Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nat Commun ; 14(1): 6699, 2023 10 23.
Article in English | MEDLINE | ID: mdl-37872167

ABSTRACT

Increasing evidence has demonstrated that drug resistance can be acquired in cancer cells by kinase rewiring, which is an obstacle for efficient cancer therapy. However, it is technically challenging to measure the expression of protein kinases on large scale due to their dynamic range in human proteome. We employ a lysine-targeted sulfonyl fluoride probe, named XO44, which binds to 133 endogenous kinases in intact lenvatinib-resistant hepatocellular carcinoma (HCC) cells. This analysis reveals cyclin-dependent kinase 6 (CDK6) upregulation, which is mediated by ERK/YAP1 signaling cascade. Functional analyses show that CDK6 is crucial in regulation of acquired lenvatinib resistance in HCC via augmentation of liver cancer stem cells with clinical significance. We identify a noncanonical pathway of CDK6 in which it binds and regulates the activity of GSK3ß, leading to activation of Wnt/ß-catenin signaling. Consistently, CDK6 inhibition by palbociclib or degradation by proteolysis targeting chimeras (PROTACs) is highly synergistic with lenvatinib in vitro. Interestingly, palbociclib not only exerts maximal growth suppressive effect with lenvatinib in lenvatinib-resistant HCC models but also reshapes the tumor immune microenvironment. Together, we unveil CDK6 as a druggable target in lenvatinib-resistant HCC and highlight the use of a chemical biology approach to understand nongenetic resistance mechanisms in cancer.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Up-Regulation , Cyclin-Dependent Kinase 6/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Cell Line, Tumor , Tumor Microenvironment
2.
STAR Protoc ; 4(3): 102506, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37594893

ABSTRACT

Cholesterol biosynthesis supports proliferation and drives resistance to tyrosine kinase inhibitor (TKI) therapy in hepatocellular carcinoma (HCC). Here, we present a protocol for using stable isotopic tracers to track the biosynthesis of cholesterol in cultured HCC cells. We describe steps for cell preparation, incubation, separation, and homogenization. We then detail lipid extraction and compound-specific isotope analysis for comparing and quantifying cholesterol synthesis between TKI-resistant HCC cells and their mock counterparts. This protocol can be expanded for use with other shorter-chained lipids.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Cholesterol , Cell Line , Isotopes
3.
JHEP Rep ; 5(1): 100604, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36440258

ABSTRACT

Background & Aims: SCY1-like pseudokinase 3 (SCYL3) was identified as a binding partner of ezrin, implicating it in metastasis. However, the clinical relevance and functional role of SCYL3 in cancer remain uncharacterized. In this study, we aimed to elucidate the role of SCYL3 in the progression of hepatocellular carcinoma (HCC). Methods: The clinical significance of SCYL3 in HCC was evaluated in publicly available datasets and by qPCR analysis of an in-house HCC cohort. The functional significance and mechanistic consequences of SCYL3 were examined in SCYL3-knockdown/overexpressing HCC cells. In vivo tumor progression was evaluated in Tp53 KO/c-Myc OE mice using the sleeping beauty transposon system. Potential downstream pathways were investigated by co-immunoprecipitation, western blotting analysis and immunofluorescence staining. Results: SCYL3 is often overexpressed in HCC; it is preferentially expressed in metastatic human HCC tumors and is associated with worse patient survival. Suppression of SCYL3 in HCC cells attenuated cell proliferation and migration as well as in vivo metastasis. Intriguingly, endogenous SCYL3 overexpression increased tumor development and metastasis in Tp53 KO/c-Myc OE mice. Mechanistic investigations revealed that SCYL3 physically binds and regulates the stability and transactivating activity of ROCK2 (Rho kinase 2) via its C-terminal domain, leading to the increased formation of actin stress fibers and focal adhesions. Conclusions: These findings reveal that SCYL3 plays a critical role in promoting the progression of HCC and have implications for developing new therapeutic strategies to tackle metastatic HCC. Impact and implications: SCYL3 was first reported to be a binding partner of a metastasis-related gene, ezrin. To date, the clinical relevance and functional role of SCYL3 in cancer remain uncharacterized. Herein, we uncover its crucial role in liver cancer progression. We show that it physically binds and regulates the stability and transactivating activity of ROCK2 leading to HCC tumor progression. Our data provide mechanistic insight that SCYL3-mediated ROCK2 protein stability plays a pivotal role in growth and metastasis of HCC cells. Targeting SCYL3/ROCK2 signaling cascade may be a novel therapeutic strategy for treatment of HCC patients.

4.
Cancer Res ; 82(17): 3102-3115, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35767704

ABSTRACT

Accumulating evidence has demonstrated that drug resistance can be acquired in cancer through the repopulation of tumors by cancer stem cell (CSC) expansion. Here, we investigated mechanisms driving resistance and CSC repopulation in hepatocellular carcinoma (HCC) as a cancer model using two drug-resistant, patient-derived tumor xenografts that mimicked the development of acquired resistance to sorafenib or lenvatinib treatment observed in patients with HCC. RNA sequencing analysis revealed that cholesterol biosynthesis was most commonly enriched in the drug-resistant xenografts. Comparison of the genetic profiles of CD133+ stem cells and CD133- bulk cells from liver regeneration and HCC mouse models showed that the cholesterol pathway was preferentially upregulated in liver CSCs compared with normal liver stem cells. Consistently, SREBP2-mediated cholesterol biosynthesis was crucial for the augmentation of liver CSCs, and loss of SREBP2 conferred sensitivity to tyrosine kinase inhibitors, suggesting a role in regulation of acquired drug resistance in HCC. Similarly, exogenous cholesterol-treated HCC cells showed enhanced cancer stemness abilities and drug resistance. Mechanistically, caspase-3 (CASP3) mediated cleavage of SREBP2 from the endoplasmic reticulum to promote cholesterol biosynthesis, which consequently caused resistance to sorafenib/lenvatinib treatment by driving activation of the sonic hedgehog signaling pathway. Simvastatin, an FDA-approved cholesterol-lowering drug, not only suppressed HCC tumor growth but also sensitized HCC cells to sorafenib. These findings demonstrate that CSC populations in HCC expand via CASP3-dependent, SREBP2-mediated cholesterol biosynthesis in response to tyrosine kinase inhibitor therapy and that targeting cholesterol biosynthesis can overcome acquired drug resistance. SIGNIFICANCE: This study finds that cholesterol biosynthesis supports the expansion of cancer stem cell populations to drive resistance to tyrosine kinase inhibitor therapy in hepatocellular carcinoma, identifying potential therapeutic approaches for improving cancer treatment.


Subject(s)
Carcinoma, Hepatocellular , Caspase 3 , Cholesterol , Liver Neoplasms , Sterol Regulatory Element Binding Protein 2 , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cholesterol/biosynthesis , Drug Resistance, Neoplasm , Hedgehog Proteins/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology , Sterol Regulatory Element Binding Protein 2/metabolism
6.
Cancer Res ; 81(12): 3229-3240, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33903122

ABSTRACT

The survival benefit derived from sorafenib treatment for patients with hepatocellular carcinoma (HCC) is modest due to acquired resistance. Targeting cancer stem cells (CSC) is a possible way to reverse drug resistance, however, inhibitors that specifically target liver CSCs are limited. In this study, we established two sorafenib-resistant, patient-derived tumor xenografts (PDX) that mimicked development of acquired resistance to sorafenib in patients with HCC. RNA-sequencing analysis of sorafenib-resistant PDXs and their corresponding mock controls identified EPH receptor B2 (EPHB2) as the most significantly upregulated kinase. EPHB2 expression increased stepwise from normal liver tissue to fibrotic liver tissue to HCC tissue and correlated with poor prognosis. Endogenous EPHB2 knockout showed attenuation of tumor development in mice. EPHB2 regulated the traits of liver CSCs; similarly, sorted EPHB2High HCC cells were endowed with enhanced CSC properties when compared with their EPHB2-Low counterparts. Mechanistically, EPHB2 regulated cancer stemness and drug resistance by driving the SRC/AKT/GSK3ß/ß-catenin signaling cascade, and EPHB2 expression was regulated by TCF1 via promoter activation, forming a positive Wnt/ß-catenin feedback loop. Intravenous administration of rAAV-8-shEPHB2 suppressed HCC tumor growth and significantly sensitized HCC cells to sorafenib in an NRAS/AKT-driven HCC immunocompetent mouse model. Targeting a positive feedback loop involving the EPHB2/ß-catenin axis may be a possible therapeutic strategy to combat acquired drug resistance in HCC. SIGNIFICANCE: This study identifies a EPHB2/ß-catenin/TCF1 positive feedback loop that augments cancer stemness and sorafenib resistance in HCC, revealing a targetable axis to combat acquired drug resistance in HCC. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3229/F1.large.jpg.


Subject(s)
Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Neoplastic Stem Cells/pathology , Receptor, EphB2/metabolism , Sorafenib/pharmacology , beta Catenin/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplastic Stem Cells/metabolism , Receptor, EphB2/genetics , Tumor Cells, Cultured , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/genetics
7.
Cell Death Dis ; 12(2): 148, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33542213

ABSTRACT

Emerging evidence indicates the role of cancer stem cells (CSCs) in tumor relapse and therapeutic resistance in patients with hepatocellular carcinoma (HCC). To identify novel targets against liver CSCs, an integrative analysis of publicly available datasets involving HCC clinical and stemness-related data was employed to select genes that play crucial roles in HCC via regulation of liver CSCs. We revealed an enrichment of an interstrand cross-link repair pathway, in which ubiquitin-conjugating enzyme E2 T (UBE2T) was the most significantly upregulated. Consistently, our data showed that UBE2T was upregulated in enriched liver CSC populations. Clinically, UBE2T overexpression in HCC was further confirmed at mRNA and protein levels and was correlated with advanced tumor stage and poor patient survival. UBE2T was found to be critically involved in the regulation of liver CSCs, as evidenced by increases in self-renewal, drug resistance, tumorigenicity, and metastasis abilities. Mule, an E3 ubiquitin ligase, was identified to be the direct protein binding partner of UBE2T. Rather than the canonical role of acting as a mediator to transfer ubiquitin to E3 ligases, UBE2T is surprisingly able to physically bind and regulate the protein expression of Mule via ubiquitination. Mule was found to directly degrade ß-catenin protein, and UBE2T was found to mediate liver CSC functions through direct regulation of Mule-mediated ß-catenin degradation; this effect was abolished when the E2 activity of UBE2T was impaired. In conclusion, we revealed a novel UBE2T/Mule/ß-catenin signaling cascade that is involved in the regulation of liver CSCs, which provides an attractive potential therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Ubiquitin-Conjugating Enzymes/metabolism , beta Catenin/metabolism , Carcinoma, Hepatocellular/pathology , Disease Progression , Female , Humans , Liver Neoplasms/pathology , Male
9.
Adv Sci (Weinh) ; 7(21): 2002157, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33173740

ABSTRACT

In hepatocellular carcinoma (HCC) patients with extrahepatic metastasis, the lung is the most frequent site of metastasis. However, how the lung microenvironment favors disseminated cells remains unclear. Here, it is found that nidogen 1 (NID1) in metastatic HCC cell-derived extracellular vesicles (EVs) promotes pre-metastatic niche formation in the lung by enhancing angiogenesis and pulmonary endothelial permeability to facilitate colonization of tumor cells and extrahepatic metastasis. EV-NID1 also activates fibroblasts, which secrete tumor necrosis factor receptor 1 (TNFR1), facilitate lung colonization of tumor cells, and augment HCC cell growth and motility. Administration of anti-TNFR1 antibody effectively diminishes lung metastasis induced by the metastatic HCC cell-derived EVs in mice. In the clinical perspective, analysis of serum EV-NID1 and TNFR1 in HCC patients reveals their positive correlation and association with tumor stages suggesting the potential of these molecules as noninvasive biomarkers for the early detection of HCC. In conclusion, these results demonstrate the interplay of HCC EVs and activated fibroblasts in pre-metastatic niche formation and how blockage of their functions inhibits distant metastasis to the lungs. This study offers promise for the new direction of HCC treatment by targeting oncogenic EV components and their mediated pathways.

11.
Cancer Lett ; 476: 48-56, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32061952

ABSTRACT

Solid evidence shows that tumor-initiating cells (T-ICs) are the root of tumor relapse and drug resistance, which lead to a poor prognosis in patients with hepatocellular carcinoma (HCC). Through an in vitro liver T-IC enrichment approach, we identified nuclear factor (erythroid-derived 2)-like 2 (NRF2) as a transcription regulator that is significantly activated in enriched liver T-IC populations. In human HCCs, NRF2 was found to be overexpressed, which was associated with poor patient survival. Through a lentiviral based knockdown approach, NRF2 was found to be critical for regulating liver T-IC properties, including self-renewal, tumorigenicity, drug resistance and expression of liver T-IC markers. Furthermore, we found that ROS-induced NRF2 activation regulates sorafenib resistance in HCC cells. Mechanistically, NRF2 was found to physically bind to the promoter of sonic hedgehog homolog (SHH), which triggers activation of the sonic hedgehog pathway. The effect of NRF2 knockdown was eliminated upon administration of recombinant SHH, demonstrating that NRF2 mediated T-IC function via upregulation of SHH expression. Our study suggests a novel regulatory mechanism for the canonical sonic hedgehog pathway that may function through the NRF2/SHH/GLI signaling axis, thus mediating T-IC phenotypes.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/metabolism , Liver Neoplasms/pathology , NF-E2-Related Factor 2/metabolism , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cell Lineage , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , NF-E2-Related Factor 2/genetics , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Prognosis , Sorafenib/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Hepatology ; 72(1): 155-168, 2020 07.
Article in English | MEDLINE | ID: mdl-31610028

ABSTRACT

BACKGROUND AND AIMS: The survival benefit of sorafenib for patients with hepatocellular carcinoma (HCC) is unsatisfactory due to the development of adaptive resistance. Increasing evidence has demonstrated that drug resistance can be acquired by cancer cells by activating a number of signaling pathways through receptor tyrosine kinases (RTKs); nevertheless, the detailed mechanism for the activation of these alternative pathways is not fully understood. APPROACH AND RESULTS: Given the physiological role of Src homology 2 domain-containing phosphatase 2 (SHP2) as a downstream effector of many RTKs for activation of various signaling cascades, we first found that SHP2 was markedly up-regulated in our established sorafenib-resistant cell lines as well as patient-derived xenografts. Upon sorafenib treatment, adaptive resistance was acquired in HCC cells through activation of RTKs including AXL, epidermal growth factor receptor, EPH receptor A2, and insulin-like growth factor 1 receptor, leading to RAS/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK), and AKT reactivation. We found that the SHP2 inhibitor SHP099 abrogated sorafenib resistance in HCC cell lines and organoid culture in vitro by blocking this negative feedback mechanism. Interestingly, this sensitization effect was also mediated by induction of cellular senescence. SHP099 in combination with sorafenib was highly efficacious in the treatment of xenografts and genetically engineered models of HCC. CONCLUSIONS: SHP2 blockade by SHP099 in combination with sorafenib attenuated the adaptive resistance to sorafenib by impeding RTK-induced reactivation of the MEK/ERK and AKT signaling pathways. SHP099 in combination with sorafenib may be a safe therapeutic strategy against HCC.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/drug effects , Liver Neoplasms/drug therapy , Piperidines/administration & dosage , Pyrimidines/administration & dosage , SH2 Domain-Containing Protein Tyrosine Phosphatases/antagonists & inhibitors , Sorafenib/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Combinations , Humans , Piperidines/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/physiology , Sorafenib/pharmacology
13.
Int J Cancer ; 145(7): 1860-1873, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30834518

ABSTRACT

Accumulating evidence illustrates the significance of cell plasticity in the molecular biology of liver cancer. Reprogramming of mature parenchymal cells to a less differentiated state by key molecular targets contributes to the pathogenesis of hepatocellular carcinoma (HCC). Hereby, we investigated the role of GATA6, a transcription factor implicated in hepatocyte lineage specification, in HCC. Our results demonstrated a lower expression of GATA6 in HCC tissues compared to the corresponding nontumoral liver tissues. Moreover, GATA6 underexpression, as observed in about 50% cases in our clinical cohort, was associated with a poorer degree of tumor cell differentiation and worse disease-free survival outcome. In vitro, silencing of GATA6 in HCC cells augmented cell migration and invasion abilities of HCC cells by activating epithelial-mesenchymal transition. Self-renewal was also enhanced in vitro. Consistently, in vivo tumorigenicity and self-renewal was promoted upon GATA6 knockdown. Notably, suppression of GATA6 converts HCC cells to a metabolic phenotype recapitulating stem-cell state. Expression of glycolytic markers was elevated in GATA6-knockdown clones accompanied by increased glucose uptake; while overexpression of GATA6 resulted in opposite effects. Further to this, we identified that GATA6 bound to the promoter region of PKM gene and regulated PKM2 transcription. Taken together, downregulation of GATA6 directs HCC cells to glycolytic metabolism and fosters tumorigenicity, self-renewal and metastasis. GATA6 is a transcriptional regulator and a genetic switch that converts the phenotypic reprogramming of HCC cells. It is a potential prognostic biomarker and therapeutic target for liver cancer.


Subject(s)
Carcinoma, Hepatocellular/pathology , Down-Regulation , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Self Renewal , Epithelial-Mesenchymal Transition , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Prognosis , Survival Analysis
14.
Cell Death Differ ; 25(8): 1426-1441, 2018 08.
Article in English | MEDLINE | ID: mdl-29445127

ABSTRACT

Identification and characterization of functional molecular targets conferring stemness properties in hepatocellular carcinoma (HCC) offers crucial insights to overcome the major hurdles of tumor recurrence, metastasis and chemoresistance in clinical management. In the current study, we investigated the significance of Cripto-1 in contributing to HCC stemness. Cripto-1 was upregulated in the sorafenib-resistant clones derived from HCC cell lines and patient-derived xenograft that we previously developed, suggesting an association between Cripto-1 and stemness. By in vitro experiments, Cripto-1 fostered cell proliferation, migration, and invasion. It also enhanced self-renewal ability and conferred chemoresistance of HCC cells. Consistently, silencing of Cripto-1 suppressed in vivo tumorigenicity on serial transplantation. On the downstream signaling mechanism, expression of major components of Wnt/ß-catenin pathway ß-catenin, AXIN2, and C-MYC, accompanied by ß-catenin activity was reduced upon Cripto-1 knockdown. The suppressive effects on stemness properties with Cripto-1 knockdown in vitro and in vivo were partially rescued by forced expression of constitutively active ß-catenin. Further elucidation revealed the binding of Cripto-1 to Frizzled-7 (FZD7), low-density lipoprotein receptor-related protein 6 (LRP6) and Dishevelled-3 (DVL3) of the Wnt/ß-catenin pathway and stabilized DVL3 protein. Analyses with clinical samples validated Cripto-1 overexpression in HCC tissues, as well as a positive correlation between Cripto-1 and AXIN2 expressions. High Cripto-1 level in tumor was associated with poorer disease-free survival of HCC patients. Taken together, Cripto-1 binds to FZD7/LRP6 and DVL3, stabilizes DVL3 expression and activates the Wnt/ß-catenin signaling cascade to confer stemness in HCC. Our study findings substantiated the role of Cripto-1 in determining stemness phenotypes of HCC and mechanistically in modulating the Wnt/ß-catenin signaling cascade, one of the most frequently deregulated pathways in liver cancer.


Subject(s)
Dishevelled Proteins/metabolism , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Apoptosis/drug effects , Axin Protein/genetics , Axin Protein/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Dishevelled Proteins/chemistry , Drug Resistance, Neoplasm/drug effects , Frizzled Receptors/chemistry , Frizzled Receptors/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Low Density Lipoprotein Receptor-Related Protein-6/chemistry , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Sorafenib/pharmacology
15.
Cancer Res ; 78(9): 2332-2342, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29483095

ABSTRACT

Frequent relapse and drug resistance in patients with hepatocellular carcinoma (HCC) can be attributed to the existence of tumor-initiating cells (TIC) within the tumor bulk. Therefore, targeting liver TICs may improve the prognosis of these patients. From transcriptome sequencing of 16 pairs of clinical HCC samples, we report that interleukin-1 receptor-associated kinase 1 (IRAK1) in the TLR/IRAK pathway is significantly upregulated in HCC. IRAK1 overexpression in HCC was further confirmed at the mRNA and protein levels and correlated with advanced tumor stages and poor patient survival. Interestingly, IRAK4, an upstream regulator of IRAK1, was also consistently upregulated. IRAK1 regulated liver TIC properties, including self-renewal, tumorigenicity, and liver TIC marker expression. IRAK1 inhibition sensitized HCC cells to doxorubicin and sorafenib treatment in vitro via suppression of the apoptotic cascade. Pharmacological inhibition of IRAK1 with a specific IRAK1/4 kinase inhibitor consistently suppressed liver TIC populations. We identified aldo-keto reductase family 1 member 10 (AKR1B10) as a novel downstream target of IRAK1, which was found to be overexpressed in HCC and significantly correlated with IRAK1 expression. Knockdown of AKR1B10 negated IRAK1-induced TIC functions via modulation of the AP-1 complex. Inhibition of IRAK1/4 inhibitor in combination with sorafenib synergistically suppressed tumor growth in an HCC xenograft model. In conclusion, targeting the IRAK4/IRAK1/AP-1/AKR1B10 signaling pathway may be a potential therapeutic strategy against HCC.Significance: IRAK4/IRAK1/AP-1/AKR1B10 signaling pathway regulates cancer stemness and drug resistance and may be a novel therapeutic target in HCC. Cancer Res; 78(9); 2332-42. ©2018 AACR.


Subject(s)
Aldehyde Reductase/metabolism , Carcinoma, Hepatocellular/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Aldo-Keto Reductases , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice
16.
Clin Transplant ; 32(3): e13203, 2018 03.
Article in English | MEDLINE | ID: mdl-29345755

ABSTRACT

BACKGROUND: Hepatic progenitor cells (HPC) are induced following liver injury to facilitate regeneration. Acute cellular rejection (ACR) is a common complication after liver transplantation as a result of immune-mediated liver injury. In this study, we characterized HPC phenotype in liver allograft biopsy with ACR. We also explored the correlation between expression HPC immunophenotype and clinicopathological parameters. METHODS: Forty-four liver allograft biopsies performed between 2008 and 2016 in a single center with histologically proven ACR were examined for immunohistochemical expression of HPC markers CK19 and Sox9. The number of positive-staining cells was assessed and correlated with clinicopathological features by statistical analysis. RESULTS: HPC phenotype expression as denoted by CK19 and Sox9 staining was detected in the liver tissue with ACR. The numbers of CK19+ and Sox9+ cells were positively correlated. A larger number of CK19+ cells were associated with higher serum aspartate aminotransferase (AST) level at biopsy. By histological rejection score, a larger number of Sox9+ cells were associated with a higher score of bile duct damage. CONCLUSION: Expression HPC markers were correlated with clinical and histological parameters in ACR. Expression of each individual marker may be more tightly associated with a particular component of the ACR process.


Subject(s)
Biomarkers/metabolism , Graft Rejection/diagnosis , Keratin-19/metabolism , Liver Transplantation/adverse effects , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Adolescent , Adult , Aged , Allografts , Cells, Cultured , Child , Female , Follow-Up Studies , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Survival , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Risk Factors , Stem Cells/pathology , Young Adult
17.
Diagn Pathol ; 12(1): 56, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28764740

ABSTRACT

BACKGROUND: Advancing knowledge in molecular pathogenesis of hepatocellular carcinoma (HCC) opens up new horizons in the diagnostic, prognostic and therapeutic perspectives. Assessing the expression of molecular targets prior to definitive treatment is gaining importance in clinical practice. In this study, we investigated the variation in expression pattern of stemness markers in synchronous multi-focal HCC. METHODS: In the first cohort, 21 liver explants with multi-focal HCC were examined for expression of stemness markers EpCAM, Sox9 and CK19 by immunohistochemistry (IHC). Expression data of 50 tumor nodules were analyzed to determine the concordance of expression among nodules in the same livers. In the second cohort, 14 tumor nodules from 6 multi-focal HCC cases proven as intra-hepatic metastasis were examined for Soc9 immunoexpression. RESULTS: In the first cohort, thirty nodules from 16 cases expressed one or more markers, with Sox9 being most frequently expressed. Complete concordance of expression pattern for all 3 markers was observed in 6 cases. Discrepancy of staining degree was noted in 4 cases for EpCAM, 14 cases for Sox9, and 6 cases for CK19. A two-tier or three-tier difference in staining scores was noted in 5 cases for Sox9 and one case for CK19. With Sox9, identical tumor morphology in terms of Edmondson grading and growth pattern did not infer the same degree of immunoexpression; and the largest tumor nodule was not representative of highest IHC score. In the second cohort of intra-hepatic metastasis, complete concordance of Sox9 expression level was observed in 5 out of 6 cases; while the remaining case showed a 1-tier difference of positive staining. CONCLUSIONS: Our findings suggested that clonality of tumor nodules is apparently an important factor to infer immunoexpression pattern. When there is limited information to discern multiple primaries versus intra-hepatic metastasis in multi-focal HCC, discordant degree of stemness markers expression among tumor nodules was commonly observed especially for markers with higher frequency of expression. Pathological features alone do not necessarily indicate the expression pattern of the synchronous nodule and in this scenario examination of each tumor nodule is justified.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Aged , Biomarkers, Tumor/analysis , Female , Humans , Immunohistochemistry , Male , Middle Aged
18.
Cancer Lett ; 403: 330-338, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28688970

ABSTRACT

The hypoxic microenvironment is well-characterized in hepatocellular carcinoma (HCC). Delineation of hypoxia-responsive events is an integral part to understand the pathogenesis of HCC. We studied the functional role and clinical relevance of Stanniocalcin 1 (STC1), a hypoxia-induced molecular target, in HCC. In our clinical cohort, STC1 transcript was up-regulated in HCC tumor tissues. Moreover, STC1 protein was detected in the sera of HCC patients. A higher serum STC1 level was correlated with larger tumor size and poorer 5-year disease-free survival. Functionally, recombinant STC1 protein (rhSTC1) promoted cell migration and cell invasion in vitro; and the effect was abolished by co-treatment of anti-STC1 neutralizing antibody. By in vivo mouse model, silencing of STC1 in HCC cells downregulated secretory STC1 level and suppressed lung metastasis. Furthermore, we found that rhSTC1 activated the JNK pathway, as evidenced by altered expression of the key molecular targets pJNK and p-c-Jun. The functional effects conferred by rhSTC1 were abrogated by co-treatment of JNK inhibitor. In summary, secretory STC1 enhances metastatic potential of HCC via JNK signaling. It potentially serves as a prognostic serum biomarker and a therapeutic target for HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/enzymology , Cell Movement , Glycoproteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/enzymology , Lung Neoplasms/enzymology , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Cell Movement/drug effects , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Glycoproteins/blood , Glycoproteins/genetics , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden , Tumor Hypoxia , Tumor Microenvironment , Up-Regulation
19.
Oncotarget ; 7(20): 29371-86, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27105493

ABSTRACT

Sox9, an SRY-related HMG box transcription factor, is a progenitor/precursor cell marker of the liver expressed during embryogenesis and following liver injury. In this study, we investigated the role of Sox9 and its molecular mechanism with reference to stemness properties in hepatocellular carcinoma (HCC). Here, we observed upregulation of Sox9 in human HCC tissues compared with the non-tumorous liver counterparts (p < 0.001). Upregulation of Sox9 transcript level was associated with poorer tumor cell differentiation (p = 0.003), venous invasion (p = 0.026), advanced tumor stage (p = 0.044) and shorter overall survival (p = 0.042). Transcript levels of Sox9 and CD24 were positively correlated. Silencing of Sox9 in HCC cells inhibited in vitro cell proliferation and tumorsphere formation, sensitized HCC cells to chemotherapeutic agents, and suppressed in vivo tumorigenicity. In addition, knockdown of Sox9 suppressed HCC cell migration, invasion, and in vivo lung metastasis. Further studies showed that Sox9 endowed stemness features through activation of Wnt/ß-catenin signaling, which was confirmed by the partial rescue effect on tumorigenicity and self-renewal upon transfection of active ß-catenin in Sox9 knockdown cells. By ChIP and luciferase promoter assays, Frizzled-7 was identified to be the direct transcriptional target of Sox9. In conclusion, Sox9 confers stemness properties of HCC through Frizzled-7 mediated Wnt/ß-catenin pathway.


Subject(s)
Carcinoma, Hepatocellular/pathology , Frizzled Receptors/metabolism , Liver Neoplasms/pathology , SOX9 Transcription Factor/metabolism , Wnt Signaling Pathway/physiology , Adult , Aged , Animals , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/physiology , Heterografts , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged
20.
Oncotarget ; 6(13): 10880-92, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25834102

ABSTRACT

Hepatocellular carcinoma (HCC) is characteristically one of the most rapidly proliferating tumors which outgrows functional blood supply and results in regional oxygen deprivation. Overexpression of PIM1, a serine/threonine kinase, has been identified recently in human cancers. Knowledge on PIM1 in HCC is however, scarce. By immunohistochemical analysis on 56 human primary HCC samples, we observed overexpression of PIM1 in 39% of the cases. In two independent cohorts of paired primary and extra-hepatic metastatic HCC tissues, PIM1 expression was higher (p=0.002) in the extra-hepatic metastatic HCC tissues as compared with the corresponding primary HCCs. PIM1 was markedly up-regulated in multiple HCC cell lines in hypoxic condition (1% O2) versus normoxia (20% O2). Silencing of PIM1 suppressed HCC cell invasion in vitro as compared to non-target control, and decreased HCC cell proliferation in vitro and tumor growth and metastatic potential in vivo. Knockdown of PIM1 significantly reduced glucose uptake by HCC cells and was associated with decreased levels of p-AKT and key molecules in the glycolytic pathway. Taken together, PIM1 is up-regulated by hypoxia in HCC and promotes tumor growth and metastasis through facilitating cancer cell glycolysis. Targeting PIM1 may have potential role in the management of HCC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Glycolysis , Liver Neoplasms/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , Active Transport, Cell Nucleus , Aged , Aged, 80 and over , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Burden , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...