Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
Hepatology ; 74(5): 2774-2790, 2021 11.
Article in English | MEDLINE | ID: mdl-34089528

ABSTRACT

BACKGROUND AND AIMS: HSCs and portal fibroblasts (PFs) are the major sources of collagen-producing myofibroblasts during liver fibrosis, depending on different etiologies. However, the mechanisms by which their dynamic gene expression directs the transition from the quiescent to the activated state-as well as their contributions to fibrotic myofibroblasts-remain unclear. Here, we analyze the activation of HSCs and PFs in CCL4 -induced and bile duct ligation-induced fibrosis mouse models, using single-cell RNA sequencing and lineage tracing. APPROACH AND RESULTS: We demonstrate that HSCs, rather than PFs, undergo dramatic transcriptomic changes, with the sequential activation of inflammatory, migrative, and extracellular matrix-producing programs. The data also reveal that HSCs are the exclusive source of myofibroblasts in CCL4 -treated liver, while PFs are the major source of myofibroblasts in early cholestatic liver fibrosis. Single-cell and lineage-tracing analysis also uncovers differential gene-expression features between HSCs and PFs; for example, nitric oxide receptor soluble guanylate cyclase is exclusively expressed in HSCs, but not in PFs. The soluble guanylate cyclase stimulator Riociguat potently reduced liver fibrosis in CCL4 -treated livers but showed no therapeutic efficacy in bile duct ligation livers. CONCLUSIONS: This study provides a transcriptional roadmap for the activation of HSCs during liver fibrosis and yields comprehensive evidence that the differential transcriptomic features of HSCs and PFs, along with their relative contributions to liver fibrosis of different etiologies, should be considered in developing effective antifibrotic therapeutic strategies.


Subject(s)
Hepatic Stellate Cells/immunology , Liver Cirrhosis, Experimental/immunology , Myofibroblasts/immunology , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Lineage/immunology , Cells, Cultured , Gene Expression Regulation/immunology , Gene Knock-In Techniques , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Male , Mice , Mice, Transgenic , Primary Cell Culture , RNA-Seq , Single-Cell Analysis
2.
Front Immunol ; 11: 580968, 2020.
Article in English | MEDLINE | ID: mdl-33013934

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) has been on a global rise. While animal models have rendered valuable insights to the pathogenesis of NAFLD, discrepancy with patient data still exists. Since non-alcoholic steatohepatitis (NASH) involves chronic inflammation, and CD4+ T cell infiltration of the liver is characteristic of NASH patients, we established and characterized a humanized mouse model to identify human-specific immune response(s) associated with NAFLD progression. Immunodeficient mice engrafted with human immune cells (HIL mice) were fed with high fat and high calorie (HFHC) or chow diet for 20 weeks. Liver histology and immune profile of HIL mice were analyzed and compared with patient data. HIL mice on HFHC diet developed steatosis, inflammation and fibrosis of the liver. Human CD4+ central and effector memory T cells increased within the liver and in the peripheral blood of our HIL mice, accompanied by marked up-regulation of pro-inflammatory cytokines (IL-17A and IFNγ). In vivo depletion of human CD4+ T cells in HIL mice reduced liver inflammation and fibrosis, but not steatosis. Our results highlight CD4+ memory T cell subsets as important drivers of NAFLD progression from steatosis to fibrosis and provides a humanized mouse model for pre-clinical evaluation of potential therapeutics.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/immunology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Cytokines/blood , Diet, High-Fat/adverse effects , Female , Fetal Stem Cells/transplantation , Hepatocytes/transplantation , Heterografts , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Liver Cirrhosis, Experimental/pathology , Lymphocyte Depletion , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Non-alcoholic Fatty Liver Disease/pathology
3.
World J Gastroenterol ; 26(21): 2810-2820, 2020 Jun 07.
Article in English | MEDLINE | ID: mdl-32550756

ABSTRACT

BACKGROUND: Liver fibrosis is a common health problem worldwide and there is still a lack of effective medicines. The Chinese herbal medicine, Gan Shen Fu Fang (GSFF) is composed of salvianolic acid B and diammonium glycyrrhizinate. In this study, we observed the effects of GSFF on liver fibrosis in vivo and in vitro in an attempt to provide some hope for the treatment. AIM: To observe the effects of GSFF on liver fibrosis in vivo and in vitro and investigate the mechanism from the perspective of the inflammatory response and extracellular signal-regulated kinase (ERK) phosphorylation. METHODS: Common bile duct-ligated rats were used for in vivo experiments. Hepatic stellate cells-T6 (HSC-T6) cells were used for in vitro experiments. Hematoxylin and eosin staining and Masson staining, biochemical assays, hydroxyproline (Hyp) assays, enzyme-linked immunoasorbent assay and western blotting were performed to evaluate the degree of liver fibrosis, liver function, the inflammatory response and ERK phosphorylation. The CCK8 assay, immunofluorescence and western blotting were applied to test the effect of GSFF on HSC-T6 cell activation and determine whether GSFF had an effect on ERK phosphorylation in HSC-T6 cells. RESULTS: GSFF improved liver function and inhibited liver fibrosis in common bile duct-ligated rats after 3 wk of treatment, as demonstrated by histological changes, hydroxyproline assays and collagen I concentrations. GSFF alleviated inflammatory cell infiltration and reduced the synthesis of pro-inflammatory cytokines [tumor necrosis factor-α (TNF-α) and interlukin-1ß] and NF-κB. In addition, GSFF decreased ERK phosphorylation. In vitro, GSFF inhibited the viability of HSC-T6 cells with and without transforming growth factor ß1 (TGF-ß1) stimulation and decreased the synthesis of collagen I. GSFF had the greatest effect at a concentration of 0.5 µmol/L. GSFF inhibited the expression of α-smooth muscle actin (α-SMA), a marker of HSC activation, in HSC-T6 cells. Consistent with the in vivo results, GSFF also inhibited the phosphorylation of ERK and downregulated the expression of NF-κB. CONCLUSION: GSFF inhibited liver fibrosis progression in vivo and HSC-T6 cell activation in vitro. These effects may be related to an alleviated inflammatory response and downregulated ERK phosphorylation.


Subject(s)
Benzofurans/pharmacology , Drugs, Chinese Herbal/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycyrrhetinic Acid/analogs & derivatives , Liver Cirrhosis, Experimental/drug therapy , Animals , Benzofurans/therapeutic use , Cell Line , Disease Progression , Drugs, Chinese Herbal/therapeutic use , Glycyrrhetinic Acid/pharmacology , Glycyrrhetinic Acid/therapeutic use , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/pathology , Humans , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Male , Phosphorylation/drug effects , Phosphorylation/immunology , Rats , Signal Transduction/drug effects , Signal Transduction/immunology
4.
Front Immunol ; 11: 354, 2020.
Article in English | MEDLINE | ID: mdl-32218781

ABSTRACT

Background and Aims: Monocyte chemotactic protein-1 (MCP-1) is a potent chemoattractant for monocytes. It is involved in pathogenesis of several inflammatory diseases. Hepatic MCP-1 is a readout of macrophage activation. While inflammation is a major driver of liver disease progression, the origin and role of circulating MCP-1 as a biomarker remains unclear. Methods: Hepatic CC-chemokine ligand 2 (CCL2) expression and F4/80 staining for Kupffer cells were measured and correlated in a mouse model of chronic liver disease (inhalative CCl4 for 7 weeks). Next, hepatic RNA levels of CCL2 were measured in explanted livers of 39 patients after transplantation and correlated with severity of disease. Changes in MCP-1 were further evaluated in a rat model of experimental cirrhosis and acute-on-chronic liver failure (ACLF). Finally, we analyzed portal and hepatic vein levels of MCP-1 in patients receiving transjugular intrahepatic portosystemic shunt insertion for complications of portal hypertension. Results: In this mouse model of fibrotic hepatitis, hepatic expression of CCL2 (P = 0.009) and the amount of F4/80 positive cells in the liver (P < 0.001) significantly increased after induction of hepatitis by CCl4 compared to control animals. Moreover, strong correlation of hepatic CCL2 expression and F4/80 positive cells were seen (P = 0.023). Furthermore, in human liver explants, hepatic transcription levels of CCL2 correlated with the MELD score of the patients, and thus disease severity (P = 0.007). The experimental model of ACLF in rats revealed significantly higher levels of MCP-1 plasma (P = 0.028) and correlation of hepatic CCL2 expression (R = 0.69, P = 0.003). Particularly, plasma MCP-1 levels did not correlate with peripheral blood monocyte CCL2 expression. Finally, higher levels of MCP-1 were observed in the hepatic compared to the portal vein (P = 0.01) in patients receiving TIPS. Similarly, a positive correlation of MCP-1 with Child-Pugh score was observed (P = 0.018). Further, in the presence of ACLF, portal and hepatic vein levels of MCP-1 were significantly higher compared to patients without ACLF (both P = 0.039). Conclusion: Circulating levels of MCP-1 mainly derive from the injured liver and are associated with severity of liver disease. Therefore, liver macrophages contribute significantly to disease progression. Circulating MCP-1 may reflect the extent of hepatic macrophage activation.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Chemokine CCL2/blood , Liver Cirrhosis, Experimental/complications , Liver/immunology , Macrophage Activation , Acute-On-Chronic Liver Failure/complications , Acute-On-Chronic Liver Failure/immunology , Animals , Chemokine CCL2/analysis , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Kupffer Cells/physiology , Liver Cirrhosis, Experimental/immunology , Male , Mice , Mice, Inbred C57BL
5.
Hepatology ; 72(5): 1800-1818, 2020 11.
Article in English | MEDLINE | ID: mdl-32064648

ABSTRACT

BACKGROUND AND AIMS: During liver injury, quiescent hepatic stellate cells (qHSCs) transdifferentiate into proliferative and fibrogenic activated myofibroblastic phenotype (activated hepatic stellate cell; aHSCs) expressing smooth muscle α-actin (αSMA) and platelet-derived growth factor beta receptor (PDGFßR). Their interactions with gut-derived bacterial lipopolysaccharide (LPS) are implicated in hepatic fibrogenesis. However, LPS can also attenuate fibrogenic characteristics of aHSCs. APPROACH AND RESULTS: We examined molecular mechanisms of antifibrogenic effects of LPS on aHSCs in vitro and in vivo. Culture-activated rat HSCs were exposed to 0-100 ng/mL of LPS or its active component, diphosphoryl-lipid A (DPLA), and parameters of fibrosis and inflammatory cytokines/chemokines were determined by qRT-PCR, western, and immunohistochemical analyses. In vivo, HSCs were activated by repeated CCl4 administration to rats every 3 days for 3 or 8 weeks, then challenged with LPS (5 mg/kg; IP). HSCs were isolated 24 hours later, and fibrogenic/inflammatory parameters were analyzed. LPS induced phenotypic changes in aHSCs (rounding, size reduction) and loss of proliferation. LPS down-regulated expression of αSMA, PDGFßR, transforming growth factor beta receptor 1 (TGFßR1), collagen 1α1 (Col1α1), and fibronectin while up-regulating tumor necrosis factor alpha, interleukin-6, and C-X-C motif chemokine ligand 1 expression. LPS did not increase peroxisome proliferation-activated receptor gamma expression or lipid accumulation typical of qHSCs. DPLA elicited the same effects as LPS on aHSCs, indicating specificity, and monophosphoryl lipid A down-regulated fibrogenic markers, but elicited very weak inflammatory response. LPS down-regulated the expression of cMyb, a transcription factor for αSMA, and up-regulated small mother against decapentaplegic (SMAD)7 and CCAAT/enhancer-binding protein (C/EBP)δ, the transcriptional inhibitors of Col1α1 expression. In vivo LPS treatment of aHSCs inhibited their proliferation, down-regulated PDGFßR, αSMA, TGFßR1, Col1α1, and cMyb expression, and increased expression of SMAD7, C/EBPα, and C/EBPδ. CONCLUSIONS: In conclusion, LPS induces a unique phenotype in aHSCs associated with down-regulation of key fibrogenic mechanisms and thus may have an important role in limiting fibrosis.


Subject(s)
Gene Expression Regulation/immunology , Hepatic Stellate Cells/immunology , Lipid A/analogs & derivatives , Liver Cirrhosis, Experimental/immunology , Liver/pathology , Animals , CCAAT-Enhancer-Binding Protein-delta/metabolism , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Transdifferentiation/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Down-Regulation , Gene Silencing , Hepatic Stellate Cells/pathology , Humans , Lipid A/immunology , Lipid A/metabolism , Liver/cytology , Liver/immunology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Mice , Mice, Knockout , Myofibroblasts/immunology , Myofibroblasts/pathology , Oxidoreductases Acting on Sulfur Group Donors/genetics , Primary Cell Culture , Proto-Oncogene Proteins c-myb/metabolism , Rats , Signal Transduction/genetics , Signal Transduction/immunology , Smad7 Protein/genetics , Smad7 Protein/metabolism , Up-Regulation/immunology
6.
Cell Mol Gastroenterol Hepatol ; 9(3): 425-446, 2020.
Article in English | MEDLINE | ID: mdl-31669262

ABSTRACT

BACKGROUND & AIMS: Liver inflammation has been recognized as a hallmark of hepatocarcinogenesis. Although Forkhead Box M1 (FoxM1) is a well-defined oncogenic transcription factor that is overexpressed in hepatocellular carcinoma (HCC), its role in liver inflammation has never been explored. METHODS: We generated hepatocyte-specific FoxM1 conditional transgenic (TG) mice by using the Cre-loxP and Tetracycline (Tet)-on systems to induce FoxM1 expression in a hepatocyte-specific and time-dependent manner. RESULTS: After treatment of Tet-derivatives doxycycline (DOX) to induce FoxM1, TG mice exhibited spontaneous development of hepatocyte death with elevated serum alanine aminotransferase levels and hepatic infiltration of macrophages. The removal of DOX in TG mice completely removed this effect, suggesting that spontaneous inflammation in TG mice occurs in a hepatocyte FoxM1-dependent manner. In addition, liver inflammation in TG mice was associated with increased levels of hepatic and serum chemokine (C-C motif) ligand 2 (CCL2). In vitro transcriptional analysis confirmed that CCL2 is a direct target of FoxM1 in murine hepatocytes. After receiving FoxM1 induction since birth, all TG mice exhibited spontaneous HCC with liver fibrosis at 12 months of age. Hepatic expression of FoxM1 was significantly increased in liver injury models. Finally, pharmacologic inhibition of FoxM1 reduced liver inflammation in models of liver injury. CONCLUSIONS: Hepatocyte FoxM1 acts as a crucial regulator to orchestrate liver inflammation linking to hepatocarcinogenesis. Thus, hepatocyte FoxM1 may be a potential target not only for the treatment of liver injury but also for the prevention toward HCC.


Subject(s)
Carcinogenesis/immunology , Forkhead Box Protein M1/metabolism , Hepatitis/pathology , Liver Neoplasms/pathology , Animals , Apoptosis/immunology , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Forkhead Box Protein M1/analysis , Forkhead Box Protein M1/genetics , Hepatectomy , Hepatitis/diagnosis , Hepatitis/immunology , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Liver/immunology , Liver/pathology , Liver/surgery , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Liver Function Tests , Liver Neoplasms/immunology , Liver Neoplasms/surgery , Mice , Mice, Transgenic , Primary Cell Culture
7.
PLoS One ; 14(7): e0215557, 2019.
Article in English | MEDLINE | ID: mdl-31291257

ABSTRACT

BACKGROUND: Chronic inflammation is the driver of liver injury and results in progressive fibrosis and eventual cirrhosis with consequences including both liver failure and liver cancer. We have previously described increased expression of the highly multifunctional glycoprotein CD147 in liver injury. This work describes a novel role of CD147 in liver inflammation and the importance of leukocyte aggregates in determining the extent of liver injury. METHODS: Non-diseased, progressive injury, and cirrhotic liver from humans and mice were examined using a mAb targeting CD147. Inflammatory cell subsets were assessed by multiparameter flow cytometry. RESULTS: In liver injury, we observe abundant, intrahepatic leukocyte clusters defined as ≥5 adjacent CD45+ cells which we have termed "leukocyte aggregates". We have shown that these leukocyte aggregates have a significant effect in determining the extent of liver injury. If CD147 is blocked in vivo, these leukocyte aggregates diminish in size and number, together with a marked significant reduction in liver injury including fibrosis. This is accompanied by no change in overall intrahepatic leukocyte numbers. Further, blocking of aggregation formation occurs prior to an appreciable increase in inflammatory markers or fibrosis. Additionally, there were no observed, "off-target" or unpredicted effects in targeting CD147. CONCLUSION: CD147 mediates leukocyte aggregation which is associated with the development of liver injury. This is not a secondary effect, but a cause of injury as aggregate formation proceeds other markers of injury. Leukocyte aggregation has been previously described in inflammation dating back over many decades. Here we demonstrate that leukocyte aggregates determine the extent of liver injury.


Subject(s)
Basigin/metabolism , Leukocytes/immunology , Liver/immunology , Liver/injuries , Animals , Basigin/genetics , Cell Aggregation/immunology , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Leukocytes/classification , Leukocytes/pathology , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Up-Regulation
8.
Immunol Lett ; 212: 37-45, 2019 08.
Article in English | MEDLINE | ID: mdl-31226359

ABSTRACT

Splenic immune cells, especially macrophages, play a key role in multiple pathological processes. With a proved anti-inflammatory and immunoregulatory function of mesencephalicastrocyte-derived neurotrophic factor (MANF) in inflammatory disorders, how MANF affects splenic immune cells in physiological and pathophysiological situations is still unknown. In this study, we constructed mono-macrophage-specific MANF knockout (Mø MANF-/-) mice and found the increased splenic M1 macrophages, but no significant change of splenic morphology and size compared with wild type (WT) mice. Also, we established the pathophysiological situation of carbon tetrachloride (CCl4)-induced hepatic fibrosis. Under the hepatic fibrosis, splenic M2 macrophages and CD138+ plasma cells were significantly increased in Mø MANF-/- mice. Consistently, we found the increased TGF-ß1 level in serum and spleen of Mø MANF-/- mice as well. Mono-macrophage-specific MANF knockout did not affect the number of splenic T and B cells under both the normal and hepatic fibrosis conditions. Our results suggest a distinct regulation of MANF on splenic immune cells and a specific regulation of MANF on the differentiation of splenic macrophages, which may exert a significant impact on physiological and pathophysiological processes of the spleen.


Subject(s)
Cell Differentiation/immunology , Liver Cirrhosis, Experimental/immunology , Macrophages/immunology , Nerve Growth Factors/metabolism , Spleen/cytology , Animals , Carbon Tetrachloride/toxicity , Humans , Liver Cirrhosis, Experimental/chemically induced , Male , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Organ Size , Spleen/immunology , Spleen/pathology , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
9.
Biomed Pharmacother ; 115: 108901, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31079002

ABSTRACT

An ingredient was isolated from Acanthus ilicifolius and identified as 4-hydroxy-2(3H)-benzoxazolone (HBOA). Its protective effects and underlying mechanism on liver fibrosis were investigated. Briefly, rats were intragastrically administrated with 50% CCl4 twice a week for 12 weeks to induce liver fibrosis. Meanwhile, the animals were treated with various medicines from weeks 8 to 12. Then the histological change, serum biochemical index, inflammatory factors and hepatocyte apoptosis were detected. Moreover, the TGF-ß1/Smads, NF-κB and ERK signaling pathways were also detected to illustrate the underlying mechanism. The results showed that HBOA significantly ameliorated CCl4-induced liver injury and collagen accumulation in rats, as evidenced by the histopathologic improvement. Moreover, HBOA markedly decreased hepatocyte apoptosis by regulating the expression levels of caspase-3, -9 and -12, as well as the Bcl-2 family. The mechanism study showed that HBOA significantly decreased the expressions of α-smooth muscle actin (α-SMA) and collagen and inhibited the generation of excessive extracellular matrix (ECM) components by restoring the balance between matrix metalloproteinases (MMPs) and its inhibitor (TIMPs). HBOA markedly alleviated oxidative stress and inflammatory cytokines through inhibiting the NF-κB pathway. In addition, HBOA significantly down-regulated the levels of TGF-ß1, Smad2/3, Smad4 and up-regulated the level of Smad7, inhibiting the TGF-ß1/Smads signaling pathway. Moreover, HBOA significantly blocked the ERK signaling pathway, leading to the inactivation of hepatic stellate cells. This study suggests that HBOA exerts a protective effect against liver fibrosis via modulating the TGF-ß1/Smads, NF-κB and ERK signaling pathways, which will be developed as a potential agent for the treatment of liver fibrosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoxazoles/pharmacology , Liver Cirrhosis, Experimental/prevention & control , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Oxazolone/pharmacology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Acanthaceae/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Benzoxazoles/isolation & purification , Carbon Tetrachloride , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Male , Medicine, Chinese Traditional , Oxazolone/isolation & purification , Oxazolone/therapeutic use , Oxidative Stress/drug effects , Oxidative Stress/immunology , Rats, Sprague-Dawley
10.
J Surg Res ; 241: 254-263, 2019 09.
Article in English | MEDLINE | ID: mdl-31035140

ABSTRACT

BACKGROUND: Clinically, liver fibrosis and cholestasis are two major disease entities, ultimately leading to hepatic failure. Although autophagy plays a substantial role in the pathogenesis of these diseases, its precise mechanism has not been determined yet. MATERIALS AND METHODS: Mouse models of liver fibrosis or cholestasis were obtained after the serial administration of thioacetamide (TAA) or surgical bile duct ligation (BDL), respectively. Then, after obtaining liver specimens at specific time points, we compared the expression of makers related to apoptosis (cleaved caspases), inflammation (CD68), necrosis (high-mobility group box 1), phospho-c-Jun N-terminal kinase (p-JNK), and autophagy (microtubule-associated protein light chain 3B and p62) in the fibrotic or cholestatic mouse livers, by polymerase chain reaction, Western blot analysis, immunohistochemistry, and immunofluorescence. RESULTS: Although cholestatic livers exhibited the tendency of progressively increasing the expression of most apoptosis-related markers (cleaved caspases), it was not prominent when it was compared with the tendency found in the livers of TAA-treated mice. Contrastingly, the necrosis-related factor (high-mobility group box 1) was significantly increased in the livers of BDL mice over time, reaching their peak values on day 7 after BDL. In addition, the inflammation-related factor (CD68) was highly expressed in BDL mice compared with TAA-treated mice over time. Autophagy marker studies indicated that autophagy was upregulated in fibrotic livers, whereas it was downregulated in cholestatic livers. We also observed mild to moderate activation of p-JNK in the livers of TAA-treated mice, whereas significantly higher p-JNK activation was detected in the livers of BDL mice. CONCLUSIONS: Unlike TAA-treated mice, BDL mice exhibited higher expression of the markers related with inflammation and necrosis, especially including p-JNK, while maintaining low levels of autophagic process. Therefore, obstructive cholestasis is characterized by higher p-JNK activation, which could be related with marked necrotic cell death resulting from extensive inflammation and little chance of compensatory autophagy.


Subject(s)
Autophagy , Cholestasis/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Cirrhosis, Experimental/immunology , Liver/pathology , Animals , Bile Ducts/surgery , Cholestasis/etiology , Cholestasis/pathology , Hepatocytes/immunology , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/immunology , Ligation , Liver/cytology , Liver/immunology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Male , Mice , Necrosis/immunology , Necrosis/pathology , Phosphorylation/immunology , Thioacetamide/toxicity
11.
J Gastroenterol Hepatol ; 34(8): 1432-1440, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30828861

ABSTRACT

BACKGROUND AND AIM: The pathological features of non-alcoholic steatohepatitis (NASH) have not been determined, so fundamental treatment has not been established. Adipose-tissue-derived stromal/stem cells (ADSCs) are beneficial for repair/regenerative therapy of impaired organs because of their immuno-modulatory capability. In this study, we assessed how liver damage progresses during the early development phase of the murine NASH model and investigated whether ADSCs are preventatively efficacious against the fibrosis progression of NASH. METHODS: C57BL/6J mice were fed with atherogenic high fat or high-fat diet 60 developing into NASH or simple steatosis. Their hepatic inflammatory cells (HICs) were analyzed by cDNA microarray. NASH mice were treated with ADSCs injected into spleen when hepatic inflammation was initially observed, and liver samples were analyzed. The effect of ADSCs on the mice hepatic stellate cell (HSC) line stimulated by recombinant IL-17 and HICs from NASH mice was analyzed. RESULTS: The gene expression features of HICs implicated as humoral cytokine mediators of lymphoid cells during NASH development, compared with a simple steatosis model. One of the featured cytokines was IL-17. The development of hepatic fibrosis was alleviated when NASH mice were treated with ADSCs as well as treated with anti-IL-17 antibody, and the frequency of IL-17-secreting HICs decreased. NASH-HICs enhanced proliferation of HSCs, in which proliferation was sensitive to IL-17 stimulation. The stimulatory effect of NASH-HICs on the activation of HSCs was attenuated by co-culture with ADSCs. CONCLUSION: ADSCs treatment prevented progression of NASH fibrosis by suppressing IL-17-mediated inflammation, which was associated with HSCs activation.


Subject(s)
Adipose Tissue/transplantation , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Liver Cirrhosis, Experimental/prevention & control , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Stem Cell Transplantation , Stem Cells/metabolism , Adipose Tissue/cytology , Animals , Cell Line , Cell Proliferation , Coculture Techniques , Diet, High-Fat , Disease Progression , Female , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Inflammation Mediators/immunology , Interleukin-17/immunology , Interleukin-17/pharmacology , Liver/immunology , Liver/pathology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Stem Cells/drug effects
12.
Immunopharmacol Immunotoxicol ; 41(1): 163-171, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30706740

ABSTRACT

Objective: Little is known about the exact underlying molecular mechanisms of the hepatoprotective effect of carvacrol against liver fibrosis. In the current study, we aimed to investigate the effect of carvacrol on the suppression of liver fibrosis progression via regulation of yes-associated protein (YAP) and transcriptional coactivators with a PDZ-binding motif (TAZ) and transforming growth factor beta (TGF-ß) pathway. Materials and methods: To fulfill our target, rats received carbon tetrachloride (CCl4) and carvacrol intraperitoneally, and orally, respectively for 10 weeks. Body weight, liver weight, serum biochemical parameters, hepatic hydroxyproline content, and histological changes were determined. Furthermore, gene expression of collagen and key elements of Hippo and TGF-ß pathways were analyzed and then the protein levels of YAP, TAZ, and TGF-ß were detected in liver tissue. Results: Carvacrol administration normalized liver and body weight, serum biochemical parameters and hepatic hydroxyproline in CCl4 treated rats. Also, carvacrol downregulated TAZ and TGF-ß signaling pathway at transcriptional levels. Furthermore, carvacrol decreased hepatic protein levels of TGF-ß, TAZ, and YAP. Low expression of TAZ and YAP were accompanied with inhibition of TGF-ß signaling pathway. Conclusion: Our data clearly revealed that carvacrol suppresses the progression of liver fibrosis via targeting of TAZ, YAP, and TGF-ß signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Liver Cirrhosis, Experimental/prevention & control , Monoterpenes/pharmacology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Acyltransferases , Animals , Carbon Tetrachloride , Cymenes , Disease Progression , Liver/drug effects , Liver/immunology , Liver/metabolism , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Male , Rats, Wistar , Signal Transduction , YAP-Signaling Proteins
13.
Gut ; 68(3): 533-546, 2019 03.
Article in English | MEDLINE | ID: mdl-29374630

ABSTRACT

OBJECTIVE: Liver injury impacts hepatic inflammation in part via Toll-like receptor (TLR) signalling. Triggering receptor expressed on myeloid cells 2 (TREM-2) modulates TLR4-mediated inflammation in bone marrow (BM)-derived macrophages but its function in liver injury is unknown. Here we hypothesised that the anti-inflammatory effects of TREM-2 on TLR signalling may limit hepatic injury. DESIGN: TREM-2 expression was analysed in livers of humans with various forms of liver injury compared with control individuals. Acute and chronic liver injury models were performed in wild type and Trem-2-/- mice. Primary liver cells from both genotypes of mice were isolated for in vitro experiments. RESULTS: TREM-2 was expressed on non-parenchymal hepatic cells and induced during liver injury in mice and man. Mice lacking TREM-2 exhibited heightened liver damage and inflammation during acute and repetitive carbon tetrachloride and acetaminophen (APAP) intoxication, the latter of which TREM-2 deficiency was remarkably associated with worsened survival. Liver damage in Trem-2-/- mice following chronic injury and APAP challenge was associated with elevated hepatic lipid peroxidation and macrophage content. BM transplantation experiments and cellular reactive oxygen species assays revealed effects of TREM-2 in the context of chronic injury depended on both immune and resident TREM-2 expression. Consistent with effects of TREM-2 on inflammation-associated injury, primary hepatic macrophages and hepatic stellate cells lacking TREM-2 exhibited augmented TLR4-driven proinflammatory responses. CONCLUSION: Our data indicate that by acting as a natural brake on inflammation during hepatocellular injury, TREM-2 is a critical regulator of diverse types of hepatotoxic injury.


Subject(s)
Liver Cirrhosis/metabolism , Liver/metabolism , Membrane Glycoproteins/physiology , Receptors, Immunologic/physiology , Acetaminophen , Aged , Animals , Carbon Tetrachloride , Case-Control Studies , Female , Hematopoietic Stem Cells/metabolism , Hepatocytes/metabolism , Humans , Inflammation Mediators/metabolism , Kupffer Cells/metabolism , Lipid Peroxidation/physiology , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Knockout , Middle Aged , Reactive Oxygen Species/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Toll-Like Receptor 4/physiology , Up-Regulation/physiology
14.
Naunyn Schmiedebergs Arch Pharmacol ; 392(3): 381-391, 2019 03.
Article in English | MEDLINE | ID: mdl-30535572

ABSTRACT

Liver fibrosis is a major health problem worldwide due to its serious complications including cirrhosis and liver cancer. 2-Methoxyestradiol (2-ME) is an end metabolite of estradiol with anti-proliferative, antioxidant, and anti-inflammatory activities. However, the protective role of 2-ME in liver fibrosis has not been fully investigated. The aim of this study was to determine the protective effect of 2-ME in carbon tetrachloride (CCl4)-induced liver fibrosis in mice. Animals were injected intraperitoneally with CCl4 twice weekly for 6 weeks. 2-ME 50 mg/kg or 100 mg/kg was administrated intraperitoneally every day over the same period. Our data showed that 2-ME reduced the extent of liver toxicity and fibrosis due to CCl4 exposure. It restored the elevated serum liver enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) levels and ameliorated oxidative status. In addition, 2-ME significantly reduced collagen deposition and alpha-smooth muscle actin (α-SMA) protein expressions. Furthermore, 2-ME markedly lowered macrophage infiltration and macrophage alternative activation marker chitinase-like molecules (CHI3L3/YM1). The results of this study indicate an important protective activity of 2-ME in liver fibrosis and highlight the role of macrophage recruitment and alternative activation as a possible target.


Subject(s)
2-Methoxyestradiol/pharmacology , 2-Methoxyestradiol/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Liver Cirrhosis, Experimental/drug therapy , Macrophages/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Actins/metabolism , Animals , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Collagen/metabolism , Liver/drug effects , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Macrophages/immunology , Male , Mice , Oxidative Stress/drug effects
15.
Chin J Integr Med ; 25(1): 51-58, 2019 Jan.
Article in English | MEDLINE | ID: mdl-26542309

ABSTRACT

OBJECTIVE: To investigate the potential antifibrotic mechanisms of Chinese medicine Ganshuang Granules (, GSG) and to provide clinical therapeutic evidence of its effects. METHODS: A cirrhotic mouse model was established by intraperitoneally injecting a mixture of CCl4 (40%) and oil (60%) at 0.2 mL per 100 g of body weight twice a week for 12 weeks. After 12-week modeling, GSG was intragastric administrated to the mice for 2 weeks, and the mice were divided into low-, medium- and high-dose groups at doses of 1, 2 and 4 g/(kg·day), respectively. Liver morphology changes were observed using Masson's trichrome staining and B-ultrasound. The levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and hyaluronic acid (HA) in serum were detected using an automatic biochemistry analyzer. The expressions of desmin, smooth muscle actin (SMA) and Foxp3 in liver were detected by immunoflfluorescence. The regulatory T cell (Treg) frequency was determined through flflow cytometry analysis. Collagen-I, SMA, IL-6, tumor necrosis factor α (TNF-α), interleukin (IL)-1ß and transforming growth factor ß1 (TGF-ß1) expression levels were measured using quantitative polymerase chain reaction (qPCR). RESULTS: Masson's staining result showed fewer pseudolobule structures and fibrous connective tissue in the GSG-treatment groups than in the spontaneous recovery group. Ultrasonography showed that GSG treatment reduced the number of punctate hyperechoic lesions in mice cirrhotic livers. The serum ALT, AST, HA levels were significantly ameliorated by GSG treatment (ALT: F=8.104, P=0.000; AST: F=7.078, P=0.002; and HA: F=7.621, P=0.001). The expression levels of collagen-I and SMA in the cirrhotic livers were also attenuated by GSG treatment (collagen-I: F=3.938, P=0.011; SMA: F=4.115, P=0.009). Tregs, which were elevated in the fibrotic livers, were suppressed by GSG treatment (F=8.268, P=0.001). The expressions of IL-6, TNF-α and IL-1ß increased, and TGF-ß levels decreased in the cirrhotic livers after GSG treatment (IL-6: F=5.457, P=0.004; TNF-α: F=6.023, P=0.002; IL-1ß: F=6.658, P=0.001; and TGF-ß1: F=11.239, P=0.000). CONCLUSIONS: GSG promoted the resolution/regression of cirrhosis and restored liver functions in part by suppressing Treg cell differentiation, which may be mediated by hepatic stellate cells.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Liver Cirrhosis, Experimental/drug therapy , T-Lymphocytes, Regulatory/drug effects , Animals , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/immunology , Male , Mice , Mice, Inbred BALB C
16.
Clin Sci (Lond) ; 132(24): 2565-2582, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30459204

ABSTRACT

Type 3 innate lymphoid cell (ILC3) has recently emerged as a crucial effector in inflammatory and fibrotic diseases. The present study was designed to determine the roles of ILC3 in liver fibrosis. By flow cytometry, we documented increased frequencies of peripheral ILC3 (Lin-CD127+CD117+CD294- lymphocytes) in patients, especially at the advanced stage of hepatitis B virus (HBV)-related chronic liver diseases, and demonstrated their correlations with disease progression. The in vitro fibrogenic effects by ILC3 were determined by co-culture experiments with LX-2 (a human hepatic stellate cell (HSC) line). The data indicate that pathogenic ILC3 can directly promote LX-2 fibrogenesis in non-contact manners by producing interleukin (IL)-17A and IL-22. Additionally, they had indirect fibrogenic effects by producing IL-22 to suppress interferon (IFN)-γ (a well-known anti-fibrotic cytokine) production by other immune cells. In carbon tetrachloride (CCl4)-induced wild-type mouse liver fibrosis models, we also documented significantly increased frequencies of both non-natural killer (NK) ILC (Lin-CD127+ lymphocytes) and ILC3 (Lin-CD127+RORγt+ lymphocytes) in liver and spleen specimens. Furthermore, the ILC3 from fibrotic mice contained more IL-17A+ILC3 and IL-22+ILC3 subsets than those from normal and less-fibrotic mice. The in vivo effects of ILC3 in liver fibrogenesis were further determined using RAG-1-/- mice with ILC depletion and further adoptive transfer of ILC3 from wild-type mice. The immunohistochemical staining of liver specimens showed the beneficial effects by ILC depletion and the detrimental effects by ILC3 transfer in CCl4-induced mouse liver fibrosis models. Collectively, ILC3 plays a pro-fibrotic role in liver fibrosis progression.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Hepatitis B, Chronic/immunology , Immunity, Innate , Liver Cirrhosis/immunology , Liver/immunology , Lymphocytes/immunology , Adult , Animals , Case-Control Studies , Cell Line , Cellular Microenvironment , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Coculture Techniques , Disease Progression , Female , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/metabolism , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/pathology , Homeodomain Proteins/genetics , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/immunology , Interleukins/metabolism , Liver/metabolism , Liver/pathology , Liver/virology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Lymphocytes/classification , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phenotype , Signal Transduction , Young Adult , Interleukin-22
17.
World J Gastroenterol ; 24(42): 4759-4772, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30479463

ABSTRACT

AIM: To investigate whether Yiguanjian decoction (YGJ) has an anti-liver cirrhotic effect and whether it regulates hepatic stem cell differentiation. METHODS: A rat model of liver cirrhosis was established via subcutaneous injection of carbon tetrachloride (CCl4) for 8 wk. From the beginning of the ninth week, the rats received 2-acetylaminofluorene (2-AAF) by oral gavage and a DLK-1+ fetal liver stem/progenitor cell (FLSPC) transplant or an FLSPC transplant in combination with YGJ treatment for 4 wk. In vitro, lipopolysaccharide (LPS)-activated macrophages were co-cultured with WB-F344 cells, and the differentiation of WB-F344 cells was observed in the presence and absence of YGJ treatment. RESULTS: FLSPC transplantation improved liver function and histopathology, and inhibited the activation of the non-canonical Wnt signaling pathway, while activating the canonical Wnt signaling pathway. YGJ enhanced the therapeutic effects of FLSPCs and also promoted the liver regeneration differentiation of FLSPCs into hepatocytes. In vitro, LPS-activated macrophages promoted the differentiation of WB-F344 cells into myofibroblasts, and the canonical Wnt signaling was inhibited while the non-canonical Wnt signaling was activated in WB-F344 cells. YGJ suppressed the activation of macrophages and then inhibited non-canonical Wnt signaling and promoted canonical Wnt signaling. CONCLUSION: YGJ enhances FLSPC-mediated repair of liver cirrhosis through regulation of macrophage activation state, and YGJ in combination with stem cell transplantation may be a suitable treatment for end-stage liver cirrhosis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Fetal Stem Cells/transplantation , Liver Cirrhosis, Experimental/therapy , Liver Regeneration/immunology , Macrophage Activation/drug effects , Animals , Carbon Tetrachloride/toxicity , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line , Coculture Techniques , Combined Modality Therapy/methods , Drugs, Chinese Herbal/therapeutic use , Hepatocytes/drug effects , Hepatocytes/immunology , Humans , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Male , Myofibroblasts , Rats , Rats, Inbred F344 , Rats, Wistar , Treatment Outcome , Wnt Signaling Pathway/drug effects
18.
World J Gastroenterol ; 24(36): 4178-4185, 2018 Sep 28.
Article in English | MEDLINE | ID: mdl-30271082

ABSTRACT

AIM: To reveal the protective mechanism of the combined use of vitamin D and puerarin in the progression of hepatic fibrosis induced by carbon tetrachloride (CCl4). METHODS: Eight-week-old male Wistar rats were randomly divided into a normal control group (C group), a CCl4 group (CCl4 group), a vitamin D group (V group), a puerarin group (P group), and a combined group of vitamin D and puerarin (V + P group), each of which contained ten rats. In this way, we built a rat model of CCl4-induced hepatic fibrosis with intervention by vitamin D, puerarin, or a combination of the two. After eight weeks, the mice were sacrificed to collect serum and liver specimens. Blood was collected to detect the hyaluronic acid (HA). We also measured hydroxyproline (Hyp) and prepared paraffin sections of liver. After Sirius red staining, the liver specimens were observed under a microscope. RT-PCR and western blot analysis were adopted to detect the mRNA and the protein levels of Collagen I, Collagen III, Wnt1, and ß-catenin in the liver tissues, respectively. RESULTS: Hepatic fibrosis was observed in the CCl4 group. In comparison, hepatic fibrosis was attenuated in the V, P, and V + P groups: the HA level in blood and the Hyp level in liver were reduced, and the mRNA levels of Collagen I, Collagen III, Wnt, and ß-catenin in liver were also decreased, as well as the protein levels of Wnt1 and ß-catenin. Among these groups, the V + P group demonstrated the greatest amelioration of hepatic fibrosis. CONCLUSION: The combined application of vitamin D and puerarin is capable of alleviating CCl4-induced hepatic fibrosis of rats. As to the mechanism, it is probably because the combined use is able to silence the Wnt1/ß-catenin pathway, suppress the activation of hepatic stellate cells, and reduce the secretion of collagen fibers, therefore improving the anti-hepatic fibrosis effect.


Subject(s)
Isoflavones/therapeutic use , Liver Cirrhosis, Experimental/drug therapy , Vasodilator Agents/therapeutic use , Vitamin D/therapeutic use , Wnt Signaling Pathway/drug effects , Animals , Carbon Tetrachloride/toxicity , Disease Progression , Drug Synergism , Drug Therapy, Combination/methods , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/immunology , Humans , Isoflavones/pharmacology , Liver/cytology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Male , Rats , Rats, Wistar , Treatment Outcome , Vasodilator Agents/pharmacology , Vitamin D/pharmacology , Wnt Signaling Pathway/immunology
19.
Biomed Pharmacother ; 106: 1332-1338, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119204

ABSTRACT

Yu Gan Long (YGL) is a Chinese traditional herbal medicine that has been used in the treatment of liver fibrosis for many years in clinical practice. However, its anti-hepatofibrotic mechanism has not been studied yet. In this study, the effect and mechanism of YGL in reducing liver fibrosis was demonstrated in vivo. Our results showed that liver fibrosis biomarkers collagen IV (Col IV), type III precollagen (PCIII), hyaluronuc acid (HA) and laminin (LN), were increased after CCl4 treatment and decreased by YGL. Among the liver fibrosis indicators, α-smooth muscle actin (α-SMA) was decreased by YGL in the CCl4-treated rats, while MMP2 and MMP9 was upregulated followed by TIMP1 downregulation. Proteins involved in liver fibrosis such as p-Smad2, p-Smad3 and Smad4 were down-regulated, while Smad7 protein was up-regulated by YGL after CCl4-induced liver damage. YGL also suppressed the increase of TGF-ß1, TNF-α, IL-1ß, IL-6, IL-4 and IL-17 A induced by CCl4 treatment, while promoted IFN-γ expression. Finally, the transcription factors ROR-γt and GATA3 were decreased, while T-bet was increased after YGL treatment. These results suggested that YGL attenuated CCl4-induced hepatic fibrosis by accelerating the extracellular matrix degradation, blocking the TGF-ß1/Smad signaling pathway and modulating the balance among IL-4, IL-17 A and IFN-γ, demonstrating YGL protective effect and its potential mechanisms in treating liver fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Drugs, Chinese Herbal/pharmacology , Extracellular Matrix/drug effects , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytoprotection , Dose-Response Relationship, Drug , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Phosphorylation , Proteolysis , Rats, Sprague-Dawley , Signal Transduction/drug effects
20.
Toxicol Sci ; 163(2): 397-408, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28204799

ABSTRACT

Epalrestat (EPS), an aldose reductase inhibitor, is widely prescribed to manage diabetic neuropathy. It is generally believed that EPS is beneficial to diabetic patients because it can protect endothelial cells, Schwann cells, or other neural cells from oxidative stress. However, several clinical studies revealed that EPS therapy led to liver dysfunction, which limited its clinical applications. Currently, the underlying mechanism by which EPS causes liver dysfunction is unknown. This study aimed to investigate the mechanism responsible for EPS-induced liver injury. In mouse liver, EPS 1) increased oxidative stress, indicated by increased expression of manganese superoxide dismutase, Ho-1, and Nqo1, 2) induced inflammation, indicated by infiltration of inflammatory cells, and induced expression of tumor necrosis factor-alpha, CD11b, and CD11c, as well as 3) predisposed to induce fibrosis, evidenced by increased mRNA and protein expression of early profibrotic biomarker genes procollagen I and alpha-smooth muscle actin, and by increased collagen deposition. In cultured mouse and human hepatoma cells, EPS treatment induced oxidative stress, decreased cell viability, and triggered apoptosis evidenced by increased Caspase-3 cleavage/activation. In addition, EPS increased mRNA and protein expression of cytoglobin in mouse liver, indicating that EPS activated hepatic stellate cells (HSCs). Furthermore, EPS treatment in cultured human HSCs increased cell viability. In summary, EPS administration induced oxidative stress and inflammation in mouse liver, and stimulated liver fibrogenesis. Therefore, cautions should be exercised during EPS therapy.


Subject(s)
Liver Cirrhosis, Experimental/chemically induced , Liver/drug effects , Oxidative Stress/drug effects , Rhodanine/analogs & derivatives , Thiazolidines/toxicity , Actins/genetics , Animals , CD11 Antigens/genetics , Cell Culture Techniques , Cell Line, Tumor , Collagen Type I/genetics , Humans , Inflammation , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/immunology , Male , Mice , Mice, Inbred C57BL , Rhodanine/toxicity , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...