Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Am J Physiol Cell Physiol ; 317(4): C788-C799, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31365294

ABSTRACT

In mouse models of biliary tract diseases, macrophages are recruited to the periductal milieu and promote injury and cholestasis. Although cell necrosis with release of biomolecules termed damage-associated molecular patterns (DAMPs) promotes recruitment and activation of macrophages, necrosis was not observed in these studies. Because extracellular vesicles (EVs) are important in cell-to-cell communication, we postulated that activated cholangiocytes may release EVs containing DAMPs as cargo. Both the human (NHC) and mouse cholangiocyte (603B) cell lines display constitutive activation with mRNA expression of chemokines. Proteomic analysis revealed that EVs from both cell lines contained the DAMP S100A11, a ligand for the receptor for advanced glycation end products (RAGE). Bone marrow-derived macrophages (BMDM) incubated with EVs derived from the mouse 603B cell line increased mRNA expression of proinflammatory cytokines. Genetic or pharmacologic inhibition of RAGE reduced BMDM expression of proinflammatory cytokines treated with EVs. RAGE signaling resulted in activation of the canonical NF-κB pathway, and consistently, proinflammatory cytokine expression was blunted by the IKKα/ß inhibitor TPCA-1 in BMDM incubated with EVs. We also demonstrated that primary mouse cholangiocyte-derived organoids express chemokines indicating cholangiocyte activation, release EVs containing S100A11, and stimulate proinflammatory cytokine expression in BMDM by a RAGE-dependent pathway. In conclusion, these observations identify a non-cell death mechanism for cellular release of DAMPs by activated cholangiocytes, namely by releasing DAMPs as EV cargo. These data also suggest RAGE inhibitors may be salutary in macrophage-associated inflammatory diseases of the bile ducts.


Subject(s)
Alarmins/metabolism , Extracellular Vesicles/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Animals , Cell Communication/physiology , Cytokines/metabolism , Epithelial Cells/metabolism , Female , Humans , Male , Mice, Inbred C57BL , Proteomics/methods , S100 Proteins/metabolism
2.
J Cell Biochem ; 119(1): 824-836, 2018 01.
Article in English | MEDLINE | ID: mdl-28661054

ABSTRACT

The Hippo pathway effector YAP is implicated in the pathogenesis of cholangiocarcinoma (CCA). The Hippo pathway relies on signaling cross talk for its regulation. Given the importance of platelet derived growth factor receptor (PDGFR) signaling in CCA biology, our aim was to examine potential YAP regulation by PDGFR. We employed human and mouse CCA specimens and cell lines for these studies. Initially, we confirmed upregulation of PDGFRß and PDGFR ligands in human and mouse CCA specimens and cell lines. YAP, a transcriptional co-activator, was localized to the nucleus in human CCA specimens and a cell line, as well as patient derived xenografts (PDX). PDGFR pharmacologic inhibition led to a redistribution of YAP from the nucleus to cytosol and downregulation of YAP target genes in a human CCA cell line. siRNA silencing of PDGFR-ß similarly downregulated YAP target genes. YAP activation (nuclear localization and target gene expression) was regulated by Src family kinases (SFKs) downstream of PDGFR. SFK activity resulted in phosphorylation of YAP on tyrosine357 (YAPY357 ). The importance of YAPY357 phosphorylation in regulating YAP activation was confirmed utilizing the SB-1 cell line, a mouse cell line expressing YAP S127A precluding canonical serine phosphorylation. PDGFR inhibition decreased cellular abundance of the survival protein Mcl-1, a known YAP target gene, and accordingly increased cell death in CCA cells in vitro and in vivo. These preclinical data demonstrate that a PDGFR-SFK cascade regulates YAP activation via tyrosine phosphorylation in CCA. Inhibiting this cascade may provide a viable therapeutic strategy for this human malignancy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Phosphoproteins/metabolism , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transcription, Genetic , Animals , Bile Duct Neoplasms/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cholangiocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Phosphorylation , Signal Transduction , Transcription Factors , Up-Regulation , YAP-Signaling Proteins
3.
J Hepatol ; 68(6): 1228-1238, 2018 06.
Article in English | MEDLINE | ID: mdl-29408314

ABSTRACT

BACKGROUND & AIMS: Myeloid cell leukemia 1 (MCL1), a prosurvival member of the BCL2 protein family, has a pivotal role in human cholangiocarcinoma (CCA) cell survival. We previously reported that fibroblast growth factor receptor (FGFR) signalling mediates MCL1-dependent survival of CCA cells in vitro and in vivo. However, the mode and mechanisms of cell death in this model were not delineated. METHODS: Human CCA cell lines were treated with the pan-FGFR inhibitor LY2874455 and the mode of cell death examined by several complementary assays. Mitochondrial oxidative metabolism was examined using a XF24 extracellular flux analyser. The efficiency of FGFR inhibition in patient-derived xenografts (PDX) was also assessed. RESULTS: CCA cells expressed two species of MCL1, a full-length form localised to the outer mitochondrial membrane, and an N terminus-truncated species compartmentalised within the mitochondrial matrix. The pan-FGFR inhibitor LY2874455 induced non-apoptotic cell death in the CCA cell lines associated with cellular depletion of both MCL1 species. The cell death was accompanied by failure of mitochondrial oxidative metabolism and was most consistent with necrosis. Enforced expression of N terminus-truncated MCL1 targeted to the mitochondrial matrix, but not full-length MCL1 targeted to the outer mitochondrial membrane, rescued cell death and mitochondrial function. LY2874455 treatment of PDX-bearing mice was associated with tumour cell loss of MCL1 and cell necrosis. CONCLUSIONS: FGFR inhibition induces loss of matrix MCL1, resulting in cell necrosis. These observations support a heretofore unidentified, alternative MCL1 survival function, namely prevention of cell necrosis, and have implications for treatment of human CCA. LAY SUMMARY: Herein, we report that therapeutic inhibition of a cell receptor expressed by bile duct cancer cells resulted in the loss of a critical survival protein termed MCL1. Cellular depletion of MCL1 resulted in the death of the cancer cells by a process characterised by cell rupture. Cell death by this process can stimulate the immune system and has implications for combination therapy using receptor inhibition with immunotherapy.


Subject(s)
Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Indazoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Bile Duct Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cholangiocarcinoma/pathology , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Necrosis , Oxidation-Reduction , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
4.
J Hepatol ; 69(3): 676-686, 2018 09.
Article in English | MEDLINE | ID: mdl-29802947

ABSTRACT

BACKGROUND & AIMS: Macrophages contribute to liver disease, but their role in cholestatic liver injury, including primary sclerosing cholangitis (PSC), is unclear. We tested the hypothesis that macrophages contribute to the pathogenesis of, and are therapeutic targets for, PSC. METHODS: Immune cell profile, hepatic macrophage number, localization and polarization, fibrosis, and serum markers of liver injury and cholestasis were measured in an acute (intrabiliary injection of the inhibitor of apoptosis antagonist BV6) and chronic (Mdr2-/- mice) mouse model of sclerosing cholangitis (SC). Selected observations were confirmed in liver specimens from patients with PSC. Because of the known role of the CCR2/CCL2 axis in monocyte/macrophage chemotaxis, therapeutic effects of the CCR2/5 antagonist cenicriviroc (CVC), or genetic deletion of CCR2 (Ccr2-/- mice) were determined in BV6-injected mice. RESULTS: We found increased peribiliary pro-inflammatory (M1-like) and alternatively-activated (M2-like) monocyte-derived macrophages in PSC compared to normal livers. In both SC models, genetic profiling of liver immune cells identified a predominance of monocytes/macrophages; immunohistochemistry confirmed peribiliary monocyte-derived macrophage recruitment (M1>M2-polarized), which paralleled injury onset and was reversed upon resolution in acute SC mice. PSC, senescent and BV6-treated human cholangiocytes released monocyte chemoattractants (CCL2, IL-8) and macrophage-activating factors in vitro. Pharmacological inhibition of monocyte recruitment by CVC treatment or CCR2 genetic deletion attenuated macrophage accumulation, liver injury and fibrosis in acute SC. CONCLUSIONS: Peribiliary recruited macrophages are a feature of both PSC and acute and chronic murine SC models. Pharmacologic and genetic inhibition of peribiliary macrophage recruitment decreases liver injury and fibrosis in mouse SC. These observations suggest monocyte-derived macrophages contribute to the development of SC in mice and in PSC pathogenesis, and support their potential as a therapeutic target. LAY SUMMARY: Primary sclerosing cholangitis (PSC) is an inflammatory liver disease which often progresses to liver failure. The cause of the disease is unclear and therapeutic options are limited. Therefore, we explored the role of white blood cells termed macrophages in PSC given their frequent contribution to other human inflammatory diseases. Our results implicate macrophages in PSC and PSC-like diseases in mice. More importantly, we found that pharmacologic inhibition of macrophage recruitment to the liver reduces PSC-like liver injury in the mouse. These exciting observations highlight potential new strategies to treat PSC.


Subject(s)
Chemokine CCL2/metabolism , Cholangitis, Sclerosing , Imidazoles/pharmacology , Liver Cirrhosis , Macrophages , Receptors, CCR2/metabolism , Receptors, CCR5/metabolism , Animals , CCR5 Receptor Antagonists/pharmacology , Chemotaxis/drug effects , Chemotaxis/immunology , Cholangitis, Sclerosing/drug therapy , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/pathology , Disease Models, Animal , Liver/immunology , Liver/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Sulfoxides , Treatment Outcome
5.
J Biol Chem ; 291(15): 8031-47, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26826125

ABSTRACT

Herein, we have identified cross-talk between the Hippo and fibroblast growth factor receptor (FGFR) oncogenic signaling pathways in cholangiocarcinoma (CCA). Yes-associated protein (YAP) nuclear localization and up-regulation of canonical target genes was observed in CCA cell lines and a patient-derived xenograft (PDX). Expression of FGFR1, -2, and -4 was identified in human CCA cell lines, driven, in part, by YAP coactivation of TBX5. In turn, FGFR signaling in a cell line with minimal basal YAP expression induced its cellular protein expression and nuclear localization. Treatment of YAP-positive CCA cell lines with BGJ398, a pan-FGFR inhibitor, resulted in a decrease in YAP activation. FGFR activation of YAP appears to be driven largely by FGF5 activation of FGFR2, as siRNA silencing of this ligand or receptor, respectively, inhibited YAP nuclear localization. BGJ398 treatment of YAP-expressing cells induced cell death due to Mcl-1 depletion. In a YAP-associated mouse model of CCA, expression of FGFR 1, 2, and 4 was also significantly increased. Accordingly, BGJ398 treatment was tumor-suppressive in this model and in a YAP-positive PDX model. These preclinical data suggest not only that the YAP and Hippo signaling pathways culminate in an Mcl-1-regulated tumor survival pathway but also that nuclear YAP expression may be a biomarker to employ in FGFR-directed therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bile Duct Neoplasms/metabolism , Bile Ducts/pathology , Cholangiocarcinoma/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts/metabolism , Cell Line, Tumor , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Mice , Mice, SCID , Phosphoproteins/analysis , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Receptors, Fibroblast Growth Factor/analysis , Receptors, Fibroblast Growth Factor/genetics , Transcription Factors , YAP-Signaling Proteins
6.
J Cell Biochem ; 118(10): 3249-3259, 2017 10.
Article in English | MEDLINE | ID: mdl-28262979

ABSTRACT

Saturated fatty acids (SFA) and their toxic metabolites contribute to hepatocyte lipotoxicity in nonalcoholic steatohepatitis (NASH). We previously reported that hepatocytes, under lipotoxic stress, express the potent macrophage chemotactic ligand C-X-C motif chemokine 10 (CXCL10), and release CXCL10-enriched extracellular vesicles (EV) by a mixed lineage kinase (MLK) 3-dependent mechanism. In the current study, we sought to examine the signaling pathway responsible for CXCL10 induction during hepatocyte lipotoxicity. Here, we demonstrate a role for signal transducer and activator of transcription (STAT) 1 in regulating CXCL10 expression. Huh7 and HepG2 cells were treated with lysophosphatidylcholine (LPC), the toxic metabolite of the SFA palmitate. In LPC-treated hepatocytes, CXCL10 induction is mediated by a mitogen activated protein kinase (MAPK) signaling cascade consisting of a relay kinase module of MLK3, MKK3/6, and p38. P38 in turn induces STAT1 Ser727 phosphorylation and CXCL10 upregulation in hepatocytes, which is reduced by genetic or pharmacological inhibition of this MAPK signaling cascade. The binding and activity of STAT1 at the CXCL10 gene promoter were identified by chromatin immunoprecipitation and luciferase gene expression assays. Promoter activation was attenuated by MLK3/STAT1 inhibition or by deletion of the consensus STAT1 binding sites within the CXCL10 promoter. In lipotoxic hepatocytes, MLK3 activates a MAPK signaling cascade, resulting in the activating phosphorylation of STAT1, and CXCL10 transcriptional upregulation. Hence, this kinase relay module and/or STAT1 inhibition may serve as a therapeutic target to reduce CXCL10 release, thereby attenuating NASH pathogenesis. J. Cell. Biochem. 118: 3249-3259, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Chemokine CXCL10/metabolism , Hepatocytes/metabolism , MAP Kinase Kinase Kinases/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , STAT1 Transcription Factor/metabolism , Hep G2 Cells , Hepatocytes/pathology , Humans , Lysophosphatidylcholines/toxicity , MAP Kinase Signaling System/drug effects , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/pathology , Palmitic Acid/toxicity , omega-Chloroacetophenone , Mitogen-Activated Protein Kinase Kinase Kinase 11
7.
Gastroenterology ; 150(4): 956-67, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26764184

ABSTRACT

BACKGROUND & AIMS: Hepatocyte cellular dysfunction and death induced by lipids and macrophage-associated inflammation are characteristics of nonalcoholic steatohepatitis (NASH). The fatty acid palmitate can activate death receptor 5 (DR5) on hepatocytes, leading to their death, but little is known about how this process contributes to macrophage-associated inflammation. We investigated whether lipid-induced DR5 signaling results in the release of extracellular vesicles (EVs) from hepatocytes, and whether these can induce an inflammatory macrophage phenotype. METHODS: Primary mouse and human hepatocytes and Huh7 cells were incubated with palmitate, its metabolite lysophosphatidylcholine, or diluent (control). The released EV were isolated, characterized, quantified, and applied to macrophages. C57BL/6 mice were placed on chow or a diet high in fat, fructose, and cholesterol to induce NASH. Some mice also were given the ROCK1 inhibitor fasudil; 2 weeks later, serum EVs were isolated and characterized by immunoblot and nanoparticle-tracking analyses. Livers were collected and analyzed by histology, immunohistochemistry, and quantitative polymerase chain reaction. RESULTS: Incubation of primary hepatocytes and Huh7 cells with palmitate or lysophosphatidylcholine increased their release of EVs, compared with control cells. This release was reduced by inactivating mediators of the DR5 signaling pathway or rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) inhibition. Hepatocyte-derived EVs contained tumor necrosis factor-related apoptosis-inducing ligand and induced expression of interleukin 1ß and interleukin 6 messenger RNAs in mouse bone marrow-derived macrophages. Activation of macrophages required DR5 and receptor-interacting protein kinase 1. Administration of the ROCK1 inhibitor fasudil to mice with NASH reduced serum levels of EVs; this reduction was associated with decreased liver injury, inflammation, and fibrosis. CONCLUSIONS: Lipids, which stimulate DR5, induce release of hepatocyte EVs, which activate an inflammatory phenotype in macrophages. Strategies to inhibit ROCK1-dependent release of EVs by hepatocytes might be developed for the treatment of patients with NASH.


Subject(s)
Extracellular Vesicles/drug effects , Hepatitis/metabolism , Hepatocytes/drug effects , Inflammation Mediators/metabolism , Liver/drug effects , Lysophosphatidylcholines/pharmacology , Non-alcoholic Fatty Liver Disease/metabolism , Palmitic Acid/pharmacology , Signal Transduction/drug effects , Animals , Caspases/metabolism , Cell Line, Tumor , Extracellular Vesicles/metabolism , HEK293 Cells , Hepatitis/drug therapy , Hepatitis/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Phenotype , Protein Kinase Inhibitors/pharmacology , RNA Interference , Rats, Sprague-Dawley , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Transfection , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
8.
Hepatology ; 63(3): 731-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26406121

ABSTRACT

UNLABELLED: Mixed lineage kinase 3 (MLK3) deficiency reduces macrophage-associated inflammation in a murine model of nonalcoholic steatohepatitis (NASH). However, the mechanistic links between MLK3 activation in hepatocytes and macrophage-driven inflammation in NASH are uncharted. Herein, we report that MLK3 mediates the release of (C-X-C motif) ligand 10 (CXCL10)-laden extracellular vesicles (EVs) from lipotoxic hepatocytes, which induce macrophage chemotaxis. Primary mouse hepatocytes (PMHs) and Huh7 cells were treated with palmitate or lysophosphatidylcholine (LPC). Released EVs were isolated by differential ultracentrifugation. LPC treatment of PMH or Huh7 cells induced release of EVs, which was prevented by either genetic or pharmacological inhibition of MLK3. Mass spectrometry identified the potent chemokine, CXCL10, in the EVs, which was markedly enriched in EVs isolated from LPC-treated hepatocytes versus untreated cells. Green fluorescent protein (GFP)-tagged CXCL10 was present in vesicular structures and colocalized with the red fluorescent protein (RFP)-tagged EV marker, CD63, after LPC treatment of cotransfected Huh-7 cells. Either genetic deletion or pharmacological inhibition of MLK3 prevented CXCL10 enrichment in EVs. Treatment of mouse bone-marrow-derived macrophages with lipotoxic hepatocyte-derived EVs induced macrophage chemotaxis, an effect blocked by incubation with CXCL10-neutralizing antisera. MLK3-deficient mice fed a NASH-inducing diet had reduced concentrations of total plasma EVs and CXCL10 containing EVs compared to wild-type mice. CONCLUSIONS: During hepatocyte lipotoxicity, activated MLK3 induces the release of CXCL10-bearing vesicles from hepatocytes, which are chemotactic for macrophages.


Subject(s)
Chemokine CXCL10/metabolism , Extracellular Vesicles/metabolism , Hepatocytes/metabolism , MAP Kinase Kinase Kinases/metabolism , Macrophages/physiology , Animals , Cell Line, Tumor , Chemotaxis , Disease Models, Animal , Humans , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Mitogen-Activated Protein Kinase Kinase Kinase 11
9.
Hepatology ; 61(5): 1627-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25580681

ABSTRACT

UNLABELLED: Cholangiocarcinoma (CCA) is a lethal hepatobiliary neoplasm originating from the biliary apparatus. In humans, CCA risk factors include hepatobiliary inflammation and fibrosis. The recently identified interleukin (IL)-1 family member, IL-33, has been shown to be a biliary mitogen which also promotes liver inflammation and fibrosis. Our aim was to generate a mouse model of CCA mimicking the human disease. Ectopic oncogene expression in the biliary tract was accomplished by the Sleeping Beauty transposon transfection system with transduction of constitutively active AKT (myr-AKT) and Yes-associated protein. Intrabiliary instillation of the transposon-transposase complex was coupled with lobar bile duct ligation in C57BL/6 mice, followed by administration of IL-33 for 3 consecutive days. Tumors developed in 72% of the male mice receiving both oncogenes plus IL-33 by 10 weeks but in only 20% of the male mice transduced with the oncogenes alone. Tumors expressed SOX9 and pancytokeratin (features of CCA) but were negative for HepPar1 (a marker of hepatocellular carcinoma). Substantive overlap with human CCA specimens was revealed by RNA profiling. Not only did IL-33 induce IL-6 expression by human cholangiocytes but it likely facilitated tumor development in vivo by an IL-6-sensitive process as tumor development was significantly attenuated in Il-6(-/-) male animals. Furthermore, tumor formation occurred at a similar rate when IL-6 was substituted for IL-33 in this model. CONCLUSION: The transposase-mediated transduction of constitutively active AKT and Yes-associated protein in the biliary epithelium coupled with lobar obstruction and IL-33 administration results in the development of CCA with morphological and biochemical features of the human disease; this model highlights the role of inflammatory cytokines in CCA oncogenesis.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic , Cholangiocarcinoma/genetics , Interleukin-6/physiology , Interleukins/physiology , Oncogenes/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Cycle Proteins , Humans , Interleukin-33 , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Phosphoproteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Tumor Cells, Cultured , YAP-Signaling Proteins
10.
J Biol Chem ; 289(49): 34074-88, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25342755

ABSTRACT

Mitochondria frequently change their shape through fission and fusion in response to physiological stimuli as well as pathological insults. Disrupted mitochondrial morphology has been observed in cholestatic liver disease. However, the role of mitochondrial shape change in cholestasis is not defined. In this study, using in vitro and in vivo models of bile acid-induced liver injury, we investigated the contribution of mitochondrial morphology to the pathogenesis of cholestatic liver disease. We found that the toxic bile salt glycochenodeoxycholate (GCDC) rapidly fragmented mitochondria, both in primary mouse hepatocytes and in the bile transporter-expressing hepatic cell line McNtcp.24, leading to a significant increase in cell death. GCDC-induced mitochondrial fragmentation was associated with an increase in reactive oxygen species (ROS) levels. We found that preventing mitochondrial fragmentation in GCDC by inhibiting mitochondrial fission significantly decreased not only ROS levels but also cell death. We also induced cholestasis in mouse livers via common bile duct ligation. Using a transgenic mouse model inducibly expressing a dominant-negative fission mutant specifically in the liver, we demonstrated that decreasing mitochondrial fission substantially diminished ROS levels, liver injury, and fibrosis under cholestatic conditions. Taken together, our results provide new evidence that controlling mitochondrial fission is an effective strategy for ameliorating cholestatic liver injury.


Subject(s)
Cholestasis/genetics , Dynamins/genetics , Liver Cirrhosis/genetics , Mitochondria, Liver/genetics , Mitochondrial Dynamics/genetics , Mutation , Adenoviridae/genetics , Animals , Cell Death , Cell Line , Cholestasis/metabolism , Cholestasis/pathology , Common Bile Duct/injuries , Dynamins/antagonists & inhibitors , Dynamins/metabolism , Gene Expression , Genetic Vectors , Glycochenodeoxycholic Acid , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Transgenic , Mitochondria, Liver/metabolism , Mitochondria, Liver/ultrastructure , Organelle Shape/genetics , Primary Cell Culture , Reactive Oxygen Species/metabolism
11.
J Biol Chem ; 289(33): 22835-22849, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24973208

ABSTRACT

Desmoplastic malignancies such as cholangiocarcinoma (CCA) are characterized by a dense stroma containing an abundance of myofibroblasts termed cancer-associated fibroblasts (CAF). The CAF phenotype represents an "activated state" in which cells are primed for cell death triggered by BH3 mimetics. Accordingly, this primed state may be therapeutically exploited. To elucidate the mechanisms underlying this poorly understood apoptotic priming, we examined the role of platelet-derived growth factor (PDGF) in CAF priming for cell death given its prominent role in CAF activation. PDGF isomers PDGF-B and PDGF-D are abundantly expressed in CCA cells derived from human specimens. Either isomer sensitizes myofibroblasts to cell death triggered by BH3 mimetics. Similar apoptotic sensitization was observed with co-culture of myofibroblasts and CCA cells. Profiling of Bcl-2 proteins expressed by PDGF-primed myofibroblasts demonstrated an increase in cellular levels of Puma. PDGF-mediated increases in cellular Puma levels induced proapoptotic changes in Bak, which resulted in its binding to Bcl-2. Short hairpin RNA-mediated down-regulation of Puma conferred resistance to PDGF-mediated apoptotic priming. Conversely, the BH3 mimetic navitoclax disrupted Bcl-2/Bak heterodimers, allowing Bak to execute the cell death program. Treatment with a Bcl-2-specific BH3 mimetic, ABT-199, reduced tumor formation and tumor burden in a murine model of cholangiocarcinoma. Collectively, these findings indicate that apoptotic priming of CAF by PDGF occurs via Puma-mediated Bak activation, which can be converted to active full-blown apoptosis by navitoclax or ABT-199 for therapeutic benefit.


Subject(s)
Apoptosis , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Fibroblasts/metabolism , Platelet-Derived Growth Factor/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Fibroblasts/pathology , Humans , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/pharmacology , Sulfonamides/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/genetics
12.
J Hepatol ; 62(5): 1156-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25445398

ABSTRACT

BACKGROUND & AIMS: Low-grade chronic inflammation is a cardinal feature of the metabolic syndrome, yet its pathogenesis is not well defined. The purpose of this study was to examine the role of TRAIL receptor (TR) signaling in the pathogenesis of obesity-associated inflammation using mice with the genetic deletion of TR. METHODS: TR knockout (TR(-/-)) mice and their littermate wild-type (WT) mice were fed a diet high in saturated fat, cholesterol and fructose (FFC) or chow. Metabolic phenotyping, liver injury, and liver and adipose tissue inflammation were assessed. Chemotaxis and activation of mouse bone marrow-derived macrophages (BMDMϕ) was measured. RESULTS: Genetic deletion of TR completely repressed weight gain, adiposity and insulin resistance in FFC-fed mice. Moreover, TR(-/-) mice suppressed steatohepatitis, with essentially normal serum ALT, hepatocyte apoptosis and liver triglyceride accumulation. Gene array data implicated inhibition of macrophage-associated hepatic inflammation in the absence of the TR. In keeping with this, there was diminished accumulation and activation of inflammatory macrophages in liver and adipose tissue. TR(-/-) BMDMϕ manifest reduced chemotaxis and diminished activation of nuclear factor-κ B signaling upon activation by palmitate and lipopolysaccharide. CONCLUSIONS: These data advance the concept that macrophage-associated hepatic and adipose tissue inflammation of nutrient excess requires TR signaling.


Subject(s)
Adipose Tissue , Inflammation , Liver , Macrophages , Obesity , Receptors, TNF-Related Apoptosis-Inducing Ligand , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Chemotaxis , Diet, High-Fat/methods , Disease Models, Animal , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , Macrophage Activation , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Obesity/complications , Obesity/metabolism , Obesity/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction
13.
J Hepatol ; 60(3): 599-605, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24239776

ABSTRACT

BACKGROUND & AIMS: The Hedgehog signaling pathway contributes to cholangiocarcinoma biology. However, canonical Hedgehog signaling requires cilia, and cholangiocarcinoma cells often do not express cilia. To resolve this paradox, we examined non-canonical (G-protein coupled, pertussis toxin sensitive) Hedgehog signaling in cholangiocarcinoma cells. METHODS: Human [non-malignant (H69), malignant (HuCC-T1 and Mz-ChA-1)] and rat [non-malignant (BDE1 and NRC), and malignant (BDEneu)] cell lines were employed for this study. A BDE(ΔLoop2) cell line with the dominant-negative receptor Patched-1 was generated with the Sleeping Beauty transposon transfection system. RESULTS: Cilia expression was readily identified in non-malignant, but not in malignant cholangiocarcinoma cell lines. Although the canonical Hh signaling pathway was markedly attenuated in cholangiocarcinoma cells, they were chemotactic to purmorphamine, a small-molecule direct Smoothened agonist. Purmorphamine also induced remodeling of the actin cytoskeleton with formation of filopodia and lamellipodia-like protrusions. All these biological features of cell migration were pertussis toxin sensitive, a feature of G-protein coupled (Gis) receptors. To further test the role of Hedgehog signaling in vivo, we employed a syngeneic orthotopic rat model of cholangiocarcinoma. In vivo, genetic inhibition of the Hedgehog signaling pathway employing BDE(ΔLoop2) cells or pharmacological inhibition with a small-molecule antagonist of Smoothened, vismodegib, was tumor and metastasis suppressive. CONCLUSIONS: Cholangiocarcinoma cells exhibit non-canonical Hedgehog signaling with chemotaxis despite impaired cilia expression. This non-canonical Hedgehog signaling pathway appears to contribute to cholangiocarcinoma progression, thereby, supporting a role for Hedgehog pathway inhibition in human cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , Chemotaxis , Cholangiocarcinoma/pathology , Hedgehog Proteins/physiology , Signal Transduction/physiology , Cell Line, Tumor , Cilia/physiology , Humans , Neoplasm Metastasis
14.
Hepatology ; 58(4): 1362-74, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23703673

ABSTRACT

UNLABELLED: Cholangiocarcinoma (CCA) cells paradoxically express the death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and thus rely on potent survival signals to circumvent cell death by TRAIL. Hedgehog (Hh) signaling is an important survival pathway in CCA. Herein, we further examine the mechanisms whereby Hh signaling mediates apoptosis resistance in CCA, revealing a pivotal role for the cell division regulating serine/threonine kinase polo-like kinase 2 (PLK2). We employed 50 human CCA samples (25 intrahepatic and 25 extrahepatic CCA) as well as human KMCH-1, Mz-CHA-1, and HUCCT-1 CCA cells for these studies. In vivo experiments were conducted using a syngeneic rat orthotopic CCA model. In human samples, polo-like kinase (PLK)1/2/3-immunoreactive cancer cells were present in the preponderance of intra- and extrahepatic CCA specimens. Inhibition of Hh signaling by cyclopamine reduced PLK2, but not PLK1 or PLK3, messenger RNA and protein expression in vehicle-treated and sonic Hh-treated CCA cells, confirming our previous microarray study. PLK2 regulation by Hh signaling appears to be direct, because the Hh transcription factors, glioma-associated oncogene 1 and 2, bind to the PLK2 promotor. Moreover, inhibition of PLK2 by the PLK inhibitor, BI 6727 (volasertib), or PLK2 knockdown was proapoptotic in CCA cells. BI 6727 administration or PLK2 knockdown decreased cellular protein levels of antiapoptotic myeloid cell leukemia 1 (Mcl-1), an effect reversed by the proteasome inhibitor, MG-132. Finally, BI 6727 administration reduced Mcl-1 protein expression in CCA cells, resulting in CCA cell apoptosis and tumor suppression in vivo. CONCLUSION: PLK2 appears to be an important mediator of Hh survival signaling. These results suggest PLK inhibitors to be of therapeutic value for treatment of human CCA.


Subject(s)
Bile Duct Neoplasms/physiopathology , Bile Ducts, Intrahepatic , Cholangiocarcinoma/physiopathology , Hedgehog Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Survival/physiology , Cholangiocarcinoma/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heterografts , Humans , Male , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/drug effects , Pteridines/pharmacology , Rats , Rats, Inbred F344 , TNF-Related Apoptosis-Inducing Ligand/physiology
15.
Am J Physiol Gastrointest Liver Physiol ; 305(9): G611-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008361

ABSTRACT

Hepatocyte apoptosis is a hallmark of nonalcoholic steatohepatitis. We have previously observed that the saturated free fatty acids (FFAs) induce hepatocyte apoptosis in part via a death receptor 5 (DR5)-mediated signaling pathway. Cellular inhibitor of apoptosis protein 1 and 2 (cIAP-1 and cIAP-2) proteins are potent inhibitors of death receptor-mediated apoptosis. However, the role of the cIAPs in FFA-mediated hepatocyte apoptosis is unexplored. Our aim was to determine whether cIAPs are dysregulated during hepatocyte lipoapoptosis. cIAP proteins underwent rapid cellular elimination following treatment with the saturated FFAs palmitate (PA) and stearate. In contrast, PA did not decrease cIAP-1 and cIAP-2 mRNA expression in the cells. Degradation of cIAPs was dependent on their E3-ligase activity, suggesting that cIAPs undergo autoubiquitination that leads to proteasomal degradation. Huh-7 cells stably expressing shRNA targeting cIAP-1, but not cIAP-2, displayed enhanced sensitivity to PA-mediated apoptosis. Incubation with the SMAC mimetic JP1584, which induces rapid degradation of cIAPs, also enhanced PA-mediated apoptosis. Hepatocytes isolated from DR5 knockout mice exhibited reduced apoptosis following treatment with PA plus JP1584, implying that degradation of cIAPs sensitizes to DR5-mediated cell death pathways. A decrease of cIAP-1 was also observed in tissue from patients with nonalcoholic steatohepatitis compared with normal obese subjects. Collectively, these results implicate proteasomal degradation of cIAPs by FFA as a mechanism contributing to hepatocyte lipoapoptosis.


Subject(s)
Apoptosis/physiology , Hepatocytes/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Fatty Liver/genetics , Fatty Liver/metabolism , Hepatocytes/cytology , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Non-alcoholic Fatty Liver Disease , Palmitates/metabolism , Signal Transduction/physiology
16.
J Biol Chem ; 286(45): 39336-48, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21941003

ABSTRACT

Nonalcoholic steatohepatitis is characterized by hepatic steatosis, elevated levels of circulating free fatty acids (FFA), endoplasmic reticulum (ER) stress, and hepatocyte lipoapoptosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor 5 (DR5) is significantly elevated in patients with nonalcoholic steatohepatitis, and steatotic hepatocytes demonstrate increased sensitivity to TRAIL-mediated cell death. Nonetheless, a role for TRAIL and/or DR5 in mediating lipoapoptotic pathways is unexplored. Here, we examined the contribution of DR5 death signaling to lipoapoptosis by free fatty acids. The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. Lipoapoptosis by palmitate was not inhibited by a soluble human recombinant DR5-Fc chimera protein suggesting that DR5 cytotoxic signaling is ligand-independent. Hepatocytes from murine TRAIL receptor knock-out mice (DR(-/-)) displayed reduced palmitate-mediated lipotoxicity. Likewise, knockdown of DR5 or caspase-8 expression by shRNA technology attenuated palmitate-induced Bax activation and apoptosis in Huh-7 cells, without altering induction of ER stress markers. Similar observations were verified in other cell models. Finally, knockdown of CHOP, an ER stress-mediated transcription factor, reduced DR5 up-regulation and DR5-mediated caspase-8 activation upon palmitate treatment. Collectively, these results suggest that ER stress-induced CHOP activation by palmitate transcriptionally up-regulates DR5, likely resulting in ligand-independent cytotoxic signaling by this death receptor.


Subject(s)
Apoptosis , Fatty Liver/metabolism , Hepatocytes/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Fatty Liver/genetics , Fatty Liver/pathology , Gene Knockdown Techniques , Hepatocytes/pathology , Humans , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Mice , Mice, Knockout , Palmitic Acid/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
17.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G77-84, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21995961

ABSTRACT

Isolated hepatocytes undergo lipoapoptosis, a feature of hepatic lipotoxicity, on treatment with saturated free fatty acids (FFA) such as palmitate (PA). However, it is unknown if palmitate is directly toxic to hepatocytes or if its toxicity is indirect via the generation of lipid metabolites such as lysophosphatidylcholine (LPC). PA-mediated hepatocyte lipoapoptosis is associated with endoplasmic reticulum (ER) stress, c-Jun NH(2)-terminal kinase (JNK) activation, and a JNK-dependent upregulation of the potent proapoptotic BH3-only protein PUMA (p53 upregulated modulator of apoptosis). Our aim was to determine which of these mechanisms of lipotoxicity are activated by PA-derived LPC. We employed Huh-7 cells and isolated murine and human primary hepatocytes. Intracellular LPC concentrations increase linearly as a function of the exogenous, extracellular PA, stearate, or LPC concentration. Incubation of Huh-7 cells or primary hepatocytes with LPC induced cell death by apoptosis in a concentration-dependent manner. Substituting LPC for PA resulted in caspase-dependent cell death that was accompanied by activating phosphorylation of JNK with c-Jun phosphorylation and an increase in PUMA expression. LPC also induced ER stress as manifest by eIF2α phosphorylation and CAAT/enhancer binding homologous protein (CHOP) induction. LPC cytotoxicity was attenuated by pharmacological inhibition of JNK or glycogen synthase kinase-3 (GSK-3). Similarly, short-hairpin RNA (shRNA)-targeted knockdown of CHOP protected Huh-7 cells against LPC-induced toxicity. The LPC-induced PUMA upregulation was prevented by JNK inhibition or shRNA-targeted knockdown of CHOP. Finally, genetic deficiency of PUMA rendered murine hepatocytes resistant to LPC-induced apoptosis. We concluded that LPC-induced lipoapoptosis is dependent on mechanisms largely indistinguishable from PA. These data suggest that FFA-mediated cytotoxicity is indirect via the generation of the toxic metabolite, LPC.


Subject(s)
Apoptosis/drug effects , Hepatocytes/metabolism , Lysophosphatidylcholines/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Hepatocytes/drug effects , Humans , Lysophosphatidylcholines/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Palmitates/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Stearates/metabolism , Transcription Factor CHOP/metabolism , Tumor Suppressor Proteins/metabolism
18.
Hepatology ; 54(6): 2076-88, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22038837

ABSTRACT

UNLABELLED: Cholangiocarcinoma (CCA) cells paradoxically express the death ligand, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and, therefore, are dependent upon potent survival signals to circumvent TRAIL cytotoxicity. CCAs are also highly desmoplastic cancers with a tumor microenvironment rich in myofibroblasts (MFBs). Herein, we examine a role for MFB-derived CCA survival signals. We employed human KMCH-1, KMBC, HuCCT-1, TFK-1, and Mz-ChA-1 CCA cells, as well as human primary hepatic stellate and myofibroblastic LX-2 cells, for these studies. In vivo experiments were conducted using a syngeneic rat orthotopic CCA model. Coculturing CCA cells with myofibroblastic human primary hepatic stellate cells or LX-2 cells significantly decreased TRAIL-induced apoptosis in CCA cells, a cytoprotective effect abrogated by neutralizing platelet-derived growth factor (PDGF)-BB antiserum. Cytoprotection by PDGF-BB was dependent upon Hedgehog (Hh) signaling, because it was abolished by the smoothened (SMO; the transducer of Hh signaling) inhibitor, cyclopamine. PDGF-BB induced cyclic adenosine monophosphate-dependent protein kinase-dependent trafficking of SMO to the plasma membrane, resulting in glioma-associated oncogene (GLI)2 nuclear translocation and activation of a consensus GLI reporter gene-based luciferase assay. A genome-wide messenger RNA expression analysis identified 67 target genes to be commonly up- (50 genes) or down-regulated (17 genes) by both Sonic hedgehog and PDGF-BB in a cyclopamine-dependent manner in CCA cells. Finally, in a rodent CCA in vivo model, cyclopamine administration increased apoptosis in CCA cells, resulting in tumor suppression. CONCLUSIONS: MFB-derived PDGF-BB protects CCA cells from TRAIL cytotoxicity by a Hh-signaling-dependent process. These results have therapeutical implications for the treatment of human CCA.


Subject(s)
Bile Duct Neoplasms/physiopathology , Bile Ducts, Intrahepatic/physiopathology , Cholangiocarcinoma/physiopathology , Hedgehog Proteins/physiology , Proto-Oncogene Proteins c-sis/physiology , Animals , Apoptosis/drug effects , Becaplermin , Bile Duct Neoplasms/genetics , Cell Line, Tumor , Cholangiocarcinoma/genetics , Coculture Techniques , Hepatic Stellate Cells/metabolism , Humans , Male , Rats , Rats, Inbred F344 , Receptors, G-Protein-Coupled/antagonists & inhibitors , Smoothened Receptor , Transcription Factors/metabolism , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1
19.
Liver Int ; 32(3): 400-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22133064

ABSTRACT

BACKGROUND: Cholangiocarcinomas (CCAs) are highly desmoplastic neoplasms with a tumour microenvironment plentiful in myofibroblasts (MFBs). MFB-derived PDGF-BB survival signalling is a mediator of CCA cell resistance to apoptotic stimuli. This raises the concept that targeting PDGFR-ß, a cognate receptor of PDGF-BB, represents a potential strategy for the treatment of human CCA. AIMS: Herein, we examine a role for inhibiting PDGFR-ß in restoring CCA cell sensitivity to apoptotic stimuli in vitro and in vivo. METHODS: We employed human CCA samples from 41 patients (19 intrahepatic and 22 extrahepatic CCA samples), the human CCA cell lines KMCH-1 and HUCCT-1 as well as shPDGFR-ß-KMCH-1 and human myofibroblastic LX-2 cells for these studies. In vivo-experiments were conducted using a syngeneic rat orthotopic CCA model. RESULTS: Of several MFB-derived growth factors profiled, PDGF-BB and CTGF were most abundantly expressed; however, only PDGF-BB attenuated tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) cytotoxicity. Co-culturing CCA cells with PDGF-BB-secreting MFBs significantly decreased TRAIL-induced CCA cell apoptosis when compared with monoculture conditions; this cytoprotective effect was abrogated in the presence of the tyrosine kinase inhibitors imatinib mesylate or linifanib, which inhibit PDGFR-ß. Consistent with these findings, MFB-imparted cytoprotection also was abolished when PDGFR-ß was knocked down as demonstrated in shPDGFR-ß-KMCH-1 cells. Finally, administration of imatinib mesylate increased CCA cell apoptosis and reduced tumour growth in a rodent in vivo-CCA model that mimics the human disease. CONCLUSIONS: Targeting PDGFR-ß sensitizes CCA cells to apoptotic stimuli and appears to be therapeutic in vivo.


Subject(s)
Apoptosis/physiology , Bile Duct Neoplasms/metabolism , Bile Ducts, Extrahepatic/metabolism , Bile Ducts, Intrahepatic/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Becaplermin , Cell Line, Tumor , Cholangiocarcinoma/physiopathology , DNA Primers/genetics , Humans , Immunoblotting , Immunohistochemistry , Keratin-7/metabolism , Microscopy, Fluorescence , Myofibroblasts/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , RNA, Small Interfering/metabolism , Rats , Real-Time Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/metabolism
20.
Exp Cell Res ; 317(1): 107-16, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20951133

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL) is a potential chemotherapeutic agent with high selectivity for malignant cells. Many tumors, however, are resistant to TRAIL cytotoxicity. Although cellular inhibitors of apoptosis 1 and 2 (cIAP-1 and -2) are often over-expressed in cancers, their role in mediating TRAIL resistance remains unclear. Here, we demonstrate that TRAIL-induced apoptosis of liver cancer cells is associated with degradation of cIAP-1 and X-linked IAP (XIAP), whereas cIAP-2 remains unchanged. Lower concentrations of TRAIL causing minimal or no apoptosis do not alter cIAP-1 or XIAP protein levels. Silencing of cIAP-1 expression, but not XIAP or cIAP-2, as well as co-treatment with a second mitochondrial activator of caspases (SMAC) mimetic (which results in rapid depletion of cIAP-1), sensitizes the cells to TRAIL. TRAIL-induced loss of cIAP-1 and XIAP requires caspase activity. In particular, caspase 8 knockdown stabilizes both cIAP-1 and XIAP, while caspase 9 knockdown prevents XIAP, but not cIAP-1 degradation. Cell-free experiments confirmed cIAP-1 is a substrate for caspase 8, with likely multiple cleavage sites. These results suggest that TRAIL-mediated apoptosis proceeds through caspase 8-dependent degradation of cIAP-1. Targeted depletion of cIAP-1 by SMAC mimetics in conjunction with TRAIL may be beneficial for the treatment of human hepatobiliary malignancies.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Inhibitor of Apoptosis Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/genetics , Apoptosis/physiology , Baculoviral IAP Repeat-Containing 3 Protein , Caspase 8/genetics , Caspase 8/physiology , Caspase 9/genetics , Caspase 9/metabolism , Caspase Inhibitors , Gene Knockdown Techniques , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Protein Processing, Post-Translational/drug effects , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL