Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 28(12): 5136-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25122558

ABSTRACT

MIF is an inflammatory cytokine but is hepatoprotective in models of hepatotoxin-induced liver fibrosis. Hepatic fibrosis can also develop from metabolic liver disease, such as nonalcoholic fatty liver disease (NASH). We investigated the role of MIF in high-fat or methionine- and choline-deficient diet mouse models of NASH. Mif(-/-) mice showed elevated liver triglyceride levels (WT, 53±14 mg/g liver; Mif(-/-), 103±7 mg/g liver; P<0.05) and a 2-3-fold increased expression of lipogenic genes. Increased fatty degeneration in the livers of Mif(-/-) mice was associated with increased hepatic inflammatory cells (1.6-fold increase in F4/80(+) macrophages) and proinflammatory cytokines (e.g., 2.3-fold increase in Tnf-α and 2-fold increase in Il-6 expression). However, inflammatory cells and cytokines were decreased by 50-90% in white adipose tissue (WAT) of Mif(-/-) mice. Subset analysis showed that macrophage phenotypes in livers of Mif(-/-) mice were skewed toward M2 (e.g., 1.7-fold and 2.5-fold increase in Arg1 and Il-13, respectively, and 2.5-fold decrease in iNos), whereas macrophages were generally reduced in WAT of these mice (70% reduction in mRNA expression of F4/80(+) macrophages). The protective MIF effect was scrutinized in isolated hepatocytes. MIF reversed inflammation-induced triglyceride accumulation in Hepa1-6 cells and primary hepatocytes and also attenuated oleic acid-elicited triglyceride increase in 3T3-L1 adipocytes. Protection from fatty hepatocyte degeneration was paralleled by a 2- to 3-fold reduction by MIF of hepatocyte proinflammatory cytokine production. Blockade of MIF receptor cluster of differentiation 74 (CD74) but not of CXCR2 or CXCR4 fully reverted the protective effect of MIF, comparable to AMPK inhibition. In summary, we demonstrate that MIF mediates hepatoprotection through the CD74/AMPK pathway in hepatocytes in metabolic models of liver injury.


Subject(s)
Macrophage Migration-Inhibitory Factors/physiology , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Base Sequence , DNA Primers , Disease Models, Animal , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
2.
PLoS One ; 8(6): e66106, 2013.
Article in English | MEDLINE | ID: mdl-23799074

ABSTRACT

Liver fibrosis is associated with infiltrating immune cells and activation of hepatic stellate cells. We here aimed to investigate the effects of the CC chemokine CCL3, also known as macrophage inflammatory protein-1α, in two different fibrosis models. To this end, we treated mice either with carbon tetrachloride or with a methionine- and choline-deficient diet to induce fibrosis in CCL3 deficient and wild-type mice. The results show that the protein expression of CCL3 is increased in wild-type mice after chronic liver injury. Deletion of CCL3 exhibited reduced liver fibrosis compared to their wild-type counterparts. We could validate these results by treating the two mouse groups with either carbon tetrachloride or by feeding a methionine- and choline-deficient diet. In these models, lack of CCL3 is functionally associated with reduced stellate cell activation and liver immune cell infiltration. In vitro, we show that CCL3 leads to increased proliferation and migration of hepatic stellate cells. In conclusion, our results define the chemokine CCL3 as a mediator of experimental liver fibrosis. Thus, therapeutic modulation of CCL3 might be a promising target for chronic liver diseases.


Subject(s)
Chemokine CCL3/physiology , Liver Cirrhosis/physiopathology , Animals , Carbon Tetrachloride/administration & dosage , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Lipogenesis , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes/cytology
3.
Int J Clin Exp Pathol ; 6(4): 678-85, 2013.
Article in English | MEDLINE | ID: mdl-23573314

ABSTRACT

Fibrosis or scarring of the liver parenchyma is a mainstay of chronic liver diseases and is associated with increased morbidity and mortality. Since complete scarring of the liver develops over several decades, therapeutic intervention with the aim of ameliorating fibrosis is of great clinical interest. In a recent study, we could identify the chemokine receptor antagonist Met-CCL5 as a potential compound to inhibit fibrosis progression and accelerate its regression. In the current study we characterized immune changes during fibrosis regression associated with the treatment with the CCL5 (RANTES) chemokine receptor antagonist Met-CCL5 in an established mouse model of chronic liver damage. Met-CCL5 or PBS was given after fibrosis induction (8 weeks of CCl(4)) and mice were sacrificed three and seven days after peak fibrosis. Mouse livers were analyzed for immune cell infiltration and cytokine gene expression. The results show that overall monocyte recruitment was not affected by Met-CCL5, but there was a significant shift to a pro-inflammatory Gr1+ monocyte population in the livers of mice treated with Met-CCL5. These monocytes were mostly iNOS +, a phenomenon which was also evident when analyzing the overall gene expression profiles in the livers. Since a shift in monocyte subpopulations has recently been identified to contribute to fibrosis regression, our results help explaining the efficacy of CCL5 chemokine antagonism as a novel treatment option for fibrotic liver diseases.


Subject(s)
Chemokine CCL5/antagonists & inhibitors , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Monocytes/drug effects , Monocytes/pathology , Receptors, CCR/antagonists & inhibitors , Animals , Carbon Tetrachloride/adverse effects , Cell Count , Chemokine CCL5/drug effects , Cytokines/metabolism , Disease Models, Animal , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Receptors, CCR/drug effects
4.
PLoS One ; 7(5): e36614, 2012.
Article in English | MEDLINE | ID: mdl-22574195

ABSTRACT

BACKGROUND: The chemokine CCL5 is involved in the recruitment of immune cells and a subsequent activation of hepatic stellate cells (HSC) after liver injury. We here investigate whether inhibition of CCL5 oligomerization and glycosaminoglycan binding by a mutated CCL5 protein ((44)AANA(47)-CCL5) has the potential to ameliorate liver cell injury and fibrosis in vivo. METHODOLOGY: Liver injury was induced in C57BL/6 mice by intraperitoneal injection of carbon tetrachloride (CCl(4)) in an acute and a chronic liver injury model. Simultaneously, mice received either (44)AANA(47)-CCL5 or vehicle. Liver cell necrosis and fibrosis was analyzed by histology, and measurement of serum transaminases and hydroxyproline. Intrahepatic mRNA expression of fibrosis and inflammation related genes were determined by quantitative RT-PCR and infiltration of immune cells was assessed by FACS analysis and immunocytochemistry. In vitro, HSC were stimulated with conditioned media of T-cell enriched splenocytes. PRINCIPAL FINDINGS: (44)AANA(47)-CCL5 treated mice displayed a significantly reduced degree of acute liver injury (liver cell necrosis, transaminases) and fibrosis (Sirus red positive area and hydroxyproline content) compared to vehicle treated mice. Ameliorated fibrosis by (44)AANA(47)-CCL5 was associated with a decreased expression of fibrosis related genes, decreased α-smoth muscle antigen (αSMA) and a reduction of infiltrating immune cells. In the acute model, (44)AANA(47)-CCL5 treated mice displayed a reduced immune cell infiltration and mRNA levels of TNF, IL-1 and CCL3 compared to vehicle treated mice. In vitro, conditioned medium of T-cell enriched splenocytes of (44)AANA(47)-CCL5 treated mice inhibited the chemotaxis and proliferation of HSC. CONCLUSIONS: The results provide evidence that inhibition of oligomerization and glycosaminoglycan binding of the chemokine CCL5 is a new therapeutic strategy for the treatment of acute and chronic liver injuries and represents an alternative to chemokine receptor antagonism.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemokine CCL5/chemistry , Chemokine CCL5/metabolism , Glycosaminoglycans/metabolism , Protein Multimerization/drug effects , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Disease Models, Animal , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Male , Mice , Mice, Inbred C57BL , Mutation , Protein Binding/drug effects , Protein Structure, Quaternary
5.
Lab Invest ; 92(5): 724-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22430509

ABSTRACT

Although acute liver failure is a rare disease, its presence is associated with high morbidity and mortality in affected patients. While a contribution of the immune system to the outcome of toxic liver failure is anticipated, functionally relevant immune cell receptors for liver cell damage need to be better defined. We here investigate the relevance of the chemokine receptor CXCR3, which is important for hepatic immune cell infiltration, in a model of experimental acute liver failure. Liver injury was induced by a single intraperitoneal injection of carbon tetrachloride (CCl(4)) in CXCR3(-/-), CCR1(-/-), CCR5(-/-) and wild-type mice. In this model, CXCR3(-/-) mice displayed augmented liver damage compared with all other mouse strains as assessed by liver histology and serum transaminases 24 and 72 h after injury. Phenotypically, CXCR3(-/-) mice had significantly reduced intrahepatic NK and NKT cells after injury at all investigated time points (all P<0.05), but strongly elevated expression levels of IL1-ß, TNF-α and IFN-γ. In line with a functional role of innate immune cells, wild-type mice depleted for NK cells with an anti-ASIALO GM1 antibody before liver injury also displayed increased liver injury after CCl(4) challenge. CXCR3(-/-) and NK cell-depleted mice show reduced apoptotic liver cells (TUNEL assay), but more necrotic hepatocytes. Functionally, the augmented liver cell necrosis in CXCR3(-/-) and NK cell-depleted mice was associated with increased expression of high mobility group 1 (HMGB1) protein and a consecutive enhanced infiltration of neutrophils into the liver. In conclusion, the results demonstrate a primarily unexpected beneficial role of CXCR3 in acute toxic liver injury. These findings should be taken into account when planning trials with CXCR3 antagonists.


Subject(s)
HMGB1 Protein/metabolism , Liver Failure, Acute/immunology , Liver Failure, Acute/metabolism , Receptors, CXCR3/metabolism , Animals , Carbon Tetrachloride/pharmacology , HMGB1 Protein/immunology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Receptors, CCR1/immunology , Receptors, CCR1/metabolism , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Transaminases/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
Hepatology ; 55(5): 1610-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22237831

ABSTRACT

UNLABELLED: Recent data suggest that the chemokine receptor CXCR3 is functionally involved in fibroproliferative disorders, including liver fibrosis. Neoangiogenesis is an important pathophysiological feature of liver scarring, but a functional role of angiostatic CXCR3 chemokines in this process is unclear. We therefore investigated neoangiogenesis in carbon tetrachloride (CCl(4))-induced liver fibrosis in Cxcr3(-/-) and wildtype mice by histological, molecular, and functional imaging methods. Furthermore, we assessed the direct role of vascular endothelial growth factor (VEGF) overexpression on liver angiogenesis and the fibroproliferative response using a Tet-inducible bitransgenic mouse model. The feasibility of attenuation of angiogenesis and associated liver fibrosis by therapeutic treatment with the angiostatic chemokine Cxcl9 was systematically analyzed in vitro and in vivo. The results demonstrate that fibrosis progression in Cxcr3(-/-) mice was strongly linked to enhanced neoangiogenesis and VEGF/VEGFR2 expression compared with wildtype littermates. Systemic VEGF overexpression led to a fibrogenic response within the liver and was associated with a significantly increased Cxcl9 expression. In vitro, Cxcl9 displayed strong antiproliferative and antimigratory effects on VEGF-stimulated endothelial cells and stellate cells by way of reduced VEGFR2 (KDR), phospholipase Cγ (PLCγ), and extracellular signal-regulated kinase (ERK) phosphorylation, identifying this chemokine as a direct counter-regulatory molecule of VEGF signaling within the liver. Accordingly, systemic administration of Cxcl9 led to a strong attenuation of neoangiogenesis and experimental liver fibrosis in vivo. CONCLUSION: The results identify direct angiostatic and antifibrotic effects of the Cxcr3 ligand Cxcl9 in a model of experimental liver fibrosis. The amelioration of liver damage by systemic application of Cxcl9 might offer a novel therapeutic approach for chronic liver diseases associated with increased neoangiogenesis.


Subject(s)
Chemokine CXCL9/pharmacology , Hepatocytes/cytology , Liver Cirrhosis/drug therapy , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Analysis of Variance , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CXCL9/metabolism , Disease Models, Animal , Flow Cytometry , Hepatocytes/metabolism , Interferon-gamma/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/drug therapy , Random Allocation
7.
Proc Natl Acad Sci U S A ; 108(42): 17444-9, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21969590

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pleiotropic inflammatory cytokine that has been implicated in various inflammatory diseases. Chronic inflammation is a mainstay of liver fibrosis, a leading cause of morbidity worldwide, but the role of MIF in liver scarring has not yet been elucidated. Here we have uncovered an unexpected antifibrotic role for MIF. Mice genetically deleted in Mif (Mif(-/-)) showed strongly increased fibrosis in two models of chronic liver injury. Pronounced liver fibrosis in Mif(-/-) mice was associated with alterations in fibrosis-relevant genes, but not by a changed intrahepatic immune cell infiltration. Next, a direct impact of MIF on hepatic stellate cells (HSC) was assessed in vitro. Although MIF alone had only marginal effects on HSCs, it markedly inhibited PDGF-induced migration and proliferation of these cells. The inhibitory effects of MIF were mediated by CD74, which we detected as the most abundant known MIF receptor on HSCs. MIF promoted the phosphorylation of AMP-activated protein kinase (AMPK) in a CD74-dependent manner and, in turn, inhibition of AMPK reversed the inhibition of PDGF-induced HSC activation by MIF. The pivotal role of CD74 in MIF-mediated antifibrotic properties was further supported by augmented liver scarring of Cd74(-/-) mice. Moreover, mice treated with recombinant MIF displayed a reduced fibrogenic response in vivo. In conclusion, we describe a previously unexplored antifibrotic function of MIF that is mediated by the CD74/AMPK signaling pathway in HSCs. The results imply MIF and CD74 as targets for treatment of liver diseases.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/physiology , Histocompatibility Antigens Class II/physiology , Intramolecular Oxidoreductases/physiology , Liver Cirrhosis, Experimental/physiopathology , Macrophage Migration-Inhibitory Factors/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Carbon Tetrachloride/toxicity , Gene Expression , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Hepatic Stellate Cells/physiology , Histocompatibility Antigens Class II/genetics , Intramolecular Oxidoreductases/deficiency , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/pharmacology , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/pathology , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet-Derived Growth Factor/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction
8.
Dig Dis ; 28(1): 192-6, 2010.
Article in English | MEDLINE | ID: mdl-20460910

ABSTRACT

BACKGROUND/AIMS: Chemokines are small chemotactic molecules which regulate the infiltration of immune cells to sites of inflammatory injury. In recent years their contribution to the initiation and perpetuation of liver injury has been better defined. However, the role of chemokines in liver diseases related to the metabolic syndrome still needs to be elucidated in detail. METHODS: Chemokines were mostly detected at the mRNA level in the liver and as proteins in the serum of patients with non-alcoholic steatohepatitis (NASH) or fatty liver. Animals with targeted deletion of chemokines have recently been subjected to NASH models to functionally dissect the role of chemokines in fatty liver diseases. RESULTS: In human liver with features of NASH, different CC and CXC chemokines have been detected at elevated mRNA levels in comparison to healthy subjects. Some of these chemokines have also been associated with NASH by demonstrating higher serum levels in affected patients. Until now, only a few animal models have been analyzed with respect to the functional role of these molecules. However, data from CCL2 and CXCR3 knockout mice suggest that these pathways are important in liver injury. CCL2 seems to influence the infiltration of macrophages to adipose tissue and thereby modulate insulin resistance. CONCLUSIONS: The further elucidation of the pathophysiology of NASH will lead to new therapeutic options to halt or reverse progressive liver disease. In this respect, chemokines are attractive target molecules, as they influence immunologic and metabolic pathways and the first oral chemokine receptor antagonists have already been licensed for humans.


Subject(s)
Chemokines/immunology , Fatty Liver/immunology , Metabolic Syndrome/complications , Animals , Chemokines/metabolism , Disease Models, Animal , Fatty Liver/complications , Fatty Liver/metabolism , Fatty Liver/pathology , Humans , Inflammation , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Diseases/complications , Liver Diseases/immunology , Liver Diseases/metabolism , Liver Diseases/pathology , Mice , Mice, Knockout , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Receptors, Scavenger/immunology , Receptors, Scavenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...