Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Hepatol Commun ; 6(7): 1574-1588, 2022 07.
Article En | MEDLINE | ID: mdl-35271760

Fibroblast growth factor 1 (FGF1) belongs to a family of growth factors involved in cellular growth and division. MicroRNA 16 (miR-16) is a regulator of gene expression, which is dysregulated during liver injury and insult. However, the role of FGF1 in the progression of biliary proliferation, senescence, fibrosis, inflammation, angiogenesis, and its potential interaction with miR-16, are unknown. In vivo studies were performed in male bile duct-ligated (BDL, 12-week-old) mice, multidrug resistance 2 knockout (Mdr2-/-) mice (10-week-old), and their corresponding controls, treated with recombinant human FGF1 (rhFGF1), fibroblast growth factor receptor (FGFR) antagonist (AZD4547), or anti-FGF1 monoclonal antibody (mAb). In vitro, the human cholangiocyte cell line (H69) and human hepatic stellate cells (HSCs) were used to determine the expression of proliferation, fibrosis, angiogenesis, and inflammatory genes following rhFGF1 treatment. PSC patient and control livers were used to evaluate FGF1 and miR-16 expression. Intrahepatic bile duct mass (IBDM), along with hepatic fibrosis and inflammation, increased in BDL mice treated with rhFGF1, with a corresponding decrease in miR-16, while treatment with AZD4547 or anti-FGF1 mAb decreased hepatic fibrosis, IBDM, and inflammation in BDL and Mdr2-/- mice. In vitro, H69 and HSCs treated with rhFGF1 had increased expression of proliferation, fibrosis, and inflammatory markers. PSC samples also showed increased FGF1 and FGFRs with corresponding decreases in miR-16 compared with healthy controls. Conclusion: Our study demonstrates that suppression of FGF1 and miR-16 signaling decreases the presence of hepatic fibrosis, biliary proliferation, inflammation, senescence, and angiogenesis. Targeting the FGF1 and miR-16 axis may provide therapeutic options in treating cholangiopathies such as PSC.


Cholangitis, Sclerosing , MicroRNAs , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Cholangitis, Sclerosing/drug therapy , Disease Models, Animal , Fibroblast Growth Factor 1/genetics , Fibrosis , Humans , Inflammation , Liver Cirrhosis/drug therapy , Male , Mice , Mice, Knockout , MicroRNAs/genetics , ATP-Binding Cassette Sub-Family B Member 4
2.
Cells ; 10(11)2021 11 09.
Article En | MEDLINE | ID: mdl-34831316

Cholangiocarcinoma (CCA), or cancer of bile duct epithelial cells, is a very aggressive malignancy characterized by early lymphangiogenesis in the tumor microenvironment (TME) and lymph node (LN) metastasis which correlate with adverse patient outcome. However, the specific roles of lymphatic endothelial cells (LECs) that promote LN metastasis remains unexplored. Here we aimed to identify the dynamic molecular crosstalk between LECs and CCA cells that activate tumor-promoting pathways and enhances lymphangiogenic mechanisms. Our studies show that inflamed LECs produced high levels of chemokine CXCL5 that signals through its receptor CXCR2 on CCA cells. The CXCR2-CXCL5 signaling axis in turn activates EMT (epithelial-mesenchymal transition) inducing MMP (matrix metalloproteinase) genes such as GLI, PTCHD, and MMP2 in CCA cells that promote CCA migration and invasion. Further, rate of mitochondrial respiration and glycolysis of CCA cells was significantly upregulated by inflamed LECs and CXCL5 activation, indicating metabolic reprogramming. CXCL5 also induced lactate production, glucose uptake, and mitoROS. CXCL5 also induced LEC tube formation and increased metabolic gene expression in LECs. In vivo studies using CCA orthotopic models confirmed several of these mechanisms. Our data points to a key finding that LECs upregulate critical tumor-promoting pathways in CCA via CXCR2-CXCL5 axis, which further augments CCA metastasis.


Bile Duct Neoplasms/metabolism , Chemokine CXCL5/metabolism , Cholangiocarcinoma/metabolism , Lymphatic System/pathology , Receptors, Interleukin-8B/metabolism , Signal Transduction , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Endothelial Cells/pathology , Energy Metabolism , Epithelial-Mesenchymal Transition/genetics , Focal Adhesions/metabolism , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Lactic Acid/biosynthesis , Lymph Nodes/pathology , Lymphangiogenesis/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Models, Biological , Reactive Oxygen Species/metabolism , Up-Regulation
3.
Hepatology ; 73(6): 2411-2428, 2021 06.
Article En | MEDLINE | ID: mdl-32964473

BACKGROUND AND AIMS: Apelin (APLN) is the endogenous ligand of its G protein-coupled receptor, apelin receptor (APJ). APLN serum levels are increased in human liver diseases. We evaluated whether the APLN-APJ axis regulates ductular reaction and liver fibrosis during cholestasis. APPROACH AND RESULTS: We measured the expression of APLN and APJ and serum APLN levels in human primary sclerosing cholangitis (PSC) samples. Following bile duct ligation (BDL) or sham surgery, male wild-type (WT) mice were treated with ML221 (APJ antagonist) or saline for 1 week. WT and APLN-/- mice underwent BDL or sham surgery for 1 week. Multidrug resistance gene 2 knockout (Mdr2-/- ) mice were treated with ML221 for 1 week. APLN levels were measured in serum and cholangiocyte supernatants, and cholangiocyte proliferation/senescence and liver inflammation, fibrosis, and angiogenesis were measured in liver tissues. The regulatory mechanisms of APLN-APJ in (1) biliary damage and liver fibrosis were examined in human intrahepatic biliary epithelial cells (HIBEpiCs) treated with APLN and (2) hepatic stellate cell (HSC) activation in APLN-treated human HSC lines (HHSteCs). APLN serum levels and biliary expression of APLN and APJ increased in PSC samples. APLN levels were higher in serum and cholangiocyte supernatants from BDL and Mdr2-/- mice. ML221 treatment or APLN-/- reduced BDL-induced and Mdr2-/- -induced cholangiocyte proliferation/senescence, liver inflammation, fibrosis, and angiogenesis. In vitro, APLN induced HIBEpiC proliferation, increased nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) expression, reactive oxygen species (ROS) generation, and extracellular signal-regulated kinase (ERK) phosphorylation. Pretreatment of HIBEpiCs with ML221, diphenyleneiodonium chloride (Nox4 inhibitor), N-acetyl-cysteine (NAC, ROS inhibitor), or PD98059 (ERK inhibitor) reduced APLN-induced cholangiocyte proliferation. Activation of HHSteCs was induced by APLN but reduced by NAC. CONCLUSIONS: The APLN-APJ axis induces cholangiocyte proliferation through Nox4/ROS/ERK-dependent signaling and HSC activation through intracellular ROS. Modulation of the APLN-APJ axis may be important for managing cholangiopathies.


Apelin Receptors/metabolism , Apelin/metabolism , Cholangitis, Sclerosing/metabolism , Cholestasis/metabolism , Liver Cirrhosis/metabolism , Nitrobenzoates/pharmacology , Pyrans/pharmacology , Acetylcysteine/pharmacology , Animals , Apelin Receptors/antagonists & inhibitors , Cell Proliferation , Cholangitis, Sclerosing/pathology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Free Radical Scavengers/pharmacology , Hepatic Stellate Cells/metabolism , Humans , Mice , NADPH Oxidase 4/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
4.
Am J Pathol ; 190(4): 900-915, 2020 04.
Article En | MEDLINE | ID: mdl-32035061

Tumor metastasis to the draining lymph nodes is critical in patient prognosis and is tightly regulated by molecular interactions mediated by lymphatic endothelial cells (LECs). The underlying mechanisms remain undefined in the head and neck squamous cell carcinomas (HNSCCs). Using HNSCC cells and LECs we determined the mechanisms mediating tumor-lymphatic cross talk. The effects of a pentacyclic triterpenoid, methyl 2-trifluoromethyl-3,11-dioxoolean-1,12-dien-30-oate (CF3DODA-Me), a potent anticancer agent, were studied on cancer-lymphatic interactions. In response to inflammation, LECs induced the chemokine (C-X-C motif) ligand 9/10/11 chemokines with a concomitant increase in the chemokine (C-X-C motif) receptor 3 (CXCR3) in tumor cells. CF3DODA-Me showed antiproliferative effects on tumor cells, altered cellular bioenergetics, suppressed matrix metalloproteinases and chemokine receptors, and the induction of CXCL11-CXCR3 axis and phosphatidylinositol 3-kinase/AKT pathways. Tumor cell migration to LECs was inhibited by blocking CXCL11 whereas recombinant CXCL11 significantly induced tumor migration, epithelial-to-mesenchymal transition, and matrix remodeling. Immunohistochemical analysis of HNSCC tumor arrays showed enhanced expression of CXCR3 and increased lymphatic vessel infiltration. Furthermore, The Cancer Genome Atlas RNA-sequencing data from HNSCC patients also showed a positive correlation between CXCR3 expression and lymphovascular invasion. Collectively, our data suggest a novel mechanism for cross talk between the LECs and HNSCC tumors through the CXCR3-CXCL11 axis and elucidate the role of the triterpenoid CF3DODA-Me in abrogating several of these tumor-promoting pathways.


Chemokine CXCL11/metabolism , Endothelial Cells/pathology , Head and Neck Neoplasms/pathology , Inflammation/pathology , Receptors, CXCR3/metabolism , Squamous Cell Carcinoma of Head and Neck/secondary , Antineoplastic Agents/pharmacology , Chemokine CXCL11/genetics , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Lymphatic Metastasis , Prognosis , Receptors, CXCR3/genetics , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Triterpenes/pharmacology , Tumor Cells, Cultured
5.
Gene Expr ; 18(3): 197-207, 2018 08 22.
Article En | MEDLINE | ID: mdl-29580318

α7-nAChR is a nicotinic acetylcholine receptor [specifically expressed on hepatic stellate cells (HSCs), Kupffer cells, and cholangiocytes] that regulates inflammation and apoptosis in the liver. Thus, targeting α7-nAChR may be therapeutic in biliary diseases. Bile duct ligation (BDL) was performed on wild-type (WT) and α7-nAChR-/- mice. We first evaluated the expression of α7-nAChR by immunohistochemistry (IHC) in liver sections. IHC was also performed to assess intrahepatic bile duct mass (IBDM), and Sirius Red staining was performed to quantify the amount of collagen deposition. Immunofluorescence was performed to assess colocalization of α7-nAChR with bile ducts (costained with CK-19) and HSCs (costained with desmin). The mRNA expression of α7-nAChR, Ki-67/PCNA (proliferation), fibrosis genes (TGF-ß1, fibronectin-1, Col1α1, and α-SMA), and inflammatory markers (IL-6, IL-1ß, and TNF-α) was measured by real-time PCR. Biliary TGF-ß1 and hepatic CD68 (Kupffer cell marker) expression was assessed using IHC. α7-nAChR immunoreactivity was observed in both bile ducts and HSCs and increased following BDL. α7-nAChR-/- BDL mice exhibited decreased (i) bile duct mass, liver fibrosis, and inflammation, and (ii) immunoreactivity of TGF-ß1 as well as expression of fibrosis genes compared to WT BDL mice. α7-nAChR activation triggers biliary proliferation and liver fibrosis and may be a therapeutic target in managing extrahepatic biliary obstruction.


Cholestasis, Extrahepatic/genetics , Liver Cirrhosis/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Animals , Bile Ducts/metabolism , Bile Ducts/pathology , Cell Line, Tumor , Cholestasis, Extrahepatic/complications , Cholestasis, Extrahepatic/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Hyperplasia , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
6.
Am J Pathol ; 187(5): 1093-1105, 2017 May.
Article En | MEDLINE | ID: mdl-28315314

Nicotine, the main addictive substance in tobacco, is known to play a role in the development and/or progression of a number of malignant tumors. However, nicotine's involvement in the pathogenesis of cholangiocarcinoma is controversial. Therefore, we studied the effects of nicotine on the growth of cholangiocarcinoma cells in vitro and the progression of cholangiocarcinoma in a mouse xenograft model. The predominant subunit responsible for nicotine-mediated proliferation in normal and cancer cells, the α7 nicotinic acetylcholine receptor (α7-nAChR), was more highly expressed in human cholangiocarcinoma cell lines compared with normal human cholangiocytes. Nicotine also stimulated the proliferation of cholangiocarcinoma cell lines and promoted α7-nAChR-dependent activation of proliferation and phosphorylation of extracellular-regulated kinase in Mz-ChA-1 cells. In addition, nicotine and PNU282987 (α7-nAChR agonist) accelerated the growth of the cholangiocarcinoma tumors in our xenograft mouse model and increased fibrosis, proliferation of the tumor cells, and phosphorylation of extracellular-regulated kinase activation. Finally, α7-nAChR was expressed at significantly higher levels in human cholangiocarcinoma compared with normal human control liver samples. Taken together, results of this study suggest that nicotine acts through α7-nAChR and plays a novel role in the pathogenesis of cholangiocarcinoma. Furthermore, nicotine may act as a mitogen in cholestatic liver disease processes, thereby facilitating malignant transformation.


Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Aged , Animals , Bile Duct Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibrosis/metabolism , Heterografts , Humans , Keratin-19/metabolism , MAP Kinase Signaling System/physiology , Male , Mice , Middle Aged , Neoplasm Transplantation , S100 Calcium-Binding Protein A4/metabolism , alpha7 Nicotinic Acetylcholine Receptor/physiology
7.
Cancer Lett ; 386: 179-188, 2017 02 01.
Article En | MEDLINE | ID: mdl-27894959

PURPOSE: Cholangiocarcinoma (CCA) is a malignancy of the biliary epithelium that is associated with low five-year survival. The apelin receptor (APLNR), which is activated by the apelin peptide, has not been studied in CCA. The purpose of this study is to determine if inhibition of the apelin/APLNR axis can inhibit CCA growth. METHODS: Immunohistochemistry, rtPCR, immunofluorescence, flow cytometry, and ELISA was used to measure APLNR expression in human CCA cells and tissues. Mz-ChA-1 cells were treated with increasing concentrations of apelin and ML221, an APLNR antagonist. Expression of proliferative and angiogenic genes were measured via rtPCR. In vivo, Mz-ChA-1 cells were injected into the flanks of nu/nu mice, which were treated with ML221 (150 µg/kg) via tail vein injection. RESULTS: Expression of the apelin/APLNR axis was increased in CCA. In vitro, CCA proliferation and angiogenesis was inhibited by ML221 treatment. ML221 treatment significantly decreased tumor growth in nu/nu mice. CONCLUSION: The apelin/APLNR axis regulates CCA proliferation and angiogenesis. Inhibition of the apelin/APLNR axis decreases tumor growth in our xenograft model. Targeting APLNR signaling has the potential to serve as a novel, tumor directed therapy for CCA.


Angiogenesis Inhibitors/pharmacology , Bile Duct Neoplasms/drug therapy , Cell Proliferation/drug effects , Cholangiocarcinoma/drug therapy , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic , Nitrobenzoates/pharmacology , Pyrans/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Adult , Aged, 80 and over , Animals , Apelin , Apelin Receptors , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
...