Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 378
Filter
Add more filters

Publication year range
1.
Gastroenterology ; 160(3): 847-862, 2021 02.
Article in English | MEDLINE | ID: mdl-33127392

ABSTRACT

BACKGROUND AND AIMS: The Hippo pathway and its downstream effectors YAP and TAZ (YAP/TAZ) are heralded as important regulators of organ growth and regeneration. However, different studies provided contradictory conclusions about their role during regeneration of different organs, ranging from promoting proliferation to inhibiting it. Here we resolve the function of YAP/TAZ during regeneration of the liver, where Hippo's role in growth control has been studied most intensely. METHODS: We evaluated liver regeneration after carbon tetrachloride toxic liver injury in mice with conditional deletion of Yap/Taz in hepatocytes and/or biliary epithelial cells, and measured the behavior of different cell types during regeneration by histology, RNA sequencing, and flow cytometry. RESULTS: We found that YAP/TAZ were activated in hepatocytes in response to carbon tetrachloride toxic injury. However, their targeted deletion in adult hepatocytes did not noticeably impair liver regeneration. In contrast, Yap/Taz deletion in adult bile ducts caused severe defects and delay in liver regeneration. Mechanistically, we showed that Yap/Taz mutant bile ducts degenerated, causing cholestasis, which stalled the recruitment of phagocytic macrophages and the removal of cellular corpses from injury sites. Elevated bile acids activated pregnane X receptor, which was sufficient to recapitulate the phenotype observed in mutant mice. CONCLUSIONS: Our data show that YAP/TAZ are practically dispensable in hepatocytes for liver development and regeneration. Rather, YAP/TAZ play an indirect role in liver regeneration by preserving bile duct integrity and securing immune cell recruitment and function.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/pathology , Liver Regeneration/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Bile Ducts/pathology , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Proliferation/genetics , Chemical and Drug Induced Liver Injury/complications , Cholestasis/etiology , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/pathology , Hippo Signaling Pathway , Humans , Liver/drug effects , Liver/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , YAP-Signaling Proteins
2.
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
3.
Hepatology ; 74(5): 2791-2807, 2021 11.
Article in English | MEDLINE | ID: mdl-34170569

ABSTRACT

BACKGROUND AND AIMS: Hepatocellular dedifferentiation is emerging as an important determinant in liver disease progression. Preservation of mature hepatocyte identity relies on a set of key genes, predominantly the transcription factor hepatocyte nuclear factor 4α (HNF4α) but also splicing factors like SLU7. How these factors interact and become dysregulated and the impact of their impairment in driving liver disease are not fully understood. APPROACH AND RESULTS: Expression of SLU7 and that of the adult and oncofetal isoforms of HNF4α, driven by its promoter 1 (P1) and P2, respectively, was studied in diseased human and mouse livers. Hepatic function and damage response were analyzed in wild-type and Slu7-haploinsufficient/heterozygous (Slu7+/- ) mice undergoing chronic (CCl4 ) and acute (acetaminophen) injury. SLU7 expression was restored in CCl4 -injured mice using SLU7-expressing adeno-associated viruses (AAV-SLU7). The hepatocellular SLU7 interactome was characterized by mass spectrometry. Reduced SLU7 expression in human and mouse diseased livers correlated with a switch in HNF4α P1 to P2 usage. This response was reproduced in Slu7+/- mice, which displayed increased sensitivity to chronic and acute liver injury, enhanced oxidative stress, and marked impairment of hepatic functions. AAV-SLU7 infection prevented liver injury and hepatocellular dedifferentiation. Mechanistically we demonstrate a unique role for SLU7 in the preservation of HNF4α1 protein stability through its capacity to protect the liver against oxidative stress. SLU7 is herein identified as a key component of the stress granule proteome, an essential part of the cell's antioxidant machinery. CONCLUSIONS: Our results place SLU7 at the highest level of hepatocellular identity control, identifying SLU7 as a link between stress-protective mechanisms and liver differentiation. These findings emphasize the importance of the preservation of hepatic functions in the protection from liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Hepatocyte Nuclear Factor 4/metabolism , RNA Splicing Factors/metabolism , Acetaminophen/administration & dosage , Acetaminophen/toxicity , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Differentiation/genetics , Cell Line , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/pathology , Humans , Liver/cytology , Liver/drug effects , Liver/pathology , Male , Mice , Oxidative Stress/genetics , Promoter Regions, Genetic , Proteolysis , Transcriptional Activation
4.
Hepatology ; 73(2): 776-794, 2021 02.
Article in English | MEDLINE | ID: mdl-32380568

ABSTRACT

BACKGROUND AND AIMS: How Wnt signaling is orchestrated in liver regeneration and tumorigenesis remains elusive. Recently, we identified transmembrane protein 9 (TMEM9) as a Wnt signaling amplifier. APPROACH AND RESULTS: TMEM9 facilitates v-ATPase assembly for vesicular acidification and lysosomal protein degradation. TMEM9 is highly expressed in regenerating liver and hepatocellular carcinoma (HCC) cells. TMEM9 expression is enriched in the hepatocytes around the central vein and acutely induced by injury. In mice, Tmem9 knockout impairs hepatic regeneration with aberrantly increased adenomatosis polyposis coli (Apc) and reduced Wnt signaling. Mechanistically, TMEM9 down-regulates APC through lysosomal protein degradation through v-ATPase. In HCC, TMEM9 is overexpressed and necessary to maintain ß-catenin hyperactivation. TMEM9-up-regulated APC binds to and inhibits nuclear translocation of ß-catenin, independent of HCC-associated ß-catenin mutations. Pharmacological blockade of TMEM9-v-ATPase or lysosomal degradation suppresses Wnt/ß-catenin through APC stabilization and ß-catenin cytosolic retention. CONCLUSIONS: Our results reveal that TMEM9 hyperactivates Wnt signaling for liver regeneration and tumorigenesis through lysosomal degradation of APC.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Membrane Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Cell Nucleus/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Gene Knockout Techniques , HEK293 Cells , Hep G2 Cells , Humans , Leupeptins/pharmacology , Liver Neoplasms/genetics , Liver Regeneration , Lysosomes/drug effects , Lysosomes/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Proteolysis/drug effects , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
5.
FASEB J ; 35(10): e21918, 2021 10.
Article in English | MEDLINE | ID: mdl-34569648

ABSTRACT

The role of LOXL1 in fibrosis via mediating ECM crosslinking and stabilization is well established; however, the role of hepatic stellate cells (HSCs)-specific LOXL1 in the development of fibrosis remains unknown. We generated HSCs-specific Loxl1-depleted mice (Loxl1Gfap-cre mice) to investigate the HSCs-specific contribution of LOXL1 in the pathogenesis of fibrosis. Loxl1fl/fl mice were used as the control. Furthermore, we used RNA sequencing to explore the underlying changes in the transcriptome. Results of the sirius red staining, type I collagen immunolabeling, and hydroxyproline content analysis, coupled with the reduced expression of profibrogenic genes revealed that Loxl1Gfap-cre mice with CCl4 -induced fibrosis exhibited decreased hepatic fibrosis. In addition, Loxl1Gfap-cre mice exhibited reduced macrophage tissue infiltration by CD68-positive cells and decreased expression of inflammatory genes compared with the controls. RNA sequencing identified integrin α8 (ITGA8) as a key modulator of LOXL1-mediated liver fibrosis. Functional analyses showed that siRNA silencing of Itga8 in cultured fibroblasts led to a decline in the LOXL1 expression and inhibition of fibroblast activation. Mechanistic analyses indicated that LOXL1 activated the FAK/PI3K/AKT/HIF1a signaling pathway, and the addition of inhibitors of FAK or PI3K reversed these results via downregulation of LOXL1. Furthermore, HIF1a directly interacted with LOXL1 and upregulated its expression, indicating that LOXL1 can positively self-regulate by forming a positive feedback loop with the FAK/PI3K/AKT/HIF1a pathway. We demonstrated that HSCs-specific Loxl1 deficiency prevented fibrosis, inflammation and that ITGA8/FAK/PI3K/AKT/HIF1a was essential for the function and expression of LOXL1. Knowledge of this approach can provide novel mechanisms and targets to treat fibrosis in the future.


Subject(s)
Amino Acid Oxidoreductases/deficiency , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , 3T3 Cells , Amino Acid Oxidoreductases/biosynthesis , Amino Acid Oxidoreductases/genetics , Animals , Base Sequence , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/adverse effects , Female , Fibroblasts/metabolism , Focal Adhesion Kinase 1/metabolism , Hepatic Stellate Cells/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Cirrhosis/pathology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Up-Regulation
6.
Acta Pharmacol Sin ; 43(1): 133-145, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33758354

ABSTRACT

N-n-Butyl haloperidol iodide (F2) is a novel compound that has antiproliferative and antifibrogenic activities. In this study we investigated the therapeutic potential of F2 against liver fibrosis in mice and the underlying mechanisms. Two widely used mouse models of fibrosis was established in mice by injection of either carbon tetrachloride (CCl4) or thioacetamide (TAA). The mice received F2 (0.75, 1.5 or 3 mg·kg-1·d-1, ip) for 4 weeks of fibrosis induction. We showed that F2 administration dose-dependently ameliorated CCl4- or TAA-induced liver fibrosis, evidenced by significant decreases in collagen deposition and c-Jun, TGF-ß receptor II (TGFBR2), α-smooth muscle actin (α-SMA), and collagen I expression in the liver. In transforming growth factor beta 1 (TGF-ß1)-stimulated LX-2 cells (a human hepatic stellate cell line) and primary mouse hepatic stellate cells, treatment with F2 (0.1, 1, 10 µM) concentration-dependently inhibited the expression of α-SMA, and collagen I. In LX-2 cells, F2 inhibited TGF-ß/Smad signaling through reducing the levels of TGFBR2; pretreatment with LY2109761 (TGF-ß signaling inhibitor) or SP600125 (c-Jun signaling inhibitor) markedly inhibited TGF-ß1-induced induction of α-SMA and collagen I. Knockdown of c-Jun decreased TGF-ß signaling genes, including TGFBR2 levels. We revealed that c-Jun was bound to the TGFBR2 promoter, whereas F2 suppressed the binding of c-Jun to the TGFBR2 promoter to restrain TGF-ß signaling and inhibit α-SMA and collagen I upregulation. In conclusion, the therapeutic benefit of F2 against liver fibrosis results from inhibition of c-Jun expression to reduce TGFBR2 and concomitant reduction of the responsiveness of hepatic stellate cells to TGF-ß1. F2 may thus be a potentially new effective pharmacotherapy for human liver fibrosis.


Subject(s)
Haloperidol/analogs & derivatives , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Animals , Carbon Tetrachloride/administration & dosage , Dose-Response Relationship, Drug , Haloperidol/administration & dosage , Haloperidol/pharmacology , Hepatic Stellate Cells/metabolism , Injections, Intraperitoneal , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Structure-Activity Relationship , Thioacetamide/administration & dosage , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
7.
Hepatology ; 72(6): 2119-2133, 2020 12.
Article in English | MEDLINE | ID: mdl-32145072

ABSTRACT

BACKGROUND AND AIMS: Hepatic sinusoidal cells are known actors in the fibrogenic response to injury. Activated hepatic stellate cells (HSCs), liver sinusoidal endothelial cells, and Kupffer cells are responsible for sinusoidal capillarization and perisinusoidal matrix deposition, impairing vascular exchange and heightening the risk of advanced fibrosis. While the overall pathogenesis is well understood, functional relations between cellular transitions during fibrogenesis are only beginning to be resolved. At single-cell resolution, we here explored the heterogeneity of individual cell types and dissected their transitions and crosstalk during fibrogenesis. APPROACH AND RESULTS: We applied single-cell transcriptomics to map the heterogeneity of sinusoid-associated cells in healthy and injured livers and reconstructed the single-lineage HSC trajectory from pericyte to myofibroblast. Stratifying each sinusoidal cell population by activation state, we projected shifts in sinusoidal communication upon injury. Weighted gene correlation network analysis of the HSC trajectory led to the identification of core genes whose expression proved highly predictive of advanced fibrosis in patients with nonalcoholic steatohepatitis (NASH). Among the core members of the injury-repressed gene module, we identified plasmalemma vesicle-associated protein (PLVAP) as a protein amply expressed by mouse and human HSCs. PLVAP expression was suppressed in activated HSCs upon injury and may hence define hitherto unknown roles for HSCs in the regulation of microcirculatory exchange and its breakdown in chronic liver disease. CONCLUSIONS: Our study offers a single-cell resolved account of drug-induced injury of the mammalian liver and identifies key genes that may serve important roles in sinusoidal integrity and as markers of advanced fibrosis in human NASH.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Endothelial Cells/pathology , Gene Regulatory Networks , Liver Cirrhosis/genetics , Non-alcoholic Fatty Liver Disease/pathology , Animals , Biopsy , Capillaries/cytology , Capillaries/pathology , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Hepatic Veins/cytology , Hepatic Veins/pathology , Humans , Liver/blood supply , Liver/pathology , Liver Cirrhosis/pathology , Membrane Proteins/genetics , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA-Seq , Single-Cell Analysis
8.
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
9.
Alcohol Clin Exp Res ; 45(10): 2103-2117, 2021 10.
Article in English | MEDLINE | ID: mdl-34486129

ABSTRACT

BACKGROUND: Magnetic resonance imaging (MRI) and MRI-based elastography (MRE) are the most promising noninvasive techniques in assessing liver diseases. The purpose of this study was to evaluate an advanced multiparametric imaging method for staging disease and assessing treatment response in realistic preclinical alcohol-associated liver disease (ALD). METHODS: We utilized four different preclinical mouse models in our study: Model 1-mice were fed a fast-food diet and fructose water for 48 weeks to induce nonalcoholic fatty liver disease; Model 2-mice were fed chronic-binge ethanol (EtOH) for 10 days or 8 weeks to induce liver steatosis/inflammation. Two groups of mice were treated with interleukin-22 at different time points to induce disease regression; Model 3-mice were administered CCl4 for 2 to 4 weeks to establish liver fibrosis followed by 2 or 4 weeks of recovery; and Model 4-mice were administered EtOH plus CCl4 for 12 weeks. Mouse liver imaging biomarkers including proton density fat fraction (PDFF), liver stiffness (LS), loss modulus (LM), and damping ratio (DR) were assessed. Liver and serum samples were obtained for histologic and biochemical analyses. Ordinal logistic regression and generalized linear regression analyses were used to model the severity of steatosis, inflammation, and fibrosis, and to assess the regression of these conditions. RESULTS: Multiparametric models with combinations of biomarkers (LS, LM, DR, and PDFF) used noninvasively to predict the histologic severity and regression of steatosis, inflammation, and fibrosis were highly accurate (area under the curve > 0.84 for all). A three-parameter model that incorporates LS, DR, and ALT predicted histologic fibrosis progression (r = 0.84, p < 0.0001) and regression (r = 0.79, p < 0.0001) as measured by collagen content in livers. CONCLUSION: This preclinical study provides evidence that multiparametric MRI/MRE can be used noninvasively to assess disease severity and monitor treatment response in ALD.


Subject(s)
Elasticity Imaging Techniques/methods , Fatty Liver, Alcoholic/diagnostic imaging , Hepatitis, Alcoholic/diagnostic imaging , Liver Cirrhosis/diagnostic imaging , Liver Diseases, Alcoholic/diagnostic imaging , Multiparametric Magnetic Resonance Imaging/methods , Animals , Carbon Tetrachloride/administration & dosage , Collagen/analysis , Disease Models, Animal , Disease Progression , Ethanol/administration & dosage , Female , Interleukins/administration & dosage , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Sensitivity and Specificity , Interleukin-22
10.
J Gastroenterol Hepatol ; 36(3): 800-810, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32870526

ABSTRACT

BACKGROUND AND AIM: The incidence of non-alcoholic steatohepatitis (NASH)-related hepatocellular carcinoma (HCC) is progressively increasing. However, the pathophysiology and etiology of NASH progression to HCC are unknown. We hypothesized that steatosis was the key factor in NASH-related hepatocarcinogenesis and aimed to evaluate the effects of long-term liver X receptor (LXR) agonist stimulation on hepatic steatosis induced by a high-fat diet and oxidative stress. METHODS: We used an LXR agonist (T0901317) and CCl4 to induce hepatic steatosis and oxidative stress, respectively. C57BL/6 mice fed with a high-fat diet were treated with either T0901317 + CCl4 (T09 + CCl4 group) or CCl4 alone (CCl4 group). T0901317 (2.5 mg/kg) and CCl4 (0.1 mL/kg) were intraperitoneally administered twice weekly for 24 weeks. RESULTS: The liver-to-body weight ratio was significantly higher in the T09 + CCl4 group than in the CCl4 group. Mice in the T09 + CCl4 group exhibited abnormal lipid metabolism and NASH-like histopathological features. Additionally, all mice in the T09 + CCl4 group developed liver tumors diagnosed as well-differentiated HCC. The genes identified via microarray analysis were related to NASH and HCC development. CONCLUSIONS: By combining long-term LXR agonist stimulation with oxidative stress and a high-fat diet, we successfully reproduced liver conditions in mice similar to those in humans with NASH and progression to HCC. Our results provide new insight into NASH-related HCC progression and therapy.


Subject(s)
Carcinoma, Hepatocellular/etiology , Hydrocarbons, Fluorinated/adverse effects , Liver Neoplasms/etiology , Liver X Receptors/agonists , Non-alcoholic Fatty Liver Disease/complications , Oxidative Stress , Sulfonamides/adverse effects , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/adverse effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Progression , Hydrocarbons, Fluorinated/administration & dosage , Injections, Intraperitoneal , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Sulfonamides/administration & dosage
11.
Int J Med Sci ; 18(6): 1382-1389, 2021.
Article in English | MEDLINE | ID: mdl-33628094

ABSTRACT

Background: Severe hepatitis is a common cause of chronic or acute liver disease and autophagy might play an important role in cellular response to inflammation and injury. It has been reported that Ginsenoside-Rg1 (G-Rg1) has strong hepatoprotective effects for acute liver injury, but its protective mechanisms have not yet been elucidated. This study aims to explore the detailed molecular mechanisms of G-Rg1 on acute liver injury via autophagy. Methods: The role of G-Rg1 by autophagic induction was studied in the mouse model of acute liver injury which induced by carbon tetrachloride (CCl4). Liver function, inflammatory reaction and apoptosis were detected when autophagy has been inhibited by 3-MA or stimulated by RPA. MCC950 and ATP were applied to investigate the role of NLRP3 inflammasome in acute liver injury. The differential expression of NF-κB, NLRP3 inflammasome, caspase 1, caspase 3, IL-1ß, IL-18, LC3-I, LC3-II, Beclin-1, PINK1 and Parkin have been detected by the quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. Results: G-Rg1 could decrease ALT, AST, TNF-α, IL-1ß and IL-6 in mice with CCl4-induced acute liver injury. The change of autophagy and apoptosis after the treatment of 3-MA or RPA demonstrated that the autophagy played a key role in the protective effect of G-Rg1 in acute liver injury. The enhancement of G-Rg1 promoted-autophagy resulted in the significant decrease in NF-κB, NLRP3 inflammasome, caspase 1, caspase 3, IL-1ß and IL-18, which suggesting that NF-κB/NLRP3 inflammasome signaling pathway was associated with the autophagy induced by G-Rg1 in acute liver injury. Conclusion: G-Rg1 ameliorated acute liver injury via the autophagy, which may be related to NF-κB/NLRP3 inflammasome signaling pathway.


Subject(s)
Autophagy/drug effects , Ginsenosides/pharmacology , Inflammasomes/antagonists & inhibitors , Liver Failure, Acute/drug therapy , Protective Agents/pharmacology , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Disease Models, Animal , Ginsenosides/therapeutic use , Humans , Inflammasomes/metabolism , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/immunology , Liver Failure, Acute/pathology , Male , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protective Agents/therapeutic use , Signal Transduction/drug effects
12.
Med Sci Monit ; 27: e931427, 2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34366426

ABSTRACT

BACKGROUND Acute chemical liver injury needs to be further explored. The present study aimed to compare the effects of intraperitoneal injection with carbon tetrachloride on acute liver toxicity after 24 h in male and female Kunming mice. MATERIAL AND METHODS In this study, female and male mice were simultaneously divided into 3 different groups. Each group was treated differently, and after 24 h, blood samples were collected to check for changes in the activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), which were used to assess liver toxicity. Liver samples were used for hematoxylin-eosin staining, and periodic acid Schiff reagent staining was performed to detect the pathological changes of each group. The expression level of biomarker molecules in liver cells was also systematically analyzed. RESULTS Our results showed that, compared with male mice, female mice showed more serious damage: reduced glycogen and higher degree of necrosis, and the levels of heatshock protein 27 (HSP27), heat-shock protein 70 (HSP70), proliferating cell nuclear antigen (PCNA) and B cell lymphoma/lewkmia-2 (Bcl-2) were significantly lower than in the male group (P<0.05 or P<0.01), while the results of Bcl-2-associated X protein (Bax), cysteinyl aspartate specific proteinase 3 (Caspase3), and cytochrome P450 2E1 (CYP2E1) were the opposite (P<0.05 or P<0.01). CONCLUSIONS The findings from this study showed that, compared with male mice, at 24 h after CCl4 toxicity, female mice showed more severe changes of hepatocyte necrosis and PAS-positivity, with significantly reduced expression of HSP27, HSP70, PCNA, and Bcl-2, and significantly increased expression of Bax, caspase-3, and CYP2E1.


Subject(s)
Carbon Tetrachloride Poisoning/diagnosis , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/diagnosis , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride Poisoning/etiology , Carbon Tetrachloride Poisoning/pathology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Female , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Injections, Intraperitoneal , Liver/drug effects , Liver/pathology , Male , Mice , Necrosis/chemically induced , Necrosis/diagnosis , Severity of Illness Index , Sex Factors , Toxicity Tests, Acute/methods
13.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805788

ABSTRACT

Persistent chronic liver diseases increase the scar formation and extracellular matrix accumulation that further progress to liver fibrosis and cirrhosis. Nevertheless, there is no antifibrotic therapy to date. The ketogenic diet is composed of high fat, moderate to low-protein, and very low carbohydrate content. It is mainly used in epilepsy and Alzheimer's disease. However, the effects of the ketogenic diet on liver fibrosis remains unknown. Through ketogenic diet consumption, ß-hydroxybutyrate (bHB) and acetoacetate (AcAc) are two ketone bodies that are mainly produced in the liver. It is reported that bHB and AcAc treatment decreases cancer cell proliferation and promotes apoptosis. However, the influence of bHB and AcAc in hepatic stellate cell (HSC) activation and liver fibrosis are still unclear. Therefore, this study aimed to investigate the effect of the ketogenic diet and ketone bodies in affecting liver fibrosis progression. Our study revealed that feeding a high-fat ketogenic diet increased cholesterol accumulation in the liver, which further enhanced the carbon tetrachloride (CCl4)- and thioacetamide (TAA)-induced liver fibrosis. In addition, more severe liver inflammation and the loss of hepatic antioxidant and detoxification ability were also found in ketogenic diet-fed fibrotic mouse groups. However, the treatment with ketone bodies (bHB and AcAc) did not suppress transforming growth factor-ß (TGF-ß)-induced HSC activation, platelet-derived growth factor (PDGF)-BB-triggered proliferation, and the severity of CCl4-induced liver fibrosis in mice. In conclusion, our study demonstrated that feeding a high-fat ketogenic diet may trigger severe steatohepatitis and thereby promote liver fibrosis progression. Since a different ketogenic diet composition may exert different metabolic effects, more evidence is necessary to clarify the effects of a ketogenic diet on disease treatment.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Acetoacetates/pharmacology , Cholesterol/biosynthesis , Diet, Ketogenic/adverse effects , Liver Cirrhosis/metabolism , Liver/drug effects , 3-Hydroxybutyric Acid/biosynthesis , Acetoacetates/metabolism , Actins/genetics , Actins/metabolism , Animals , Becaplermin/pharmacology , Carbon Tetrachloride/administration & dosage , Catalase/genetics , Catalase/metabolism , Cell Proliferation/drug effects , Cholesterol/blood , Collagen Type I/genetics , Collagen Type I/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Desmin/genetics , Desmin/metabolism , Disease Progression , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture , Severity of Illness Index , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Thioacetamide/administration & dosage , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/pharmacology
14.
J Hepatol ; 72(4): 736-745, 2020 04.
Article in English | MEDLINE | ID: mdl-31786256

ABSTRACT

BACKGROUND & AIMS: Acute-on-chronic liver failure (ACLF) is a clinical syndrome defined by liver failure on pre-existing chronic liver disease. It is often associated with bacterial infection and high short-term mortality. Experimental models that fully reproduce ACLF are lacking, so too are effective pharmacological therapies for this condition. METHODS: To mimic ACLF conditions, we developed a severe liver injury model by combining chronic injury (chronic carbon tetrachloride [CCl4] injection), acute hepatic insult (injection of a double dose of CCl4), and bacterial infection (intraperitoneal injection of bacteria). Serum and liver samples from patients with ACLF or acute drug-induced liver injury (DILI) were used. Liver injury and regeneration were assessed to ascertain the potential benefits of interleukin-22 (IL-22Fc) administration. RESULTS: This severe liver injury model recapitulated some of the key features of clinical ACLF, including acute-on-chronic liver injury, bacterial infection, multi-organ injury, and high mortality. Liver regeneration in this model was severely impaired because of a shift from the activation of the pro-regenerative IL-6/STAT3 pathway to the anti-regenerative IFN-γ/STAT1 pathway. The impaired IL-6/STAT3 activation was due to the inability of Kupffer cells to produce IL-6; whereas the enhanced STAT1 activation was due to a strong innate immune response and subsequent production of IFN-γ. Compared to patients with DILI, patients with ACLF had higher levels of IFN-γ but lower liver regeneration. IL-22Fc treatment improved survival in ACLF mice by reversing the STAT1/STAT3 pathway imbalance and enhancing expression of many antibacterial genes in a manner involving the anti-apoptotic protein BCL2. CONCLUSIONS: Acute-on-chronic liver injury or bacterial infection is associated with impaired liver regeneration due to a shift from a pro-regenerative to an anti-regenerative pathway. IL-22Fc therapy reverses this shift and attenuates bacterial infection, thus IL-22Fc may have therapeutic potential for ACLF treatment. LAY SUMMARY: A mouse model combining chronic liver injury, acute hepatic insult, and bacterial infection recapitulates some of the key features of acute-on-chronic liver failure (ACLF) in patients. Both fibrosis and bacterial infection contribute to the impaired regenerative capacity of the liver in patients with ACLF. Herein, we show that IL-22Fc therapy improves ACLF by reprogramming impaired regenerative pathways and attenuating bacterial infection. Thus, it may have therapeutic potential for patients with ACLF.


Subject(s)
Acute-On-Chronic Liver Failure/blood , Acute-On-Chronic Liver Failure/drug therapy , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/drug therapy , Interleukins/administration & dosage , Klebsiella Infections/drug therapy , Klebsiella pneumoniae , Liver Regeneration/drug effects , Acute Disease , Acute-On-Chronic Liver Failure/chemically induced , Acute-On-Chronic Liver Failure/microbiology , Adult , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/adverse effects , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Female , Hepatocytes/metabolism , Humans , Klebsiella Infections/microbiology , Kupffer Cells/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Treatment Outcome , Interleukin-22
15.
Apoptosis ; 25(5-6): 412-425, 2020 06.
Article in English | MEDLINE | ID: mdl-32409930

ABSTRACT

Liver injury is the early stage of liver disease, which is caused by multiple factors. Baicalein has shown extensive bioactivity. But whether baicalein has a protective effect on liver injury has not been reported thus far. In this study, we aim to investigate the protective effects of baicalein on liver injury induced by oxidative stress. H2O2 and CCl4 were employed to establish liver injury models in vivo and in vitro, respectively. The protective effect of baicalein on oxidative stress-induced liver injury was evaluated by detecting the mitochondrial dynamics, the level of autophagy and apoptosis, the histopathology of liver, the indicators of liver function, and the level of oxidative stress in vitro and in vivo. March5 is the key regulator during liver injury induced by oxidative stress. March5 can ubiquitinate Drp1 and promote Drp1 degradation, then maintain the homeostasis of mitochondrial dynamics, keep the balance of autophagy, and reduce apoptosis. Baicalein is able to effectively reduce liver injury; it can contribute to the expression of March5 by regulating KLF4 during liver injury. These results indicate that baicalein plays a key role in salvaging liver from injury induced by oxidative stress via regulating the KLF4-March5-Drp1 signal pathway.


Subject(s)
Antioxidants/pharmacology , Carbon Tetrachloride/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/genetics , Flavanones/pharmacology , Hepatocytes/drug effects , Mitochondrial Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carbon Tetrachloride/administration & dosage , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dynamins/genetics , Dynamins/metabolism , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
16.
BMC Gastroenterol ; 20(1): 14, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31941457

ABSTRACT

BACKGROUND: To investigate the merit of T1rho relaxation for the evaluation of liver fibrosis, inflammatory activity, and liver injury monitoring in a carbon tetrachloride (CCl4)-induced rat model. METHODS: Model rats from CCl4-induced liver fibrosis (fibrosis group: n = 41; regression group: n = 20) and control (n = 11) groups underwent black blood T1rho magnetic resonance (MR) imaging (MRI). Injection of CCl4 was done twice weekly for up to 12 weeks in the fibrosis group and for up to 6 weeks in the regression group. MR scanning time points were at baseline and at 2, 4, 6, 8, 10 and 12 weeks after CCl4 injection in the fibrosis group and at baseline and at 2, 4, 6 (CCl4 withdrawal), 7, 8, 10 and 12 weeks in the regression group. RESULTS: In the fibrosis group, liver T1rho values increased gradually within week 8 and then decreased. In the regression group, T1rho values dropped gradually after the withdrawal of CCl4 and fell below those at baseline. The T1rho values at S0 were lower than those at any other stage (all P < 0.05). The T1rho values at G0 were significantly lower than those at any other grade, and G1 was lower than G2 (all P < 0.01). The T1rho values mildly correlated with fibrosis stages (r = 0.362) and moderately correlated with grades of inflammation (r = 0.568). The T1rho values of rats with the same inflammation grades showed no significant difference among different fibrosis stages, and the T1rho values at S3 showed a significant difference among different grades of inflammation (P = 0.024). Inflammation grade was an independent variable associated with T1rho values (P < 0.001). CONCLUSION: T1rho MRI can be used to monitor CCl4-induced liver injury, and inflammatory activity had a greater impact on liver T1rho values than fibrosis.


Subject(s)
Image Processing, Computer-Assisted/methods , Liver Cirrhosis/diagnostic imaging , Liver/diagnostic imaging , Magnetic Resonance Imaging/methods , Animals , Carbon Tetrachloride/administration & dosage , Disease Models, Animal , Inflammation , Liver/drug effects , Liver Cirrhosis/chemically induced , Rats
17.
Bioorg Chem ; 99: 103804, 2020 06.
Article in English | MEDLINE | ID: mdl-32272365

ABSTRACT

Many tests have shown cyclooxygenase-2 (COX-2) was closely related to the activation of hepatic stellate cells (HSCs), which further promoting the onset and development of hepatic fibrosis. According to these research findings, a series of glycyrrhetinic acid derivatives were designed and synthesized. Meanwhile, their anti-hepaticfibrotic activities were evaluated in vitro and in vivo. Firstly, in the tests of the cell models, all the compounds displayed anti-proliferative effect on the HSC-T6 activated by (transforming growth factor beta) TGF-ß1 (10 ng/mL). Among them, compounds 2 and 16 exhibited a stronger activity than the others, and their IC50 values were 17.6 µM and 30.3 µM, respectively; both of them were low toxicity to normal HSC-T6 cells and WI38 cells, and they inhibited the activated HSC-T6 cells proliferation by promoting apoptosis and resting them at the G0/G1 phase. Secondly, compounds 2 and 16 displayed strong inhibitory effect on activation of HSCs; they not only inhibited the expression of α-SMA and Col1 in the activated HSC-T6 cells, but also decreased the levels of COX-2, TGF-ß1 and (reactive oxygen species) ROS in a concentration-dependent manner; they down-regulated the levels of three biomarkers in the process of test, but this decrease did not change linearly with the action time of compound. Thirdly, for the rats which induced with (carbontetrachloride) CCl4, the symptoms of liver fibrosis in rats were significantly alleviated after successive administration the tested compound for 14d; the α-SMA level in liver tissue decreased in a concentration dependent manner; and the liver cell necrosis and the fat collagen fiber decreased significantly compared with the positive control group; furthermore, inflammatory infiltration was significantly lower than that of the control. This suggests the compounds possibly are candidates for hepatic fibrosis with promising application in clinic.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Glycyrrhetinic Acid/pharmacology , Liver Cirrhosis/drug therapy , Administration, Oral , Animals , Apoptosis/drug effects , Carbon Tetrachloride/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Cytokines/analysis , Dose-Response Relationship, Drug , Glycyrrhetinic Acid/chemical synthesis , Glycyrrhetinic Acid/chemistry , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Molecular Structure , Rats , Rats, Wistar , Structure-Activity Relationship
18.
Bioorg Chem ; 99: 103803, 2020 06.
Article in English | MEDLINE | ID: mdl-32251945

ABSTRACT

Many peroxisome proliferator-activated receptors (PPARs) agonists have been developed for the treatment of metabolic disorders, while several PPARs agonists were discontinued in clinical trials because of PPARγ related side effects. In order to increase the selectivity against PPARγ, we performed a structure-activity relationship study based on PPARα/γ/δ agonist MHY2013. These efforts eventually led to the identification of compound 4, a dual PPARα/δ agonist with considerable potencies on PPARα/δ and high selectivity against PPARγ. In the Western Diet and CCl4-induced non-alcoholic steatohepatitis model, compound 4 alleviates the hepatic steatosis, inflammation, and fibrosis. These results indicated that dual PPARα/δ agonist 4 might be a promising lead compound for further investigations.


Subject(s)
Benzimidazoles/pharmacology , Drug Discovery , Fatty Liver/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , PPAR alpha/agonists , PPAR gamma/agonists , Administration, Oral , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Carbon Tetrachloride/administration & dosage , Dose-Response Relationship, Drug , Fatty Liver/chemically induced , Fatty Liver/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Structure-Activity Relationship
19.
Digestion ; 101(5): 522-535, 2020.
Article in English | MEDLINE | ID: mdl-31600750

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a continuous diseases spectrum associated with obesity, type 2 diabetes, insulin resistance, and hyperlipidemia. Simple hepatic steatosis may progress to nonalcoholic steatohepatitis (NASH), even fibrosis and cirrhosis, and finally hepatocellular carcinoma. In recent years, NAFLD has become a public health concern with increasing prevalence. However, the mechanisms underlying the pathogenesis remain incompletely understood, and few effective therapeutic approaches are available. Summary and Key Messages: A myriad of different rodent models has been developed to elucidate pathophysiology of NAFLD/NASH and guide therapeutic strategy. To date, no single rodent model can display the whole disease spectrum and metabolic features associated with human NASH, but can imitate particular characteristics. In this paper, we review the most commonly used dietary, genetic, and chemical rodent models for NAFLD referring to their advantages and disadvantages. Also, we illustrate the status of latest treatment strategy using various NAFLD rodent models. We hope to provide critical guidance for researchers to select appropriate animal models.


Subject(s)
Disease Models, Animal , Liver Cirrhosis/prevention & control , Liver/pathology , Non-alcoholic Fatty Liver Disease/therapy , Animals , Antioxidants/therapeutic use , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cholesterol, Dietary/adverse effects , Combined Modality Therapy , Diet, Carbohydrate Loading/adverse effects , Diet, High-Fat/adverse effects , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Insulin/therapeutic use , Lipid Metabolism/genetics , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Mice, Obese , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Probiotics/administration & dosage , Rats , Rats, Transgenic , Streptozocin/administration & dosage , Streptozocin/toxicity , Tetracycline/administration & dosage , Tetracycline/toxicity
20.
BMC Vet Res ; 16(1): 308, 2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32843036

ABSTRACT

BACKGROUND: Portal hypertension is a severe complication caused by various chronic liver diseases. The standard methods for detecting portal hypertension (hepatic venous pressure gradient and free portal pressure) are available in only a few hospitals due to their technical difficulty and invasiveness; thus, non-invasive measuring methods are needed. This study aimed to establish and assess a novel model to calculate free portal pressure based on biofluid mechanics. RESULT: Comparison of each dog's virtual and actual free portal pressure showed that a biofluid mechanics-based model could accurately predict free portal pressure (mean difference: -0.220, 95% CI: - 0.738 to 0.298; upper limit of agreement: 2.24, 95% CI: 1.34 to 3.14; lower limit of agreement: -2.68, 95% CI: - 3.58 to - 1.78; intraclass correlation coefficient: 0.98, 95% CI: 0.96 to 0.99; concordance correlation coefficient: 0.97, 95% CI: 0.93 to 0.99) and had a high AUC (0.984, 95% CI: 0.834 to 1.000), sensitivity (92.3, 95% CI: 64.0 to 99.8), specificity (91.7, 95% CI: 61.5 to 99.8), positive likelihood ratio (11.1, 95% CI: 1.7 to 72.8), and low negative likelihood ratio (0.08, 95% CI: 0.01 to 0.6) for detecting portal hypertension. CONCLUSIONS: Our study suggests that the biofluid mechanics-based model was able to accurately predict free portal pressure and detect portal hypertension in canines. With further research and validation, this model might be applicable for calculating human portal pressure, detecting portal hypertensive patients, and evaluating disease progression and treatment efficacy.


Subject(s)
Dog Diseases/diagnosis , Hypertension, Portal/veterinary , Portal Pressure , Portal Vein/diagnostic imaging , Animals , Biomechanical Phenomena , Blood Flow Velocity , Carbon Tetrachloride/administration & dosage , Dog Diseases/diagnostic imaging , Dogs , Hypertension, Portal/chemically induced , Hypertension, Portal/diagnosis , Hypertension, Portal/diagnostic imaging , Male , Models, Theoretical , Sensitivity and Specificity , Tomography, X-Ray Computed/veterinary , Ultrasonography, Doppler/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL