Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Commun Signal ; 21(1): 162, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37381005

ABSTRACT

BACKGROUND: Adherens junctions (AJs) facilitate cell-cell contact and contribute to cellular communication as well as signaling under physiological and pathological conditions. Aberrant expression of AJ proteins is frequently observed in human cancers; however, how these factors contribute to tumorigenesis is poorly understood. In addition, for some factors such as α-catenin contradicting data has been described. In this study we aim to decipher how the AJ constituent α-catenin contributes to liver cancer formation. METHODS: TCGA data was used to detect transcript changes in 23 human tumor types. For the detection of proteins, liver cancer tissue microarrays were analyzed by immunohistochemistry. Liver cancer cell lines (HLF, Hep3B, HepG2) were used for viability, proliferation, and migration analyses after RNAinterference-mediated gene silencing. To investigate the tumor initiating potential, vectors coding for α-catenin and myristoylated AKT were injected in mice by hydrodynamic gene delivery. A BioID assay combined with mass spectrometry was performed to identify α-catenin binding partners. Results were confirmed by proximity ligation and co-immunoprecipitation assays. Binding of transcriptional regulators at gene promoters was investigated using chromatin-immunoprecipitation. RESULTS: α-catenin mRNA was significantly reduced in many human malignancies (e.g., colon adenocarcinoma). In contrast, elevated α-catenin expression in other cancer entities was associated with poor clinical outcome (e.g., for hepatocellular carcinoma; HCC). In HCC cells, α-catenin was detectable at the membrane as well as cytoplasm where it supported tumor cell proliferation and migration. In vivo, α-catenin facilitated moderate oncogenic properties in conjunction with AKT overexpression. Cytokinesis regulator centrosomal protein 55 (CEP55) was identified as a novel α-catenin-binding protein in the cytoplasm of HCC cells. The physical interaction between α-catenin and CEP55 was associated with CEP55 stabilization. CEP55 was highly expressed in human HCC tissues and its overexpression correlated with poor overall survival and cancer recurrence. Next to the α-catenin-dependent protein stabilization, CEP55 was transcriptionally induced by a complex consisting of TEA domain transcription factors (TEADs), forkhead box M1 (FoxM1), and yes-associated protein (YAP). Surprisingly, CEP55 did not affect HCC cell proliferation but significantly supported migration in conjunction with α-catenin. CONCLUSION: Migration-supporting CEP55 is induced by two independent mechanisms in HCC cells: stabilization through interaction with the AJ protein α-catenin and transcriptional activation via the FoxM1/TEAD/YAP complex.


Cell­cell contact in epithelial cells is important for cell polarity, cellular compartmentalisation, as well as tissue architecture during development, homeostasis, and regeneration of adult tissues in metazoans. In this context, adherens junctions (AJs) mechanically sense cell contact information with direct impact on cytoskeletal remodelling, the regulation of signalling pathways, and eventually cell biology. Indeed, the loss of cell­cell contact and cellular polarity are key features in human carcinogenesis and important pathological parameters for the identification of many epithelial tumors.We demonstrate in this study, that overexpression of the AJ constituent α­catenin is frequently observed in human hepatocellular carcinoma (HCC). α­catenin supports HCC cell proliferation and migration. Together with the oncogene AKT, α­catenin moderately facilitates tumor initiation in mouse livers. Using mass spectrometry, we identified several new α­catenin interaction partners in the cytosol of liver cancer cells, including the cytokinesis regulator centrosomal protein 55 (CEP55). CEP55 mediates pro-migratory effects and its overexpression in HCC cells is controlled by two molecular mechanisms: α­catenin-dependent protein stabilization and transcriptional induction by the TEA domain transcription factors (TEADs)/forkhead box M1 (FoxM1)/yes-associated protein (YAP) complex.In summary, we here describe a new mechanism how changes in cell­cell contact support liver cancer formation and progression. This study demonstrates that dysregulation of the AJ component α­catenin contributes to liver carcinogenesis via distinct molecular mechanisms. Video Abstract.


Subject(s)
Adenocarcinoma , Carcinoma, Hepatocellular , Cell Cycle Proteins , Colonic Neoplasms , Liver Neoplasms , Animals , Humans , Mice , alpha Catenin , Cell Line , Cell Movement , Neoplasm Recurrence, Local , Proto-Oncogene Proteins c-akt
2.
Elife ; 112022 10 18.
Article in English | MEDLINE | ID: mdl-36255405

ABSTRACT

The Hippo signaling pathway controls cell proliferation and tissue regeneration via its transcriptional effectors yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). The canonical pathway topology is characterized by sequential phosphorylation of kinases in the cytoplasm that defines the subcellular localization of YAP and TAZ. However, the molecular mechanisms controlling the nuclear/cytoplasmic shuttling dynamics of both factors under physiological and tissue-damaging conditions are poorly understood. By implementing experimental in vitro data, partial differential equation modeling, as well as automated image analysis, we demonstrate that nuclear phosphorylation contributes to differences between YAP and TAZ localization in the nucleus and cytoplasm. Treatment of hepatocyte-derived cells with hepatotoxic acetaminophen (APAP) induces a biphasic protein phosphorylation eventually leading to nuclear protein enrichment of YAP but not TAZ. APAP-dependent regulation of nuclear/cytoplasmic YAP shuttling is not an unspecific cellular response but relies on the sequential induction of reactive oxygen species (ROS), RAC-alpha serine/threonine-protein kinase (AKT, synonym: protein kinase B), as well as elevated nuclear interaction between YAP and AKT. Mouse experiments confirm this sequence of events illustrated by the expression of ROS-, AKT-, and YAP-specific gene signatures upon APAP administration. In summary, our data illustrate the importance of nuclear processes in the regulation of Hippo pathway activity. YAP and TAZ exhibit different shuttling dynamics, which explains distinct cellular responses of both factors under physiological and tissue-damaging conditions.


Subject(s)
Chemical and Drug Induced Liver Injury , Proto-Oncogene Proteins c-akt , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation , Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Acetaminophen/toxicity , Reactive Oxygen Species/metabolism , Protein Serine-Threonine Kinases , YAP-Signaling Proteins , Nuclear Proteins/metabolism , Threonine/metabolism , Serine/metabolism
3.
Int J Biol Sci ; 18(9): 3800-3817, 2022.
Article in English | MEDLINE | ID: mdl-35813467

ABSTRACT

Background : Acetaminophen (APAP)-induced liver injury (AILI) is a common cause of drug-induced liver injury (DILI). The mechanism underlying protection in AILI or DILI remains to be elucidated, and the role of early growth response 1 (Egr1) in AILI and potential mechanisms remain to be known. Methods : The role of Egr1 was studied both in vivo and in vitro. Liver-specific Egr1-knockout (Egr1LKO) mice and those overexpressing Egr1 via tail vein injection of Egr1-expressing adenovirus (Ad-Egr1) were utilized with AILI. Chromatin immunoprecipitation-sequencing, RNA-sequencing, seahorse XF analysis, and targeted fatty acid analysis were performed. EGR1 levels were also studied in liver tissues and serum samples from AILI/DILI patients. Results: In this study, we have demonstrated that Egr1 was upregulated in AILI models in vivo and in vitro. liver-specific Egr1 knockout aggravated AILI; however, Ad-Egr1 treatment ameliorated this. Mechanistically, Egr1 deficiency inhibited, whereas overexpression promoted, mitochondrial respiratory function and fatty acid ß-oxidation (FAO) activity in AILI. Egr1 transcriptionally upregulated FAO-related genes in hepatocytes. Notably, the knockdown of acetyl-coenzyme A acyltransferase 2 (Acaa2), a key gene involved in FAO, diminished this protective effect of Egr1. Clinically, EGR1 was markedly increased in liver tissues from AILI patients. Interestingly, EGR1 levels of liver tissues and serum samples were also obviously higher in idiosyncratic DILI patients. Conclusions: Egr1 confers adaptive protection in AILI, mediated via the transcriptional upregulation of Acaa2, which improves mitochondrial FAO, and might be a potential biomarker and novel therapeutic target for AILI.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Acetaminophen/toxicity , Acetyl-CoA C-Acyltransferase , Acyltransferases/pharmacology , Animals , Chemical and Drug Induced Liver Injury/genetics , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/pharmacology , Fatty Acids , Liver , Mice , Mice, Inbred C57BL
4.
Liver Int ; 41(12): 3011-3023, 2021 12.
Article in English | MEDLINE | ID: mdl-34459091

ABSTRACT

BACKGROUND & AIM: The development of hepatocellular carcinoma (HCC) is associated with the formation of communication networks leading to the recruitment of disease-modifying macrophages. However, how oncogenes in tumour cells control paracrine communication is not fully understood. METHODS: Transgenic mice with liver-specific expression of the constitutively active yes-associated protein (YAPS127A ) or an orthotopic implantation model served as tumour models. FACS-sorted F4/80+ /CD11bdim /CD146- /retinoid- macrophages from healthy and tumour-bearing livers were used for transcriptomic profiling. Expression data of 242 human HCCs and a tissue microarray consisting of 91 HCCs and seven liver tissues were analyzed. RESULTS: Screening of primary tumour cells expressing YAPS127A identified CC chemokine ligand 2 (Ccl2) as a macrophage chemoattractant, whose expression was regulated in a YAP/TEA domain family member 4 (TEAD4)-dependent manner. Ccl2 expression was associated with a loss of Kupffer cells (KCs) and an increase in immature macrophages (Mɸimm ) in hepatocarcinogenesis. Recruited Mɸimm were characterized by a lack of functional polarization (M0 signature) and high expression of the Ccl2 receptors C-C motif chemokine receptor 2 (Ccr2), C-X3-C motif chemokine receptor 1 (Cx3cr1) and pro-angiogenic platelet-derived growth factors (Pdgfa/Pdgfb). Mɸimm formed cellular clusters in the perivascular space, which correlated with vascular morphometric changes indicative for angiogenesis. In human HCCs, the M0 signature served as an identifier for poor clinical outcome and CCL2 correlated with YAP expression and vascular network formation. CONCLUSIONS: In conclusion, YAP/TEAD4-regulated Ccl2 associates with perivascular recruitment of unpolarized Mɸimm and may contribute to a proangiogenic microenvironment in liver cancer.


Subject(s)
Carcinoma, Hepatocellular , Chemokine CCL2 , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins , Chemokine CCL2/metabolism , Humans , Kupffer Cells/metabolism , Ligands , Liver Neoplasms/pathology , Macrophages/metabolism , Mice , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Transcription Factors , Tumor Microenvironment , Vascular Remodeling , YAP-Signaling Proteins
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1293-1304, 2019 10.
Article in English | MEDLINE | ID: mdl-31170503

ABSTRACT

BACKGROUND: Bile acids (BAs) participate in lipid absorption and serve as metabolic regulatory factors in gut-liver communication. To date, there are no studies on the systemic patterns of BAs in the serum, liver, and gut in the same non-alcoholic fatty liver disease (NAFLD) model. METHODS: A targeted metabolomics approach and 16S rRNA sequencing were used to identify the profile of BAs and connection between BAs and microbiota. The role and mechanism of altered BAs on hepatic steatosis were investigated. FINDINGS: In the liver, the composition of taurocholic acid (TCA) was increased, but taurohyodeoxycholic acid (THDCA) and ursodeoxycholic acid (UDCA) were decreased. In the gut, the deconjugated form of TCA (cholic acid (CA)) was increased, while the deconjugated forms of THDCA (α-hyodeoxycholic acid (HDCA)) and ω-muricholic acid (ωMCA) were decreased. In the serum, the composition of TCA was increased, while both HDCA and THDCA were decreased. THDCA induced the gene expression of apolipoprotein, bile secretion-related proteins, and cytochrome P450 family but suppressed inflammatory response genes expression in steatotic hepatocytes by RNAseq analysis. THDCA ameliorated neutral lipid accumulation and improved insulin sensitivity in primary rat hepatocytes. The decreased HDCA level correlated with the level of Bacteroidetes, while the level of CA correlated with the levels of Firmicutes and Verrucomicrobia but correlated inversely with Bacteroidetes. CONCLUSION: BAs profiles in the serum, liver and caecal content were altered in a rat NAFLD model, which may affect hepatic lipid accumulation and correlate with gut dysbiosis.


Subject(s)
Bile Acids and Salts/metabolism , Cecum/metabolism , Fatty Liver/metabolism , Liver/metabolism , Animals , Bile Acids and Salts/blood , Cells, Cultured , Diet, High-Fat , Fatty Liver/blood , Gastrointestinal Microbiome , Male , Rats, Sprague-Dawley
6.
Cell Mol Life Sci ; 76(1): 129-145, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30151693

ABSTRACT

As an analgesic and antipyretic drug, acetaminophen (APAP) is commonly used and known to be safe at therapeutic doses. In many countries, the overuse of APAP provokes acute liver injury and even liver failure. APAP-induced liver injury (AILI) is the most used experimental model of drug-induced liver injury (DILI). Here, we have demonstrated elevated levels of growth arrest and DNA damage-inducible 45α (GADD45α) in the livers of patients with DILI/AILI, in APAP-injured mouse livers and in APAP-treated hepatocytes. GADD45α exhibited a protective effect against APAP-induced liver injury and alleviated the accumulation of small lipid droplets in vitro and in vivo. We found that GADD45α promoted the activation of AMP-activated protein kinase α and induced fatty acid beta-oxidation, tricarboxylic acid cycle (TCA) and glycogenolysis-related gene expression after APAP exposure. Liquid chromatography-mass spectrometry (LC-MS) analysis showed that GADD45α increased the levels of TCA cycle metabolites. Co-immunoprecipitation analysis showed that Ppp2cb, a catalytic subunit of protein phosphatase 2A, could interact directly with GADD45α. Our results indicate that hepatocyte GADD45α might represent a therapeutic target to prevent and rescue liver injury caused by APAP.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetaminophen/adverse effects , Antipyretics/adverse effects , Cell Cycle Proteins/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Liver/drug effects , Nuclear Proteins/metabolism , AMP-Activated Protein Kinases/analysis , Analgesics, Non-Narcotic/adverse effects , Animals , Cell Cycle Proteins/analysis , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Citric Acid Cycle/drug effects , Enzyme Activation/drug effects , Fatty Acids/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/analysis , Signal Transduction/drug effects
7.
Toxicol Lett ; 283: 69-76, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29174983

ABSTRACT

Acetaminophen (APAP) overdose is a leading cause of drug-induced acute liver failure in many countries. In the present study, we developed stable mouse models of acute drug-induced hepatic injury (DILI) and acute drug-induced hepatic failure (DILF) by sub-lethal and lethal APAP injection respectively. The differences in hepatic transcriptome profiling between these two models were compared by RNA sequencing, which were validated by qPCR, western-blot and ELISA. In results, serum IL-6, TNF-a and IL-10 levels are higher in DILF than in DILI. The upregulated genes in DILF compared with DILI were mostly enriched in the areas of "cellular development process", "cell division", "multicellular organism development," etc. The downregulated genes in DILF compared with DILI were mostly enriched in the areas of "cellular response to chemical stimulus", "cellular response to stress", "cell activation," etc. Sub-lethal doses of APAP increased Myc, Bag3 and Btc expression in mouse liver, but lethal doses of APAP did not, which suggested that these three genes might play important roles in adaptive protection reactions in DILI. The serum Btc level might be a potential biomarker of drug induced liver injury with good prognosis. Our data can help us better understand the mechanisms of hepatotoxicity that influence prognosis and seek novel prognostic indicators of DILI.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/genetics , Liver Failure, Acute/chemically induced , Liver Failure, Acute/genetics , Transcriptome/drug effects , Animals , Biomarkers , Gene Expression Profiling , Gene Expression Regulation/drug effects , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...