Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Clin Liver Dis ; 26(2): 165-179, 2022 05.
Article in English | MEDLINE | ID: mdl-35487603

ABSTRACT

Hepatorenal syndrome (HRS) is defined as a functional renal failure without major histologic changes in individuals with severe liver disease and it is associated with a high mortality rate. Renal hypoperfusion due to marked vasoconstriction as a result of complex circulatory dysfunction has been suggested to be the cornerstone of HRS. Splanchnic and peripheral arterial vasodilation and cirrhotic cardiomyopathy result in effective arterial hypovolemia and compensatory activation of vasoconstrictor mechanisms. The efficacy of current therapeutic strategies targeting this circulatory dysfunction is limited. Increasing evidence suggests a substantial role of systemic inflammation in HRS via either vascular or direct renal effects. Here we summarize the current understanding of HRS pathophysiology.


Subject(s)
Hepatorenal Syndrome , Female , Hepatorenal Syndrome/etiology , Hepatorenal Syndrome/therapy , Humans , Liver Cirrhosis/complications , Male , Vasoconstrictor Agents/pharmacology , Vasoconstrictor Agents/therapeutic use , Vasodilation/physiology
2.
Lab Invest ; 101(12): 1540-1549, 2021 12.
Article in English | MEDLINE | ID: mdl-34453120

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease globally. miRNAs (miRs) regulate various cellular events that lead to NAFLD. In this study we tested the hypothesis that miR-155 is an important regulator of steatohepatitis and fibrosis pathways. Wild type (WT) or miR-155 deficient (KO) mice received a high fat-high cholesterol-high sugar-diet (HF-HC-HS) for 34 weeks and liver tissues were analyzed. In patients with nonalcoholic steatohepatitis and in the mouse model of HF-HC-HS diet we found increased miR-155 levels in the liver compared to normal livers. Upon HF-HC-HS diet feeding, miR-155 KO mice displayed less liver injury, decreased steatosis, and attenuation in fibrosis compared to WT mice. ALT, triglyceride levels, and genes involved in fatty acid metabolic pathway were increased in WT mice whereas miR-155 KO mice showed attenuation in these parameters. HF-HC-HS diet-induced significant increase in the expression of NLRP3 inflammasome components in the livers of WT mice compared to chow fed diet. Compared to WT mice, miR-155 KO showed attenuated induction in the NLRP3, ASC, and caspase1 inflammasome expression on HF-HC-HS diet. Fibrosis markers such as collagen content and deposition, αSMA, Zeb2, and vimentin were all increased in WT mice and miR-155 KO mice showed attenuated fibrosis marker expression. Overall, our findings highlight a role for miR-155 in HF-HC-HS diet-induced steatosis and liver fibrosis.


Subject(s)
Inflammasomes/metabolism , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Diet, High-Fat , Dietary Sugars , Fibrosis , Gene Expression Regulation , Lipid Metabolism , Liver/pathology , Male , Mice, Knockout , Non-alcoholic Fatty Liver Disease/pathology
3.
J Leukoc Biol ; 102(2): 487-498, 2017 08.
Article in English | MEDLINE | ID: mdl-28584078

ABSTRACT

Inflammation promotes the progression of alcoholic liver disease. Alcohol sensitizes KCs to gut-derived endotoxin (LPS); however, signaling pathways that perpetuate inflammation in alcoholic liver disease are only partially understood. We found that chronic alcohol feeding in mice induced miR-155, an inflammatory miRNA in isolated KCs. We hypothesized that miR-155 might increase the responsiveness of KCs to LPS via targeting the negative regulators of LPS signaling. Our results revealed that KCs that were isolated from alcohol-fed mice showed a decrease in IRAK-M, SHIP1, and PU.1, and an increase in TNF-α levels. This was specific to KCs, as no significant differences were observed in these genes in hepatocytes. We found a causal effect of miR-155 deficiency on LPS responsiveness, as KCs that were isolated from miR-155 KO mice showed a greater induction of IRAK-M, SHIP1, and suppressor of cytokine signaling 1 after LPS treatment. C/EBPß, a validated miR-155 target, stimulates IL-10 transcription. We found a higher induction of C/EBPß and IL-10 in KCs that were isolated from miR-155 KO mice after LPS treatment. Gain- and loss-of-function studies affirmed that alcohol-induced miR-155 directly regulates IRAK-M, SHIP1, suppressor of cytokine signaling 1, and C/EBPß, as miR-155 inhibition increased and miR-155 overexpression decreased these genes in LPS or alcohol-pretreated wild-type KCs. HDAC11, a regulator of IL-10, was significantly increased and IL-10 was decreased in KCs that were isolated from alcohol-fed mice. Functionally, knockdown of HDAC11 with small interfering RNA resulted in an IL-10 increase in LPS or alcohol-pretreated Mϕ. We found that acetaldehyde and NF-κB pathways regulate HDAC11 levels. Collectively, our results indicate that the alcohol-induced responsiveness of KCs to LPS, in part, is governed by miR-155 and HDAC11.


Subject(s)
Gene Expression Regulation/immunology , Histone Deacetylases/metabolism , Kupffer Cells/metabolism , Liver Diseases, Alcoholic/metabolism , MicroRNAs/metabolism , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Gene Knockdown Techniques , Inflammation/immunology , Inflammation/metabolism , Kupffer Cells/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Liver Diseases, Alcoholic/immunology , Liver Diseases, Alcoholic/pathology , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
4.
Liver Int ; 37(7): 968-973, 2017 07.
Article in English | MEDLINE | ID: mdl-28345165

ABSTRACT

BACKGROUND & AIMS: Inflammation and impaired hepatocyte regeneration contribute to liver failure in alcoholic hepatitis (AH). Interleukin (IL)-1 is a key inflammatory cytokine in the pathobiology of AH. The role of IL-1 in liver regeneration in the recovery phase of alcohol-induced liver injury is unknown. METHODS: In this study, we tested IL-1 receptor antagonist to block IL-1 signalling in a mouse model of acute-on-chronic liver injury on liver inflammation and hepatocyte regeneration in AH. RESULTS: We observed that inhibition of IL-1 signalling decreased liver inflammation and neutrophil infiltration, and resulted in enhanced regeneration of hepatocytes and increased rate of recovery from liver injury in AH. CONCLUSION: Our novel findings suggest that IL-1 drives sustained liver inflammation and impaired hepatocyte regeneration even after cessation of ethanol exposure.


Subject(s)
Acute-On-Chronic Liver Failure/drug therapy , Cell Proliferation/drug effects , Hepatitis, Alcoholic/drug therapy , Hepatocytes/drug effects , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1/antagonists & inhibitors , Liver Regeneration/drug effects , Liver/drug effects , Acute-On-Chronic Liver Failure/metabolism , Acute-On-Chronic Liver Failure/pathology , Acute-On-Chronic Liver Failure/physiopathology , Animals , Disease Models, Animal , Female , Hepatitis, Alcoholic/metabolism , Hepatitis, Alcoholic/pathology , Hepatitis, Alcoholic/physiopathology , Hepatocytes/metabolism , Hepatocytes/pathology , Interleukin-1/metabolism , Liver/metabolism , Liver/pathology , Liver/physiopathology , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Recovery of Function , Signal Transduction/drug effects , Time Factors
5.
J Hepatol ; 64(6): 1378-87, 2016 06.
Article in English | MEDLINE | ID: mdl-26867493

ABSTRACT

BACKGROUND & AIMS: Alcoholic liver disease (ALD) ranges from fatty liver to inflammation and cirrhosis. miRNA-155 is an important regulator of inflammation. In this study, we describe the in vivo role of miR-155 in ALD. METHODS: Wild-type (WT) (C57/BL6J) or miR-155 knockout (KO) and TLR4 KO mice received Lieber DeCarli diet for 5weeks. Some mice received corn oil or CCl4 for 2 or 9weeks. RESULTS: We found that miR-155 KO mice are protected from alcohol-induced steatosis and inflammation. The reduction in alcohol-induced fat accumulation in miR-155 KO mice was associated with increased peroxisome proliferator-activated receptor response element (PPRE) and peroxisome proliferator-activated receptors (PPAR)α (miR-155 target) binding and decreased MCP1 production. Treatment with a miR-155 inhibitor increased PPARγ expression in naïve and alcohol treated RAW macrophages. Alcohol increased lipid metabolism gene expression (FABP4, LXRα, ACC1 and LDLR) in WT mice and this was prevented in KO mice. Alcohol diet caused an increase in the number of CD163(+) CD206(+) infiltrating macrophages and neutrophils in WT mice, which was prevented in miR-155 KO mice. Kupffer cells isolated from miR-155 KO mice exhibited predominance of M2 phenotype when exposed to M1 polarized signals and this was due to increased C/EBPß. Pro-fibrotic genes were attenuated in miR-155 KO mice after alcohol diet or CCl4 treatment. Compared to WT mice, attenuation in CCl4 induced hydroxyproline and α-SMA was observed in KO mice. Finally, we show TLR4 signaling regulates miR-155 as TLR4 KO mice showed no induction of miR-155 after alcohol diet. CONCLUSIONS: Collectively our results demonstrated the role of miR-155 in alcohol-induced steatohepatitis and fibrosis in vivo.


Subject(s)
Fatty Liver, Alcoholic/etiology , Liver Cirrhosis, Experimental/etiology , MicroRNAs/physiology , Animals , Carbon Tetrachloride , DNA/metabolism , Female , Interferon-gamma/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/metabolism , Response Elements , Toll-Like Receptor 4/physiology
6.
Dig Dis Sci ; 61(5): 1314-24, 2016 May.
Article in English | MEDLINE | ID: mdl-26769057

ABSTRACT

MicroRNAs (miRNAs) are highly conserved, small, 18-25 nucleotide, non-coding RNAs that regulate gene expression at the post-transcriptional level. Each miRNA can regulate hundreds of target genes, and vice versa each target gene can be regulated by numerous miRNAs, suggesting a very complex network and explaining how miRNAs play pivotal roles in fine-tuning essentially all biological processes in all cell types in the liver. Here, we summarize the current knowledge on the role of miRNAs in the pathogenesis and diagnosis of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) with an outlook to the broader aspects of metabolic syndrome. Furthermore, we discuss the role of miRNAs as potential biomarkers and therapeutic targets in NAFLD/NASH.


Subject(s)
Fatty Liver/genetics , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Biomarkers , Fatty Liver/diagnosis , Fatty Liver/pathology , Humans , Metabolic Syndrome/diagnosis , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology
7.
J Transl Med ; 13: 193, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26077675

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is becoming a pandemic. While multiple 'hits' have been reported to contribute to NAFLD progression to non-alcoholic steatohepatitis (NASH), fibrosis and liver cancer, understanding the natural history of the specific molecular signals leading to hepatocyte damage, inflammation and fibrosis, is hampered by the lack of suitable animal models that reproduce disease progression in humans. The purpose of this study was first, to develop a mouse model that closely mimics progressive NAFLD covering the spectrum of immune, metabolic and histopathologic abnormalities present in human disease; and second, to characterize the temporal relationship between sterile/exogenous danger signals, inflammation, inflammasome activation and NAFLD progression. METHODS: Male C57Bl/6 mice were fed a high fat diet with high cholesterol and a high sugar supplement (HF-HC-HSD) for 8, 27, and 49 weeks and the extent of steatosis, liver inflammation, fibrosis and tumor development were evaluated at each time point. RESULTS: The HF-HC-HSD resulted in liver steatosis at 8 weeks, progressing to steatohepatitis and early fibrosis at 27 weeks, and steatohepatitis, fibrosis, and tumor development at 49 weeks compared to chow diet. Steatohepatitis was characterized by increased levels of MCP-1, TNFα, IL-1ß and increased liver NASH histological score. We found increased serum levels of sterile danger signals, uric acid and HMGB1, as early as 8 weeks, while endotoxin and ATP levels increased only after 49 weeks. Increased levels of these sterile and microbial danger signals paralleled upregulation and activation of the multiprotein complex inflammasome. At 27, 49 weeks of HF-HC-HSD, activation of M1 macrophages and loss of M2 macrophages as well as liver fibrosis were present. Finally, similar to human NASH, liver tumors occurred in 41% of mice in the absence of cirrhosis and livers expressed increased p53 and detectable AFP. CONCLUSIONS: HF-HC-HSD over 49 weeks induces the full spectrum of liver pathophysiologic changes that characterizes the progression of NAFLD in humans. NAFLD progression to NASH, fibrosis and liver tumor follows progressive accumulation of sterile and microbial danger signals, inflammasome activation, altered M1/M2 cell ratios that likely contribute to NASH progression and hepatic tumor formation.


Subject(s)
Disease Progression , Inflammation/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Liver/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Biomarkers/metabolism , Body Weight , Cholesterol/adverse effects , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dietary Carbohydrates/adverse effects , Disease Models, Animal , Feeding Behavior , Female , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Insulin/metabolism , Kupffer Cells/pathology , Liver Cirrhosis/complications , Liver Neoplasms/complications , Macrophage Activation , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/complications , Organ Size , Time Factors , Up-Regulation
8.
PLoS One ; 10(6): e0129251, 2015.
Article in English | MEDLINE | ID: mdl-26042593

ABSTRACT

BACKGROUND & AIM: MicroRNAs (miRs) regulate hepatic steatosis, inflammation and fibrosis. Fibrosis is the consequence of chronic tissue damage and inflammation. We hypothesized that deficiency of miR-155, a master regulator of inflammation, attenuates steatohepatitis and fibrosis. METHODS: Wild type (WT) and miR-155-deficient (KO) mice were fed methionine-choline-deficient (MCD) or -supplemented (MCS) control diet for 5 weeks. Liver injury, inflammation, steatosis and fibrosis were assessed. RESULTS: MCD diet resulted in steatohepatitis and increased miR-155 expression in total liver, hepatocytes and Kupffer cells. Steatosis and expression of genes involved in fatty acid metabolism were attenuated in miR-155 KO mice after MCD feeding. In contrast, miR-155 deficiency failed to attenuate inflammatory cell infiltration, nuclear factor κ beta (NF-κB) activation and enhanced the expression of the pro-inflammatory cytokines tumor necrosis factor alpha (TNFα) and monocyte chemoattractant protein-1 (MCP1) in MCD diet-fed mice. We found a significant attenuation of apoptosis (cleaved caspase-3) and reduction in collagen and α smooth muscle actin (αSMA) levels in miR-155 KO mice compared to WTs on MCD diet. In addition, we found attenuation of platelet derived growth factor (PDGF), a pro-fibrotic cytokine; SMAD family member 3 (Smad3), a protein involved in transforming growth factor-ß (TGFß) signal transduction and vimentin, a mesenchymal marker and indirect indicator of epithelial-to-mesenchymal transition (EMT) in miR-155 KO mice. Nuclear binding of CCAAT enhancer binding protein ß (C/EBPß) a miR-155 target involved in EMT was significantly increased in miR-155 KO compared to WT mice. CONCLUSIONS: Our novel data demonstrate that miR-155 deficiency can reduce steatosis and fibrosis without decreasing inflammation in steatohepatitis.


Subject(s)
Fatty Liver/complications , Fatty Liver/genetics , Inflammation/complications , Liver Cirrhosis/complications , Liver Cirrhosis/genetics , MicroRNAs/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Nucleus/metabolism , Choline , Diet , Disease Models, Animal , Female , Gene Expression Regulation , Lipid Metabolism/genetics , Lipopolysaccharides/pharmacology , Liver/pathology , Methionine , Mice, Inbred C57BL , MicroRNAs/genetics , Protein Binding , Smad3 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vimentin/metabolism
9.
Sci Rep ; 5: 10721, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26024046

ABSTRACT

Circulating miRNAs can be found in extracellular vesicles (EV) and could be involved in intercellular communication. Here, we report the biodistribution of EV associated miR-155 using miR-155 KO mouse model. Administration of exosomes loaded with synthetic miR-155 mimic into miR-155 KO mice resulted in a rapid accumulation and clearance of miR-155 in the plasma with subsequent distribution in the liver, adipose tissue, lung, muscle and kidney (highest to lowest, respectively). miR-155 expression was detected in isolated hepatocytes and liver mononuclear cells of recipient KO mice suggesting its cellular uptake. In vitro, exosome-mediated restoration of miR-155 in Kupffer cells from miR-155 deficient mice augmented their LPS-induced MCP1 mRNA increase. The systemic delivery of wild type plasma to miR-155 KO mice also resulted in a rapid accumulation of miR-155 in the circulation and distribution to the liver and adipose tissue. In summary, our results demonstrate tissue biodistribution and biologic function of EV-associated miR-155.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Exosomes/metabolism , Extracellular Space/metabolism , Kupffer Cells/metabolism , Mice , Mice, Knockout , MicroRNAs/blood , Tissue Distribution
10.
Liver Int ; 35(2): 532-41, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25040043

ABSTRACT

BACKGROUND & AIMS: miR-122 is the most abundant miRNA in the liver particularly in hepatocytes where it targets cholesterol metabolism. Steatosis, a key component of non-alcoholic fatty liver disease, is regulated by hypoxia-inducible factor-1α (HIF-1α). Here, we hypothesized that reduced miR-122 has a pathogenic role in steatohepatitis. METHODS: miR-122 and its target genes were evaluated in mouse livers and/or isolated hepatocytes after methionine-choline-deficient (MCD) or methionine-choline-supplemented (MCS) diet. RESULTS: Liver and hepatocyte miR-122 expression was significantly decreased in steatohepatitis. A maximum reduction in miR-122 occurred at the fibrosis stage (8 weeks of MCD diet). MAP3K3, a miR-122 target gene, was induced at all stages of non-alcoholic steatohepatitis (NASH; 3-8 weeks) only at the mRNA level. Increased NF-κB activation was found in MCD diet-fed mice and MAP3K3 regulated the NF-κB DNA binding in naive hepatocytes. HIF-1α mRNA and DNA binding and expression of the HIF-1α target gene, profibrotic lysyl oxidase, was increased in advanced steatohepatitis (8 weeks). In addition, increase in vimentin and Sirius red staining (liver fibrosis) was found at 8 weeks of MCD diet. Using miR-122 overexpression and inhibition approaches, we confirmed that HIF-1α, vimentin and MAP3K3 are novel miR-122 targets in hepatocytes. We report transcriptional repression of miR-122 in NASH. Decreased liver miR-122 was associated with elevated circulating miR-122 in both exosome-rich and protein-rich serum fractions. CONCLUSIONS: Our novel data suggest that decreased liver miR-122 contributes to upregulation of modulators of tissue remodelling (HIF-1α, vimentin and MAP3K3) and might play a role in NASH-induced liver fibrosis.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Liver Cirrhosis/pathology , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Vimentin/metabolism , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Hepatocytes/metabolism , Immunohistochemistry , Liver Cirrhosis/etiology , MAP Kinase Kinase Kinase 3/metabolism , Mice , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/complications , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
11.
World J Gastroenterol ; 20(26): 8525-34, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-25024607

ABSTRACT

AIM: To develop an animal model that encompasses the different facets of non-alcoholic steatohepatitis (NASH), which has been a challenge. METHODS: In this study, we used a high fat diet (HFD) feeding supplemented with fructose and sucrose in the water mimicking the high-fructose corn syrup that is abundant in the diet in the United States. We used C57Bl/6 wild-type mice for short and long-term feedings of 6 and 16 wk respectively, and evaluated the extent of liver damage, steatosis, and inflammasome activation. Our methods included histopathological analysis to assess liver damage and steatosis, which involved H and E and oil-red-o staining; biochemical studies to look at ALT and triglyceride levels; RNA analysis using quantitative polymerase chain reaction; and cytokine analysis, which included the enzyme-linked immunosorbent assay method to look at interleukin (IL)-1ß and tumor necrosis factor-α (TNFα) levels. Furthermore, at each length of feeding we also looked at insulin resistance and glucose tolerance using insulin tolerance tests (ITT) and glucose tolerance tests. RESULTS: There was no insulin resistance, steatosis, or inflammasome activation at 6 wk. In contrast, at 16 wk we found significant insulin resistance demonstrated by impaired glucose and ITT in male, but not female mice. In males, elevated alanine aminotransferase and triglyceride levels, indicated liver damage and steatosis, respectively. Increased liver TNFα and monocyte chemoattractant protein-1 mRNA and protein, correlated with steatohepatitis. The inflammasome components, adaptor molecule, Aim2, and NOD-like receptor 4, increased at the mRNA level, and functional inflammasome activation was indicated by increased caspase-1 activity and IL-1ß protein levels in male mice fed a long-term HFD. Male mice on HFD had increased α-smooth muscle actin and pro-collagen-1 mRNA indicating evolving fibrosis. In contrast, female mice displayed only elevated triglyceride levels, steatosis, and no fibrosis. CONCLUSION: Our data indicate gender differences in NASH. Male mice fed a long-term HFD display steatohepatitis and inflammasome activation, whereas female mice have steatosis without inflammation.


Subject(s)
Diet, High-Fat/adverse effects , Inflammasomes/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Actins/genetics , Actins/metabolism , Alanine Transaminase/blood , Animals , Blood Glucose/metabolism , Caspase 1/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dietary Sucrose/adverse effects , Female , Fructose/adverse effects , Inflammasomes/genetics , Inflammasomes/immunology , Inflammation Mediators/metabolism , Insulin/blood , Insulin Resistance , Interleukin-1beta/metabolism , Liver/immunology , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Procollagen/genetics , Procollagen/metabolism , RNA, Messenger/metabolism , Sex Factors , Time Factors , Triglycerides/blood , Tumor Necrosis Factor-alpha/metabolism , Weight Gain
12.
Liver Int ; 34(9): 1402-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24650018

ABSTRACT

BACKGROUND & AIMS: Inflammation promotes the progression of non-alcoholic steatohepatitis (NASH). Toll-like receptor 4 (TLR4) and TLR9 activation through myeloid differentiation primary response gene 88 (MyD88) and production of mature interleukin-1ß (IL-1ß) via inflammasome activation contribute to steatohepatitis. Here, we investigated the inter-relationship between TLR signalling and inflammasome activation in dietary steatohepatitis. METHODS: Wild type (WT), TLR4- and MyD88-deficient (KO) mice received methionine-choline-deficient (MCD) or -supplemented (MCS) diets for 5 weeks and a subset was challenged with TLR9 ligand CpG-DNA. RESULTS: TLR4, TLR9, AIM2 (absent in melanoma 2) and NLRP3 (NLR family pyrin domain containing 3) inflammasome mRNA, and mature IL-1ß protein levels were increased in MCD diet-induced steatohepatitis compared to MCS controls. TLR9 stimulation resulted in greater up-regulation of the DNA-sensing AIM2 expression and IL-1ß production in livers of MCD compared to MCS diet-fed mice. High mobility group box 1 (HMGB1), a TLR9-activating danger molecule and phospho-HMGB1 protein levels were also increased in livers of MCD diet-fed mice. MyD88- but not TLR4-deficiency prevented up-regulation of AIM2, NLRP3 mRNA and IL-1ß protein production in dietary steatohepatitis. Selective MyD88 deficiency either in bone marrow (BM)-derived or non-BM-derived cells attenuated hepatic up-regulation of inflammasome mRNA, caspase-1 activation and IL-1ß protein production, but only BM-derived cell-specific MyD88-deficiency attenuated liver injury. CONCLUSIONS: Our data demonstrate that both bone marrow-derived and non-BM-derived cells contribute to inflammasome activation in a MyD88-dependent manner in dietary steatohepatitis. We show that AIM2 inflammasome expression and activation are further augmented by TLR9 ligands in dietary steatohepatitis.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Inflammasomes/metabolism , Myeloid Differentiation Factor 88/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Bone Marrow Cells/metabolism , Choline Deficiency , Diet , Immunoprecipitation , Methionine/deficiency , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
13.
Proc Natl Acad Sci U S A ; 110(41): 16544-9, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24052526

ABSTRACT

Emerging evidence suggests that innate immunity drives alcoholic liver disease (ALD) and that the interferon regulatory factor 3 (IRF3),a transcription factor regulating innate immune responses, is indispensable for the development of ALD. Here we report that IRF3 mediates ALD via linking endoplasmic reticulum (ER) stress with apoptotic signaling in hepatocytes. We found that ethanol induced ER stress and triggered the association of IRF3 with the ER adaptor, stimulator of interferon genes (STING), as well as subsequent phosphorylation of IRF3. Activated IRF3 associated with the proapoptotic molecule Bax [B-cell lymphoma 2 (Bcl2)-associated X protein] and contributed to hepatocyte apoptosis. Deficiency of STING prevented IRF3 phosphorylation by ethanol or ER stress, and absence of IRF3 prevented hepatocyte apoptosis. The pathogenic role of IRF3 in ALD was independent of inflammation or Type-I interferons. Thus, STING and IRF3 are key determinants of ALD, linking ER stress signaling with the mitochondrial pathway of hepatocyte apoptosis.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Immunity, Innate/immunology , Interferon Regulatory Factor-3/metabolism , Liver Diseases, Alcoholic/physiopathology , Membrane Proteins/metabolism , Signal Transduction/immunology , Animals , Hepatocytes/physiology , Liver/metabolism , Liver Diseases, Alcoholic/immunology , Mice , Mice, Knockout , Phosphorylation , Regression Analysis , Triglycerides/metabolism
14.
PLoS One ; 8(8): e70945, 2013.
Article in English | MEDLINE | ID: mdl-23951048

ABSTRACT

INTRODUCTION: Alcohol-induced neuroinflammation is mediated by pro-inflammatory cytokines and chemokines including tumor necrosis factor-α (TNFα), monocyte chemotactic protein-1 (MCP1) and interleukin-1-beta (IL-1ß). Toll-like receptor-4 (TLR4) pathway induced nuclear factor-κB (NF-κB) activation is involved in the pathogenesis of alcohol-induced neuroinflammation. Inflammation is a highly regulated process. Recent studies suggest that microRNAs (miRNAs) play crucial role in fine tuning gene expression and miR-155 is a major regulator of inflammation in immune cells after TLR stimulation. AIM: To evaluate the role of miR-155 in the pathogenesis of alcohol-induced neuroinflammation. METHODS: Wild type (WT), miR-155- and TLR4-knockout (KO) mice received 5% ethanol-containing or isocaloric control diet for 5 weeks. Microglia markers were measured by q-RTPCR; inflammasome activation was measured by enzyme activity; TNFα, MCP1, IL-1ß mRNA and protein were measured by q-RTPCR and ELISA; phospho-p65 protein and NF-κB were measured by Western-blotting and EMSA; miRNAs were measured by q-PCR in the cerebellum. MiR-155 was measured in immortalized and primary mouse microglia after lipopolysaccharide and ethanol stimulation. RESULTS: Chronic ethanol feeding up-regulated miR-155 and miR-132 expression in mouse cerebellum. Deficiency in miR-155 protected mice from alcohol-induced increase in inflammatory cytokines; TNFα, MCP1 protein and TNFα, MCP1, pro-IL-1ß and pro-caspase-1 mRNA levels were reduced in miR-155 KO alcohol-fed mice. NF-κB was activated in WT but not in miR-155 KO alcohol-fed mice. However increases in cerebellar caspase-1 activity and IL-1ß levels were similar in alcohol-fed miR-155-KO and WT mice. Alcohol-fed TLR4-KO mice were protected from the induction of miR-155. NF-κB activation measured by phosphorylation of p65 and neuroinflammation were reduced in alcohol-fed TLR4-KO compared to control mice. TLR4 stimulation with lipopolysaccharide in primary or immortalized mouse microglia resulted in increased miR-155. CONCLUSION: Chronic alcohol induces miR-155 in the cerebellum in a TLR4-dependent manner. Alcohol-induced miR-155 regulates TNFα and MCP1 expression but not caspase-dependent IL-1ß increase in neuroinflammation.


Subject(s)
Alcoholism/genetics , Cerebellum/metabolism , Ethanol/adverse effects , MicroRNAs/genetics , Microglia/metabolism , Toll-Like Receptor 4/genetics , Alcoholism/immunology , Alcoholism/pathology , Animals , Cerebellum/immunology , Cerebellum/pathology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chronic Disease , Female , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Knockout , MicroRNAs/immunology , Microglia/immunology , Microglia/pathology , Primary Cell Culture , Signal Transduction , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
15.
J Gastroenterol Hepatol ; 28 Suppl 1: 93-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23855302

ABSTRACT

The similar histopathological characteristics of alcoholic steatohepatitis (ASH) and non-alcoholic steatohepatitis (NASH), and the crucial role of the innate immune response in both conditions may lead to the assumption that ASH and NASH represent the same pathophysiological entities caused by different risk factors. In this review paper, we elaborate on the pathophysiological differences between these two entities and highlight the disease-specific involvement of signaling molecules downstream of the Toll-like receptor 4, and the differential mechanism by which the inflammasome contributes to ASH versus NASH. Our findings emphasize that ASH and NASH have disease-specific mechanisms and therefore represent distinct biological entities. Further studies are needed to dissect the emerging differences in pathogenesis of these two conditions.


Subject(s)
Fatty Liver, Alcoholic/immunology , Fatty Liver/immunology , Immunity, Innate/immunology , Signal Transduction/immunology , Bacteria , Fatty Liver/genetics , Fatty Liver, Alcoholic/genetics , Humans , Immunity, Innate/genetics , Immunity, Innate/physiology , Inflammasomes , Interleukin-1 , Intestines/microbiology , Lipopolysaccharides , Non-alcoholic Fatty Liver Disease , Risk Factors , Signal Transduction/genetics , Signal Transduction/physiology , Toll-Like Receptor 4
16.
J Leukoc Biol ; 94(1): 171-82, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23625200

ABSTRACT

Alcohol-induced neuroinflammation is mediated by proinflammatory cytokines, including IL-1ß. IL-1ß production requires caspase-1 activation by inflammasomes-multiprotein complexes that are assembled in response to danger signals. We hypothesized that alcohol-induced inflammasome activation contributes to increased IL-1ß in the brain. WT and TLR4-, NLRP3-, and ASC-deficient (KO) mice received an ethanol-containing or isocaloric control diet for 5 weeks, and some received the rIL-1ra, anakinra, or saline treatment. Inflammasome activation, proinflammatory cytokines, endotoxin, and HMGB1 were measured in the cerebellum. Expression of inflammasome components (NLRP1, NLRP3, ASC) and proinflammatory cytokines (TNF-α, MCP-1) was increased in brains of alcohol-fed compared with control mice. Increased caspase-1 activity and IL-1ß protein in ethanol-fed mice indicated inflammasome activation. TLR4 deficiency protected from TNF-α, MCP-1, and attenuated alcohol-induced IL-1ß increases. The TLR4 ligand, LPS, was not increased in the cerebellum. However, we found up-regulation of acetylated and phosphorylated HMGB1 and increased expression of the HMGB1 receptors (TLR2, TLR4, TLR9, RAGE) in alcohol-fed mice. NLRP3- or ASC-deficient mice were protected from caspase-1 activation and alcohol-induced IL-1ß increase in the brain. Furthermore, in vivo treatment with rIL-1ra prevented alcohol-induced inflammasome activation and IL-1ß, TNF-α, and acetylated HMGB1 increases in the cerebellum. Conversely, intracranial IL-1ß administration induced TNF-α and MCP-1 in the cerebellum. In conclusion, alcohol up-regulates and activates the NLRP3/ASC inflammasome, leading to caspase-1 activation and IL-1ß increase in the cerebellum. IL-1ß amplifies neuroinflammation, and disruption of IL-1/IL-1R signaling prevents alcohol-induced inflammasome activation and neuroinflammation. Increased levels of acetylated and phosphorylated HMGB1 may contribute to alcoholic neuroinflammation.


Subject(s)
Brain/pathology , Carrier Proteins/physiology , Cytoskeletal Proteins/physiology , Ethanol/toxicity , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Toll-Like Receptor 4/physiology , Acetylation/drug effects , Animals , Anti-Infective Agents, Local/toxicity , Apoptosis Regulatory Proteins , Blotting, Western , Brain/immunology , Brain/metabolism , CARD Signaling Adaptor Proteins , Cytokines/genetics , Cytokines/metabolism , Female , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Immunoprecipitation , Inflammasomes/drug effects , Inflammasomes/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphorylation/drug effects , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
17.
Adv Clin Chem ; 59: 155-201, 2013.
Article in English | MEDLINE | ID: mdl-23461136

ABSTRACT

Activation of inflammatory signaling pathways is of central importance in the pathogenesis of alcoholic liver disease (ALD) and nonalcoholic steatohepatitis (NASH). Recent studies demonstrated that Toll-like receptors, the sensors of microbial and endogenous danger signals, are expressed and activated in innate immune cells as well as in parenchymal cells in the liver and thereby contribute to ALD and NASH. In this review, we emphasize the importance of gut-derived endotoxin and its recognition by TLR4 in the liver. The significance of TLR-induced intracellular signaling pathways and cytokine production as well as the contribution of individual cell types to the inflammation is evaluated. The contribution of TLR signaling to the induction of liver fibrosis and to the progression of liver pathology mediated by viral pathogens is reviewed in the context of ALD and NASH.


Subject(s)
Liver Diseases/etiology , Toll-Like Receptors/physiology , Animals , Cytokines/biosynthesis , Fatty Liver/etiology , Humans , Immunity, Innate , Liver Diseases, Alcoholic/etiology , Non-alcoholic Fatty Liver Disease , Signal Transduction
18.
J Clin Invest ; 122(10): 3476-89, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945633

ABSTRACT

Alcoholic liver disease (ALD) is characterized by steatosis and upregulation of proinflammatory cytokines, including IL-1ß. IL-1ß, type I IL-1 receptor (IL-1R1), and IL-1 receptor antagonist (IL-1Ra) are all important regulators of the IL-1 signaling complex, which plays a role in inflammation. Furthermore, IL-1ß maturation is dependent on caspase-1 (Casp-1). Using IL-1Ra-treated mice as well as 3 mouse models deficient in regulators of IL-1ß activation (Casp-1 and ASC) or signaling (IL-1R1), we found that IL-1ß signaling is required for the development of alcohol-induced liver steatosis, inflammation, and injury. Increased IL-1ß was due to upregulation of Casp-1 activity and inflammasome activation. The pathogenic role of IL-1 signaling in ALD was attributable to the activation of the inflammasome in BM-derived Kupffer cells. Importantly, in vivo intervention with a recombinant IL-1Ra blocked IL-1 signaling and markedly attenuated alcohol-induced liver inflammation, steatosis, and damage. Furthermore, physiological doses of IL-1ß induced steatosis, increased the inflammatory and prosteatotic chemokine MCP-1 in hepatocytes, and augmented TLR4-dependent upregulation of inflammatory signaling in macrophages. In conclusion, we demonstrated that Casp-1-dependent upregulation of IL-1ß and signaling mediated by IL-1R1 are crucial in ALD pathogenesis. Our findings suggest a potential role of IL-1R1 inhibition in the treatment of ALD.


Subject(s)
Fatty Liver, Alcoholic/drug therapy , Inflammasomes/physiology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Caspase 1/physiology , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Drug Evaluation, Preclinical , Ethanol/toxicity , Fatty Liver, Alcoholic/etiology , Female , Hepatocytes/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/blood , Interleukin-1beta/biosynthesis , Interleukin-1beta/blood , Interleukin-1beta/physiology , Interleukin-1beta/toxicity , Kupffer Cells/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/physiology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Toll-Like Receptor 4/physiology , Up-Regulation/drug effects
19.
J Hepatol ; 57(3): 642-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22634126

ABSTRACT

Inflammation is a common element in the pathogenesis of most chronic liver diseases that lead to fibrosis and cirrhosis. Inflammation is characterized by activation of innate immune cells and production of pro-inflammatory cytokines IL-1α, IL-1ß, and TNFα. Inflammasomes are intracellular multiprotein complexes expressed in both parenchymal and non-parenchymal cells of the liver that in response to cellular danger signals activate caspase-1, and release IL-1ß and IL-18. The importance of inflammasome activation in various forms of liver diseases in relation to liver damage, steatosis, inflammation and fibrosis is discussed in this review.


Subject(s)
Caspase 1/metabolism , Inflammasomes/metabolism , Liver Diseases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins , Humans , Interleukin-1beta/metabolism , Liver Diseases/enzymology , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Nuclear Proteins/metabolism
20.
World J Gastroenterol ; 17(43): 4772-8, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-22147977

ABSTRACT

AIM: To examine the activation of the Nalp3 inflammasome and its downstream targets following lipopolysaccharide (LPS)-induced stimulation in the liver. METHODS: Six-to-eight-week-old C57BL/6 chow fed mice were injected intraperitoneally with 0.5 µg/g bodyweight LPS and sacrificed 2, 4, 6, 18 or 24 h later. LPS-induced liver damage was confirmed by a biochemical assay to detect alanine aminotransferase (ALT) levels. To determine if LPS stimulation in the liver led to activation of the inflammasome, real-time quantitative polymerase chain reaction was used to evaluate the mRNA expression of components of the Nalp3 inflammasome. Enzyme-linked immunosorbent assays were used to determine the protein expression levels of several downstream targets of the Nalp3 inflammasome, including caspase-1 and two cytokine targets of caspase-1, interleukin (IL)-1ß and IL-18. RESULTS: We found that LPS injection resulted in liver damage as indicated by elevated ALT levels. This was associated with a significant increase in both mRNA and protein levels of the proinflammatory cytokine tumor necrosis factor (TNF)-α in the liver, as well as increased levels of TNFs in serum. We showed that LPS stimulation led to upregulation of mRNA levels in the liver for all the receptor components of the inflammasome, including Nalp3, Nalp1, pannexin-1 and the adaptor molecule apoptosis-associated speck-like, caspase recruitment domain-domain containing protein. We also found increased levels of mRNA and protein for caspase-1, a downstream target of the inflammasome. In addition, LPS challenge led to increased levels of both mRNA and protein in the liver for two cytokine targets of caspase-1, IL-1ß and IL-18. Interestingly, substantial baseline expression of pre-IL-1ß and pre-IL-18 was found in the liver. Inflammasome and caspase-1 activation was indicated by the significant increase in the active forms of IL-1ß and IL-18 after LPS stimulation. CONCLUSION: Our results show that the Nalp3 inflammasome is upregulated and activated in the liver in response to LPS stimulation.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Lipopolysaccharides/immunology , Liver/metabolism , Liver/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...