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










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Hepatol Int ; 7 Suppl 2: 771-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24587847

ABSTRACT

Immune and inflammatory pathways have a central role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Both the innate and adaptive immune systems contribute to the development of NAFLD. Pathogen-associated molecular patterns and danger-associated molecular patterns are known to activate a variety of pattern-recognition receptors that result in inflammation. The key features of the immune system and inflammatory pathways in the development of NAFLD are discussed in this review.

7.
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
8.
Hepatology ; 54(1): 133-44, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21488066

ABSTRACT

UNLABELLED: The pathogenesis of nonalcoholic steatohepatitis (NASH) and inflammasome activation involves sequential hits. The inflammasome, which cleaves pro-interleukin-1ß (pro-IL-1ß) into secreted IL-1ß, is induced by endogenous and exogenous danger signals. Lipopolysaccharide (LPS), a toll-like receptor 4 ligand, plays a role in NASH and also activates the inflammasome. In this study, we hypothesized that the inflammasome is activated in NASH by multiple hits involving endogenous and exogenous danger signals. Using mouse models of methionine choline-deficient (MCD) diet-induced NASH and high-fat diet-induced NASH, we found up-regulation of the inflammasome [including NACHT, LRR, and PYD domains-containing protein 3 (NALP3; cryopyrin), apoptosis-associated speck-like CARD-domain containing protein, pannexin-1, and pro-caspase-1] at the messenger RNA (mRNA) level increased caspase-1 activity, and mature IL-1ß protein levels in mice with steatohepatitis in comparison with control livers. There was no inflammasome activation in mice with only steatosis. The MCD diet sensitized mice to LPS-induced increases in NALP3, pannexin-1, IL-1ß mRNA, and mature IL-1ß protein levels in the liver. We demonstrate for the first time that inflammasome activation occurs in isolated hepatocytes in steatohepatitis. Our novel data show that the saturated fatty acid (FA) palmitic acid (PA) activates the inflammasome and induces sensitization to LPS-induced IL-1ß release in hepatocytes. Furthermore, PA triggers the release of danger signals from hepatocytes in a caspase-dependent manner. These hepatocyte-derived danger signals, in turn, activate inflammasome, IL-1ß, and tumor necrosis factor α release in liver mononuclear cells. CONCLUSION: Our novel findings indicate that saturated FAs represent an endogenous danger in the form of a first hit, up-regulate the inflammasome in NASH, and induce sensitization to a second hit with LPS for IL-ß release in hepatocytes. Furthermore, hepatocytes exposed to saturated FAs release danger signals that trigger inflammasome activation in immune cells. Thus, hepatocytes play a key role in orchestrating tissue responses to danger signals in NASH.


Subject(s)
Endotoxins/pharmacology , Fatty Acids/pharmacology , Fatty Liver/etiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Immune System/physiology , Inflammasomes/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 1/metabolism , Choline Deficiency/complications , Dietary Fats/adverse effects , Disease Models, Animal , Endotoxins/adverse effects , Fatty Acids/adverse effects , Fatty Liver/metabolism , Fatty Liver/physiopathology , Female , Hepatocytes/pathology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Methionine/deficiency , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...