Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.025
1.
Int J Biol Sci ; 20(7): 2422-2439, 2024.
Article En | MEDLINE | ID: mdl-38725842

Background & Aims: Reactive oxygen species (ROS) act as modulators triggering cellular dysfunctions and organ damage including liver fibrosis in which hepatic stellate cell (HSC) activation plays a key role. Previous studies suggest that microRNA-144 (miR-144) acts as a pro-oxidant molecule; however, whether and how miR-144 affects HSC activation and liver fibrosis remain unknown. Methods: Carbon tetrachloride (CCl4) and bile duct ligation (BDL)-induced experimental liver fibrosis models were used. Hepatic miR-144 expression was analyzed by miRNA in situ hybridization with RNAscope probe. The in vivo effects of silencing or overexpressing miR-144 were examined with an adeno-associated virus 6 (AAV6) carrying miR-144 inhibitor or mimics in fibrotic mouse experimental models. Results: In this study, we demonstrated that ROS treatment significantly upregulated miR-144 in HSCs, which further promoted HSC activation in vitro. Interestingly, miR-144 was preferentially elevated in HSCs of experimental liver fibrosis in mice and in human liver fibrotic tissues. Furthermore, in vivo loss or gain-of-function experiments via AAV6 carrying miR-144 antagomir or agomir revealed that blockade of miR-144 in HSCs mitigated, while overexpression of miR-144 in HSCs accelerated the development of experimental liver fibrosis. Mechanistically, SIN3 transcription regulator family member A (SIN3A), a transcriptional repressor, was identified to be the target of miR-144 in HSCs. MiR-144 downregulated Sin3A, and in line with this result, specific knockdown of Sin3a in HSCs remarkedly activated p38 MAPK signaling pathway to promote HSC activation, eventually exacerbating liver fibrosis. Conclusions: Oxidative stress-driven miR-144 fuels HSC activation and liver fibrogenesis by limiting the SIN3A-p38 axis. Thus, a specific inhibition of miR-144 in HSCs could be a novel therapeutic strategy for the treatment of liver fibrosis.


Hepatic Stellate Cells , Liver Cirrhosis , MicroRNAs , Oxidative Stress , Reactive Oxygen Species , Sin3 Histone Deacetylase and Corepressor Complex , p38 Mitogen-Activated Protein Kinases , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Humans , p38 Mitogen-Activated Protein Kinases/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Reactive Oxygen Species/metabolism , Male , Mice, Inbred C57BL , Repressor Proteins/metabolism , Repressor Proteins/genetics , Carbon Tetrachloride
2.
Int J Biol Sci ; 20(7): 2339-2355, 2024.
Article En | MEDLINE | ID: mdl-38725853

Chronic cholestatic damage is associated to both accumulation of cytotoxic levels of bile acids and expansion of adult hepatic progenitor cells (HPC) as part of the ductular reaction contributing to the regenerative response. Here, we report a bile acid-specific cytotoxic response in mouse HPC, which is partially impaired by EGF signaling. Additionally, we show that EGF synergizes with bile acids to trigger inflammatory signaling and NLRP3 inflammasome activation in HPC. Aiming at understanding the impact of this HPC specific response on the liver microenvironment we run a proteomic analysis of HPC secretome. Data show an enrichment in immune and TGF-ß regulators, ECM components and remodeling proteins in HPC secretome. Consistently, HPC-derived conditioned medium promotes hepatic stellate cell (HSC) activation and macrophage M1-like polarization. Strikingly, EGF and bile acids co-treatment leads to profound changes in the secretome composition, illustrated by an abolishment of HSC activating effect and by promoting macrophage M2-like polarization. Collectively, we provide new specific mechanisms behind HPC regulatory action during cholestatic liver injury, with an active role in cellular interactome and inflammatory response regulation. Moreover, findings prove a key contribution for EGFR signaling jointly with bile acids in HPC-mediated actions.


Bile Acids and Salts , ErbB Receptors , Inflammation , Mice, Inbred C57BL , Signal Transduction , Animals , Bile Acids and Salts/metabolism , ErbB Receptors/metabolism , Mice , Inflammation/metabolism , Stem Cells/metabolism , Liver/metabolism , Liver/pathology , Male , Proteomics , Macrophages/metabolism , Hepatic Stellate Cells/metabolism
3.
Commun Biol ; 7(1): 558, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730013

Whether and how the reactive oxygen species generated by hepatic stellate cells (HSCs) promote immune evasion of hepatocellular carcinoma (HCC) remains mysterious. Therefore, investigating the function of superoxide anion (O2•-), the firstly generated reactive oxygen species, during the immune evasion become necessary. In this work, we establish a novel in situ imaging method for visualization of O2•- changes in HSCs based on a new two-photon fluorescence probe TPH. TPH comprises recognition group for O2•- and HSCs targeting peptides. We observe that O2•- in HSCs gradually rose, impairing the infiltration of CD8+ T cells in HCC mice. Further studies reveal that the cyclin-dependent kinase 4 is deactivated by O2•-, and then cause the up-regulation of PD-L1. Our work provides molecular insights into HSC-mediated immune evasion of HCC, which may represent potential targets for HCC immunotherapy.


Hepatic Stellate Cells , Superoxides , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/immunology , Animals , Superoxides/metabolism , Mice , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Optical Imaging/methods , Immune Evasion , CD8-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Tumor Escape , Male
4.
PLoS One ; 19(5): e0302786, 2024.
Article En | MEDLINE | ID: mdl-38722973

A role for exportin 4 (XPO4) in the pathogenesis of liver fibrosis was recently identified. We sought to determine changes in hepatic XPO4 promoter methylation levels during liver fibrosis. The quantitative real-time RT-PCR technique was used to quantify the mRNA level of XPO4. Additionally, pyrosequencing was utilized to assess the promoter methylation status of XPO4. The methylation rate of the XPO4 promoter was significantly increased with fibrosis in human and mouse models, while XPO4 mRNA expression negatively correlated with methylation of its promoter. DNA methyltransferases (DNMTs) levels (enzymes that drive DNA methylation) were upregulated in patients with liver fibrosis compared to healthy controls and in hepatic stellate cells upon transforming growth factor beta (TGFß) stimulation. The DNA methylation inhibitor 5-Aza or specific siRNAs for these DNMTs led to restoration of XPO4 expression. The process of DNA methylation plays a crucial role in the repression of XPO4 transcription in the context of liver fibrosis development.


DNA Methylation , Karyopherins , Liver Cirrhosis , Promoter Regions, Genetic , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Humans , Karyopherins/genetics , Karyopherins/metabolism , Animals , Mice , Male , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mice, Inbred C57BL
5.
World J Gastroenterol ; 30(18): 2391-2396, 2024 May 14.
Article En | MEDLINE | ID: mdl-38764773

This editorial contains comments on the article by Zhao et al in print in the World Journal of Gastroenterology. The mechanisms responsible for hepatic fibrosis are also involved in cancerogenesis. Here, we recapitulated the complexity of the renin-angiotensin system, discussed the role of hepatic stellate cell (HSC) autophagy in liver fibrogenesis, and analyzed the possible implications in the development of hepatocarcinoma (HCC). Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers definitively contribute to reducing hepatic fibrogenesis, whereas their involvement in HCC is more evident in experimental conditions than in human studies. Angiotensin-converting enzyme 2 (ACE2), and its product Angiotensin (Ang) 1-7, not only regulate HSC autophagy and liver fibrosis, but they also represent potential targets for unexplored applications in the field of HCC. Finally, ACE2 overexpression inhibits HSC autophagy through the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway. In this case, Ang 1-7 acts binding to the MasR, and its agonists could modulate this pathway. However, since AMPK utilizes different targets to suppress the mTOR downstream complex mTOR complex 1 effectively, we still need to unravel the entire pathway to identify other potential targets for the therapy of fibrosis and liver cancer.


AMP-Activated Protein Kinases , Angiotensin-Converting Enzyme 2 , Autophagy , Carcinoma, Hepatocellular , Hepatic Stellate Cells , Liver Cirrhosis , Liver Neoplasms , Renin-Angiotensin System , Signal Transduction , TOR Serine-Threonine Kinases , Humans , TOR Serine-Threonine Kinases/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , AMP-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Autophagy/drug effects , Hepatic Stellate Cells/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/enzymology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Angiotensin I/metabolism , Animals , Peptidyl-Dipeptidase A/metabolism , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Peptide Fragments/metabolism , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin Receptor Antagonists/pharmacology , Liver/pathology , Liver/drug effects , Liver/metabolism
6.
PeerJ ; 12: e17356, 2024.
Article En | MEDLINE | ID: mdl-38766485

Background: Hepatic stellate cell (HSC) activation and hepatic fibrosis mediated biliary atresia (BA) development, but the underlying molecular mechanisms are poorly understood. This study aimed to investigate the roles of circRNA hsa_circ_0009096 in the regulation of HSC proliferation and hepatic fibrosis. Methods: A cellular hepatic fibrosis model was established by treating LX-2 cells with transforming growth factor ß (TGF-ß1). RNaseR and actinomycin D assays were performed to detect hsa_circ_0009096 stability. Expression of hsa_circ_0009096, miR-370-3p, and target genes was detected using reverse transcription-qPCR. Direct binding of hsa_circ_0009096 to miR-370-3p was validated using dual luciferase reporter assay. Cell cycle progression and apoptosis of LX-2 cells were assessed using flow cytometry. The alpha-smooth muscle actin (α-SMA), collagen 1A1 (COL1A1), and TGF beta receptor 2 (TGFBR2) protein levels in LX-2 cells were analyzed using immunocytochemistry and western blotting. Results: Hsa_circ_0009096 exhibited more resistance to RNase R and actinomycinD digestion than UTRN mRNA. Hsa_circ_0009096 expression increased significantly in LX-2 cells treated with TGF-ß1, accompanied by elevated α-SMA and COL1A1 expression. Hsa_circ_0009096 siRNAs effectively promoted miR-370-3p and suppressed TGFBR2 expression in LX-2 cells, mediated by direct association of hsa_circ_0009096 with miR-370-3p. Hsa_circ_0009096 siRNA interfered with the cell cycle progression, promoted apoptosis, and reduced α-SMA and COL1A1 expression in LX-2 cells treated with TGF-ß1. MiR-370-3p inhibitors mitigated the alterations in cell cycle progression, apoptosis, and α-SMA, COL1A1, and TGFBR2 expression in LX-2 cells caused by hsa_circ_0009096 siRNA. In conclusion, hsa_circ_0009096 promoted HSC proliferation and hepatic fibrosis during BA pathogenesis by accelerating TGFBR2 expression by sponging miR-370-3p.


Biliary Atresia , Cell Proliferation , Hepatic Stellate Cells , Liver Cirrhosis , MicroRNAs , RNA, Circular , Receptor, Transforming Growth Factor-beta Type II , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Biliary Atresia/pathology , Biliary Atresia/genetics , Biliary Atresia/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Apoptosis , Cell Line , Actins/metabolism , Actins/genetics , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics
7.
BMC Gastroenterol ; 24(1): 163, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745150

BACKGROUND: The liver regeneration is a highly complicated process depending on the close cooperations between the hepatocytes and non-parenchymal cells involving various inflammatory cells. Here, we explored the role of myeloid-derived suppressor cells (MDSCs) in the processes of liver regeneration and liver fibrosis after liver injury. METHODS: We established four liver injury models of mice including CCl4-induced liver injury model, bile duct ligation (BDL) model, concanavalin A (Con A)-induced hepatitis model, and lipopolysaccharide (LPS)-induced hepatitis model. The intrahepatic levels of MDSCs (CD11b+Gr-1+) after the liver injury were detected by flow cytometry. The effects of MDSCs on liver tissues were analyzed in the transwell co-culture system, in which the MDSCs cytokines including IL-10, VEGF, and TGF-ß were measured by ELISA assay and followed by being blocked with specific antibodies. RESULTS: The intrahepatic infiltrations of MDSCs with surface marker of CD11b+Gr-1+ remarkably increased after the establishment of four liver injury models. The blood served as the primary reservoir for hepatic recruitment of MDSCs during the liver injury, while the bone marrow appeared play a compensated role in increasing the number of MDSCs at the late stage of the inflammation. The recruited MDSCs in injured liver were mainly the M-MDSCs (CD11b+Ly6G-Ly6Chigh) featured by high expression levels of cytokines including IL-10, VEGF, and TGF-ß. Co-culture of the liver tissues with MDSCs significantly promoted the proliferation of both hepatocytes and hepatic stellate cells (HSCs). CONCLUSIONS: The dramatically and quickly infiltrated CD11b+Gr-1+ MDSCs in injured liver not only exerted pro-proliferative effects on hepatocytes, but also accounted for the activation of profibrotic HSCs.


CD11b Antigen , Liver Cirrhosis , Liver Regeneration , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells , Animals , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Mice , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Regeneration/physiology , CD11b Antigen/metabolism , Male , Disease Models, Animal , Liver/pathology , Liver/metabolism , Vascular Endothelial Growth Factor A/metabolism , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Concanavalin A , Ligation , Lipopolysaccharides , Interleukin-10/metabolism , Transforming Growth Factor beta/metabolism , Hepatic Stellate Cells/metabolism , Coculture Techniques , Hepatocytes/metabolism , Hepatocytes/pathology , Bile Ducts
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 652-659, 2024 Apr 20.
Article Zh | MEDLINE | ID: mdl-38708497

OBJECTIVE: To investigate the protective effect of arbutin against CCl4-induced hepatic fibrosis in mice and explore the underlying mechanisms. METHODS: Twenty-four C57BL/6 mice were randomly divided into control group, model group, and low- and high-dose arbutin treatment (25 and 50 mg/kg, respectively) groups. Mouse models of liver fibrosis were established by intraperitoneal injection of CCl4, and arbutin was administered daily via gavage for 6 weeks. After the treatments, serum biochemical parameters of the mice were tested, and liver tissues were taken for HE staining, Sirius Red staining and immunohistochemical staining. RT-qPCR was used to detect the mRNA levels of α-SMA, Pdgfb, Col1α1, Timp-1, Ccl2 and Tnf-a, and Western blotting was performed to detect α-SMA protein expression in the liver tissues. In the cell experiment, the effect of arbutin treatment for 24 h on THP-1 and RAW264.7 cell migration and recruitment was examined using Transwell migration assay and DAPI staining; The changes in protein levels of Akt, p65, Smad3, p-Akt, p-p65, p-Smad3 and α-SMA in arbutintreated LX-2 cells were detected with Western blotting. RESULTS: Arbutin treatment significantly lowered serum alanine aminotransferase and aspartate aminotransferase levels, alleviated liver tissue damage and collagen deposition, and reduced macrophage infiltration and α-SMA protein expression in the liver of the mouse models (P < 0.05 or 0.001). Arbutin treatment also significantly reduced CCl4-induced elevation of a-SMA, Pdgfb, Col1α1, Timp-1, Ccl2 and Tnf-a mRNA levels in mice (P < 0.05). In the cell experiment, arbutin treatment obviously inhibited migration and recruitment of THP-1 and RAW264.7 cells and lowered the phosphorylation levels of Akt, p65 and Smad3 and the protein expression level of α-SMA in LX-2 cells. CONCLUSION: Arbutin ameliorates liver inflammation and fibrosis in mice by inhibiting hepatic stellate cell activation via reducing macrophage recruitment and infiltration and suppressing activation of the Akt/NF-κB and Smad signaling pathways.


Arbutin , Liver Cirrhosis , Macrophages , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Mice , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Arbutin/pharmacology , Arbutin/therapeutic use , Macrophages/metabolism , Macrophages/drug effects , NF-kappa B/metabolism , Smad Proteins/metabolism , Carbon Tetrachloride , RAW 264.7 Cells , Cell Movement/drug effects , Liver/metabolism , Liver/pathology , Liver/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Male , Disease Models, Animal
9.
J Immunol Res ; 2024: 6343757, 2024.
Article En | MEDLINE | ID: mdl-38715844

This study aims to explore the influence of coinfection with HCV and HIV on hepatic fibrosis. A coculture system was set up to actively replicate both viruses, incorporating CD4 T lymphocytes (Jurkat), hepatic stellate cells (LX-2), and hepatocytes (Huh7.5). LX-2 cells' susceptibility to HIV infection was assessed through measurements of HIV receptor expression, exposure to cell-free virus, and cell-to-cell contact with HIV-infected Jurkat cells. The study evaluated profibrotic parameters, including programed cell death, ROS imbalance, cytokines (IL-6, TGF-ß, and TNF-α), and extracellular matrix components (collagen, α-SMA, and MMP-9). The impact of HCV infection on LX-2/HIV-Jurkat was examined using soluble factors released from HCV-infected hepatocytes. Despite LX-2 cells being nonsusceptible to direct HIV infection, bystander effects were observed, leading to increased oxidative stress and dysregulated profibrotic cytokine release. Coculture with HIV-infected Jurkat cells intensified hepatic fibrosis, redox imbalance, expression of profibrotic cytokines, and extracellular matrix production. Conversely, HCV-infected Huh7.5 cells exhibited elevated profibrotic gene transcriptions but without measurable effects on the LX-2/HIV-Jurkat coculture. This study highlights how HIV-infected lymphocytes worsen hepatic fibrosis during HCV/HIV coinfection. They increase oxidative stress, profibrotic cytokine levels, and extracellular matrix production in hepatic stellate cells through direct contact and soluble factors. These insights offer valuable potential therapies for coinfected individuals.


Bystander Effect , Coculture Techniques , Coinfection , Cytokines , HIV Infections , Hepacivirus , Hepatic Stellate Cells , Hepatitis C , Liver Cirrhosis , Humans , Hepatic Stellate Cells/metabolism , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/virology , HIV Infections/immunology , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatitis C/complications , Hepatitis C/immunology , Jurkat Cells , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Liver Cirrhosis/etiology , Cytokines/metabolism , Hepatocytes/metabolism , Hepatocytes/virology , HIV/physiology , Oxidative Stress , Cell Communication , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Extracellular Matrix/metabolism
10.
PLoS One ; 19(5): e0303296, 2024.
Article En | MEDLINE | ID: mdl-38753743

AIM: Metabolic dysfunction-associated steatohepatitis (MASH) is one of the most prevalent liver diseases and is characterized by steatosis and the accumulation of bioactive lipids. This study aims to understand the specific lipid species responsible for the progression of liver fibrosis in MASH. METHODS: Changes in bioactive lipid levels were examined in the livers of MASH mice fed a choline-deficient diet (CDD). Additionally, sphingosine kinase (SphK)1 mRNA, which generates sphingosine 1 phosphate (S1P), was examined in the livers of patients with MASH. RESULTS: CDD induced MASH and liver fibrosis were accompanied by elevated levels of S1P and increased expression of SphK1 in capillarized liver sinusoidal endothelial cells (LSECs) in mice. SphK1 mRNA also increased in the livers of patients with MASH. Treatment of primary cultured mouse hepatic stellate cells (HSCs) with S1P stimulated their activation, which was mitigated by the S1P receptor (S1PR)2 inhibitor, JTE013. The inhibition of S1PR2 or its knockout in mice suppressed liver fibrosis without reducing steatosis or hepatocellular damage. CONCLUSION: S1P level is increased in MASH livers and contributes to liver fibrosis via S1PR2.


Fatty Liver , Hepatic Stellate Cells , Liver Cirrhosis , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor) , Sphingosine-1-Phosphate Receptors , Sphingosine , Animals , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/etiology , Mice , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Humans , Sphingosine-1-Phosphate Receptors/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Male , Mice, Knockout , Mice, Inbred C57BL , Liver/metabolism , Liver/pathology , Choline Deficiency/complications , Choline Deficiency/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/genetics , Pyrazoles , Pyridines
11.
Cells ; 13(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38727301

Liver fibrosis, characterized by excessive extracellular matrix (ECM) deposition, can progress to cirrhosis and increases the risk of liver cancer. Hepatic stellate cells (HSCs) play a pivotal role in fibrosis progression, transitioning from a quiescent to activated state upon liver injury, wherein they proliferate, migrate, and produce ECM. Calcium signaling, involving the inositol 1,4,5-trisphosphate receptor (IP3R), regulates HSC activation. This study investigated the efficacy of a novel IP3R inhibitor, desmethylxestospongin B (dmXeB), in preventing HSC activation. Freshly isolated rat HSCs were activated in vitro in the presence of varying dmXeB concentrations. The dmXeB effectively inhibited HSC proliferation, migration, and expression of fibrosis markers without toxicity to the primary rat hepatocytes or human liver organoids. Furthermore, dmXeB preserved the quiescent phenotype of HSCs marked by retained vitamin A storage. Mechanistically, dmXeB suppressed mitochondrial respiration in activated HSCs while enhancing glycolytic activity. Notably, methyl pyruvate, dimethyl α-ketoglutarate, and nucleoside supplementation all individually restored HSC proliferation despite dmXeB treatment. Overall, dmXeB demonstrates promising anti-fibrotic effects by inhibiting HSC activation via IP3R antagonism without adverse effects on other liver cells. These findings highlight dmXeB as a potential therapeutic agent for liver fibrosis treatment, offering a targeted approach to mitigate liver fibrosis progression and its associated complications.


Cell Proliferation , Hepatic Stellate Cells , Inositol 1,4,5-Trisphosphate Receptors , Liver Cirrhosis , Animals , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Rats , Humans , Cell Proliferation/drug effects , Male , Rats, Sprague-Dawley , Cell Movement/drug effects
12.
Sci Rep ; 14(1): 11404, 2024 05 18.
Article En | MEDLINE | ID: mdl-38762616

Non-alcoholic steatohepatitis (NASH), caused by fat buildup, can lead to liver inflammation and damage. Elucidation of the spatial distribution of fibrotic tissue in the fatty liver in NASH can be immensely useful to understand its pathogenesis. Thus, we developed a novel serial section-3D (SS3D) technique that combines high-resolution image acquisition with 3D construction software, which enabled highly detailed analysis of the mouse liver and extraction and quantification of stained tissues. Moreover, we studied the underexplored mechanism of fibrosis progression in the fatty liver in NASH by subjecting the mice to a high-fat diet (HFD), followed by lipopolysaccharide (LPS) administration. The HFD/LPS (+) group showed extensive fibrosis compared with control; additionally, the area of these fibrotic regions in the HFD/LPS (+) group was almost double that of control using our SS3D technique. LPS administration led to an increase in Tnfα and Il1ß mRNA expression and the number of macrophages in the liver. On the other hand, transforming growth factor-ß1 (Tgfß1) mRNA increased in HFD group compared to that of control group without LPS-administration. In addition, COL1A1 levels increased in hepatic stellate cell (HSC)-like XL-2 cells when treated with recombinant TGF-ß1, which attenuated with recombinant latency-associated protein (rLAP). This attenuation was rescued with LPS-activated macrophages. Therefore, we demonstrated that fatty liver produced "latent-form" of TGF-ß1, which activated by macrophages via inflammatory cytokines such as TNFα and IL1ß, resulting in activation of HSCs leading to the production of COL1A1. Moreover, we established the effectiveness of our SS3D technique in creating 3D images of fibrotic tissue, which can be used to study other diseases as well.


Diet, High-Fat , Lipopolysaccharides , Liver Cirrhosis , Macrophages , Non-alcoholic Fatty Liver Disease , Transforming Growth Factor beta1 , Animals , Transforming Growth Factor beta1/metabolism , Mice , Macrophages/metabolism , Macrophages/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Diet, High-Fat/adverse effects , Male , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Macrophage Activation , Imaging, Three-Dimensional/methods , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Interleukin-1beta/metabolism
13.
Sci Signal ; 17(830): eade4335, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38564492

Serum ferritin concentrations increase during hepatic inflammation and correlate with the severity of chronic liver disease. Here, we report a molecular mechanism whereby the heavy subunit of ferritin (FTH) contributes to hepatic inflammation. We found that FTH induced activation of the NLRP3 inflammasome and secretion of the proinflammatory cytokine interleukin-1ß (IL-1ß) in primary rat hepatic stellate cells (HSCs) through intercellular adhesion molecule-1 (ICAM-1). FTH-ICAM-1 stimulated the expression of Il1b, NLRP3 inflammasome activation, and the processing and secretion of IL-1ß in a manner that depended on plasma membrane remodeling, clathrin-mediated endocytosis, and lysosomal destabilization. FTH-ICAM-1 signaling at early endosomes stimulated Il1b expression, implying that this endosomal signaling primed inflammasome activation in HSCs. In contrast, lysosomal destabilization was required for FTH-induced IL-1ß secretion, suggesting that lysosomal damage activated inflammasomes. FTH induced IL-1ß production in liver slices from wild-type mice but not in those from Icam1-/- or Nlrp3-/- mice. Thus, FTH signals through its receptor ICAM-1 on HSCs to activate the NLRP3 inflammasome. We speculate that this pathway contributes to hepatic inflammation, a key process that stimulates hepatic fibrogenesis associated with chronic liver disease.


Inflammasomes , Liver Diseases , Rats , Mice , Animals , Inflammasomes/genetics , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Hepatic Stellate Cells/metabolism , Ferritins/genetics , Ferritins/metabolism , Interleukin-1beta/metabolism , Inflammation/genetics , Inflammation/metabolism
14.
Cell Metab ; 36(5): 1030-1043.e7, 2024 May 07.
Article En | MEDLINE | ID: mdl-38670107

The mechanisms of hepatic stellate cell (HSC) activation and the development of liver fibrosis are not fully understood. Here, we show that deletion of a nuclear seven transmembrane protein, TM7SF3, accelerates HSC activation in liver organoids, primary human HSCs, and in vivo in metabolic-dysfunction-associated steatohepatitis (MASH) mice, leading to activation of the fibrogenic program and HSC proliferation. Thus, TM7SF3 knockdown promotes alternative splicing of the Hippo pathway transcription factor, TEAD1, by inhibiting the splicing factor heterogeneous nuclear ribonucleoprotein U (hnRNPU). This results in the exclusion of the inhibitory exon 5, generating a more active form of TEAD1 and triggering HSC activation. Furthermore, inhibiting TEAD1 alternative splicing with a specific antisense oligomer (ASO) deactivates HSCs in vitro and reduces MASH diet-induced liver fibrosis. In conclusion, by inhibiting TEAD1 alternative splicing, TM7SF3 plays a pivotal role in mitigating HSC activation and the progression of MASH-related fibrosis.


DNA-Binding Proteins , Liver Cirrhosis , TEA Domain Transcription Factors , Transcription Factors , TEA Domain Transcription Factors/metabolism , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Alternative Splicing , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Hepatic Stellate Cells/metabolism , Male , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/genetics , Mice, Knockout
15.
Biomolecules ; 14(4)2024 Mar 27.
Article En | MEDLINE | ID: mdl-38672422

Alcohol-associated liver disease (ALD) is a substantial cause of morbidity and mortality worldwide and represents a spectrum of liver injury beginning with hepatic steatosis (fatty liver) progressing to inflammation and culminating in cirrhosis. Multiple factors contribute to ALD progression and disease severity. Here, we overview several crucial mechanisms related to ALD end-stage outcome development, such as epigenetic changes, cell death, hemolysis, hepatic stellate cells activation, and hepatic fatty acid binding protein 4. Additionally, in this review, we also present two clinically relevant models using human precision-cut liver slices and hepatic organoids to examine ALD pathogenesis and progression.


Disease Progression , Liver Diseases, Alcoholic , Humans , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Animals , Liver/metabolism , Liver/pathology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Epigenesis, Genetic
16.
Tissue Eng Regen Med ; 21(4): 609-624, 2024 Jun.
Article En | MEDLINE | ID: mdl-38568409

BACKGROUND: Hepatic fibrosis (HF) is a common pathological feature of chronic hepatic diseases. We aimed to illuminate the significance of amniotic mesenchymal stem cells (AMSCs)-derived extracellular vesicles (AMSCs-EVs) in HF. METHODS: Human AMSCs-EVs were isolated and identified. HF mice were constructed and treated with EVs. The fibrosis was observed by staining experiments and Western blot (WB) assay. Alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), and hepatic hydroxyproline (Hyp) were detected to confirm liver function. For the in vitro experiments, human hepatic stellate cells were induced with transforming growth factor-ß and treated with EVs. To measure the degree of HF, the expression of alpha-smooth muscle actin (α-SMA) and Collagen I was detected by WB assay, and cell proliferation was detected by cell counting kit 8 assay. The levels of miR-200a, Zinc finger E-box binding homeobox 1 (ZEB1), and phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) were detected by WB and real-time quantitative polymerase chain reaction. The binding of ZEB1 to PIK3R3 and miR-200a to ZEB1 was analyzed by chromatin immunoprecipitation and dual luciferase assays to validate their relationships. RESULTS: Human AMSCs and AMSCs-EVs were obtained. Serum ALT, AST, TBIL, and hepatic Hyp were increased, implying the fibrosis degree was aggravated in HF mice, which was decreased again after EV treatment. EVs inhibited HF degree by reducing α-SMA and Collagen I and promoting cell proliferation. AMSCs-EVs delivered miR-200a into hepatocytes, which up-regulated miR-200a expression, inhibited ZEB1 expression, and reduced its enrichment on the PIK3R3 promoter, therefore inhibiting PIK3R3 expression and alleviating HF. Overexpression of ZEB1 or PIK3R3 attenuated the anti-fibrotic effect of AMSCs-EVs. CONCLUSION: Human AMSCs-derived EVs mediated miR-200a delivery and inhibition of intracellular ZEB1/PIK3R3 axis to exert anti-fibrosis effects.


Extracellular Vesicles , Liver Cirrhosis , Mesenchymal Stem Cells , MicroRNAs , Zinc Finger E-box-Binding Homeobox 1 , Animals , Liver Cirrhosis/therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Humans , Mice , Zinc Finger E-box-Binding Homeobox 1/metabolism , Hepatic Stellate Cells/metabolism , Cell Proliferation , Male , Mice, Inbred C57BL
17.
Environ Toxicol ; 39(6): 3760-3771, 2024 Jun.
Article En | MEDLINE | ID: mdl-38558500

Liver fibrosis is an invertible pathophysiologic process featured by excessive accumulation of extracellular matrix (ECM) which injures liver cells and activates hepatic stellate cells (HSCs). Besides, inducing ferroptosis in activated HSCs can alleviate liver fibrosis. LncRNAs modulate ferroptosis in activated HSCs and ECM deposition in liver fibrosis. However, the role of lncRNA FRMD6-AS1 in liver fibrosis is not discovered. In this study, lncRNA FRMD6-AS1 was dramatically up-regulated in activated HSCs. Knockdown of FRMD6-AS1 markedly increased iron ion, ROS and MDA levels, decreased GSH level, SLC7A11 and GPX4 protein expressions in activated HSCs. In addition, HSCs activation markers α-SMA and COL1α1 expressions were up-regulated in activated HSCs; knockdown of FRMD6-AS1 markedly down-regulated α-SMA and COL1α1 expressions in HSCs. Besides, lncRNA FRMD6-AS1 could interact with miR-491-5p, and negatively modulate miR-491-5p expression. USP13 was a target of miR-491-5p, and could be negatively modulated by miR-491-5p. Moreover, FRMD6-AS1 knockdown increased iron ion and ROS levels, decreased SLC7A11 and GPX4 protein expressions, facilitated HSCs viability, and up-regulated α-SMA and COL1α1 expressions via miR-491-5p/USP13 pathway. Finally, FRMD6-AS1 knockdown restored liver tissue structure and abrogated fibrosis in livers in a CCL4 liver fibrosis mouse model. Hence, lncRNA FRMD6-AS1/miR-491-5p/USP13 pathway repressed ferroptosis, promoted ECM deposition and facilitated liver fibrosis in vitro and in vivo models.


Ferroptosis , Hepatic Stellate Cells , Liver Cirrhosis , MicroRNAs , RNA, Long Noncoding , Ferroptosis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Mice , Mice, Inbred C57BL , Male , Carbon Tetrachloride/toxicity , Humans , Cell Line , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism
18.
Biochem Biophys Res Commun ; 712-713: 149958, 2024 Jun 18.
Article En | MEDLINE | ID: mdl-38640731

Hepatic stellate cells (HSCs) perform a significant function in liver regeneration (LR) by becoming active. We propose to investigate if activated HSCs enhance glycolysis via PFKFB3, an essential glycolytic regulator, and whether targeting this pathway could be beneficial for LR. The liver and isolated HSCs of mice subjected to 2/3 partial hepatectomy (PHx) exhibited a significant rise in PFKFB3 expression, as indicated by quantitative RT-PCR analyses and Western blotting. Also, the primary HSCs of mice subjected to PHx have a significant elevation of the glycolysis level. Knocking down PFKFB3 significantly diminished the enhancement of glycolysis by PDGF in human LX2 cells. The hepatocyte proliferation in mice treated with PHx was almost completely prevented when the PFKFB3 inhibitor 3PO was administered, emerging that PFKFB3 is essential in LR. Furthermore, there was a decline in mRNA expression of immediate early genes and proinflammatory cytokines. In terms of mechanism, both the p38 MAP kinase and ERK1/2 phosphorylation in LO2 cells and LO2 proliferation were significantly reduced by the conditioned medium (CM) obtained from LX2 cells with either PFKFB3 knockdown or inhibition. Compared to the control group, isolated hepatocytes from 3PO-treated mice showed decreased p38 MAP kinase and ERK1/2 phosphorylation and proliferation. Thus, LR after PHx involves the activation of PFKFB3 in HSCs, which enhances glycolysis and promotes lactate production, thereby facilitating hepatocyte proliferation via the p38/ERK MAPK signaling pathway.


Cell Proliferation , Glycolysis , Hepatic Stellate Cells , Liver Regeneration , Mice, Inbred C57BL , Phosphofructokinase-2 , Phosphofructokinase-2/metabolism , Phosphofructokinase-2/genetics , Animals , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/cytology , Humans , Mice , Male , Cell Line , Hepatectomy , Cells, Cultured , Liver/metabolism
19.
Sci Transl Med ; 16(744): eadk6213, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38657025

The Fontan operation is the current standard of care for single-ventricle congenital heart disease. Individuals with a Fontan circulation (FC) exhibit central venous hypertension and face life-threatening complications of hepatic fibrosis, known as Fontan-associated liver disease (FALD). The fundamental biology and mechanisms of FALD are little understood. Here, we generated a transcriptomic and epigenomic atlas of human FALD at single-cell resolution using multiomic snRNA-ATAC-seq. We found profound cell type-specific transcriptomic and epigenomic changes in FC livers. Central hepatocytes (cHep) exhibited the most substantial changes, featuring profound metabolic reprogramming. These cHep changes preceded substantial activation of hepatic stellate cells and liver fibrosis, suggesting cHep as a potential first "responder" in the pathogenesis of FALD. We also identified a network of ligand-receptor pairs that transmit signals from cHep to hepatic stellate cells, which may promote their activation and liver fibrosis. We further experimentally demonstrated that activins A and B promote fibrotic activation in vitro and identified mechanisms of activin A's transcriptional activation in FALD. Together, our single-cell transcriptomic and epigenomic atlas revealed mechanistic insights into the pathogenesis of FALD and may aid identification of potential therapeutic targets.


Fontan Procedure , Hepatic Stellate Cells , Hepatocytes , Liver Diseases , Single-Cell Analysis , Transcriptome , Humans , Fontan Procedure/adverse effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Transcriptome/genetics , Liver Diseases/pathology , Liver Diseases/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Epigenomics , Liver/pathology , Liver/metabolism , Multiomics
20.
Biochem Pharmacol ; 224: 116205, 2024 Jun.
Article En | MEDLINE | ID: mdl-38615918

Nitazoxanide is an FDA-approved antiprotozoal drug. Our previous studies find that nitazoxanide and its metabolite tizoxanide affect AMPK, STAT3, and Smad2/3 signals which are involved in the pathogenesis of liver fibrosis, therefore, in the present study, we examined the effect of nitazoxanide on experimental liver fibrosis and elucidated the potential mechanisms. The in vivo experiment results showed that oral nitazoxanide (75, 100 mg·kg-1) significantly improved CCl4- and bile duct ligation-induced liver fibrosis in mice. Oral nitazoxanide activated the inhibited AMPK and inhibited the activated STAT3 in liver tissues from liver fibrosis mice. The in vitro experiment results showed that nitazoxanide and its metabolite tizoxanide activated AMPK and inhibited STAT3 signals in LX-2 cells (human hepatic stellate cells). Nitazoxanide and tizoxanide inhibited cell proliferation and collagen I expression and secretion of LX-2 cells. Nitazoxanide and tizoxanide inhibited transforming growth factor-ß1 (TGF-ß1)- and IL-6-induced increases of cell proliferation, collagen I expression and secretion, inhibited TGF-ß1- and IL-6-induced STAT3 and Smad2/3 activation in LX-2 cells. In mouse primary hepatic stellate cells, nitazoxanide and tizoxanide also activated AMPK, inhibited STAT3 and Smad2/3 activation, inhibited cell proliferation, collagen I expression and secretion. In conclusion, nitazoxanide inhibits liver fibrosis and the underlying mechanisms involve AMPK activation, and STAT3 and Smad2/3 inhibition.


Antiprotozoal Agents , Nitro Compounds , Thiazoles , Animals , Mice , Thiazoles/pharmacology , Thiazoles/therapeutic use , Male , Humans , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Cell Line , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/chemically induced , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Smad3 Protein/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/prevention & control , Mice, Inbred C57BL , Smad2 Protein/metabolism
...