Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Semin Liver Dis ; 43(1): 1-12, 2023 02.
Article in English | MEDLINE | ID: mdl-36522162

ABSTRACT

Biliary epithelium (i.e., cholangiocytes) is a heterogeneous population of epithelial cells in the liver, which line small and large bile ducts and have individual responses and functions dependent on size and location in the biliary tract. We discuss the recent findings showing that the intrahepatic biliary tree is heterogeneous regarding (1) morphology and function, (2) hormone expression and signaling (3), response to injury, and (4) roles in liver regeneration. This review overviews the significant characteristics and differences of the small and large cholangiocytes. Briefly, it outlines the in vitro and in vivo models used in the heterogeneity evaluation. In conclusion, future studies addressing biliary heterogeneity's role in the pathogenesis of liver diseases characterized by ductular reaction may reveal novel therapeutic approaches.


Subject(s)
Biliary Tract , Liver Diseases , Humans , Bile Ducts, Intrahepatic/metabolism , Epithelium/metabolism , Epithelium/pathology , Epithelial Cells , Liver , Liver Diseases/metabolism
2.
Hepatol Commun ; 6(7): 1574-1588, 2022 07.
Article in English | MEDLINE | ID: mdl-35271760

ABSTRACT

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.


Subject(s)
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
3.
Cells ; 10(11)2021 11 09.
Article in English | MEDLINE | ID: mdl-34831316

ABSTRACT

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.


Subject(s)
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
4.
Hepatology ; 73(6): 2411-2428, 2021 06.
Article in English | MEDLINE | ID: mdl-32964473

ABSTRACT

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.


Subject(s)
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
5.
Semin Liver Dis ; 40(4): 403-410, 2020 11.
Article in English | MEDLINE | ID: mdl-32906164

ABSTRACT

Cholestatic liver disease affects millions of people worldwide and stems from a plethora of causes such as immune dysfunction, genetics, cancerous growths, and lifestyle choices. While not considered a classical lymphatic organ, the liver plays a vital role in the lymph system producing up to half of the body's lymph per day. The lymphatic system is critical to the health of an organism with its networks of vessels that provide drainage for lymphatic fluid and routes for surveilling immune cells. Cholestasis results in an increase of inflammatory cytokines, growth factors, and inflammatory infiltrate. Left unchecked, further disease progression will include collagen deposition which impedes both the hepatic and lymphatic ducts, eventually resulting in an increase in hepatic decompensation, increasing portal pressures, and accumulation of fluid within the abdominal cavity (ascites). Despite the documented interplay between these vital systems, little is known about the effect of liver disease on the lymph system and its biological response. This review looks at the current cholestatic literature from the perspective of the lymphatic system and summarizes what is known about the role of the lymph system in liver pathogenesis during hepatic injury and remodeling, immune-modulating events, or variations in interstitial pressures.


Subject(s)
Cholestasis , Lymphatic Vessels , Humans , Liver , Lymph , Lymphatic System
6.
Hepatology ; 71(3): 990-1008, 2020 03.
Article in English | MEDLINE | ID: mdl-31344280

ABSTRACT

BACKGROUND AND AIMS: Serotonin (5HT) is a neuroendocrine hormone synthetized in the central nervous system (CNS) as well as enterochromaffin cells of the gastrointestinal tract. Tryptophan hydroxylase (TPH1) and monoamine oxidase (MAO-A) are the key enzymes for the synthesis and catabolism of 5HT, respectively. Previous studies demonstrated that 5-hydroxytryptamine receptor (5HTR)1A/1B receptor agonists inhibit biliary hyperplasia in bile-duct ligated (BDL) rats, whereas 5HTR2B receptor antagonists attenuate liver fibrosis (LF) in mice. Our aim was to evaluate the role of 5HTR2A/2B/2C agonists/antagonists in cholestatic models. APPROACH AND RESULTS: While in vivo studies were performed in BDL rats and the multidrug resistance gene 2 knockout (Mdr2-/- ) mouse model of PSC, in vitro studies were performed in cell lines of cholangiocytes and hepatic stellate cells (HSCs). 5HTR2A/2B/2C and MAO-A/TPH1 are expressed in cholangiocytes and HSCs from BDL rats and Mdr2-/- - mice. Ductular reaction, LF, as well as the mRNA expression of proinflammatory genes increased in normal, BDL rats, and Mdr2-/- - mice following treatment 5HTR2A/2B/2C agonists, but decreased when BDL rats and Mdr2-/- mice were treated with 5HTR2A/2B/2C antagonists compared to BDL rats and Mdr2-/- mice, respectively. 5HT levels increase in Mdr2-/- mice and in PSC human patients compared to their controls and decrease in serum of Mdr2-/- mice treated with 5HTR2A/2B/2C antagonists compared to untreated Mdr2-/- mice. In vitro, cell lines of murine cholangiocytes and human HSCs express 5HTR2A/2B/2C and MAO-A/TPH1; treatment of these cell lines with 5HTR2A/2B/2C antagonists or TPH1 inhibitor decreased 5HT levels as well as expression of fibrosis and inflammation genes compared to controls. CONCLUSIONS: Modulation of the TPH1/MAO-A/5HT/5HTR2A/2B/2C axis may represent a therapeutic approach for management of cholangiopathies, including PSC.


Subject(s)
Bile Ducts/pathology , Cholestasis/pathology , Liver Cirrhosis/etiology , Monoamine Oxidase/physiology , Receptors, Serotonin/physiology , Serotonin/physiology , Tryptophan Hydroxylase/physiology , ATP Binding Cassette Transporter, Subfamily B/physiology , Animals , Cell Proliferation , Cholangitis, Sclerosing/etiology , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/physiology , Receptor, Serotonin, 5-HT2B/physiology , Receptor, Serotonin, 5-HT2C/physiology , Serotonin/blood , ATP-Binding Cassette Sub-Family B Member 4
7.
Cancers (Basel) ; 11(11)2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31731549

ABSTRACT

Liver cancer is a devastating cancer that ranges from relatively rare (around 2% of all cancers in the United States) to commonplace (up to 50% of cancers in underdeveloped countries). Depending upon the stage of pathogenesis, prognosis, or functional liver tissue present, transplantation or partial hepatectomy may be the only available treatment option. However, due to the rise in metabolic syndrome and the increasing demand for livers, patients often wait months or years for available organs. Due to this shortage, doctors must have other treatment options available. One promising area of cancer research lies in understanding the role of regulatory non-coding RNAs (ncRNAs) as oncogenic drivers and potential targets for prospective therapies. While the role of these ncRNAs was not initially clear, many of them have since been recognized to function as important players in the regulation of gene expression, epigenetic modification, and signal transduction in both normal and cancer cell cycles. Dysregulation of these different ncRNA subtypes has been implicated in the pathogenesis and progression of many major cancers including hepatocellular carcinoma. This review summarizes current findings on the roles noncoding RNAs play in the progression of liver cancer and the various animal models used in current research to elucidate those data.

8.
EBioMedicine ; 48: 130-142, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31522982

ABSTRACT

BACKGROUND: Cholangiocytes are the target cells of cholangiopathies including primary sclerosing cholangitis (PSC). Vimentin is an intermediate filament protein that has been found in various types of mesenchymal cells. The aim of this study is to evaluate the role of vimentin in the progression of biliary damage/liver fibrosis and whether there is a mesenchymal phenotype of cholangiocytes in the Mdr2-/- model of PSC. METHODS: In vivo studies were performed in 12 wk. Mdr2-/- male mice with or without vimentin Vivo-Morpholino treatment and their corresponding control groups. Liver specimens from human PSC patients, human intrahepatic biliary epithelial cells (HIBEpiC) and human hepatic stellate cell lines (HHSteCs) were used to measure changes in epithelial-to-mesenchymal transition (EMT). FINDINGS: There was increased mesenchymal phenotype of cholangiocytes in Mdr2-/- mice, which was reduced by treatment of vimentin Vivo-Morpholino. Concomitant with reduced vimentin expression, there was decreased liver damage, ductular reaction, biliary senescence, liver fibrosis and TGF-ß1 secretion in Mdr2-/- mice treated with vimentin Vivo-Morpholino. Human PSC patients and derived cell lines had increased expression of vimentin and other mesenchymal markers compared to healthy controls and HIBEpiC, respectively. In vitro silencing of vimentin in HIBEpiC suppressed TGF-ß1-induced EMT and fibrotic reaction. HHSteCs had decreased fibrotic reaction and increased cellular senescence after stimulation with cholangiocyte supernatant with reduced vimentin levels. INTERPRETATION: Our study demonstrated that knockdown of vimentin reduces mesenchymal phenotype of cholangiocytes, which leads to decreased biliary senescence and liver fibrosis. Inhibition of vimentin may be a key therapeutic target in the treatment of cholangiopathies including PSC. FUND: National Institutes of Health (NIH) awards, VA Merit awards.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Cholangitis, Sclerosing/genetics , Epithelial Cells/metabolism , Gene Knockdown Techniques , Vimentin/genetics , Animals , Biomarkers , Cell Line , Cholangitis, Sclerosing/diagnosis , Cholangitis, Sclerosing/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Fibrosis , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Phenotype , ATP-Binding Cassette Sub-Family B Member 4
9.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1525-1539, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30890428

ABSTRACT

Melatonin, a neuroendocrine hormone synthesized by the pineal gland and cholangiocytes, decreases biliary hyperplasia and liver fibrosis during cholestasis-induced biliary injury via melatonin-dependent autocrine signaling through increased biliary arylalkylamine N-acetyltransferase (AANAT) expression and melatonin secretion, downregulation of miR-200b and specific circadian clock genes. Melatonin synthesis is decreased by pinealectomy (PINX) or chronic exposure to light. We evaluated the effect of PINX or prolonged light exposure on melatonin-dependent modulation of biliary damage/ductular reaction/liver fibrosis. Studies were performed in male rats with/without BDL for 1 week with 12:12 h dark/light cycles, continuous light or after 1 week of PINX. The expression of AANAT and melatonin levels in serum and cholangiocyte supernatant were increased in BDL rats, while decreased in BDL rats following PINX or continuous light exposure. BDL-induced increase in serum chemistry, ductular reaction, liver fibrosis, inflammation, angiogenesis and ROS generation were significantly enhanced by PINX or light exposure. Concomitant with enhanced liver fibrosis, we observed increased biliary senescence and enhanced clock genes and miR-200b expression in total liver and cholangiocytes. In vitro, the expression of AANAT, clock genes and miR-200b was increased in PSC human cholangiocyte cell lines (hPSCL). The proliferation and activation of HHStecs (human hepatic stellate cell lines) were increased after stimulating with BDL cholangiocyte supernatant and further enhanced when stimulated with BDL rats following PINX or continuous light exposure cholangiocyte supernatant via intracellular ROS generation. Conclusion: Melatonin plays an important role in the protection of liver against cholestasis-induced damage and ductular reaction.


Subject(s)
Arylalkylamine N-Acetyltransferase/genetics , CLOCK Proteins/genetics , Cholestasis/genetics , Liver Cirrhosis/genetics , Melatonin/biosynthesis , MicroRNAs/genetics , Animals , Arylalkylamine N-Acetyltransferase/metabolism , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Bile Ducts, Intrahepatic/surgery , CLOCK Proteins/metabolism , Cell Line , Cell Proliferation/radiation effects , Cholestasis/metabolism , Cholestasis/pathology , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/radiation effects , Humans , Kupffer Cells/metabolism , Kupffer Cells/radiation effects , Light , Liver/metabolism , Liver/pathology , Liver/surgery , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , MicroRNAs/metabolism , Pineal Gland/metabolism , Pineal Gland/radiation effects , Pineal Gland/surgery , Pinealectomy/methods , Primary Cell Culture , Rats , Rats, Inbred F344 , Signal Transduction
10.
F1000Res ; 72018.
Article in English | MEDLINE | ID: mdl-30109019

ABSTRACT

Cholestatic liver disease encompasses a detrimental group of diseases that are non-discriminatory in nature. These diseases occur over every age range from infancy (biliary atresia) to geriatrics (hepatitis). They also cover both genders in the form of primary sclerosing cholangitis in men and primary biliary cholangitis in women. Oftentimes, owing to the disease progression and extensive scarring, the treatment of last resort becomes a liver transplant. In this review, we will briefly discuss and explore new avenues of understanding in the progression of cholestatic liver disease and possible therapeutic targets for intervention. The greater our understanding into the idiopathic nature of cholestatic liver disease, the better our chances of discovering treatment options to halt or reverse the progression, reducing or eliminating the need for expensive and risky transplants.


Subject(s)
Bile Ducts/pathology , Cholestasis/complications , Cholestasis/pathology , Fibrosis , Humans , Liver Diseases/complications , Signal Transduction
11.
Gene Expr ; 18(3): 197-207, 2018 08 22.
Article in English | MEDLINE | ID: mdl-29580318

ABSTRACT

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.


Subject(s)
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
12.
Am J Pathol ; 187(5): 1093-1105, 2017 May.
Article in English | MEDLINE | ID: mdl-28315314

ABSTRACT

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.


Subject(s)
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
13.
Cancer Lett ; 386: 179-188, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27894959

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...