Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacol Res ; 177: 106125, 2022 03.
Article in English | MEDLINE | ID: mdl-35149186

ABSTRACT

Alcohol-induced liver injury (ALI) is associated with inflammatory responses regulated by macrophages. Activation of macrophages plays a crucial role in ALI while DNA methylation-regulated gene silencing is associated with inflammation processes in macrophages. Proline-Serine-Threonine Phosphatase Interacting Protein 2 (PSTPIP2), which belongs to the Fes/CIP4 homology-Bin/Amphiphysin/Rvs domain family of proteins and plays a role in macrophages. Previous studies have shown that Pstpip2 can be methylated. Herein, its expression was found to be significantly downregulated in primary liver macrophages isolated from EtOH-fed mice and EtOH-induced RAW264.7 cells. Overexpression of PSTPIP2 using liver-specific recombinant AAV serotype 9 (rAAV9)-PSTPIP2 in EtOH-fed mice dramatically alleviated liver injury and inflammatory responses. In addition, silencing of PSTPIP2 aggravated the alcohol-induced inflammatory response in vitro. Mechanistically, PSTPIP2 might affect macrophage-induced inflammatory responses by regulating the STAT1 and NF-κB signaling pathways. The downregulation of PSTPIP2 in ALI may be associated with DNA methylation. Methylation-specific PCR and western blotting analyses showed that EtOH induced abnormal DNA methylation patterns and increased the protein expression levels of DNMT1, DNMT3a, and DNMT3b. The chromatin immunoprecipitation assay showed that DNMT3a could directly bind to the Pstpip2 promoter and act as a principal regulator of PSTPIP2 expression. Moreover, silencing of DNMT3a significantly restored the EtOH-induced low expression of PSTPIP2 and inhibited EtOH-induced inflammation. Overall, these findings provide a detailed understanding of the possible functions and mechanisms of PSTPIP2 in ALI, thus providing new substantive research to elucidate the pathogenesis of ALI and investigate potential targeted treatment strategies.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , NF-kappa B , Animals , Chemical and Drug Induced Liver Injury, Chronic/genetics , DNA Methylation , DNA Modification Methylases/genetics , Ethanol/toxicity , Inflammation/genetics , Mice , NF-kappa B/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
2.
Front Immunol ; 12: 585412, 2021.
Article in English | MEDLINE | ID: mdl-34262554

ABSTRACT

Proline-serine-threonine-phosphatase-interacting protein 2 (PSTPIP2) belongs to the Fes/CIP4 homology-Bin/Amphiphysin/Rvs (F-BAR) domain family. It exhibits lipid-binding, membrane deformation, and F-actin binding activity, suggesting broader roles at the membrane-cytoskeleton interface. PSTPIP2 is known to participate in macrophage activation, neutrophil migration, cytokine production, and osteoclast differentiation. In recent years, it has been observed to play important roles in innate immune diseases and autoinflammatory diseases (AIDs). Current research indicates that the protein tyrosine phosphatase PTP-PEST, Src homology domain-containing inositol 5'-phosphatase 1 (SHIP1), and C-terminal Src kinase (CSK) can bind to PSTPIP2 and inhibit the development of AIDs. However, the mechanisms underlying the function of PSTPIP2 have not been fully elucidated. This article reviews the research progress and mechanisms of PSTPIP2 in AIDs. PSTPIP2 also provides a new therapeutic target for the treatment of AIDs.


Subject(s)
Inflammation/genetics , Inflammation/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/immunology , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/genetics , Humans , Inflammation/physiopathology , Mice , Phosphorylation , Protein Binding , Signal Transduction
3.
Int Immunopharmacol ; 95: 107471, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33756231

ABSTRACT

Alcoholic liver disease (ALD) is the most common chronic liver disease worldwide. Currently, there is no definitive treatment for alcohol-induced liver injury (ALI). Inflammatory response and oxidative stress play a crucial role in ALI. Cyclooxygenase 2 (COX-2) can be induced by inflammation and it has been reported that the enhanced expression of COX-2 in alcoholic liver injury. Rutaecarpine (RUT) was extracted from evodia rutaecarpa. RUT has a wide range of pharmacological activities. In order to increase its anti-inflammatory activity, our group introduced sulfonyl group to synthesized the 3-[2-(trifluoromethoxy)benzenesulfonamide]-rutaecarpine (3-B-RUT). In this study, we explored the protective effect of 3-B-RUT on alcoholic liver injury in vivo and in vitro and preliminarily explore its mechanism. Mice ALI model was established according to the chronic-plus-binge ethanol model. Results showed that 3-B-RUT (20 µg/kg) attenuated alcohol-induced liver injury and suppressed liver inflammation and oxidative stress, and the effect was comparable to RUT (20 mg/kg). In vitro results are consistent with in vivo results. Mechanistically, the 3-B-RUT might suppress inflammatory response and oxidative stress by regulating activation of NF-κB/COX-2 pathway. In summary, 3-B-RUT, a derivative of RUT, may be a promising clinical candidate for ALI treatment.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Indole Alkaloids/therapeutic use , Liver Diseases, Alcoholic/drug therapy , Quinazolines/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Indole Alkaloids/pharmacology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Diseases, Alcoholic/immunology , Liver Diseases, Alcoholic/pathology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Oxidative Stress/drug effects , Quinazolines/pharmacology , RAW 264.7 Cells
4.
Clin Sci (Lond) ; 134(14): 1935-1956, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32639005

ABSTRACT

The regulation of macrophages during inflammatory responses is a crucial process in alcoholic liver disease (ALD) and aberrant macrophage DNA methylation is associated with inflammation. Our preliminary screening results of macrophage methylation in the present study demonstrated the zinc finger SWI2/SNF2 and MuDR (SWIM)-domain containing 3 (ZSWIM3) were hypermethylated in the 5' untranslated region (5'-UTR) region. ZSWIM3, a novel zinc finger-chelate domain of SWIM, is predicted to function in DNA-binding and protein-binding interactions. Its expression was found to be consistently decreased in macrophages isolated from livers of ethyl alcohol (EtOH)-fed mice and in EtOH+lipopolysaccharide (LPS)-induced RAW264.7 cells. Over-expression of ZSWIM3 was found to attenuate chronic+binge ethanol feeding-induced liver injury and inhibit inflammatory responses in vivo. Enforced expression of ZSWIM3 in vitro was also found to have anti-inflammatory effects. Aberrant expression of ZSWIM3 in alcohol-induced liver injury (ALI) was found to be associated with hypermethylation. Analysis of CpG prediction indicated the presence of two methylated sites in the ZSWIM3 promoter region and methylation inhibitor and DNA methyltransferases (DNMTs)-siRNA transfection were found to restore down-regulated ZSWIM3. Chromatin immunoprecipitation (ChIP) assay and molecular docking affirmed the role of DNMT 3b (DNMT3b) as a principal regulator of ZSWIM3 expression. Mechanistically, ZSWIM3 might affect inflammation by binding with tumor necrosis factor receptor-associated factor 2 (TRAF2), which further mediates the activation of the nuclear transcription factor κB (NF-κB) pathway. The present study, therefore, provides detailed insights into the possible structure and function of ZSWIM3 and thus, contributes new substantial research in the elucidation of the pathogenesis of ALI.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Liver Diseases, Alcoholic/metabolism , Macrophages/metabolism , Animals , DNA Methylation , Disease Models, Animal , Liver Diseases, Alcoholic/immunology , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , NF-kappa B/metabolism , TNF Receptor-Associated Factor 2/metabolism , DNA Methyltransferase 3B
5.
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
6.
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
7.
Int Immunopharmacol ; 75: 105671, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31377590

ABSTRACT

Liver fibrosis is the representative features of liver chronic inflammation and the characteristic of early cirrhosis. To date, effective therapy for liver fibrosis is lacking. Recently, Traditional Chinese Medicine (TCM) has attracted increasing attention due to its wide pharmacological effects and more uses in clinical. Wogonin, as one major active constituent of Scutellaria radix, has been reported it plays an important role in anti-inflammatory, anti-cancer, anti-viral, anti-angiogenesis, anti-oxidant and neuro-protective effects. However, the anti-fibrotic effect of wogonin is never covered in liver. In this study, we evaluated the protect effect of wogonin in liver fibrosis. Wogonin significantly attenuated liver fibrosis both in CCl4-induced mice and TGF-ß1 activated HSCs. Meanwhile, wogonin can enhances apoptosis of TGF-ß1 activated HSC-T6 cell from rat and LX-2 cell from human detected by flow cytometry. Additionally, wogonin can largely enhances cle-caspase3, cle-caspase9 expression and the ratio of Bax/Bcl-2 in T6 cells. Pro-apoptosis effect of wogonin in vivo was further verified in situ. In conclusion, wogonin can attenuate liver fibrosis via regulating the activation and apoptosis of hepatic stellate cells, and may be an effective drug to treat and prevent liver fibrosis.


Subject(s)
Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/drug therapy , Flavanones/therapeutic use , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Animals , Carbon Tetrachloride , Cell Line , Flavanones/pharmacology , Humans , Male , Mice, Inbred C57BL , Rats , Transforming Growth Factor beta1
8.
J Cell Physiol ; 234(9): 14709-14720, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30701547

ABSTRACT

Alcoholic liver disease (ALD) is a complex process with high morbitity and can cause liver dysfunction, which contains a wide spectrum of hepatic lesions, including steatohepatitis, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. To date, the molecular mechanisms for ALD have not been fully explored and an effective therapy is still missing. Overwhelming evidence shows dysregulation of noncoding RNAs (ncRNAs), particularly microRNAs (miRNAs), is correlated with etiopathogenesis and progress of ALD including hepatocyte damage, disrupted lipid metabolism, aggressive inflammatory responses, oxidative stress, programmed cell death, fibrosis, and epigenetic changes induced by alcohol. For example, circulating miRNA-122 is a marker of hepatocyte damage, and miRNA-155 is a potential marker of inflammation, indicating their diagnosis therapeutic potential in ALD. In addition, roles for long noncoding RNAs (lncRNAs) and circular RNAs in ALD are being uncovered. Further, circulating ncRNAs and exosome-derived ncRNAs have attracted more attention lately, suggesting a role in the prevention and treatment of ALD. This review covers the roles of ncRNAs in ALD, and the potential uses as markers for diagnosis and therapeutic options.

SELECTION OF CITATIONS
SEARCH DETAIL
...