Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Sci Transl Med ; 11(490)2019 05 01.
Article in English | MEDLINE | ID: mdl-31043571

ABSTRACT

Interference with immune cell proliferation represents a successful treatment strategy in T cell-mediated autoimmune diseases such as rheumatoid arthritis and multiple sclerosis (MS). One prominent example is pharmacological inhibition of dihydroorotate dehydrogenase (DHODH), which mediates de novo pyrimidine synthesis in actively proliferating T and B lymphocytes. Within the TERIDYNAMIC clinical study, we observed that the DHODH inhibitor teriflunomide caused selective changes in T cell subset composition and T cell receptor repertoire diversity in patients with relapsing-remitting MS (RRMS). In a preclinical antigen-specific setup, DHODH inhibition preferentially suppressed the proliferation of high-affinity T cells. Mechanistically, DHODH inhibition interferes with oxidative phosphorylation (OXPHOS) and aerobic glycolysis in activated T cells via functional inhibition of complex III of the respiratory chain. The affinity-dependent effects of DHODH inhibition were closely linked to differences in T cell metabolism. High-affinity T cells preferentially use OXPHOS during early activation, which explains their increased susceptibility toward DHODH inhibition. In a mouse model of MS, DHODH inhibitory treatment resulted in preferential inhibition of high-affinity autoreactive T cell clones. Compared to T cells from healthy controls, T cells from patients with RRMS exhibited increased OXPHOS and glycolysis, which were reduced with teriflunomide treatment. Together, these data point to a mechanism of action where DHODH inhibition corrects metabolic disturbances in T cells, which primarily affects profoundly metabolically active high-affinity T cell clones. Hence, DHODH inhibition may promote recovery of an altered T cell receptor repertoire in autoimmunity.


Subject(s)
Crotonates/therapeutic use , Mitochondria/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Toluidines/therapeutic use , Aerobiosis/drug effects , Animals , Cell Proliferation/drug effects , Cell Respiration/drug effects , Crotonates/pharmacology , Dihydroorotate Dehydrogenase , Electron Transport Complex III/metabolism , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Humans , Hydroxybutyrates , Lymphocyte Activation/drug effects , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Mitochondria/drug effects , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis, Relapsing-Remitting/immunology , Nitriles , Oxidative Phosphorylation/drug effects , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , Toluidines/pharmacology
2.
Proc Natl Acad Sci U S A ; 113(41): E6182-E6191, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27671636

ABSTRACT

Molecular mechanisms that determine lesion localization or phenotype variation in multiple sclerosis are mostly unidentified. Although transmigration of activated encephalitogenic T cells across the blood-brain barrier (BBB) is a crucial step in the disease pathogenesis of CNS autoimmunity, the consequences on brain endothelial barrier integrity upon interaction with such T cells and subsequent lesion formation and distribution are largely unknown. We made use of a transgenic spontaneous mouse model of CNS autoimmunity characterized by inflammatory demyelinating lesions confined to optic nerves and spinal cord (OSE mice). Genetic ablation of a single immune-regulatory molecule in this model [i.e., B7-homolog 1 (B7-H1, PD-L1)] not only significantly increased incidence of spontaneous CNS autoimmunity and aggravated disease course, especially in the later stages of disease, but also importantly resulted in encephalitogenic T-cell infiltration and lesion formation in normally unaffected brain regions, such as the cerebrum and cerebellum. Interestingly, B7-H1 ablation on myelin oligodendrocyte glycoprotein-specific CD4+ T cells, but not on antigen-presenting cells, amplified T-cell effector functions, such as IFN-γ and granzyme B production. Therefore, these T cells were rendered more capable of eliciting cell contact-dependent brain endothelial cell dysfunction and increased barrier permeability in an in vitro model of the BBB. Our findings suggest that a single immune-regulatory molecule on T cells can be ultimately responsible for localized BBB breakdown, and thus substantial changes in lesion topography in the context of CNS autoimmunity.


Subject(s)
Autoimmunity/genetics , B7-H1 Antigen/genetics , Brain/immunology , Brain/metabolism , Endothelial Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , B7-H1 Antigen/metabolism , Blood-Brain Barrier/metabolism , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental , Gene Knockout Techniques , Genetic Predisposition to Disease , Mice , Mice, Transgenic , Mortality , Permeability , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
3.
J Immunol ; 195(8): 3584-95, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26378076

ABSTRACT

It is currently acknowledged that TH17 cells are critically involved in the pathogenesis of autoimmune diseases such as multiple sclerosis (MS). In this article, we demonstrate that signals delivered by the coinhibitory molecule B7-homologue 1 (B7-H1) via a B7-homologue 1 mouse-IgG2aFc (B7-H1-Ig) fusion protein nearly abolish TH17, but not TH1 and TH2, differentiation via direct interaction with the T cell. These effects were equally pronounced in the absence of programmed death-1 or B7.1 and B7.2 on the T cell side, thus providing clear evidence that B7-H1 modulates T cell differentiation via a novel receptor. Mechanistically, B7-H1 interfered with early TCR-mediated signaling and cytokine-mediated induction of the TH17-determining transcription factors retinoic acid-related orphan receptor γ t and IFN regulator factor-4 in a programmed death-1 and B7-independent fashion. In an animal model of MS, active myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, B7-H1-Ig exhibited a significant and long-lasting effect on disease severity upon administration during the first 5 d of the priming phase, which was accompanied by reduced TH17 responses in the periphery and within the CNS. Importantly, B7-H1-Ig was even capable of interfering with T cell encephalitogenicity when interaction with the T cells occurred after priming using an adoptive transfer experimental autoimmune encephalomyelitis model. In line with this, both naive human CD4(+) T cells and differentiated TH17 effector cells from MS patients were highly sensitive toward B7-H1-Ig-mediated TH17 suppression. Together, we propose the existence of a novel B7-H1-mediated immune-regulatory pathway in T cells, which selectively limits murine and human TH17 cell responses and might be therapeutically exploited to control TH17-mediated autoimmunity.


Subject(s)
B7-H1 Antigen/immunology , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Programmed Cell Death 1 Receptor/immunology , Th17 Cells/immunology , Animals , B7-H1 Antigen/genetics , Cell Differentiation/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Programmed Cell Death 1 Receptor/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Th17 Cells/pathology
4.
PLoS One ; 9(12): e115920, 2014.
Article in English | MEDLINE | ID: mdl-25541968

ABSTRACT

Human biospecimen collection, processing and preservation are rapidly emerging subjects providing essential support to clinical as well as basic researchers. Unlike collection of other biospecimens (e.g. DNA and serum), biobanking of viable immune cells, such as peripheral blood mononuclear cells (PBMC) and/or isolated immune cell subsets is still in its infancy. While certain aspects of processing and freezing conditions have been studied in the past years, little is known about the effect of blood transportation on immune cell survival, phenotype and specific functions. However, especially for multicentric and cooperative projects it is vital to precisely know those effects. In this study we investigated the effect of blood shipping and pre-processing delay on immune cell phenotype and function both on cellular and subcellular levels. Peripheral blood was collected from healthy volunteers (n = 9): at a distal location (shipped overnight) and in the central laboratory (processed immediately). PBMC were processed in the central laboratory and analyzed post-cryopreservation. We analyzed yield, major immune subset distribution, proliferative capacity of T cells, cytokine pattern and T-cell receptor signal transduction. Results show that overnight transportation of blood samples does not globally compromise T- cell subsets as they largely retain their phenotype and proliferative capacity. However, NK and B cell frequencies, the production of certain PBMC-derived cytokines and IL-6 mediated cytokine signaling pathway are altered due to transportation. Various control experiments have been carried out to compare issues related to shipping versus pre-processing delay on site. Our results suggest the implementation of appropriate controls when using multicenter logistics for blood transportation aiming at subsequent isolation of viable immune cells, e.g. in multicenter clinical trials or studies analyzing immune cells/subsets. One important conclusion might be that despite changes due to overnight shipment, highly standardized central processing (and analysis) could be superior to multicentric de-central processing with more difficult standardization.


Subject(s)
Blood Preservation , Leukocytes, Mononuclear/cytology , Adult , Blood Banks , Blood Specimen Collection , Cell Proliferation , Cryopreservation , Cytokines/analysis , Cytokines/immunology , Female , Humans , Interleukin-6/analysis , Interleukin-6/immunology , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Signal Transduction
5.
Neurology ; 81(10): 865-71, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23925765

ABSTRACT

OBJECTIVE: To find biomarkers identifying patients at risk for the development of progressive multifocal leukoencephalopathy (PML) during natalizumab treatment. METHODS: Patients were recruited from 10 European and US cohorts. Of 289 patients with multiple sclerosis (MS), 224 had been treated with natalizumab (18-80 months), 21 received other immune-modulatory treatments, and 28 were untreated. We had access to samples from 16 natalizumab PML patients. Eight of these patients had given blood before the diagnosis of PML. We also analyzed non-natalizumab-treated patients who developed PML (n = 10) and age- and sex-matched healthy donors (n = 31). All flow cytometric assessments were done on previously cryopreserved, viable peripheral blood mononuclear cells. RESULTS: The percentage of l-selectin-expressing CD4+ T cells was significantly lower in patients treated long-term with natalizumab (40.2%) when compared with patients not receiving natalizumab treatment (47.2%; p = 0.016) or healthy controls (61.0%; p < 0.0001). An unusually low percentage (9-fold lower; 4.6%) was highly correlated with the risk of developing PML in the patient group with available pre-PML samples when compared with non-PML natalizumab-treated patients (p ≤ 0.0001). Samples were gathered between 4 and 26 months before PML diagnosis. CONCLUSIONS: The cell-based assessment of the percentage of l-selectin-expressing CD4 T cells could provide an urgently needed biomarker for individual PML risk assessment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , L-Selectin/blood , Leukoencephalopathy, Progressive Multifocal/blood , Multiple Sclerosis/blood , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers/blood , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cohort Studies , Humans , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/pathology , Multiple Sclerosis/diagnosis , Multiple Sclerosis/pathology , Natalizumab , Risk Factors , Treatment Outcome
6.
Cancer Immunol Immunother ; 60(1): 23-35, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20857101

ABSTRACT

Frequent expression of cancer testis antigens (CTA) has been consistently observed in head and neck squamous cell carcinomas (HNSCC). For instance, in 52 HNSCC patients, MAGE-A3 and -A4 CTA were expressed in over 75% of tumors, regardless of the sites of primary tumors such as oral cavity or hypopharynx. Yet, T-cell responses against these CTA in tumor-bearing patients have not been investigated in detail. In this study, we assessed the naturally acquired T-cell response against MAGE-A3 and -A4 in nonvaccinated HNSCC patients. Autologous antigen-presenting cells pulsed with overlapping peptide pools were used to detect and isolate MAGE-A3 and MAGE-A4 specific CD4(+) T cells from healthy donors and seven head and neck cancer patients. CD4(+) T-cell clones were characterized by cytokine secretion. We could detect and isolate MAGE-A3 and MAGE-A4 specific CD4(+) T cells from 7/7 cancer patients analyzed. Moreover, we identified six previously described and three new epitopes for MAGE-A3. Among them, the MAGE-A3(111-125) and MAGE-A3(161-175) epitopes were shown to be naturally processed and presented by DC in association with HLA-DP and DR, respectively. All of the detected MAGE-A4 responses were specific for new helper epitopes. These data suggest that naturally acquired CD4(+) T-cell responses against CT antigens often occur in vivo in HNSCC cancer patients and provide a rationale for the development of active immunotherapeutic approaches in this type of tumor.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Squamous Cell/immunology , Epitopes, T-Lymphocyte/metabolism , Head and Neck Neoplasms/immunology , Immunotherapy, Adoptive , Aged , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Female , HLA-DP Antigens/metabolism , HLA-DR Antigens/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , Lymphocyte Activation , Male , Middle Aged , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding
7.
J Immunol ; 184(9): 4936-46, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20351194

ABSTRACT

Protective adaptive immune responses rely on TCR-mediated recognition of Ag-derived peptides presented by self-MHC molecules. However, self-Ag (tumor)-specific TCRs are often of too low affinity to achieve best functionality. To precisely assess the relationship between TCR-peptide-MHC binding parameters and T cell function, we tested a panel of sequence-optimized HLA-A(*)0201/NY-ESO-1(157-165)-specific TCR variants with affinities lying within physiological boundaries to preserve antigenic specificity and avoid cross-reactivity, as well as two outliers (i.e., a very high- and a low-affinity TCR). Primary human CD8 T cells transduced with these TCRs demonstrated robust correlations between binding measurements of TCR affinity and avidity and the biological response of the T cells, such as TCR cell-surface clustering, intracellular signaling, proliferation, and target cell lysis. Strikingly, above a defined TCR-peptide-MHC affinity threshold (K(D) < approximately 5 muM), T cell function could not be further enhanced, revealing a plateau of maximal T cell function, compatible with the notion that multiple TCRs with slightly different affinities participate equally (codominantly) in immune responses. We propose that rational design of improved self-specific TCRs may not need to be optimized beyond a given affinity threshold to achieve both optimal T cell function and avoidance of the unpredictable risk of cross-reactivity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/genetics , HLA-A Antigens/genetics , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Peptide Fragments/genetics , Protein Binding/genetics , Protein Binding/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics
8.
Methods Mol Biol ; 626: 231-49, 2010.
Article in English | MEDLINE | ID: mdl-20099132

ABSTRACT

Large numbers and functionally competent T cells are required to protect from diseases for which antibody-based vaccines have consistently failed (1), which is the case for many chronic viral infections and solid tumors. Therefore, therapeutic vaccines aim at the induction of strong antigen-specific T-cell responses. Novel adjuvants have considerably improved the capacity of synthetic vaccines to activate T cells, but more research is necessary to identify optimal compositions of potent vaccine formulations. Consequently, there is a great need to develop accurate methods for the efficient identification of antigen-specific T cells and the assessment of their functional characteristics directly ex vivo. In this regard, hundreds of clinical vaccination trials have been implemented during the last 15 years, and monitoring techniques become more and more standardized.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Drug Evaluation, Preclinical/methods , Lymphocyte Activation/drug effects , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Cells, Cultured , Humans
9.
J Immunol ; 180(3): 1634-42, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209059

ABSTRACT

The transmembrane adaptor protein SIT is a negative regulator of TCR-mediated signaling. However, little is known about the functional role of SIT in mature T cells. In this study, we show that mice deficient for SIT display a decreased number of naive CD8(+) T cells and a progressive accumulation of memory-like (CD44(high)) CD8(+) T lymphocytes that resemble cells undergoing homeostatic proliferation. Indeed, when transferred into lymphopenic hosts, SIT(-/-) naive CD8(+) T cells undergo enhanced homeostatic proliferation and express a higher level of CD44 in comparison to wild-type T cells. By using class-I-restricted TCR transgenic models with different ligand affinity/avidity, we show that lymphopenia-induced homeostatic proliferation is more pronounced in cells carrying low-affinity TCRs. Strikingly, the loss of SIT induces homeostatic proliferation of HY TCR transgenic cells, which are normally unable to proliferate in lymphopenic mice. Collectively, these data demonstrate that SIT negatively regulates T cell homeostasis. Finally, we show that SIT-deficient T cells develop a mechanism analogous to sensory adaptation as they up-regulate CD5, down-regulate the coreceptor, and display impaired TCR-mediated ZAP-70 activation.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Homeostasis/genetics , Hyaluronan Receptors/analysis , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Knockout , Signal Transduction
10.
Mol Cell Biol ; 25(17): 7557-68, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16107703

ABSTRACT

SIT is a transmembrane adapter protein that modulates signals emanating from the T-cell receptor (TCR). Here, we have used gene-targeted mice to assess the role of SIT for T-cell development and peripheral T-cell functions. SIT(-/-) double-positive thymocytes show an upregulation of the activation markers CD5 and CD69, suggesting that SIT negatively regulates TCR-mediated signals at the CD4(+) CD8(+) stage of thymic development. This assumption is further supported by the observation that in female H-Y TCR transgenic mice, positive selection is enhanced and even converted to negative selection. Similarly, mature peripheral T cells are hyperresponsive towards TCR-mediated stimuli and produce larger amounts of T-helper 1 (TH1) cytokines, and SIT-deficient mice show an increased susceptibility to develop experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. These results demonstrate that SIT is a critical negative regulator of TCR-mediated signaling and finely tunes the signals required for thymic selection and peripheral T-cell activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Membrane Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Up-Regulation
11.
Semin Immunol ; 17(1): 35-49, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15582487

ABSTRACT

The basic mechanisms of lymphocyte activation are well established, with stimulation via the antigen receptor inducing a rapid wave of tyrosine phosphorylation that requires the coordinated action of multiple protein tyrosine kinase families. This in turn leads to the generation of second messengers like Ca(2+), diacylglycerol (DAG) and inositoltrisphosphate (IP(3)) as well as the activation of effector molecules like Ras and the mitogen-activated protein kinases (MAPKs) and ultimately in activation of the transcription factors NFAT, AP-1, and NF-kappaB, resulting in gene transcription. Researchers, hoping to interconnect these events, have identified a multitude of proteins, among which is a relatively new group, the transmembrane adaptor proteins (TRAPs). TRAPs are unique in that they lack extracellular ligands and possess neither enzymatic nor transcriptional activity, but rather serve as scaffolds providing docking sites for other proteins and thereby serving to coordinate signals proximal to the membrane. Our study of these novel molecules is shedding new insights into the positive and negative regulatory mechanisms which fine tune antigen receptor signaling.


Subject(s)
Lymphocyte Activation/immunology , Phosphotyrosine/immunology , Protein-Tyrosine Kinases/immunology , Second Messenger Systems/immunology , Signal Transduction/immunology , Cell Membrane/immunology , Humans , Transcription Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...