Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
FASEB J ; 34(12): 16163-16178, 2020 12.
Article in English | MEDLINE | ID: mdl-33063394

ABSTRACT

Hepatitis is an important health problem worldwide. Novel molecular targets are in demand for detection and management of hepatitis. Hepatoma-derived growth factor (HDGF) has been delineated to participate in hepatic fibrosis and liver carcinogenesis. However, the relationship between hepatitis and HDGF remains unclear. This study aimed to elucidate the role of HDGF during hepatitis using concanavalin A (ConA)-induced hepatitis model. In cultured hepatocytes, ConA treatment-elicited HDGF upregulation at transcriptional level and promoted HDGF secretion while reducing intracellular HDGF protein level and cellular viability. Similarly, mice receiving ConA administration exhibited reduced hepatic HDGF expression and elevated circulating HDGF level, which was positively correlated with serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. By using HDGF knockout (KO) mice, it was found the ConA-evoked cell death was prominently alleviated in KO compared with control. Besides, it was delineated HDGF ablation conferred protection by suppressing the ConA-induced neutrophils recruitment in livers. Above all, the ConA-mediated activation of tumor necrosis factor-α (TNF-α)/interleukin-1ß (IL-1ß)/interleukin-6 (IL-6)/cyclooxygenase-2 (COX-2) inflammatory signaling was significantly abrogated in KO mice. Treatment with recombinant HDGF (rHDGF) dose-dependently stimulated the expression of TNF-α/IL-1ß/IL-6/COX-2 in hepatocytes, further supporting the pro-inflammatory function of HDGF. Finally, application of HDGF antibody not only attenuated the ConA-mediated inflammatory cascade in hepatocytes, but also ameliorated the ConA-induced hepatic necrosis and AST elevation in mice. In summary, HDGF participates in ConA-induced hepatitis via neutrophils recruitment and may constitute a therapeutic target for acute hepatitis.


Subject(s)
Concanavalin A/pharmacology , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Rats , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
2.
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
3.
Am J Pathol ; 186(3): 524-38, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26762581

ABSTRACT

The interplay between the inflammatory infiltrate and tissue resident cell populations invokes fibrogenesis. However, the temporal and mechanistic contributions of these cells to fibrosis are obscure. To address this issue, liver inflammation, ductular reaction (DR), and fibrosis were induced in C57BL/6 mice by thioacetamide administration for up to 12 weeks. Thioacetamide treatment induced two phases of liver fibrosis. A rapid pericentral inflammatory infiltrate enriched in F4/80(+) monocytes co-localized with SMA(+) myofibroblasts resulted in early collagen deposition, marking the start of an initial fibrotic phase (1 to 6 weeks). An expansion of bone marrow-derived macrophages preceded a second phase, characterized by accelerated progression of fibrosis (>6 weeks) after DR migration from the portal tracts to the centrilobular site of injury, in association with an increase in DR/macrophage interactions. Although chemokine (C-C motif) ligand 2 (CCL2) mRNA was induced rapidly in response to thioacetamide, CCL2 deficiency only partially abrogated fibrosis. In contrast, colony-stimulating factor 1 receptor blockade diminished C-C chemokine receptor type 2 [CCR2(neg) (Ly6C(lo))] monocytes, attenuated the DR, and significantly reduced fibrosis, illustrating the critical role of colony-stimulating factor 1-dependent monocyte/macrophage differentiation and linking the two phases of injury. In response to liver injury, colony-stimulating factor 1 drives early monocyte-mediated myofibroblast activation and collagen deposition, subsequent macrophage differentiation, and their association with the advancing DR, the formation of fibrotic septa, and the progression of liver fibrosis to cirrhosis.


Subject(s)
Hepatitis, Animal/pathology , Liver Cirrhosis, Experimental/pathology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Animals , Chemokines/genetics , Chemokines/metabolism , Collagen Type I/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation , Hepatitis, Animal/genetics , Hepatitis, Animal/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , Thioacetamide
4.
Hepatology ; 64(2): 508-21, 2016 08.
Article in English | MEDLINE | ID: mdl-26991125

ABSTRACT

UNLABELLED: Death receptor (DR) ligands such as tumor necrosis factor (TNF) have been identified as fundamental mediators of liver damage both in mouse models and in humans. While the essential site of function of DR signaling is conceivably the hepatocyte, a systematic analysis is missing. Using mice with conditional gene ablation, we analyzed the tissue-specific function of DR signaling in T cell-dependent (concanavalin A) and independent (lipopolysaccharide/galactosamine) hepatitis and in models of bacterial infection (Listeria monocytogenes, lipopolysaccharide). We report that lipopolysaccharide/galactosamine-induced liver injury depends on hepatocyte-intrinsic TNF receptor 1 (p55, TNFR1). In contrast, we show that T cell-induced hepatitis was independent of TNFR1 signaling in hepatocytes, T cells, or endothelial cells. Moreover, T cell-induced hepatitis was independent of hepatocyte-intrinsic Fas-associated protein with death domain, TNF-related apoptosis-inducing ligand receptor, or Fas signaling. Instead, concanavalin A-induced hepatitis was completely prevented in mice with myeloid-derived cell (MDC)-specific deletion of TNFR1. Significantly, however, mice lacking TNFR1 in MDCs succumbed to listeria infection, although they displayed similar sensitivity toward endotoxin-induced septic shock when compared to control mice. These results suggest that TNFR1 signaling in MDCs is a critical mediator of both the detrimental and the protective functions of TNF in T cell-induced hepatitis and bacterial infection, respectively. CONCLUSION: The critical site of action of DRs is completely dependent on the nature of hepatitis; the data specify MDCs as the essential cell type of TNFR1 function in T cell-mediated hepatitis and in the response to listeria, thereby identifying the opposing role of MDC TNFR1 in autoimmunity and bacterial infection. (Hepatology 2016;64:508-521).


Subject(s)
Hepatitis, Animal/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Concanavalin A , Endothelial Cells/metabolism , Hepatitis, Animal/immunology , Hepatitis, Animal/microbiology , Lipopolysaccharides , Listeria monocytogenes , Liver/metabolism , Male , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/metabolism , T-Lymphocytes/physiology
5.
Scand J Immunol ; 86(3): 156-164, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29072876

ABSTRACT

An autoimmune background is suspected for Doberman hepatitis (DH). It is based on the finding of mononuclear cell infiltrates in the liver, strong female bias, association to the homozygous risk factor dog leucocyte antigen (DLA) allele DRB1*00601 and aberrant major histocompatibility complex (MHC) class II expression on hepatocytes that correlates with the degree of inflammation in the liver. The aim of this study was to search for autoantibodies against liver-related antigens associated with DH. Twenty-five Dobermans with subclinical DH (SDH), 13 that clinically manifest DH (CDH) and 17 healthy controls were studied. Immunoblotting analysis detected specific antibodies in the DH sera. By mass spectrometry the targets were identified as liver-related enzymes glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and alcohol dehydrogenase (ADH). Using ELISA, anti-GAPDH IgG was detected in 36% (9/25) of SDH dogs and 69.2% (9/13) of the CDH dogs compared to healthy controls (0/17) (P < 0.0005). Anti-ADH IgG was detected in 72% (18/25) of SDH dogs and 76.9% (10/13) of CDH dogs and only in one (1/17) control (P < 0.0005). The finding of novel autoantigens, GAPDH and ADH strengthen the hypothesis that DH is an autoimmune disease of the liver. These findings suggest that DH could be diagnosed by screening for autoantibodies against the defined antigens.


Subject(s)
Alcohol Dehydrogenase/immunology , Glyceraldehyde 3-Phosphate/immunology , Hepatitis, Animal/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/immunology , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Immunoblotting , Male , Proteome , Proteomics/methods
6.
Molecules ; 22(8)2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28786950

ABSTRACT

Purple sweet potato color (PSPC), a class of naturally occurring anthocyanins, exhibits beneficial effects on metabolic syndrome. Sustained inflammation plays a crucial role in the pathogenesis of metabolic syndrome. Here we explored the effects of PSPC on high-fat diet (HFD)-induced hepatic inflammation and the mechanisms underlying these effects. Mice were divided into four groups: Control group, HFD group, HFD + PSPC group, and PSPC group. PSPC was administered by daily oral gavage at doses of 700 mg/kg/day for 20 weeks. Nicotinamide riboside (NR) was used to increase NAD⁺ levels. Our results showed that PSPC effectively ameliorated obesity and liver injuries in HFD-fed mice. Moreover, PSPC notably blocked hepatic oxidative stress in HFD-treated mice. Furthermore, PSPC dramatically restored NAD⁺ level to abate endoplasmic reticulum stress (ER stress) in HFD-treated mouse livers, which was confirmed by NR treatment. Consequently, PSPC remarkably suppressed the nuclear factor-κB (NF-κB) p65 nuclear translocation and nucleotide oligomerization domain protein1/2 (NOD1/2) signaling in HFD-treated mouse livers. Thereby, PSPC markedly diminished the NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation, ultimately lowering the expressions of inflammation-related genes in HFD-treated mouse livers. In summary, PSPC protected against HFD-induced hepatic inflammation by boosting NAD⁺ level to inhibit NLRP3 inflammasome activation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hepatitis, Animal/drug therapy , Hepatitis, Animal/metabolism , Inflammasomes/metabolism , Ipomoea batatas/chemistry , NAD/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pigments, Biological/pharmacology , Plant Extracts/pharmacology , Animals , Anthocyanins/chemistry , Anthocyanins/pharmacology , Anti-Inflammatory Agents/chemistry , Diet, High-Fat , Endoplasmic Reticulum Stress , Gene Expression Regulation/drug effects , Hepatitis, Animal/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , NF-kappa B/metabolism , Nod Signaling Adaptor Proteins/genetics , Nod Signaling Adaptor Proteins/metabolism , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Oxidative Stress/drug effects , Pigments, Biological/chemistry , Plant Extracts/chemistry , Protein Transport
7.
Pharm Biol ; 55(1): 198-205, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27927057

ABSTRACT

CONTEXT: Duck virus hepatitis (DVH) caused by duck hepatitis A virus type 1 (DHAV-1) is an acute and lethal disease of young ducklings. However, there is still no effective drug to treat DVH. OBJECTIVE: This study assessed the curative effect on DVH of a flavonoid prescription baicalin-linarin-icariin-notoginsenoside R1 (BLIN) as well as the hepatoprotective and antioxidative effects of BLIN. MATERIALS AND METHODS: MTT method was used to test the anti-DHAV-1 ability of BLIN in vitro. We then treated ducklings by BLIN (3 mg per duckling, once a day for 5 days) to evaluate the in vivo efficacy. To study the hepatoprotective and antioxidative roles of BLIN in its curative effect on DVH, we investigated the hepatic injury evaluation biomarkers and the oxidative stress evaluation indices of the ducklings. RESULTS: On duck embryonic hepatocytes, DHAV-1 inhibitory rate of BLIN at 20 µg/mL was 69.3%. The survival rate of ducklings treated by BLIN was about 35.5%, which was significantly higher than that of virus control (0.0%). After the treatment of BLIN, both the hepatic injury and the oxidative stress of infected ducklings alleviated. At the same time, a significant positive correlation (p < 0.05) existed between the hepatic injury indices and the oxidative stress indices. CONCLUSIONS: BLIN showed a significant curative effect on DVH. The antioxidative and hepatoprotective effects of BLIN made great contributions to the treatment of DVH. Furthermore, BLIN is expected to be exploited as a new drug for the clinical treatment of DVH.


Subject(s)
Antioxidants/pharmacology , Antiviral Agents/pharmacology , Ducks , Flavonoids/pharmacology , Hepatitis Virus, Duck/drug effects , Hepatitis, Animal/drug therapy , Hepatocytes/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Poultry Diseases/drug therapy , Animals , Animals, Newborn , Biomarkers/metabolism , Cells, Cultured , Drug Combinations , Ginsenosides/pharmacology , Glycosides/pharmacology , Hepatitis Virus, Duck/pathogenicity , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Hepatitis, Animal/virology , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Liver/metabolism , Liver/pathology , Liver/virology , Poultry Diseases/metabolism , Poultry Diseases/pathology , Poultry Diseases/virology , Time Factors
8.
J Infect Dis ; 213(3): 361-9, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26238685

ABSTRACT

Disruption of the mucosal epithelium during lentivirus infections permits translocation of microbial products into circulation, causing immune activation and driving disease. Although the liver directly filters blood from the intestine and is the first line of defense against gut-derived antigens, the effects of microbial products on the liver are unclear. In livers of normal macaques, minute levels of bacterial products were detectable, but increased 20-fold in simian immunodeficiency virus (SIV)-infected animals. Increased microbial products in the liver induced production of the chemoattractant CXCL16 by myeloid dendritic cells (mDCs), causing subsequent recruitment of hypercytotoxic natural killer (NK) cells expressing the CXCL16 receptor, CXCR6. Microbial accumulation, mDC activation, and cytotoxic NK cell frequencies were significantly correlated with markers of liver damage, and SIV-infected animals consistently had evidence of hepatitis and fibrosis. Collectively, these data indicate that SIV-associated accumulation of microbial products in the liver initiates a cascade of innate immune activation, resulting in liver damage.


Subject(s)
Killer Cells, Natural/physiology , Liver/metabolism , Myeloid Cells/physiology , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus , Animals , Apoptosis , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Gene Expression Regulation , Hepatitis, Animal/etiology , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Liver/microbiology , Macaca mulatta , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/microbiology
9.
Development ; 140(3): 639-48, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23293295

ABSTRACT

Congenital biliary atresia is an incurable disease of newborn infants, of unknown genetic causes, that results in congenital deformation of the gallbladder and biliary duct system. Here, we show that during mouse organogenesis, insufficient SOX17 expression in the gallbladder and bile duct epithelia results in congenital biliary atresia and subsequent acute 'embryonic hepatitis', leading to perinatal death in ~95% of the Sox17 heterozygote neonates in C57BL/6 (B6) background mice. During gallbladder and bile duct development, Sox17 was expressed at the distal edge of the gallbladder primordium. In the Sox17(+/-) B6 embryos, gallbladder epithelia were hypoplastic, and some were detached from the luminal wall, leading to bile duct stenosis or atresia. The shredding of the gallbladder epithelia is probably caused by cell-autonomous defects in proliferation and maintenance of the Sox17(+/-) gallbladder/bile duct epithelia. Our results suggest that Sox17 plays a dosage-dependent function in the morphogenesis and maturation of gallbladder and bile duct epithelia during the late-organogenic stages, highlighting a novel entry point to the understanding of the etiology and pathogenesis of human congenital biliary atresia.


Subject(s)
Biliary Atresia/genetics , Gene Expression Regulation, Developmental , HMGB Proteins/metabolism , Haploinsufficiency , SOXF Transcription Factors/metabolism , Animals , Animals, Newborn , Bile Ducts/metabolism , Bile Ducts/pathology , Biliary Atresia/pathology , Cell Proliferation , Cholestasis/genetics , Cholestasis/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endoplasmic Reticulum Stress , Epithelium/metabolism , Epithelium/pathology , Female , Gallbladder/metabolism , Gallbladder/ultrastructure , HMGB Proteins/genetics , Hepatitis, Animal/genetics , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Hepatocytes/metabolism , Heterozygote , Immunohistochemistry , Liver/metabolism , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pregnancy , SOXF Transcription Factors/genetics , Time Factors
10.
J Biol Chem ; 289(18): 12457-66, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24634219

ABSTRACT

Overexpression of tumor necrosis factor α (TNFα) is a hallmark of many inflammatory diseases, including rheumatoid arthritis, inflammatory bowel disease, and septic shock and hepatitis, making it a potential therapeutic target for clinical interventions. To explore chemical inhibitors against TNFα activity, we applied computer-aided drug design combined with in vitro and cell-based assays and identified a lead chemical compound, (E)-4-(2-(4-chloro-3-nitrophenyl) (named as C87 thereafter), which directly binds to TNFα, potently inhibits TNFα-induced cytotoxicity (IC50 = 8.73 µM) and effectively blocks TNFα-triggered signaling activities. Furthermore, by using a murine acute hepatitis model, we showed that C87 attenuates TNFα-induced inflammation, thereby markedly reducing injuries to the liver and improving animal survival. Thus, our results lead to a novel and highly specific small-molecule TNFα inhibitor, which can be potentially used to treat TNFα-mediated inflammatory diseases.


Subject(s)
Hepatitis, Animal/prevention & control , Inflammation/prevention & control , Small Molecule Libraries/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , HL-60 Cells , Hepatitis, Animal/genetics , Hepatitis, Animal/metabolism , Humans , Hydrazones/chemistry , Hydrazones/pharmacology , Inflammation/genetics , Inflammation/metabolism , Jurkat Cells , K562 Cells , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred BALB C , Molecular Structure , Protein Binding/drug effects , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance , Thiazoles/chemistry , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Biochem Biophys Res Commun ; 462(3): 245-50, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25964086

ABSTRACT

Juglone as a natural production mainly extracted from green walnut husks of Juglans mandshurica has been defined as the functional composition among a series of compounds. It showed powerful protective effect in various diseases by inhibiting inflammation and tumor cells growth. However, studies on its anti-inflammatory effect based on high-fat diet-induced hepatitis and neuroinflammation are still not available. In this regard, we first investigated whether juglone suppresses high-fat diet-stimulated liver injury, hypothalamus inflammation and underlying mechanisms by which they may recover them. SD rats were orally treated with or without high-fat diet, 0.25 mg/kg or 1 mg/kg juglone for 70 days. Subsequently, blood, hypothalamus and liver tissue were collected for different analysis. Also, the primary astrocytes were isolated and used to analyze the inhibitory effect of juglone in vitro. Analysis of inflammatory cytokines declared that the inhibition of TNF-α, IL-1ß and IL-6 could be carried by juglone in response to high-fat diet rats. Meanwhile, TLR4 expression and NF-kappa activity also have been confirmed to be the key link in the development of hepatitis and nerve inflammation. The activation was significantly suppressed in treatment group as compared with model. These results indicated that juglone prevents high-fat diet-induced liver injury and nerve inflammation in mice through inhibition of inflammatory cytokine secretion, NF-kappa B activation and endotoxin production.


Subject(s)
Endotoxemia/complications , Hepatitis, Animal/prevention & control , Inflammation/prevention & control , NF-kappa B/metabolism , Naphthoquinones/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Diet, High-Fat/adverse effects , Endotoxemia/metabolism , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Nervous System Diseases/metabolism , Nervous System Diseases/prevention & control , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
13.
Hepatology ; 60(5): 1741-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24723460

ABSTRACT

UNLABELLED: Liver eosinophilia has been associated with incidences of drug-induced liver injury (DILI) for more than 50 years, although its role in this disease has remained largely unknown. In this regard, it was recently shown that eosinophils played a pathogenic role in a mouse model of halothane-induced liver injury (HILI). However, the signaling events that drove hepatic expression of eosinophil-associated chemokines, eotaxins, eosinophil infiltration, and subsequent HILI were unclear. We now provide evidence implicating hepatic epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) and type 2 immunity, in particular, interleukin-4 (IL-4) production, in mediating hepatic eosinophilia and injury during HILI. TSLP was constitutively expressed by mouse hepatocytes and increased during HILI. Moreover, the severity of HILI was reduced in mice deficient in either the TSLP receptor (TSLPR) or IL-4 and was accompanied by decreases in serum levels of eotaxins and hepatic eosinophilia. Similarly, concanavalin A-induced liver injury, where type 2 cytokines and eosinophils play a significant role in its pathogenesis, was also reduced in TSLPR-deficient mice. Studies in vitro revealed that mouse and human hepatocytes produce TSLP and eotaxins in response to treatment with combinations of IL-4 and proinflammatory cytokines IL-1ß and tumor necrosis factor alpha. CONCLUSION: This report provides the first evidence implicating roles for hepatic TSLP signaling, type 2 immunity, and eosinophilia in mediating liver injury caused by a drug.


Subject(s)
Anesthetics, Inhalation/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Cytokines/metabolism , Halothane/adverse effects , Interleukin-4/metabolism , Animals , Concanavalin A , Female , Hepatitis, Animal/metabolism , Hepatocytes/metabolism , Humans , Mice, Inbred BALB C , Thymic Stromal Lymphopoietin
14.
J Immunol ; 191(1): 480-7, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740948

ABSTRACT

An emerging body of evidence suggests a pivotal role of CD3(+) T cells in mediating early ischemia reperfusion injury (IRI). However, the precise phenotype of T cells involved and the mechanisms underlying such T cell-mediated immune responses in IRI, as well as their clinical relevance, are poorly understood. In this study, we investigated early immunological events in a model of partial warm hepatic IRI in genetically targeted mice to study the precise pathomechanistic role of RORγt(+) T cells. We found that unconventional CD27(-)γδTCR(+) and CD4(-)CD8(-) double-negative T cells are the major RORγt-expressing effector cells in hepatic IRI that play a mechanistic role by being the main source of IRI-mediating IL-17A. We further show that unconventional IRI-mediating T cells are contingent on RORγt, as highlighted by the fact that a genetic deficiency for RORγt, or its therapeutic antagonization via digoxin, is protective against hepatic IRI. Therefore, identification of CD27(-)γδTCR(+) and CD4(-)CD8(-) double-negative T cells as the major source of IL-17A via RORγt in hepatic IRI opens new therapeutic options to improve liver transplantation outcomes.


Subject(s)
Hepatitis, Animal/immunology , Hepatitis, Animal/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Animals , Disease Models, Animal , Genes, Reporter , Hepatitis, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Reperfusion Injury/metabolism , T-Lymphocyte Subsets/metabolism , Time Factors
15.
J Immunol ; 191(4): 1835-44, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23842754

ABSTRACT

Liver fibrosis is a severe, life-threatening clinical condition resulting from nonresolving hepatitis of different origins. IL-17A is critical in inflammation, but its relation to liver fibrosis remains elusive. We find increased IL-17A expression in fibrotic livers from HBV-infected patients undergoing partial hepatectomy because of cirrhosis-related early-stage hepatocellular carcinoma in comparison with control nonfibrotic livers from uninfected patients with hepatic hemangioma. In fibrotic livers, IL-17A immunoreactivity localizes to the inflammatory infiltrate. In experimental carbon tetrachloride-induced liver fibrosis of IL-17RA-deficient mice, we observe reduced neutrophil influx, proinflammatory cytokines, hepatocellular necrosis, inflammation, and fibrosis as compared with control C57BL/6 mice. IL-17A is produced by neutrophils and T lymphocytes expressing the Th17 lineage-specific transcription factor Retinoic acid receptor-related orphan receptor γt. Furthermore, hepatic stellate cells (HSCs) isolated from naive C57BL/6 mice respond to IL-17A with increased IL-6, α-smooth muscle actin, collagen, and TGF-ß mRNA expression, suggesting an IL-17A-driven fibrotic process. Pharmacologic ERK1/2 or p38 inhibition significantly attenuated IL-17A-induced HSC activation and collagen expression. In conclusion, IL-17A(+) Retinoic acid receptor-related orphan receptor γt(+) neutrophils and T cells are recruited into the injured liver driving a chronic, fibrotic hepatitis. IL-17A-dependent HSC activation may be critical for liver fibrosis. Thus, blockade of IL-17A could potentially benefit patients with chronic hepatitis and liver fibrosis.


Subject(s)
Hepatic Stellate Cells/metabolism , Interleukin-17/physiology , Liver Cirrhosis/etiology , Actins/biosynthesis , Actins/genetics , Adult , Animals , Carbon Tetrachloride Poisoning/complications , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/surgery , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Collagen/biosynthesis , Collagen/genetics , Cytokines/biosynthesis , Cytokines/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hemangioma/chemistry , Hemangioma/surgery , Hepatectomy , Hepatic Stellate Cells/pathology , Hepatitis B, Chronic/complications , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Humans , Interleukin-17/analysis , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukin-17/pharmacology , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/chemistry , Liver Neoplasms/surgery , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Middle Aged , Neutrophils/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Protein Kinase Inhibitors/pharmacology , Receptors, Interleukin-17/deficiency , Recombinant Proteins/pharmacology , Th17 Cells/immunology , Th17 Cells/metabolism , Transforming Growth Factor beta/biosynthesis
16.
J Lipid Res ; 55(3): 421-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24347528

ABSTRACT

Obesity-induced liver inflammation can drive insulin resistance. HDL has anti-inflammatory properties, so we hypothesized that low levels of HDL would perpetuate inflammatory responses in the liver and that HDL treatment would suppress liver inflammation and insulin resistance. The aim of this study was to investigate the effects of lipid-free apoAI on hepatic inflammation and insulin resistance in mice. We also investigated apoAI as a component of reconstituted HDLs (rHDLs) in hepatocytes to confirm results we observed in vivo. To test our hypothesis, C57BL/6 mice were fed a high-fat diet (HFD) for 16 weeks and administered either saline or lipid-free apoAI. Injections of lipid-free apoAI twice a week for 2 or 4 weeks with lipid-free apoAI resulted in: i) improved insulin sensitivity associated with decreased systemic and hepatic inflammation; ii) suppression of hepatic mRNA expression for key transcriptional regulators of lipogenic gene expression; and iii) suppression of nuclear factor κB (NF-κB) activation. Human hepatoma HuH-7 cells exposed to rHDLs showed suppressed TNFα-induced NF-κB activation, correlating with decreased NF-κB target gene expression. We conclude that apoAI suppresses liver inflammation in HFD mice and improves insulin resistance via a mechanism that involves a downregulation of NF-κB activation.


Subject(s)
Hepatitis, Animal/prevention & control , Insulin Resistance , Lipoproteins, HDL/pharmacology , Liver/drug effects , Animals , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/pharmacology , Blood Glucose/metabolism , Cell Line, Tumor , Diet, High-Fat , Gene Expression/drug effects , Glucose/metabolism , Glucose Tolerance Test , Hepatitis, Animal/genetics , Hepatitis, Animal/metabolism , Humans , Insulin/blood , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-6/blood , Interleukin-6/genetics , Lipoproteins, HDL/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
17.
Hepatology ; 57(1): 277-88, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22806577

ABSTRACT

UNLABELLED: NKG2D activation plays an important role in initiating and maintaining liver inflammation, and blockade of NKG2D recognition becomes a promising approach to alleviate liver inflammation. Treatment by silencing NKG2D ligands on hepatocytes, but not NKG2D on circulating immune cells, is more liver-specific, and simultaneous knockdown of multiple NKG2D ligands on hepatocytes will be more efficient in liver disease intervention. Here, we constructed a single vector that could simultaneously express multiple short hairpin RNAs (shRNAs) against all murine NKG2D ligands including Rae1, Mult1, and H60. After hydrodynamic injection of plasmid containing the three shRNA sequences (shRae1-shMult1-shH60), also called pRNAT-shRMH, we found the expression of all three NKG2D ligands on hepatocytes was downregulated both on messenger RNA and protein levels. Moreover, natural killer (NK) cell-mediated NKG2D-dependent fulminant hepatitis of the mice was alleviated, along with inactivation of hepatic NK cells, by pRNAT-shRMH if compared with its counterpart RNA interference vectors against single or double ligands. The therapeutic efficacy of pRNAT-shRMH was equivalent to that of injecting three monoclonal antibodies against Rae1, Mult1, and H60. For better in vivo application, we constructed a recombinant adenovirus containing pRNAT-shRMH (called Ad-RMH) with efficient hepatotropic infection capacity and observed that Ad-RMH intravenous injection exerted a similar therapeutic efficiency as plasmid pRNAT-shRMH hydrodynamic injection. Noticeably, simultaneous knockdown of multiple human NKG2D ligands (MICA/B, ULBP2, and ULBP3) also significantly attenuated NK cell cytolysis against human NKG2D ligand-positive hepatocyte L-02 cells, suggesting a possible translation into human settings. CONCLUSION: Simultaneous knockdown of multiple ligands of NKG2D prevents NK cell-mediated fulminant hepatitis and is a potential therapeutic approach to treat liver diseases.


Subject(s)
Hepatitis, Animal/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Animals , Gene Knockdown Techniques , Genetic Vectors , Hepatitis, Animal/metabolism , Hepatocytes/immunology , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , NK Cell Lectin-Like Receptor Subfamily K/genetics , RNA Interference , RNA, Small Interfering
18.
Hepatology ; 57(1): 373-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22821628

ABSTRACT

UNLABELLED: Acetaminophen overdose causes acute liver inflammation with neutrophil infiltration; however, the mechanism of damage-associated inflammation has not been elucidated. In this study we found that the HMGB1-TLR4-IL-23-IL-17A axis played a crucial role in acetaminophen-induced infiltration of neutrophils and liver injury. Notably, interleukin (IL)-17A and IL-23 significantly increased after acetaminophen challenge. A neutralizing antibody against IL-17A attenuated the recruitment of neutrophils, accompanied by reduced liver injury. Only IL-17A(+) CD3(+) γδ T cell receptor (TCR)(+) cells were significantly increased in the liver, and depletion of γδ T cells, but not CD4(+) T cells or natural killer (NK)T cells significantly reduced IL-17A production, attenuated liver injury, and decreased the number of neutrophils in the liver. Furthermore, a neutralizing IL-23 p19 antibody or p40-deficiency significantly decreased the levels of IL-17A and infiltration of neutrophils. After in vitro stimulation, the percentage of IL-17A-producing γδ T cells and the levels of supernatant IL-17A from total hepatic lymphocytes or purified γδ T cells markedly increased in the presence with IL-23. Importantly, IL-23 and IL-17A were reduced after inhibition of macrophages and could not be induced in Toll-like receptor TLR4(-/-) mice after acetaminophen challenge. Meanwhile, serum high-mobility group box 1 (HMGB1), a damage-associated molecule released from necrotic hepatocytes, increased after acetaminophen challenge, and the HMGB1 inhibitor glycyrrhizin markedly reduced the production of IL-23 and IL-17A and the recruitment of hepatic neutrophils. HMGB1 stimulated the production of IL-23 by TLR4(+/+) but not by TLR4(-/-) macrophages. CONCLUSION: The HMGB1-TLR4-IL-23 pathway in macrophages makes the generation of IL-17-producing γδ T cells, which mediates neutrophil infiltration and damage-induced liver inflammation.


Subject(s)
HMGB1 Protein/blood , Hepatitis, Animal/immunology , Macrophages/metabolism , Neutrophil Infiltration , T-Lymphocytes/metabolism , Acetaminophen , Analgesics, Non-Narcotic , Animals , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Hepatocytes/metabolism , Interleukin-17/metabolism , Interleukin-23/metabolism , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/metabolism
19.
Hepatology ; 57(1): 228-38, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22821642

ABSTRACT

UNLABELLED: Negative feedback immune mechanisms are essential for maintenance of hepatic homeostasis and prevention of immune-mediated liver injury. We show here that scavenger receptor A (SRA/CD204), a pattern recognition molecule, is highly up-regulated in the livers of patients with autoimmune or viral hepatitis, and of mice during concanavalin A (Con A)-induced hepatitis (CIH). Strikingly, genetic SRA ablation strongly sensitizes mice to Con A-induced liver injury. SRA loss, increased mortality and liver pathology correlate with excessive production of IFN-γ and heightened activation of T cells. Increased liver expression of SRA primarily occurs in mobilized hepatic myeloid cells during CIH, including CD11b(+) Gr-1(+) cells. Mechanistic studies establish that SRA on these cells functions as a negative regulator limiting T-cell activity and cytokine production. SRA-mediated protection from CIH is further validated by adoptive transfer of SRA(+) hepatic mononuclear cells or administration of a lentivirus-expressing SRA, which effectively ameliorates Con A-induced hepatic injury. Also, CIH and clinical hepatitis are associated with increased levels of soluble SRA. This soluble SRA displays a direct T-cell inhibitory effect and is capable of mitigating Con A-induced liver pathology. CONCLUSION: Our findings demonstrate an unexpected role of SRA in attenuation of Con A-induced, T-cell-mediated hepatic injury. We propose that SRA serves as an important negative feedback mechanism in liver immune homeostasis, and may be exploited for therapeutic treatment of inflammatory liver diseases.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Concanavalin A/toxicity , Hepatitis, Animal/immunology , Scavenger Receptors, Class A/metabolism , T-Lymphocytes/physiology , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Hepatitis, Animal/metabolism , Interferon-gamma/metabolism , Liver/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Hepatology ; 57(1): 362-72, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22936459

ABSTRACT

UNLABELLED: Concanavalin A (Con A) treatment induces severe hepatitis in mice in a manner dependent on T cells, interferon (IFN)-gamma, and tumor necrosis factor (TNF). Treatment with the anticoagulant heparin protects against hepatitis, despite healthy production of IFN-γ and TNF. Here, we investigated molecular and cellular mechanisms for hypercoagulation-mediated hepatitis. After Con A challenge, liver of wild-type (WT) mice showed prompt induction of Ifnγ and Tnf, followed by messenger RNA expression of tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1), which initiate blood coagulation and inhibit clot lysis, respectively. Mice developed dense intrahepatic fibrin deposition and massive liver necrosis. In contrast, Ifnγ(-/-) mice and Ifnγ(-/-) Tnf(-/-) mice neither induced Pai1 or Tf nor developed hepatitis. In WT mice TF blockade with an anti-TF monoclonal antibody protected against Con A-induced hepatitis, whereas Pai1(-/-) mice were not protected. Both hepatic macrophages and sinusoidal endothelial cells (ECs) expressed Tf after Con A challenge. Macrophage-depleted WT mice reconstituted with hematopoietic cells, including macrophages deficient in signal transducer and activator of transcription-1 (STAT1) essential for IFN-γ signaling, exhibited substantial reduction of hepatic Tf and of liver injuries. This was also true for macrophage-depleted Stat1(-/-) mice reconstituted with WT macrophages. Exogenous IFN-γ and TNF rendered T-cell-null, Con A-resistant mice deficient in recombination-activating gene 2, highly susceptible to Con A-induced liver injury involving TF. CONCLUSIONS: Collectively, these results strongly suggest that proinflammatory signals elicited by IFN-γ, TNF, and Con A in both hepatic macrophages and sinusoidal ECs are necessary and sufficient for the development of hypercoagulation-mediated hepatitis.


Subject(s)
Hepatitis, Animal/etiology , Interferon-gamma , Liver/metabolism , Thrombophilia/complications , Tumor Necrosis Factor-alpha/metabolism , Animals , Concanavalin A , Endothelial Cells/metabolism , Fibrin/metabolism , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Interferon-gamma/metabolism , Liver/immunology , Liver/pathology , Macrophages/metabolism , Mice , Mice, Knockout , Mitogens , Necrosis , Plasminogen Activator Inhibitor 1/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes/physiology , Thrombophilia/chemically induced , Thromboplastin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL