Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; : e2403387, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39018261

ABSTRACT

RNA splicing is a dynamic molecular process in response to environmental stimuli and is strictly regulated by the spliceosome. Sm proteins, constituents of the spliceosome, are key components that mediate splicing reactions; however, their potential role in hepatocellular carcinoma (HCC) is poorly understood. In the study, SNRPD2 (PD2) is found to be the most highly upregulated Sm protein in HCC and to act as an oncogene. PD2 modulates DDX39A intron retention together with HNRNPL to sustain the DDX39A short variant (39A_S) expression. Mechanistically, 39A_S can mediate MYC mRNA nuclear export to maintain high MYC protein expression, while MYC in turn potentiates PD2 transcription. Importantly, digitoxin can directly interact with PD2 and has a notable cancer-suppressive effect on HCC. The study reveals a novel mechanism by which DDX39A senses oncogenic MYC signaling and undergoes splicing via PD2 to form a positive feedback loop in HCC, which can be targeted by digitoxin.

2.
Oncogenesis ; 12(1): 54, 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-37957153

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is frequently reported to be hyperactivated in hepatocellular carcinoma (HCC) and contributes to HCC recurrence. However, the underlying regulatory mechanisms of mTORC1 signaling in HCC are not fully understood. In the present study, we found that the expression of kinesin family member 18B (KIF18B) was positively correlated with mTORC1 signaling in HCC, and the upregulation of KIF18B and p-mTOR was associated with a poor prognosis and HCC recurrence. Utilizing in vitro and in vivo assays, we showed that KIF18B promoted HCC cell proliferation and migration through activating mTORC1 signaling. Mechanistically, we identified Actin gamma 1 (γ-Actin) as a binding partner of KIF18B. KIF18B and γ-Actin synergistically modulated lysosome positioning, promoted mTORC1 translocation to lysosome membrane, and prohibited p70 S6K from entering lysosomes for degradation, which finally led to the enhancement of mTORC1 signaling transduction. Moreover, we found that KIF18B was a direct target of Forkhead box M1, which explains the potential mechanism of KIF18B overexpression in HCC. Our study highlights the potential of KIF18B as a therapeutic target for the treatment of HCC.

3.
Biochem Pharmacol ; 210: 115451, 2023 04.
Article in English | MEDLINE | ID: mdl-36758707

ABSTRACT

Circular RNAs (circRNAs) are a newly identified form of non-coding RNA that play a crucial role in various pathological processes. However, the expression profile and function of circRNAs in hepatic fibrosis (HF) remain largely unknown. In this study, we showed that a novel circRNA ASPH (circASPH) mediates HF by targeting the miR-139-5p/Notch1 axis. We investigated the expression profile of circRNAs in hepatocyte exosomes of mice with HF using circRNA-sequencing and found significant upregulation of circASPH. Loss- and gain-of-function analysis of circASPH was performed to assess its role in HF. Furthermore, we performed luciferase reporter assay, RNA pull-down, and fluorescence in situ hybridization analyses and confirmed that circASPH directly binds to miR-139-5p. We also found that circASPH was upregulated in liver fibrogenesis. Downregulation of circASPH expression inhibited hepatic stellate cell (HSC) activation and proliferation, induced apoptosis, and attenuated mouse liver fibrogenic injury. Mechanistically, circASPH directly targeted miR-139-5p to regulate the expression of Notch1 in HF. Thus, downregulation of circASPH may suppress the activation of HSCs and HF through the circASPH/miR-139-5p/Notch1 axis. Our findings indicated that circASPH may be a potential biomarker for HF diagnosis and therapy.


Subject(s)
MicroRNAs , RNA, Circular , Mice , Animals , RNA, Circular/genetics , RNA, Circular/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Hepatic Stellate Cells/metabolism , In Situ Hybridization, Fluorescence , Liver Cirrhosis/metabolism , Apoptosis , Cell Proliferation/genetics
4.
Cells ; 11(20)2022 10 20.
Article in English | MEDLINE | ID: mdl-36291167

ABSTRACT

Cell death is a vital physiological or pathological phenomenon in the development process of the organism. Ferroptosis is a kind of newly-discovered regulated cell death (RCD), which is different from other RCD patterns, such as apoptosis, necrosis and autophagy at the morphological, biochemical and genetic levels. It is a kind of iron-dependent mode of death mediated by lipid peroxides and lipid reactive oxygen species aggregation. Noteworthily, the number of studies focused on ferroptosis has been increasing exponentially since ferroptosis was first found in 2012. The liver is the organ that stores the most iron in the human body. Recently, it was frequently found that there are different degrees of iron metabolism disorder and lipid peroxidation and other ferroptosis characteristics in various liver diseases. Numerous investigators have discovered that the progression of various liver diseases can be affected via the regulation of ferroptosis, which may provide a potential therapeutic strategy for clinical hepatic diseases. This review aims to summarize the mechanism and update research progress of ferroptosis, so as to provide novel promising directions for the treatment of liver diseases.


Subject(s)
Ferroptosis , Liver Diseases , Humans , Reactive Oxygen Species/metabolism , Lipid Peroxides , Iron/metabolism , Liver Diseases/therapy
5.
Mol Ther Methods Clin Dev ; 26: 191-206, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-35859692

ABSTRACT

Liver fibrosis is a wound-healing response that results from various chronic damages. If the causes of damage are not removed or effective treatments are not given in a timely manner, it will progress to cirrhosis, even liver cancer. Currently, there are no specific medical therapies for liver fibrosis. Adeno-associated virus (AAV)-mediated gene therapy, one of the frontiers of modern medicine, has gained more attention in many fields due to its high safety profile, low immunogenicity, long-term efficacy in mediating gene expression, and increasingly known tropism. Notably, increasing evidence suggests a promising therapeutic potential for AAV-mediated gene therapy in different liver fibrosis models, which helps to correct abnormally changed target genes in the process of fibrosis and improve liver fibrosis at the molecular level. Moreover, the addition of cell-specific promoters to the genome of recombinant AAV helps to limit gene expression in specific cells, thereby producing better therapeutic efficacy in liver fibrosis. However, animal models are considered to be powerless predictive of tissue tropism, immunogenicity, and genotoxic risks in humans. Thus, AAV-mediated gene therapy will face many challenges. This review systemically summarizes the recent advances of AAV-mediated gene therapy in liver fibrosis, especially focusing on cellular and molecular mechanisms of transferred genes, and presents prospective challenges.

6.
Toxicology ; 466: 153087, 2022 01 30.
Article in English | MEDLINE | ID: mdl-34974135

ABSTRACT

Alcoholic liver injury (ALI) is a major risk factor for alcoholic liver disease, characterized by excessive inflammatory response and abnormal liver dysfunction. Previous studies have indicated that O-alkyl and o-benzyl hesperetin derivative-1 L (HD-1 L) has anti-inflammatory and hepato-protective effects in CCl4-induced liver injury. However, its effect on ALI and underlying mechanism has not been elucidated. This study was designed to evaluate the protective effects of HD-1 L on alcoholic liver injury and reveal the underlying mechanisms. ALI model was established in male C57BL/6 J mice (aged 6-8 weeks) by Gao-Binge protocol. The mice were received different doses of HD-1 L (25 mg/kg, 50 mg/kg, 100 mg/kg) by daily intragastric administration, respectively. Liver function and inflammation were measured. Mechanism underlying the anti-inflammatory and hepato-protective effect of HD-1 L were studied in RAW264.7 cells. In alcoholic liver injury mice, HD-1 L effectively improved the liver pathology, and remarkably reduced the levels of alanine transaminase (ALT), aspartate transaminase (AST), triglyceride (TG) and total cholesterol (T-CHO) in serum. Moreover, HD-1 L markedly suppressed inflammation in vivo and inhibited the secretion of inflammatory factors in vitro. Our results showed that HD-1 L decreased the activity of Bromodomain-containing Protein 2 (BRD2) and inhibited expression of BRD2 in vivo and in vitro. Furthermore, HD-1 L further alleviated alcohol-induced inflammation after blocking BRD2 with inhibitor (JQ1) or BRD2 small interfering (si)-RNA in RAW264.7 cells. Besides, HD-1 L failed to effectively exert its anti-inflammatory effects after over expression of BRD2. In addition, HD-1 L significantly inhibited the phosphorylation and activation of NF-κB-P65 mediated by BRD2. In conclusion, HD-1 L alleviated liver injury and inflammation mainly by inhibiting BRD2-NF-κB signaling pathway, and HD-1 L may be a potential anti-inflammatory compound in treatment of alcoholic liver disease.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Hesperidin/pharmacology , Inflammation/drug therapy , Liver/drug effects , Liver/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents/pharmacology , Aspartate Aminotransferases/blood , Cholesterol/blood , Cytokines/metabolism , Hesperidin/chemistry , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction/drug effects , Triglycerides/blood
7.
Toxicol Lett ; 355: 88-99, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34838997

ABSTRACT

Liver fibrosis is a reversible wound healing reaction characterized by abnormal accumulation of extracellular matrix (ECM) in response to liver injury. Recent studies have shown that it can be epigenetically regulated, especially by microRNAs (miRNAs). It has been acknowledged that activation of hepatic stellate cells (HSCs) is a pivotal step in the initiation and progression of liver fibrosis. Notably, our results showed that miR-195-3p was increased in HSCs isolated from CCl4-treated mice and that the increase was more pronounced as the degree of liver fibrosis increased. Moreover, treatment of LX-2 cells, a human immortalized hepatic stellate cell line, with TGF-ß1 resulted remarkable upregulation of miR-195-3p. Gain-of-function and loss-of-function experiments have suggested that the increased levels of miR-195-3p inhibit the expression of phosphatase and tension homolog deleted on chromosome 10 (PTEN), a negative regulator of the PI3K/Akt/mTOR signaling pathway in liver fibrosis, thereby contributing to HSC activation and proliferation and promoting the expression of profibrotic genes, such as α-SMA and collagen I, in LX-2 cells, which accelerates the accumulation of fibrous extracellular matrix deposition in the liver, while knockdown of miR-195-3p induced the opposite effect. Taken together, these results provide evidence for the harmful role of miR-195-3p in CCl4-treated mouse liver fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/chemically induced , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , 3' Untranslated Regions , Animals , Carbon Tetrachloride Poisoning/pathology , Cell Line , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation
8.
Eur J Pharmacol ; 915: 174530, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34902361

ABSTRACT

Liver fibrosis, a chronic inflammatory healing reaction, progresses to hepatocirrhosis without effective intervention. Hesperetin derivative (HD-16), a monomer compound derived from hesperitin, exerts anti-inflammatory and hepatoprotective effects against a spectrum of liver diseases. However, the anti-fibrotic potential of HD-16 in liver fibrosis and its underlying mechanism have not yet been elucidated. In this study, we investigated the anti-fibrotic effect of HD-16 on mouse liver fibrosis induced by CCl4 and on LX-2 cells (human immortalized HSCs) stimulated by TGF-ß1, in vivo and in vitro. HD-16 exerted an anti-fibrotic effect via regulation of the AMPK/SIRT3 pathway. Pharmacodynamic results showed that HD-16 alleviated the degree of injury and inflammation in CCl4-induced mouse liver fibrosis. Consistently, HD-16 also effectively inhibited the expression of α-SMA, Col1α1, Col3α1, and TIMP-1 in TGF-ß1-activated LX-2 cells. Mechanistically, HD-16 promoted SIRT3 expression and activity in fibrotic liver and activated LX-2 cells. Furthermore, SIRT3 depletion attenuated the anti-fibrotic effects of HD-16. Intriguingly, HD-16 increased AMPK phosphorylation, whereas inhibition of SIRT3 expression did not affect AMPK phosphorylation. In contrast, AMPK silencing suppressed SIRT3 expression, suggesting that SIRT3 is a downstream target of AMPK in liver fibrosis. Overall, HD-16 attenuated CCl4-induced liver inflammation and fibrosis by activating the AMPK/SIRT3 pathway, and HD-16 may be a potential anti-fibrotic compound in the treatment of liver fibrosis.


Subject(s)
Hesperidin
9.
Front Pharmacol ; 12: 741151, 2021.
Article in English | MEDLINE | ID: mdl-34887753

ABSTRACT

CircRNAs (circRNAs) are commonly dysregulated in a variety of human diseases and are involved in the development and progression of cancer. However, the role of circRNAs in hepatic fibrosis (HF) is still unclear. Our previous high throughput screen revealed changes in many circRNAs in mice with carbon tetrachloride (CCl4)-induced HF. For example, circCREBBP was significantly down-regulated in primary hepatic stellate cells (HSCs) and liver tissue of HF mice induced by CCl4 compared to those in the vehicle group. Overexpression of circCREBBP with AAV8-circCREBBP in vivo prevented CCl4-induced HF worsening by reducing serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) contents, liver hydroxyproline levels, collagen deposition, and levels of pro-fibrosis genes and pro-inflammatory cytokines. Furthermore, in vitro function loss and function gain analysis showed that circCREBBP inhibited HSCs activation and proliferation. Mechanically, circCREBBP acts as a sponge for hsa-miR-1291 and subsequently promotes LEFTY2 expression. In conclusion, our current results reveal a novel mechanism by which circCREBBP alleviates liver fibrosis by targeting the hsa-miR-1291/LEFTY2 axis, and also suggest that circCREBBP may be a potential biomarker for heart failure.

10.
FASEB J ; 35(7): e21700, 2021 07.
Article in English | MEDLINE | ID: mdl-34105828

ABSTRACT

Histone deacetylases (HDACs), especially HDAC2, play a role in alleviating liver fibrosis; however, the specific upstream regulation mechanism is unknown. Herein, TargetScan was used to predict the potential upstream targets of HDAC2, and the role of miR-455-3p was explored. The dual luciferase reporter assay showed that miR-455-3p binds to the 3' UTR of HDAC2 mRNA. Additionally, miR-455-3p was downregulated in the liver tissues of patients with cirrhosis and mice with liver fibrosis, as well as in primary HSCs isolated from fibrotic mouse livers and TGF-ß-treated LX-2 cells. In contrast, it is highly expressed in the reversal stage of hepatic fibrosis and MDI-cultured LX-2 cells. Our functional analyses showed that miR-455-3p overexpression facilitated apoptosis and reduced the expression of pro-fibrotic markers and the proliferation of activated LX-2 cells. On the contrary, miR-455-3p inhibition converted inactivated LX-2 cells into activated, proliferative, fibrogenic cells. Interestingly, restoration of HDAC2 expression partially blocked the function of miR-455-3p. Downregulated miR-455-3p expression can be restored by DNA methyltransferases in activated LX-2 cells. Methylation-specific PCR, bisulfite sequencing PCR, and chromatin immunoprecipitation assays indicated that the methylation level of miR-455-3p promoter CpG islands was elevated in TGF-ß-treated LX-2 cells and that miR-455-3p was downregulated in activated LX-2 cells by DNA hypermethylation, which is mediated by DNMT3b and DNMT1. In conclusion, miR-455-3p acts as a liver fibrosis suppressor by targeting HDAC2, and its deficiency further aggravates the reversal phase of fibrosis. Thus, the epigenetics mediated miR-455-3p/HDAC2 axis may serve as a novel potential therapeutic target for clinical treatment of hepatic fibrosis.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Histone Deacetylase 2/metabolism , Liver Cirrhosis/prevention & control , MicroRNAs/genetics , Animals , Apoptosis , Carbon Tetrachloride/toxicity , Cell Proliferation , Hepatic Stellate Cells/cytology , Histone Deacetylase 2/genetics , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
11.
Mol Ther Nucleic Acids ; 23: 847-862, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33614234

ABSTRACT

Recently, circular RNAs (circRNAs) have been frequently reported to be involved in hepatocellular carcinoma (HCC) development and progression. However, the role of circRNAs in hepatic fibrosis (HF) is still unclear. Our previous high-throughput screen revealed changes in many circRNAs in mice with carbon tetrachloride (CCl4)-induced HF. For instance, the expression of circPSD3, a circRNA derived from the Pleckstrin and Sec7 domain-containing 3 (PSD3) gene, was considerably downregulated in primary hepatic stellate cells (HSCs) and liver tissues of mice with CCl4-induced HF compared to those in the vehicle group. In vivo overexpression of circPSD3 using AAV8-circPSD3 arrested the deterioration of CCl4-induced HF as indicated by reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) content, liver hydroxyproline level, collagen deposition, and pro-fibrogenic gene and pro-inflammatory cytokine levels. Moreover, in vitro loss-of-function and gain-of-function analyses suggested that circPSD3 inhibited the activation and proliferation of HSCs. Mechanistically, circPSD3 served as a sponge for miR-92b-3p, subsequently promoting the expression of Smad7. In conclusion, our present findings reveal a novel mechanism by which circPSD3 alleviates hepatic fibrogenesis by targeting the miR-92b-3p/Smad7 axis, and they also indicate that circPSD3 may serve as a potential biomarker for HF.

12.
Int Immunopharmacol ; 88: 106968, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33182058

ABSTRACT

Alcohol-induced liver injury is characterized by abnormal liver dysfunction and excessive inflammation response. Recent years a wealth of data have been yielded indicating that EtOH (ethyl alcohol)-induced macrophage activation along with liver inflammation plays a dominating role in the progression of alcohol-induced liver injury. Here we found high expression of NLRP12 (Nucleotide-binding oligomerization domain protein 12, which is generally considered to be a negative regulator of inflammatory response) in EtOH-fed mouse liver tissue, primary Kupffer cells and EtOH-induced RAW264.7 cells. Additionally, overexpression of NLRP12 following Ad (adenovirus)-NLRP12-EGFP contributed to the attenuation of steatosis and inflammation in EtOH-fed mice model and EtOH-primed RAW264.7 cells. In parallel, Knockdown of NLRP12 aggravated the inflammatory response in RAW264.7 cells triggered by EtOH. Meanwhile, after administration of overexpression or inhibition of NLRP12 expression in vitro, the expression of phosphorylated protein of NF-kB signaling pathway was significantly affected. After increasing or decreasing the expression of NLRP12 in RAW264.7 cells, AML-12 cells were cultured with the supernatant of RAW264.7 cells stimulated by EtOH, and the percent of apoptosis ratio of AML-12 cells was remarkably altered. The study suggested that reduced inflammatory response induced by NLRP12-mediated inhibition of NF-kB pathway participated in the decrease of hepatocyte apoptosis in alcohol-induced liver injury. Collectively, these findings suggested the significance of NLRP12-mediated macrophage activation in alcohol-induced liver injury.


Subject(s)
Apoptosis , Hepatocytes/immunology , Intracellular Signaling Peptides and Proteins/immunology , Liver Diseases, Alcoholic/immunology , Macrophage Activation , NF-kappa B/immunology , Animals , Cell Line , Cytokines/immunology , Ethanol , Liver/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
13.
Liver Int ; 40(11): 2612-2626, 2020 11.
Article in English | MEDLINE | ID: mdl-32745314

ABSTRACT

With advances in genome and transcriptome research technology, the function and mechanism of lncRNAs in physiological and pathological states have been gradually revealed. Nuclear Enriched Abundant Transcript 1 (NEAT1, a long non-coding RNA), a vital component of paraspeckles, plays an indispensable role in the formation and integrity of paraspeckles. Throughout the research history, NEAT1 is mostly aberrantly upregulated in various cancers, and high expression of NEAT1 often contributes to poor prognosis of patients. Notably, the role and mechanism of NEAT1 in liver diseases have been increasingly reported. NEAT1 accelerates the progression of non-alcoholic fatty liver disease (NAFLD), liver fibrosis and hepatocellular carcinoma, while exerting a protective role in the pathogenesis of acute-on-chronic liver failure by inhibiting the inflammatory response. In this review, we will elaborate on relevant studies on the different casting of NEAT1 in liver diseases, especially focusing on its regulatory mechanisms and new opportunities for alcoholic liver disease.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , RNA, Long Noncoding , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/genetics , Non-alcoholic Fatty Liver Disease/genetics , RNA, Long Noncoding/genetics
14.
Mol Immunol ; 126: 31-39, 2020 10.
Article in English | MEDLINE | ID: mdl-32745796

ABSTRACT

Activated hepatic stellate cells (HSCs) are the major cell type involved in the deposition of extracellular matrix (ECM) during the development of hepatic fibrosis. In this study, we revealed that left-right determination factor 2 (LEFTY2), one of the proteins belonging to the transforming growth factor-ß (TGF-ß) protein superfamily, was remarkedly decreased in human hepatic fibrosis tissues and in a carbon tetrachloride (CCl4)-induced liver fibrosis mouse model. In addition, TGF-ß1 treatment markedly reduced the level of LEFTY2 in HSCs. Importantly, overexpression of LEFTY2 suppressed the activation and proliferation of HSCs. LEFTY2 inhibited the expression of TGF-ß1-induced fibrosis-associated genes (α-SMA and COL1a1) in human (LX-2) and rat (HSC-T6) HSC cell lines in vitro. Mechanistically, we demonstrated, for the first time, the role of LEFTY2 in inhibiting TGF-ß1/Smad3 signaling, suggesting that there is a mutual antagonism between LEFTY2 and TGF-ß1/Smad3 signaling during liver fibrosis. Similarly, we observed that LEFTY2 has a negative effect on its downstream genes, including c-MYC, CDK4, and cyclin D1, in liver fibrosis. Collectively, our data strongly indicated that LEFTY2 plays an important role in controlling the proliferation and activation of HSCs in the progression of liver fibrosis and this could be a potential therapeutic target for its treatment.


Subject(s)
Hepatic Stellate Cells/pathology , Left-Right Determination Factors/metabolism , Liver Cirrhosis/pathology , Liver/pathology , Aged , Animals , Carbon Tetrachloride/toxicity , Cell Line , Cell Proliferation , Down-Regulation , Female , Humans , Liver/drug effects , Liver Cirrhosis/chemically induced , Male , Mice , Middle Aged , Rats , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
15.
J Cell Mol Med ; 24(13): 7405-7416, 2020 07.
Article in English | MEDLINE | ID: mdl-32463161

ABSTRACT

As an outcome of chronic liver disease, liver fibrosis involves the activation of hepatic stellate cells (HSCs) caused by a variety of chronic liver injuries. It is important to explore approaches to inhibit the activation and proliferation of HSCs for the treatment of liver fibrosis. PLK1 is overexpressed in many human tumour cells and has become a popular drug target in tumour therapy. Therefore, further study of the function of PLK1 in the cell cycle is valid. In the present study, we found that PLK1 expression was elevated in primary HSCs isolated from CCl4 -induced liver fibrosis mice and LX-2 cells stimulated with TGF-ß1. Knockdown of PLK1 inhibited α-SMA and Col1α1 expression and reduced the activation of HSCs in CCl4 -induced liver fibrosis mice and LX-2 cells stimulated with TGF-ß1. We further showed that inhibiting the expression of PLK1 reduced the proliferation of HSCs and promoted HSCs apoptosis in vivo and in vitro. Furthermore, we found that the Wnt/ß-catenin signalling pathway may be essential for PLK1-mediated HSCs activation. Together, blocking PLK1 effectively suppressed liver fibrosis by inhibiting HSC activation, which may provide a new treatment strategy for liver fibrosis.


Subject(s)
Cell Cycle Proteins/metabolism , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/pathology , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Signaling Pathway , Animals , Apoptosis , Carbon Tetrachloride , Cell Line , Cell Proliferation , Humans , Male , Mice, Inbred C57BL , Models, Biological , Transforming Growth Factor beta1/metabolism , Up-Regulation , Polo-Like Kinase 1
16.
Theranostics ; 10(11): 4851-4870, 2020.
Article in English | MEDLINE | ID: mdl-32308754

ABSTRACT

Rationale: Circular RNAs (circRNAs) are a new form of noncoding RNAs that play crucial roles in various pathological processes. However, the expression profile and function of circRNAs in hepatic fibrosis (HF) remain largely unknown. In this study, we show a novel circFBXW4 mediates HF via targeting the miR-18b-3p/FBXW7 axis. Methods: We investigated the expression profile of circRNAs, microRNAs and mRNAs in hepatic stellate cells (HSCs) from HF progression and regression mice by circRNAs-seq and microarray analysis. We found a significantly dysregulated circFBXW4 in HF. Loss-of-function and gain-of-function analysis of circFBXW4 were performed to assess the role of circFBXW4 in HF. Furthermore, we confirmed that circFBXW4 directly binds to miR-18b-3p by luciferase reporter assay, RNA pull down and fluorescence in situ hybridization analysis. Results: We found that circFBXW4 downregulated in liver fibrogenesis. Enforcing the expression of circFBXW4 inhibited HSCs activation, proliferation and induced apoptosis, attenuated mouse liver fibrogenesis injury and showed anti-inflammation effect. Mechanistically, circFBXW4 directly targeted to miR-18b-3p to regulate the expression of FBXW7 in HF. Conclusions: circFBXW4 may act as a suppressor of HSCs activation and HF through the circFBXW4/miR-18b-3p/FBXW7 axis. Our findings identify that circFBXW4 serves as a potential biomarker for HF therapy.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/metabolism , Liver Cirrhosis/prevention & control , MicroRNAs/genetics , RNA, Circular/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Down-Regulation , F-Box-WD Repeat-Containing Protein 7/genetics , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
17.
Toxicol Lett ; 319: 11-21, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31711802

ABSTRACT

Alcoholic liver injury (ALI) is a part of alcohol-related liver diseases. These diseases include steatohepatitis, alcoholic fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Accumulating data indicates that alcohol metabolism and circulating endotoxin/lipopolysaccharide (LPS) contribute to macrophage activation, which leads to the development of ALI. Protein tyrosine phosphatase 1B (PTP1B) has been shown to be involved in many tissue inflammations as well as liver fibrosis; however, the role of PTP1B in ALI is still unclear. In this study, PTP1B expression was elevated in liver tissues and primary macrophages isolated from EtOH-fed mice. Moreover, PTP1B expression was elevated in RAW264.7 cells stimulated with alcohol and LPS. Additional studies showed that silencing of PTP1B reduced the inflammatory response and expression of inflammatory cytokines such as IL-1ß, IL-6 and TNF-α, while overexpression of PTP1B induced inflammation in RAW264.7 cells. In addition, we found that NF-κB pathway was activated in RAW264.7 cells stimulated with alcohol and LPS, and PTP1B silencing or overexpression could regulate NF-κB signaling. In conclusion, this study revealed the function of PTP1B in ALI via its regulation of the NF-κB signaling pathway and may provide theoretical support for further research on ALI.


Subject(s)
Liver Diseases, Alcoholic/genetics , Macrophage Activation , NF-kappa B/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction/genetics , Animals , Central Nervous System Depressants/pharmacology , Cytokines/biosynthesis , Ethanol/pharmacology , Lipopolysaccharides/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , RAW 264.7 Cells , Up-Regulation
18.
Int Immunopharmacol ; 76: 105838, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31473406

ABSTRACT

Hepatic fibrosis, a common pathological feature and leading cause of various chronic liver diseases, still lacks effective therapy. Hesperetin derivative (HD) is a derivative of Traditional Chinese Medicine monomer isolated from the fruit peel of Citrusaurantium L. (Rutaceae). In the present study, we revealed the anti-fibrotic effects of HD in CCl4-induced mouse hepatic fibrosis model and in TGF-ß1-activated LX-2 cells, in vivo and in vitro. Results showed that HD prevented CCl4-induced liver injury and histological damage. Consistently, HD inhibited the up-regulation of liver fibrogenesis markers α-SMA, Col1α1, Col3α1 and TIMP-1 in primary hepatic stellate cells (HSCs) and suppressed inflammatory responses in primary liver macrophages from hepatic fibrosis mice. Furthermore, HD promoted the apoptosis of activated HSCs, a key step in the onset of fibrosis regression. Mechanistically, the Hedgehog pathway was involved in HD-treated hepatic fibrosis, and HD specifically contributed to attenuate the aberrant expression of Glioma associated oncogene-1 (Gli-1). Interestingly, blockade of Gli-1 removed the inhibitory effect of HD on activated HSCs, indicating that Gli-1 may play a pivotal role in mediating the anti-fibrotic effect of HD in hepatic fibrosis. Collectively, our results suggest that HD may be a potential anti-fibrotic Traditional Chinese Medicine monomer for the treatment of hepatic fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Hesperidin/therapeutic use , Liver Cirrhosis/drug therapy , Protective Agents/therapeutic use , Animals , Apoptosis/drug effects , Carbon Tetrachloride , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Cytokines/genetics , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hesperidin/pharmacology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Protective Agents/pharmacology , Transforming Growth Factor beta1/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
19.
Oncol Lett ; 17(2): 1401-1408, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30675193

ABSTRACT

The incidence of cancer is increasing at an alarming rate despite recent advances in prevention strategies, diagnostics and therapeutics for various types of cancer. The identification of novel biomarkers to aid in prognosis and treatment for cancer is urgently required. Uncontrolled proliferation and dysregulated apoptosis are characteristics exhibited by cancer cells in the initiation of various types of cancer. Notably, aberrant expression of crucial oncogenes or cancer suppressors is a defining event in cancer occurrence. Research has demonstrated that SAD1/UNC84 domain protein-2 (SUN2) serves a suppressive role in breast cancer, atypical teratoid/rhabdoid tumors and lung cancer progression. Furthermore, SUN2 inhibits cancer cell proliferation, migration and promotes apoptosis. Recent reports have also shown that SUN2 serves prominent roles in resistance to the excessive DNA damage that destabilizes the genome and promotes cancer development, and these functions of SUN2 are critical for evading initiation of cancer. Additionally, increasing evidence has demonstrated that SUN2 is involved in maintaining cell nuclear structure and appears to be a central component for organizing the natural nuclear architecture in cancer cells. The focus of the present review is to provide an overview on the pharmacological functions of SUN2 in cancers. These findings suggest that SUN2 may serve as a promising therapeutic target and novel predictive marker in various types of cancer.

20.
Mol Immunol ; 107: 29-40, 2019 03.
Article in English | MEDLINE | ID: mdl-30639476

ABSTRACT

Yes-associated protein (YAP) is a significant downstream protein in the Hippo signaling pathway with important functions in cell proliferation, apoptosis, invasion and migration. YAP also plays a role in the progression and development of various liver diseases. In hepatic fibrosis development and reversion, the proliferation and apoptosis of activated hepatic stellate cells (HSCs) play a critical role. However, the contribution of YAP to hepatic fibrosis progression and reversion and the underlying mechanism have not been investigated. Here we investigated the expression and function of YAP in the proliferation and apoptosis of activated HSCs. We found that YAP expression was increased in liver fibrosis tissues from CCl4-induced model mice and restored to normal level after stopping CCl4 injection and 6 weeks of spontaneously recovery. YAP expression was elevated in HSC-T6 cells treated with TGF-ß1 and recovered after MDI treatment. Silencing of YAP inhibited the activation and proliferation of HSC-T6 cells stimulated by TGF-ß1. In addition, the apoptosis of activated HSC-T6 cells silenced for YAP was slightly enhanced. Furthermore, over-expression of YAP repressed the reversion of activated HSC-T6 cells mediated by MDI reversal. We found that HSC-T6 cells activated by TGF-ß1 showed higher levels of nuclear YAP compared with MDI-treated cells, indicating that YAP was activated in HSC-T6 cells treated by TGF-ß1. We also found that loss of YAP attenuated Wnt/ß-catenin pathway activity in activated HSC-T6 cells. Treatment of VP, an inhibitor of the YAP-TEAD complex, reduced both activation and proliferation of HSC-T6 cells and increased apoptosis. Together these results indicated that reduced expression of YAP contributes to acquisition of the quiescent phenotype in HSCs. Our results suggest that YAP may be a useful target in HSCs activation and reversion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Phosphoproteins/metabolism , Animals , Apoptosis/drug effects , Carbon Tetrachloride , Cell Cycle Proteins , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Gene Silencing/drug effects , Hepatic Stellate Cells/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Models, Biological , Transforming Growth Factor beta1/metabolism , Verteporfin/pharmacology , Wnt Signaling Pathway/drug effects , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...