Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 309(7): G542-53, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26251474

ABSTRACT

Interleukin (IL)-33 is crucially involved in liver pathology and drives hepatoprotective functions. However, the regulation of IL-33 by cytokines of the IL-6 family, including oncostatin M (OSM) and IL-6, is not well studied. The aim of the present study was to determine whether OSM mediates regulation of IL-33 expression in liver cells. Intramuscular administration in mice of an adenovirus encoding OSM (AdOSM) leads to increase in expression of OSM in muscles, liver, and serum of AdOSM-infected mice compared with control mice. The increase of circulating OSM markedly regulated mRNA of genes associated with blood vessel biology, chemotaxis, cellular death, induction of cell adhesion molecules, and the alarmin cytokine IL-33 in liver. Steady-state IL-33 mRNA was upregulated by OSM at an early phase (8 h) following AdOSM infection. At the protein level, the expression of IL-33 was significantly induced in liver endothelial cells [liver sinusoidal endothelial cells (LSEC) and vascular endothelial cells] with a peak at 8 days post-AdOSM infection in mice. In addition, we found OSM-stimulated human microvascular endothelial HMEC-1 cells and human LSEC/TRP3 cells showed a significant increase in expression of IL-33 mRNA in a dose-dependent manner in cell culture. The OSM-mediated overexpression of IL-33 was associated with the activation/enrichment of CD4(+)ST2(+) cells in liver of AdOSM-infected mice compared with adenovirus encoding green fluorescent protein-treated control mice. In summary, these data suggest that the cytokine OSM regulates the IL-33 expression in liver endothelial cells in vivo and in HMEC-1/TRP3 cells in vitro and may specifically expand the target CD4(+)ST2(+) cells in liver.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Endothelial Cells/metabolism , Growth Inhibitors/pharmacology , Interleukin-33/metabolism , Liver/drug effects , Oncostatin M/pharmacology , Animals , Cell Culture Techniques , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Flow Cytometry , Hepatocytes/drug effects , Humans , Interleukin-33/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
J Mol Med (Berl) ; 93(8): 867-78, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25747661

ABSTRACT

UNLABELLED: Interleukin-33 (IL-33), a cytokine belonging to the IL-1 family, is crucially involved in inflammatory pathologies including liver injury and linked to various modes of cell death. However, a link between IL-33 and necroptosis or programmed necrosis in liver pathology remains elusive. We aimed to investigate the regulation of IL-33 during necroptosis-associated liver injury. The possible regulation of IL-33 during liver injury by receptor-interacting protein kinase 1 (RIPK1) and poly(ADP-ribose) polymerase 1 (PARP-1) was investigated in mice in vivo and in hepatic stellate cells in vitro. The liver immunohistopathology, flow cytometry, serum transaminase measurement, ELISA, and qPCR-based cytokine measurement were carried out. By using a chemical approach, we showed that pretreatment of mice with Necrostatin-1 (Nec-1) (inhibitor of RIPK1) and/or PJ34 (inhibitor of PARP-1) significantly protected mice against concanavalin A (ConA) liver injury (aspartate amino-transferase (AST)/alanine amino-transferase (ALT)) associated with down-regulated hepatocyte-specific IL-33 expression. In contrast, the expression level of most systemic cytokines (except for IL-6) or activation of liver immune cells was not altered by chemical inhibitors rather an increased infiltration of neutrophils in the liver. During polyinosine-polycytidylic acid (Poly(I:C))-induced acute hepatitis, liver injury and hepatocyte-specific IL-33 expression was also inhibited by PJ34 without any protective effect of PJ34 in CCl4-induced liver injury. Moreover, PJ34 down-regulated the protein expression of IL-33 in activated hepatic stellate cells by cocktail of cytokines or staurosporine in vitro. In conclusion, we evidenced that the Nec-1/PJ34 is a potent inhibitor of liver injury and Nec-1/PJ34 down-regulated hepatocyte-specific IL-33 expression in the liver in vivo or in hepatic stellate cells in vitro, suggesting IL-33 as a possible readout of necroptosis-involved liver pathologies. KEY MESSAGE: Necroptosis inhibitors can protect mice against liver injury induced by ConA or Poly(I:C). IL-33 expression in liver injury in vivo is inhibited by PJ34. IL-33 expression in hepatic stellate cells in vitro is inhibited by PJ34. Hepatocyte-specific IL-33 expression is down-regulated by Nec-1/PJ34 during hepatitis. IL-33 is a new marker of necroptosis-associated liver injuries.


Subject(s)
Cell Death/drug effects , Down-Regulation/drug effects , Imidazoles/pharmacology , Indoles/pharmacology , Interleukin-33/genetics , Liver/drug effects , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , Cells, Cultured , Hepatitis/genetics , Hepatitis/pathology , Hepatitis/prevention & control , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Imidazoles/therapeutic use , Indoles/therapeutic use , Liver/injuries , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Phenanthrenes/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
3.
J Hepatol ; 60(2): 245-52, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24036009

ABSTRACT

BACKGROUND & AIMS: Infection with hepatitis C virus is a worldwide health problem. An inadequate Th2 cytokine response promotes the fibrosis-cirrhosis fate. Immune-modulating molecules favoring a Th2 profile, such as HLA-G molecules of the HLA class Ib family, may play a role in chronic hepatitis. HLA-G contributes to the escape of tumors, and their involvement in viral infections has been increasingly described. The aim of this work was to study the expression of HLA-G in the liver, its cellular source and its regulation in cases of chronic C hepatitis. METHODS: HLA-G cells in blocks of liver derived from patients infected with HCV were labeled by immunohistochemistry and enumerated. Double immunofluorescence allowed the identification of the cellular source. HLA-G secretion by a human mast cell line was quantified by ELISA after various stimulations. After treatment with IFN-α, real-time PCR was performed to determine the kinetics of cytokine expression profiles, followed by heat map clustering analysis. RESULTS: The number of HLA-G+ cells was significantly associated with the area of fibrosis. For the first time, we identify the HLA-G+ cells as being mast cells. HLA-G secretion was significantly induced in human mast cells stimulated by IL-10 or interferons of class I. The transcriptome of the secretome of this cell line stimulated by IFN-α revealed that (i) the HLA-G gene is upregulated late, and that (ii) T lymphocytes and NK cells are recruited. CONCLUSIONS: These findings suggest an autocrine loop in the genesis of HCV liver fibrosis, based on mast cells expressing HLA-G.


Subject(s)
HLA-G Antigens/metabolism , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/immunology , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Mast Cells/immunology , Adult , Aged , Aged, 80 and over , Chemokines/genetics , Cytokines/genetics , Disease Progression , Female , Gene Expression , HLA-G Antigens/genetics , Hepatitis C, Chronic/genetics , Humans , Immunohistochemistry , Interferon-alpha/metabolism , Liver/immunology , Liver/pathology , Liver Cirrhosis/genetics , Male , Mast Cells/pathology , Middle Aged , Th2 Cells/immunology
4.
PLoS One ; 8(9): e74278, 2013.
Article in English | MEDLINE | ID: mdl-24058536

ABSTRACT

The IL-33/ST2 axis is known to be involved in liver pathologies. Although, the IL-33 levels increased in sera of viral hepatitis patients in human, the cellular sources of IL-33 in viral hepatitis remained obscure. Therefore, we aimed to investigate the expression of IL-33 in murine fulminant hepatitis induced by a Toll like receptor (TLR3) viral mimetic, poly(I:C) or by pathogenic mouse hepatitis virus (L2-MHV3). The administration of poly(I:C) plus D-galactosamine (D-GalN) in mice led to acute liver injury associated with the induction of IL-33 expression in liver sinusoidal endothelial cells (LSEC) and vascular endothelial cells (VEC), while the administration of poly(I:C) alone led to hepatocyte specific IL-33 expression in addition to vascular IL-33 expression. The hepatocyte-specific IL-33 expression was down-regulated in NK-depleted poly(I:C) treated mice suggesting a partial regulation of IL-33 by NK cells. The CD1d KO (NKT deficient) mice showed hepatoprotection against poly(I:C)-induced hepatitis in association with increased number of IL-33 expressing hepatocytes in CD1d KO mice than WT controls. These results suggest that hepatocyte-specific IL-33 expression in poly(I:C) induced liver injury was partially dependent of NK cells and with limited role of NKT cells. In parallel, the L2-MHV3 infection in mice induced fulminant hepatitis associated with up-regulated IL-33 expression as well as pro-inflammatory cytokine microenvironment in liver. The LSEC and VEC expressed inducible expression of IL-33 following L2-MHV3 infection but the hepatocyte-specific IL-33 expression was only evident between 24 to 32h of post infection. In conclusion, the alarmin cytokine IL-33 was over-expressed during fulminant hepatitis in mice with LSEC, VEC and hepatocytes as potential sources of IL-33.


Subject(s)
Endothelial Cells/immunology , Hepatitis, Viral, Animal/immunology , Hepatitis/immunology , Interleukins/genetics , Liver/immunology , Murine hepatitis virus/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Endothelial Cells/pathology , Endothelial Cells/virology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Galactosamine/administration & dosage , Gene Deletion , Gene Expression/immunology , Hepatitis/etiology , Hepatitis/genetics , Hepatitis/pathology , Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Interleukin-33 , Interleukins/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Liver/pathology , Liver/virology , Mice , Mice, Knockout , Murine hepatitis virus/pathogenicity , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Natural Killer T-Cells/virology , Poly I-C/administration & dosage
5.
Hepatology ; 56(6): 2353-62, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22961755

ABSTRACT

UNLABELLED: Interleukin (IL)-33, a member of the IL-1 cytokine family, positively correlates with acute hepatitis and chronic liver failure in mice and humans. IL-33 is expressed in hepatocytes and is regulated by natural killer T (NKT) cells during concanavalin A (ConA)-induced acute liver injury. Here, we investigated the molecular mechanisms underlying the expression of IL-33 during acute hepatitis. The expression of IL-33 and its regulation by death receptor pathways was investigated after the induction of ConA-acute hepatitis in wildtype (WT), perforin(-/-) , tumor necrosis factor related apoptosis inducing ligand (TRAIL)(-/-) , and NKT cell-deficient (CD1d(-/-) ) mice. In addition, we used a model of acute liver injury by administering Jo2/Fas-antibody or D-galactosamine-tumor necrosis factor alpha (TNFα) in WT mice. Finally, the effect of TRAIL on IL-33 expression was assessed in primary cultured murine hepatocytes. We show that IL-33 expression in hepatocytes is partially controlled by perforin during acute liver injury, but not by TNFα or Fas ligand (FasL). Interestingly, the expression of IL-33 in hepatocytes is blocked during ConA-acute hepatitis in TRAIL-deficient mice compared to WT mice. In contrast, administration of recombinant murine TRAIL associated with ConA-priming in CD1d-deficient mice or in vitro stimulation of murine hepatocytes by TRAIL but not by TNFα or Jo2 induced IL-33 expression in hepatocytes. The IL-33-deficient mice exhibited more severe ConA liver injury than WT controls, suggesting a protective effect of IL-33 in ConA-hepatitis. CONCLUSION: The expression of IL-33 during acute hepatitis is dependent on TRAIL, but not on FasL or TNFα.


Subject(s)
Acute Lung Injury/metabolism , Hepatitis, Animal/metabolism , Interleukins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Acute Lung Injury/chemically induced , Animals , Antibodies, Monoclonal , Antibodies, Monoclonal, Murine-Derived , Concanavalin A , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Galactosamine , Gene Expression , Hepatitis, Animal/chemically induced , Hepatitis, Animal/immunology , Hepatocytes/metabolism , Interleukin-33 , Interleukins/genetics , Mice , Natural Killer T-Cells , Perforin/genetics , Perforin/metabolism , Primary Cell Culture , RNA, Messenger , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Eur J Cancer ; 48(1): 138-48, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21778049

ABSTRACT

Development of hepatocellular carcinoma (HCC) is a complex and progressive disease that involves cycles of liver cell death, inflammation, and tissue regeneration/remodelling. Chemokines and chemokine receptors play numerous and integral roles in the disease progression of HCC. Here we investigated the novel chemokine receptor CXCR7/RDC1 in HCC progression, its two known ligands CXCL12 and CXCL11, as well as the other CXCL12 receptor, CXCR4. Our results show that in a cohort of 408 human HCCs, CXCR7 and CXCL11 were significantly higher in tumours compared to normal liver controls (5- and 10-fold, respectively). Immunohistochemical (IHC) staining on human HCC sections confirmed that both CXCL11 and CXCR7 were much higher in cancer tissues. Furthermore, IHC staining revealed that CXCR7 protein was only expressed in endothelial cells whereas CXCL11 exhibited a much broader tissue expression. At the cellular level we observed that in vitro, human microvascular endothelial cells (HMEC-1) up-regulated CXCR7 under hypoxic and acidic pH conditions, which are well known characteristics of the HCC tumour micro-environment. As for its ligand, we observed that IFNγ robustly induced CXCL11 in hepatic stellate cells, hepatocytes, and HMEC-1s. In addition, in the mouse Diethylnitrosamine model of hepatocarcinogenesis we observed a very strong induction of CXCR7 and CXCL11 transcripts, confirming that CXCR7/CXCL11 up-regulation is conserved between human and mice liver cancer. Altogether, our results strongly support the hypothesis that the CXCL11/CXCR7 pathway is involved HCC progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Endothelial Cells/metabolism , Liver Neoplasms/genetics , Receptors, CXCR/genetics , Aged , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Disease Progression , Endothelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Organ Specificity/genetics , Receptors, CXCR/metabolism , Tumor Cells, Cultured
7.
Am J Respir Cell Mol Biol ; 45(6): 1125-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21642589

ABSTRACT

IL-33, a novel IL-1 family member, is crucially expressed and involved in pulmonary diseases, but its regulation in viral diseases such as influenza A virus (IAV) remains unclear. This study aimed to characterize the expression and release of IL-33 in lungs of IAV-infected mice in vivo and in murine respiratory epithelial cells (MLE-15) in vitro. Our results provide evidence of up-regulation of IL-33 mRNA in IAV-infected murine lungs, compared with noninfected control mice. The overexpression of IL-33 was positively correlated with a significant increase in mRNA encoding the proinflammatory cytokines TNF-α, IFN-γ, IL-1ß, and IL-6, and was also associated with an increase in IFN-ß mRNA. A profound overexpression of IL-33 protein was evident in IAV-infected murine lungs and bronchoalveolar lavages of influenza-infected mice, compared with low concentrations in naive lungs in vivo. Immunolocalization highlighted the cellular expression of IL-33 in alveolar epithelial and endothelial cells, along with increased infiltrate cells in virus-infected lungs. Further in vitro experiments showed an induction of IL-33 transcript-in MLE-15 cells and human epithelial cells (A549) infected with different strains of IAV in comparison with noninfected cells. In conclusion, our findings evidenced a profound expression of IL-33 in lungs during both in vivo and in vitro IAV infections, suggesting a role for IL-33 in virus-induced lung infections.


Subject(s)
Gene Expression Regulation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Interleukins/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Animals , Bronchoalveolar Lavage , Cell Line , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/virology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Interleukin-33 , Interleukin-6/biosynthesis , Interleukin-6/immunology , Interleukins/biosynthesis , Lung/metabolism , Lung/virology , Mice , Orthomyxoviridae Infections/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
8.
Eur J Immunol ; 41(8): 2341-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21557213

ABSTRACT

Interleukin-33 (IL-33) is thought to be released during cellular death as an alarming cytokine during the acute phase of disease, but its regulation in vivo is poorly understood. We investigated the expression of IL-33 in two mouse models of acute hepatitis by administering either carbon tetrachloride (CCl(4) ) or concanavalin A (ConA). IL-33 was overexpressed in both models but with a stronger induction in ConA-induced hepatitis. IL-33 was weakly expressed in vascular and sinusoidal endothelial cells from normal liver and was clearly induced in CCl(4) -treated mice. Surprisingly, we found that hepatocytes strongly expressed IL-33 exclusively in the ConA model. CD1d knock-out mice, which are deficient in NKT cells and resistant to ConA-induced hepatitis, no longer expressed IL-33 in hepatocytes following ConA administration. Interestingly, invariant NKT (iNKT) cells adoptively transferred into ConA-treated CD1d KO mouse restored IL-33 expression in hepatocytes. This strongly suggests that NKT cells are responsible for the induction of IL-33 in hepatocytes.


Subject(s)
Hepatitis, Animal/genetics , Hepatocytes/metabolism , Interleukins/genetics , Natural Killer T-Cells/metabolism , Acute Disease , Adoptive Transfer , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Carbon Tetrachloride , Concanavalin A , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-33 , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Eur J Immunol ; 41(6): 1720-32, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469102

ABSTRACT

Invariant natural killer T (iNKT) cells are involved in the intrahepatic immune response and in hepatitis. In particular, iNKT lymphocytes are responsible for hepatocyte death in concanavalin A-induced hepatitis in mice. We examined the role of iNKT cells in acute hepatitis induced by a hepatotoxic agent, carbon tetrachloride (CCl(4) ). WT and iNKT cell-deficient (Jα18(-/-) ) mice were challenged with a single dose of 2.4 g/kg CCl(4) and both hepatic physiopathology and immune responses were studied. Plasma alanine and aspartate amino-transferase levels were significantly higher in Jα18(-/-) mice than in WT mice two days after CCl(4) administration. Chemokine CXCL1/keratinocyte-derived chemokine (KC) and MMP-8 were significantly higher in iNKT cell-deficient mice than in control mice. The more severe liver injury in Jα18(-/-) mice was associated with greater leukocyte infiltrate, which was enriched in neutrophils (CD11b(+) CD11c(-) Gr-1(+) cells), in agreement with CXCL1/KC and MMP-8 levels. Complementary experiments with NK-depleted animals indicate a minor role for NK cells in the liver damage found in iNKT-deficient mice. Thus, unlike for ConA-induced hepatitis, we report that iNKT cells protect the liver against acute hepatitis induced by CCl(4) and limit neutrophil infiltration.


Subject(s)
Chemokine CXCL1/metabolism , Hepatitis, Animal/immunology , Liver/metabolism , Natural Killer T-Cells/metabolism , Neutrophils/metabolism , Acute Disease , Alanine/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Hepatitis, Animal/blood , Hepatitis, Animal/chemically induced , Hepatocytes/pathology , Liver/drug effects , Liver/immunology , Liver/pathology , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/immunology , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Receptors, Antigen, T-Cell, alpha-beta/genetics
10.
J Cell Mol Med ; 14(6B): 1726-39, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19508382

ABSTRACT

Interleukin-33 (IL-33), the most recently identified member of the IL-1 family, induces synthesis of T Helper 2 (Th2)-type cytokines via its heterodimeric ST2/IL-1RAcP receptor. Th2-type cytokines play an important role in fibrosis; thus, we investigated the role of IL-33 in liver fibrosis. IL-33, ST2 and IL-1RAcP gene expression was analysed in mouse and human normal (n= 6) and fibrotic livers (n= 28), and in human hepatocellular carcinoma (HCC; n= 22), using real-time PCR. IL-33 protein was detected in normal and fibrotic liver sections and in isolated liver cells using Western blotting and immunolocalization approaches. Our results showed that IL-33 and ST2 mRNA was overproduced in mouse and human fibrotic livers, but not in human HCC. IL-33 expression correlated with ST2 expression and also with collagen expression in fibrotic livers. The major sources of IL-33 in normal liver from both mice and human beings are the liver sinusoidal endothelial cells and, in fibrotic liver, the activated hepatic stellate cells (HSC). Moreover, IL-33 expression was increased in cultured HSC when stimulated by pro-inflammatory cytokines. In conclusion, IL-33 is strongly associated with fibrosis in chronic liver injury and activated HSC are a source of IL-33.


Subject(s)
Interleukins/metabolism , Liver Cirrhosis/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Nucleus/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/metabolism , Interleukin-33 , Interleukins/genetics , Liver/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Middle Aged , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
J Cell Mol Med ; 13(8A): 1526-35, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19432809

ABSTRACT

Chemokines are the inflammatory mediators that modulate liver fibrosis, a common feature of chronic inflammatory liver diseases. CX3CL1/fractalkine is a membrane-associated chemokine that requires step processing for chemotactic activity and has been recently implicated in liver disease. Here, we investigated the potential shedding activities involved in the release of the soluble chemotactic peptides from CX3CL1 in the injured liver. We showed an increased expression of the sheddases ADAM10 and ADAM17 in patients with chronic liver diseases that was associated with the severity of liver fibrosis. We demonstrated that hepatic stellate cells (HSC) were an important source of ADAM10 and ADAM17 and that treatment with the inflammatory cytokine inter-feron-gamma induced the expression of CX3CL1 and release of soluble peptides. This release was inhibited by the metalloproteinase inhibitor batimastat; however, ADAM10/ADAM17 inhibitor GW280264X only partially affected shedding activity. By using selective tissue metalloprotease inhibitors and overexpression analyses, we showed that CX3CL1 was mainly processed by matrix metalloproteinase (MMP)-2, a metalloprotease highly expressed by HSC. We further demonstrated that the CX3CL1 soluble peptides released from stimulated HSC induced the activation of the CX3CR1-dependent signalling pathway and promoted chemoattraction of monocytes in vitro. We conclude that ADAM10, ADAM17 and MMP-2 synthesized by activated HSC mediate CX3CL1 shedding and release of chemotactic peptides, thereby facilitating recruitment of inflammatory cells and paracrine stimulation of HSC in chronic liver diseases.


Subject(s)
Chemokine CX3CL1/metabolism , Hepatic Stellate Cells/metabolism , Inflammation/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Cell Line , Chemokine CX3CL1/genetics , Chronic Disease , Collagen Type I/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/pathology , Humans , Interferon-gamma/pharmacology , Liver/drug effects , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Matrix Metalloproteinase 2/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solubility/drug effects , Up-Regulation/drug effects
12.
Mol Biol Cell ; 19(11): 4738-49, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18716062

ABSTRACT

Reepithelialization during cutaneous wound healing involves numerous signals that result in basal keratinocyte activation, spreading, and migration, all linked to a loosening of cell-cell adhesion structures. The transcription factor Slug is required for this process, and EGF treatment of human keratinocytes induced activating phosphorylation of Erk5 that coincides with slug transcription. Accordingly, ectopic activation of Erk5 led to increased Slug mRNA levels and faster wound healing, whereas keratinocyte migration was totally blocked by Erk5 pathway inhibition. Expression of a shRNA specific for Erk5 strongly diminished Erk5 levels in keratinocytes and significantly decreased their motility response to EGF, along with induction of Slug expression. These Erk5-deprived keratinocytes showed an altered, more compact morphology, along with disruption of desmosome organization. Accordingly, they displayed an altered ability to form cell aggregates. These results implicate a novel EGFR/Erk5/Slug pathway in the control of cytoskeleton organization and cell motility in keratinocytes treated with EGF.


Subject(s)
Keratinocytes/cytology , Keratinocytes/enzymology , Mitogen-Activated Protein Kinase 7/metabolism , Transcription Factors/genetics , Wound Healing , Animals , CHO Cells , Cell Adhesion/drug effects , Cricetinae , Cricetulus , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epidermal Growth Factor/pharmacology , Epithelium/drug effects , Epithelium/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Humans , Keratinocytes/drug effects , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Snail Family Transcription Factors , Transcription Factors/deficiency , Transcription Factors/metabolism , Wound Healing/drug effects
13.
J Biol Chem ; 283(38): 26000-9, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18621736

ABSTRACT

ADAM12 belongs to a disintegrin-like and metalloproteinase-containing protein family that possesses multidomain structures composed of a pro-domain, a metalloprotease, disintegrin-like, cysteine-rich, epidermal growth factor-like, and transmembrane domains, and a cytoplasmic tail. Overexpression of several ADAMs has been reported in human cancer, and we recently described the involvement of ADAM12 in liver injury (Le Pabic, H., Bonnier, D., Wewer, U. M., Coutand, A., Musso, O., Baffet, G., Clement, B., and Theret, N. (2003) Hepatology 37, 1056-1066). In this study, we used a yeast two-hybrid screening of a cDNA library from human hepatocellular carcinoma to analyze binding partners of ADAM12. We identify RACK1, a receptor for activated protein kinase C (PKC), as a new ADAM12 interacting protein. RACK1 is up-regulated in patients with hepatocellular carcinoma and is highly expressed by activated hepatic stellate cells. We demonstrate the involvement of RACK1 in mediating the PKC-dependent translocation of ADAM12 to membranes of activated hepatic stellate cells. In particular, treatment of cells with phorbol esters enhances ADAM12 immunostaining in the membrane fractions and the co-immunoprecipitation of ternary complexes containing RACK1, ADAM12, and PKC. By using RNA interference, we demonstrate that inhibition of RACK1 expression diminishes the phorbol 12-myristate 13-acetate-dependent translocation of ADAM12 to membranes of hepatic stellate cells. Finally, hepatic stellate cells cultured on coated type I collagen induces relocalization of ADAM12 in the membrane, suggesting that this major matrix component in liver cancer and fibrogenesis might stimulate ADAM12 translocation to the cell membrane where its shedding activity takes place.


Subject(s)
ADAM Proteins/metabolism , GTP-Binding Proteins/physiology , Liver/metabolism , Liver/pathology , Membrane Proteins/metabolism , Neoplasm Proteins/physiology , Receptors, Cell Surface/physiology , ADAM12 Protein , Amino Acid Sequence , Biotinylation , Cell Membrane/metabolism , Fibrin/chemistry , GTP-Binding Proteins/chemistry , Humans , Integrin beta1/metabolism , Liver/cytology , Models, Genetic , Molecular Sequence Data , Neoplasm Proteins/chemistry , Protein Biosynthesis , Protein Transport , Receptors for Activated C Kinase , Receptors, Cell Surface/chemistry , Tetradecanoylphorbol Acetate/pharmacology , Two-Hybrid System Techniques
14.
J Cell Biol ; 178(2): 201-8, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17620406

ABSTRACT

Transforming growth factor-beta (TGF-beta) regulates a wide variety of biological processes through two types of Ser/Thr transmembrane receptors: the TGF-beta type I receptor and the TGF-beta type II receptor (TbetaRII). Upon ligand binding, TGF-beta type I receptor activated by TbetaRII propagates signals to Smad proteins, which mediate the activation of TGF-beta target genes. In this study, we identify ADAM12 (a disintegrin and metalloproteinase 12) as a component of the TGF-beta signaling pathway that acts through association with TbetaRII. We found that ADAM12 functions by a mechanism independent of its protease activity to facilitate the activation of TGF-beta signaling, including the phosphorylation of Smad2, association of Smad2 with Smad4, and transcriptional activation. Furthermore, ADAM12 induces the accumulation of TbetaRII in early endosomal vesicles and stabilizes the TbetaRII protein presumably by suppressing the association of TbetaRII with Smad7. These results define ADAM12 as a new partner of TbetaRII that facilitates its trafficking to early endosomes in which activation of the Smad pathway is initiated.


Subject(s)
ADAM Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , ADAM12 Protein , Animals , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Fluorescent Antibody Technique , Genes, Reporter , Humans , Luciferases/metabolism , Lung/cytology , Mice , Mink , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Plasmids , Transfection , Transforming Growth Factor beta , Two-Hybrid System Techniques
15.
Hepatology ; 44(5): 1296-307, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17058241

ABSTRACT

The molecular mechanisms underlying the progression of cirrhosis toward hepatocellular carcinoma were investigated by a combination of DNA microarray analysis and literature data mining. By using a microarray screening of suppression subtractive hybridization cDNA libraries, we first analyzed genes differentially expressed in tumor and nontumor livers with cirrhosis from 15 patients with hepatocellular carcinomas. Seventy-four genes were similarly recovered in tumor (57.8% of differentially expressed genes) and adjacent nontumor tissues (64% of differentially expressed genes) compared with histologically normal livers. Gene ontology analyses revealed that downregulated genes (n = 35) were mostly associated with hepatic functions. Upregulated genes (n = 39) included both known genes associated with extracellular matrix remodeling, cell communication, metabolism, and post-transcriptional regulation gene (e.g., ZFP36L1), as well as the tumor suppressor gene menin (multiple endocrine neoplasia type 1; MEN1). MEN1 was further identified as an important node of a regulatory network graph that integrated array data with array-independent literature mining. Upregulation of MEN1 in tumor was confirmed in an independent set of samples and associated with tumor size (P = .016). In the underlying liver with cirrhosis, increased steady-state MEN1 mRNA levels were correlated with those of collagen alpha2(I) mRNA (P < .01). In addition, MEN1 expression was associated with hepatic stellate cell activation during fibrogenesis and involved in transforming growth factor beta (TGF-beta)-dependent collagen alpha2(I) regulation. In conclusion, menin is a key regulator of gene networks that are activated in fibrogenesis associated with hepatocellular carcinoma through the modulation of TGF-beta response.


Subject(s)
Carcinoma, Hepatocellular/genetics , Collagen Type I/metabolism , Hepatocytes/physiology , Liver Cirrhosis/metabolism , Proto-Oncogene Proteins/genetics , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Gene Library , Genes, Tumor Suppressor , Humans , Liver Cirrhosis/complications , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/biosynthesis , Transfection , Transforming Growth Factor beta/metabolism , Up-Regulation
16.
J Hepatol ; 43(6): 1038-44, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16139919

ABSTRACT

BACKGROUND/AIMS: In chronic liver injury, quiescent hepatic stellate cells change into proliferative myofibroblast-like cells, which are a main source of fibrosis. We have recently reported that these cells synthesize ADAM12, a disintegrin and metalloprotease whose expression is up-regulated by TGF-beta1 in liver cancers. Here, we studied the role of the serine/threonine p70S6 kinase (p70S6K) in regulating TGF-beta1-induced ADAM12 expression. RESULTS: The phophatidylinositol 3-kinase (PI3K) inhibitor LY294002 and the mitogen-activated protein kinase inhibitor, UO126, decreased the TGF-beta1-dependent ADAM12 expression and prevented the phosphorylation of p70S6K. In addition, TGF-beta1-induced ADAM12 up-regulation was blocked by the Frap/mTOR inhibitor rapamycin, which abrogated the phosphorylation of p70S6K. In untreated cells, LY294002 but not rapamycin diminished the basal ADAM12 expression related to inhibition of Akt and the glycogen synthase kinase-3 phosphorylation. CONCLUSIONS: The data suggest that TGF-beta1 induces ADAM12 gene expression through both the PI3K/Frap-mTOR/p70S6K and MEK/ERK pathways. In addition, activation of the PI3 pathway might be involved in the basal ADAM12 expression in cultured hepatic stellate cells. The involvement of PI3K in ADAM12 expression, similar to that previously observed for collagen I and fibronectin, suggests common pathways for gene up-regulation in hepatic stellate cells that occur during liver fibrogenesis and contribute to tumor progression.


Subject(s)
ADAM Proteins/genetics , Membrane Proteins/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Transforming Growth Factor beta/metabolism , ADAM Proteins/metabolism , ADAM12 Protein , Cells, Cultured , Gene Expression , Humans , Liver/cytology , MAP Kinase Kinase Kinases/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...