Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Heliyon ; 10(5): e26129, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38434258

ABSTRACT

Background: Yi-Qi-Jian-Pi Formula (YQJPF) is a herbal medicine that is used to treat patients with liver failure. However, scientific evidence supporting the treatment of hepatic fibrosis with YQJPF has not been forthcoming. The present study aimed to determine the mechanisms underlying the anti-fibrotic effects of YQJPF in mouse models of hepatic fibrosis. Methods: Mice were randomly assigned to control, hepatic fibrosis model, silymarin (positive treated), and low-, medium- and high-dose YQJPF (7.5, 15, and 30 g/kg, respectively) groups. Liver function, inflammatory cytokines, and oxygen stress were analyzed using ELISA kits. Sections were histopathologically stained with hematoxylin-eosin, Masson trichrome, and Sirius red. Macrophage polarization was measured by flow cytometry and immunofluorescence. Potential targets of YQJPF against hepatic fibrosis were analyzed by network pharmacology of Chinese herbal compound and the effects of YQJPF on the transforming growth factor-beta (TGF-ß)/Suppressor of Mothers against Decapentaplegic family member 3 (Smad3) signaling pathway were assessed using qRT-PCR and immunohistochemical staining. Finally, metagenomics and LC-MS/MS were used to detect the intestinal flora and metabolites of the mice, and an in-depth correlation analysis was performed by spearman correlation analysis. The data were compared by one-way ANOVA and least significant differences (LSDs) or ANOVA-Dunnett's T3 method used when no homogeneity was detected. Results: We induced hepatic fibrosis using CCl4 to establish mouse models and found that YQJPF dose-dependently increased body weight, improved liver function, and reversed hepatic fibrosis. Elevated levels of the pro-inflammatory factors IL-1ß, IL-6, and TNF-α in the model mice were substantially decreased by YQJPF, particularly at the highest dose. Levels of serum malondialdehyde and superoxide dismutase (SOD) activity were elevated and reduced, respectively. The malondialdehyde concentration decreased and SOD activity increased in the high-dose group. M1 polarized macrophages (CD86) in the mouse models were significantly decreased and M2 polarization was mildly decreased without significance. However, high-dose YQJPF increased the numbers of M2 macrophages and inhibited TGF-ß/Smad3 signaling. Metagenomic and non-targeted metabolomics detection results showed that YQJPF could regulate intestinal homeostasis, and Spearman correlation analysis showed that the abundance of Calditerrivibrio_nitroreducens was significantly negatively correlated with 18ß-glycyrrhetinic acid. It is suggested that Calditerrivibrio_nitroreducens may reduce the anti-fibrosis effect of licorice and other Chinese herbs by digesting 18ß-glycyrrhetinic acid. Conclusions: YQJPF can reverse liver fibrosis by inhibiting inflammation, suppressing oxidative stress, regulating the immunological response initiated by macrophages, inhibiting TGF-ß/Smad3 signaling and regulating intestinal flora homeostasis. Therefore, YQJPF may be included in clinical regimens to treat hepatic fibrosis.

2.
J Ethnopharmacol ; 317: 116683, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37315653

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Yi-Qi-Jian-Pi formula (YQJPF) is a commonly used traditional Chinese medicine (TCM) compound used to treat acute-on-chronic liver failure (ACLF) in China, but its specific mechanism of action has not been fully clarified. AIM OF THE STUDY: The aim of this study was to determine the effect of YQJPF on liver injury and hepatocyte pyroptosis in rats and further explore its molecular mechanism of action. MATERIALS AND METHODS: This study established carbon tetrachloride (CCl4)-, lipopolysaccharide (LPS)- and D-galactose (D-Gal)-induced in vivo models of ACLF in rats and in vitro LPS-induced hepatocyte injury models. Animal experiments were divided into the following groups: control, ACLF model, groups with different doses of YQJPF (5.4, 10.8, and 21.6 g/kg), and western medicine (methylprednisolone). There were 7 rats in the control group and 11 in the other groups. Serological, immunohistochemical, and pathological analyses were used to observe the effect of YQJPF on the liver of ACLF rats. The protective effect of YQJPF on hepatocytes was further verified by RT-qPCR, western blotting, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and other methods. RESULTS: YQJPF significantly improved liver injury in vivo and in vitro, which depended on the regulation of hepatocyte NLRP3/GSDMD-induced pyroptosis. In addition, we found that mitochondrial membrane potential and ATP production decreased after LPS treatment of hepatocytes, which suggested that YQJPF may improve mitochondrial energy metabolism disorders in hepatocytes. We administered a hepatocyte mitochondrial uncoupling agent, FCCP, to determine whether mitochondrial metabolic disorders affected cell pyroptosis. The results showed that the expression of IL-18, IL-1ß, and NLRP3 proteins increased significantly, indicating that the effect of this drug on hepatocyte pyroptosis may be related to mitochondrial metabolism disorders. We found that YQJPF significantly restored the tricarboxylic acid (TCA) cycle rate-limiting enzyme activity and affected the content of TCA metabolites. Furthermore, we revealed that the IDH2 gene, which plays a unique role in ACLF, is a key factor in the regulation of the mitochondrial TCA cycle and can be upregulated under the action of YQJPF. CONCLUSIONS: YQJPF can inhibit classical pyroptosis in hepatocytes by regulating TCA cycle metabolism, thus alleviating liver injury, and IDH2 may be a potential upstream regulatory target of YQJPF.


Subject(s)
Acute-On-Chronic Liver Failure , Rats , Animals , Acute-On-Chronic Liver Failure/drug therapy , Acute-On-Chronic Liver Failure/metabolism , Acute-On-Chronic Liver Failure/pathology , Pyroptosis , Citric Acid Cycle , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Lipopolysaccharides/pharmacology , Hepatocytes
3.
J Ethnopharmacol ; 308: 116276, 2023 May 23.
Article in English | MEDLINE | ID: mdl-36806340

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: A key event in the pathogenesis of acute-on-chronic liver failure (ACLF) is the imbalance in the systemic immune response; immunosuppression in patients with ACLF contributes to poor prognosis. The Yi-Qi-Jian-Pi formula (YQJPF) may improve T lymphocyte immune function in patients with ACLF. AIM OF THE STUDY: To investigate the immune mechanism of YQJPF in vivo and in vitro. MATERIALS AND METHODS: An ACLF rat model was established by injection of CCl4, lipopolysaccharide, and D-galactosamine. We examined the effect of different doses of YQJPF (6.43, 12.87, 25.74 g/kg) on liver injury and immune function in the ACLF rat model. Magnetic-activated cell sorting was used to sort the CD8+ T lymphocytes in the spleen for lymphocyte function detection. In primary CD8+ T lymphocytes and Jurkat cell lines, the expression of mitochondrial function and biogenesis and autophagy related markers were detected using molecular biological methods and flow cytometry analysis. RESULTS: YQJPF improved the peripheral blood lymphocyte count and proportion of CD8+ T lymphocytes in ACLF rats, increased pro-inflammatory factors (IL-2, IFN-λ, and TNF-α), and reduced anti-inflammatory factors (IL-10 and TGF ß1). YQJPF also improved metabolism and mitochondrial homeostasis in CD8+ T lymphocytes, alleviated lymphocyte immune dysfunction by promoting autophagy, upregulated mitochondrial biogenesis by promoting PGC-1α, NRF-1, and TFAM expression, and regulated the relationship between autophagy and mitochondrial biogenesis via PGC-1α. CONCLUSIONS: Our results suggest that YQJPF could improve immune function in a rat model of ACLF, possibly via affecting the homeostasis of lymphatic mitochondria in CD8+ T lymphocytes. YQJPF may enhance lymphocyte mitochondrial biosynthesis and promote lymphocyte autophagy. PGC-1α is a possible upstream regulatory target of YQJPF.


Subject(s)
Acute-On-Chronic Liver Failure , Rats , Animals , Acute-On-Chronic Liver Failure/pathology , Organelle Biogenesis , CD8-Positive T-Lymphocytes , Autophagy , Immunity
4.
J Clin Transl Hepatol ; 10(6): 1107-1116, 2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36381090

ABSTRACT

Background and Aims: Recognition of excessive activation of hepatic stellate cells (HSCs) in liver fibrosis prompted us to investigate the regulatory mechanisms of HSCs. We aimed to examine the role of O-GlcNAcylation modification of alanine, serine, cysteine transporter 2 (ASCT2) in HSCs and liver fibrosis. Methods: The expression of O-GlcNAcylation modification in fibrotic mice livers and activated HSCs was analyzed by western blotting. Immunoprecipitation was used to assess the interaction of ASCT2 and O-GlcNAc transferase (OGT). In addition, ASCT2 protein stability was assayed after cycloheximide (CHX) treatment. The O-GlcNAcylation site of ASCT2 was predicted and mutated by site-directed mutagenesis. Real-time PCR, immunofluorescence, kit determinations and Seahorse assays were used to clarify the effect of ASCT2 O-GlcNAcylation on HSC glutaminolysis and HSC activation. Western blotting, immunochemistry, and immunohistofluorescence were used to analyze the effect of ASCT2 O-GlcNAcylation in vivo. Results: We observed significantly increased O-GlcNAcylation modification of ASCT2. ASCT2 was found to interact with OGT to regulate ASCT2 stability. We predicted and confirmed that O-GlcNAcylation of ASCT2 at Thr122 site resulted in HSCs activation. We found Thr122 O-GlcNAcylation of ASCT2 mediated membrane trafficking of glutamine transport and attenuated HSC glutaminolysis. Finally, we validated the expression and function of ASCT2 O-GlcNAcylation after injection of AAV8-ASCT2 shRNA in CCl4-induced liver fibrosis mice in vivo. Conclusions: Thr122 O-GlcNAcylation regulation of ASCT2 resulted in stability and membrane trafficking-mediated glutaminolysis in HSCs and liver fibrosis. Further studies are required to assess its role as a putative therapeutic target.

5.
Front Genet ; 12: 777931, 2021.
Article in English | MEDLINE | ID: mdl-34899862

ABSTRACT

Background: Globally, hepatocellular carcinoma (HCC) is the sixth most frequent malignancy with a high incidence and a poor prognosis. Immune cell infiltration (ICI) underlies both the carcinogenesis and immunogenicity of tumors. However, a comprehensive classification system based on the immune features for HCC remains unknown. Methods: The HCC dataset from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts was used in this study. The ICI patterns of 571 patients were characterized using two algorithms: the patterns were determined based on the ICI using the ConsensusClusterPlus package, and principal component analysis (PCA) established the ICI scores. Differences in the immune landscape, biological function, and somatic mutations across ICI scores were evaluated and compared, followed by a predictive efficacy evaluation of ICI scores for immunotherapy by the two algorithms and validation using an external immunotherapy cohort. Results: Based on the ICI profile of the HCC patients, three ICI patterns were identified, including three subtypes having different immunological features. Individual ICI scores were determined; the high ICI score subtype was characterized by enhanced activation of immune-related signaling pathways and a significantly high tumor mutation burden (TMB); concomitantly, diminished immunocompetence and enrichment of pathways associated with cell cycle and RNA degradation were found in the low ICI score subtype. Taken together, our results contribute to a better understanding of an active tumor and plausible reasons for its poor prognosis. Conclusion: The present study reveals that ICI scores may serve as valid prognostic biomarkers for immunotherapy in HCC.

6.
Biochem Pharmacol ; 192: 114730, 2021 10.
Article in English | MEDLINE | ID: mdl-34400125

ABSTRACT

Activation of hepatic stellate cells (HSCs) is a central event in the pathogenesis of liver fibrosis and is often accompanied by the disappearance of lipid droplets (LDs). Although interference with LD metabolism can effectively reverse the activation of HSCs, there is currently no effective therapy for liver fibrosis. Our previous evidence indicates that long non-coding RNA (lncRNA)-H19 plays an essential role in LD metabolism of HSC. In this study, we investigated the potential molecular mechanism of dihydroartemisinin (DHA) inhibits LD metabolism and liver fibrosis by regulating H19-AMPK pathway. We found that DHA restores LDs content in activated HSCs via reducing the transcription of H19 driven by hypoxia inducible factor 1 subunit alpha (HIF1α) and inhibiting the lipid oxidation signal mediated by AMP-activated protein kinase (AMPK) phosphorylation. In vivo experiments, we have proved that DHA reduced the deposition of extracellular matrix (ECM) and reduce the level of liver fibrosis in CCl4-induced liver fibrosis of mice. In summary, our results emphasize the importance of H19 in liver fibrosis and the potential of DHA to regulate H19 to treat liver fibrosis, providing a new direction for the prevention and treatment of liver fibrosis.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Artemisinins/therapeutic use , Hepatic Stellate Cells/drug effects , Lipid Droplets/drug effects , Lipid Metabolism/drug effects , RNA, Long Noncoding/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Artemisinins/pharmacology , Cell Line , Hepatic Stellate Cells/metabolism , Humans , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred ICR , RNA, Long Noncoding/metabolism
7.
J Ethnopharmacol ; 280: 114411, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34265380

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Acute-on-chronic liver failure (ACLF) is a key complication of chronic hepatitis, with a relatively high mortality rate and limited treatment options, which dramatically threatens human lives. Yi-Qi-Jian-Pi formula (YQJPF) is a herbal compound commonly used to treat liver failure. AIM OF THE STUDY: The purpose of this research is to discuss the potential molecular biological effect and mechanism of YQJPF in ACLF. MATERIALS AND METHODS: In this study, we created a rat model of ACLF by CCl4-, LPS- and D-Galactosamine (D-Gal) and an in vitro model of LPS-induced hepatocyte damage. The specific components of YQJPF and potential mechanism were explored based on bioinformatics analyses. Furthermore, we verified the effect of YQJPF on ACLF using immunohistochemistry, RT-qPCR, western blotting, and flow cytometry. RESULTS: Our research demonstrated that, after YQJPF treatment, hepatocyte injury in rats was relieved. Bioinformatics analysis showed that PI3K/AKT, HIF-1, mitochondrial apoptosis pathways played prominent roles. YQJPF promoted HIF-1α protein expression and exerted protective effects against hypoxic injury, simultaneously reducing mitochondrial ROS production, suppressing hepatocyte apoptosis. Furthermore, we showed that YQJPF accelerates PI3K/AKT pathway activation, a known broad-spectrum inhibitor of PI3K. LY294002, which was used for reverse verification, suppressed the effect of YQJPF on hypoxic injury and ROS-mediated hepatocyte apoptosis. CONCLUSIONS: YQJPF ameliorates liver injury by suppressing hypoxic injury and ROS-mediated hepatocyte apoptosis by modulating the PI3K/AKT pathway.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Drugs, Chinese Herbal/therapeutic use , End Stage Liver Disease/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Chemical and Drug Induced Liver Injury/metabolism , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , End Stage Liver Disease/chemically induced , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Liver/metabolism , Male , Methylprednisolone/therapeutic use , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Random Allocation , Rats , Rats, Sprague-Dawley
8.
IUBMB Life ; 73(9): 1166-1179, 2021 09.
Article in English | MEDLINE | ID: mdl-34173707

ABSTRACT

AIMS: This study was designated to illustrate the underlying mechanisms of emodin anti-liver fibrosis via network pharmacology and experiment. METHODS: The TSMCP and Genecards database were applied to screen the relevant targets of emodin or liver fibrosis. The essential target was selected by using Cytoscape to analyze the topological network of potential targets. Furthermore, we constructed a preliminary molecule docking study to explore the binding site by Surflex-Dock suite SYBYL X 2.0. The DAVID database was selected for gene functional annotations and KEGG enrichment analysis. Moreover, we demonstrated the ameliorating effect of emodin on carbon tetrachloride (CCl4 )-induced liver injury in mice. We also verified the network predictions in vitro via various techniques. RESULTS: The collected results showed that 35 targets were related to emodin, and 6,198 targets were associated with liver fibrosis. The Venn analysis revealed that 17 intersection targets were correlated with emodin anti-liver fibrosis. The topological network analysis suggested that the p53 was the remarkable crucial target. Besides, the molecule docking results showed that emodin could directly interact with p53 by binding the active site residues ASN345, GLN331, and TYR347. Finally, KEGG pathway enrichment results indicated that essential genes were mainly enriched in mitogen-activated protein kinase (MAPK) signaling pathways. Moreover, our study confirmed that emodin alleviated CCl4 -induced liver injury in mice, inducing hepatic stellate cells (HSCs) apoptosis via regulating the p53/ERK/p38 axis. CONCLUSIONS: This study partially verified the network pharmacological prediction of emodin inducing HSCs cell apoptosis through the p53/ERK/p38 axis.


Subject(s)
Emodin/pharmacology , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Network Pharmacology/methods , Animals , Apoptosis/drug effects , Carbon Tetrachloride/toxicity , Caspases/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred ICR , Molecular Docking Simulation , Molecular Sequence Annotation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
9.
Front Pharmacol ; 12: 658811, 2021.
Article in English | MEDLINE | ID: mdl-33967802

ABSTRACT

Acute-on-chronic liver failure (ACLF) is described as a characteristic of acute jaundice and coagulation dysfunction. Effective treatments for ACLF are unavailable and hence are urgently required. We aimed to define the effect of Yi-Qi-Jian-Pi Formula (YQJPF) on liver injury and further examine the molecular mechanisms. In this study, we established CCl4-, LPS-, and d-galactosamine (D-Gal)-induced ACLF rat models in vivo and LPS- and D-Gal-induced hepatocyte injury models in vitro. We found that YQJPF significantly ameliorates liver injury in vivo and in vitro that is associated with the regulation of hepatocyte necroptosis. Specifically, YQJPF decreased expression of receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and pseudokinase mixed lineage kinase domain-like (MLKL) to inhibit the migration of RIPK1 and RIPK3 into necrosome. YQJPF also reduces the expression of inflammatory cytokines IL-6, IL-8, IL-1ß, and TNF-α, which were regulated by RIPK3 mediates cell death. RIPK1 depletion was found to enhance the protective effect of YQJPF. Furthermore, we showed that YQJPF significantly downregulates the mitochondrial reactive oxygen species (ROS) production and mitochondrial depolarization, with ROS scavenger, 4-hydroxy-TEMPO treatment recovering impaired RIPK1-mediated necroptosis and reducing the expression of IL-6, IL-8, IL-1ß, and TNF-α. In summary, our study revealed the molecular mechanism of protective effect of YQJPF on hepatocyte necroptosis, targeting RIPK1/RIPK3-complex-dependent necroptosis via ROS signaling. Overall, our results provided a novel perspective to indicate the positive role of YQJPF in ACLF.

10.
Medicine (Baltimore) ; 100(5): e24075, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33592861

ABSTRACT

BACKGROUND: Hepatitis B virus-related acute-on-chronic liver failure (HBV-ACLF) is a life-threatening syndrome with high mortality. Biomarkers are urgently needed to predict the prognosis of HBV-ACLF. Recent evidence suggests a key role for immune system in the pathology of HBV-ACLF. Here, we analyzed the correlation between peripheral blood T lymphocytes and the severity and prognosis in HBV-ACLF patients. METHOD: Sixty-six patients with HBV-ACLF received conventional medical treatments for 4 weeks. Twenty-five healthy subjects and 20 HBV patients were enrolled for comparison. We determined white blood cell count, lymphocytes, CD3+, CD4+ and CD8+ T cells, and CD4+CD25+ Treg cells in the blood of all subjects. Their associations with laboratory parameters before or after treatments were statistically analyzed. RESULT: The results showed that compare normal subjects and chronic hepatitis B patients, HBV-ACLF patients had significantly increased white blood count, CD4+ T cells and decreased lymphocytes, CD3+ T cells, and Treg cells. Correlation analysis showed that white blood cell, lymphocytes, and peripheral T lymphocytes were correlated with prothrombin activity (PTA) and model for end-stage liver disease (MELD) scores. After treatment, white blood cell, lymphocytes, and peripheral T lymphocytes were also correlated with PTA and MELD scores. Additionally, total bilirubin (TBIL), alanine aminotransferase (ALT), international standard ratio (INR), MELD, and white blood cell count were potential prognostic criteria for HBV-ACLF patients. CONCLUSION: HBV-ACLF patients had depletion and dysfunction of immune system. Changes of peripheral T lymphocytes were closely related to the pathogenesis and prognosis of disease. Our results may contribute to predict the severity of HBV-ACLF, and provide a prognosis response to improve the treatment of HBV-ACLF.


Subject(s)
Acute-On-Chronic Liver Failure , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hepatitis B, Chronic/complications , Lymphocyte Count/methods , T-Lymphocytes, Regulatory/immunology , Acute-On-Chronic Liver Failure/blood , Acute-On-Chronic Liver Failure/diagnosis , Acute-On-Chronic Liver Failure/etiology , Acute-On-Chronic Liver Failure/mortality , China/epidemiology , Correlation of Data , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Severity of Illness Index
11.
Toxicology ; 452: 152707, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33549628

ABSTRACT

Liver pathological angiogenesis is considered to be one of the key events in the development of liver fibrosis. Autophagy is a defense and stress regulation mechanism. However, whether autophagy regulates pathological angiogenesis in liver fibrosis is still questionable. Here, we aimed to study how curcumol regulated liver sinusoidal endothelial cells (LSECs) angiogenesis through autophagy. We found that curcumol (10, 20 and 40 µM) could inhibit the expression of angiogenesis markers in the LSECs. Importantly, we showed that curcumol might influence LSEC pathological angiogenesis by regulating autophagy level. Furthermore, we indicated that the transcription factor Krüppel-like factor 5 (KLF5) was considered as a key target for curcumol to regulate LSEC angiogenesis. Interestingly, we also suggested that autophagy was as a potential mechanism for curcumol to restrain KLF5 expression. Increased autophagy level could impair the suppression effect of curcumol on KLF5. Fascinatingly, our results indicated that curcumol inhibited autophagy and led to p62 accumulation, which might be a regulation mechanism of KLF5 degradation. Finally, in mice liver fibrosis model, we unanimously showed that curcumol (30 mg/kg) inhibited pathological angiogenesis by reducing LSEC autophagy level and suppressing KLF5 expression. Collectively, these results provided a deeper insight into the molecular mechanism of curcumol to inhibit LSEC pathological angiogenesis during liver fibrosis.


Subject(s)
Autophagy/drug effects , Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Neovascularization, Pathologic/metabolism , RNA-Binding Proteins/metabolism , Sesquiterpenes/therapeutic use , Animals , Autophagy/physiology , Capillaries/drug effects , Capillaries/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Kruppel-Like Transcription Factors/antagonists & inhibitors , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred ICR , Neovascularization, Pathologic/prevention & control , Sesquiterpenes/pharmacology
12.
Life Sci ; 264: 118696, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33157090

ABSTRACT

AIMS: Liver fibrosis is a difficult problem in the medical field. We previously reported that curcumol, a bioactive substance, may inhibit the pathological angiogenesis of liver sinusoidal endothelial cells (LSECs) and play a good anti-hepatic fibrosis effect. However, the mechanism of curcumol inhibiting angiogenesis in LSEC needs to be further clarified. Here, we focus on how curcumol inhibits LSEC angiogenesis in liver fibrosis. MATERIALS AND METHODS: Primary rat LSECs were cultured in vitro, and various molecular experiments including real-time PCR, western blot, immunofluorescence, tube formation assay and transwell migration assay were used to clarify the potential mechanism of curcumol. Carbon tetrachloride (CCl4) was applied to create a mouse liver fibrosis model. Blood and livers were taken to elucidate the efficacy of curcumol in vivo. KEY FINDINGS: We found that curcumol could effectively inhibit LSEC angiogenesis in vitro. Interestingly, this process may depend on curcumol's inhibition of the expression of transcription factor KLF5. Mice experiment also showed that curcumol could alleviate chronic liver injury by reducing KLF5 expression. In addition, we suggested that curcumol could reduce the production of mitochondrial ROS and improve mitochondrial morphology in LSEC. More importantly, we proved that curcumol could suppress KLF5-mediated LSEC angiogenesis by inhibiting ROS/ERK signaling. SIGNIFICANCE: We suggested that transcription factor KLF5 could be considered as a new target molecule of curcumol in improving liver fibrosis, and pointed out that curcumol targeted ROS/ERK-mediated KLF5 expression could inhibit LSEC angiogenesis. This provided a new theoretical basis for curcumol to ameliorate liver fibrosis.


Subject(s)
Endothelial Cells/pathology , Kruppel-Like Transcription Factors/metabolism , Liver/pathology , MAP Kinase Signaling System , Neovascularization, Pathologic/drug therapy , Reactive Oxygen Species/metabolism , Sesquiterpenes/therapeutic use , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Neovascularization, Pathologic/metabolism , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Sesquiterpenes/pharmacology
13.
Br J Pharmacol ; 178(11): 2246-2265, 2021 06.
Article in English | MEDLINE | ID: mdl-33085791

ABSTRACT

BACKGROUND AND PURPOSE: Portal hypertension is a lethal complication of cirrhosis. Its mechanism and therapeutic targets remain largely unknown. Hepatic stellate cell (HSC) contraction increases intrahepatic vascular resistance contributing to portal hypertension. We investigated how HSC contraction was regulated by Wnt signalling and the therapeutic implications. EXPERIMENTAL APPROACH: Liver tissues from cirrhotic patients were examined. Cirrhotic mice with genetic or pharmacological treatments were used for in vivo assessments, and their primary cells were isolated. Cellular functions and signalling pathways were analysed in human HSC-LX2 cells using real-time PCR, Western blotting, siRNA, luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation and site-directed mutagenesis. KEY RESULTS: Wnt/ß-catenin correlated with HSC contraction in human cirrhotic liver. Wnt3a stimulated Smo-independent Gli1 nuclear translocation followed by LARG-mediated RhoA activation leading to HSC contraction. Suppressor of fused (Sufu) negatively mediated Wnt3a-induced Gli1 nuclear translocation. Wnt/ß-catenin repressed transcription of Sufu dependent on ß-catenin/TCF4 interaction and TCF4 binding to Sufu promoter. Molecular simulation and site-directed mutagenesis identified the ß-catenin residues Lys312 and Lys435 critically involved in this interaction. TCF4 binding to the sequence CACACCTTCC at Sufu promoter was required for transrepression of Sufu. In cirrhotic mice, short-term liver-targeting ß-catenin deficiency or acute treatment with ß-catenin inhibitors reduced portal pressure via restriction of HSC contraction rather than inhibiting HSC activation. Long-term deficiency or treatments also ameliorated liver injury, fibrosis and inflammation. CONCLUSION AND IMPLICATIONS: Interaction between Wnt/ß-catenin and Smo-independent Gli1 pathways promoted HSC contraction via TCF4-dependent transrepression of Sufu. HSC-specific inhibition of ß-catenin may have therapeutic benefits for cirrhotic portal hypertension.


Subject(s)
Hepatic Stellate Cells , Hypertension, Portal , Wnt Signaling Pathway , Zinc Finger Protein GLI1 , Animals , Cell Line , Humans , Hypertension, Portal/pathology , Liver Cirrhosis/pathology , Mice , Transcription Factor 4
14.
Pharmacol Res ; 161: 105218, 2020 11.
Article in English | MEDLINE | ID: mdl-33007418

ABSTRACT

Endoplasmic reticulum (ER) stress is easily observed in chronic liver disease, which often causes accumulation of unfolded or misfolded proteins in the ER, leading to unfolded protein response (UPR). Regulating protein degradation is an integral part of UPR to relieve ER stress. The major protein degradation system includes the ubiquitin-proteasome system (UPS) and autophagy. All three arms of UPR triggered in response to ER stress can regulate UPS and autophagy. Accumulated misfolded proteins could activate these arms, and then generate various transcription factors to regulate the expression of UPS-related and autophagy-related genes. The protein degradation process regulated by UPR has great significance in many chronic liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, liver fibrosis, and hepatocellular carcinoma(HCC). In most instances, the degradation of excessive proteins protects cells with ER stress survival from apoptosis. According to the specific functions of protein degradation in chronic liver disease, choosing to promote or inhibit this process is promising as a potential method for treating chronic liver disease.


Subject(s)
Endoplasmic Reticulum Stress , Liver Diseases/metabolism , Liver/metabolism , Proteostasis , Animals , Autophagy , Chronic Disease , Endoplasmic Reticulum Stress/drug effects , Humans , Liver/drug effects , Liver/pathology , Liver Diseases/drug therapy , Liver Diseases/pathology , Proteolysis , Proteostasis/drug effects , Unfolded Protein Response
15.
Redox Biol ; 36: 101619, 2020 09.
Article in English | MEDLINE | ID: mdl-32863216

ABSTRACT

Ferroptosis is a recently discovered form of programmed cell death, but its regulatory mechanisms are not fully understood. In the current study, we reported that the BRD7-P53-SLC25A28 axis played a crucial role in regulating ferroptosis in hepatic stellate cells (HSCs). Upon exposure to ferroptosis inducers, bromodomain-containing protein 7 (BRD7) protein expression was remarkably increased through the inhibition of the ubiquitin-proteasome pathway. CRISPR/Cas9-mediated BRD7 knockout conferred resistance to HSC ferroptosis, whereas specific BRD7 plasmid-mediated BRD7 overexpression facilitated HSC ferroptosis. Interestingly, the elevated BRD7 expression exhibited to promote p53 mitochondrial translocation via direct binding with p53 N-terminal transactivation domain (TAD), which may be the underlying mechanisms for BRD7-enhanced HSC ferroptosis. Site-directed mutations of serine 392 completely blocked the binding of BRD7 to p53, and, in turn, prevented p53 mitochondrial translocation and HSC ferroptosis. Importantly, mitochondrial p53 interacted with solute carrier family 25 member 28 (SLC25A28) to form complex and enhanced the activity of SLC25A28, which could lead to the abnormal accumulation of redox-active iron and hyperfunction of electron transfer chain (ETC). SLC25A28 knockdown impaired BRD7-or p53-mediated ferroptotic events. In mice, erastin treatment ameliorated pathological damage of liver fibrosis through inducing HSC ferroptosis. HSC-specific blockade of BRD7-P53-SLC25A28 axis could abrogate erastin-induced HSC ferroptosis. Of note, we analyzed the effect of sorafenib on HSC ferroptosis in advanced fibrotic patients with hepatocellular carcinoma receiving sorafenib monotherapy. Attractively, BRD7 upregulation, p53 mitochondrial translocation, combination of SLC25A28 and p53, and ferroptosis induction occurred in primary human HSCs. Overall, these findings reveal novel signal transduction and regulatory mechanism of ferroptosis, and also suggest BRD7-P53-SLC25A28 axis as potential targets for liver fibrosis.


Subject(s)
Carcinoma, Hepatocellular , Cation Transport Proteins , Ferroptosis , Liver Neoplasms , Animals , Chromosomal Proteins, Non-Histone , Hepatic Stellate Cells , Humans , Mice , Tumor Suppressor Protein p53/genetics
16.
Free Radic Biol Med ; 160: 845-859, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32947011

ABSTRACT

BACKGROUND: Currently, the existing treatments have not cured the liver fibrosis thoroughly. Ferroptosis is a newly discovered way of cell death, which is closely related to many diseases. Previous studies have shown that ferroptosis plays an important role in the occurrence and development of liver fibrosis, but the further mechanism remains to be discovered. METHODS: LX-2 cells were used as the research object, fibrosis activation index was detected by Western blot, PCR and Immunofluorescence, ferroptosis was detected by kits, the binding and interaction between IRP2 (iron regulatory protein 2) and STUB1 (STIP1 homology and U-box containing protein 1) were detected by Immunoprecipitation and ubiquitin test, and IRP2 knockdown mice were constructed by interfering plasmid to verify the results of in vitro experiment. RESULT: Our research showed that ART (artemether) had a good anti-fibrosis effect in vivo and in vitro, and ferroptosis played an important role in this process. Further studies have found that ART could lead to the accumulation of IRP 2 a in hepatic stellate cell by inhibiting the ubiquitination of it, thus inducing the increase of iron in HSC (hepatic stellate cell), which could product a large number of ROS (reactive oxide species), resulting the occurrence of ferroptosis in cells. Our findings provided an experimental basis for ART to become a drug for the treatment of liver fibrosis. CONCLUSION: Our results show that IRP2-Iron-ROS axis is necessary for ART to induce ferroptosis in HSC and play an anti-fibrotic effect.


Subject(s)
Artemether , Ferroptosis , Iron Regulatory Protein 2 , Liver Cirrhosis , Animals , Hepatic Stellate Cells/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Mice
17.
Int Immunopharmacol ; 85: 106637, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32512269

ABSTRACT

More and more evidence showed that autophagy is an inflammation-related defense mechanism against a variety of diseases including liver fibrosis. However, the essential mechanisms remain poorly understood. In this study, we sought to elucidate the impact of Oroxylin A on autophagy and further to identify the potential mechanism of its anti-inflammatory activity. We found that Oroxylin A played a critical role in controlling inflammation in murine liver fibrosis. Moreover, Oroxylin A could inhibit the secretion of pro-inflammatory cytokines in activated hepatic stellate cell (HSCs). We previously reported that Oroxylin A can induce autophagy to alleviate the pathological changes of liver fibrosis and the activation of HSC. Here we further revealed that the inhibition of the PI3K/Akt/mTOR signaling was required for Oroxylin A to induce autophagy activation, which may be the underlying mechanism of the anti-inflammatory activity of Oroxylin A. Interestingly, mTOR overexpression completely impaired the Oroxylin A-mediated autophagy activation, and in turn, damaged the anti-inflammatory activity. Importantly, Oroxylin A inhibited PI3K/Akt/mTOR signaling by scavenging reactive oxygen species (ROS). ROS accumulation by buthionine sulfoximine (BSO) could abrogate the Oroxylin A-mediated ROS elimination, the inhibition of PI3K/Akt/mTOR signaling, and anti-inflammatory activities. Overall, our results provided reliable evidence for the molecular mechanism of Oroxylin A-mediated anti-fibrosis activity, and also identified a new target for drug therapy of liver fibrosis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Flavonoids/therapeutic use , Liver Cirrhosis/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Carbon Tetrachloride , Cells, Cultured , Cytokines/genetics , Flavonoids/pharmacology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
18.
Life Sci ; 255: 117818, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32445757

ABSTRACT

Activation of hepatic stellate cells (HSCs) is a central event in the pathogenesis of liver fibrosis and is characterized by the disappearance of lipid droplets. Although the exogenous supplementation of lipid droplet content can effectively reverse the activation of HSCs, the underlying molecular mechanisms are largely unknown. In our current study, we sought to investigate the role of lncRNA-H19 in the process of lipid droplets disappearance and to further examine the underlying molecular mechanisms. We found that the lncRNA-H19 level was increased in CCl4-induced fibrotic liver, which activated HSCs. Further research showed that hypoxia inducible factor-1α (HIF-1α) significantly increased lncRNA-H19 expression by binding to the lncRNA-H19 promoter at two hypoxia response element (HRE) sites located at 492-499 and 515-522 bp. Importantly, lncRNA-H19 knockdown markedly inhibited HSC activation and alleviated liver fibrosis, indicating that lncRNA-H19 may be a potential target for anti-fibrosis therapeutic approaches. Moreover, lncRNA-H19 knockdown could reverse the lipid droplet phenotype of activated HSCs, inhibiting the phosphorylated AMPKα-mediated lipid oxidation signaling pathway. The AMPK agonist AICAR promoted AMPKα phosphorylation and abrogated lipid droplets restoration in HSCs transfected with the lncRNA-H19 knockdown plasmid. Experimental molecular analysis showed that lncRNA-H19 triggered AMPKα to interact with LKB1 and resulted in AMPKα phosphorylation, which accelerating lipid droplets degradation and lipid oxidation. Taken together, our results highlighted the role of lncRNA-H19 in the metabolism of lipid droplets in HSCs, and revealed a new molecular target for alleviating liver fibrosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hepatic Stellate Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Cirrhosis/pathology , RNA, Long Noncoding/genetics , Animals , Cell Line , Gene Knockdown Techniques , Humans , Lipid Droplets/metabolism , Liver Cirrhosis/genetics , Male , Mice , Mice, Inbred ICR , Response Elements/genetics
19.
Int Immunopharmacol ; 84: 106470, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32304991

ABSTRACT

Activation of hepatic stellate cells (HSCs) is a pivotal event in liver fibrosis, characterized by enhanced retinoic acid signals. Although up-regulated retinoic acid signal responds further to maintain HSC activation, the underlying molecular mechanisms are largely unknown. In this study, we sought to investigate the role of lncRNA-H19 in regulation of retinoic acid signals, and to further examine the underlying mechanism in this molecular context. We found that lncRNA-H19 upregulation could enhance retinoic acid signals to induce HSC activation, whereas lncRNA-H19 knockdown completely disturbed retinoic acid signals. Moreover, the activation of retinoic acid signals impaired the lncRNA-H19 knockdown mediated HSC inactivation. Interestingly, we also found that enhanced retinoic acid signals by lncRNA-H19 was associated with a coordinate increase in retinol metabolism during HSC activation. Increased retinol metabolism contributed to obvious lipid droplet consumption. Importantly, we identified that alcohol dehydrogenase III (ADH3) was essential for lncRNA-H19 to enhance retinoic acid signals. The inhibition of ADH3 completely abrogated the lncRNA-H19 mediated retinoic acid signals and HSC activation. Of note, we identified dihydroartemisinin (DHA) as a natural inhibitor for lncRNA-H19. Treatment with DHA significantly decreased the expression of lncRNA-H19, reduced the expression of ADH3, blocked retinoic acid signals, and in turn, inhibited HSC activation. Overall, these results provided novel implications to reveal the molecular mechanism of increased retinoic acid signals during HSC activation, and identify lncRNA-H19/ADH3 pathway as a potential target for the treatment of liver fibrosis.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Hepatic Stellate Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tretinoin/metabolism , Animals , Artemisinins/pharmacology , Carbon Tetrachloride/adverse effects , Cell Line , Gene Knockdown Techniques , Lipid Metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred ICR , RNA, Long Noncoding/antagonists & inhibitors , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Vitamin A/metabolism
20.
Toxicology ; 440: 152475, 2020 07.
Article in English | MEDLINE | ID: mdl-32344006

ABSTRACT

OBJECTIVES: Curcumol, a guaiane-type sesquiterpenoid hemiketal extracted from the herb Rhizoma Curcumae, exhibits multiple-pharmacological activities. We previously reported that curcumol ameliorated hepatic fibrosis by inhibiting hepatic stellate cell (HSC) activation. In this study, we aimed to investigate the effect of curcumol on HSC migration and adhesion, and reveal its regulation mechanisms. MATERIALS AND METHODS: Cellular viability was determined by Cell Counting Kit-8. Cell migration was detected by boyden chamber and cell scratch experiment. Recombinant human periostin (rh POSTN) and adeno-associated viral (AAV)-GFP-periostin were used to achieve POSTN overexpression in vitro and in vivo, respectively. Nuclear factor kappa B (NF-κB)-p65 overexpression was achieved by using plasmid. ELISA was conducted to detect POSTN level. Immunohistochemistry, qRT-PCR, Western blotting, and immunofluorescence were performed to assess associated factor expression. RESULTS: Curcumol suppressed HSC migration and adhesion, and reduced the secretion and expression of POSTN. By gain of function POSTN in HSCs, using rh POSTN, we found that the inhibition of HSC migration and adhesion by curcumol depended on the decrease of POSTN. Besides, curcumol protection against chronic CCl4-caused hepatic fibrosis could be impaired by POSTN overexpression. Moreover, we showed that curcumol repressed NF-κB signaling and the production of pro-inflammatory factor. Importantly, curcumol down-regulation of POSTN was rescued by knock-in of NF-κB, as well as the inhibition of HSC migration and adhesion. CONCLUSION: These findings reveal the molecular mechanism of curcumol-reduced HSC migration and adhesion, by which points to the possibility of using curcumol based on NF-κB dependent POSTN for the treatment of fibrogenesis.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Hepatic Stellate Cells/drug effects , Sesquiterpenes/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Animals , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/prevention & control , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Count , Cell Line , Cell Movement/drug effects , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Down-Regulation/drug effects , Humans , Male , Mice , Mice, Inbred ICR , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...