Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Hepatol ; 67(5): 979-990, 2017 11.
Article in English | MEDLINE | ID: mdl-28647567

ABSTRACT

BACKGROUND & AIMS: We investigated the functional role and clinical significance of stearoyl-CoA desaturase-1 (SCD1) mediated endoplasmic reticulum (ER) stress in regulating liver tumor-initiating cells (T-ICs) and sorafenib resistance, with the aim of developing a novel therapeutic strategy against hepatocellular carcinomas (HCCs). METHODS: We evaluated the clinic-pathological relevance of SCD1 and its correlation with sorafenib resistance in large cohorts of HCC clinical samples by qPCR and immunohistochemical analyses. Lentiviral-based overexpression and knockdown approaches were performed to characterize the functional roles of SCD1 in regulating liver T-ICs and sorafenib resistance. Molecular pathways mediating the phenotypic alterations were identified through RNA sequencing analysis and functional rescue experiments. The combinatorial effect of SCD1 inhibition and sorafenib was tested using a patient-derived tumor xenograft (PDTX) model. RESULTS: SCD1 overexpression was found in HCC, which was associated with shorter disease-free survival (p = 0.008, log rank test). SCD1 was found to regulate the populations of liver T-ICs; while its suppression by a SCD1 inhibitor suppressed liver T-ICs and sorafenib resistance. Interestingly, SCD1 was markedly upregulated in our established sorafenib-resistant PDTX model, and its overexpression predicts the clinical response of HCC patients to sorafenib treatment. Suppression of SCD1 forces liver T-ICs to differentiate via ER stress-induced unfolded protein response, resulting in an enhanced sensitivity to sorafenib. The PDTX#1 model, combined with sorafenib treatment and a novel SCD1 inhibitor (SSI-4), showed a maximal growth suppressive effect. CONCLUSIONS: SCD1-mediated ER stress regulates liver T-ICs and sorafenib sensitivity. Targeting SCD1 alone or in combination with sorafenib might be a novel personalized medicine against HCC. Lay summary: In this study, SCD1 was found to play a critical role in regulating liver tumor-initiating cells and sorafenib resistance through the regulation of ER stress-mediated differentiation. Targeting SCD1 in combination with sorafenib may be a novel therapeutic strategy against liver cancer.


Subject(s)
Carcinoma, Hepatocellular , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms , Niacinamide/analogs & derivatives , Phenylurea Compounds , Stearoyl-CoA Desaturase/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm/genetics , Hong Kong , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Niacinamide/administration & dosage , Niacinamide/pharmacokinetics , Pharmacogenomic Testing , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacokinetics , Sorafenib , Survival Analysis
2.
Cell Rep ; 15(6): 1175-89, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27134167

ABSTRACT

Like normal stem cells, tumor-initiating cells (T-ICs) are regulated extrinsically within the tumor microenvironment. Because HCC develops primarily in the context of cirrhosis, in which there is an enrichment of activated fibroblasts, we hypothesized that cancer-associated fibroblasts (CAFs) would regulate liver T-ICs. We found that the presence of α-SMA(+) CAFs correlates with poor clinical outcome. CAF-derived HGF regulates liver T-ICs via activation of FRA1 in an Erk1,2-dependent manner. Further functional analysis identifies HEY1 as a direct downstream effector of FRA1. Using the STAM NASH-HCC mouse model, we find that HGF-induced FRA1 activation is associated with the fibrosis-dependent development of HCC. Thus, targeting the CAF-derived, HGF-mediated c-Met/FRA1/HEY1 cascade may be a therapeutic strategy for the treatment of HCC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Separation , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/pharmacology , Humans , Liver Neoplasms/genetics , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
3.
Liver Int ; 36(5): 737-45, 2016 05.
Article in English | MEDLINE | ID: mdl-26351778

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is often associated with metastasis and recurrence leading to a poor prognosis. Therefore, development of novel treatment regimens is urgently needed to improve the survival of HCC patients. In this study, we aimed to investigate the in vitro and in vivo effects of anti-CD47 antibody alone and in combination with chemotherapy in HCC. METHODS: In this study, we examined the functional effects of anti-CD47 antibody (B6H12) on cell proliferation, sphere formation, migration and invasion, chemosensitivity, macrophage-mediated phagocytosis and tumourigenicity both in vitro and in vivo. The therapeutic efficacy of anti-CD47 antibody alone or in combination with doxorubicin was examined in patient-derived HCC xenograft. RESULTS: Blocking CD47 with anti-CD47 monoclonal antibody (B6H12) at 10 µg/ml could suppress self-renewal, tumourigenicity and migration and invasion abilities of MHCC-97L and Huh-7 cells. Interestingly, anti-CD47 antibody synergized the effect of HCC cells to chemotherapeutic drugs including doxorubicin and cisplatin. Blockade of CD47 by anti-CD47 antibody induced macrophage-mediated phagocytosis. Using a patient-derived HCC xenograft mouse model, we found that anti-CD47 antibody (400 µg/mouse) in combination with doxorubicin (2 mg/kg) exerted maximal effects on tumour suppression, as compared with doxorubicin and anti-CD47 antibody alone. CONCLUSIONS: Anti-CD47 antibody treatment could complement chemotherapy which may be a promising therapeutic strategy for the treatment of HCC patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD47 Antigen/immunology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Macrophages/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Humans , Mice , Xenograft Model Antitumor Assays
4.
Oncotarget ; 6(41): 43483-95, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26536659

ABSTRACT

Hepatocellular carcinoma (HCC) is frequently complicated by the occurrence of intrahepatic and extrahepatic metastases, leading to poor prognosis. To improve the prognosis for HCC patients, there is an urgent need to understand the molecular mechanisms of metastasis in HCC. Since protein Serine/Threonine phosphorylation emerges to be an important posttranslational modification critical in signaling process associated with cell proliferation, survival and metastasis, we employed a pair of primary tumor-derived and corresponding lung-metastatic counterparts (PLC/PRF/5-PT and PLC/PRF/5-LM) and aimed to identify these changes using CelluSpot Serine/Threonine kinase peptide array. Upon analysis, we found phosphorylated level of nucleophosmin (NPM) at Threonine 234/237 (p-NPM-Thr234/237) had remarkably high level in metastatic HCC cells (PLC-LM) than the corresponding primary HCC cell line (PLC-PT). Similar observation was observed in another match primary and their metastatic counterparts (MHCC-97L and MHCC-97H). By immunohistochemical staining, p-NPM-Thr234/237 was consistently found to be preferentially expressed in metastatic HCCs when compared with primary HCC in 28 HCC cases (p < 0.0001). By overexpressing Flag-tagged NPM and its phosphorylation site mutant (Thr234/237A) into low p-NPM-Thr234/237 expressing cells (Hep3B and Huh7) using a lentiviral based approach, we demonstrated that p-NPM-Thr234/237 is critical in invasion and migration of HCC cells, and this effect was mediated by cyclin-dependent kinase 1 (CDK1). Wild-type NPM was found to physically interact with a metastatic gene, ROCK2, and defective in Thr234/237 phosphorylation decreased its binding affinity, resulting in decrease in ROCK2 mediated signaling pathway. Identification of CDK1/p-NPM/ROCK2 signaling pathway provides a novel target for molecular therapy against HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Blotting, Western , CDC2 Protein Kinase , Carcinoma, Hepatocellular/pathology , Cyclin-Dependent Kinases/metabolism , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Liver Neoplasms/pathology , Neoplasm Invasiveness , Nucleophosmin , Phosphorylation , Polymerase Chain Reaction , Signal Transduction/physiology , Threonine/metabolism , rho-Associated Kinases/metabolism
5.
Hepatology ; 62(2): 534-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25902734

ABSTRACT

UNLABELLED: Sorafenib is a new standard treatment for patients with advanced hepatocellular carcinoma (HCC). However, the survival benefit of this treatment is modest, partly owing to drug resistance. Recent evidence has demonstrated the existence of tumor-initiating cells (T-ICs) as the culprit for treatment resistance. To examine whether sorafenib resistance was a result of the presence of liver T-ICs, we developed sorafenib-resistant HCC cells both in vitro and in vivo through continuous exposure to sorafenib. Using these models, we found that sorafenib-resistant clones demonstrated enhanced T-IC properties, including tumorigenicity, self-renewal, and invasiveness. In addition, several T-IC markers were found to be up-regulated, among which CD47 was found to be most significant. Using chromatin immunoprecipitation assays and expression analyses, CD47 expression was found to be regulated by nuclear factor kappa B (NF-κB) through a specific response element in the promoter of CD47, and the site occupancy and expression were increased and decreased upon stimulation and inhibition of NF-κB, respectively. Consistently, NF-κB was activated in sorafenib-resistant HCC cells, and this finding was confirmed in clinical HCC samples, which showed a positive correlation between NF-κB and CD47 expression. Functional characterization of CD47 in sorafenib-resistant HCC cells was evaluated using a lentivirus-based knockdown approach and showed increased sensitization to sorafenib upon CD47 knockdown. Furthermore, blockade of CD47 using anti-CD47 antibody (Ab) showed a similar effect. Using a patient-derived HCC xenograft mouse model, we found that anti-CD47 Ab (500 µg/mouse) in combination with sorafenib (100 mg/kg, orally) exerted synergistic effects on tumor suppression, as compared with sorafenib and anti-CD47 Ab alone. CONCLUSIONS: NF-κB-mediated CD47 up-regulation promotes sorafenib resistance, and targeting CD47 in combination with sorafenib is an attractive therapeutic regimen for the treatment of HCC patients.


Subject(s)
CD47 Antigen/genetics , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/genetics , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Disease Models, Animal , Humans , Liver Neoplasms/genetics , Mice , Mice, SCID , Molecular Targeted Therapy , NF-kappa B/drug effects , NF-kappa B/metabolism , Niacinamide/pharmacology , Random Allocation , Signal Transduction/drug effects , Sorafenib , Treatment Outcome , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...