

The T-cell receptor (TCR) consists of a TCRαß heterodimer, a TCRζ homodimer, and CD3γε and CD3δε heterodimers. The precise mechanism of T-cell triggering following TCR ligand engagement remains elusive. Previous studies reported that the cytoplasmic tail of CD3ε binds to the plasma membrane through a basic residue-rich stretch (BRS) and proposed that dissociation from the membrane is required for phosphorylation thereof. In this report we show that BRS motifs within the cytoplasmic tail of TCRζ mediate association with the plasma membrane and that TCR engagement results in TCRζ dissociation from the membrane. This dissociation requires phosphorylation of the TCRζ immunoreceptor tyrosine-based activation motifs by lymphocyte cell-specificprotein tyrosine kinase (Lck) but not ζ-chain-associated protein kinase 70 binding. Mutations of the TCRζ BRS motifs that disrupt this membrane association attenuate proximal and distal responses induced by TCR engagement. These mutations appear to alter the localization of TCRζ with respect to Lck as well as the mobility of the TCR complex. This study reveals that tyrosine phosphorylation of the TCRζ cytoplasmic domain regulates its association with the plasma membrane and highlights the functional importance of TCRζ BRS motifs.


Subject(s)
Cell Membrane/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , Amino Acid Motifs/genetics , Animals , Blotting, Western , Cell Line , Cytoplasm/metabolism , Flow Cytometry , Fluorescence Resonance Energy Transfer , Immunoprecipitation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Microscopy, Confocal , Mutation/genetics , Phosphorylation , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...