Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2743: 81-92, 2024.
Article in English | MEDLINE | ID: mdl-38147209

ABSTRACT

Phosphotyrosine phosphatase non-receptor type 22 (PTPN22) is a key regulator of immune cell activation and responses. Genetic polymorphisms of PTPN22 have been strongly linked with an increased risk of developing autoimmune diseases, while analysis of PTPN22-deficient mouse strains has determined that PTPN22 serves as a negative regulator of T cell antigen receptor signaling. As well as these key roles in maintaining immune tolerance, PTPN22 acts as an intracellular checkpoint for T cell responses to cancer, suggesting that PTPN22 might be a useful target to improve T cell immunotherapies. To assess the potential for targeting PTPN22, we have crossed Ptpn22-deficient mice to an OT-I TCR transgenic background and used adoptive T cell transfer approaches in mouse cancer models. We provide basic methods for the in vitro expansion of effector OT-I cytotoxic T lymphocytes, in vitro phenotypic analysis, and in vivo adoptive T cell transfer models to assess the role of PTPN22 in anti-cancer immunity.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell , Animals , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Neoplasms/genetics , Neoplasms/therapy , Signal Transduction , Disease Models, Animal , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
2.
J Immunother Cancer ; 11(12)2023 12 06.
Article in English | MEDLINE | ID: mdl-38056892

ABSTRACT

BACKGROUND: Adoptive cell therapy (ACT) is a promising strategy for treating cancer, yet it faces several challenges such as lack of long-term protection due to T cell exhaustion induced by chronic TCR stimulation in the tumor microenvironment. One benefit of ACT, however, is that it allows for cellular manipulations, such as deletion of the phosphotyrosine phosphatase non-receptor type 22 (PTPN22), which improves CD8+ T cell antitumor efficacy in ACT. We tested whether Ptpn22KO cytolytic T cells (CTLs) were also more effective than Ptpn22WT CTL in controlling tumors in scenarios that favor T cell exhaustion. METHODS: Tumor control by Ptpn22WT and Ptpn22KO CTL was assessed following adoptive transfer of low numbers of CTL to mice with subcutaneously implanted MC38 tumors. Tumor infiltrating lymphocytes were isolated for analysis of effector functions. An in vitro assay was established to compare CTL function in response to acute and chronic restimulation with antigen-pulsed tumor cells. The expression of effector and exhaustion-associated proteins by Ptpn22WT and Ptpn22KO T cells was followed over time in vitro and in vivo using the ID8 tumor model. Finally, the effect of PD-1 and TIM-3 blockade on Ptpn22KO CTL tumor control was assessed using monoclonal antibodies and CRISPR/Cas9-mediated knockout. RESULTS: Despite having improved effector function at the time of transfer, Ptpn22KO CTL became more exhausted than Ptpn22WT CTL, characterized by more rapid loss of effector functions, and earlier and higher expression of inhibitory receptors (IRs), particularly the terminal exhaustion marker TIM-3. TIM-3 expression, under the control of the transcription factor NFIL3, was induced by IL-2 signaling which was enhanced in Ptpn22KO cells. Antitumor responses of Ptpn22KO CTL were improved following PD-1 blockade in vivo, yet knockout or antibody-mediated blockade of TIM-3 did not improve but further impaired tumor control, indicating TIM-3 signaling itself did not drive the diminished function seen in Ptpn22KO CTL. CONCLUSIONS: This study questions whether TIM-3 plays a role as an IR and highlights that genetic manipulation of T cells for ACT needs to balance short-term augmented effector function against the risk of T cell exhaustion in order to achieve longer-term protection.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Neoplasms , Mice , Animals , Programmed Cell Death 1 Receptor , T-Cell Exhaustion , Protein Tyrosine Phosphatases , Cell- and Tissue-Based Therapy , Tumor Microenvironment
3.
Nat Immunol ; 24(8): 1217-1219, 2023 08.
Article in English | MEDLINE | ID: mdl-37365385

Subject(s)
B-Lymphocytes , Sugars
4.
Blood Adv ; 7(15): 3874-3890, 2023 08 08.
Article in English | MEDLINE | ID: mdl-36867577

ABSTRACT

Multiple myeloma (MM) shows constitutive activation of canonical and noncanonical nuclear factor κB (NF-κB) signaling via genetic mutations or tumor microenvironment (TME) stimulations. A subset of MM cell lines showed dependency for cell growth and survival on the canonical NF-κB transcription factor RELA alone, suggesting a critical role for a RELA-mediated biological program in MM pathogenesis. Here, we determined the RELA-dependent transcriptional program in MM cell lines and found the expression of the cell surface molecules interleukin-27 receptor-α (IL-27Rα) and the adhesion molecule JAM2 to be responsive to RELA at the messenger RNA and protein levels. IL-27Rα and JAM2 were expressed on primary MM cells at higher levels than on healthy long-lived plasma cells (PCs) in the bone marrow. IL-27 activated STAT1, and to a lesser extent STAT3, in MM cell lines and in PCs generated from memory B cells in an IL-21-dependent in vitro PC differentiation assay. Concomitant activity of IL-21 and IL-27 enhanced differentiation into PCs and increased the cell-surface expression of the known STAT target gene CD38. In accordance, a subset of MM cell lines and primary MM cells cultured with IL-27 upregulated CD38 cell-surface expression, a finding with potential implications for enhancing the efficacy of CD38-directed monoclonal antibody therapies by increasing CD38 expression on tumor cells. The elevated expression of IL-27Rα and JAM2 on MM cells compared with that on healthy PCs may be exploited for the development of targeted therapeutic strategies that modulate the interaction of MM cells with the TME.


Subject(s)
Interleukin-27 , Multiple Myeloma , Humans , Interleukin-27/metabolism , Multiple Myeloma/genetics , NF-kappa B/metabolism , Receptors, Cytokine/metabolism , Tumor Microenvironment , Up-Regulation
5.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33822775

ABSTRACT

T cell receptor (TCR) triggering by antigen results in metabolic reprogramming that, in turn, facilitates the exit of T cells from quiescence. The increased nutrient requirements of activated lymphocytes are met, in part, by upregulation of cell surface transporters and enhanced uptake of amino acids, fatty acids, and glucose from the environment. However, the role of intracellular pathways of amino acid biosynthesis in T cell activation is relatively unexplored. Asparagine is a nonessential amino acid that can be synthesized intracellularly through the glutamine-hydrolyzing enzyme asparagine synthetase (ASNS). We set out to define the requirements for uptake of extracellular asparagine and ASNS activity in CD8+ T cell activation. At early time points of activation in vitro, CD8+ T cells expressed little or no ASNS, and, as a consequence, viability and TCR-stimulated growth, activation, and metabolic reprogramming were substantially impaired under conditions of asparagine deprivation. At later time points (more than 24 hours of activation), TCR-induced mTOR-dependent signals resulted in ASNS upregulation that endowed CD8+ T cells with the capacity to function independently of extracellular asparagine. Thus, our data suggest that the coordinated upregulation of ASNS expression and uptake of extracellular asparagine is involved in optimal T cell effector responses.


Subject(s)
Asparagine/metabolism , Aspartate-Ammonia Ligase/metabolism , CD8-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/physiology , Receptors, Antigen, T-Cell/metabolism , Animals , Aspartate-Ammonia Ligase/genetics , Cell Survival , In Vitro Techniques , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
6.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33707311

ABSTRACT

Patients with glioblastoma (GBM) have a poor prognosis, and inefficient delivery of drugs to tumors represents a major therapeutic hurdle. Hematopoietic stem cell (HSC)-derived myeloid cells efficiently home to GBM and constitute up to 50% of intratumoral cells, making them highly appropriate therapeutic delivery vehicles. Because myeloid cells are ubiquitously present in the body, we recently established a lentiviral vector containing matrix metalloproteinase 14 (MMP14) promoter, which is active specifically in tumor-infiltrating myeloid cells as opposed to myeloid cells in other tissues, and resulted in a specific delivery of transgenes to brain metastases in HSC gene therapy. Here, we used this novel approach to target transforming growth factor beta (TGFß) as a key tumor-promoting factor in GBM. Transplantation of HSCs transduced with lentiviral vector expressing green fluorescent protein (GFP) into lethally irradiated recipient mice was followed by intracranial implantation of GBM cells. Tumor-infiltrating HSC progeny was characterized by flow cytometry. In therapy studies, mice were transplanted with HSCs transduced with lentiviral vector expressing soluble TGFß receptor II-Fc fusion protein under MMP14 promoter. This TGFß-blocking therapy was compared with the targeted tumor irradiation, the combination of the two therapies, and control. Tumor growth and survival were quantified (statistical significance determined by t-test and log-rank test). T cell memory response was probed through a repeated tumor challenge. Myeloid cells were the most abundant HSC-derived population infiltrating GBM. TGFß-blocking HSC gene therapy in combination with irradiation significantly reduced tumor burden as compared with monotherapies and the control, and significantly prolonged survival as compared with the control and TGFß-blocking monotherapy. Long-term protection from GBM was achieved only with the combination treatment (25% of the mice) and was accompanied by a significant increase in CD8+ T cells at the tumor implantation site following tumor rechallenge. We demonstrated a preclinical proof-of-principle for tumor myeloid cell-specific HSC gene therapy in GBM. In the clinic, HSC gene therapy is being successfully used in non-cancerous brain disorders and the feasibility of HSC gene therapy in patients with glioma has been demonstrated in the context of bone marrow protection. This indicates an opportunity for clinical translation of our therapeutic approach.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/therapy , Hematopoietic Stem Cell Transplantation , Immunoglobulin Fc Fragments/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Immunoglobulin Fc Fragments/metabolism , Matrix Metalloproteinase 14/genetics , Mice, Inbred C57BL , Promoter Regions, Genetic , Proof of Concept Study , Radiotherapy, Adjuvant , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Burden
7.
J Natl Cancer Inst ; 112(6): 617-627, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31501884

ABSTRACT

BACKGROUND: Brain metastases (BrM) develop in 20-40% of cancer patients and represent an unmet clinical need. Limited access of drugs into the brain because of the blood-brain barrier is at least partially responsible for therapeutic failure, necessitating improved drug delivery systems. METHODS: Green fluorescent protein (GFP)-transduced murine and nontransduced human hematopoietic stem cells (HSCs) were administered into mice (n = 10 and 3). The HSC progeny in mouse BrM and in patient-derived BrM tissue (n = 6) was characterized by flow cytometry and immunofluorescence. Promoters driving gene expression, specifically within the BrM-infiltrating HSC progeny, were identified through differential gene-expression analysis and subsequent validation of a series of promoter-green fluorescent protein-reporter constructs in mice (n = 5). One of the promoters was used to deliver tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to BrM in mice (n = 17/21 for TRAIL vs control group). RESULTS: HSC progeny (consisting mostly of macrophages) efficiently homed to macrometastases (mean [SD] = 37.6% [7.2%] of all infiltrating cells for murine HSC progeny; 27.9% mean [SD] = 27.9% [4.9%] of infiltrating CD45+ hematopoietic cells for human HSC progeny) and micrometastases in mice (19.3-53.3% of all macrophages for murine HSCs). Macrophages were also abundant in patient-derived BrM tissue (mean [SD] = 8.8% [7.8%]). Collectively, this provided a rationale to optimize the delivery of gene therapy to BrM within myeloid cells. MMP14 promoter emerged as the strongest promoter construct capable of limiting gene expression to BrM-infiltrating myeloid cells in mice. TRAIL delivered under MMP14 promoter statistically significantly prolonged survival in mice (mean [SD] = 19.0 [3.4] vs mean [SD] = 15.0 [2.0] days for TRAIL vs control group; two-sided P = .006), demonstrating therapeutic and translational potential of our approach. CONCLUSIONS: Our study establishes HSC gene therapy using a myeloid cell-specific promoter as a new strategy to target BrM. This approach, with strong translational value, has potential to overcome the blood-brain barrier, target micrometastases, and control multifocal lesions.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Myeloid Cells/physiology , Animals , Female , Gene Transfer Techniques , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Lentivirus/genetics , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/genetics
8.
JCI Insight ; 52019 07 23.
Article in English | MEDLINE | ID: mdl-31335326

ABSTRACT

Adoptive T cell therapy (ACT) has been established as an efficacious methodology for the treatment of cancer. Identifying targets to enhance the antigen recognition, functional capacity and longevity of T cells has the potential to broaden the applicability of these approaches in the clinic. We previously reported that targeting expression of phosphotyrosine phosphatase, non-receptor type (PTPN) 22 in effector CD8+ T cells enhances the efficacy of ACT for tumor clearance in mice. In the current work, we demonstrate that, upon ACT, PTPN22-deficient effector CD8+ T cells afford greater protection against tumors expressing very low affinity antigen, but do not survive long-term in vivo. Persistence of CD8+ T cells following tumor clearance is improved by ACT of memory phenotype cells that have a distinct metabolic phenotype as compared to effector T cells. Importantly, PTPN22-deficient T cells have comparable capacity to form long-lived memory cells in vivo but enhanced anti-tumor activity in vivo and effector responses ex vivo. These findings provide key insight into the regulation of effector and memory T cell responses in vivo, and indicate that PTPN22 is a rationale target to improve ACT for cancer.


Subject(s)
Immunologic Memory , Immunotherapy, Adoptive/methods , Ovarian Neoplasms/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Mice, Knockout , Ovarian Neoplasms/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/transplantation
9.
PLoS Biol ; 17(3): e2006716, 2019 03.
Article in English | MEDLINE | ID: mdl-30856173

ABSTRACT

The interdependence of selective cues during development of regulatory T cells (Treg cells) in the thymus and their suppressive function remains incompletely understood. Here, we analyzed this interdependence by taking advantage of highly dynamic changes in expression of microRNA 181 family members miR-181a-1 and miR-181b-1 (miR-181a/b-1) during late T-cell development with very high levels of expression during thymocyte selection, followed by massive down-regulation in the periphery. Loss of miR-181a/b-1 resulted in inefficient de novo generation of Treg cells in the thymus but simultaneously permitted homeostatic expansion in the periphery in the absence of competition. Modulation of T-cell receptor (TCR) signal strength in vivo indicated that miR-181a/b-1 controlled Treg-cell formation via establishing adequate signaling thresholds. Unexpectedly, miR-181a/b-1-deficient Treg cells displayed elevated suppressive capacity in vivo, in line with elevated levels of cytotoxic T-lymphocyte-associated 4 (CTLA-4) protein, but not mRNA, in thymic and peripheral Treg cells. Therefore, we propose that intrathymic miR-181a/b-1 controls development of Treg cells and imposes a developmental legacy on their peripheral function.


Subject(s)
MicroRNAs/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Flow Cytometry , Mice , Mice, Knockout , MicroRNAs/genetics , Microscopy, Confocal , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Thymocytes/metabolism
10.
Immunology ; 154(3): 377-382, 2018 07.
Article in English | MEDLINE | ID: mdl-29512901

ABSTRACT

A number of polymorphisms in immune-regulatory genes have been identified as risk factors for the development of autoimmune disease. PTPN22 (that encodes a tyrosine phosphatase) has been associated with the development of several autoimmune diseases, including type 1 diabetes, rheumatoid arthritis and systemic lupus erythematosus. PTPN22 regulates the activity and effector functions of multiple important immune cell types, including lymphocytes, granulocytes and myeloid cells. In this review, we describe the role of PTPN22 in regulating T-cell activation and effector responses. We discuss progress in our understanding of the impact of PTPN22 in autoimmune disease in humans and mouse models, as well as recent evidence suggesting that genetic manipulation of PTPN22 expression might enhance the efficacy of anti-tumour T-cell responses.


Subject(s)
Autoimmunity/genetics , Immunomodulation , Neoplasms/etiology , Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Neoplasms/pathology , Polymorphism, Single Nucleotide
11.
Proc Natl Acad Sci U S A ; 115(7): E1540-E1549, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386395

ABSTRACT

Inhibition of immune checkpoints programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on T cells results in durable antitumor activity in melanoma patients. Despite high frequency of melanoma brain metastases (BrM) and associated poor prognosis, the activity and mechanisms of immune checkpoint inhibitors (ICI) in metastatic tumors that develop within the "immune specialized" brain microenvironment, remain elusive. We established a melanoma tumor transplantation model with intracranial plus extracranial (subcutaneous) tumor, mimicking the clinically observed coexistence of metastases inside and outside the brain. Strikingly, intracranial ICI efficacy was observed only when extracranial tumor was present. Extracranial tumor was also required for ICI-induced increase in CD8+ T cells, macrophages, and microglia in brain tumors, and for up-regulation of immune-regulatory genes. Combined PD-1/CTLA-4 blockade had a superior intracranial efficacy over the two monotherapies. Cell depletion studies revealed that NK cells and CD8+ T cells were required for intracranial anti-PD-1/anti-CTLA-4 efficacy. Rather than enhancing CD8+ T cell activation and expansion within intracranial tumors, PD-1/CTLA-4 blockade dramatically (∼14-fold) increased the trafficking of CD8+ T cells to the brain. This was mainly through the peripheral expansion of homing-competent effector CD8+ T cells and potentially further enhanced through up-regulation of T cell entry receptors intercellular adhesion molecule 1 and vascular adhesion molecule 1 on tumor vasculature. Our study indicates that extracranial activation/release of CD8+ T cells from PD-1/CTLA-4 inhibition and potentiation of their recruitment to the brain are paramount to the intracranial anti-PD-1/anti-CTLA-4 activity, suggesting augmentation of these processes as an immune therapy-enhancing strategy in metastatic brain cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/secondary , Female , Granzymes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/immunology , Skin Neoplasms/secondary , Skin Neoplasms/therapy , Tumor Burden , Tumor Cells, Cultured
12.
Nat Commun ; 8(1): 1343, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116089

ABSTRACT

Transforming growth factor ß (TGFß) is important in maintaining self-tolerance and inhibits T cell reactivity. We show that CD8+ T cells that lack the tyrosine phosphatase Ptpn22, a major predisposing gene for autoimmune disease, are resistant to the suppressive effects of TGFß. Resistance to TGFß suppression, while disadvantageous in autoimmunity, helps Ptpn22 -/- T cells to be intrinsically superior at clearing established tumors that secrete TGFß. Mechanistically, loss of Ptpn22 increases the capacity of T cells to produce IL-2, which overcomes TGFß-mediated suppression. These data suggest that a viable strategy to improve anti-tumor adoptive cell therapy may be to engineer tumor-restricted T cells with mutations identified as risk factors for autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Transforming Growth Factor beta/pharmacology , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/transplantation , Female , Homeodomain Proteins/genetics , Interleukin-2/metabolism , Male , Mice, Mutant Strains , Mice, Transgenic , Ovalbumin/pharmacology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Peptide Fragments/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
13.
Sci Signal ; 9(448): ra99, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27703032

ABSTRACT

Integrins are heterodimeric transmembrane proteins that play a fundamental role in the migration of leukocytes to sites of infection or injury. We found that protein tyrosine phosphatase nonreceptor type 22 (PTPN22) inhibits signaling by the integrin lymphocyte function-associated antigen-1 (LFA-1) in effector T cells. PTPN22 colocalized with its substrates at the leading edge of cells migrating on surfaces coated with the LFA-1 ligand intercellular adhesion molecule-1 (ICAM-1). Knockout or knockdown of PTPN22 or expression of the autoimmune disease-associated PTPN22-R620W variant resulted in the enhanced phosphorylation of signaling molecules downstream of integrins. Superresolution imaging revealed that PTPN22-R620 (wild-type PTPN22) was present as large clusters in unstimulated T cells and that these disaggregated upon stimulation of LFA-1, enabling increased association of PTPN22 with its binding partners at the leading edge. The failure of PTPN22-R620W molecules to be retained at the leading edge led to increased LFA-1 clustering and integrin-mediated cell adhesion. Our data define a previously uncharacterized mechanism for fine-tuning integrin signaling in T cells, as well as a paradigm of autoimmunity in humans in which disease susceptibility is underpinned by inherited phosphatase mutations that perturb integrin function.


Subject(s)
Autoimmunity/physiology , Intercellular Adhesion Molecule-1/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , T-Lymphocytes , Amino Acid Substitution , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Knockout , Mutation, Missense , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
J Immunol ; 197(2): 429-40, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27288531

ABSTRACT

The cytoplasmic phosphatase, protein tyrosine phosphatase nonreceptor type 22 (PTPN22), is a negative regulator of T cell signaling. Genome-wide association studies have shown that single-nucleotide polymorphisms in PTPN22 confer an increased risk of developing multiple autoimmune diseases in humans. The precise function of PTPN22 and how the variant protein contributes to autoimmunity is not well understood. To address this issue, we investigated the effect of PTPN22 deficiency on disease susceptibility in a mouse model of autoimmune arthritis. The SKG mouse expresses a hypomorphic mutant allele of ZAP70, which, upon exposure to fungal Ags, predisposes the mice to a CD4(+) T cell-mediated autoimmune arthritis that closely resembles rheumatoid arthritis in humans. Surprisingly, SKG Ptpn22(-/-) mice developed less severe mannan-induced arthritis compared with SKG mice. Diminution of disease was not due to significant alterations in thymocyte development or repertoire selection in SKG Ptpn22(-/-) mice, even though T cell-mediated signal transduction was improved. Instead, Ptpn22 deficiency appeared to bias CD4 Th cell differentiation away from the Th17 lineage, which is pathogenic in this setting, to a more Th1/T regulatory-focused response. These data show that even small perturbations in TCR signal transduction pathways can have profound consequences on the differentiation of T cell lineages and thus for the development of autoimmune diseases.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Flow Cytometry , Mannans/toxicity , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , Receptors, Antigen, T-Cell/immunology , Th17 Cells/immunology
15.
J Immunol ; 195(10): 4615-22, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26453749

ABSTRACT

Ag-dependent activation of naive T cells induces dramatic changes in cellular metabolism that are essential for cell growth, division, and differentiation. In recent years, the serine/threonine kinase mechanistic target of rapamycin (mTOR) has emerged as a key integrator of signaling pathways that regulate these metabolic processes. However, the role of specific downstream effectors of mTOR function in T cells is poorly understood. Ribosomal protein S6 (rpS6) is an essential component of the ribosome and is inducibly phosphorylated following mTOR activation in eukaryotic cells. In the current work, we addressed the role of phosphorylation of rpS6 as an effector of mTOR function in T cell development, growth, proliferation, and differentiation using knockin and TCR transgenic mice. Surprisingly, we demonstrate that rpS6 phosphorylation is not required for any of these processes either in vitro or in vivo. Indeed, rpS6 knockin mice are completely sensitive to the inhibitory effects of rapamycin and an S6 kinase 1 (S6K1)-specific inhibitor on T cell activation and proliferation. These results place the mTOR complex 1-S6K1 axis as a crucial determinant of T cell activation independently of its ability to regulate rpS6 phosphorylation.


Subject(s)
Lymphocyte Activation/immunology , Multiprotein Complexes/metabolism , Ribosomal Protein S6 Kinases/metabolism , Ribosomal Protein S6/metabolism , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Signal Transduction/immunology , Sirolimus/pharmacology , T-Lymphocytes/cytology
16.
Cell Cycle ; 14(5): 705-11, 2015.
Article in English | MEDLINE | ID: mdl-25715232

ABSTRACT

The non-receptor tyrosine phosphatase PTPN22 has a vital function in inhibiting antigen-receptor signaling in T cells, while polymorphisms in the PTPN22 gene are important risk alleles in human autoimmune diseases. We recently reported that a key physiological function of PTPN22 was to prevent naïve T cell activation and effector cell responses in response to low affinity antigens. PTPN22 also has a more general role in limiting T cell receptor-induced proliferation. Here we present new data emphasizing this dual function for PTPN22 in T cells. Furthermore, we show that T cell activation modulates the expression of PTPN22 and additional inhibitory phosphatases. We discuss the implication of these findings for our understanding of the roles of PTPN22 in regulating T cell responses and in autoimmunity.


Subject(s)
Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Homeostasis , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Antigens/immunology , Cell Proliferation , Humans , Lymphocyte Activation/immunology , Lymphopenia/immunology , Mice , Models, Animal , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Up-Regulation
17.
Nat Immunol ; 15(9): 875-883, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108421

ABSTRACT

T cells must be tolerant of self antigens to avoid autoimmunity but responsive to foreign antigens to provide protection against infection. We found that in both naive T cells and effector T cells, the tyrosine phosphatase PTPN22 limited signaling via the T cell antigen receptor (TCR) by weak agonists and self antigens while not impeding responses to strong agonist antigens. T cells lacking PTPN22 showed enhanced formation of conjugates with antigen-presenting cells pulsed with weak peptides, which led to activation of the T cells and their production of inflammatory cytokines. This effect was exacerbated under conditions of lymphopenia, with the formation of potent memory T cells in the absence of PTPN22. Our data address how loss-of-function PTPN22 alleles can lead to the population expansion of effector and/or memory T cells and a predisposition to human autoimmunity.


Subject(s)
Autoimmunity/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Alleles , Animals , Antigen-Presenting Cells/immunology , Lymphocyte Activation/immunology , Mice
18.
Nat Rev Immunol ; 13(4): 257-69, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23524462

ABSTRACT

Engagement of antigen-specific T cell receptors (TCRs) is a prerequisite for T cell activation. Acquisition of appropriate effector T cell function requires the participation of multiple signals from the T cell microenvironment. Trying to understand how these signals integrate to achieve specific functional outcomes while maintaining tolerance to self is a major challenge in lymphocyte biology. Several recent publications have provided important insights into how dysregulation of T cell signalling and the development of autoreactivity can result if the branching and integration of signalling pathways are perturbed. We discuss how these findings highlight the importance of spatial segregation of individual signalling components as a way of regulating T cell responsiveness and immune tolerance.


Subject(s)
Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Humans , Immune Tolerance/immunology , Integrins/immunology , Integrins/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Models, Immunological , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
19.
J Immunol ; 189(12): 5694-702, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23150718

ABSTRACT

Fcγ receptors (FcγR) provide important immunoregulation. Targeting inhibitory FcγRIIb may therefore prolong allograft survival, but its role in transplantation has not been addressed. FcγRIIb signaling was examined in murine models of acute or chronic cardiac allograft rejection by transplanting recipients that either lacked FcγRIIb expression (FcγRIIb(-/-)) or overexpressed FcγRIIb on B cells (B cell transgenic [BTG]). Acute heart allograft rejection occurred at the same tempo in FcγRIIb(-/-) C57BL/6 (B6) recipients as wild type recipients, with similar IgG alloantibody responses. In contrast, chronic rejection of MHC class II-mismatched bm12 cardiac allografts was accelerated in FcγRIIb(-/-) mice, with development of more severe transplant arteriopathy and markedly augmented effector autoantibody production. Autoantibody production was inhibited and rejection was delayed in BTG recipients. Similarly, whereas MHC class I-mismatched B6.K(d) hearts survived indefinitely and remained disease free in B6 mice, much stronger alloantibody responses and progressive graft arteriopathy developed in FcγRIIb(-/-) recipients. Notably, FcγRIIb-mediated inhibition of B6.K(d) heart graft rejection was abrogated by increasing T cell help through transfer of additional H2.K(d)-specific CD4 T cells. Thus, inhibitory FcγRIIb signaling regulates chronic but not acute rejection, most likely because the supra-optimal helper CD4 T cell response in acute rejection overcomes FcγRIIb-mediated inhibition of the effector B cell population. Immunomodulation of FcγRIIb in clinical transplantation may hold potential for inhibiting progression of transplant arteriopathy and prolonging transplant survival.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Immunoglobulin G/physiology , Isoantibodies/biosynthesis , Receptors, IgG/antagonists & inhibitors , Receptors, IgG/physiology , Signal Transduction/immunology , Acute Disease , Animals , Chronic Disease , Graft Rejection/metabolism , Heart Transplantation/immunology , Hep G2 Cells , Humans , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Receptors, IgG/deficiency
20.
Sci Signal ; 5(252): ra87, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23193160

ABSTRACT

The cytoplasmic phosphatase PTPN22 (protein tyrosine phosphatase nonreceptor type 22) plays a key role in regulating lymphocyte homeostasis, which ensures that the total number of lymphocytes in the periphery remains relatively constant. Mutations in PTPN22 confer an increased risk of developing autoimmune diseases; however, the precise function of PTPN22 and how mutations contribute to autoimmunity remain controversial. Loss-of-function mutations in PTPN22 are associated with increased numbers of effector T cells and autoreactive B cells in humans and mice; however, the complete absence of PTPN22 in mice does not result in spontaneous autoimmunity. We found that PTPN22 was a key regulator of regulatory T cell (T(reg)) function that fine-tuned the signaling of the T cell receptor and integrins. PTPN22(-/-) T(regs) were more effective at immunosuppression than were wild-type T(regs), and they suppressed the activity of PTPN22(-/-) effector T cells, preventing autoimmunity. Compared to wild-type T(regs), PTPN22(-/-) T(regs) produced increased amounts of the immunosuppressive cytokine interleukin-10 and had enhanced adhesive properties mediated by the integrin lymphocyte function-associated antigen-1, processes that are critical for T(reg) function. This previously undiscovered role of PTPN22 in regulating integrin signaling and T(reg) function suggests that PTPN22 may be a useful therapeutic target for manipulating T(reg) function in human disease.


Subject(s)
Immune Tolerance , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cell Adhesion/genetics , Cell Adhesion/immunology , Humans , Integrins/genetics , Integrins/immunology , Integrins/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Mice , Mice, Knockout , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Signal Transduction/genetics , T-Lymphocytes, Regulatory/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...