Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Int J Cancer ; 154(11): 1999-2013, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38308587

ABSTRACT

The global pandemic of metabolic diseases has increased the incidence of hepatocellular carcinoma (HCC) in the context of non-alcoholic steatohepatitis (NASH). The downregulation of the E3 ubiquitin ligase TRIM21 has been linked to poor prognosis in different cancers including HCC. In order to investigate the role of TRIM21 in liver cancer progression on NASH, Trim21+/+ and Trim21-/- male mice were injected with streptozotocin at the neonatal stage. The hypoinsulinemic mice were then fed with a high-fat high-cholesterol diet (HFHCD) for 4, 8 or 12 weeks. All mice developed NASH which systematically resulted in HCC progression. Interestingly, compared to the Trim21+/+ control mice, liver damage was worsened in Trim21-/- mice, with more HCC nodules found after 12 weeks on HFHCD. Immune population analysis in the spleen and liver revealed a higher proportion of CD4+PD-1+ and CD8+PD-1+ T cells in Trim21-/- mice. The liver and HCC tumors of Trim21-/- mice also exhibited an increase in the number of PD-L1+ and CD68+ PD-L1+ cells. Thus, TRIM21 limits the emergence of HCC nodules in mice with NASH by potentially restricting the expression of PD-1 in lymphocytes and PD-L1 in tumors.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Ribonucleoproteins , Animals , Male , Mice , B7-H1 Antigen/metabolism , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/complications , Disease Models, Animal , Liver Neoplasms/genetics , Liver Neoplasms/complications , Non-alcoholic Fatty Liver Disease/complications , Programmed Cell Death 1 Receptor/metabolism , Up-Regulation , Ribonucleoproteins/deficiency , Ribonucleoproteins/genetics
2.
Int J Mol Sci ; 23(13)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35806372

ABSTRACT

Some life-threatening acute hepatitis originates from drug-induced liver injury (DILI). Carbon tetrachloride (CCl4)-induced acute liver injury in mice is the widely used model of choice to study acute DILI, which pathogenesis involves a complex interplay of oxidative stress, necrosis, and apoptosis. Since the receptor interacting protein kinase-1 (RIPK1) is able to direct cell fate towards survival or death, it may potentially affect the pathological process of xenobiotic-induced liver damage. Two different mouse lines, either deficient for Ripk1 specifically in liver parenchymal cells (Ripk1LPC-KO) or for the kinase activity of RIPK1 (Ripk1K45A, kinase dead), plus their respective wild-type littermates (Ripk1fl/fl, Ripk1wt/wt), were exposed to single toxic doses of CCl4. This exposure led in similar injury in Ripk1K45A mice and their littermate controls. However, Ripk1LPC-KO mice developed more severe symptoms with massive hepatocyte apoptosis as compared to their littermate controls. A pretreatment with a TNF-α receptor decoy exacerbated liver apoptosis in both Ripk1fl/fl and Ripk1LPC-KO mice. Besides, a FasL antagonist promoted hepatocyte apoptosis in Ripk1fl/fl mice but reduced it in Ripk1LPC-KO mice. Thus, the scaffolding properties of RIPK1 protect hepatocytes from apoptosis during CCl4 intoxication. TNF-α and FasL emerged as factors promoting hepatocyte survival. These protective effects appeared to be independent of RIPK1, at least in part, for TNF-α, but dependent on RIPK1 for FasL. These new data complete the deciphering of the molecular mechanisms involved in DILI in the context of research on their prevention or cure.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Hepatitis , Animals , Apoptosis , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Hepatitis/metabolism , Hepatocytes/metabolism , Liver/metabolism , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
J Hepatol ; 66(6): 1205-1213, 2017 06.
Article in English | MEDLINE | ID: mdl-28088582

ABSTRACT

BACKGROUND & AIMS: The severity of liver diseases is exacerbated by the death of hepatocytes, which can be induced by the sensing of pathogen associated molecular patterns (PAMPs) derived from the gut microbiota. The molecular mechanisms regulating these cell death pathways are poorly documented. In this study, we investigated the role of the receptor interacting protein kinase 1 (RIPK1), a protein known to regulate cell fate decisions, in the death of hepatocytes using two in vivo models of PAMP-induced hepatitis. METHODS: Hepatitis was induced in mice by independent injections of two different bacterial PAMPs: lipopolysaccharide (LPS) and unmethylated CpG oligodeoxynucleotide (CpG-DNA) motifs. The role of RIPK1 was evaluated by using mice specifically lacking RIPK1 in liver parenchymal cells (Ripk1LPC-KO). Administration of liposome-encapsulated clodronate served to investigate the role of Kupffer cells in the establishment of the disease. Etanercept, a tumor necrosis factor (TNF)-decoy receptor, was used to study the contribution of TNF-α during LPS-mediated liver injury. RESULTS: Whereas RIPK1 deficiency in liver parenchymal cells did not trigger basal hepatolysis, it greatly sensitized hepatocytes to apoptosis and liver damage following a single injection of LPS or CpG-DNA. Importantly, hepatocyte death was prevented by previous macrophage depletion or by TNF inhibition. CONCLUSIONS: Our data highlight the pivotal function of RIPK1 in maintaining liver homeostasis in conditions of macrophage-induced TNF burst in response to PAMPs sensing. LAY SUMMARY: Excessive death of hepatocytes is a characteristic of liver injury. A new programmed cell death pathway has been described involving upstream death ligands such as TNF and downstream kinases such as RIPK1. Here, we show that in the presence of LPS liver induced hepatic injury was due to secretion of TNF by liver macrophages, and that RIPK1 acts as a powerful protector of hepatocyte death. This newly identified pathway in the liver may be helpful in the management of patients to predict their risk of developing acute liver failure.


Subject(s)
Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Pathogen-Associated Molecular Pattern Molecules/toxicity , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Hepatitis, Animal/etiology , Hepatocytes/drug effects , Kupffer Cells/drug effects , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
4.
Am J Physiol Gastrointest Liver Physiol ; 313(5): G399-G409, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28751426

ABSTRACT

Excessive or persistent inflammation and hepatocyte death are the key triggers of liver diseases. The poly(ADP-ribose) polymerase (PARP) proteins induce cell death and inflammation. Chemical inhibition of PARP activity protects against liver injury during concanavalin A (ConA)-induced hepatitis. In this mice model, ConA activates immune cells, which promote inflammation and induce hepatocyte death, mediated by the activated invariant natural killer T (iNKT) lymphocyte population. We analyzed immune cell populations in the liver and several lymphoid organs, such as the spleen, thymus, and bone marrow in Parp2-deficient mice to better define the role of PARP proteins in liver immunity and inflammation at steady state and during ConA-induced hepatitis. We show that 1) the genetic inactivation of Parp2, but not Parp1, protected mice from ConA hepatitis without deregulating cytokine expression and leucocyte recruitment; 2) cellularity was lower in the thymus, but not in spleen, liver, or bone marrow of Parp2-/- mice; 3) spleen and liver iNKT lymphocytes, as well as thymic T and NKT lymphocytes were reduced in Parp2 knockout mice. In conclusion, our results suggest that the defect of T-lymphocyte maturation in Parp2 knockout mice leads to a systemic reduction of iNKT cells, reducing hepatocyte death during ConA-mediated liver damage, thus protecting the mice from hepatitis.NEW & NOTEWORTHY The genetic inactivation of Parp2, but not Parp1, protects mice from concanavalin A hepatitis. Immune cell populations are lower in the thymus, but not in the spleen, liver, or bone marrow of Parp2-deficient mice compared with wild-type mice. Spleen and liver invariant natural killer T (NKT) lymphocytes, as well as thymic T and NKT lymphocytes, are reduced in Parp2-deficient mice.


Subject(s)
Chemical and Drug Induced Liver Injury , Hepatocytes , Natural Killer T-Cells , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Thymus Gland , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Cell Death/drug effects , Cell Death/physiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/pharmacology , Disease Models, Animal , Hepatitis/etiology , Hepatitis/immunology , Hepatitis/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Inflammation/chemically induced , Inflammation/immunology , Mice , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/physiology , Protective Factors , Spleen/immunology , Spleen/pathology , Thymus Gland/immunology , Thymus Gland/pathology
5.
Mediators Inflamm ; 2017: 1359064, 2017.
Article in English | MEDLINE | ID: mdl-28607531

ABSTRACT

The alarmin IL-33 has been described to be upregulated in human and murine viral hepatitis. However, the role of endogenous IL-33 in viral hepatitis remains obscure. We aimed to decipher its function by infecting IL-33-deficient mice (IL-33 KO) and their wild-type (WT) littermates with pathogenic mouse hepatitis virus (L2-MHV3). The IL-33 KO mice were more sensitive to L2-MHV3 infection exhibiting higher levels of AST/ALT, higher tissue damage, significant weight loss, and earlier death. An increased depletion of B and T lymphocytes, NKT cells, dendritic cells, and macrophages was observed 48 h postinfection (PI) in IL-33 KO mice than that in WT mice. In contrast, a massive influx of neutrophils was observed in IL-33 KO mice at 48 h PI. A transcriptomic study of inflammatory and cell-signaling genes revealed the overexpression of IL-6, TNFα, and several chemokines involved in recruitment/activation of neutrophils (CXCL2, CXCL5, CCL2, and CCL6) at 72 h PI in IL-33 KO mice. However, the IFNγ was strongly induced in WT mice with less profound expression in IL-33 KO mice demonstrating that endogenous IL-33 regulated IFNγ expression during L2-MHV3 hepatitis. In conclusion, we demonstrated that endogenous IL-33 had multifaceted immunoregulatory effect during viral hepatitis via induction of IFNγ, survival effect on immune cells, and infiltration of neutrophils in the liver.


Subject(s)
Hepatitis/immunology , Hepatitis/metabolism , Interleukin-33/metabolism , Liver/metabolism , Neutrophils/metabolism , Animals , B-Lymphocytes/metabolism , Chemokine CCL2/metabolism , Chemokine CXCL2/metabolism , Chemokine CXCL5/metabolism , Chemokines, CC/metabolism , Interferon-gamma/metabolism , Interleukin-33/deficiency , Interleukin-6/metabolism , Mice , Mice, Knockout , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Immunology ; 149(2): 204-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27273587

ABSTRACT

Viral replication in the liver is generally detected by cellular endosomal Toll-like receptors (TLRs) and cytosolic helicase sensors that trigger antiviral inflammatory responses. Recent evidence suggests that surface TLR2 may also contribute to viral detection through recognition of viral coat proteins but its role in the outcome of acute viral infection remains elusive. In this study, we examined in vivo the role of TLR2 in acute infections induced by the highly hepatotrophic mouse hepatitis virus (MHV) type 3 and weakly hepatotrophic MHV-A59 serotype. To address this, C57BL/6 (wild-type; WT) and TLR2 knockout (KO) groups of mice were intraperitoneally infected with MHV3 or MHV-A59. MHV3 infection provoked a fulminant hepatitis in WT mice, characterized by early mortality and high alanine and aspartate transaminase levels, histopathological lesions and viral replication whereas infection of TLR2 KO mice was markedly less severe. MHV-A59 provoked a comparable mild and subclinical hepatitis in WT and TLR2 KO mice. MHV3-induced fulminant hepatitis in WT mice correlated with higher hepatic expression of interferon-ß, interleukin-6, tumour necrosis factor-α, CXCL1, CCL2, CXCL10 and alarmin (interleukin-33) than in MHV-A59-infected WT mice and in MHV3-infected TLR2 KO mice. Intrahepatic recruited neutrophils, natural killer cells, natural killer T cells or macrophages rapidly decreased in MHV3-infected WT mice whereas they were sustained in MHV-A59-infected WT mice and MHV3-infected TLR2 KO. MHV3 in vitro infection of macrophagic cells induced rapid and higher viral replication and/or interleukin-6 induction in comparison to MHV-A59, and depended on viral activation of TLR2 and p38 mitogen-activated protein kinase. Taken together, these results support a new aggravating inflammatory role for TLR2 in MHV3-induced acute fulminant hepatitis.


Subject(s)
Hepatitis, Viral, Animal/immunology , Murine hepatitis virus/physiology , Toll-Like Receptor 2/metabolism , Acute Disease , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cells, Cultured , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 2/genetics , Virus Replication/genetics , p38 Mitogen-Activated Protein Kinases/administration & dosage , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G313-23, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27340126

ABSTRACT

The IL-33/ST2 axis plays a protective role in T-cell-mediated hepatitis, but little is known about the functional impact of endogenous IL-33 on liver immunopathology. We used IL-33-deficient mice to investigate the functional effect of endogenous IL-33 in concanavalin A (Con A)-hepatitis. IL-33(-/-) mice displayed more severe Con A liver injury than wild-type (WT) mice, consistent with a hepatoprotective effect of IL-33. The more severe hepatic injury in IL-33(-/-) mice was associated with significantly higher levels of TNF-α and IL-1ß and a larger number of NK cells infiltrating the liver. The expression of Th2 cytokines (IL-4, IL-10) and IL-17 was not significantly varied between WT and IL-33(-/-) mice following Con A-hepatitis. The percentage of CD25(+) NK cells was significantly higher in the livers of IL-33(-/-) mice than in WT mice in association with upregulated expression of CXCR3 in the liver. Regulatory T cells (Treg cells) strongly infiltrated the liver in both WT and IL-33(-/-) mice, but Con A treatment increased their membrane expression of ST2 and CD25 only in WT mice. In vitro, IL-33 had a significant survival effect, increasing the total number of splenocytes, including B cells, CD4(+) and CD8(+) T cells, and the frequency of ST2(+) Treg cells. In conclusion, IL-33 acts as a potent immune modulator protecting the liver through activation of ST2(+) Treg cells and control of NK cells.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Hepatitis/immunology , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-33/deficiency , Killer Cells, Natural/immunology , Liver/innervation , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Chemotaxis, Leukocyte , Concanavalin A , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Hepatitis/metabolism , Hepatitis/pathology , Hepatitis/prevention & control , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Killer Cells, Natural/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , T-Lymphocytes, Regulatory/metabolism
8.
Infect Immun ; 83(5): 1738-48, 2015 May.
Article in English | MEDLINE | ID: mdl-25712928

ABSTRACT

Interleukin-33 (IL-33) has now emerged as a cytokine with diverse and pleiotropic functions in various infectious and inflammatory diseases. IL-33 is expressed by epithelial cells, endothelial cells, fibroblasts, and hepatocytes. The target cells of IL-33 are Th2 cells, basophils, dendritic cells, mast cells, macrophages, NKT cells, and nuocytes, newly discovered natural helper cells/innate lymphoid cells bearing the ST2 receptor. IL-33 has dual functions, both as a traditional cytokine and as a nuclear factor that regulates gene transcription. IL-33 functions as an "alarmin" released following cell death, as a biomarker, and as a vaccine adjuvant, with proinflammatory and protective effects during various infections. The exacerbated or protective role of the IL-33/ST2 axis during different infections is dependent upon the organ involved, type of infectious agent, whether the infection is acute or chronic, the invasiveness of the infectious agent, the host immune compartment, and cellular and cytokine microenvironments. In this review, we focus on recent advances in the understanding of the role of the IL-33/ST2 axis in various viral, bacterial, fungal, helminth, and protozoal infectious diseases gained from animal models and studies in human patients. The functional role of IL-33 and ST2 during experimentally induced infections has been summarized by accumulating the data for IL-33- and ST2-deficient mice or for mice exogenously administered IL-33. In summary, exploring the crucial and diverse roles of the IL-33/ST2 axis during infections may help in the development of therapeutic interventions for a wide range of infectious diseases.


Subject(s)
Communicable Diseases/immunology , Interleukins/metabolism , Receptors, Cell Surface/metabolism , Animals , Disease Models, Animal , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Mice, Knockout , Receptors, Interleukin/metabolism
10.
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
11.
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
12.
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
13.
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
14.
Liver Int ; 32(8): 1200-10, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22530772

ABSTRACT

'Alarmins' are a group of proteins or molecules that are released from cells during cellular demise to alert the host immune system. Two of them, Interleukin-33 (IL-33) and high-mobility group box-1 (HMGB1), share many similarities of cellular localization, functions and involvement in various inflammatory pathologies including hepatitis. The expressions of IL-33 and HMGB1, and their receptors ST2 and receptor for advanced glycation end products (RAGE), are substantially up-regulated during acute and chronic hepatitis. Recent data evidence a possible protective role of IL-33/ST2 axis during liver injury. A contrast in expression of IL-33 and HMGB1 alarmins were associated with type of hepatocellular death mediated by immune cells or hepato-toxic agents. The massive release of active form of IL-33 from hepatocytes may affect the recruitment and activation of its ST2-positive target immune cells in the liver to confer its alarmin functions. This review highlights the emerging roles of alarmin proteins in various liver pathologies, by focusing on classical HMGB1 and a newly discovered alarmin, the IL-33.


Subject(s)
Cell Death/immunology , HMGB1 Protein/immunology , Hepatitis, Chronic/immunology , Interleukins/immunology , Liver/immunology , Animals , HMGB1 Protein/metabolism , Hepatitis, Chronic/metabolism , Hepatitis, Chronic/pathology , Humans , Interleukin-33 , Interleukins/metabolism , Liver/metabolism , Liver/pathology
15.
Exp Cell Res ; 317(13): 1922-34, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21570965

ABSTRACT

The nuclear zinc finger protein ZFPIP/Zfp462 is an important factor involved in cell division during the early embryonic development of vertebrates. In pluripotent P19 cells, ZFPIP/Zfp462 takes part in cell proliferation, likely via its role in maintaining chromatin structure. To further define the function of ZFPIP/Zfp462 in the mechanisms of pluripotency and cell differentiation, we constructed a stable P19 cell line in which ZFPIP/Zfp462 knockdown is inducible. We report that ZFPIP/Zfp462 was vital for mitosis and self-renewal in pluripotent P19 cells. Its depletion induced substantial decreases in the expression of the pluripotency genes Nanog, Oct4 and Sox2 and was associated with the transient expression of specific neuronal differentiation markers. We also demonstrated that ZFPIP/Zfp462 expression appears to be unnecessary after neuronal differentiation is induced in P19 cells. Taken together, our results strongly suggest that ZFPIP/Zfp462 is a key chromatin factor involved in maintaining P19 pluripotency and in the early mechanisms of neural differentiation but that it is dispensable in differentiated P19 cells.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins , Mice , Nerve Tissue Proteins/genetics
16.
Nutrients ; 14(2)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35057565

ABSTRACT

The globally prevalent disease, non-alcoholic steatohepatitis (NASH), is characterized by a steatotic and inflammatory liver. In NASH patients, tissue repair mechanisms, activated by the presence of chronic liver damage, lead to the progressive onset of hepatic fibrosis. This scar symptom is a key prognostic risk factor for liver-related morbidity and mortality. Conflicting reports discuss the efficiency of dietary interventions on the reversibility of advanced fibrosis established during NASH. In the present study, the effect of dietary interventions was investigated in the outcome of the fibrosis settled in livers of C57BL/6J mice on a high-fat, high-cholesterol diet (HFHCD) for 5 or 12 consecutive weeks. Various clinico-pathological investigations, including a histological analysis of the liver, measurement of plasma transaminases, steatosis and fibrosis, were performed. To assess the effectiveness of the dietary intervention on established symptoms, diseased mice were returned to a standard diet (SD) for 4 or 12 weeks. This food management resulted in a drastic reduction in steatosis, liver injuries, inflammatory markers, hepatomegaly and oxidative stress and a gradual improvement in the fibrotic state of the liver tissue. In conclusion, our results demonstrated that dietary intervention can partially reverse liver fibrosis induced by HFHCD feeding.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Liver Cirrhosis/diet therapy , Liver Cirrhosis/etiology , Non-alcoholic Fatty Liver Disease/complications , Animals , Cholesterol, Dietary/administration & dosage , Liver/pathology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL
17.
J Mol Med (Berl) ; 100(7): 1027-1038, 2022 07.
Article in English | MEDLINE | ID: mdl-35476028

ABSTRACT

Non-alcoholic steatohepatitis (NASH), a chronic liver disease that emerged in industrialized countries, can further progress into liver fibrosis, cirrhosis, and hepatocellular carcinoma. In the next decade, NASH is predicted to become the leading cause of liver transplantation, the only current interventional therapeutic option. Hepatocyte death, triggered by different death ligands, plays key role in its progression. Previously, we showed that the receptor-interacting protein kinase-1 (RIPK1) in hepatocytes exhibits a protective role in ligand-induced death. Now, to decipher the role of RIPK1 in NASH, Ripk1LPC-KO mice, deficient for RIPK1 only in liver parenchymal cells, and their wild-type littermates (Ripk1fl/fl) were fed for 3, 5, or 12 weeks with high-fat high-cholesterol diet (HFHCD). The main clinical signs of NASH were analyzed to compare the pathophysiological state established in mice. Most of the symptoms evolved similarly whatever the genotype, whether it was the increase in liver to body weight ratio, the steatosis grade or the worsening of liver damage revealed by serum transaminase levels. In parallel, inflammation markers followed the same kinetics with significant equivalent inductions of cytokines (hepatic mRNA levels and blood cytokine concentrations) and a main peak of hepatic infiltration of immune cells at 3 weeks of HFHCD. Despite this identical inflammatory response, more hepatic fibrosis was significantly evidenced at week 12 in Ripk1LPC-KO mice. This coincided with over-induced rates of transcripts of genes implied in fibrosis development (Tgfb1, Tgfbi, Timp1, and Timp2) in Ripk1LPC-KO animals. In conclusion, our results show that RIPK1 in hepatocyte limits the progression of liver fibrosis during NASH.


Subject(s)
Liver Cirrhosis , Non-alcoholic Fatty Liver Disease , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Cytokines/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Hepatocytes/metabolism , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
18.
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
19.
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
20.
J Hepatol ; 52(4): 560-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20207439

ABSTRACT

BACKGROUND & AIMS: Liver resection includes temporal vascular inflow occlusion resulting in ischemia/reperfusion injury in the remnant liver. Here, we developed a rat model of selective lobe occlusion to isolate reperfusion stress from ischemia and to analyze its effect on liver regeneration. METHODS: Left lateral and median lobes of liver were either mobilized or subjected twice for 10min to ischemia followed by 5min reperfusion prior to resection while the regenerative lobes were only subjected to reperfusion. RESULTS: Although intermittent reperfusion stress induced higher levels of serum transaminases, analysis of cell cycle regulators revealed accelerated regenerative response compared to standard partial hepatectomy. The G0/G1 transition occurred before tissue resection, as evidenced by c-fos, junB, and IL-6 induction. Following hepatectomy, Cyclin D1 up-regulation, G1/S transition, and cell division occurred earlier than normal. Unexpectedly, liver mobilization, a component of the clamping procedure, also resulted in earlier G1/S transition. The shortened G1-phase was driven by the c-Jun N-terminal Kinase pathway and was associated with an oxidative stress response as evidenced by the expression of inducible nitric oxide synthase. CONCLUSION: Intermittent selective clamping of lobes to be resected induced reperfusion stress on remnant liver that was beneficial for liver regeneration, suggesting this procedure could be applied in clinical practice.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Liver Regeneration/physiology , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Stress, Physiological/physiology , Animals , Cell Division/physiology , Cyclin D1/genetics , G1 Phase/physiology , Gene Expression/physiology , Heme Oxygenase-1/metabolism , Hepatectomy/methods , Hepatocytes/cytology , Hepatocytes/metabolism , Interleukin-6/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Male , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley , Resting Phase, Cell Cycle/physiology , S Phase/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Superoxide Dismutase/metabolism , Surgical Instruments
SELECTION OF CITATIONS
SEARCH DETAIL