Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Innate Immun ; 18(5): 745-52, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22436844

ABSTRACT

Innate immunity is crucial for an effective host defense against pathogenic microorganisms in periodontal tissues. As periodontal ligament (PDL) cells synthesize immunomodulatory cytokines, the aim of this in vitro study was to investigate whether these cells can interact with innate immune cells. Resting and inflammatory primed (IL-1ß, TNF-α, HMGB1) human PDL cells were co-cultured with human monocyte-derived dendritic cells or macrophages. Migration, phenotypic maturation and modulation of phagocytosis of Porphyromonas gingivalis by immune cells were investigated upon co-culture with PDL cells and/or their released soluble factors. PDL cells interacted with immune cells under both non-inflammatory and inflammatory conditions. Immune cell migration was significantly enhanced by co-culture with PDL cells, which also affected their phenotypic maturation both through cell-cell contact and through released soluble mediators. The dendritic cell maturation markers CD83 and CD86 were upregulated as much as both 'alternatively activated' M2 macrophage maturation markers CD23 and CD163. In contrast, the 'classically activated' M1 macrophage maturation marker CD64 was downregulated. Finally, PDL cells significantly enhanced the phagocytosis of Porphyromonas gingivalis by immune cells. Our experiments revealed that PDL cells are not only structural elements of the periodontium, but actively influence immune responses by interaction with innate immune cells.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/immunology , Macrophages/immunology , Periodontal Diseases/immunology , Periodontal Ligament/immunology , Porphyromonas gingivalis/immunology , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Coculture Techniques , HMGB1 Protein/immunology , Humans , Immunity, Innate , Immunomodulation , Interleukin-1beta , Phagocytosis , Tumor Necrosis Factor-alpha/immunology
2.
Hepatology ; 55(1): 287-97, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21898480

ABSTRACT

UNLABELLED: Adeno-associated viral vectors (rAAV) are frequently used in gene therapy trials. Although rAAV vectors are of low immunogenicity, humoral as well as T cell responses may be induced. While the former limits vector reapplication, the expansion of cytotoxic T cells correlates with liver inflammation and loss of transduced hepatocytes. Because adaptive immune responses are a consequence of recognition by the innate immune system, we aimed to characterize cell autonomous immune responses elicited by rAAV in primary human hepatocytes and nonparenchymal liver cells. Surprisingly, Kupffer cells, but also liver sinusoidal endothelial cells, mounted responses to rAAV, whereas neither rAAV2 nor rAAV8 were recognized by hepatocytes. Viral capsids were sensed at the cell surface as pathogen-associated molecular patterns by Toll-like receptor 2. In contrast to the Toll-like receptor 9-mediated recognition observed in plasmacytoid dendritic cells, immune recognition of rAAV in primary human liver cells did not induce a type I interferon response, but up-regulated inflammatory cytokines through activation of nuclear factor κB. CONCLUSION: Using primary human liver cells, we identified a novel mechanism of rAAV recognition in the liver, demonstrating that alternative means of sensing rAAV particles have evolved. Minimizing this recognition will be key to improving rAAV-mediated gene transfer and reducing side effects in clinical trials due to immune responses against rAAV.


Subject(s)
Dependovirus/immunology , Genetic Therapy/methods , Genetic Vectors/immunology , Hepatocytes/immunology , Immunity, Innate/immunology , Toll-Like Receptor 2/immunology , Biopsy , Capsid/immunology , Cytokines/immunology , Dependovirus/genetics , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/virology , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/virology , Humans , Kupffer Cells/cytology , Kupffer Cells/immunology , Kupffer Cells/virology , NF-kappa B/immunology , NF-kappa B/metabolism , Primary Cell Culture , Signal Transduction/immunology , Toll-Like Receptor 2/metabolism , Up-Regulation/immunology
3.
Dig Dis ; 29(4): 384-90, 2011.
Article in English | MEDLINE | ID: mdl-21894009

ABSTRACT

The liver has a pivotal role in glucose, lipid and protein metabolism as well as in removal of toxins and waste products. A unique microanatomy and a network of resident scavenger cell populations specialized in endocytic uptake of antigens and macromolecules cooperatively mediate these salient hepatic functions together with parenchymal hepatocytes. Antigens taken up by hepatic scavenger cell populations, such as Kupffer cells, hepatic dendritic cells, stellate cells and liver sinusoidal endothelial cells (LSECs), can be (cross-)presented on MHC class I and II molecules, which leads to modulation of T cell immune functions. Among these cell populations, LSECs are endowed with the highest scavenger activity and are the most efficient cell population in cross-presenting soluble exogenous antigens to CD8 T cells. Together with their large number and the high cumulative surface area, LSECs represent the hepatic cell population that is best situated to interact with circulating T cells. Under physiological conditions, antigen-specific interaction of LSECs with CD8 T cells induces tolerance that is characterized by nonresponsiveness towards T cell receptor-mediated stimulation. In contrast to functional maturation of dendritic cells by activation through pattern recognition receptors, there is no such maturation in antigen-presenting LSECs, demonstrating that even under inflammatory conditions induction of CD8 T cell tolerance is preserved. However, upon viral infection of LSECs, a unique program of T cell differentiation into effector cytotoxic T cells is initiated that is independent of currently known costimulatory signals. These results highlight specific mechanisms operative in liver-resident antigen-presenting cells governing the local balance between tolerance and immunity.


Subject(s)
Immune Tolerance/immunology , Immunity/immunology , Liver/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Humans , Liver/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
4.
PLoS One ; 6(7): e22034, 2011.
Article in English | MEDLINE | ID: mdl-21779369

ABSTRACT

BACKGROUND: The transcription factor Tcfap2c has been demonstrated to be essential for various processes during mammalian development. It has been found to be upregulated in various undifferentiated tumors and is implicated with poor prognosis. Tcfap2c is reported to impinge on cellular proliferation, differentiation and apoptosis. However, the physiological consequences of Tcfap2c-expression remain largely unknown. METHODOLOGY/PRINCIPAL FINDINGS: Therefore we established a gain of function model to analyze the role of Tcfap2c in development and disease. Induction of the transgene led to robust expression in all tissues (except brain and testis) and lead to rapid mortality within 3-7 days. In the liver cellular proliferation and apoptosis was detected. Accumulation of microvesicular lipid droplets and breakdown of major hepatic metabolism pathways resulted in steatosis. Serum analysis showed a dramatic increase of enzymes indicative for hepatic failure. After induction of Tcfap2c we identified a set of 447 common genes, which are deregulated in both liver and primary hepatocyte culture. Further analysis showed a prominent repression of the cytochrome p450 system, PPARA, Lipin1 and Lipin2. These data indicate that in the liver Tcfap2c represses pathways, which are responsible for fatty acid metabolism. In the intestine, Tcfap2c expression resulted in expansion of Sox9 positive and proliferative active epithelial progenitor cells resulting in dysplastic growth of mucosal crypt cells and loss of differentiated mucosa. CONCLUSIONS: The transgenic mice show that ectopic expression of Tcfap2c is not tolerated. Due to the phenotype observed, iTcfap2c-mice represent a model system to study liver failure. In intestine, Tcfap2c induced cellular hyperplasia and suppressed terminal differentiation indicating that Tcfap2c serves as a repressor of differentiation and inducer of proliferation. This might be achieved by the Tcfap2c mediated activation of Sox9 known to be expressed in intestinal and hepatic stem/progenitor cell populations.


Subject(s)
Intestinal Diseases/metabolism , Liver Failure/metabolism , Transcription Factor AP-2/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Intestinal Diseases/etiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Liver Failure/etiology , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , PPAR alpha/metabolism , Phosphatidate Phosphatase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-2/genetics
5.
Hepatology ; 51(4): 1430-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20373369

ABSTRACT

UNLABELLED: In vivo evaluation of CD8 T cell effector (cytotoxic T lymphocyte [CTL]) function in peripheral organs such as the liver is currently not possible but would greatly improve our understanding of local immune regulation, because simple determination of antigen-specific CTL numbers does not predict the outcome of immune responses. In particular, measurement of alanine aminotransferase serum levels is not sensitive enough to detect T cell immunity against low numbers of target hepatocytes. We developed a procedure that detects virus-specific effector function of CTLs in the liver after simultaneous adenoviral transfer of reporter and immune target genes into hepatocytes, followed by bioluminescence imaging of reporter genes. Bioluminescence imaging enabled detection of as few as 10,000 infected hepatocytes in vivo, and even more importantly, quantification of antiviral effector function of as few as 50,000 CTLs. CONCLUSION: Our results provide evidence that low numbers of antigen-specific CTLs are sufficient to control viral gene expression and eliminate viral infection from hepatocytes. The experimental system established here is a highly sensitive method to simultaneously detect viral infection of hepatocytes and to quantify antiviral CTL function in the liver in vivo and will help in characterizing principles of hepatic immune regulation.


Subject(s)
Liver/immunology , T-Lymphocytes, Cytotoxic/immunology , Alanine Transaminase/blood , Animals , Hepatocytes/physiology , Immunization , Luminescent Measurements , Mice , Mice, Inbred C57BL
6.
J Immunol ; 184(8): 4107-14, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20212092

ABSTRACT

Cross-presentation of soluble Ag on MHC class I molecules to naive CD8 T cells by liver sinusoidal endothelial cells (LSECs) leads to induction of T cell tolerance that requires interaction between coinhibitory B7-H1 on LSECs and programmed cell death-1 on CD8 T cells. In this study, we investigate whether cross-presentation of high as well as low Ag concentrations allowed for LSEC-induced tolerance. Ag concentration directly correlated with the cross-presentation capacity of murine LSECs and thus strength of TCR stimulation. Although LSEC cross-presentation at low-Ag concentrations resulted in tolerance, they induced differentiation into effector T cells (CTL) at high-Ag concentrations. CTL differentiation under these conditions was not caused by increased expression of costimulatory CD80/86 on cross-presenting LSECs but was determined by early IL-2 release from naive CD8 T cells. B7-H1 signals from LSECs and TCR avidity reciprocally controlled early T cell release of IL-2 and CTL differentiation. B7-H1 expression directly correlated with cross-presentation at low- but not high-Ag concentrations, indicating an imbalance between TCR and coinhibitory signals regulating T cell release of IL-2. Exogenous IL-2 overrode coinhibitory B7-H1-mediated signals by LSECs and induced full CTL differentiation. Our results imply that LSEC-mediated T cell tolerance can be broken in situations where T cells bearing high-avidity TCR encounter LSECs cross-presenting high numbers of cognate MHC class I peptide molecules, such as during viral infection of the liver. Furthermore, we attribute a novel costimulatory function to IL-2 acting in a T cell autonomous fashion to promote local induction of immunity in the liver even in the absence of CD80/86 costimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Immune Tolerance/immunology , Liver/cytology , Liver/immunology , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Cross-Priming/immunology , Cytotoxicity Tests, Immunologic , Endothelial Cells/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Ovalbumin/metabolism , Resting Phase, Cell Cycle/immunology
7.
Immunology ; 128(1 Suppl): e728-37, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740334

ABSTRACT

Adenosine is a well-described anti-inflammatory modulator of immune responses within peripheral tissues. Extracellular adenosine accumulates in inflamed and damaged tissues and inhibits the effector functions of various immune cell populations, including CD8 T cells. However, it remains unclear whether extracellular adenosine also regulates the initial activation of naïve CD8 T cells by professional and semi-professional antigen-presenting cells, which determines their differentiation into effector or tolerant CD8 T cells, respectively. We show that adenosine inhibited the initial activation of murine naïve CD8 T cells after alphaCD3/CD28-mediated stimulation. Adenosine caused inhibition of activation, cytokine production, metabolic activity, proliferation and ultimately effector differentiation of naïve CD8 T cells. Remarkably, adenosine interfered efficiently with CD8 T-cell priming by professional antigen-presenting cells (dendritic cells) and semi-professional antigen-presenting cells (liver sinusoidal endothelial cells). Further analysis of the underlying mechanisms demonstrated that adenosine prevented rapid tyrosine phosphorylation of the key kinase ZAP-70 as well as Akt and ERK1/2 in naïve alphaCD3/CD28-stimulated CD8 cells. Consequently, alphaCD3/CD28-induced calcium-influx into CD8 cells was reduced by exposure to adenosine. Our results support the notion that extracellular adenosine controls membrane-proximal T-cell receptor signalling and thereby also differentiation of naïve CD8 T cells. These data raise the possibility that extracellular adenosine has a physiological role in the regulation of CD8 T-cell priming and differentiation in peripheral organs.


Subject(s)
Adenosine/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antigen-Presenting Cells/drug effects , CD8-Positive T-Lymphocytes/drug effects , Immunologic Factors/pharmacology , Lymphocyte Activation , Receptors, Antigen, T-Cell/antagonists & inhibitors , Animals , Antigen-Presenting Cells/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Calcium/metabolism , Cell Differentiation , Mice , Mice, Inbred C57BL , Protein Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
8.
Hepatology ; 50(3): 909-19, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19610048

ABSTRACT

UNLABELLED: Cross-presentation is an important function of immune competent cells, such as dendritic cells (DCs), macrophages, and an organ-resident liver cell population, i.e., liver sinusoidal endothelial cells (LSECs). Here, we characterize in direct comparison to DCs the distinct dynamics and kinetics of cross-presentation employed by LSECs, which promote tolerance induction in CD8 T cells. We found that LSECs were as competent in cross-presenting circulating soluble antigen ex vivo as DCs at a per-cell basis. However, antigen uptake in vivo was 100-fold more pronounced in LSECs, indicating distinct mechanisms of cross-presentation. In contrast to mannose-receptor-mediated antigen uptake and routing into stable endosomes dedicated to cross-presentation in DCs, we observed distinct antigen-uptake and endosomal routing with high antigen turnover in LSECs that resulted in short-lived cross-presentation. Receptor-mediated endocytosis did not always lead to cross-presentation, because immune-complexed antigen taken up by the Fc-receptor was not cross-presented by LSECs, indicating that induction of CD8 T cell tolerance by LSECs is impaired in the presence of preexisting immunity. CONCLUSION: These results provide a mechanistic explanation how organ-resident LSECs accommodate continuous scavenger function with the capacity to cross-present circulating antigens using distinct kinetics and dynamics of antigen-uptake, routing and cross-presentation compared to DCs.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Liver/immunology , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Endocytosis/physiology , Lectins, C-Type/metabolism , Liver/cytology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/metabolism , Receptors, Cell Surface/metabolism
9.
Hepatology ; 49(5): 1664-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19205034

ABSTRACT

UNLABELLED: Peripheral CD8 T-cell tolerance can be generated outside lymphatic tissue in the liver, but the course of events leading to tolerogenic interaction of hepatic cell populations with circulating T-cells remain largely undefined. Here we demonstrate that preferential uptake of systemically circulating antigen by murine liver sinusoidal endothelial cells (LSECs), and not by other antigen-presenting cells in the liver or spleen, leads to cross-presentation on major histocompatibility complex (MHC) I molecules, which causes rapid antigen-specific naïve CD8 T-cell retention in the liver but not in other organs. Using bone-marrow chimeras and a novel transgenic mouse model (Tie2-H-2K(b) mice) with endothelial cell-specific MHC I expression, we provide evidence that cross-presentation by organ-resident and radiation-resistant LSECs in vivo was both essential and sufficient to cause antigen-specific retention of naïve CD8 T-cells under noninflammatory conditions. This was followed by sustained CD8 T-cell proliferation and expansion in vivo, but ultimately led to the development of T-cell tolerance. CONCLUSION: Our results show that cross-presentation of circulating antigens by LSECs caused antigen-specific retention of naïve CD8 T-cells and identify antigen-specific T-cell adhesion as the first step in the induction of T-cell tolerance.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Endothelial Cells/immunology , Liver/immunology , Animals , Antigens/metabolism , Cell Migration Inhibition , Cells, Cultured , Endothelial Cells/metabolism , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology
10.
FEBS J ; 272(21): 5623-34, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16262700

ABSTRACT

We examined whether the fast release of replicon initiation after sudden O2 recovery of hypoxically incubated mammalian cells depends on kinase activity of Cdk2. We used a system based on starved/refed T24 cells elaborated previously for such investigations [van Betteraey-Nikoleit M, Eisele KH, Stabenow D and Probst H (2003) Eur J Biochem270, 3880-3890]. Cells subjected to hypoxia concurrently with refeeding accumulate the G1 DNA content within 5-6 h. In this state they are ready to perform, within 1-2 min after O2 recovery, a burst of replicon initiations that marks the start of a synchronous S-phase. We found that Cdk2 binds to the chromatin fraction within 4-6 h after refeeding with fresh medium, irrespective of whether the cells were incubated normoxically or hypoxically. However, inhibition of Cdk2 by olomoucine, roscovitine or the Cdk2/cyclin inhibitory peptide II had no influence on the synchronous burst of replicon initiations. Cdc6 and pRb, possible targets of Cdk2 phosphorylation, behaved differentially. Inhibition did not affect phosphorylation of Cdc6 after reoxygenation, whilst chromatin bound pRb remained hypophosphorylated beyond the initiation burst. Thus, neither Cdk2 activity, though present at the end of the hypoxic period, nor pRb phosphorylation are necessary for releasing the burst of replicon initiations upon oxygen recovery. Consequentially, Cdk2 dependent phosphorylation(s) cannot be a critical trigger of replicon initiation in response to reoxygenation after several hours of hypoxia, at least in the T24 cells studied.


Subject(s)
Cell Hypoxia/genetics , Cyclin-Dependent Kinase 2/metabolism , DNA Replication/genetics , Replicon/genetics , Cell Cycle Proteins/metabolism , Cell Hypoxia/physiology , Cell Line , Chromatin/drug effects , Chromatin/metabolism , Culture Media/pharmacology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , DNA Replication/drug effects , Humans , Oxygen/metabolism , Oxygen/pharmacology , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Replicon/drug effects , Retinoblastoma Protein/metabolism
11.
Eur J Biochem ; 270(19): 3880-90, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14511370

ABSTRACT

It was shown previously [Riedinger, H. J., van Betteraey-Nikoleit, M & Probst, H. (2002) Eur. J. Biochem.269, 2383-2393] that initiation of in vivo SV40 DNA replication is reversibly suppressed by hypoxia in a state where viral minichromosomes exhibit a nearly complete set of replication proteins. Reoxygenation triggers fast completion and post-translational modifications. Trying to reveal such fast changes of chromatin-bound replication proteins in the much more complex replication of the cellular genome itself, we developed a protocol to extend these studies using the human bladder carcinoma cell line T24, which was presynchronized in G1 by starvation. Concomitantly with stimulation of the cells by medium renewal, hypoxia was established. This treatment induced T24 cells to contain a large amount of replicons arrested in the 'hypoxic preinitiation state', ready to initiate replication as soon as normal pO2 was restored. Replicons in other stages of replicative activity were not detectable. Consequently the arrested replicons were rapidly released into synchronous initiation and succeeding elongation. Extraction of T24 nuclei with a Triton X-100 buffer yielded a fraction containing the cellular chromatin, including DNA-bound replication proteins, while unbound proteins were removed. The usefulness of this protocol was tested by the proliferation marker PCNA. We demonstrate here that this protein switches from the remainder cellular protein pool into the Triton-extracted nuclear fraction upon reoxygenation. Employing this protocol, analyses of chromatin-bound MCM2, MCM3, Cdc6 and cdk2 suggests that the 'classical' prereplication complex is already formed during hypoxia.


Subject(s)
Chromatin/metabolism , DNA Replication/physiology , Nuclear Proteins/metabolism , Replicon , Biochemistry/methods , Blotting, Western , Carcinoma/genetics , Carcinoma/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Hypoxia , Chromatin/chemistry , DNA/analysis , DNA/biosynthesis , DNA-Binding Proteins , Flow Cytometry/methods , Humans , Macromolecular Substances , Minichromosome Maintenance Complex Component 2 , Minichromosome Maintenance Complex Component 3 , Mitotic Index , Nuclear Proteins/genetics , Octoxynol/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...