Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.265
Filter
1.
J Ethnopharmacol ; 285: 114826, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34767833

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Indigofera linifolia (L.f.) Retz. is used in subcontinent for liver disorders, in wounds, febrile eruption and as diuretic. AIM OF STUDY: The current study evaluates the protective effects of the methanol extract of Indigofera linifolia (ILM) on CCl4-induced endoplasmic reticulum (ER) stress in liver of rat. METHODS: ILM was analyzed for phytochemical classes, total phenolic (TPC) and flavonoid content (TFC) as well as multidimensional in vitro antioxidant assays. Male (Sprague Dawley) rats were dispersed into seven groups (6 rats/group) receiving 0.9% saline (1 ml/kg bw), CCl4 (1 ml/kg bw) diluted in olive oil (3:7 v/v), silymarin (200 mg/kg bw) + CCl4 (30% v/v), ILM (150 mg/kg bw) + CCl4 (30% v/v), ILM (300 mg/kg bw) + CCl4 and ILM alone (either 150 mg/kg bw or 300 mg/kg bw). RESULTS: ILM extract was constituted of different phytochemical classes. Co-administration of ILM along with CCl4 to rat revert the level of alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP) and total bilirubin in blood serum and antioxidant parameters in liver. Further, CCl4 increased the level of ER stress markers and inflammatory mediators while decreased level of GCLC and Nrf-2 in liver tissues of rat. CCl4-induced histopathological variations were reduced with ILM co-administration in liver tissues. CONCLUSION: The results suggest that active phyto-constituents of I. linifolia might be responsible for its antioxidant, anti-inflammatory and gene-regulating activities.


Subject(s)
Carbon Tetrachloride Poisoning , Endoplasmic Reticulum Stress/drug effects , Glutamate-Cysteine Ligase/metabolism , Indigofera , Liver , NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Carbon Tetrachloride/adverse effects , Carbon Tetrachloride/metabolism , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Liver/drug effects , Liver/metabolism , Rats , Rats, Sprague-Dawley
2.
Life Sci ; 289: 120235, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34914932

ABSTRACT

AIMS: Liver fibrosis is a growing public health concern without effective medical treatment. Recent reports have indicated that inhibitors of apoptosis proteins (IAPs) were potential targets for idiopathic pulmonary fibrosis therapy. However, their roles have not been well identified in liver fibrosis. METHODS: The expression of IAPs were examined in human liver tissue and experimental mouse models. Liver fibrosis in CCl4-induced mouse models were investigated by Sirius red staining, RT-PCR, Western blotting after hepatocytes-specific cIAP2 knockout or IAPs inhibitor APG-1387 treatment. The underlying molecular mechanism of APG-1387 action was explored by apoptosis analysis, matrix metalloprotein 9 (MMP9) inhibition, neutrophils depletion, and CC Motif Chemokine Ligand 5 (CCL5) gene knockout in vitro and in vivo. FINDINGS: Our study showed that increased expression of cIAP2 was associated with liver fibrosis severity in liver tissues. Deletion of cIAP2 from hepatocytes or degrading cIAPs by APG-1387 ameliorated liver fibrosis induced by CCl4. APG-1387 treatment exhibited increased expression of MMP9 and resulted in higher ratio of MMP9 to tissue inhibitor of metalloproteinase-1. MMP9 was mainly derived from CCL5 chemotactic neutrophils. Further, MMP9 inhibition by CTT peptide, neutrophil depletion by Ly6G antibody or CCL5 deficiency blocked the anti-fibrotic effects of APG-1387 in vivo. SIGNIFICANCE: These results suggested that cIAPs, especially cIAP2, might play a novel role in the pathogenesis of liver fibrosis, and targeting cIAPs represented a promising therapeutic strategy for liver fibrosis by increasing MMP9 expression induced by CCL5 chemotactic neutrophils.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Carbon Tetrachloride Poisoning/metabolism , Gene Expression Regulation, Enzymologic , Liver Cirrhosis/metabolism , Matrix Metalloproteinase 9/biosynthesis , Neutrophils/metabolism , Animals , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/pathology , Gene Deletion , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Neutrophils/pathology
3.
Int J Mol Sci ; 22(24)2021 Dec 12.
Article in English | MEDLINE | ID: mdl-34948151

ABSTRACT

Chronic liver injury may result in hepatic fibrosis, which can progress to cirrhosis and eventually liver failure. There are no drugs that are specifically approved for treating hepatic fibrosis. The natural product honokiol (HNK), a bioactive compound extracted from Magnolia grandiflora, represents a potential tool in the management of hepatic fibrosis. Though HNK has been reported to exhibit suppressive effects in a rat fibrosis model, the mechanisms accounting for this suppression remain unclear. In the present study, the anti-fibrotic effects of HNK on the liver were evaluated in vivo and in vitro. In vivo studies utilized a murine liver fibrosis model, in which fibrosis is induced by treatment with carbon tetrachloride (CCl4). For in vitro studies, LX-2 human hepatic stellate cells (HSCs) were treated with HNK, and expression of markers of fibrosis, cell viability, the transforming growth factor-ß (TGF-ß1)/SMAD signaling pathway, and autophagy were analyzed. HNK was well tolerated and significantly attenuated CCl4-induced liver fibrosis in vivo. Moreover, HNK decreased HSC activation and collagen expression by downregulating the TGF-ß1/SMAD signaling pathway and autophagy. These results suggest that HNK is a new potential candidate for the treatment of hepatic fibrosis through suppressing both TGF-ß1/SMAD signaling and autophagy in HSCs.


Subject(s)
Autophagy/drug effects , Biphenyl Compounds/pharmacology , Carbon Tetrachloride Poisoning , Hepatic Stellate Cells , Lignans/pharmacology , Liver Cirrhosis , Liver/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice
4.
Ann Hepatol ; 26: 100560, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34653689

ABSTRACT

INTRODUCTION AND OBJECTIVES: Cirrhosis has gradually become a serious public health issue, especially the national prevalence of cirrhosis was 29.2% in northwest China. Recent evidence has revealed that intestinal barrier (IB) dysfunction results from and contributes to cirrhosis. Our previous results have indicated that insulin-like growth factors (IGF-1) improved the impaired IB function and downregulated high mobility group protein box-1 (HMGB-1). Nevertheless, the role of the IGF-1/HMGB1 axis in cirrhosis remains largely unknown. MATERIALS AND METHODS: Western blotting and qRT-PCR were used to detect protein and mRNA levels of related genes. The levels of AST, ALT, IL-1ß, and TNF-α were examined using commercial kits. Immunofluorescence was used to evaluate the expression of HMGB1 in tissues. RESULTS: In carbon tetrachloride (CCl4)-treated rat, the levels of AST (380.12 vs. 183.97), ALT (148.12 vs. 53.56), IL-1ß (155.94 vs. 55.60), and TNF-α (155.00 vs. 48.90) were significantly increased compared with the control group, while IGF-1 treatment significantly alleviated CCL4-induced inflammatory response and IB dysfunction by downregulating HMGB1-mediated the TLR4/MyD88/NF-κB signaling pathway. In vitro experiments, HMGB1 treatment promoted inflammatory cytokines secretion and reduced cell viability and tight junctions by activating the TLR4/MyD88/NF-κB signaling pathway in Caco-2 cells, but IGF-1 alleviated these effects. CONCLUSION: Our findings suggest that IGF-1 might serve as a potential therapeutic target for cirrhosis and IB dysfunction via inactivation of the TLR4/MyD88/NF-κB pathway through down-regulation HMGB1.


Subject(s)
Carbon Tetrachloride Poisoning/complications , Down-Regulation , Gene Expression Regulation , HMGB1 Protein/genetics , Insulin-Like Growth Factor I/therapeutic use , Intestinal Mucosa/metabolism , Liver Cirrhosis, Experimental/genetics , Animals , Caco-2 Cells , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/metabolism , HMGB1 Protein/biosynthesis , Humans , Intestinal Mucosa/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/therapy , Male , RNA/genetics , Rats
5.
Mol Biol Rep ; 48(11): 7153-7163, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34536190

ABSTRACT

BACKGROUND: Previous study reports that fibroblast growth factor 21 (FGF21) could ameliorate hepatic fibrosis, but its mechanisms have not been fully investigated. METHODS AND RESULTS: In this study, three models were used to investigate the mechanism by which FGF21 alleviates liver fibrosis. Hepatic fibrosis animal models were respectively induced by CCL4 and dimethylnitrosamine. Our results demonstrated that liver index and liver function were deteriorated in both models. Hematoxylin and eosin and Masson's staining showed that the damaged tissue architectonics were observed in the mice of both models. Treatment with FGF21 significantly ameliorated these changes. ELISA analysis showed that the serum levels of IL-1ß, IL-6 and TNF-α were significantly elevated in both models. However, administration of FGF21 significantly reduced these inflammatory cytokines. Real-time PCR and Western blot analysis showed that treatment with FGF21 significantly decreased mRNA and protein expressions of collagenI, α-SMA and TGF-ß. Platelet-derived growth factor-BB (PDGF-BB) stimulant was used to establish the experimental cell model in hepatic stellate cells (HSCs). Real-time PCR and Western blot analysis demonstrated that the expression of collagenI and α-SMA were significantly upregulated by this stimulant in model group. Interestingly, our results showed that mRNA and protein expressions of leptin were also significantly induced in PDGF-BB treated HSCs. Administration of FGF21 significantly reduced leptin expression in a dose dependent manner and these effects were reversed in siRNA (against ß-klotho) transfected HSCs. Furthermore, the leptin signaling pathways related protein p-ERK/t-ERK, p-STAT3/STAT3 and TGF-ß were significantly downregulated by FGF21 treatment in a dose dependent manner. The expressions of SOCS3 and Nrf-2 were enhanced by treatment with FGF21. The underlying mechanism may be that FGF21 regulates leptin-STAT3 axis via Nrf-2 and SOCS3 pathway in activated HSCs. CONCLUSIONS: FGF21 ameliorates hepatic fibrosis by multiple mechanisms.


Subject(s)
Carbon Tetrachloride Poisoning , Fibroblast Growth Factors/pharmacology , Liver Cirrhosis , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/metabolism , Gene Expression Regulation/drug effects , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred ICR , Tumor Necrosis Factor-alpha/metabolism
6.
J Pharm Pharmacol ; 73(11): 1562-1570, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34387320

ABSTRACT

OBJECTIVES: Spirulina platensis (SP) is an edible Cyanobacterium with ethnomedicinal significance. This study aims at evaluating the beneficial effect of SP against carbon tetrachloride (CCl4)-induced liver toxicity in male rats. METHODS: Rats received intraperitoneal injections of CCl4 (2 ml/kg body weight [b.w.] per every other day) for 40 days, alone or in combination with oral treatments of SP (400 mg/kg b.w. per day). KEY FINDINGS: SP attenuated haematological disturbances, serum liver markers, hepatic necrosis and inflammation, and dyslipidemia in CCl4-intoxicated rats. SP also reduced CCl4-induced oxidative stress by increasing the activities of antioxidant enzymes, such as glutathione peroxidase, superoxide dismutase and catalase and glutathione content, and inhibiting lipid peroxidation products and nitric oxide levels in the rat liver. Further investigations revealed that SP counteracted CCl4-induced increased hepatic levels of Ki-67 (a parameter of cell proliferation), interleukin-6, and tumour necrosis factor-alpha and cyclooxygenase-2 messenger RNA expression. Noticeably, the supplementation of SP restored the decrease of proapoptotic p53 protein levels in the liver of rats treated with CCl4. CONCLUSIONS: SP prevented liver damage in CCl4-treated rats via augmentation of antioxidant defense mechanisms and inhibition of inflammatory cytokines/mediators and antiproliferative effects.


Subject(s)
Antioxidants/therapeutic use , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Liver/drug effects , Oxidative Stress/drug effects , Phytotherapy , Spirulina , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Carbon Tetrachloride Poisoning/complications , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/prevention & control , Chemical and Drug Induced Liver Injury/complications , Chemical and Drug Induced Liver Injury/metabolism , Cytokines/metabolism , Dietary Supplements , Dyslipidemias/prevention & control , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Inflammation/metabolism , Inflammation/prevention & control , Lipid Peroxidation , Liver/metabolism , Male , Nitric Oxide/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats, Wistar , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Sci Rep ; 11(1): 16575, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400737

ABSTRACT

Carbon tetrachloride (CCl4) is an abundant environmental pollutant that can generate free radicals and induce oxidative stress in different human and animal organs like the kidney, lung, brain, and spleen, causing toxicity. The present study evaluated the alleviative mechanism of the isolated polyphenolic fraction from seedless (pulp and skin) black Vitis vinifera (VVPF) on systemic oxidative and necroinflammatory stress in CCl4-intoxicated rats. Here, we found that the administration of VVPF to CCl4-intoxicated rats for ten days was obviously ameliorated the CCl4-induced systemic elevation in ROS, NO and TBARS levels, as well as MPO activity. Also, it upregulated the cellular activities of the enzymatic (SOD, and GPx) and non-enzymatic (TAC and GSH) antioxidants. Furthermore, the gene expression of the ROS-related necroinflammatory mediators (NF-κB, iNOS, COX-2, and TNF-α) in the kidney, brain, and spleen, as well as IL-1ß, and IL-8 in the lung were greatly restored. The histopathological studies confirmed these biochemical results and showed a noticeable enhancing effect in the architecture of the studied organs after VVPF intake. Thus, this study indicated that VVPF had an alleviative effect on CCl4-induced necroinflammation and oxidative stress in rat kidney, lung, brain, and spleen via controlling the ROS/NF-κB pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Carbon Tetrachloride Poisoning/drug therapy , NF-kappa B/antagonists & inhibitors , Phytotherapy , Polyphenols/therapeutic use , Reactive Oxygen Species/antagonists & inhibitors , Vitis/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Brain/drug effects , Brain/metabolism , Carbon Tetrachloride Poisoning/metabolism , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cytokines/biosynthesis , Cytokines/genetics , Drug Evaluation, Preclinical , Fruit/chemistry , Inhibitory Concentration 50 , Kidney/drug effects , Kidney/metabolism , Lung/drug effects , Lung/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Polyphenols/chemistry , Polyphenols/isolation & purification , Rats , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Thiobarbituric Acid Reactive Substances/analysis
8.
Cells ; 10(6)2021 06 21.
Article in English | MEDLINE | ID: mdl-34205789

ABSTRACT

Toll-like receptor 4 (TLR4) signaling plays a key role in liver inflammation and fibrosis. The therapeutic effects of eritoran, a TLR4 antagonist, in mice with chronic liver injury remained unclear. C57BL/6 mice were fed a fast-food diet (FFD) or treated with carbon tetrachloride (CCl4) to induce chronic liver injury. Eritoran (10 mg/kg) or a vehicle was randomly intraperitoneally administered to the FFD-fed mice and the CCl4-injured mice. Primary mouse liver cells were cultured with lipopolysaccharide (LPS) or eritoran. In both FFD and CCl4 mouse models, eritoran significantly reduced serum ALT levels and decreased hepatic inflammatory cell infiltration without altering hepatic steatosis. Additionally, eritoran attenuated liver fibrosis by decreasing hepatic stellate cells (HSCs) activation and the abundance of α-smooth muscle actin and transforming growth factor-ß1. Hepatic TLR4 downstream signaling including MyD88 expression, NF-κB p65 nuclear translocation, p38 and JNK phosphorylation were successfully inhibited by eritoran. In the in vitro study, LPS-induced nuclear translocation of NF-κB in primary HSCs and Kupffer cells was significantly suppressed by eritoran. In conclusion, eritoran attenuated hepatic inflammation and fibrosis by inhibition of the TLR4 signaling pathway in mice with chronic liver injury. Eritoran may serve as a potential drug for chronic liver disease.


Subject(s)
Carbon Tetrachloride Poisoning , Disaccharides/pharmacology , End Stage Liver Disease , Liver Cirrhosis , Sugar Phosphates/pharmacology , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , End Stage Liver Disease/chemically induced , End Stage Liver Disease/drug therapy , End Stage Liver Disease/metabolism , Hepatic Stellate Cells/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Kupffer Cells/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Transcription Factor RelA/metabolism , Transforming Growth Factor beta1/metabolism
9.
Signal Transduct Target Ther ; 6(1): 270, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262012

ABSTRACT

Mesenchymal stem cell (MSC)-mediated immunomodulation has been harnessed for the treatment of human diseases, but its underlying mechanism has not been fully understood. Dead cells, including apoptotic cells have immunomodulatory properties. It has been repeatedly reported that the proportion of nonviable MSCs in a MSC therapeutic preparation varied from 5~50% in the ongoing clinical trials. It is conceivable that the nonviable cells in a MSC therapeutic preparation may play a role in the therapeutic effects of MSCs. We found that the MSC therapeutic preparation in the present study had about 5% dead MSCs (DMSCs), characterized by apoptotic cells. Namely, 1 × 106 MSCs in the preparation contained about 5 × 104 DMSCs. We found that the treatment with even 5 × 104 DMSCs alone had the equal therapeutic effects as with 1 × 106 MSCs. This protective effect of the dead MSCs alone was confirmed in four mouse models, including concanavalin A (ConA)- and carbon tetrachloride (CCl4)-induced acute liver injury, LPS-induced lung injury and spinal cord injury. We also found that the infused MSCs died by apoptosis in vivo. Furthermore, the therapeutic effect was attributed to the elevated level of phosphatidylserine (PS) upon the injection of MSCs or DMSCs. The direct administration of PS liposomes (PSLs) mimic apoptotic cell fragments also exerted the protective effects as MSCs and DMSCs. The Mer tyrosine kinase (MerTK) deficiency or the knockout of chemokine receptor C-C motif chemokine receptor 2 (CCR2) reversed these protective effects of MSCs or DMSCs. These results revealed that DMSCs alone in the therapeutic stem cell preparation or the apoptotic cells induced in vivo may exert the same immunomodulatory property as the "living MSCs preparation" through releasing PS, which was further recognized by MerTK and participated in modulating immune cells.


Subject(s)
Apoptosis , Carbon Tetrachloride Poisoning , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Phosphatidylserines/metabolism , Animals , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/therapy , Female , Mice , Mice, Knockout
10.
Sci Rep ; 11(1): 9730, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33958644

ABSTRACT

The liver is an exclusive organ with tremendous regenerative capacity. Liver metabolic functions exhibit spatial heterogeneity, reflecting liver zonation. The mechanisms controlling the proliferation of hepatocytes and the accompanying matrix reconstruction during regeneration have been well explored, but the recovery potential of differentiated metabolic functions and zonation after liver injury remains unclear. We employed a mouse model of carbon tetrachloride (CCl4) induced-acute liver injury with clodronate-induced macrophage depletion to clarify the impact of liver injury on liver metabolism and recovery dynamics of metabolic function and liver zonation during regeneration. Depleting macrophages suppressed tissue remodelling and partially delayed cell proliferation during regeneration after liver injury. In addition, recovery of metabolic functions was delayed by suppressing the tissue remodelling caused by the depleted macrophages. The model revealed that drug metabolic function was resilient against the dysfunction caused by liver injury, but glutamine synthesis was not. Metabolomic analysis revealed that liver branched-chain amino acid (BCAA) and carbohydrate metabolism were suppressed by injury. The plasma BCAA concentration reflected recovery of hepatic function during regeneration. Our study reveals one aspect of the regenerative machinery for hepatic metabolism following acute liver injury.


Subject(s)
Carbon Tetrachloride Poisoning/immunology , Chemical and Drug Induced Liver Injury/immunology , Liver/immunology , Macrophages/immunology , Amino Acids, Branched-Chain/blood , Animals , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/physiopathology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Clodronic Acid/administration & dosage , Liver/metabolism , Liver/physiopathology , Liver Regeneration/drug effects , Mice , Mice, Inbred C57BL
11.
Int J Biol Sci ; 17(4): 1153-1167, 2021.
Article in English | MEDLINE | ID: mdl-33867837

ABSTRACT

Background & Aims: Liver fibrosis is a common consequence of chronic liver injury and is characterized by the accumulation of extracellular matrix mainly generated from activated hepatic stellate cells (HSCs). At present, the mechanisms underlying liver fibrogenesis remain obscure and effective pharmacological therapies are lacking. Neutrophil-specific microRNA-223 (miR-223) plays an important role in controlling the development of various liver diseases; however, its role in HSC activation and liver fibrosis remains unclear. Methods: Liver fibrosis was induced by chronic carbon tetrachloride (CCl4) injection of miR-223 knockout (miR-223KO) mice and littermate wild-type controls. MiR-223 was overexpressed in cultured HSCs to determine its function and targets during HSC activation and proliferation. The expression of miR-223 and pri-miR-223 was examined in primary HSCs isolated from CCl4-treated mice and in cultured HSCs. The communication between HSCs and neutrophils was studied by performing in vitro co-culture experiments. Results: Genetic deletion of miR-223 exacerbated chronic CCl4-induced liver fibrosis. Administration of miR-223 inhibited liver fibrosis by inhibiting the transcriptional coactivator with PDZ-binding motif (TAZ)-Indian hedgehog (IHH)-GLI Family Zinc Finger 2 (GLI2) pathway via the crosstalk between hepatocytes and HSCs. Overexpression of miR-223 also directly attenuated Gli2 as well as platelet-derived growth factor receptor α/ß (Pdgfra/b) expression in HSCs, thereby suppressing HSC activation and proliferation. The expression of pri-miR-223 and miR-223 was downregulated during HSC activation in vitro. Expression of pri-miR-223 was also decreased in activated HSCs in vivo in fibrotic livers but mature miR-223 expression was not reduced. Finally, in co-culture experiments, activated HSCs were able to take up miR-223-enriched extracellular vesicles from neutrophils, resulting in elevation of miR-223. Conclusion: MiR-223 restricts liver fibrosis by targeting multiple genes in hepatocytes and HSCs, providing potential therapeutic targets for the treatment of liver fibrosis.


Subject(s)
Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , MicroRNAs/metabolism , Acyltransferases/metabolism , Animals , Carbon Tetrachloride Poisoning/metabolism , Cell Line , Extracellular Vesicles/metabolism , Hedgehog Proteins/metabolism , Humans , Mice, Knockout , Receptor Cross-Talk , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Zinc Finger Protein Gli2/metabolism
12.
Biosci Biotechnol Biochem ; 85(4): 775-785, 2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33686395

ABSTRACT

This research aimed to evaluate the antihepatic fibrosis effect and explore the mechanism of Qiwei Qinggan Powder (QGS-7) in vivo and in vitro. Carbon tetrachloride (CCl4)-treated rats and hepatic stellate cells (HSCs) were used. QGS-7 treatment significantly improved the liver function of rats as indicated by decreased serum enzymatic activities of alanine aminotransferase, aspartate transaminase, and alkaline phosphatase. Meanwhile, the hydroxyproline of liver was significantly decreased. Histopathological results indicated that QGS-7 alleviated liver damage and reduced the formation of fibrosis septa. Moreover, QGS-7 significantly attenuated expressions of Alpha smooth muscle actin, Collagen I, Janus kinase 2 (JAK2), phosphorylation-JAK2, signal transducer and activator of transcription 3 (STAT3), phosphorylation-STAT3 in the rat hepatic fibrosis model. QGS-7 inhibited HSC proliferation and promoted it apoptosis. QGS-7 may affect hepatic fibrosis through JAK2/STAT3 signaling pathway so as to play an antihepatic fibrosis role.


Subject(s)
Carbon Tetrachloride Poisoning/drug therapy , Liver Cirrhosis/drug therapy , Medicine, Mongolian Traditional , Animals , Carbon Tetrachloride Poisoning/metabolism , Cell Proliferation/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Hydroxyproline/metabolism , Janus Kinase 2/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Function Tests , Mongolia , Phosphorylation , Powders , Rats , STAT3 Transcription Factor/metabolism
13.
Cytokine ; 136: 155288, 2020 12.
Article in English | MEDLINE | ID: mdl-32980687

ABSTRACT

Hepatic fibrosis is characterized by abnormal accumulation of extracellular matrix (ECM). Hepatic stellate cells (HSCs) are the primary cells that produce ECM in response to hepatic injury, and transforming growth factor-beta (TGF-ß) has been regarded as the central stimulus responsible for HSC-mediated ECM production. In the present study, we attempted to identify a critical factor in HSC activation and the underlying mechanism. By analyzing online microarray expression profiles, we found that the expression of high-affinity cationic amino acid transporter 1 (CAT1) was upregulated in hepatic fibrosis models and activated HSCs. We isolated and identified mouse HSCs (MHSCs) and found that in these cells, CAT1 was most highly upregulated by TGF-ß1 stimulation in both time- and dose-dependent manners. In vitro, CAT1 overexpression further enhanced, while CAT1 silencing inhibited, the effect of TGF-ß1 in promoting MHSC activation. In vivo, CAT1 silencing significantly improved the hepatic fibrosis induced by both CCl4 and non-alcoholic fatty liver disease (NAFLD). In summary, CAT1 was significantly upregulated in TGF-ß1-activated MHSCs and mice with hepatic fibrosis. CAT1 silencing inhibited TGF-ß1-induced MHSC activation in vitro and fibrogenic changes in vivo. CAT1 is a promising target for hepatic fibrosis treatment that requites further investigation in human cells and clinical practice.


Subject(s)
Calcium Channels/metabolism , Extracellular Matrix/metabolism , Gene Silencing , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , TRPV Cation Channels/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Calcium Channels/genetics , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Cell Line , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , TRPV Cation Channels/genetics , Transforming Growth Factor beta1/genetics
14.
Cytokine ; 136: 155250, 2020 12.
Article in English | MEDLINE | ID: mdl-32882667

ABSTRACT

BACKGROUND: Liver fibrosis is a serious health problem which may lead to advanced liver cirrhosis and hepatocellular carcinoma. OBJECTIVE: The present study aimed to investigate the role of Wnt/ß-catenin signaling pathway and glutamine aminohydrolase enzyme (l-glutaminase) in the pathogenesis of liver fibrosis and the potential benefits of niclosamide in treating liver fibrosis. METHODS: Ninety male Albino rats were divided into 6 equal groups (n = 15) as follows: a normal control group (NC), CCl4-only treated group (Fib.) which received 1 mg/kg CCl4 two times weekly, niclosamide-treated group (Niclo.) which received 5 mg/kg of niclosamide one time daily, lithium chloride-treated group (LiCl) which received 100 mg/kg of LiCl one time daily, niclosamide-and-CCl4-treated group (Niclo. + Fib.) which received same doses of niclosamide and CCl4 given to other groups, and finally lithium chloride-and-CCl4-treated rat group (LiCl + Fib.) which received same doses of LiCl and CCl4 given to other groups. All treatments were administered orally for 8 weeks. Liver tissue was assessed for l-hydroxyproline, beta-catenin (ß-catenin), l-glutaminase activity, as well as the gene expression of transforming growth factor beta-1 (TGF-ß1) and Dishevelled-2 (Dvl2). Histopathological and immunohistochemical analyses of alpha smooth muscle actin α-SMA were performed. Serum alanine transaminase (ALT), aspartate transaminase (AST), and total bilirubin were measured. RESULTS: The group of niclosamide-and-CCl4-treated rats showed a significant decrease in total bilirubin, ALT and AST, ß-catenin, l-hydroxyproline, l-glutaminase activity, and gene expression of TGF-ß1 and Dvl2. Moreover, the liver tissue in this group of rats showed mild α-SMA reactivity compared with the rats treated with CCl4 only (fibrosis group). On the other hand, lithium chloride-and-CCl4-treated rats showed a significant increase in liver indices, TGF-ß1 expression, ß-catenin, l-hydroxyproline, and l-glutaminase activity with severe α-SMA reactivity and apoptosis in the liver tissue. CONCLUSIONS: Niclosamide protected rats against liver fibrosis by inhibiting the Wnt/ß-catenin pathway and glutaminolysis.


Subject(s)
Carbon Tetrachloride Poisoning , Drug Repositioning , Glutamic Acid/metabolism , Lithium Chloride/pharmacology , Liver Cirrhosis , Niclosamide/pharmacology , Wnt Signaling Pathway/drug effects , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male , Rats
15.
Nat Metab ; 2(6): 514-531, 2020 06.
Article in English | MEDLINE | ID: mdl-32694734

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is characterized by lipotoxicity, inflammation and fibrosis, ultimately leading to end-stage liver disease. The molecular mechanisms promoting NASH are poorly understood, and treatment options are limited. Here, we demonstrate that hepatic expression of bone morphogenetic protein 8B (BMP8B), a member of the transforming growth factor beta (TGFß)-BMP superfamily, increases proportionally to disease stage in people and animal models with NASH. BMP8B signals via both SMAD2/3 and SMAD1/5/9 branches of the TGFß-BMP pathway in hepatic stellate cells (HSCs), promoting their proinflammatory phenotype. In vivo, the absence of BMP8B prevents HSC activation, reduces inflammation and affects the wound-healing responses, thereby limiting NASH progression. Evidence is featured in primary human 3D microtissues modelling NASH, when challenged with recombinant BMP8. Our data show that BMP8B is a major contributor to NASH progression. Owing to the near absence of BMP8B in healthy livers, inhibition of BMP8B may represent a promising new therapeutic avenue for NASH treatment.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Carbon Tetrachloride Poisoning/metabolism , Diet, High-Fat , Diet, Western , Hepatic Stellate Cells/metabolism , Humans , Inflammation/genetics , Liver Regeneration/drug effects , Liver Regeneration/genetics , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Recombinant Proteins/pharmacology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing/genetics
16.
Int J Mol Sci ; 21(15)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32718038

ABSTRACT

The lack of Lipocalin (LCN2) provokes overwhelming endoplasmic reticulum (ER) stress responses in vitro and in acute toxic liver injury models, resulting in hepatocyte apoptosis. LCN2 is an acute phase protein produced in hepatocytes in response to acute liver injuries. In line with these findings we investigated ER stress responses of Lcn2-/- mice in chronic ER stress using a long-term repetitive carbon tetrachloride (CCl4) injection model. We found chronic CCl4 application to enhance ER stress and unfolded protein responses (UPR), including phosphorylation of eukaryotic initiation factor 2α (eIF2α), increased expression of binding immunoglobulin protein (BiP) and glucose-regulated protein 94 (GRP94). IRE1α/TRAF2/JNK signaling enhanced mitochondrial apoptotic pathways, and showed slightly higher in Lcn2-/- mice compared to the wild type counterparts, leading to increased hepatocyte apoptosis well evidenced by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Hepatocyte injuries were confirmed by significant high serum alanine transaminase (ALT) levels in CCl4-treated Lcn2-/- mice. Lcn2-/- mice furthermore developed mild hepatic steatosis, supporting our finding that ER stress promotes lipogenesis. In a previous report we demonstrated that the pharmacological agent tunicamycin (TM) induced ER stress through altered protein glycosylation and induced high amounts of C/EBP-homologous protein (CHOP), resulting in hepatocyte apoptosis. We compared TM-induced ER stress in wild type, Lcn2-/-, and Chop null (Chop-/-) primary hepatocytes and found Chop-/- hepatocytes to attenuate ER stress responses and resist ER stress-induced hepatocyte apoptosis through canonical eIF2α/GADD34 signaling, inhibiting protein synthesis. Unexpectedly, in later stages of TM incubation, Chop-/- hepatocytes resumed activation of IRE1α/JNK/c-Jun and p38/ATF2 signaling, leading to late hepatocyte apoptosis. This interesting observation indicates Chop-/- mice to be unable to absolutely prevent all types of liver injury, while LCN2 protects the hepatocytes by maintaining homeostasis under ER stress conditions.


Subject(s)
Apoptosis , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride/toxicity , Endoplasmic Reticulum Stress , Hepatocytes/metabolism , Lipocalin-2/deficiency , Unfolded Protein Response , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Hepatocytes/pathology , Lipocalin-2/metabolism , Lipogenesis/drug effects , Lipogenesis/genetics , Mice , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/genetics , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
17.
Int J Biol Macromol ; 162: 533-547, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32565302

ABSTRACT

This study aimed to explore the basic structural features of phosphorylated Pleurotus ostreatus polysaccharide (PPOP) and study the protective effect of PPOP on liver injury induced by carbon tetrachloride in male Kunming mice. The phosphorylated polysaccharide was prepared from the natural polysaccharide extracted from Pleurotus ostreatus (POP). The structures of PPOP and POP were characterized by FT-IR, ESEM spectroscopy, and Congo red test. Chemical composition analysis revealed that PPOP was mainly composed of rhamnose, galacturonic acid, and xylose in a molar ratio of 0.10: 1.98: 1.00. Structural analysis indicated that PPOP had multi-strand structure and the absorption peaks of PO and P-O-C. Furthermore, animal experiments showed that the hepatoprotective effect of PPOP against liver injury was reflected by decreasing the levels of alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, total cholesterol, trilaurin, and low-density lipoprotein cholesterol in the serum, increasing the content of high-density lipoprotein cholesterol and albumin in blood, reducing the content of malondialdehyde and promoting the activity of antioxidant enzymes in liver. PPOP exhibited stronger hepatoprotective effect and antioxidant activity in vivo than POP. The final results indicated that PPOP could be used in the treatment of chemical-induced hepatotoxicity based on the above biological research.


Subject(s)
Carbon Tetrachloride Poisoning/drug therapy , Chemical and Drug Induced Liver Injury/drug therapy , Fungal Polysaccharides , Pleurotus/chemistry , Animals , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/isolation & purification , Fungal Polysaccharides/pharmacology , Male , Mice , Structure-Activity Relationship
18.
Blood ; 136(6): 726-739, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32374849

ABSTRACT

Although the serum-abundant metal-binding protein transferrin (encoded by the Trf gene) is synthesized primarily in the liver, its function in the liver is largely unknown. Here, we generated hepatocyte-specific Trf knockout mice (Trf-LKO), which are viable and fertile but have impaired erythropoiesis and altered iron metabolism. Moreover, feeding Trf-LKO mice a high-iron diet increased their susceptibility to developing ferroptosis-induced liver fibrosis. Importantly, we found that treating Trf-LKO mice with the ferroptosis inhibitor ferrostatin-1 potently rescued liver fibrosis induced by either high dietary iron or carbon tetrachloride (CCl4) injections. In addition, deleting hepatic Slc39a14 expression in Trf-LKO mice significantly reduced hepatic iron accumulation, thereby reducing ferroptosis-mediated liver fibrosis induced by either a high-iron diet or CCl4 injections. Finally, we found that patients with liver cirrhosis have significantly lower levels of serum transferrin and hepatic transferrin, as well as higher levels of hepatic iron and lipid peroxidation, compared with healthy control subjects. Taken together, these data indicate that hepatic transferrin plays a protective role in maintaining liver function, providing a possible therapeutic target for preventing ferroptosis-induced liver fibrosis.


Subject(s)
Ferroptosis/physiology , Iron/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Transferrin/physiology , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cyclohexylamines/pharmacology , Cytokines/analysis , Erythropoiesis/physiology , Erythropoietin/analysis , Female , Ferroptosis/drug effects , Hepatocytes/metabolism , Homeostasis , Iron Overload/complications , Iron, Dietary/toxicity , Lipid Peroxidation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/analysis , Phenylenediamines/pharmacology , Transferrin/analysis
19.
Sci Rep ; 10(1): 3850, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123215

ABSTRACT

Hepatic fibrosis is a major consequence of chronic liver disease such as non-alcoholic steatohepatitis which is undergoing a dramatic evolution given the obesity progression worldwide, and has no treatment to date. Hepatic stellate cells (HSCs) play a key role in the fibrosis process, because in chronic liver damage, they transdifferentiate from a "quiescent" to an "activated" phenotype responsible for most the collagen deposition in liver tissue. Here, using a diet-induced liver fibrosis murine model (choline-deficient amino acid-defined, high fat diet), we characterized a specific population of HSCs organized as clusters presenting simultaneously hypertrophy of retinoid droplets, quiescent and activated HSC markers. We showed that hypertrophied HSCs co-localized with fibrosis areas in space and time. Importantly, we reported the existence of this phenotype and its association with collagen deposition in three other mouse fibrosis models, including CCl4-induced fibrosis model. Moreover, we have also shown its relevance in human liver fibrosis associated with different etiologies (obesity, non-alcoholic steatohepatitis, viral hepatitis C and alcoholism). In particular, we have demonstrated a significant positive correlation between the stage of liver fibrosis and HSC hypertrophy in a cohort of obese patients with hepatic fibrosis. These results lead us to conclude that hypertrophied HSCs are closely associated with hepatic fibrosis in a metabolic disease context and may represent a new marker of metabolic liver disease progression.


Subject(s)
Carbon Tetrachloride Poisoning , Dietary Fats/adverse effects , Hepatic Stellate Cells , Liver Cirrhosis , Animals , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Dietary Fats/pharmacology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice
20.
Mol Med Rep ; 21(3): 1390-1398, 2020 03.
Article in English | MEDLINE | ID: mdl-31922209

ABSTRACT

Carbon tetrachloride (CCl4) is widely used to induce hepatic fibrosis. Therapeutic agents alleviate hepatic fibrosis by inhibiting signal transducer and activator of transcription 3 (STAT3) activation. To understand the direct effects of CCl4 on STAT3 expression in the liver, the present study incubated cultured hepatocytes expressing connective tissue growth factor (CTGF) with CCl4. Rats exposed to CCl4 for 8 weeks exhibited hepatic fibrosis, which was confirmed through the assessment of plasma biomarkers. Isolated liver samples were used to determine the protein levels of CTGF and STAT3 using western blotting. In addition, STAT3 expression was silenced in α mouse liver 12 (AML­12) cells using small interfering RNA transfection. In addition, a pharmacological inhibitor, stattic, was used to inhibit STAT3 expression. The incubation of AML­12 cells with CCl4 induced a dose­dependent increase in CTGF expression and STAT3 activation. Notably, silymarin, an extract from milk thistle, inhibited these changes in AML­12 cells and the antioxidant tiron produced similar effects. Silencing of STAT3 reduced the CTGF expression promoted by CCl4 in the hepatocytes. Additionally, similar to tiron, stattic inhibited CTGF expression induced by CCl4. In conclusion, CCl4 may activate STAT3 through oxidative stress to promote CTGF expression, which is one of the main factors contributing to the risk of hepatic fibrosis.


Subject(s)
Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride/toxicity , Connective Tissue Growth Factor/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , STAT3 Transcription Factor/metabolism , Animals , Carbon Tetrachloride Poisoning/pathology , Cell Line , Hepatocytes/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...