Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1388962, 2024.
Article in English | MEDLINE | ID: mdl-38720895

ABSTRACT

Introduction: Chronic inflammation is a hallmark of chronic wounds and inflammatory skin diseases. Due to a hyperactive and prolonged inflammation triggered by proinflammatory immune cells, transitioning to the repair and healing phase is halted. T cells may exacerbate the proinflammatory milieu by secreting proinflammatory cytokines. Chamomilla recutita L. (chamomile) has been suggested for use in several inflammatory diseases, implying a capability to modulate T cells. Here, we have characterized and compared the effects of differently prepared chamomile extracts and characteristic pure compounds on the T cell redox milieu as well as on the migration, activation, proliferation, and cytokine production of primary human T cells. Methods: Phytochemical analysis of the extracts was carried out by LC-MS/MS. Primary human T cells from peripheral blood (PBTs) were pretreated with aqueous or hydroethanolic chamomile extracts or pure compounds. Subsequently, the effects on intracellular ROS levels, SDF-1α induced T cell migration, T cell activation, proliferation, and cytokine production after TCR/CD3 and CD28 costimulation were determined. Gene expression profiling was performed using nCounter analysis, followed by ingenuity pathway analysis, and validation at protein levels. Results: The tested chamomile extracts and pure compounds differentially affected intracellular ROS levels, migration, and activation of T cells. Three out of five differently prepared extracts and two out of three pure compounds diminished T cell proliferation. In line with these findings, LC-MS/MS analysis revealed high heterogeneity of phytochemicals among the different extracts. nCounter based gene expression profiling identified several genes related to T cell functions associated with activation and differentiation to be downregulated. Most prominently, apigenin significantly reduced granzyme B induction and cytotoxic T cell activity. Conclusion: Our results demonstrate an anti-inflammatory effect of chamomile- derived products on primary human T cells. These findings provide molecular explanations for the observed anti-inflammatory action of chamomile and imply a broader use of chamomile extracts in T cell driven chronic inflammatory diseases such as chronic wounds and inflammatory skin diseases. Importantly, the mode of extract preparation needs to be considered as the resulting different phytochemicals can result in differential effects on T cells.


Subject(s)
Anti-Inflammatory Agents , Cytokines , Flowers , Lymphocyte Activation , Matricaria , Plant Extracts , T-Lymphocytes , Humans , Plant Extracts/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Matricaria/chemistry , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Flowers/chemistry , Lymphocyte Activation/drug effects , Plant Roots/chemistry , Cells, Cultured , Cell Proliferation/drug effects , Cell Movement/drug effects
2.
Front Immunol ; 13: 1063313, 2022.
Article in English | MEDLINE | ID: mdl-36591284

ABSTRACT

Use of chimeric antigen receptor (CAR) T cells to treat B cell lymphoma and leukemia has been remarkably successful. Unfortunately, the therapeutic efficacy of CAR T cells against solid tumors is very limited, with immunosuppression by the pro-oxidative tumor microenvironment (TME) a major contributing factor. High levels of reactive oxygen species are well-tolerated by tumor cells due to their elevated expression of antioxidant proteins; however, this is not the case for T cells, which consequently become hypo-responsive. The aim of this study was to improve CAR T cell efficacy in solid tumors by empowering the antioxidant capacity of CAR T cells against the pro-oxidative TME. To this end, HER2-specific human CAR T cells stably expressing two antioxidant systems: thioredoxin-1 (TRX1), and glutaredoxin-1 (GRX1) were generated and characterized. Thereafter, antitumor functions of CAR T cells were evaluated under control or pro-oxidative conditions. To provide insights into the role of antioxidant systems, gene expression profiles as well as global protein oxidation were analyzed. Our results highlight that TRX1 is pivotal for T cell redox homeostasis. TRX1 expression allows CAR T cells to retain their cytolytic immune synapse formation, cytokine release, proliferation, and tumor cell-killing properties under pro-oxidative conditions. Evaluation of differentially expressed genes and the first comprehensive redoxosome analysis of T cells by mass spectrometry further clarified the underlying mechanisms. Taken together, enhancement of the key antioxidant TRX1 in human T cells opens possibilities to increase the efficacy of CAR T cell treatment against solid tumors.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Oxidative Stress , T-Lymphocytes , Tumor Microenvironment , Humans , Antioxidants/metabolism , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Oxidation-Reduction , Oxidative Stress/genetics , Oxidative Stress/immunology , T-Lymphocytes/immunology , Thioredoxins/genetics , Thioredoxins/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
3.
Cell Mol Life Sci ; 78(7): 3543-3564, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33449151

ABSTRACT

The integrin LFA-1 is crucial for T-cell/ APC interactions and sensitive recognition of antigens. Precise nanoscale organization and valency regulation of LFA-1 are mandatory for an appropriate function of the immune system. While the inside-out signals regulating the LFA-1 affinity are well described, the molecular mechanisms controlling LFA-1 avidity are still not fully understood. Here, we show that activation of the actin-bundling protein L-plastin (LPL) through phosphorylation at serine-5 enables the formation of clusters containing LFA-1 in high-affinity conformation. Phosphorylation of LPL is induced by an nPKC-MEK-p90RSK pathway and counter-regulated by the serine-threonine phosphatase PP2A. Interestingly, recruitment of LFA-1 into the T-cell/APC contact zone is not affected by LPL phosphorylation. Instead, for this process, activation of the actin-remodeling protein cofilin through dephosphorylation is essential. Together, this study reveals a dichotomic spatial regulation of LFA-1 clustering and microscale movement in T-cells by two different actin-binding proteins, LPL and cofilin.


Subject(s)
Actins/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Microfilament Proteins/metabolism , Protein Phosphatase 2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , T-Lymphocytes/metabolism , Cells, Cultured , Humans , Phosphorylation , T-Lymphocytes/immunology
4.
Nat Commun ; 10(1): 4073, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31501427

ABSTRACT

Several antitumor therapies work by increasing reactive oxygen species (ROS) within the tumor micromilieu. Here, we reveal that L-plastin (LPL), an established tumor marker, is reversibly regulated by ROS-induced thiol oxidation on Cys101, which forms a disulfide bridge with Cys42. LPL reduction is mediated by the Thioredoxin1 (TRX1) system, as shown by TRX1 trapping, TRX1 knockdown and blockade of Thioredoxin1 reductase (TRXR1) with auranofin. LPL oxidation diminishes its actin-bundling capacity. Ratiometric imaging using an LPL-roGFP-Orp1 fusion protein and a dimedone-based proximity ligation assay (PLA) reveal that LPL oxidation occurs primarily in actin-based cellular extrusions and strongly inhibits cell spreading and filopodial extension formation in tumor cells. This effect is accompanied by decreased tumor cell migration, invasion and extracellular matrix (ECM) degradation. Since LPL oxidation occurs following treatment of tumors with auranofin or γ-irradiation, it may be a molecular mechanism contributing to the effectiveness of tumor treatment with redox-altering therapies.


Subject(s)
Actins/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Neoplasms/metabolism , Alkylation , Cell Line, Tumor , Cell Movement/drug effects , Cell Surface Extensions/metabolism , Cysteine/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Hydrogen Peroxide/toxicity , Models, Biological , Mutation/genetics , Oxidation-Reduction , Sulfhydryl Compounds/metabolism , Thioredoxin Reductase 1/metabolism
5.
J Vis Exp ; (143)2019 01 07.
Article in English | MEDLINE | ID: mdl-30663655

ABSTRACT

The immune synapse is the area of communication between T cells and antigen-presenting cells (APCs). T cells polarize surface receptors and proteins towards the immune synapse to assure a stable binding and signal exchange. Classical confocal, TIRF, or super-resolution microscopy have been used to study the immune synapse. Since these methods require manual image acquisition and time-consuming quantification, the imaging of rare events is challenging. Here, we describe a workflow that enables the morphological analysis of tens of thousands of cells. Immune synapses are induced between primary human T cells in pan-leukocyte preparations and Staphylococcus aureus enterotoxin B (SEB)-loaded Raji cells as APCs. Image acquisition is performed with imaging flow cytometry, also called In-Flow microscopy, which combines features of a flow cytometer and a fluorescence microscope. A complete gating strategy for identifying T cell/APC couples and analyzing the immune synapses is provided. As this workflow allows the analysis of immune synapses in unpurified pan-leukocyte preparations and hence requires only a small volume of blood (i.e., 1 mL), it can be applied to samples from patients. Importantly, several samples can be prepared, measured, and analyzed in parallel.


Subject(s)
Flow Cytometry/methods , Immunological Synapses/immunology , Humans
6.
Mol Cell Biol ; 38(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30181394

ABSTRACT

While several protein serine/threonine kinases control cytokine production by T cells, the roles of serine/threonine phosphatases are largely unexplored. Here, we analyzed the involvement of protein phosphatase 1α (PP1α) in cytokine synthesis following costimulation of primary human T cells. Small interfering RNA (siRNA)-mediated knockdown of PP1α (PP1KD) or expression of a dominant negative PP1α (D95N-PP1) drastically diminished interleukin-10 (IL-10) production. Focusing on a key transcriptional activator of human IL-10, we demonstrate that nuclear translocation of NF-κB was significantly inhibited in PP1KD or D95N-PP1 cells. Interestingly, knockdown of cofilin, a known substrate of PP1 containing a nuclear localization signal, also prevented nuclear accumulation of NF-κB. Expression of a constitutively active nonphosphorylatable S3A-cofilin in D95N-PP1 cells restored nuclear translocation of NF-κB and IL-10 expression. Subpopulation analysis revealed that defective nuclear translocation of NF-κB was most prominent in CD4+ CD45RA- CXCR3- T cells that included IL-10-producing TH2 cells. Together these findings reveal novel functions for PP1α and its substrate cofilin in T cells namely the regulation of the nuclear translocation of NF-κB and promotion of IL-10 production. These data suggest that stimulation of PP1α could limit the overwhelming immune responses seen in chronic inflammatory diseases.


Subject(s)
Actin Depolymerizing Factors/metabolism , Anti-Inflammatory Agents/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Interleukin-10/metabolism , NF-kappa B/metabolism , Protein Phosphatase 1/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Humans , Immunity/physiology , Inflammation/metabolism , Protein Transport/physiology , Th2 Cells
7.
J Cell Biochem ; 118(9): 2528-2533, 2017 09.
Article in English | MEDLINE | ID: mdl-28252214

ABSTRACT

The clearance of tumors or virus infected cells is a crucial task of the immune system. Cytotoxic T-cells (CTLs) are able to detect and to kill such altered host cells. Given the recent success of checkpoint inhibitors for tumor therapy, it becomes more and more important to understand the biology of T-cell mediated target cell killing. Tests that allow analyzing the biology of CTLs are either based on flow cytometry or fluorescence microscopy. Thus, they either lack image-based information or have a poor statistical robustness. Therefore, we describe an approach to quantify CTL-mediated cytotoxicity using imaging flow cytometry. Using activated primary human cytotoxic T-cells as CTLs and P815 as target cells, we show that both the evaluation of target cell death and the biology of CTLs can be evaluated in parallel. This enables to gain information about CTL-mediated cytotoxicity in samples from patients important for translational medicine. J. Cell. Biochem. 118: 2528-2533, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Flow Cytometry/methods , Immunity, Cellular , CD8-Positive T-Lymphocytes/cytology , Cell Line , Humans
8.
Methods ; 112: 25-38, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27693880

ABSTRACT

Neutrophils or polymorphonuclear cells (PMN) eliminate bacteria via phagocytosis and/or NETosis. Apart from these conventional roles, PMN also have immune-regulatory functions. They can transdifferentiate and upregulate MHCII as well as ligands for costimulatory receptors which enables them to behave as antigen presenting cells (APC). The initial step for activating T-cells is the formation of an immune synapse between T-cells and antigen-presenting cells. However, the immune synapse that develops at the PMN/T-cell contact zone is as yet hardly investigated due to the non-availability of methods for analysis of large number of PMN interactions. In order to overcome these obstacles, we introduce here a workflow to analyse the immune synapse of primary human PMN and T-cells using multispectral imaging flow cytometry (InFlow microscopy) and super-resolution microscopy. For that purpose, we used CD3 and CD66b as the lineage markers for T-cells and PMN, respectively. Thereafter, we applied and critically discussed various "masks" for identification of T-cell PMN interactions. Using this approach, we found that a small fraction of transdifferentiated PMN (CD66b+CD86high) formed stable PMN/T-cell conjugates. Interestingly, while both CD3 and CD66b accumulation in the immune synapse was dependent on the maturation state of the PMN, only CD3 accumulation was greatly enhanced by the presence of superantigen. The actin cytoskeleton was weakly rearranged at the PMN side on the immune synapse upon contact with a T-cell in the presence of superantigen. A more detailed analysis using super-resolution microscopy (structured-illumination microscopy, SIM) confirmed this finding. Together, we present an InFlow microscopy based approach for the large scale analysis of PMN/T-cell interactions and - combined with SIM - a possibility for an in-depth analysis of protein translocation at the site of interactions.


Subject(s)
Antigen-Presenting Cells/metabolism , Cell Communication/immunology , Flow Cytometry/methods , Image Cytometry/methods , Microscopy/methods , T-Lymphocytes/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/immunology , Actin Cytoskeleton/ultrastructure , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/ultrastructure , Antigens, CD/genetics , Antigens, CD/immunology , Biomarkers/metabolism , CD3 Complex/genetics , CD3 Complex/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Communication/genetics , Cell Transdifferentiation , Coculture Techniques , Flow Cytometry/instrumentation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression , Granulocytes/immunology , Granulocytes/metabolism , Granulocytes/ultrastructure , Humans , Image Cytometry/instrumentation , Immunological Synapses/genetics , Immunological Synapses/ultrastructure , Immunomagnetic Separation/methods , Microscopy/instrumentation , Primary Cell Culture , T-Lymphocytes/immunology , T-Lymphocytes/ultrastructure
9.
J Immunol Methods ; 423: 29-39, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25795421

ABSTRACT

The actin cytoskeleton is a main component to preserve the cell shape. It represents a cellular machinery that enables morphological changes and orchestrates important dynamic cellular functions. Thereby, it supports T-cell migration, immune synapse formation, activation and execution of effector functions. The analysis of actin rearrangements in T-cells is therefore an important field of basic and clinical research. Actin reorganization is traditionally performed using flow cytometry or confocal microscopy. However, while flow cytometry lacks spatial and structural information, confocal microscopy is time consuming and not feasible for the characterization of rare events or of un-purified primary cell populations. Here we describe a methodology to analyze actin rearrangements using InFlow microscopy, which is a hybrid technique consisting of flow cytometric and microscopic features. We show that InFlow microscopy is a valuable tool for quantification of the amount and distribution of F-actin in human T-cells after stimulation with chemokines or antigen-presenting cells.


Subject(s)
Actins/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Synapses/metabolism , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Cell Shape/physiology , Cells, Cultured , Flow Cytometry/methods , Humans , Microscopy, Confocal/methods , T-Lymphocytes/metabolism
10.
Mol Cancer ; 13: 10, 2014 Jan 18.
Article in English | MEDLINE | ID: mdl-24438191

ABSTRACT

BACKGROUND: Tumor cell migration and metastasis require dynamic rearrangements of the actin cytoskeleton. Interestingly, the F-actin cross-linking and stabilizing protein L-plastin, originally described as a leukocyte specific protein, is aberrantly expressed in several non-hematopoietic malignant tumors. Therefore, it has been discussed as a tumor marker. However, systematic in vivo analyses of the functional relevance of L-plastin for tumor cell metastasis were so far lacking. METHODS: We investigated the relevance of L-plastin expression and phosphorylation by ectopical expression of L-plastin in human melanoma cells (MV3) and knock-down of endogenous L-plastin in prostate cancer (PC3M). The growth and metastatic potential of tumor cells expressing no L-plastin, phosphorylatable or non-phosphorylatable L-plastin was analyzed in a preclinical mouse model after subcutaneous and intracardial injection of the tumor cells. RESULTS: Knock-down of endogenous L-plastin in human prostate carcinoma cells led to reduced tumor cell growth and metastasis. Vice versa, and in line with these findings, ectopic expression of L-plastin in L-plastin negative melanoma cells significantly increased the number of metastases. Strikingly, the metastasis promoting effect of L-plastin was not observed if a non-phosphorylatable L-plastin mutant was expressed. CONCLUSIONS: Our data provide the first in vivo evidence that expression of L-plastin promotes tumor metastasis and, importantly, that this effect depends on an additionally required phosphorylation of L-plastin. In conclusion, these findings imply that for determining the importance of tumor-associated proteins like L-plastin a characterization of posttranslational modifications is indispensable.


Subject(s)
Biomarkers, Tumor/metabolism , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Prostatic Neoplasms/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cytoskeleton/metabolism , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Phosphorylation , Prostatic Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 288(41): 29430-9, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24003227

ABSTRACT

Oxidative stress can lead to T cell hyporesponsiveness. A reducing micromilieu (e.g. provided by dendritic cells) can rescue T cells from such oxidant-induced dysfunction. However, the reducing effects on proteins leading to restored T cell activation remained unknown. One key molecule of T cell activation is the actin-remodeling protein cofilin, which is dephosphorylated on serine 3 upon T cell costimulation and has an essential role in formation of mature immune synapses between T cells and antigen-presenting cells. Cofilin is spatiotemporally regulated; at the plasma membrane, it can be inhibited by phosphatidylinositol 4,5-bisphosphate (PIP2). Here, we show by NMR spectroscopy that a reducing milieu led to structural changes in the cofilin molecule predominantly located on the protein surface. They overlapped with the PIP2- but not actin-binding sites. Accordingly, reduction of cofilin had no effect on F-actin binding and depolymerization and did not influence the cofilin phosphorylation state. However, it did prevent inhibition of cofilin activity through PIP2. Therefore, a reducing milieu may generate an additional pool of active cofilin at the plasma membrane. Consistently, in-flow microscopy revealed increased actin dynamics in the immune synapse of untransformed human T cells under reducing conditions. Altogether, we introduce a novel mechanism of redox regulation: reduction of the actin-remodeling protein cofilin renders it insensitive to PIP2 inhibition, resulting in enhanced actin dynamics.


Subject(s)
Actin Depolymerizing Factors/metabolism , Cell Membrane/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , T-Lymphocytes/metabolism , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/genetics , Actins/metabolism , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Dynamics Simulation , Mutation , Oxidation-Reduction , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphorylation , Polymerization , Protein Binding , Protein Conformation , Protein Structure, Tertiary
12.
Eur J Immunol ; 41(11): 3157-69, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21805466

ABSTRACT

Activation of naïve T cells requires costimulation via TCR/CD3 plus accessory receptors, which enables the dynamic rearrangement of the actin cytoskeleton and immune synapse maturation. Signaling events induced following costimulation may thus be valuable targets for therapeutic immunosuppression. Phosphorylation of the actin-bundling protein L-plastin represents such a costimulatory signal in primary human T cells. Phosphorylated L-plastin has a higher affinity toward F-actin. However, the importance of the L-plastin phosphorylation for actin cytoskeleton regulation upon antigen recognition remained unclear. Here, we demonstrate that phosphorylation of L-plastin is important for immune synapse maturation. Thus, expression of nonphosphorylatable L-plastin in untransformed human peripheral blood T cells leads to reduced accumulation of LFA-1 in the immune synapse and to a diminished F-actin increase upon T-cell activation. Interestingly, L-plastin phosphorylation is inhibited by the glucocorticoid dexamethasone. In line with this finding, dexamethasone treatment leads to a reduced F-actin content in stimulated T cells and prevents maturation of the immune synapse. This inhibitory effect of dexamethasone could be reverted by expression of a phospho-mimicking L-plastin mutant. In conclusion, our data introduce costimulation-induced L-plastin phosphorylation as an important event for immune synapse formation and its inhibition by dexamethasone as a novel mode of function of this immunosuppressive glucocorticoid.


Subject(s)
Dexamethasone/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , T-Lymphocytes/metabolism , Blotting, Western , Cell Separation , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Phosphorylation , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Eur J Immunol ; 40(9): 2437-49, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20683899

ABSTRACT

Formation of immune synapses (IS) between T cells and APC requires multiple rearrangements in the actin cytoskeleton and selective receptor accumulation in supramolecular activation clusters (SMAC). The inner cluster (central SMAC) contains the TCR/CD3 complex. The outer cluster (peripheral SMAC) contains the integrin LFA-1 and Talin. Molecular mechanisms selectively stabilizing receptors in the IS remained largely unknown. Here, we demonstrate that sustained LFA-1 clustering in the IS is a consequence of the combined activities of the actin-bundling protein L-plastin (LPL) and calmodulin. Thus, upon antigen-recognition of T cells, LPL accumulated predominantly in the peripheral SMAC. siRNA-mediated knock-down of LPL led to a failure of LFA-1 and Talin redistribution - however, not TCR/CD3 relocalization - into the IS. As a result of this LPL knock-down, the T-cell/APC interface became smaller over time and T-cell proliferation was inhibited. Importantly, binding of calmodulin to LPL was required for the maintenance of LPL in the IS and consequently inhibition of calmodulin also prevented stable accumulation of LFA-1 and Talin, but not CD3, in the IS.


Subject(s)
Actins/metabolism , Calmodulin/metabolism , Immunological Synapses/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , T-Lymphocytes/metabolism , Actins/genetics , Actins/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation , Cloning, Molecular , Enterotoxins/metabolism , Humans , Immunological Synapses/genetics , Immunological Synapses/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Microscopy, Confocal , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Transport/drug effects , Protein Transport/genetics , RNA, Small Interfering/genetics , Sequence Deletion/genetics , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
14.
Basic Res Cardiol ; 104(3): 341-51, 2009 May.
Article in English | MEDLINE | ID: mdl-19139946

ABSTRACT

BACKGROUND: Cardiac troponins provide excellent risk stratification in unstable angina (UA), but no reliable markers are available in troponin-negative patients. Beta2-integrin mediated T cell recruitment plays a pivotal role in coronary atherosclerotic plaque rupture. The present study investigates beta2-integrin activation on T cell subsets as a risk marker in UA. METHODS: Functional activation (affinity/avidity) of beta2-integrins on T cells was measured using a flow cytometry-based whole blood assay in 87 patients with UA. RESULTS: Beta2-integrin activation was significantly higher in patients with severe coronary artery disease (sC) and myocardial infarction (MI) compared to patients with no/minimal coronary atherosclerosis (no/mC), irrespective of troponin status. Adjusted for cardiovascular risk factors, medication, left ventricular function, MI at enrollment and high sensitivity C-reactive protein (hsCRP), beta2-integrin activation was independently associated with incidence of revascularization, hospitalization and all major cardiovascular events during 9 months of follow-up after index investigation. The highest prognostic value of beta2-integrin activation was seen in troponin-and hsCRP-negative patients. CONCLUSION: Quantitative assessment of T cell beta2-integrin activation allows to identify high risk patients with UA and sC without established MI; furthermore, it is associated with incidence of future cardiovascular events independent of conventional risk factors (troponin, hsCRP).


Subject(s)
Angina, Unstable/metabolism , Biomarkers/metabolism , CD18 Antigens/metabolism , T-Lymphocyte Subsets/metabolism , Aged , Angina, Unstable/immunology , Angina, Unstable/mortality , Coronary Artery Disease/complications , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis
15.
Immunity ; 29(3): 404-13, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18771940

ABSTRACT

Oxidative stress leads to impaired T cell activation. A central integrator of T cell activation is the actin-remodelling protein cofilin. Cofilin is activated through dephosphorylation at Ser3. Activated cofilin enables actin dynamics through severing and depolymerization of F-actin. Binding of cofilin to actin is required for formation of the immune synapse and T cell activation. Here, we showed that oxidatively stressed human T cells were impaired in chemotaxis- and costimulation-induced F-actin modulation. Although cofilin was dephosphorylated, steady-state F-actin levels increased under oxidative stress conditions. Mass spectrometry revealed that cofilin itself was a target for oxidation. Cofilin oxidation induced formation of an intramolecular disulfide bridge and loss of its Ser3 phosphorylation. Importantly, dephosphorylated oxidized cofilin, although still able to bind to F-actin, did not mediate F-actin depolymerization. Impairing actin dynamics through oxidation of cofilin provides a molecular explanation for the T cell hyporesponsiveness caused by oxidative stress.


Subject(s)
Actin Depolymerizing Factors/metabolism , Actins/metabolism , Lymphocyte Activation , Neutrophils/immunology , Oxidative Stress , T-Lymphocytes/immunology , Actin Depolymerizing Factors/chemistry , CD28 Antigens/immunology , CD3 Complex/immunology , Chemotaxis, Leukocyte , Humans , Hydrogen Peroxide/metabolism , Lim Kinases/metabolism , Neutrophil Activation , Neutrophils/metabolism , Oxidation-Reduction , Phosphorylation , T-Lymphocytes/metabolism
16.
Eur J Immunol ; 37(10): 2881-91, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17899554

ABSTRACT

T cell activation requires costimulation of TCR/CD3 plus accessory receptors (e.g. CD28). A hallmark of costimulation is the dynamic reorganization of the actin cytoskeleton, important for receptor polarization in the immunological synapse. The classical model of T cell costimulation was challenged by the detection of superagonistic anti-CD28 antibodies. These induce T cell proliferation and--as demonstrated here--production of IFN-gamma, CD25 and CD69 even in the absence of TCR/CD3 coligation. Here, we analyzed whether superagonistic CD28 stimulation induces costimulatory signaling events. Costimulation leads to phosphorylation of the actin-bundling protein L-plastin and dephosphorylation of the actin-reorganizing protein cofilin. Cofilin binds to F-actin only in its dephosphorylated form. Binding of cofilin to F-actin leads to depolymerization or severing of F-actin. The latter ends up in smaller F-actin fragments, which can be elongated at the free barbed ends. This results in enhanced actin polymerization. Dephosphorylation of cofilin requires activation of Ras and PI3Kinase. Interestingly, superagonistic CD28 stimulation activates human peripheral blood T cells independently of Ras and PI3Kinase. Accordingly, it does not lead to cofilin dephosphorylation and receptor polarization. Likewise, L-plastin is not phosphorylated. Thus, superagonistic CD28 stimulation does not mimic costimulation. Instead, it leads to a Ras/PI3Kinase/cofilin-independent state of "unpolarized T cell activation".


Subject(s)
Actin Depolymerizing Factors/physiology , CD28 Antigens/immunology , CD28 Antigens/metabolism , Leukocyte Common Antigens/biosynthesis , Lymphocyte Activation , Phosphatidylinositol 3-Kinases/physiology , T-Lymphocyte Subsets/immunology , ras Proteins/physiology , Animals , Antibodies/pharmacology , Cell Polarity/immunology , Cells, Cultured , Enzyme Activation/immunology , Humans , Lymphocyte Activation/immunology , Mitogens/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rabbits , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/metabolism , ras Proteins/metabolism
17.
J Immunol Methods ; 327(1-2): 30-9, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17719602

ABSTRACT

Adhesion of leukocytes is an early step in the formation of adaptive or innate immunity. In chronic inflammatory pathologies like atherosclerosis, regulation of adhesiveness is pivotal for the accumulation of leukocytes within the vessel wall. Therefore, the quantification of adhesion is crucial for the understanding and monitoring of immune responses in patients. However, so far, functional analysis of leukocyte adhesion has been time consuming and required prior purification of cell populations from peripheral blood. This reduced the number of samples and cell populations that could be analysed from limited patient material. Here, we introduce a novel method involving rapid quantification of integrin-mediated leukocyte adhesion in human whole blood using flow cytometry. The quantification relies on soluble multivalent immunocomplexes and is thus called "ligand-complex-based adhesion assay" (LC-AA). LC-AA evaluates both integrin affinity and avidity in T-cells, NK-cells and monocytes from as little as 20 mul of whole blood. In marked contrast to T-cells and NK-cells, unstimulated monocytes show non-blockable background binding of the complexes. Therefore, for this subset only, the stimulation-induced integrin activation is measurable. With the LC-AA, for the first time, measurement of adhesiveness of extremely rare cell populations like CD34+ peripheral blood stem cells can be assessed in the absence of prior purification steps. Finally, the small blood volumes needed for adhesion analysis with the LC-AA allow the evaluation of multiple cell subpopulations in large sample collectives, e.g. required in clinical studies.


Subject(s)
Cell Adhesion , Flow Cytometry/methods , Leukocytes/cytology , Stem Cells/cytology , Antigens, CD34 , Blood Cells/cytology , Cell Separation/methods , Humans , Integrins , Intercellular Adhesion Molecule-1 , Sensitivity and Specificity , Vascular Cell Adhesion Molecule-1
SELECTION OF CITATIONS
SEARCH DETAIL
...