Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Res Sq ; 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34580668

ABSTRACT

Patients with cancer have higher COVID-19 morbidity and mortality. Here we present the prospective CAPTURE study (NCT03226886) integrating longitudinal immune profiling with clinical annotation. Of 357 patients with cancer, 118 were SARS-CoV-2-positive, 94 were symptomatic and 2 patients died of COVID-19. In this cohort, 83% patients had S1-reactive antibodies, 82% had neutralizing antibodies against WT, whereas neutralizing antibody titers (NAbT) against the Alpha, Beta, and Delta variants were substantially reduced. Whereas S1-reactive antibody levels decreased in 13% of patients, NAbT remained stable up to 329 days. Patients also had detectable SARS-CoV-2-specific T cells and CD4+ responses correlating with S1-reactive antibody levels, although patients with hematological malignancies had impaired immune responses that were disease and treatment-specific, but presented compensatory cellular responses, further supported by clinical. Overall, these findings advance the understanding of the nature and duration of immune response to SARS-CoV-2 in patients with cancer.

2.
Med ; 2(9): 1093-1109.e6, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34414384

ABSTRACT

BACKGROUND: Differences in humoral immunity to coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), between children and adults remain unexplained, and the effect of underlying immune dysfunction or suppression is unknown. Here, we sought to examine the antibody immune competence of children and adolescents with prevalent inflammatory rheumatic diseases, juvenile idiopathic arthritis (JIA), juvenile dermatomyositis (JDM), and juvenile systemic lupus erythematosus (JSLE) against the seasonal human coronavirus (HCoV)-OC43 that frequently infects this age group. METHODS: Sera were collected from JIA (n = 118), JDM (n = 49), and JSLE (n = 30) patients and from healthy control (n = 54) children and adolescents prior to the coronavirus disease 19 (COVID-19) pandemic. We used sensitive flow-cytometry-based assays to determine titers of antibodies that reacted with the spike and nucleoprotein of HCoV-OC43 and cross-reacted with the spike and nucleoprotein of SARS-CoV-2, and we compared them with respective titers in sera from patients with multisystem inflammatory syndrome in children and adolescents (MIS-C). FINDINGS: Despite immune dysfunction and immunosuppressive treatment, JIA, JDM, and JSLE patients maintained comparable or stronger humoral responses than healthier peers, which was dominated by immunoglobulin G (IgG) antibodies to HCoV-OC43 spike, and harbored IgG antibodies that cross-reacted with SARS-CoV-2 spike. In contrast, responses to HCoV-OC43 and SARS-CoV-2 nucleoproteins exhibited delayed age-dependent class-switching and were not elevated in JIA, JDM, and JSLE patients, which argues against increased exposure. CONCLUSIONS: Consequently, autoimmune rheumatic diseases and their treatment were associated with a favorable ratio of spike to nucleoprotein antibodies. FUNDING: This work was supported by a Centre of Excellence Centre for Adolescent Rheumatology Versus Arthritis grant, 21593, UKRI funding reference MR/R013926/1, the Great Ormond Street Children's Charity, Cure JM Foundation, Myositis UK, Lupus UK, and the NIHR Biomedical Research Centres at GOSH and UCLH. This work was supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK, the UK Medical Research Council, and the Wellcome Trust.


Subject(s)
Autoimmune Diseases , COVID-19 , Coronavirus OC43, Human , Rheumatic Diseases , Adolescent , Adult , Antibodies, Viral , Antibody Formation , COVID-19/complications , Child , Humans , Immunoglobulin G , Nucleoproteins , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Systemic Inflammatory Response Syndrome
3.
Nat Cancer ; 2(12): 1321-1337, 2021 12.
Article in English | MEDLINE | ID: mdl-35121900

ABSTRACT

Patients with cancer have higher COVID-19 morbidity and mortality. Here we present the prospective CAPTURE study, integrating longitudinal immune profiling with clinical annotation. Of 357 patients with cancer, 118 were SARS-CoV-2 positive, 94 were symptomatic and 2 died of COVID-19. In this cohort, 83% patients had S1-reactive antibodies and 82% had neutralizing antibodies against wild type SARS-CoV-2, whereas neutralizing antibody titers against the Alpha, Beta and Delta variants were substantially reduced. S1-reactive antibody levels decreased in 13% of patients, whereas neutralizing antibody titers remained stable for up to 329 days. Patients also had detectable SARS-CoV-2-specific T cells and CD4+ responses correlating with S1-reactive antibody levels, although patients with hematological malignancies had impaired immune responses that were disease and treatment specific, but presented compensatory cellular responses, further supported by clinical recovery in all but one patient. Overall, these findings advance the understanding of the nature and duration of the immune response to SARS-CoV-2 in patients with cancer.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/immunology , Neoplasms/complications , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/blood , COVID-19/mortality , Female , Follow-Up Studies , Humans , Immunity, Cellular , Male , Middle Aged , Neoplasms/blood , Neoplasms/immunology , Prospective Studies , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Young Adult
4.
Science ; 370(6522): 1339-1343, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33159009

ABSTRACT

Zoonotic introduction of novel coronaviruses may encounter preexisting immunity in humans. Using diverse assays for antibodies recognizing SARS-CoV-2 proteins, we detected preexisting humoral immunity. SARS-CoV-2 spike glycoprotein (S)-reactive antibodies were detectable using a flow cytometry-based method in SARS-CoV-2-uninfected individuals and were particularly prevalent in children and adolescents. They were predominantly of the immunoglobulin G (IgG) class and targeted the S2 subunit. By contrast, SARS-CoV-2 infection induced higher titers of SARS-CoV-2 S-reactive IgG antibodies targeting both the S1 and S2 subunits, and concomitant IgM and IgA antibodies, lasting throughout the observation period. SARS-CoV-2-uninfected donor sera exhibited specific neutralizing activity against SARS-CoV-2 and SARS-CoV-2 S pseudotypes. Distinguishing preexisting and de novo immunity will be critical for our understanding of susceptibility to and the natural course of SARS-CoV-2 infection.


Subject(s)
Antibodies, Viral/blood , COVID-19/immunology , Immunity, Humoral , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , COVID-19/blood , Epitope Mapping , Female , HEK293 Cells , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Middle Aged , SARS-CoV-2/chemistry , Spike Glycoprotein, Coronavirus/chemistry , Viral Zoonoses/blood , Viral Zoonoses/immunology , Young Adult
5.
Front Immunol ; 11: 376, 2020.
Article in English | MEDLINE | ID: mdl-32194571

ABSTRACT

Dendritic cells (DCs) are specialized antigen presenting cells that instruct T cell responses through sensing environmental and inflammatory danger signals. Maintaining the homeostasis of the multiple functionally distinct conventional dendritic cells (cDC) subsets that exist in vivo is crucial for regulating immune responses, with changes in numbers sufficient to break immune tolerance. Using Ptpn22-/- mice we demonstrate that the phosphatase PTPN22 is a highly selective, negative regulator of cDC2 homeostasis, preventing excessive population expansion from as early as 3 weeks of age. Mechanistically, PTPN22 mediates cDC2 homeostasis in a cell intrinsic manner by restricting cDC2 proliferation. A single nucleotide polymorphism, PTPN22R620W, is one of the strongest genetic risk factors for multiple autoantibody associated human autoimmune diseases. We demonstrate that cDC2 are also expanded in mice carrying the orthologous PTPN22619W mutation. As a consequence, cDC2 dependent CD4+ T cell proliferation and T follicular helper cell responses are increased. Collectively, our data demonstrate that PTPN22 controls cDC2 homeostasis, which in turn ensures appropriate cDC2-dependent T cell responses under antigenic challenge. Our findings provide a link between perturbations in DC development and susceptibility to a broad spectrum of PTPN22R620W associated human autoimmune diseases.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Homeostasis/immunology , Immune Tolerance/immunology , Mice , Mice, Transgenic , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
6.
J Cell Sci ; 133(5)2019 10 16.
Article in English | MEDLINE | ID: mdl-31471459

ABSTRACT

Effector T-cells rely on integrins to drive adhesion and migration to facilitate their immune function. The heterodimeric transmembrane integrin LFA-1 (αLß2 integrin) regulates adhesion and migration of effector T-cells through linkage of the extracellular matrix with the intracellular actin treadmill machinery. Here, we quantified the velocity and direction of F-actin flow in migrating T-cells alongside single-molecule localisation of transmembrane and intracellular LFA-1. Results showed that actin retrograde flow positively correlated and immobile actin negatively correlated with T-cell velocity. Plasma membrane-localised LFA-1 forms unique nano-clustering patterns in the leading edge, compared to the mid-focal zone, of migrating T-cells. Deleting the cytosolic phosphatase PTPN22, loss-of-function mutations of which have been linked to autoimmune disease, increased T-cell velocity, and leading-edge co-clustering of pY397 FAK, pY416 Src family kinases and LFA-1. These data suggest that differential nanoclustering patterns of LFA-1 in migrating T-cells may instruct intracellular signalling. Our data presents a paradigm where T-cells modulate the nanoscale organisation of adhesion and signalling molecules to fine tune their migration speed, with implications for the regulation of immune and inflammatory responses.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cell Movement , Lymphocyte Function-Associated Antigen-1/metabolism , T-Lymphocytes/cytology , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion , Cell Membrane/metabolism , Cells, Cultured , Female , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Signal Transduction
7.
Sci Rep ; 8(1): 12692, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30139951

ABSTRACT

A C1858T single nucleotide polymorphism within PTPN22 (which encodes PTPN22R620W) is associated with an enhanced susceptibility to multiple autoimmune diseases including type 1 diabetes and rheumatoid arthritis. Many of the associated autoimmune diseases have an autoantibody component to their pathology. Fc receptors (FcRs) recognise autoantibodies when they bind to autoantigens and form immune complexes. After immune complex binding and receptor crosslinking, FcRs signal via Src and Syk family kinases, leading to antigen uptake, presentation and cytokine secretion. Ptpn22 encodes a protein tyrosine phosphatase that negatively regulates Src and Syk family kinases proximal to immunoreceptor signalling cascades. We therefore hypothesised that PTPN22 regulates immune complex stimulated FcR responses in dendritic cells (DCs). Bone marrow derived DCs (BMDCs) from wild type (WT) or Ptpn22-/- mice were pulsed with ovalbumin:anti-ovalbumin immune complexes (ova ICs). Co-culture with WT OT-II T cells revealed that ova IC pulsed Ptpn22-/- BMDCs have an enhanced capability to induce T cell proliferation. This was associated with an increased capability of Ptpn22-/- BMDCs to present immune complex derived antigens and to form ova IC dependent DC-T cell conjugates. These findings highlight PTPN22 as a regulator of FcR mediated responses and provide a link between the association of PTPN22R620W with autoantibody associated autoimmune diseases.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Animals , Antigen-Antibody Complex/metabolism , Bone Marrow Cells/cytology , Cell Proliferation/physiology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Genetic Predisposition to Disease/genetics , Humans , Mice , Mice, Knockout , Polymorphism, Single Nucleotide/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Syk Kinase/genetics , Syk Kinase/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
8.
J Autoimmun ; 94: 45-55, 2018 11.
Article in English | MEDLINE | ID: mdl-30054208

ABSTRACT

A missense C1858T single nucleotide polymorphism within PTPN22 is a strong genetic risk factor for the development of multiple autoimmune diseases. PTPN22 encodes a protein tyrosine phosphatase that negatively regulates immuno-receptor proximal Src and Syk family kinases. Notably, PTPN22 negatively regulates kinases downstream of T-cell receptor (TCR) and LFA-1, thereby setting thresholds for T-cell activation. Alterations to the quality of TCR and LFA-1 engagement at the immune synapse and the regulation of downstream signals can have profound effects on the type of effector T-cell response induced. Here we describe how IFNγ+ Th1 responses are potentiated in Ptpn22-/- T-cells and in T-cells from mice expressing Ptpn22R619W (the mouse orthologue of the human genetic variant) as they age, or following repeated immune challenge, and explore the mechanisms contributing to the expansion of Th1 cells. Specifically, we uncover two LFA-1-ICAM dependent mechanisms; one T-cell intrinsic, and one T-cell extrinsic. Firstly, we found that in vitro anti-CD3/LFA-1 induced Th1 responses were enhanced in Ptpn22-/- T-cells compared to WT, whereas anti-CD3/anti-CD28 induced IFNy responses were similar. These data were associated with an enhanced ability of Ptpn22-/- T-cells to engage ICAM-1 at the immune synapse when incubated on planar lipid bilayers, and to form conjugates with dendritic cells. Secondly, we observed a T-cell extrinsic mechanism whereby repeated stimulation of WT OT-II T-cells with LPS and OVA323-339 pulsed Ptpn22-/- bone marrow derived dendritic cells (BMDCs) was sufficient to enhance Th1 cell development compared to WT BMDCs. Furthermore, this response could be reversed by LFA-1 blockade. Our data point to two related but distinct mechanisms by which PTPN22 regulates LFA-1 dependent signals to enhance Th1 development, highlighting how perturbations to PTPN22 function over time to regulate the balance of the immune response.


Subject(s)
Arthritis, Experimental/immunology , Dendritic Cells/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Th1 Cells/immunology , Animals , Antibodies/pharmacology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/antagonists & inhibitors , CD3 Complex/genetics , CD3 Complex/immunology , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/pathology , Gene Expression Regulation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Lipid Bilayers/chemistry , Lipid Bilayers/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Function-Associated Antigen-1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/pharmacology , Peptide Fragments/pharmacology , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/drug effects , Th1 Cells/pathology
9.
Eur J Immunol ; 48(2): 306-315, 2018 02.
Article in English | MEDLINE | ID: mdl-28948613

ABSTRACT

A single nucleotide polymorphism within the PTPN22 gene is a strong genetic risk factor predisposing to the development of multiple autoimmune diseases. PTPN22 regulates Syk and Src family kinases downstream of immuno-receptors. Fungal ß-glucan receptor dectin-1 signals via Syk, and dectin-1 stimulation induces arthritis in mouse models. We investigated whether PTPN22 regulates dectin-1 dependent immune responses. Bone marrow derived dendritic cells (BMDCs) generated from C57BL/6 wild type (WT) and Ptpn22-/- mutant mice, were pulsed with OVA323-339 and the dectin-1 agonist curdlan and co-cultured in vitro with OT-II T-cells or adoptively transferred into OT-II mice, and T-cell responses were determined by immunoassay. Dectin-1 activated Ptpn22-/- BMDCs enhanced T-cell secretion of IL-17 in vitro and in vivo in an IL-1ß dependent manner. Immunoblotting revealed that compared to WT, dectin-1 activated Ptpn22-/- BMDCs displayed enhanced Syk and Erk phosphorylation. Dectin-1 activation of BMDCs expressing Ptpn22R619W (the mouse orthologue of human PTPN22R620W ) also resulted in increased IL-1ß secretion and T-cell dependent IL-17 responses, indicating that in the context of dectin-1 Ptpn22R619W operates as a loss-of-function variant. These findings highlight PTPN22 as a novel regulator of dectin-1 signals, providing a link between genetically conferred perturbations of innate receptor signaling and the risk of autoimmune disease.


Subject(s)
Autoimmune Diseases/genetics , Dendritic Cells/physiology , Lectins, C-Type/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Th17 Cells/immunology , Animals , Cells, Cultured , Coculture Techniques , Humans , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Risk , Signal Transduction
10.
PLoS One ; 12(10): e0186625, 2017.
Article in English | MEDLINE | ID: mdl-29040339

ABSTRACT

The PTPN22R620W single nucleotide polymorphism increases the risk of developing multiple autoimmune diseases including type 1 diabetes, rheumatoid arthritis and lupus. PTPN22 is highly expressed in antigen presenting cells (APCs) where the expression of the murine disease associated variant orthologue (Ptpn22R619W) is reported to dysregulate pattern recognition receptor signalling in dendritic cells (DCs) and promote T-cell proliferation. Because T-cell activation is dependent on DC antigen uptake, degradation and presentation, we analysed the efficiency of these functions in splenic and GM-CSF bone marrow derived DC from wild type (WT), Ptpn22-/- or Ptpn22R619W mutant mice. Results indicated no differential ability of DCs to uptake antigen via macropinocytosis or receptor-mediated endocytosis. Antigen degradation and presentation was also equal as was WT T-cell conjugate formation and subsequent T-cell proliferation. Despite the likely presence of multiple phosphatase-regulated pathways in the antigen uptake, processing and presentation pathways that we investigated, we observed that Ptpn22 and the R619W autoimmune associated variant were dispensable. These important findings indicate that under non-inflammatory conditions there is no requirement for Ptpn22 in DC dependent antigen uptake and T-cell activation. Our findings reveal that perturbations in antigen uptake and processing, a fundamental pathway determining adaptive immune responses, are unlikely to provide a mechanism for the risk associated with the Ptpn22 autoimmune associated polymorphism.


Subject(s)
Antigen Presentation , Antigens, Bacterial/chemistry , Dendritic Cells/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , T-Lymphocytes/immunology , Animals , Antigens, Bacterial/immunology , Coculture Techniques , Dendritic Cells/cytology , Endocytosis/immunology , Fluorescent Dyes/chemistry , Gene Expression , Immunological Synapses , Listeria monocytogenes/chemistry , Listeria monocytogenes/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/chemistry , Ovalbumin/immunology , Pinocytosis/immunology , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Signal Transduction , Staining and Labeling , T-Lymphocytes/cytology
11.
Bioconjug Chem ; 28(2): 481-495, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27966893

ABSTRACT

Tris(hydroxypyridinone) chelators conjugated to peptides can rapidly complex the positron-emitting isotope gallium-68 (68Ga) under mild conditions, and the resulting radiotracers can delineate peptide receptor expression at sites of diseased tissue in vivo. We have synthesized a dendritic bifunctional chelator containing nine 1,6-dimethyl-3-hydroxypyridin-4-one groups (SCN-HP9) that can coordinate up to three Ga3+ ions. This derivative has been conjugated to a trimeric peptide (RGD3) containing three peptide groups that target the αvß3 integrin receptor. The resulting dendritic compound, HP9-RGD3, can be radiolabeled in 97% radiochemical yield at a 3-fold higher specific activity than its homologues HP3-RGD and HP3-RGD3 that contain only a single metal binding site. PET scanning and biodistribution studies show that [68Ga(HP9-RGD3)] demonstrates higher receptor-mediated tumor uptake in animals bearing U87MG tumors that overexpress αvß3 integrin than [68Ga(HP3-RGD)] and [68Ga(HP3-RGD3)]. However, concomitant nontarget organ retention of [68Ga(HP9-RGD3)] results in low tumor to nontarget organ contrast in PET images. On the other hand, the trimeric peptide homologue containing a single tris(hydroxypyridinone) chelator, [68Ga(HP3-RGD3)], clears nontarget organs and exhibits receptor-mediated uptake in mice bearing tumors and in mice with induced rheumatoid arthritis. PET imaging with [68Ga(HP3-RGD3)] enables clear delineation of αvß3 integrin receptor expression in vivo.


Subject(s)
Chelating Agents/chemistry , Gallium Radioisotopes/chemistry , Integrin alphaVbeta3/analysis , Oligopeptides/chemistry , Positron-Emission Tomography/methods , Pyridines/chemistry , Animals , Arthritis, Rheumatoid/diagnostic imaging , Chelating Agents/pharmacokinetics , Female , Gallium Radioisotopes/pharmacokinetics , Joints/diagnostic imaging , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/diagnostic imaging , Oligopeptides/pharmacokinetics , Pyridines/pharmacokinetics , Tissue Distribution
12.
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
13.
PLoS One ; 7(2): e32193, 2012.
Article in English | MEDLINE | ID: mdl-22363815

ABSTRACT

NF-κB signalling plays an essential role in T cell activation and generation of regulatory and memory populations in vivo. In the present study, we aimed to investigate the role of NF-κB signalling in post-activation T cells using tissue specific ablation of inhibitor of kappa-B kinase 2 expression, an important component of the inhibitor of kappa-B kinase complex in canonical NF-κB signalling. The OX40 antigen is expressed on activated T cells. Therefore, we used previously described mouse strain expressing Cre recombinase from the endogenous OX40 locus. Ablation of IKK2 expression using OX40(Cre) mice resulted in the development of an inflammatory response in the skin epidermis causing wide spread skin lesions. The inflammatory response was characterised by extensive leukocytic infiltrate in skin tissue, hyperplasia of draining lymph nodes and widespread activation in the T cell compartment. Surprisingly, disease development did not depend on T cells but was rather associated with an unanticipated expression of Cre in skin epidermis, and activation of the T cell compartment did not require Ikbk2 deletion in T cells. Employment of Cre reporter strains revealed extensive Cre activity in skin epidermis. Therefore, development of skin lesions was rather more likely explained by deletion of Ikbk2 in skin keratinocytes in OX40(Cre) mice.


Subject(s)
Epidermis/enzymology , Gene Deletion , Genetic Loci/genetics , I-kappa B Kinase/metabolism , Integrases/metabolism , Receptors, OX40/metabolism , Animals , Apoptosis , Epidermis/immunology , Epidermis/pathology , Epithelium/pathology , Genes, Reporter/genetics , Homeodomain Proteins/metabolism , Hypertrophy , Inflammation/pathology , Lymphocyte Activation/immunology , Lymphoid Tissue/pathology , Mice , Organ Specificity , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/biosynthesis
14.
Proc Natl Acad Sci U S A ; 107(45): 19461-6, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20978210

ABSTRACT

CD31 is an Ig-like molecule expressed by leukocytes and endothelial cells with an established role in the regulation of leukocyte trafficking. Despite genetic deletion of CD31 being associated with exacerbation of T cell-mediated autoimmunity, the contribution of this molecule to T-cell responses is largely unknown. Here we report that tumor and allograft rejection are significantly enhanced in CD31-deficient mice, which are also resistant to tolerance induction. We propose that these effects are dependent on an as yet unrecognized role for CD31-mediated homophilic interactions between T cells and antigen-presenting cells (APCs) during priming. We show that loss of CD31 interactions leads to enhanced primary clonal expansion, increased killing capacity, and diminished regulatory functions by T cells. Immunomodulation by CD31 signals correlates with a partial inhibition of proximal T-cell receptor (TCR) signaling, specifically Zap-70 phosphorylation. However, CD31-deficient mice do not develop autoimmunity due to increased T-cell death following activation, and we show that CD31 triggering induces Erk-mediated prosurvival activity in T cells either in conjunction with TCR signaling or autonomously. We conclude that CD31 functions as a nonredundant comodulator of T-cell responses, which specializes in sizing the ensuing immune response by setting the threshold for T-cell activation and tolerance, while preventing memory T-cell death.


Subject(s)
Immune Tolerance , Platelet Endothelial Cell Adhesion Molecule-1/immunology , T-Lymphocytes/immunology , Animals , Cell Survival , Clone Cells , Cytotoxicity, Immunologic , Genes, Immunoglobulin , Immunologic Memory , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics
15.
Immunol Cell Biol ; 88(2): 213-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19949423

ABSTRACT

Interleukin (IL)-7 and IL-15 have non-redundant roles in promoting development of memory CD8(+) T cells. STAT5 is activated by receptors of both cytokines and has also been implicated as a requirement for generation of memory. To determine whether STAT5 activity was required for IL-7 and IL-15-mediated generation of memory, we expressed either wild type (WT) or constitutively active (CA) forms of STAT5a in normal effector cells and then observed their ability to form memory in cytokine replete or deficient hosts. Receptor-independent CA-STAT5a significantly enhanced memory formation in the absence of either cytokine but did not mediate complete rescue. Interestingly, WT-STAT5a expression enhanced memory formation in a strictly IL-7-dependent manner, suggesting that IL-7 is a more potent activator of STAT5 than IL-15 in vivo. These data suggest that the non-redundant requirement for IL-7 and IL-15 is mediated through differential activation of both STAT5-dependent and STAT5-independent pathways.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-15/immunology , Interleukin-7/immunology , STAT5 Transcription Factor/immunology , Animals , Mice , Mice, Transgenic , Mutant Proteins/metabolism , Phosphorylation , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-7/metabolism , Retroviridae/genetics , Transduction, Genetic
16.
Nat Immunol ; 9(5): 513-21, 2008 May.
Article in English | MEDLINE | ID: mdl-18391955

ABSTRACT

Phosphatidylinositol-3-OH kinase (PI(3)K) and the nutrient sensor mTOR are evolutionarily conserved regulators of cell metabolism. Here we show that PI(3)K and mTOR determined the repertoire of adhesion and chemokine receptors expressed by T lymphocytes. The key lymph node-homing receptors CD62L (L-selectin) and CCR7 were highly expressed on naive T lymphocytes but were downregulated after immune activation. CD62L downregulation occurred through ectodomain proteolysis and suppression of gene transcription. The p110delta subunit of PI(3)K controlled CD62L proteolysis through mitogen-activated protein kinases, whereas control of CD62L transcription by p110delta was mediated by mTOR through regulation of the transcription factor KLF2. PI(3)K-mTOR nutrient-sensing pathways also determined expression of the chemokine receptor CCR7 and regulated lymphocyte trafficking in vivo. Hence, lymphocytes use PI(3)K and mTOR to match metabolism and trafficking.


Subject(s)
Phosphatidylinositol 3-Kinases/physiology , Protein Kinases/physiology , T-Lymphocytes/immunology , Animals , Cell Movement , L-Selectin/metabolism , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR7/metabolism , Signal Transduction , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases
17.
Blood ; 108(2): 600-8, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16569767

ABSTRACT

Although interleukin 2 (IL-2) and IL-15 signal through the common gamma chain (gammac) and through IL-2 receptor beta-chain (CD122) subunits, they direct distinct physiologic and immunotherapeutic responses in T cells. The present study provides some insight into why IL-2 and IL-15 differentially regulate T-cell function by revealing that these cytokines are strikingly distinct in their ability to control protein synthesis and T-cell mass. IL-2 and IL-15 are shown to be equivalent mitogens for antigen-stimulated CD8(+) T cells but not for equivalent growth factors. Antigen-primed T cells cannot autonomously maintain amino acid incorporation or de novo protein synthesis without exogenous cytokine stimulation. Both IL-2 and IL-15 induce amino acid uptake and protein synthesis in antigen-activated T cells; however, the IL-2 response is strikingly more potent than the IL-15 response. The differential action of IL-2 and IL-15 on amino acid uptake and protein synthesis is explained by temporal differences in signaling induced by these 2 cytokines. Hence, the present results show that cytokines that are equivalent mitogens can have different potency in terms of regulating protein synthesis and cell growth.


Subject(s)
Cell Proliferation/drug effects , Interleukin-15/pharmacology , Interleukin-2/pharmacology , T-Lymphocytes/cytology , Animals , Cells, Cultured , Mice , Mice, Transgenic , Mitogens/pharmacology , Protein Biosynthesis/drug effects , Signal Transduction
18.
J Virol ; 76(10): 4919-27, 2002 May.
Article in English | MEDLINE | ID: mdl-11967309

ABSTRACT

To identify cell proteins regulated by the Epstein-Barr virus (EBV) transcription factor EBNA-2, we analyzed a cell line with conditional EBNA-2 activity by using microarray expression profiling. This led to the identification of two novel target genes induced by EBNA-2. The first of these, interleukin-16, is an immunomodulatory cytokine involved in the regulation of CD4 T cells. The second, AML-2, is a member of the Runt domain family of transcription factors. Quiescent B cells initially expressed AML-1 but, 48 h after virus infection, the levels of AML-1 decreased dramatically, whereas the amount of AML-2 protein increased. Analysis of a panel of B-cell lines indicated that AML-2 expression is normally predominant in EBV latency III, whereas AML-1 is associated with EBV latency I or EBV-negative cells. The AML genes are the first example of cell transcription factors whose expression correlates with the latency I/III phenotype.


Subject(s)
B-Lymphocytes/virology , Cell Transformation, Viral/genetics , DNA-Binding Proteins/biosynthesis , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human , Proto-Oncogene Proteins , Transcription Factors/biosynthesis , Adaptor Proteins, Signal Transducing , B-Lymphocytes/metabolism , Burkitt Lymphoma , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Core Binding Factor Alpha 2 Subunit , Core Binding Factor Alpha 3 Subunit , Cytoskeletal Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Humans , Interleukin-16/metabolism , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Lymphocyte Activation/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Virus Latency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...