Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
Hepatol Commun ; 7(9)2023 09 01.
Article En | MEDLINE | ID: mdl-37556373

BACKGROUND: We previously showed that loss of yes-associated protein 1 (YAP) in early liver development (YAPKO) leads to an Alagille syndrome-like phenotype, with failure of intrahepatic bile duct development, severe cholestasis, and chronic hepatocyte adaptations to reduce liver injury. TAZ, a paralog of YAP, was significantly upregulated in YAPKO hepatocytes and interacted with TEA domain family member (TEAD) transcription factors, suggesting possible compensatory activity. METHODS: We deleted both Yap1 and Wwtr1 (which encodes TAZ) during early liver development using the Foxa3 promoter to drive Cre expression, similar to YAPKO mice, resulting in YAP/TAZ double knockout (DKO) and YAPKO with TAZ heterozygosity (YAPKO TAZHET). We evaluated these mice using immunohistochemistry, serum biochemistry, bile acid profiling, and RNA sequencing. RESULTS: DKO mice were embryonic lethal, but their livers were similar to YAPKO, suggesting an extrahepatic cause of death. Male YAPKO TAZHET mice were also embryonic lethal, with insufficient samples to determine the cause. However, YAPKO TAZHET females survived and were phenotypically similar to YAPKO mice, with increased bile acid hydrophilicity and similar global gene expression adaptations but worsened the hepatocellular injury. TAZ heterozygosity in YAPKO impacted the expression of canonical YAP targets Ctgf and Cyr61, and we found changes in pathways regulating cell division and inflammatory signaling correlating with an increase in hepatocyte cell death, cell cycling, and macrophage recruitment. CONCLUSIONS: YAP loss (with or without TAZ loss) aborts biliary development. YAP and TAZ play a codependent critical role in foregut endoderm development outside the liver, but they are not essential for hepatocyte development. TAZ heterozygosity in YAPKO livers increased cell cycling and inflammatory signaling in the setting of chronic injury, highlighting genes that are especially sensitive to TAZ regulation.


Adaptor Proteins, Signal Transducing , Carcinoma, Hepatocellular , Cholestasis , Liver Neoplasms , YAP-Signaling Proteins , Animals , Male , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Endoderm/metabolism , Intracellular Signaling Peptides and Proteins , Trans-Activators/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins/genetics , Female
2.
Clin Transl Med ; 11(4): e379, 2021 04.
Article En | MEDLINE | ID: mdl-33931972

BACKGROUND AND AIMS: 4-phenylbutyric acid (4-PBA) is a low molecular weight fatty acid that is used in clinical practice to treat inherited urea cycle disorders. In previous reports, it acted as a chemical chaperone inhibiting endoplasmic reticulum (ER) stress and unfolded protein response signaling. A few studies have suggested its function against hepatic fibrosis in mice models. However, its role in hepatocarcinogenesis remained unknown. METHODS: 4-PBA was administered alone or in combination with diethylnitrosamine to investigate its long-term effect on liver tumorigenesis. The role of 4-PBA in oncogene-induced hepatocellular carcinoma (HCC) mice model using sleeping beauty system co-expressed with hMet and ß-catenin point mutation (S45Y) was also observed. RNA-seq and PCR array were used to screen the pathways and genes involved. In vitro and in vivo studies were conducted to explore the effect of 4-PBA on liver and validate the underlying mechanism. RESULTS: 4-PBA alone didn't cause liver tumor in long term. However, it promoted liver tumorigenesis in HCC mice models via initiation of liver cancer stem cells (LCSCs) through Wnt5b-Fzd5 mediating ß-catenin signaling. Peroxisome proliferator-activated receptors (PPAR)-α induced by 4-PBA was responsible for the activation of ß-catenin signaling. Thus, intervention of PPAR-α reversed 4-PBA-induced initiation of LCSCs and HCC development in vivo. Further study revealed that 4-PBA could not only upregulate the expression of PPAR-α transcriptionally but also enhance its stabilization via protecting it from proteolysis. Moreover, high PPAR-α expression predicted poor prognosis in HCC patients. CONCLUSIONS: 4-PBA could upregulate PPAR-α to initiate LCSCs by activating ß-catenin signaling pathway, promoting HCC at early stage. Therefore, more discretion should be taken to monitor the potential tumor-promoting effect of 4-PBA under HCC-inducing environment.


Carcinoma, Hepatocellular/chemically induced , Liver Neoplasms/chemically induced , Neoplastic Stem Cells/drug effects , PPAR alpha/metabolism , Phenylbutyrates/pharmacology , Animals , Carcinogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Wnt Signaling Pathway/drug effects
3.
J Cancer ; 11(15): 4614-4624, 2020.
Article En | MEDLINE | ID: mdl-32489479

Platelet-derived growth receptor α (PDGFRα) is a key factor in many pathophysiological processes. The expression level of PDGFRα is significantly elevated in the early stage of liver development and maintained at a lower level in adult normal livers. In this study, we constructed a liver-specific PDGFRαD842 mutant transgenic (TG) mice model to explore the effect of continuous activation of PDGFRα on liver regeneration and hepatocarcinogenesis. 14-day-old TG and wild-type (WT) mice were intraperitoneally injected with diethylnitrosamine (DEN) at a dose of 25 µg/g body weight. Two-month-old male TG and WT mice were subjected to partial hepatectomy (PH). The liver tissues were collected for further analysis at different time points. Overexpression of PDGFRα D842V and its target genes, Akt, c-myc and cyclin D1 in hepatocytes with no overt phenotype versus WT mice were compared. Unexpectedly, a dramatic decrease in hepatocyte proliferation was noted after PH in TG versus WT mice, possibly due to the downregulation of hepatocyte growth factor receptor (MET) and epidermal growth factor receptor (EGFR). No TG mice developed HCC spontaneously after 14 months follow-up. However, TG mice were more resistant to DEN-induced hapatocarcinogenesis at 6, 10, and 12 months of age, showing delayed hepatocyte proliferation and apoptosis, lower tumor incidence, smaller size and fewer number, compared with age-matched WTs, partially through downregulation of MET and EGFR. In conclusion, continuous activation of PDGFRα signaling by expression of PDGFRα D842V does not promote, but inhibit hepatic regeneration and hepatocarcinogenesis, possibly through compensatory downregulation of MET and EGFR.

4.
Oncotarget ; 10(15): 1475-1490, 2019 Feb 19.
Article En | MEDLINE | ID: mdl-30863496

Hepatoblastoma (HB) is the most common pediatric liver malignancy. Around 80% of HB demonstrate simultaneous activation of ß-catenin and Yes-associated protein 1 (Yap1). The mechanism by which these signaling pathways contribute to HB pathogenesis remain obscure. Recently, mTORC1 activation was reported in human HB cells and in a murine HB model driven by ß-catenin and Yap1. Here, we directly investigate the therapeutic impact of mTOR inhibition following HB development in the Yap1-ß-catenin model. HB were established by hydrodynamic tail vein injection of Sleeping Beauty transposase and plasmids coding for ΔN90-ß-catenin and S127A-Yap1. Five weeks after injection, when HB were evident, mice were randomized into Rapamycin diet-fed or basal-diet-fed groups for 5-weeks. Tumor growth was monitored via ultrasound imaging and mice in both groups were euthanized after 5-weeks for molecular analysis. Transcriptomic analysis showed a strong correlation in gene expression between HB in the Yap1-ß-catenin model and HB patient cohorts. Rapamycin treatment decreased HB burden, almost normalizing liver weight to body weight ratio. Ultrasound imaging showed reduction in tumor growth over the duration of Rapamycin treatment as compared to controls. Majority of HB in the controls exhibited crowded fetal or embryonal histology, while remnant tumors in the experimental group showed well-differentiated fetal morphology. Immunohistochemistry confirmed inhibition of mTORC1 in the Rapamycin group. Thus, Rapamycin reduces HB in a clinically relevant model driven by ß-catenin and Yap1, supporting use of mTORC1 inhibitors in their therapy. We also show the utility of standard and 3D ultrasound imaging for monitoring liver tumors in mice.

5.
Cancer Lett ; 410: 20-31, 2017 12 01.
Article En | MEDLINE | ID: mdl-28942012

Sex affects the risk, treatment responses and outcome of many types of cancers. The mechanism of gender disparity in development of hepatocellular carcinoma (HCC) remains obscure. Sex-determining region on Y chromosome (SRY) was overexpressed in approximate 84% male patient HCC. Moreover, we are the first to generate a liver-specific transgenic (TG) murine model with overexpression of the male specific gene SRY. Subject to a single intraperitoneal injection N-nitrosodiethylamine (DEN) at day 14, TG and wildtype (WT) mice of both genders were sacrificed at different time points (6-13.5 months). Overexpression of SRY in male TG and ectopic expression of SRY in female TG livers promoted DEN-induced hepatocarcinogenesis compared to age- and sex-matched WT. This accelerated tumorigenesis in TG of both genders was a consequence of increased injury and inflammation, fibrosis, and compensatory enhancement in hepatocytes proliferation secondary to activation of downstream targets Sox9 and platelet-derived growth factor receptor α (PDGFRα)/phosphoinositide 3-kinase (PI3K)/Akt and c-myc/CyclinD1. In conclusion, activation of SRY and its downstream Sox9 and PDGFRα pathways are commonly involved in male hepatocarcinogenesis, which provides novel insights into gender disparity and sex-specific therapeutic strategies of HCC.


Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Health Status Disparities , Liver Neoplasms/metabolism , Sex-Determining Region Y Protein/metabolism , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclin D1/metabolism , Diethylnitrosamine , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOX9 Transcription Factor/metabolism , Sex Factors , Sex-Determining Region Y Protein/genetics , Signal Transduction , Time Factors , Tumor Microenvironment , Up-Regulation
6.
Gene Expr ; 17(1): 19-34, 2016.
Article En | MEDLINE | ID: mdl-27226410

Triiodothyronine (T3) induces hepatocyte proliferation in rodents. Recent work has shown molecular mechanism for T3's mitogenic effect to be through activation of ß-catenin signaling. Since systemic side effects of T3 may preclude its clinical use, and hepatocytes mostly express T3 hormone receptor ß (TRß), we investigated if selective TRß agonists like GC-1 may also have ß-catenin-dependent hepatocyte mitogenic effects. Here we studied the effect of GC-1 and T3 in conditional knockouts of various Wnt pathway components. We also assessed any regenerative advantage of T3 or GC-1 when given prior to partial hepatectomy in mice. Mice administered GC-1 showed increased pSer675-ß-catenin, cyclin D1, BrdU incorporation, and PCNA. No abnormalities in liver function tests were noted. GC-1-injected liver-specific ß-catenin knockouts (ß-catenin LKO) showed decreased proliferation when compared to wild-type littermates. To address if Wnt signaling was required for T3- or GC-1-mediated hepatocyte proliferation, we used LRP5-6-LKO, which lacks the two redundant Wnt coreceptors. Surprisingly, decreased hepatocyte proliferation was also evident in LRP5-6-LKO in response to T3 and GC-1, despite increased pSer675-ß-catenin. Further, increased levels of active ß-catenin (hypophosphorylated at Ser33, Ser37, and Thr41) were evident after T3 and GC-1 treatment. Finally, mice pretreated with T3 or GC-1 for 7 days followed by partial hepatectomy showed a significant increase in hepatocyte proliferation both at the time (T0) and 24 h after surgery. In conclusion, like T3, TRß-selective agonists induce hepatocyte proliferation through ß-catenin activation via both PKA- and Wnt-dependent mechanisms and confer a regenerative advantage following surgical resection. Hence, these agents may be useful regenerative therapies in liver transplantation or other surgical settings.


Acetates/pharmacology , Cell Proliferation/drug effects , Hepatocytes/drug effects , Liver Regeneration/drug effects , Phenols/pharmacology , Thyroid Hormone Receptors beta/agonists , Triiodothyronine/pharmacology , beta Catenin/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Hepatectomy/methods , Hepatocytes/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Thyroid Hormone Receptors beta/metabolism , Wnt Signaling Pathway/drug effects
7.
Hepatology ; 64(5): 1587-1605, 2016 11.
Article En | MEDLINE | ID: mdl-27097116

Hepatocellular cancer (HCC) remains a significant therapeutic challenge due to its poorly understood molecular basis. In the current study, we investigated two independent cohorts of 249 and 194 HCC cases for any combinatorial molecular aberrations. Specifically we assessed for simultaneous HMET expression or hMet activation and catenin ß1 gene (CTNNB1) mutations to address any concomitant Met and Wnt signaling. To investigate cooperation in tumorigenesis, we coexpressed hMet and ß-catenin point mutants (S33Y or S45Y) in hepatocytes using sleeping beauty transposon/transposase and hydrodynamic tail vein injection and characterized tumors for growth, signaling, gene signatures, and similarity to human HCC. Missense mutations in exon 3 of CTNNB1 were identified in subsets of HCC patients. Irrespective of amino acid affected, all exon 3 mutations induced similar changes in gene expression. Concomitant HMET overexpression or hMet activation and CTNNB1 mutations were evident in 9%-12.5% of HCCs. Coexpression of hMet and mutant-ß-catenin led to notable HCC in mice. Tumors showed active Wnt and hMet signaling with evidence of glutamine synthetase and cyclin D1 positivity and mitogen-activated protein kinase/extracellular signal-regulated kinase, AKT/Ras/mammalian target of rapamycin activation. Introduction of dominant-negative T-cell factor 4 prevented tumorigenesis. The gene expression of mouse tumors in hMet-mutant ß-catenin showed high correlation, with subsets of human HCC displaying concomitant hMet activation signature and CTNNB1 mutations. CONCLUSION: We have identified cooperation of hMet and ß-catenin activation in a subset of HCC patients and modeled this human disease in mice with a significant transcriptomic intersection; this model will provide novel insight into the biology of this tumor and allow us to evaluate novel therapies as a step toward precision medicine. (Hepatology 2016;64:1587-1605).


Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Point Mutation , Proto-Oncogene Proteins c-met/genetics , beta Catenin/genetics , Animals , Gene Expression Regulation, Neoplastic , Male , Mice , Models, Biological
8.
PLoS One ; 11(4): e0152695, 2016.
Article En | MEDLINE | ID: mdl-27100093

AIM: ß-catenin signaling is a major oncogenic pathway in hepatocellular carcinoma (HCC). Since ß-catenin phosphorylation by glycogen synthase kinase 3ß (GSK3ß) and casein kinase 1ε (CK1ε) results in its degradation, mutations affecting these phosphorylation sites cause ß-catenin stabilization. However, the relevance of missense mutations in non-phosphorylation sites in exon 3 remains unclear. The current study explores significance of such mutations in addition to addressing the clinical and biological implications of ß-catenin activation in human HCC. METHODS: Gene alteration in exon3 of CTNNB1, gene expression of ß-catenin targets such as glutamate synthetase (GS), axin2, lect2 and regucalcin (RGN), and protein expression of ß-catenin were examined in 125 human HCC tissues. RESULTS: Sixteen patients (12.8%) showed conventional missense mutations affecting codons 33, 37, 41, and 45. Fifteen additional patients (12.0%) had other missense mutations in codon 32, 34, and 35. Induction of exon3 mutation caused described ß-catenin target gene upregulation in HCC cell line. Interestingly, conventional and non-phosphorylation site mutations were equally associated with upregulation of ß-catenin target genes. Nuclear localization of ß-catenin was associated with poor overall survival (p = 0.0461). Of these patients with nuclear ß-catenin localization, loss of described ß-catenin target gene upregulation showed significant poorer overall survival than others (p = 0.0001). CONCLUSION: This study suggests that both conventional and other missense mutations in exon 3 of CTNNB1 lead to ß-catenin activation in human HCC. Additionally, the mechanism of nuclear ß-catenin localization without upregulation of described ß-catenin target genes might be of clinical importance depending on distinct mechanism.


Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , beta Catenin/genetics , Axin Protein/genetics , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Exons/genetics , Gene Expression/genetics , Glutamate Synthase/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation/genetics , Phosphorylation/genetics , Prognosis , Signal Transduction/genetics , Up-Regulation/genetics
9.
Oncotarget ; 7(21): 30379-95, 2016 May 24.
Article En | MEDLINE | ID: mdl-27105497

Establishing c-Myc's (Myc) role in liver regeneration has proven difficult particularly since the traditional model of partial hepatectomy may provoke an insufficiently demanding proliferative stress. We used a model of hereditary tyrosinemia whereby the affected parenchyma can be gradually replaced by transplanted hepatocytes, which replicate 50-100-fold, over several months. Prior to transplantation, livers from myc-/- (KO) mice were smaller in young animals and larger in older animals relative to myc+/+ (WT) counterparts. KO mice also consumed more oxygen, produced more CO2 and generated more heat. Although WT and KO hepatocytes showed few mitochondrial structural differences, the latter demonstrated defective electron transport chain function. RNAseq revealed differences in transcripts encoding ribosomal subunits, cytochrome p450 members and enzymes for triglyceride and sterol biosynthesis. KO hepatocytes also accumulated neutral lipids. WT and KO hepatocytes repopulated recipient tyrosinemic livers equally well although the latter were associated with a pro-inflammatory hepatic environment that correlated with worsening lipid accumulation, its extracellular deposition and parenchymal oxidative damage. Our results show Myc to be dispensable for sustained in vivo hepatocyte proliferation but necessary for maintaining normal lipid homeostasis. myc-/- livers resemble those encountered in non-alcoholic fatty liver disease and, under sustained proliferative stress, gradually acquire the features of non-alcoholic steatohepatitis.


Hepatocytes/metabolism , Lipid Metabolism/genetics , Liver Regeneration , Proto-Oncogene Proteins c-myc/genetics , Animals , Cell Proliferation , Cell Size , Cells, Cultured , Gene Expression Profiling/methods , Hepatocytes/cytology , Hepatocytes/transplantation , Liver/cytology , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Triglycerides/metabolism
10.
Am J Pathol ; 186(3): 587-99, 2016 Mar.
Article En | MEDLINE | ID: mdl-26772417

All serious liver injuries alter metabolism and initiate hepatic regeneration. Recent studies using partial hepatectomy (PH) and other experimental models of liver regeneration implicate the metabolic response to hepatic insufficiency as an important source of signals that promote regeneration. Based on these considerations, the analyses reported here were undertaken to assess the impact of interrupting the hypoglycemic response to PH on liver regeneration in mice. A regimen of parenteral dextrose infusion that delays PH-induced hypoglycemia for 14 hours after surgery was identified, and the hepatic regenerative response to PH was compared between dextrose-treated and control mice. The results showed that regenerative recovery of the liver was postponed in dextrose-infused mice (versus vehicle control) by an interval of time comparable to the delay in onset of PH-induced hypoglycemia. The regulation of specific liver regeneration-promoting signals, including hepatic induction of cyclin D1 and S-phase kinase-associated protein 2 expression and suppression of peroxisome proliferator-activated receptor γ and p27 expression, was also disrupted by dextrose infusion. These data support the hypothesis that alterations in metabolism that occur in response to hepatic insufficiency promote liver regeneration, and they define specific pro- and antiregenerative molecular targets whose regenerative regulation is postponed when PH-induced hypoglycemia is delayed.


Gene Expression Regulation/drug effects , Glucose/administration & dosage , Hepatic Insufficiency/prevention & control , Hypoglycemia/drug therapy , Liver Regeneration/drug effects , Animals , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Models, Animal , Hepatectomy/adverse effects , Hydrogen-Ion Concentration , Hypoglycemia/etiology , Liver/cytology , Liver/metabolism , Liver/surgery , Male , Mice , Mice, Inbred C57BL , Models, Biological , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism
11.
J Hepatol ; 64(2): 316-325, 2016 Feb.
Article En | MEDLINE | ID: mdl-26505118

BACKGROUND & AIMS: During liver regeneration, hepatocytes are derived from pre-existing hepatocytes. However, if hepatocyte proliferation is compromised, biliary epithelial cells (BECs) become the source of new hepatocytes. We recently reported on a zebrafish liver regeneration model in which BECs extensively contribute to hepatocytes. Using this model, we performed a targeted chemical screen to identify important factors that regulate BEC-driven liver regeneration, the mechanisms of which remain largely unknown. METHODS: Using Tg(fabp10a:CFP-NTR) zebrafish, we examined the effects of 44 selected compounds on BEC-driven liver regeneration. Liver size was assessed by fabp10a:DsRed expression; liver marker expression was analyzed by immunostaining, in situ hybridization and quantitative PCR. Proliferation and apoptosis were also examined. Moreover, we used a mouse liver injury model, choline-deficient, ethionine-supplemented (CDE) diet. RESULTS: We identified 10 compounds that affected regenerating liver size. Among them, only bromodomain and extraterminal domain (BET) inhibitors, JQ1 and iBET151, blocked both Prox1 and Hnf4a induction in BECs. BET inhibition during hepatocyte ablation blocked BEC dedifferentiation into hepatoblast-like cells (HB-LCs). Intriguingly, after JQ1 washout, liver regeneration resumed, indicating temporal, but not permanent, perturbation of liver regeneration by BET inhibition. BET inhibition after hepatocyte ablation suppressed the proliferation of newly generated hepatocytes and delayed hepatocyte maturation. Importantly, Myca overexpression, in part, rescued the proliferation defect. Furthermore, oval cell numbers in mice fed CDE diet were greatly reduced upon JQ1 administration, supporting the zebrafish findings. CONCLUSIONS: BET proteins regulate BEC-driven liver regeneration at multiple steps: BEC dedifferentiation, HB-LC proliferation, the proliferation of newly generated hepatocytes, and hepatocyte maturation.


Azepines/metabolism , Epithelial Cells/physiology , Hepatocytes/physiology , Heterocyclic Compounds, 4 or More Rings/metabolism , Liver Regeneration/physiology , Triazoles/metabolism , Animals , Biliary Tract/pathology , Cell Line , Cell Proliferation/physiology , Cell Transdifferentiation/physiology , Liver/metabolism , Liver/pathology , Mice , Organ Size , Transcription Factors/antagonists & inhibitors , Transcriptional Activation/physiology , Zebrafish
12.
J Hepatol ; 62(2): 380-7, 2015 Feb.
Article En | MEDLINE | ID: mdl-25457204

BACKGROUND & AIMS: Hepatocellular cancer (HCC) remains a disease of poor prognosis, highlighting the relevance of elucidating key molecular aberrations that may be targeted for novel therapies. Wnt signalling activation, chiefly due to mutations in CTNNB1, have been identified in a major subset of HCC patients. While several in vitro proof of concept studies show the relevance of suppressing Wnt/ß-catenin signalling in HCC cells or tumour xenograft models, no study has addressed the impact of ß-catenin inhibition in a relevant murine HCC model driven by Ctnnb1 mutations. METHODS: We studied the in vivo impact of ß-catenin suppression by locked nucleic acid (LNA) antisense treatment, after establishing Ctnnb1 mutation-driven HCC by diethylnitrosamine and phenobarbital (DEN/PB) administration. RESULTS: The efficacy of LNA directed against ß-catenin vs. scrambled on Wnt signalling was demonstrated in vitro in HCC cells and in vivo in normal mice. The DEN/PB model leads to HCC with Ctnnb1 mutations. A complete therapeutic response in the form of abrogation of HCC was observed after ten treatments of tumour-bearing mice with ß-catenin LNA every 48h as compared to the scrambled control. A decrease in ß-catenin activity, cell proliferation and increased cell death was evident after ß-catenin suppression. No effect of ß-catenin suppression was evident in non-Ctnnb1 mutated HCC, observed after DEN-only administration. CONCLUSIONS: Thus, we provide the in vivo proof of concept that ß-catenin suppression in HCC will be of significant therapeutic benefit, provided the tumours display Wnt activation via mechanisms like CTNNB1 mutations.


Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , DNA, Neoplasm/genetics , Liver Neoplasms, Experimental/genetics , Mutation , Oligonucleotides/metabolism , beta Catenin/genetics , Alkylating Agents/therapeutic use , Animals , Blotting, Western , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , DNA Mutational Analysis , Diethylnitrosamine/pharmacology , Enzyme-Linked Immunosorbent Assay , Excitatory Amino Acid Antagonists/therapeutic use , Immunohistochemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred C3H , Phenobarbital/pharmacology , Tumor Cells, Cultured , beta Catenin/drug effects , beta Catenin/metabolism
13.
Am J Pathol ; 184(11): 3013-25, 2014 Nov.
Article En | MEDLINE | ID: mdl-25193591

Acetaminophen (APAP) overdose results in acute liver failure and has limited treatment options. Previous studies show that stimulating liver regeneration is critical for survival after APAP overdose, but the mechanisms remain unclear. In this study, we identified major signaling pathways involved in liver regeneration after APAP-induced acute liver injury using a novel incremental dose model. Liver injury and regeneration were studied in C57BL/6 mice treated with either 300 mg/kg (APAP300) or 600 mg/kg (APAP600) APAP. Mice treated with APAP300 developed extensive liver injury and robust liver regeneration. In contrast, APAP600-treated mice exhibited significant liver injury but substantial inhibition of liver regeneration, resulting in sustained injury and decreased survival. The inhibition of liver regeneration in the APAP600 group was associated with cell cycle arrest and decreased cyclin D1 expression. Several known regenerative pathways, including the IL-6/STAT-3 and epidermal growth factor receptor/c-Met/mitogen-activated protein kinase pathways, were activated, even at APAP600, where regeneration was inhibited. However, canonical Wnt/ß-catenin and NF-κB pathways were activated only in APAP300-treated mice, where liver regeneration was stimulated. Furthermore, overexpression of a stable form of ß-catenin, where serine 45 is mutated to aspartic acid, in mice resulted in improved liver regeneration after APAP overdose. Taken together, our incremental dose model has identified a differential role of several signaling pathways in liver regeneration after APAP overdose and highlighted canonical Wnt signaling as a potential target for regenerative therapies for APAP-induced acute liver failure.


Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Liver Regeneration/physiology , Signal Transduction/physiology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
14.
Gastroenterology ; 147(3): 690-701, 2014 Sep.
Article En | MEDLINE | ID: mdl-24837480

BACKGROUND & AIMS: Aberrant activation of ß-catenin and Yes-associated protein 1 (Yap1) signaling pathways have been associated with the development of multiple tumor types. Yap functions as a transcriptional coactivator by interacting with TEA domain DNA binding proteins. We investigated the interactions among these pathways during hepatic tumorigenesis. METHODS: We used immunohistochemical analysis to determine expression of ß-catenin and Yap1 in liver cancer specimens collected from patients in Europe and the United States, consisting of 104 hepatocellular carcinoma, 62 intrahepatic cholangiocarcinoma, and 94 hepatoblastoma samples. We assessed ß-catenin and Yap1 signaling and interactions in hepatoblastoma cell lines ((HuH6, HepG2, HepT1, HC-AFW1, HepG2, and HC-AFW1); proteins were knocked down with small interfering RNAs, and effects on proliferation and cell death were measured. Sleeping beauty-mediated hydrodynamic transfection was used to overexpress constitutively active forms of ß-catenin (ΔN90/ß-catenin) and Yap1 (YapS127A) in livers of mice; tissues were collected, and histological and immunohistochemical analyses were performed. RESULTS: We observed nuclear localization of ß-catenin and Yap1 in 79% of hepatoblastoma samples but not in most hepatocellular carcinoma or intrahepatic cholangiocarcinoma samples. Yap1 and ß-catenin coprecipitated in hepatoblastoma but not hepatocellular carcinoma cells. Small interfering RNA-mediated knockdown of Yap1 or ß-catenin in hepatoblastoma cells reduced proliferation in an additive manner. Knockdown of Yap1 reduced its ability to coactivate transcription with ß-catenin; ß-catenin inhibitors inactivated Yap1. Overexpression of constitutively active forms of Yap1 and ß-catenin in mouse liver led to rapid tumorigenesis, with 100% mortality by 11 weeks. Tumor cells expressed both proteins, and human hepatoblastoma cells expressed common targets of their 2 signaling pathways. Yap1 binding of TEA domain factors was required for tumorigenesis in mice. CONCLUSIONS: ß-catenin and the transcriptional regulator Yap1 interact physically and are activated in most human hepatoblastoma tissues; overexpression of activated forms of these proteins in livers of mice leads to rapid tumor development. Further analysis of these mice will allow further studies of these pathways in hepatoblastoma pathogenesis and could lead to the identification of new therapeutic targets.


Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic/metabolism , Hepatoblastoma/metabolism , Liver Neoplasms/metabolism , Phosphoproteins/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Death , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Europe , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Hepatoblastoma/genetics , Hepatoblastoma/pathology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Phosphoproteins/genetics , Protein Binding , RNA Interference , Signal Transduction , Time Factors , Transcription Factors , Transcription, Genetic , Transfection , United States , YAP-Signaling Proteins , beta Catenin/genetics
15.
Am J Pathol ; 184(2): 306-8, 2014 Feb.
Article En | MEDLINE | ID: mdl-24418096

This Guest Editorial introduces this month's special Liver Pathobiology Theme Issue, a series of reviews that encompass the discipline of hepatic regenerative medicine.


Liver Regeneration , Regenerative Medicine , Extracellular Matrix/metabolism , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Liver/cytology , Liver/metabolism , Polyploidy , Tissue Engineering
16.
Curr Cancer Drug Targets ; 13(8): 867-78, 2013 Oct.
Article En | MEDLINE | ID: mdl-23822752

Hepatocellular cancer (HCC) is the third cause of death by cancer worldwide. In the current study we target ß- catenin, an oncogene mutated and constitutively active in 20-30% of HCCs, via a novel, cell permeable gamma guanidine-based peptide nucleic acid (γGPNA) antisense oligonucleotide designed against either the transcription or the translation start site of the human ß-catenin gene. Using TOPflash, a luciferase reporter assay, we show that γGPNA targeting the transcription start site showed more robust activity against ß-catenin activity in liver tumor cells that harbor ß-catenin gene mutations (HepG2 & Snu-449). We identified concomitant suppression of ß-catenin expression and of various Wnt targets including glutamine synthetase (GS) and cyclin-D1. Concurrently, γGPNA treatment reduced proliferation, survival and viability of HCC cells. Intriguingly, an angiogenesis quantitative Real-Time-PCR array identified decreased expression of several pro-angiogenic secreted factors such as EphrinA1, FGF-2, and VEGF-A upon ß-catenin inhibition in liver tumor cells. Conversely, transfection of stabilized-ß-catenin mutants enhanced the expression of angiogenic factors like VEGF-A. Conditioned media from HepG2 cells treated with ß-catenin but not the mismatch γGPNA significantly diminished spheroid and tubule formation by SK-Hep1 cells, an HCC-associated endothelial cell line. Thus, we report a novel class of cell permeable and efficacious γGPNAs that effectively targets ß-catenin, a known oncogene in the liver. Our study also identifies a novel role of ß-catenin in liver tumor angiogenesis through paracrine mechanisms in addition to its roles in proliferation, survival, metabolism and cancer stem cell biology, thus further strengthening its effectiveness as a therapeutic target in HCC.


Carcinoma, Hepatocellular/drug therapy , Cell-Penetrating Peptides/pharmacology , Guanidine/chemistry , Liver Neoplasms/drug therapy , Peptide Nucleic Acids/pharmacology , beta Catenin/antagonists & inhibitors , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Gene Knockdown Techniques , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Luciferases/metabolism , Neovascularization, Pathologic , Peptide Nucleic Acids/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Initiation Site , Tumor Cells, Cultured , beta Catenin/genetics
18.
J Biol Chem ; 288(29): 21389-21398, 2013 Jul 19.
Article En | MEDLINE | ID: mdl-23740243

Tissue regeneration requires the activation of a set of specific growth signaling pathways. The identity of these cascades and their biological roles are known; however, the molecular mechanisms regulating the interplay between these pathways remain poorly understood. Here, we define a new role for SULFATASE 2 (SULF2) in regulating tissue regeneration and define the WNT-GLI1 axis as a novel downstream effector for this sulfatase in a liver model of tissue regeneration. SULF2 is a heparan sulfate 6-O-endosulfatase, which releases growth factors from extracellular storage sites turning active multiple signaling pathways. We demonstrate that SULF2-KO mice display delayed regeneration after partial hepatectomy (PH). Mechanistic analysis of the SULF2-KO phenotype showed a decrease in WNT signaling pathway activity in vivo. In isolated hepatocytes, SULF2 deficiency blocked WNT-induced ß-CATENIN nuclear translocation, TCF activation, and proliferation. Furthermore, we identified the transcription factor GLI1 as a novel target of the SULF2-WNT cascade. WNT induces GLI1 expression in a SULF2- and ß-CATENIN-dependent manner. GLI1-KO mice phenocopied the SULF2-KO, showing delayed regeneration and decreased hepatocyte proliferation. Moreover, we identified CYCLIN D1, a key mediator of cell growth during tissue regeneration, as a GLI1 transcriptional target. GLI1 binds to the cyclin d1 promoter and regulates its activity and expression. Finally, restoring GLI1 expression in the liver of SULF2-KO mice after PH rescues CYCLIN D1 expression and hepatocyte proliferation to wild-type levels. Thus, together these findings define a novel pathway in which SULF2 regulates tissue regeneration in part via the activation of a novel WNT-GLI1-CYCLIN D1 pathway.


Kruppel-Like Transcription Factors/metabolism , Liver Regeneration , Sulfatases/metabolism , Wnt Signaling Pathway , Animals , Cell Proliferation/drug effects , Down-Regulation/drug effects , Hedgehog Proteins/metabolism , Hepatectomy , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Kruppel-Like Transcription Factors/genetics , Liver Regeneration/drug effects , Mice , Mice, Inbred C57BL , Models, Biological , Sulfatases/deficiency , Wnt Signaling Pathway/drug effects , Wnt3A Protein/pharmacology , Zinc Finger Protein GLI1 , beta Catenin/metabolism
19.
Cancer Res ; 73(15): 4781-90, 2013 Aug 01.
Article En | MEDLINE | ID: mdl-23749643

Hypoxia promotes angiogenesis, proliferation, invasion, and metastasis of pancreatic cancer. Essentially, all studies of the hypoxia pathway in pancreatic cancer research to date have focused on fully malignant tumors or cancer cell lines, but the potential role of hypoxia inducible factors (HIF) in the progression of premalignant lesions has not been critically examined. Here, we show that HIF2α is expressed early in pancreatic lesions both in human and in a mouse model of pancreatic cancer. HIF2α is a potent oncogenic stimulus, but its role in Kras-induced pancreatic neoplasia has not been discerned. We used the Ptf1aCre transgene to activate Kras(G12D) and delete Hif2α solely within the pancreas. Surprisingly, loss of Hif2α in this model led to markedly higher, rather than reduced, number of low-grade pancreatic intraepithelial neoplasia (mPanIN) lesions. These lesions, however, failed to progress to high-grade mPanINs, and displayed exclusive loss of ß-catenin and SMAD4. The relationship among HIF2α, ß-catenin, and Smad4 was further confirmed in vitro, where silencing of Hif2α resulted in reduced ß-catenin and Smad4 transcript levels. Thus, with oncogenic Ras expressed in the pancreas, HIF2α modulates Wnt-signaling during mPanIN progression by maintaining appropriate levels of both Smad4 and ß-catenin.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Wnt Signaling Pathway/physiology , Animals , Blotting, Western , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Pancreatic Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Smad Proteins/metabolism , beta Catenin/metabolism
20.
Neoplasia ; 15(4): 421-34, 2013 Apr.
Article En | MEDLINE | ID: mdl-23555187

ß-Catenin is important in liver homeostasis as a part of Wnt signaling and adherens junctions (AJs), while its aberrant activation is observed in hepatocellular carcinoma (HCC). We have reported hepatocyte-specific ß-catenin knockout (KO) mice to lack adhesive defects as γ-catenin compensated at AJ. Because γ-catenin is a desmosomal protein, we asked if its increase in KO might deregulate desmosomes. No changes in desmosomal proteins or ultrastructure other than increased plakophilin-3 were observed. To further elucidate the role and regulation of γ-catenin, we contemplate an in vitro model and show γ-catenin increase in HCC cells upon ß-catenin knockdown (KD). Here, γ-catenin is unable to rescue ß-catenin/T cell factor (TCF) reporter activity; however, it sufficiently compensates at AJs as assessed by scratch wound assay, centrifugal assay for cell adhesion (CAFCA), and hanging drop assays. γ-Catenin increase is observed only after ß-catenin protein decrease and not after blockade of its transactivation. γ-Catenin increase is associated with enhanced serine/threonine phosphorylation and abrogated by protein kinase A (PKA) inhibition. In fact, several PKA-binding sites were detected in γ-catenin by in silico analysis. Intriguingly γ-catenin KD led to increased ß-catenin levels and transactivation. Thus, γ-catenin compensates for ß-catenin loss at AJ without affecting desmosomes but is unable to fulfill functions in Wnt signaling. γ-Catenin stabilization after ß-catenin loss is brought about by PKA. Catenin-sensing mechanism may depend on absolute ß-catenin levels and not its activity. Anti-ß-catenin therapies for HCC affecting total ß-catenin may target aberrant Wnt signaling without negatively impacting intercellular adhesion, provided mechanisms leading to γ-catenin stabilization are spared.


Adherens Junctions/metabolism , Desmoplakins/metabolism , beta Catenin/genetics , Animals , Cell Adhesion , Cell Membrane/metabolism , Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Desmogleins/metabolism , Desmoplakins/genetics , Desmosomes/metabolism , Gene Expression , Gene Knockdown Techniques , Hep G2 Cells , Humans , Liver Neoplasms , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plakophilins/metabolism , Protein Processing, Post-Translational , Protein Stability , RNA, Small Interfering/genetics , Wnt Signaling Pathway , beta Catenin/metabolism , gamma Catenin
...