57 research outputs found

    Die Rolle von CD152 (CTLA-4) bei der Begrenzung von T-Zellantworten

    Get PDF
    Bei der adaptiven Immunantwort wird ein breites Repertoire an Effektor-T-Zellen gebildet, das sich durch spezifische und vielfältige funktionelle Fähigkeiten auszeichnet. Neben der Aktivierung der Immunantwort werden Mechanismen benötigt, die Immunantworten regulieren und abschalten können, um unerwünschte Immunreaktion zu verhindern. Die Arbeit befasst sich mit der Rolle von CD152 (CTLA-4), einem Homolog zu CD28, bei der Begrenzung von T-Zellantworten. Die Inhibition der Proliferationsbegrenzung der T-Zellen durch CD152 war ursprünglich auf eine späte Abschaltung der T-Zellproliferation zurückgeführt worden. Wir konnten zeigen, dass CD152 bereits die T-Zellaktivierung abschalten kann und somit die Aktivierungsschwelle der T-Zelle heraufsetzt. Wir konnten auch zeigen, dass CD152 die frühe T-Zellproliferation auf zwei Ebenen inhibiert: Durch die Transkriptionsinitiierung des autologen, Proliferation-induzierenden Zytokins IL-2 und durch die Expression von G1-Kinasen, die für das voranschreiten des Zellzyklus unerläßlich sind. Bisher war es nicht möglich, individuelle, CD152-oberflächen-exprimierende T-Zellen zu detektieren. Um die Expression von CD152 auf der Oberfläche von T-Zellen zu analysieren, haben wir eine sensitive Färbemethode für Oberflächen-exprimiertes CD152 etabliert. Wir konnten damit zeigen, dass während einer antigen-spezifischen Stimulation Zellmembran-gebundenes CD152 lediglich auf einer Subpopulation von aktivierten Zellen (CD152+ T-Zellen) exprimiert wird. Isolierte, aktivierte CD152+ T-Zellen waren im Gegensatz zu aktivierten CD152- T-Zellen bei Restimulation in ihrer Proliferation inhibiert. Dies zeigt auch, dass CD152 in der Lage ist, bereits aktivierte T-Zellen zu inhibieren. Die heterogene Expression von CD152 auf der Oberfläche lässt vermuten, dass die CD152 exprimierenden Zellen, wenn sie ein CD152-Signal bekommen, eine andere Zelldifferenzierung einschlagen als Zellen ohne CD152. Wiederholte oder chronische Aktivierung von Th-Zellen führt zu einer Form von Apoptose, dem Aktivierungs-induzierter Zelltod, gegen den Th2-Zellen resistent sind. Aktivierte Th2-Zellen exprimieren, im Gegensatz zu Th1-Zellen, häufiger CD152 auf ihrer Oberfläche. Wir konnten zeigen, dass CD152-Kreuzvernetzung von aktivierten T-Zellen direkt Resistenz gegen Apoptose vermittelt. Dies geschieht, indem ein Signaltransduktionsmolekül, die PI3´Kinase, aktiviert wird. Dies führt zur Inaktivierung von Apoptose-unterstützenden Molekülen (Phosphorylierung von FKHRL1 und Herunterregulation von FasL) und zur Induktion des Apoptose-verhindernden Moleküls Bcl-2. Vermeidung von Apoptose ist eine zentrale Voraussetzung zur Induktion von Gedächtniszellen. CD152 exprimierende Zellen wären somit gute Kandidaten, um zu Gedächtniszellen zu differenzieren. Um die Rolle von CD152 bei der späten T-Zelldifferenzierung in vivo untersuchen zu können, wurde das CD152 Gen konditionell in der Maus mutagenisiert.During adaptive immune responses a broad repertoire of effector T-cells is generated, characterized by diverse functional capabilities. Besides activation of the immune response other mechanisms are needed in order to regulate and terminate responses, thus preventing unwanted immune reactions. Here I focus on the role of CD152 (CTLA-4), a homologue of CD28, in the limitation of T-cell responses. Inhibition of T-cell-proliferation by CD152 was originally attributed to a late regulation of the T-cell proliferation. We now show that CD152 is already able to prevent the activation of T-cells and to set the threshold for their activation. We also show that CD152 inhibits T-cell activation in two ways: It inhibits the induction of the growth-factor IL-2 and it inhibits the expression of G1-kinases mandatory for the progression of the cell cycle. Until now, it has not been possible to detect individual T-cells expressing CD152 at their surface. To analyze the expression of CD152 at the surface of individual cells, we developed a sensitive staining method. Using this technique we could show that antigen-specific stimulation of T-cells leads to the expression of surface-bound CD152 only on a fraction of the activated T-cells. Isolated, activated CD152+ T-cells were inhibited in their proliferation whereas CD152- T cells were not. This also shows that CD152 is indeed able to inhibit already activated T-cells. The heterogenous expression of CD152 at the cell surface of already activated T-cells also suggests that CD152+ T-cells will differentiate differently compared to CD152-T-cells. Repeated or chronic activation of Th-cells leads to one form of apoptosis, activation-induced cell death (AICD), against which Th2-cells are resistant. Activated Th2-cells express surface CD152 at higher frequencies than Th1-cells. We show here, that CD152-crosslinking of activated T-cells directly induces resistance against AICD by a mechanism requiring PI3´kinase. This leads to the inactivation of pro-apoptotic molecules (phosphorylation of FKHRL1 and downregulation of FasL). It also leads to the induction of the survival molecule Bcl-2. Prevention of apoptosis is a central prerequisite for the generation of memory cells. Therefore, surface CD152+ T-cells might be good candidates to differentiate into memory cells. To investigate the role of CD152 during the differentiation of T-cells in vivo, the CD152 gene was conditional mutagenese of the CD152 gene was generated

    CTLA-4 and PD-1 Control of T-Cell Motility and Migration: Implications for Tumor Immunotherapy

    Get PDF
    CTLA-4 is a co-receptor on T-cells that controls peripheral tolerance and the development of autoimmunity. Immune check-point blockade (ICB) uses monoclonal antibodies (MAbs) to block the binding of inhibitory receptors (IRs) to their natural ligands. A humanized antibody to CTLA-4 was first approved clinically followed by the use of antibody blockade against PD-1 and its ligand PD-L1. Effective anti-tumor immunity requires the activation of tumor-specific effector T-cells, the blockade of regulatory cells and the migration of T-cells into the tumor. Here, we review data implicating CTLA-4 and PD-1 in the motility of T-cells with a specific reference to the potential exploitation of these pathways for more effective tumor infiltration and eradication

    CD152 (CTLA-4) Determines the Unequal Resistance of Th1 and Th2 Cells against Activation-induced Cell Death by a Mechanism Requiring PI3 Kinase Function

    Get PDF
    Survival of antigen-experienced T cells is essential for the generation of adaptive immune responses. Here, we show that the genetic and antibody-mediated inactivation of CD152 (cytotoxic T lymphocyte antigen 4) in T helper (Th) effector cells reduced the frequency of nonapoptotic cells in a completely Fas/Fas ligand (FasL)–dependent manner. CD152 cross-linking together with stimulation of CD3 and CD28 on activated Th2 cells prevented activation-induced cell death (AICD) as a result of reduced Fas and FasL expression. Apoptosis protection conferred by CD152 correlated with the up-regulation of Bcl-2 and was mediated by phosphatidylinositol 3 kinase, which prevented FasL expression through the inhibitory phosphorylation of Forkhead transcription factor FKHRL1. We show that signals induced by CD152 act directly on activated T lymphocytes and, due to its differential surface expression on activated Th1 and Th2 cells, induce resistance to AICD mainly in Th2 cells

    Differential trafficking of ligands trogocytosed via CD28 versus CTLA4 promotes collective cellular control of co-stimulation

    Get PDF
    Intercellular communication is crucial for collective regulation of cellular behaviors. While clustering T cells have been shown to mutually control the production of key communication signals, it is unclear whether they also jointly regulate their availability and degradation. Here we use newly developed reporter systems, bioinformatic analyses, protein structure modeling and genetic perturbations to assess this. We find that T cells utilize trogocytosis by competing antagonistic receptors to differentially control the abundance of immunoregulatory ligands. Specifically, ligands trogocytosed via CD28 are shuttled to the T cell surface, enabling them to co-stimulate neighboring T cells. In contrast, CTLA4-mediated trogocytosis targets ligands for degradation. Mechanistically, this fate separation is controlled by different acid-sensitivities of receptor-ligand interactions and by the receptor intracellular domains. The ability of CD28 and CTLA4 to confer different fates to trogocytosed ligands reveals an additional layer of collective regulation of cellular behaviors and promotes the robustness of population dynamics.Fil: Zenke, Simon. Albert Ludwigs University of Freiburg; AlemaniaFil: Sica, Mauricio Pablo. Consejo Nacional de Investigaciones Científicas y Técnicas; ArgentinaFil: Steinberg, Florian. Albert Ludwigs University of Freiburg; AlemaniaFil: Braun, Julia. Albert Ludwigs University of Freiburg; AlemaniaFil: Zink, Alicia. Albert Ludwigs University of Freiburg; AlemaniaFil: Gavrilov, Alina. Max Planck Institute of Immunobiology and Epigenetics; AlemaniaFil: Hilger, Alexander. Albert Ludwigs University of Freiburg; AlemaniaFil: Arra, Aditya. Otto-von-Guericke-Universität Magdeburg; AlemaniaFil: Brunner Weinzierl, Monika. Otto-von-Guericke-Universität Magdeburg; AlemaniaFil: Elling, Roland. Albert Ludwigs University of Freiburg; AlemaniaFil: Beyersdorf, Niklas. Universität Würzburg; AlemaniaFil: Lämmermann, Tim. Albert Ludwigs University of Freiburg; AlemaniaFil: Smulski, Cristian Roberto. Consejo Nacional de Investigaciones Científicas y Técnicas; ArgentinaFil: Rohr, Jan C.. Albert Ludwigs University of Freiburg; Alemani

    CD152 (CTLA-4) Determines CD4 T Cell Migration In Vitro and In Vivo

    Get PDF
    BACKGROUND:Migration of antigen-experienced T cells to secondary lymphoid organs and the site of antigenic-challenge is a mandatory prerequisite for the precise functioning of adaptive immune responses. The surface molecule CD152 (CTLA-4) is mostly considered as a negative regulator of T cell activation during immune responses. It is currently unknown whether CD152 can also influence chemokine-driven T cell migration. METHODOLOGY/PRINCIPAL FINDINGS:We analyzed the consequences of CD152 signaling on Th cell migration using chemotaxis assays in vitro and radioactive cell tracking in vivo. We show here that the genetic and serological inactivation of CD152 in Th1 cells reduced migration towards CCL4, CXCL12 and CCL19, but not CXCL9, in a G-protein dependent manner. In addition, retroviral transduction of CD152 cDNA into CD152 negative cells restored Th1 cell migration. Crosslinking of CD152 together with CD3 and CD28 stimulation on activated Th1 cells increased expression of the chemokine receptors CCR5 and CCR7, which in turn enhanced cell migration. Using sensitive liposome technology, we show that mature dendritic cells but not activated B cells were potent at inducing surface CD152 expression and the CD152-mediated migration-enhancing signals. Importantly, migration of CD152 positive Th1 lymphocytes in in vivo experiments increased more than 200% as compared to CD152 negative counterparts showing that indeed CD152 orchestrates specific migration of selected Th1 cells to sites of inflammation and antigenic challenge in vivo. CONCLUSIONS/SIGNIFICANCE:We show here, that CD152 signaling does not just silence cells, but selects individual ones for migration. This novel activity of CD152 adds to the already significant role of CD152 in controlling peripheral immune responses by allowing T cells to localize correctly during infection. It also suggests that interference with CD152 signaling provides a tool for altering the cellular composition at sites of inflammation and antigenic challenge

    Migration of Th1 Lymphocytes Is Regulated by CD152 (CTLA-4)-Mediated Signaling via PI3 Kinase-Dependent Akt Activation

    Get PDF
    Efficient adaptive immune responses require the localization of T lymphocytes in secondary lymphoid organs and inflamed tissues. To achieve correct localization of T lymphocytes, the migration of these cells is initiated and directed by adhesion molecules and chemokines. It has recently been shown that the inhibitory surface molecule CD152 (CTLA-4) initiates Th cell migration, but the molecular mechanism underlying this effect remains to be elucidated. Using CD4 T lymphocytes derived from OVA-specific TCR transgenic CD152-deficient and CD152-competent mice, we demonstrate that chemokine-triggered signal transduction is differentially regulated by CD152 via phosphoinositide 3-kinase (PI3K)-dependent activation of protein kinase B (PKB/Akt). In the presence of CD152 signaling, the chemoattractant CCL4 selectively induces the full activation of Akt via phosphorylation at threonine 308 and serine 473 in pro-inflammatory Th lymphocytes expressing the cognate chemokine receptor CCR5. Akt signals lead to cytoskeleton rearrangements, which are indispensable for migration. Therefore, this novel Akt-modulating function of CD152 signals affecting T cell migration demonstrates that boosting CD152 or its down-stream signal transduction could aid therapies aimed at sensitizing T lymphocytes for optimal migration, thus contributing to a precise and effective immune response

    Immune Privilege as an Intrinsic CNS Property: Astrocytes Protect the CNS against T-Cell-Mediated Neuroinflammation

    Get PDF
    Astrocytes have many functions in the central nervous system (CNS). They support differentiation and homeostasis of neurons and influence synaptic activity. They are responsible for formation of the blood-brain barrier (BBB) and make up the glia limitans. Here, we review their contribution to neuroimmune interactions and in particular to those induced by the invasion of activated T cells. We discuss the mechanisms by which astrocytes regulate pro- and anti-inflammatory aspects of T-cell responses within the CNS. Depending on the microenvironment, they may become potent antigen-presenting cells for T cells and they may contribute to inflammatory processes. They are also able to abrogate or reprogram T-cell responses by inducing apoptosis or secreting inhibitory mediators. We consider apparently contradictory functions of astrocytes in health and disease, particularly in their interaction with lymphocytes, which may either aggravate or suppress neuroinflammation
    • …
    corecore