Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mult Scler ; 23(7): 1025-1030, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27481205

ABSTRACT

BACKGROUND: Distinct lesion topography in relapsing-remitting multiple sclerosis (RRMS) might be due to different antigen presentation and/or trafficking routes of immune cells into the central nervous system (CNS). OBJECTIVE: To investigate whether distinct lesion patterns in multiple sclerosis (MS) might be associated with a predominance of distinct circulating T-helper cell subset as well as their innate counterparts. METHODS: Flow cytometric analysis of lymphocytes derived from the peripheral blood of patients with exclusively cerebral (n = 20) or predominantly spinal (n = 12) disease manifestation. RESULTS: Patients with exclusively cerebral or preferential spinal lesion manifestation were associated with increased proportions of circulating granulocyte-macrophage colony-stimulating factor (GM-CSF) producing TH1 cells or interleukin (IL)-17-producing TH17 cells, respectively. In contrast, proportions of peripheral IL-17/IL-22-producing lymphoid tissue inducer (LTi), the innate counterpart of TH17 cells, were enhanced in RRMS patients with exclusively cerebral lesion topography. CONCLUSIONS: Distinct T-helper and T-helper-like innate lymphoid cell (ILC) subsets are associated with different lesion topography in RRMS.


Subject(s)
Brain/immunology , Immunity, Innate , Lymphocytes/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Spinal Cord/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Aged , Biomarkers/blood , Brain/diagnostic imaging , Brain/metabolism , Case-Control Studies , Disability Evaluation , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Humans , Immunophenotyping/methods , Interleukin-17/blood , Interleukins/blood , Lymphocytes/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Phenotype , Spinal Cord/diagnostic imaging , Spinal Cord/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism , Young Adult , Interleukin-22
2.
Proc Natl Acad Sci U S A ; 113(21): E2973-82, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162345

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) resulting from a breakdown in peripheral immune tolerance. Although a beneficial role of natural killer (NK)-cell immune-regulatory function has been proposed, it still needs to be elucidated whether NK cells are functionally impaired as part of the disease. We observed NK cells in active MS lesions in close proximity to T cells. In accordance with a higher migratory capacity across the blood-brain barrier, CD56(bright) NK cells represent the major intrathecal NK-cell subset in both MS patients and healthy individuals. Investigating the peripheral blood and cerebrospinal fluid of MS patients treated with natalizumab revealed that transmigration of this subset depends on the α4ß1 integrin very late antigen (VLA)-4. Although no MS-related changes in the migratory capacity of NK cells were observed, NK cells derived from patients with MS exhibit a reduced cytolytic activity in response to antigen-activated CD4(+) T cells. Defective NK-mediated immune regulation in MS is mainly attributable to a CD4(+) T-cell evasion caused by an impaired DNAX accessory molecule (DNAM)-1/CD155 interaction. Both the expression of the activating NK-cell receptor DNAM-1, a genetic alteration consistently found in MS-association studies, and up-regulation of the receptor's ligand CD155 on CD4(+) T cells are reduced in MS. Therapeutic immune modulation of IL-2 receptor restores impaired immune regulation in MS by increasing the proportion of CD155-expressing CD4(+) T cells and the cytolytic activity of NK cells.


Subject(s)
Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Multiple Sclerosis/immunology , Receptors, Interleukin-2/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Blood-Brain Barrier/pathology , CD4-Positive T-Lymphocytes/pathology , Cell Movement/drug effects , Female , Humans , K562 Cells , Killer Cells, Natural/pathology , Male , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Natalizumab/administration & dosage , Receptors, Virus/immunology
3.
Neurol Neuroimmunol Neuroinflamm ; 3(1): e183, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26767188

ABSTRACT

OBJECTIVE: To evaluate the effect of dimethyl fumarate (DMF; Tecfidera, Biogen, Weston, MA) on CD4(+) and CD8(+) T cell subsets in patients with multiple sclerosis (MS). METHODS: Peripheral lymphocyte subsets, including CD4(+) and CD8(+) memory cells and T helper (TH) cells TH1, TH2, TH17, and peripheral regulatory T cell (pTreg) subpopulations were analyzed before and 6 months after onset of DMF treatment. RESULTS: CD4(+) and CD8(+) memory T cells were preferentially decreased compared to naive CD4(+) and CD8(+) T cell populations. Within the CD4(+) memory T cell population, frequencies of TH1 cells were decreased, whereas those of TH2 cells were increased and those of TH17 cells remained unaltered. Accordingly, we observed decreased production of interferon γ, granulocyte-macrophage colony-stimulating factor, tumor necrosis factor α, and interleukin (IL)-22 by CD4(+) T cells under DMF treatment, whereas the frequency of IL-4- and IL-17A-producing CD4(+) T cells remained unchanged. With regard to regulatory T cells, proportions of pTreg increased following DMF treatment. CONCLUSION: Our data demonstrate that DMF treatment of patients with MS affects predominantly memory T cells accompanied by a shift in TH cell populations, resulting in a shift toward anti-inflammatory responses. These findings indicate that monitoring of memory subsets might enhance vigilance of impaired antiviral immunity and that patients with TH1-driven disease might preferentially benefit from DMF treatment. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that DMF might preferentially reduce CD4(+) and CD8(+) memory T cells in MS.

4.
Mult Scler ; 22(8): 1048-60, 2016 07.
Article in English | MEDLINE | ID: mdl-26432858

ABSTRACT

BACKGROUND: Natalizumab treatment is associated with progressive multifocal leukoencephalopathy (PML) development. Treatment duration, prior immunosuppressant use, and JCV serostatus are currently used for risk stratification, but PML incidence stays high. Anti-JCV antibody index and L-selectin (CD62L) have been proposed as additional risk stratification parameters. OBJECTIVE: This study aimed at verifying and integrating both parameters into one algorithm for risk stratification. METHODS: Multicentric, international cohorts of natalizumab-treated MS patients were assessed for JCV index (1921 control patients and nine pre-PML patients) and CD62L (1410 control patients and 17 pre-PML patients). RESULTS: CD62L values correlate with JCV serostatus, as well as JCV index values. Low CD62L in natalizumab-treated patients was confirmed and validated as a biomarker for PML risk with the risk factor "CD62L low" increasing a patient's relative risk 55-fold (p < 0.0001). Validation efforts established 86% sensitivity/91% specificity for CD62L and 100% sensitivity/59% specificity for JCV index as predictors of PML. Using both parameters identified 1.9% of natalizumab-treated patients in the reference center as the risk group. CONCLUSIONS: Both JCV index and CD62L have merit for risk stratification and share a potential biological relationship with implications for general PML etiology. A risk algorithm incorporating both biomarkers could strongly reduce PML incidence.


Subject(s)
Antibodies, Viral/blood , JC Virus/immunology , L-Selectin/blood , Leukoencephalopathy, Progressive Multifocal/chemically induced , Multiple Sclerosis/drug therapy , Natalizumab/adverse effects , Opportunistic Infections/chemically induced , Algorithms , Biomarkers/blood , Europe , Humans , Immunocompromised Host , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/prevention & control , Leukoencephalopathy, Progressive Multifocal/virology , Multiple Sclerosis/blood , Multiple Sclerosis/diagnosis , Multiple Sclerosis/immunology , Opportunistic Infections/immunology , Opportunistic Infections/virology , Retrospective Studies , Risk Assessment , Risk Factors , Serologic Tests , Treatment Outcome
5.
BMC Neurol ; 15: 96, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26099927

ABSTRACT

BACKGROUND: In light of the increased risk of progressive multifocal encephalopathy (PML) development under long-term treatment with the monoclonal antibody natalizumab which is approved for treatment of active relapsing remitting multiple sclerosis (RRMS), there is a clear need for alternative treatment options with comparable efficacy and reduced PML risk. One such option is fingolimod, a functional sphingosin-1-receptor antagonist that has been approved as first oral drug for treatment of active RRMS. However, the optimal switching design in terms of prevention of disease reoccurrence is still unknown. Moreover, potential additive effects of both drugs on immune functions, especially with regard to migration, have not yet been evaluated. METHODS/DESIGN: This is an exploratory, open-label, monocentric, investigator-initiated clinical trial. Fifteen RRMS patients under stable treatment with natalizumab will receive one last natalizumab infusion followed by a wash-out period of 8 weeks before fingolimod treatment initiation for a period of 24 weeks. Disease activity under natalizumab and during switching will be closely monitored by assessment of relapse rate and disease severity as well as high-frequent high-resolution magnetic resonance imaging including quantitative diffusion tensor imaging. Immunological assays include longitudinal assessment of adhesion molecule expression, functional evaluation of the migratory capacity of immune cells in an in-vitro model of the blood-brain-barrier, and the quality of cellular antiviral immune responses. DISCUSSION: Our trial represents the first detailed and longitudinal functional analysis of key immunological parameters in the process of switching from natalizumab and fingolimod, especially with regard to potential additive effects of both drugs on trafficking and immune surveillance. Moreover, our study will generate valuable information about even subtle disease exacerbations as consequence of natalizumab cessation, which will help to understand whether a switching protocol containing a wash-out period of 8 weeks before fingolimod treatment is appropriate in terms of disease stability.


Subject(s)
Fingolimod Hydrochloride/therapeutic use , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/therapeutic use , Disease Progression , Humans , Prospective Studies , Risk
6.
Ann Clin Transl Neurol ; 2(2): 119-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25750917

ABSTRACT

OBJECTIVE: To evaluate the influence of Fingolimod treatment on B-cell subset composition and function in multiple sclerosis patients and its potential clinical relevance. METHODS: Subset composition and cytokine production of B cells derived from peripheral blood mononuclear cells from multiple sclerosis patients under Fingolimod treatment, untreated multiple sclerosis patients and healthy controls were analyzed by flow cytometry and ELISA. Migration of lymphocyte subsets across primary human brain microvascular endothelial cells was assessed in an in vitro transmigration assay. Cell numbers and composition of B-cell subsets in cerebrospinal fluid and peripheral blood were determined by flow cytometry. Regulatory B-cell frequencies were correlated with parameters of disease stability. RESULTS: Within the peripheral B-cell compartment of Fingolimod-treated patients, the proportion of regulatory B cells (CD38(+)CD27(-)CD24(+)CD5(+)) was significantly increased as compared to treatment-naïve multiple sclerosis patients and to healthy controls, and significantly more regulatory B cells produced Interleukin-10. Fingolimod treatment enhanced the capacity of regulatory B cells to transmigrate across brain endothelial cells in an in vitro model of the blood-brain-barrier. In line with these findings, the cerebrospinal fluid/blood ratio of total B cells and regulatory B cells was strongly increased by Fingolimod treatment, and patients exhibited increased regulatory B-cell frequencies in the cerebrospinal fluid. Finally, elevated regulatory B-cell percentages in the periphery significantly correlated with clinical and paraclinical disease stability. INTERPRETATION: These data suggest a novel and as yet unrecognized role of Fingolimod in correction of the imbalance between regulatory and effector B-cell functions in multiple sclerosis both by direct effects and indirect partitioning effects on B-cell subpopulations.

7.
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
9.
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
10.
Mult Scler ; 19(9): 1137-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23388163

ABSTRACT

BACKGROUND: The StratifyJCV® test is a qualitative assay to classify MS patients as anti-JC virus (JCV) antibody positive or negative. Quantification of anti-JCV antibody levels in serum and cerebrospinal fluid (CSF) of multiple sclerosis (MS) patients might add to the progressive multifocal leukoencephalopathy (PML) risk assessment. OBJECTIVE: The objective of this study is to test sera of patients in a quantitative anti-JCV antibody assay, and to compare the results with preexisting data from the StratifyJCV® test. METHODS: Sera of a total of 175 MS patients and matched non-MS-controls were tested for anti-JCV antibodies using glutathione S-transferase-tagged-VP1 as antigen. Antibody reactivity was quantified in arbitrary units using human immunoglobulin as standard. RESULTS: The comparison of our assay with StratifyJCV® showed good inter-assay agreement (kappa 0.6), and strong correlation for antibody reactivity (r (2) = 0.94). Discordant samples had low-reactive positivity, and a higher proportion (13% vs. 4%) tested positive in the StratifyJCV® test only. CONCLUSIONS: The method presented is a tool for the reliable quantification of anti-JCV antibodies, which demonstrates good agreement with results from StratifyJCV®. In contrast to StratifyJCV®, we pre-adsorbed all of the sera with BK virus (BKV) VP1 protein to reduce cross-reactivity. This step may account for a higher species-specificity of our assay. As such, our assay might be a promising additional tool for PML risk assessment.


Subject(s)
Antibodies, Viral/analysis , Enzyme-Linked Immunosorbent Assay/methods , JC Virus/immunology , Multiple Sclerosis/virology , Adult , Antibody Specificity/immunology , BK Virus/immunology , Capsid Proteins/immunology , Cross Reactions/immunology , Female , Humans , Male
12.
PLoS One ; 6(9): e23978, 2011.
Article in English | MEDLINE | ID: mdl-21915273

ABSTRACT

Src family kinases (SFK) control multiple processes during brain development and function. We show here that the phosphoprotein associated with glycosphigolipid-enriched microdomains (PAG)/Csk binding protein (Cbp) modulates SFK activity in the brain. The timing and localization of PAG expression overlap with Fyn and Src, both of which we find associated to PAG. We demonstrate in newborn (P1) mice that PAG negatively regulates Src family kinases (SFK). P1 Pag1(-/-) mouse brains show decreased recruitment of Csk into lipid rafts, reduced phosphorylation of the inhibitory tyrosines within SFKs, and an increase in SFK activity of >/ = 50%. While in brain of P1 mice, PAG and Csk are highly and ubiquitously expressed, little Csk is found in adult brain suggesting altered modes of SFK regulation. In adult brain Pag1-deficiency has no effect upon Csk-distribution or inhibitory tyrosine phosphorylation, but kinase activity is now reduced (-20-30%), pointing to the development of a compensatory mechanism that may involve PSD93. The distribution of the Csk-homologous kinase CHK is not altered. Importantly, since the activities of Fyn and Src are decreased in adult Pag1(-/-) mice, thus presenting the reversed phenotype of P1, this provides the first in vivo evidence for a Csk-independent positive regulatory function for PAG in the brain.


Subject(s)
Brain/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , src-Family Kinases/metabolism , Animals , Animals, Newborn , Blotting, Western , Glycosphingolipids , Immunoprecipitation , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphoproteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , src-Family Kinases/genetics
13.
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
14.
Nat Immunol ; 9(3): 272-81, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18264101

ABSTRACT

The paracaspase MALT1 is pivotal in antigen receptor-mediated lymphocyte activation and lymphomagenesis. MALT1 contains a caspase-like domain, but it is unknown whether this domain is proteolytically active. Here we report that MALT1 had arginine-directed proteolytic activity that was activated after T cell stimulation, and we identify the signaling protein Bcl-10 as a MALT1 substrate. Processing of Bcl-10 after Arg228 was required for T cell receptor-induced cell adhesion to fibronectin. In contrast, MALT1 activity but not Bcl-10 cleavage was essential for optimal activation of transcription factor NF-kappaB and production of interleukin 2. Thus, the proteolytic activity of MALT1 is central to T cell activation, which suggests a possible target for the development of immunomodulatory or anticancer drugs.


Subject(s)
Caspases/physiology , Lymphocyte Activation/immunology , NF-kappa B/metabolism , Neoplasm Proteins/physiology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , Cell Line , Electrophoresis, Gel, Two-Dimensional , Humans , Jurkat Cells , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Peptide Hydrolases/metabolism , Protein Isoforms/metabolism
15.
Eur J Immunol ; 38(1): 250-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18085663

ABSTRACT

PAG, the phosphoprotein associated with glycosphingolipid-enriched microdomains (GEM), negatively regulates Src family kinases by recruiting C-terminal Src kinase (Csk) to the membrane, where Csk phosphorylates the inhibitory tyrosine of the Src kinases. S-acylation of a CxxC motif juxtaposed to the transmembrane domain within PAG has been proposed to be responsible for targeting PAG to the lipid rafts. Here, we present the characterization of a mutant PAG molecule lacking the palmitoylation motif. We demonstrate that the mutant protein is expressed at the plasma membrane, but does not localize within the GEM. Despite being displaced, the mutant PAG molecule still binds the Src kinase Fyn and the cytoskeletal adaptor ezrin-radixin-moesin-binding phosphoprotein of 50 kDa, becomes tyrosine-phosphorylated, and recruits Csk to the membrane. Functional characterization of the mutant shows that, unlike WT PAG, it does not block proximal TCR signaling, and surprisingly enhances stromal cell-derived factor 1 (CXCL12)-induced migration. The mutant functions by depleting Csk from the GEM fractions, as apparent by changes in the phosphorylation of the inhibitory tyrosines within the Src kinases. Indeed this mechanism is supported by RNA interference of PAG, which results in enhanced migration and Src kinase activity. Our results therefore support a functional role for the compartmentalization of Src kinases within the membrane.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , Chemokine CXCL12/metabolism , Membrane Microdomains/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Blotting, Western , Chemokine CXCL12/immunology , Humans , Immunoprecipitation , Jurkat Cells , Lymphocyte Activation/immunology , Molecular Sequence Data , Mutation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Transfection
16.
Blood ; 110(2): 596-615, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17389760

ABSTRACT

In primary human T cells, anergy induction results in enhanced p59Fyn activity. Because Fyn is the kinase primarily responsible for the phosphorylation of PAG (the phosphoprotein associated with glycosphingolipid-enriched microdomains), which negatively regulates Src-kinase activity by recruiting Csk (the C-terminal Src kinase) to the membrane, we investigated whether anergy induction also affects PAG. Analysis of anergic T cells revealed that PAG is hyperphosphorylated at the Csk binding site, leading to enhanced Csk recruitment and inhibitory tyrosine phosphorylation within Fyn. This together with enhanced phosphorylation of a tyrosine within the SH2 domain of Fyn leads to the formation of a hyperactive conformation, thus explaining the enhanced Fyn kinase activity. In addition, we have also identified the formation of a multiprotein complex containing PAG, Fyn, Sam68, and RasGAP in stimulated T cells. We demonstrate that PAG-Fyn overexpression is sufficient to suppress Ras activation in Jurkat T cells and show that this activity is independent of Csk binding. Thus, in addition to negatively regulating Src family kinases by recruiting Csk, PAG also negatively regulates Ras by recruiting RasGAP to the membrane. Finally, by knocking down PAG, we demonstrate both enhanced Src kinase activity and Ras activation, thereby establishing PAG as an important negative regulator of T-cell activation.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Membrane Proteins/immunology , Phosphoproteins/antagonists & inhibitors , T-Lymphocytes/immunology , ras Proteins/metabolism , Cell Division , Clonal Anergy , Cyclic AMP/metabolism , Enzyme Activation , Homeostasis/immunology , Humans , Kinetics , Models, Immunological , Phosphorylation , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Tetradecanoylphorbol Acetate/pharmacology , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...