Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(7)2020 Apr 05.
Article in English | MEDLINE | ID: mdl-32260486

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) regulates target gene expression upon ligand binding. Apart from its effects on metabolism, PPARγ activity can inhibit the production of pro-inflammatory cytokines by several immune cells, including dendritic cells and macrophages. In chronic inflammatory disease models, PPARγ activation delays the onset and ameliorates disease severity. Here, we investigated the effect of PPARγ activation by the agonist Pioglitazone on the function of hepatic immune cells and its effect in a murine model of immune-mediated hepatitis. Cytokine production by both liver sinusoidal endothelial cells (IL-6) and in T cells ex vivo (IFNγ) was decreased in cells from Pioglitazone-treated mice. However, PPARγ activation did not decrease pro-inflammatory tumor necrosis factor alpha TNFα production by Kupffer cells after Toll-like receptor (TLR) stimulation ex vivo. Most interestingly, although PPARγ activation was shown to ameliorate chronic inflammatory diseases, it did not improve hepatic injury in a model of immune-mediated hepatitis. In contrast, Pioglitazone-induced PPARγ activation exacerbated D-galactosamine (GalN)/lipopolysaccharide (LPS) hepatitis associated with an increased production of TNFα by Kupffer cells and increased sensitivity of hepatocytes towards TNFα after in vivo Pioglitazone administration. These results unravel liver-specific effects of Pioglitazone that fail to attenuate liver inflammation but rather exacerbate liver injury in an experimental hepatitis model.


Subject(s)
Hepatitis, Autoimmune/immunology , PPAR gamma/agonists , Pioglitazone/pharmacology , Animals , Cells, Cultured , Interferon-gamma/metabolism , Kupffer Cells/drug effects , Kupffer Cells/immunology , Lymphocyte Activation , Macrophage Activation , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
J Hepatol ; 62(5): 1085-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25529619

ABSTRACT

BACKGROUND & AIMS: During pregnancy, acetaminophen is one of the very few medications recommended by physicians to treat fever or pain. Recent insights from epidemiological studies suggest an association between prenatal acetaminophen medication and an increased risk for development of asthma in children later in life. The underlying pathogenesis of such association is still unknown. METHODS: We aimed to develop a mouse model to provide insights into the effect of prenatal acetaminophen on maternal, fetal and adult offspring's health. The toxic effect of acetaminophen was studied in mice on 1) maternal liver; mirrored by biomarkers of liver injury, centrilobular necrosis, and infiltration of granulocytes; 2) fetal liver; reflected by the frequency of hematopoietic stem cells, and 3) postnatal health; evaluated by the severity of allergic airway inflammation among offspring. RESULTS: We observed an increased susceptibility towards acetaminophen-induced liver damage in pregnant mice compared to virgins. Moreover, hematopoietic stem cell frequency in fetal liver declined in response to acetaminophen. Furthermore, a greater severity of airway inflammation was observed in offspring of dams upon prenatal acetaminophen treatment, identified lung infiltration by leukocytes and eosinophil infiltration into the airways. CONCLUSION: Our newly developed mouse model on prenatal use of acetaminophen reflects findings from epidemiological studies in humans. The availability of this model will allow improvement in our understanding of how acetaminophen-related hepatotoxicity is operational in pregnant individuals and how an increased risk for allergic diseases in response to prenatal acetaminophen is mediated. Such insights, once available, may change the recommendations for prenatal acetaminophen use.


Subject(s)
Acetaminophen , Asthma , Chemical and Drug Induced Liver Injury , Fetal Stem Cells , Prenatal Exposure Delayed Effects , Acetaminophen/administration & dosage , Acetaminophen/adverse effects , Adult , Adult Children , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/adverse effects , Animals , Asthma/etiology , Asthma/physiopathology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Chemical and Drug Induced Liver Injury/prevention & control , Disease Models, Animal , Female , Fetal Stem Cells/drug effects , Fetal Stem Cells/pathology , Humans , Inflammation/etiology , Inflammation/physiopathology , Liver/drug effects , Liver/pathology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/prevention & control , Severity of Illness Index
3.
J Leukoc Biol ; 96(4): 571-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24970859

ABSTRACT

The liver plays a pivotal role in maintaining immunological tolerance, although the exact molecular mechanism is still largely unknown. The induction of systemic tolerance by liver resident APCs has been attributed to peripheral deletion and to the induction of Tregs. HCs, the parenchymal cells in the liver, could function as nonprofessional APCs and interact and establish cell-cell contact with T lymphocytes. We hypothesized that HCs from healthy or regenerated livers may contribute to induction of functional Tregs. Here, we show that murine HCs induced Foxp3(+) Tregs within CD4(+) T cells in vitro, which increased in the presence of TGF-ß. Interestingly, a further Foxp3(+) Treg expansion was observed if HCs were isolated from regenerated livers. Additionally, the induction of Foxp3(+) Tregs was associated with the Notch signaling pathway, as the ability of HCs to enhance Foxp3 was abolished by γ-secretase inhibition. Furthermore, HC-iTregs showed ability to suppress the proliferative response of CD4(+) T cells to anti-CD3 stimulation in vitro. Thus, HCs may play a pivotal role in the induction of tolerance via Notch-mediated conversion of CD4(+) T cells into Foxp3(+) Tregs upon TCR stimulation.


Subject(s)
Hepatocytes/metabolism , Receptors, Notch/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Animals , Calcium-Binding Proteins/metabolism , Cytokines/biosynthesis , Forkhead Transcription Factors/metabolism , Hepatocytes/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Membrane Proteins/metabolism , Mice , Models, Biological , Serrate-Jagged Proteins , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/pharmacology
4.
J Immunol ; 191(11): 5574-82, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24140644

ABSTRACT

The "liver tolerance effect" has been attributed to a unique potential of liver-resident nonprofessional APCs including hepatocytes (HCs) to suppress T cell responses. The exact molecular mechanism of T cell suppression by liver APCs is still largely unknown. In mice, IL-10-dependent T cell suppression is observed after Th1-mediated hepatitis induced by Con A. In this study, we show that HCs, particularly those from regenerating livers of Con A-pretreated mice, induced a regulatory phenotype in naive CD4(+) T cells in vitro. Using reporter mice, we observed that these T regulatory cells released substantial amounts of IL-10, produced IFN-γ, failed to express Foxp3, but suppressed proliferation of responder T cells upon restimulation with anti-CD3 mAb. Hence, these regulatory cells feature a similar phenotype as the recently described IL-10-producing Th1 cells, which are generated upon activation of Notch signaling. Indeed, inhibition of γ-secretase and a disintegrin and metalloproteinase 17 but not a disintegrin and metalloproteinase 10, respectively, which blocked Notch activation, prevented IL-10 secretion. HCs from Con A-pretreated mice showed enhanced expression of the Notch ligand Jagged1 and significantly increased receptor density of Notch1 on CD4(+) T cells. However, HCs from Con A-pretreated IFN regulatory factor 1(-/-) mice, which cannot respond to IFN-γ, as well as those from IFN-γ(-/-) mice failed to augment IL-10 production by CD4(+) T cells. In conclusion, it seems that HCs fine-tune liver inflammation by upregulation of Jagged1 and activation of Notch signaling in Th1 cells. This mechanism might be of particular importance in the regenerating liver subsequent to Th1-mediated hepatitis.


Subject(s)
Hepatitis/immunology , Hepatocytes/immunology , Receptors, Notch/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disintegrins/pharmacology , Gene Expression Regulation/immunology , Hepatocytes/pathology , Immune Tolerance , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interferon Regulatory Factor-1/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/metabolism , Jagged-1 Protein , Liver/pathology , Lymphocyte Activation/drug effects , Male , Matrix Metalloproteinase 17/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Serrate-Jagged Proteins , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/drug effects
5.
J Am Soc Nephrol ; 24(6): 930-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23641052

ABSTRACT

Regulatory T cells (Tregs) exert their immunosuppressive activity through several immunoregulatory mechanisms, including the production of anti-inflammatory cytokines such as IL-10. Although several studies suggest a role for Tregs in modulating crescentic GN, the underlying mechanisms are not well understood. Here, using IL-10 reporter mice, we detected IL-10-producing Foxp3(+) T cells in the kidney, blood, and secondary lymphoid tissue in a mouse model of crescentic GN. Specific inactivation of Il10 in Foxp3(+) Tregs eliminated the ability of these cells to suppress renal and systemic production of IFNγ and IL-17; these IL-10-deficient Tregs lost their capacity to attenuate renal tissue injury. These data highlight the suppressive functions of Tregs in crescentic GN and suggest the importance of Treg-derived IL-10 in ameliorating disease severity and in modulating both the Th1 and most notably Th17 immune response.


Subject(s)
Glomerulonephritis/immunology , Interleukin-10/genetics , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Blood Proteins/immunology , Blood Proteins/toxicity , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Glomerulonephritis/chemically induced , Interleukin-10/metabolism , Kidney/immunology , Kidney/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Severity of Illness Index , Sheep , Spleen/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
6.
J Reprod Immunol ; 97(1): 128-39, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23432879

ABSTRACT

Counter-intuitively, over-the-counter medication is commonly taken by pregnant women. In this context, acetaminophen (APAP, e.g. Paracetamol, Tylenol) is generally recommended by physicians to treat fever and pain during pregnancy. Thus, APAP ranks at the top of the list of medications taken prenatally. Insights on an increased risk for pregnancy complications such as miscarriage, stillbirth, preterm birth or fetal malformations upon APAP exposure are rather ambiguous. However, emerging evidence arising from human trials clearly reveals a significant correlation between APAP use during pregnancy and an increased risk for the development of asthma in children later in life. Pathways through which APAP increases this risk are still elusive. APAP can be liver toxic and since APAP appears to freely cross the placenta, therapeutic and certainly toxic doses could not only affect maternal, but also fetal hepatocytes. It is noteworthy that during fetal development, the liver transiently functions as the main hematopoietic organ. We here review the effect of APAP on metabolic and immunological parameters in pregnant women and on fetal development and immune ontogeny in order to delineate novel, putative and to date underrated pathways through which APAP use during pregnancy can impair maternal, fetal and long term children's health. We conclude that future studies are urgently needed to reconsider the safety and dosage of APAP during pregnancy and - based on the advances made in the field of reproduction as well as APAP metabolism - we propose pathways, which should be addressed in future research and clinical endeavors.


Subject(s)
Acetaminophen/adverse effects , Antipyretics/adverse effects , Liver/drug effects , Maternal Exposure/adverse effects , Pregnancy Complications/drug therapy , Prenatal Exposure Delayed Effects/etiology , Acetaminophen/administration & dosage , Antipyretics/administration & dosage , Child , Female , Fetal Development/drug effects , Fetus , Humans , Mothers , Pregnancy , Prenatal Exposure Delayed Effects/prevention & control
7.
PLoS One ; 7(1): e29993, 2012.
Article in English | MEDLINE | ID: mdl-22253853

ABSTRACT

TNFα stimulates both pro- and anti-apoptotic signalling in hepatocytes. Anti-apoptotic signalling depends on a cascade of ubiquitylation steps leading to NFκB activation. Using Sharpin-deficient mice, we show that the ubiquitin binding protein Sharpin interacts with Hoip, an E3 ligase which generates linear ubiquitin chains. Sharpin-deficiency sensitized hepatocytes to induction of apoptosis by TNFα even in the absence of transcriptional inhibition. TNFα induced activation of NFκB was strongly reduced in hepatocytes from Sharpin-deficient mice, due to reduced and delayed phosphorylation and degradation of IκBα. Injection of TNFα-inducing lipopolysaccharides led to strongly exacerbated liver damage and premature death in Sharpin-deficient mice. Our findings point to an essential role of Sharpin in linear ubiquitin chain formation, NFκB activation, and protection of the liver against inflammatory damaging signals.


Subject(s)
Apoptosis/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Gene Expression Profiling , Humans , Lipopolysaccharides/pharmacology , Liver Failure/metabolism , Liver Failure/pathology , Mice , Mice, Inbred C57BL , Organ Specificity/drug effects , Organ Specificity/genetics , Protein Binding/drug effects , Rats , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
Hepatology ; 55(2): 553-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953613

ABSTRACT

UNLABELLED: Induction or overexpression of the heme-degrading enzyme, heme oxygenase 1 (HO-1), has been shown to protect mice from liver damage induced by acute inflammation. We have investigated the effects of HO-1 induction in a mouse model of chronic liver inflammation and fibrogenesis with progression to hepatocellular carcinoma (HCC) (Mdr2ko; FVB.129P2-Abcb4(tm1Bor)). HO-1 was induced in vivo by treatment with cobalt protoporphyrin IX, starting at week 5 or 12 of mice lifespan, and continued for 7 weeks. Our results showed that HO-1 induction reduced liver damage and chronic inflammation by regulating immune cell infiltration or proliferation as well as tumor necrosis factor receptor signaling. Fibrosis progression was significantly reduced by HO-1 induction in mice with mild, as well as established, portal and lobular fibrosis. HO-1 induction significantly suppressed hepatic stellate cell activation. During established fibrosis, HO-1 induction was able to revert portal inflammation and fibrosis below levels observed at the start of treatment. Moreover, hepatocellular proliferation and signs of dysplasia were decreased after HO-1 induction. CONCLUSION: Induction of HO-1 interferes with chronic inflammation and fibrogenesis and, in consequence, might delay progression to HCC.


Subject(s)
Heme Oxygenase-1/metabolism , Liver Cirrhosis/enzymology , Membrane Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Cell Proliferation , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Liver Cirrhosis/immunology , Mice , Mice, Knockout , Phosphorylation , Protoporphyrins , Receptors, Tumor Necrosis Factor/metabolism , ATP-Binding Cassette Sub-Family B Member 4
10.
Arterioscler Thromb Vasc Biol ; 31(10): 2240-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21817100

ABSTRACT

OBJECTIVE: Inhibitor of apoptosis proteins (IAPs), such as X-linked or cellular IAP 1/2 (XIAP, cIAP1/2), are important regulators of apoptosis. IAP antagonists are currently under clinical investigation as anticancer agents. Interestingly, IAPs participate in the inflammation-associated TNF receptor signaling complex and regulate NFκB signaling. This raises the question about the role of IAPs in inflammation. Here, we investigated the anti-inflammatory potential of IAP inhibitors and the role of IAPs in inflammatory processes of endothelial cells. METHODS AND RESULTS: In mice, the small molecule IAP antagonist A-4.10099.1 (ABT) suppressed antigen-induced arthritis, leukocyte infiltration in concanavalin A-evoked liver injury, and leukocyte transmigration in the TNFα-activated cremaster muscle. In vitro, we observed an attenuation of leukocyte-endothelial cell interaction by downregulation of the intercellular adhesion molecule-1. ABT did not impair NFκB signaling but decreased the TNFα-induced activation of the TGF-ß-activated kinase 1, p38, and c-Jun N-terminal kinase. These effects are based on the proteasomal degradation of cIAP1/2 accompanied by an altered ratio of the levels of membrane-localized TNF receptor-associated factors 2 and 5. CONCLUSIONS: Our results reveal IAP antagonism as a profound anti-inflammatory principle in vivo and highlight IAPs as important regulators of inflammatory processes in endothelial cells.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/prevention & control , Chemical and Drug Induced Liver Injury/prevention & control , Endothelial Cells/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , Caspases/metabolism , Cell Adhesion/drug effects , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Concanavalin A , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Enzyme Activation , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Intercellular Adhesion Molecule-1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , RNA Interference , Serum Albumin, Bovine , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 5/metabolism , Time Factors , Transendothelial and Transepithelial Migration/drug effects , Transfection , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases , Ubiquitination , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Kidney Int ; 80(2): 154-64, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525855

ABSTRACT

Crescentic glomerulonephritis is mediated by inappropriate humoral and cellular immune responses toward self-antigens that may result from defects in central and peripheral tolerance. Evidence now suggests that regulatory T cells (Tregs) may be of pathophysiological importance in proliferative and crescentic forms of glomerulonephritis. To analyze the role of endogenous Tregs in a T cell-dependent glomerulonephritis model of nephrotoxic nephritis, we used 'depletion of regulatory T cell' (DEREG) mice that express the diphtheria toxin receptor under control of the FoxP3 (forkhead box P3) gene promoter. Toxin injection into these mice efficiently depleted renal and splenic FoxP3(+) Treg cells as determined by fluorescent-activated cell sorting (FACS) and immunohistochemical analyses. Treg depletion exacerbated systemic and renal interferon-γ (IFNγ) expression and increased recruitment of IFNγ-producing Th1 cells into the kidney without an effect on the Th17 immune response. The enhanced Th1 response, following Treg cell depletion, was associated with an aggravated course of glomerulonephritis as measured by glomerular crescent formation. Thus, our results establish the functional importance of endogenous Tregs in the control of a significantly enhanced systemic and renal Th1 immune response in experimental glomerulonephritis.


Subject(s)
Glomerulonephritis/immunology , Immunity, Cellular/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Chemotaxis , Disease Models, Animal , Interferon-gamma/biosynthesis , Kidney/immunology , Kidney/pathology , Lymphocyte Depletion , Mice
12.
Hepatology ; 53(4): 1259-69, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21480331

ABSTRACT

UNLABELLED: Chronic diseases of the biliary system are common and may cause fibrosis and eventually progression to liver cirrhosis. The aim was to define a new mouse model of a cholangiopathy leading to liver fibrosis in fra-1tg mice. Liver pathology of fra-1tg mice was analyzed in detail by histology and flow cytometry. Transcript levels of fibrosis-related genes and matrix metalloproteinase (MMP) activities were quantified and immunohistochemical analysis additionally applied. The role of the immune system in this model was analyzed by crossing fra-1tg mice with rag2(-/-) mice. Furthermore, expression of Fra-1 in corresponding human liver diseases was investigated on transcription level and histologically. Fra-1tg mice spontaneously develop biliary fibrosis preceded by ductular proliferation and infiltration of inflammatory cells. Fra-1 protein is present in cholangiocytes and inflammatory cells within the liver. These findings were replicated in human biopsies of patients with advanced liver fibrosis. The inflammatory infiltrate showed a strong increase in activated T cells and decreased natural killer (NK), natural killer T cells (NKT), and B cells in fra-1tg mice as compared to wildtype mice. Moreover, fra-1tg mice develop biliary fibrosis with a time-dependent increase in hepatic collagen content and increase in relative messenger RNA (mRNA) expression of profibrotic genes. Attenuation but not complete prevention of collagen accumulation in liver was observed in the fra-1tg × rag2(-/-) mice. However, transplantation of fra-1tg bone marrow cells into wildtype mice could not induce disease. CONCLUSION: Fra-1tg mice spontaneously develop a progressive biliary disease. These mice are an attractive model for the investigation of cholangiopathies and their interaction with the immune system.


Subject(s)
Cholangitis/chemically induced , Liver Cirrhosis/chemically induced , Proto-Oncogene Proteins c-fos/physiology , Animals , Chemokines/biosynthesis , Disease Models, Animal , Fibrosis , Humans , Liver/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Transgenic , Transcription Factor AP-1/physiology
13.
J Immunol ; 186(9): 5284-93, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21441449

ABSTRACT

The chemokine receptor CXCR3 is preferentially expressed by Th1 cells and critically involved in their recruitment to inflamed tissue. In a mouse model of immune-mediated liver injury inducible by Con A, we investigated the role of CXCR3 in acute IFN-γ-mediated hepatitis as well as in tolerance induction, which has been shown to depend on IL-10-producing CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Induction of Con A hepatitis resulted in increased intrahepatic expression of the CXCR3 ligands CXCL9, CXCL10, and CXCL11. CXCR3(-/-) mice developed a more severe liver injury with higher plasma transaminase activities and a more pronounced Th1/Th17 response compared with wild-type (wt) animals upon Con A injection. Moreover, CXCR3(-/-) mice did not establish tolerance upon Con A restimulation, although Tregs from CXCR3(-/-) mice were still suppressive in an in vitro suppression assay. Instead, Tregs failed to accumulate in livers of CXCR3(-/-) mice upon Con A restimulation in contrast to those from wt animals. Con A-tolerant wt mice harbored significantly increased numbers of intrahepatic CXCR3(+)T-bet(+) Tregs that produced IL-10 compared with nontolerant animals. IFN-γ deficiency or anti-IFN-γ Ab treatment demonstrated that conversion to CXCR3(+)T-bet(+) Tregs depended on a Th1 response. Accordingly, in an immunotherapeutic approach, CD4(+)CD25(+)Foxp3(+) Tregs from Con A-pretreated CXCR3-deficient mice failed to protect against Con A-induced hepatitis, whereas Tregs from Con A-tolerant wt mice allowed CXCR3-deficient mice to recover from Con A hepatitis. In summary, CXCR3(+)T-bet(+)IL-10(+) Tregs are generated in the liver in dependence of IFN-γ, then disseminated into the organism and specifically migrate into the liver, where they limit immune-mediated liver damage.


Subject(s)
Hepatitis/immunology , Immune Tolerance/immunology , Receptors, CXCR3/deficiency , T-Lymphocytes, Regulatory/immunology , Animals , Cell Separation , Concanavalin A/toxicity , Flow Cytometry , Interferon-gamma/immunology , Male , Mice , Mice, Inbred C57BL , Mitogens/toxicity , Receptors, CXCR3/immunology , Reverse Transcriptase Polymerase Chain Reaction
14.
Dig Dis ; 28(1): 86-92, 2010.
Article in English | MEDLINE | ID: mdl-20460895

ABSTRACT

BACKGROUND/AIMS: The liver plays an important role in immunological tolerance due to its anatomical location, as it links the gastrointestinal tract and the systemic venous circulation. Therefore, immune reactions against dietary or bacterial antigens from the gut have to be avoided. However, immune responses resulting in elimination of harmful hepatotrophic pathogens have to be induced. We investigated mechanisms of tolerance induction in response to liver inflammation in a murine model of immune-mediated liver injury. METHODS: Liver damage was induced by injection of the plant lectin concanavalin A (ConA). Cytokine levels were measured in plasma and liver tissue. The frequencies of intrahepatic and splenic cell subsets were measured by FACS analyses. RESULTS: ConA hepatitis was mediated by activation of CD4+ T cells, NKT cells and Kupffer cells releasing IFN-gamma and TNF-alpha. Tolerance developed towards ConA rechallenge within 8 days, lasted for several weeks and was characterized by significantly reduced plasma transaminase activities, decreased Th1/Th17 responses and an increased IL-10 release, the latter being produced by CD4+CD25+FoxP3+ regulatory T cells and Kupffer cells. Moreover, regulatory T cells from ConA-tolerant mice displayed a higher immunosuppressive potential in vitro and in vivo compared to those from non-tolerant animals. Interestingly, ConA hepatitis was aggravated in CCR5(-/-) and CXCR3(-/-) mice. CONCLUSION: These results suggest that ConA tolerance is mediated by induced IL10+ regulatory T cells, probably trafficking into the liver depending on the IFN-gamma-inducible chemokine receptors CCR5 and CXCR3.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Disease Models, Animal , Immune Tolerance , Liver/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Chemokines/immunology , Concanavalin A/immunology , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/therapy , Lymphocyte Activation , Mice , Receptors, Chemokine/immunology
15.
J Am Soc Nephrol ; 21(6): 974-85, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20299360

ABSTRACT

T cells recruited to the kidney contribute to tissue damage in crescentic and proliferative glomerulonephritides. Chemokines and their receptors regulate T cell trafficking, but the expression profile and functional importance of chemokine receptors for renal CD4+ T cell subsets are incompletely understood. In this study, we observed that renal FoxP3+CD4+ regulatory T cells (Tregs) and IL-17-producing CD4+ T (Th17) cells express the chemokine receptor CCR6, whereas IFNgamma-producing Th1 cells are CCR6-. Induction of experimental glomerulonephritis (nephrotoxic nephritis) in mice resulted in upregulation of the only CCR6 ligand, CCL20, followed by T cell recruitment, renal tissue injury, albuminuria, and loss of renal function. CCR6 deficiency aggravated renal injury and increased mortality (from uremia) among nephritic mice. Compared with wild-type (WT) mice, CCR6 deficiency reduced infiltration of Tregs and Th17 cells but did not affect recruitment of Th1 cells in the setting of glomerulonephritis. Adoptive transfer of WT but not CCR6-deficient Tregs attenuated morphologic and functional renal injury in nephritic mice. Furthermore, reconstitution with WT Tregs protected CCR6-/- mice from aggravated nephritis. Taken together, these data suggest that CCR6 mediates renal recruitment of both Tregs and Th17 cells and that the reduction of anti-inflammatory Tregs in the presence of a fully functional Th1 response aggravates experimental glomerulonephritis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Glomerulonephritis/pathology , Interleukin-17/metabolism , Kidney/pathology , Receptors, CCR6/metabolism , T-Lymphocytes, Regulatory/pathology , Animals , Chemokine CCL20/metabolism , Disease Models, Animal , Glomerulonephritis/metabolism , Immune System/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, CCR6/genetics , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/metabolism
16.
J Hepatol ; 51(2): 342-53, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19464067

ABSTRACT

BACKGROUND/AIMS: Calcitonin gene-related peptide (CGRP) is a potent vasodilator and supposed to be responsible for neurogenic inflammation involved in migraine. Its role in inflammatory diseases of other organs is controversial and poorly investigated regarding liver inflammation, although the organ is innervated by CGRP containing primary sensory nerve fibers. METHODS: Male Balb/c and IL-10(-/-) mice were pretreated with either alphaCGRP or the CGRP receptor antagonists CGRP(8-37) or BIBN4096BS. Immune-mediated liver injury was induced by administration of lipopolysaccharide (LPS) or tumor necrosis factor-alpha (TNFalpha) to galactosamine (GalN)-sensitized mice and evaluated by serum transaminase activities and cytokine levels. Furthermore, intrahepatic CGRP receptor expression and hepatic CGRP concentrations were examined. RESULTS: CGRP receptor 1 was expressed by immune cells and hepatocytes in human and murine liver. During liver injury CGRP receptor expression was increased whereas hepatic CGRP concentrations concomitantly decreased. While CGRP receptor antagonists failed to affect liver damage, pretreatment with alphaCGRP protected mice from GalN/LPS-induced liver injury by suppression of the pro-inflammatory cytokine response independently from IL-10 but related to the induction of the transcriptional repressor inducible cAMP early repressor (ICER). In contrast, alphaCGRP failed to protect against GalN/TNFalpha-induced liver failure. CONCLUSION: In the liver, CGRP exerts anti-inflammatory properties, which are characterized by a reduced production of pro-inflammatory cytokines.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide/physiology , Hepatitis/prevention & control , Adult , Aged , Animals , Base Sequence , Calcitonin Gene-Related Peptide Receptor Antagonists , Cytokines/biosynthesis , Cytokines/genetics , DNA Primers/genetics , Female , Galactosamine/immunology , Hepatitis/etiology , Hepatitis/immunology , Humans , Immunization , Inflammation Mediators/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/immunology , Liver/injuries , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Tumor Necrosis Factor-alpha/pharmacology
17.
J Biol Chem ; 284(1): 495-504, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-19001362

ABSTRACT

Tumor necrosis factor-alpha (TNFalpha) stimulation of hepatocytes induces either cell survival or apoptosis, which seems to be regulated by the ubiquitin-proteasome system. Here we investigated the role of TNFalpha-induced down-modulation of the de-ubiquitinating enzyme USP2 for hepatocyte survival. Inhibition of hepatocyte apoptosis by pre-treatment with TNFalpha (TNFalpha tolerance) was analyzed in the mouse model of galactosamine/TNFalpha-induced liver injury and in actinomycin D/TNFalpha-treated primary mouse hepatocytes. The role of USP2 for TNFalpha-induced hepatocyte survival was studied using small interference RNA or an expression clone. Injection of mice or preincubation of hepatocytes with TNFalpha caused a rapid down-regulation of hepatic USP2-41kD, the predominant USP2 isoform in the liver. In vitro an artificial knockdown of USP2 inhibited actinomycin D/TNFalpha-induced hepatocyte apoptosis, which was associated with elevated levels of the anti-apoptotic protein c-Flip(L/S) and a concomitant decrease of cellular levels of the ubiquitinligase Itch, a negative regulator of c-Flip. USP2-41kD overexpression abrogated TNFalpha tolerance in vitro, prevented accumulation of c-Flip(L/S) and resulted in elevated levels of Itch. Accordingly, c-Flip(L/S) protein levels were elevated in livers of TNFalpha-tolerant mice, which correlated to a switch from JNK and ERK to p38 signaling after galactosamine/TNF re-challenge. Our results indicate that TNFalpha-induced USP2 down-regulation is an effective cytoprotective mechanism in hepatocytes. Hence, USP2 could be a novel pharmacological target, and specific USP2 inhibitors might be potential candidates for the treatment of inflammation-related apoptotic liver damage.


Subject(s)
Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/enzymology , Endopeptidases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/drug therapy , Dactinomycin/toxicity , Down-Regulation/drug effects , Hepatocytes/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/enzymology , Liver/pathology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Protein Synthesis Inhibitors/toxicity , RNA, Small Interfering , Ubiquitin Thiolesterase , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Hepatology ; 45(2): 475-85, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17256743

ABSTRACT

UNLABELLED: The liver appears to play an important role in immunological tolerance, for example, during allo-transplantation. We investigated tolerance mechanisms in the model of concanavalin A (ConA)-induced immune-mediated liver injury in mice. We found that a single injection of a sublethal ConA dose to C57BL/6 mice induced tolerance toward ConA-induced liver damage within 8 days. This tolerogenic state was characterized by suppression of the typical Th1 response in this model and increased IL-10 production. Tolerance induction was fully reversible in IL-10 -/- mice and after blockade of IL-10 responses by anti-IL10R antibody. Co-cultures of CD4+CD25+ regulatory T cells (T(reg)s) and CD4+CD25- responder cells revealed T(reg) from ConA-tolerant mice being more effective in suppressing polyclonal T cell responses than T(reg) from control mice. Moreover, T(reg) from tolerant but not from control mice were able to augment in vitro IL-10 expression. Depletion by anti-CD25 monoclonal antibody (MAb) indicated a functional role of T(reg)s in ConA tolerance in vivo. Cell depletion studies revealed T(reg)S and Kupffer cells (KC) to be crucial for IL-10 expression in ConA tolerance. Studies with CD1d -/- mice lacking natural killer T (NKT) cells disclosed these cells as irrelevant for the tolerogenic effect. Finally, cellular immune therapy with CD4+CD25+ cells prevented ConA-induced liver injury, with higher protection by Treg from ConA-tolerized mice. CONCLUSION: The immunosuppressive cytokine IL-10 is crucial for tolerance induction in ConA hepatitis and is mainly expressed by CD4+CD25+ T(reg) and KC. Moreover, T(reg)s exhibit therapeutic potential against immune-mediated liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Concanavalin A/adverse effects , Interleukin-10/metabolism , Kupffer Cells/physiology , Mitogens/adverse effects , T-Lymphocytes, Regulatory/physiology , Animals , CD4 Antigens/metabolism , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Drug Tolerance/physiology , Interleukin-10/genetics , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Knockout , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...