Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38712079

ABSTRACT

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths, and commonly associated with hepatic fibrosis or cirrhosis. This study aims to establish a rat model mimicking the progression from liver fibrosis to cirrhosis and subsequently to HCC using thioacetamide (TAA). We utilized male Lewis rats, treating them with intra-peritoneal injections of TAA. These rats received bi-weekly injections of either 200 mg/kg TAA or saline (as a control) over a period of 34 weeks. The development of cirrhosis and hepatocarcinogenesis was monitored through histopathological examinations, biochemical markers, and immunohistochemical analyses. Our results demonstrated that chronic TAA administration induced cirrhosis and well-differentiated HCC, characterized by increased fibrosis, altered liver architecture, and enhanced hepatocyte proliferation. Biochemical analyses revealed significant alterations in liver function markers, including elevated alpha-fetoprotein (AFP) levels, without affecting kidney function or causing significant weight loss or mortality in rats. This TAA-induced cirrhosis and HCC rat model successfully replicates the clinical progression of human HCC, including liver function impairment and early-stage liver cancer characteristics. It presents a valuable tool for future research on the mechanisms of antitumor drugs in tumor initiation and development.

2.
Organogenesis ; 20(1): 2313696, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38357804

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality globally. HCC is highly heterogenous with diverse etiologies leading to different driver mutations potentiating unique tumor immune microenvironments. Current therapeutic options, including immune checkpoint inhibitors and combinations, have achieved limited objective response rates for the majority of patients. Thus, a precision medicine approach is needed to tailor specific treatment options for molecular subsets of HCC patients. Lipid nanovesicle platforms, either liposome- (synthetic) or extracellular vesicle (natural)-derived present are improved drug delivery vehicles which may be modified to contain specific cargos for targeting specific tumor sites, with a natural affinity for liver with limited toxicity. This mini-review provides updates on the applications of novel lipid nanovesicle-based therapeutics for HCC precision medicine and the challenges associated with translating this therapeutic subclass from preclinical models to the clinic.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Vesicles , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Precision Medicine , Extracellular Vesicles/pathology , Lipids/therapeutic use , Tumor Microenvironment
3.
Am J Pathol ; 193(5): 558-566, 2023 05.
Article in English | MEDLINE | ID: mdl-36773785

ABSTRACT

Hepatic zonation is critical for most metabolic functions in liver. Wnt signaling plays an important role in establishing and maintaining liver zonation. Yet, the anatomic expression of Wnt signaling components, especially all 10 Frizzled (Fzd) receptors, has not been characterized in adult liver. To address this, the spatial expression of Fzd receptors was quantitatively mapped in adult mouse liver via multiplex fluorescent in situ hybridization. Although all 10 Fzd receptors were expressed within a metabolic unit, Fzd receptors 1, 4, and 6 were the highest expressed. Although most Wnt signaling occurs in zone 3, expression of most Fzd receptors was not zonated. In contrast, Fzd receptor 6 was preferentially expressed in zone 1. Wnt2 and Wnt9b expression was highly zonated and primarily found in zone 3. Therefore, the current results suggest that zonated Wnt/ß-catenin signaling at baseline occurs primarily due to Wnt2 and Wnt9b rather than zonation of Fzd mRNA expression. Finally, the study showed that Fzd receptors and Wnts are not uniformly expressed by all hepatic cell types. Instead, there is broad distribution among both hepatocytes and nonparenchymal cells, including endothelial cells. Overall, this establishment of a definitive mRNA expression atlas, especially of Fzd receptors, opens the door to future functional characterization in healthy and diseased liver states.


Subject(s)
Receptors, Wnt , Wnt Proteins , Mice , Animals , Receptors, Wnt/genetics , Receptors, Wnt/metabolism , Wnt Proteins/genetics , In Situ Hybridization, Fluorescence , Endothelial Cells/metabolism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Liver/metabolism , Wnt Signaling Pathway , RNA, Messenger/genetics , RNA, Messenger/metabolism , beta Catenin/metabolism
4.
Pharmacol Ther ; 241: 108334, 2023 01.
Article in English | MEDLINE | ID: mdl-36535346

ABSTRACT

The liver possesses extraordinary regenerative capacity mainly attributable to the ability of hepatocytes (HCs) and biliary epithelial cells (BECs) to self-replicate. This ability is left over from their bipotent parent cell, the hepatoblast, during development. When this innate regeneration is compromised due to the absence of proliferative parenchymal cells, such as during cirrhosis, HCs and BEC can transdifferentiate; thus, adding another layer of complexity to the process of liver repair. In addition, dysregulated lineage maintenance in these two cell populations has been shown to promote malignant growth in experimental conditions. Here, malignant transformation, driven in part by insufficient maintenance of lineage reprogramming, contributes to end-stage liver disease. Epigenetic changes are key drivers for cell fate decisions as well as transformation by finetuning overall transcription and gene expression. In this review, we address how altered DNA methylation contributes to the initiation and progression of hepatic cell fate conversion and cancer formation. We also discussed the diagnostic and therapeutic potential of targeting DNA methylation in liver cancer, its current limitations, and what future research is necessary to facilitate its contribution to clinical translation.


Subject(s)
Cell Plasticity , Liver Neoplasms , Humans , DNA Methylation , Cell Proliferation , Liver/metabolism , Liver Neoplasms/genetics
5.
Hepatology ; 77(5): 1593-1611, 2023 05 01.
Article in English | MEDLINE | ID: mdl-35862186

ABSTRACT

BACKGROUND AND AIMS: Liver regeneration (LR) following partial hepatectomy (PH) occurs via activation of various signaling pathways. Disruption of a single pathway can be compensated by activation of another pathway to continue LR. The Wnt-ß-catenin pathway is activated early during LR and conditional hepatocyte loss of ß-catenin delays LR. Here, we study mechanism of LR in the absence of hepatocyte-ß-catenin. APPROACH AND RESULTS: Eight-week-old hepatocyte-specific Ctnnb1 knockout mice (ß-catenin ΔHC ) were subjected to PH. These animals exhibited decreased hepatocyte proliferation at 40-120 h and decreased cumulative 14-day BrdU labeling of <40%, but all mice survived, suggesting compensation. Insulin-mediated mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) activation was uniquely identified in the ß-catenin ΔHC mice at 72-96 h after PH. Deletion of hepatocyte regulatory-associated protein of mTOR (Raptor), a critical mTORC1 partner, in the ß-catenin ΔHC mice led to progressive hepatic injury and mortality by 30 dys. PH on early stage nonmorbid Raptor ΔHC -ß-catenin ΔHC mice led to lethality by 12 h. Raptor ΔHC mice showed progressive hepatic injury and spontaneous LR with ß-catenin activation but died by 40 days. PH on early stage nonmorbid Raptor ΔHC mice was lethal by 48 h. Temporal inhibition of insulin receptor and mTORC1 in ß-catenin ΔHC or controls after PH was achieved by administration of linsitinib at 48 h or rapamycin at 60 h post-PH and completely prevented LR leading to lethality by 12-14 days. CONCLUSIONS: Insulin-mTORC1 activation compensates for ß-catenin loss to enable LR after PH. mTORC1 signaling in hepatocytes itself is critical to both homeostasis and LR and is only partially compensated by ß-catenin activation. Dual inhibition of ß-catenin and mTOR may have notable untoward hepatotoxic side effects.


Subject(s)
Liver Regeneration , beta Catenin , Mice , Animals , Liver Regeneration/physiology , beta Catenin/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Insulin/metabolism , Hepatocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/physiology , Mice, Knockout , Cell Proliferation , Sirolimus/pharmacology
6.
Am J Pathol ; 192(9): 1259-1281, 2022 09.
Article in English | MEDLINE | ID: mdl-35718058

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is an epidemic affecting 30% of the US population. It is characterized by insulin resistance, and by defective lipid metabolism and mitochondrial dysfunction in the liver. SLC25A34 is a major repressive target of miR-122, a miR that has a central role in NAFLD and liver cancer. However, little is known about the function of SLC25A34. To investigate SLC25A34 in vitro, mitochondrial respiration and bioenergetics were examined using hepatocytes depleted of Slc25a34 or overexpressing Slc25a34. To test the function of SLC25A34 in vivo, a hepatocyte-specific knockout mouse was generated, and loss of SLC25A34 was assessed in mice maintained on a chow diet and a fast-food diet (FFD), a model for NAFLD. Hepatocytes depleted of Slc25a34 displayed increased mitochondrial biogenesis, lipid synthesis, and ADP/ATP ratio; Slc25a34 overexpression had the opposite effect. In the knockout model on chow diet, SLC25A34 loss modestly affected liver function (altered glucose metabolism was the most pronounced defect). RNA-sequencing revealed changes in metabolic processes, especially fatty acid metabolism. After 2 months on FFD, knockouts had a more severe phenotype, with increased lipid content and impaired glucose tolerance, which was attenuated after longer FFD feeding (6 months). This work thus presents a novel model for studying SLC25A34 in vivo in which SLC25A34 plays a role in mitochondrial respiration and bioenergetics during NAFLD.


Subject(s)
MicroRNAs , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat , Glucose/metabolism , Hepatocytes/metabolism , Homeostasis , Lipid Metabolism , Lipids , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/metabolism
7.
Cell Mol Gastroenterol Hepatol ; 13(2): 565-582, 2022.
Article in English | MEDLINE | ID: mdl-34756982

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic fatty liver disease is a frequent cause of hepatic dysfunction and is now a global epidemic. This ailment can progress to an advanced form called nonalcoholic steatohepatitis (NASH) and end-stage liver disease. Currently, the molecular basis of NASH pathogenesis is poorly understood, and no effective therapies exist to treat NASH. These shortcomings are due to the paucity of experimental NASH models directly relevant to humans. METHODS: We used chimeric mice with humanized liver to investigate nonalcoholic fatty liver disease in a relevant model. We carried out histologic, biochemical, and molecular approaches including RNA-Seq. For comparison, we used side-by-side human NASH samples. RESULTS: Herein, we describe a "humanized" model of NASH using transplantation of human hepatocytes into fumarylacetoacetate hydrolase-deficient mice. Once fed a high-fat diet, these mice develop NAFLD faithfully, recapitulating human NASH at the histologic, cellular, biochemical, and molecular levels. Our RNA-Seq analyses uncovered that a variety of important signaling pathways that govern liver homeostasis are profoundly deregulated in both humanized and human NASH livers. Notably, we made the novel discovery that hepatocyte growth factor (HGF) function is compromised in human and humanized NASH at several levels including a significant increase in the expression of the HGF antagonists known as NK1/NK2 and marked decrease in HGF activator. Based on these observations, we generated a potent, human-specific, and stable agonist of human MET that we have named META4 (Metaphor) and used it in the humanized NASH model to restore HGF function. CONCLUSIONS: Our studies revealed that the humanized NASH model recapitulates human NASH and uncovered that HGF-MET function is impaired in this disease. We show that restoring HGF-MET function by META4 therapy ameliorates NASH and reinstates normal liver function in the humanized NASH model. Our results show that the HGF-MET signaling pathway is a dominant regulator of hepatic homeostasis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat , Hepatocytes/metabolism , Mice , Non-alcoholic Fatty Liver Disease/pathology
8.
Mol Cell Biol ; 41(4)2021 03 24.
Article in English | MEDLINE | ID: mdl-33526450

ABSTRACT

IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a ubiquitously expressed scaffolding protein that is overexpressed in a number of cancers, including liver cancer, and is associated with protumorigenic processes, such as cell proliferation, motility, and adhesion. IQGAP1 can integrate multiple signaling pathways and could be an effective antitumor target. Therefore, we examined the role of IQGAP1 in tumor initiation and promotion during liver carcinogenesis. We found that ectopic overexpression of IQGAP1 in the liver is not sufficient to initiate tumorigenesis. Moreover, we report that the tumor burden and cell proliferation in the diethylnitrosamine-induced liver carcinogenesis model in Iqgap1-/- mice may be driven by MET signaling. In contrast, IQGAP1 overexpression enhanced YAP activation and subsequent NUAK2 expression to accelerate and promote hepatocellular carcinoma (HCC) in a clinically relevant model expressing activated (S45Y) ß-catenin and MET. Here, increasing IQGAP1 expression in vivo does not alter ß-catenin or MET activation; instead, it promotes YAP activity. Overall, we demonstrate that although IQGAP1 expression is not required for HCC development, the gain of IQGAP1 function promotes the rapid onset and increased liver carcinogenesis. Our results show that an adequate amount of IQGAP1 scaffold is necessary to maintain the quiescent status of the liver.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , ras GTPase-Activating Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Liver/metabolism , Liver Neoplasms/genetics , Mice, Knockout , YAP-Signaling Proteins , ras GTPase-Activating Proteins/genetics
9.
Cell Rep ; 31(9): 107711, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492423

ABSTRACT

The availability of an autologous transplantable auxiliary liver would dramatically affect the treatment of liver disease. Assembly and function in vivo of a bioengineered human liver derived from induced pluripotent stem cells (iPSCs) has not been previously described. By improving methods for liver decellularization, recellularization, and differentiation of different liver cellular lineages of human iPSCs in an organ-like environment, we generated functional engineered human mini livers and performed transplantation in a rat model. Whereas previous studies recellularized liver scaffolds largely with rodent hepatocytes, we repopulated not only the parenchyma with human iPSC-hepatocytes but also the vascular system with human iPS-endothelial cells, and the bile duct network with human iPSC-biliary epithelial cells. The regenerated human iPSC-derived mini liver containing multiple cell types was tested in vivo and remained functional for 4 days after auxiliary liver transplantation in immunocompromised, engineered (IL2rg-/-) rats.


Subject(s)
Hepatocytes/transplantation , Tissue Engineering , Activins/genetics , Activins/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Fetus/cytology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Immunocompromised Host , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Rats , Rats, Sprague-Dawley , Tissue Scaffolds/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Am J Pathol ; 190(2): 372-387, 2020 02.
Article in English | MEDLINE | ID: mdl-31843499

ABSTRACT

Aging is associated with inflammation and metabolic syndrome, which manifests in the liver as nonalcoholic fatty liver disease (NAFLD). NAFLD can range in severity from steatosis to fibrotic steatohepatitis and is a major cause of hepatic morbidity. However, the pathogenesis of NAFLD in naturally aged animals is unclear. Herein, we performed a comprehensive study of lipid content and inflammatory signature of livers in 19-month-old aged female mice. These animals exhibited increased body and liver weight, hepatic triglycerides, and inflammatory gene expression compared with 3-month-old young controls. The aged mice also had a significant increase in F4/80+ hepatic macrophages, which coexpressed CD11b, suggesting a circulating monocyte origin. A global knockout of the receptor for monocyte chemoattractant protein (CCR2) prevented excess steatosis and inflammation in aging livers but did not reduce the number of CD11b+ macrophages, suggesting changes in macrophage accumulation precede or are independent from chemokine (C-C motif) ligand-CCR2 signaling in the development of age-related NAFLD. RNA sequencing further elucidated complex changes in inflammatory and metabolic gene expression in the aging liver. In conclusion, we report a previously unknown accumulation of CD11b+ macrophages in aged livers with robust inflammatory and metabolic transcriptomic changes. A better understanding of the hallmarks of aging in the liver will be crucial in the development of preventive measures and treatments for end-stage liver disease in elderly patients.


Subject(s)
Aging/pathology , Chemokine CCL2/metabolism , Disease Models, Animal , Inflammation/pathology , Non-alcoholic Fatty Liver Disease/pathology , Receptors, CCR2/metabolism , Aging/metabolism , Animals , Body Weight , Chemokine CCL2/genetics , Female , Gene Expression Profiling , Inflammation/etiology , Inflammation/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Organ Size , Receptors, CCR2/genetics
11.
Am J Pathol ; 189(6): 1241-1255, 2019 06.
Article in English | MEDLINE | ID: mdl-30928253

ABSTRACT

The liver contains diploid and polyploid hepatocytes (tetraploid, octaploid, etc.), with polyploids comprising ≥90% of the hepatocyte population in adult mice. Polyploid hepatocytes form multipolar spindles in mitosis, which lead to chromosome gains/losses and random aneuploidy. The effect of aneuploidy on liver function is unclear, and the degree of liver aneuploidy is debated, with reports showing aneuploidy affects 5% to 60% of hepatocytes. To study relationships among liver polyploidy, aneuploidy, and adaptation, mice lacking E2f7 and E2f8 in the liver (LKO), which have a polyploidization defect, were used. Polyploids were reduced fourfold in LKO livers, and LKO hepatocytes remained predominantly diploid after extensive proliferation. Moreover, nearly all LKO hepatocytes were euploid compared with control hepatocytes, suggesting polyploid hepatocytes are required for production of aneuploid progeny. To determine whether reduced polyploidy impairs adaptation, LKO mice were bred onto a tyrosinemia background, a disease model whereby the liver can develop disease-resistant, regenerative nodules. Although tyrosinemic LKO mice were more susceptible to morbidities and death associated with tyrosinemia-induced liver failure, they developed regenerating nodules similar to control mice. Analyses revealed that nodules in the tyrosinemic livers were generated by aneuploidy and inactivating mutations. In summary, we identified new roles for polyploid hepatocytes and demonstrated that they are required for the formation of aneuploid progeny and can facilitate adaptation to chronic liver disease.


Subject(s)
Adaptation, Physiological , Hepatocytes/metabolism , Liver Regeneration , Lung Injury/metabolism , Polyploidy , Animals , E2F7 Transcription Factor/deficiency , Gene Knockdown Techniques , Hepatocytes/pathology , Lung Injury/genetics , Lung Injury/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Repressor Proteins/deficiency
12.
Hepatology ; 69(3): 1242-1258, 2019 03.
Article in English | MEDLINE | ID: mdl-30244478

ABSTRACT

The liver contains a mixture of hepatocytes with diploid or polyploid (tetraploid, octaploid, etc.) nuclear content. Polyploid hepatocytes are commonly found in adult mammals, representing ~90% of the entire hepatic pool in rodents. The cellular and molecular mechanisms that regulate polyploidization have been well characterized; however, it is unclear whether diploid and polyploid hepatocytes function similarly in multiple contexts. Answering this question has been challenging because proliferating hepatocytes can increase or decrease ploidy, and animal models with healthy diploid-only livers have not been available. Mice lacking E2f7 and E2f8 in the liver (liver-specific E2f7/E2f8 knockout; LKO) were recently reported to have a polyploidization defect, but were otherwise healthy. Herein, livers from LKO mice were rigorously characterized, demonstrating a 20-fold increase in diploid hepatocytes and maintenance of the diploid state even after extensive proliferation. Livers from LKO mice maintained normal function, but became highly tumorigenic when challenged with tumor-promoting stimuli, suggesting that tumors in LKO mice were driven, at least in part, by diploid hepatocytes capable of rapid proliferation. Indeed, hepatocytes from LKO mice proliferate faster and out-compete control hepatocytes, especially in competitive repopulation studies. In addition, diploid or polyploid hepatocytes from wild-type (WT) mice were examined to eliminate potentially confounding effects associated with E2f7/E2f8 deficiency. WT diploid cells also showed a proliferative advantage, entering and progressing through the cell cycle faster than polyploid cells, both in vitro and during liver regeneration (LR). Diploid and polyploid hepatocytes responded similarly to hepatic mitogens, indicating that proliferation kinetics are unrelated to differential response to growth stimuli. Conclusion: Diploid hepatocytes proliferate faster than polyploids, suggesting that the polyploid state functions as a growth suppressor to restrict proliferation by the majority of hepatocytes.


Subject(s)
Cell Proliferation/genetics , Hepatocytes/cytology , Liver Regeneration/genetics , Polyploidy , Animals , Female , Male , Mice , Mice, Inbred C57BL
13.
Gynecol Oncol ; 141(2): 348-356, 2016 05.
Article in English | MEDLINE | ID: mdl-26946093

ABSTRACT

OBJECTIVE: Nuclear receptors (NRs) play a vital role in the development and progression of several cancers including breast and prostate. Using TCGA data, we sought to identify critical nuclear receptors in high grade serous ovarian cancers (HGSOC) and to confirm these findings using in vitro approaches. METHODS: In silico analysis of TCGA data was performed to identify relevant NRs in HGSOC. Ovarian cancer cell lines were screened for NR expression and functional studies were performed to determine the significance of these NRs in ovarian cancers. NR expression was analyzed in ovarian cancer tissue samples using immunohistochemistry to identify correlations with histology and stage of disease. RESULTS: The NR4A family of NRs was identified as a potential driver of ovarian cancer pathogenesis. Overexpression of NR4A1 in particular correlated with worse progression free survival. Endogenous expression of NR4A1 in normal ovarian samples was relatively high compared to that of other tissue types, suggesting a unique role for this orphan receptor in the ovary. Expression of NR4A1 in HGSOC cell lines as well as in patient samples was variable. NR4A1 primarily localized to the nucleus in normal ovarian tissue while co-localization within the cytoplasm and nucleus was noted in ovarian cancer cell lines and patient tissues. CONCLUSIONS: NR4A1 is highly expressed in a subset of HGSOC samples from patients that have a worse progression free survival. Studies to target NR4A1 for therapeutic intervention should include HGSOC.


Subject(s)
Neoplasms, Glandular and Epithelial/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/biosynthesis , Ovarian Neoplasms/metabolism , Animals , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Genome , Heterografts , Humans , Immunohistochemistry , Mice , Mice, SCID , Neoplasms, Glandular and Epithelial/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Ovarian Neoplasms/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Hepatology ; 64(2): 599-615, 2016 08.
Article in English | MEDLINE | ID: mdl-27016325

ABSTRACT

UNLABELLED: A defining feature of the mammalian liver is polyploidy, a numerical change in the entire complement of chromosomes. The first step of polyploidization involves cell division with failed cytokinesis. Although polyploidy is common, affecting ∼90% of hepatocytes in mice and 50% in humans, the specialized role played by polyploid cells in liver homeostasis and disease remains poorly understood. The goal of this study was to identify novel signals that regulate polyploidization, and we focused on microRNAs (miRNAs). First, to test whether miRNAs could regulate hepatic polyploidy, we examined livers from Dicer1 liver-specific knockout mice, which are devoid of mature miRNAs. Loss of miRNAs resulted in a 3-fold reduction in binucleate hepatocytes, indicating that miRNAs regulate polyploidization. Second, we surveyed age-dependent expression of miRNAs in wild-type mice and identified a subset of miRNAs, including miR-122, that is differentially expressed at 2-3 weeks, a period when extensive polyploidization occurs. Next, we examined Mir122 knockout mice and observed profound, lifelong depletion of polyploid hepatocytes, proving that miR-122 is required for complete hepatic polyploidization. Moreover, the polyploidy defect in Mir122 knockout mice was ameliorated by adenovirus-mediated overexpression of miR-122, underscoring the critical role miR-122 plays in polyploidization. Finally, we identified direct targets of miR-122 (Cux1, Rhoa, Iqgap1, Mapre1, Nedd4l, and Slc25a34) that regulate cytokinesis. Inhibition of each target induced cytokinesis failure and promoted hepatic binucleation. CONCLUSION: Among the different signals that have been associated with hepatic polyploidy, miR-122 is the first liver-specific signal identified; our data demonstrate that miR-122 is both necessary and sufficient in liver polyploidization, and these studies will serve as the foundation for future work investigating miR-122 in liver maturation, homeostasis, and disease. (Hepatology 2016;64:599-615).


Subject(s)
Hepatocytes/physiology , MicroRNAs/physiology , Polyploidy , Animals , Cytokinesis , Liver/cytology , Liver/growth & development , Mice, Inbred C57BL , Mice, Knockout
15.
J Hepatol ; 62(2): 380-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25457204

ABSTRACT

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.


Subject(s)
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
16.
PLoS One ; 9(6): e98817, 2014.
Article in English | MEDLINE | ID: mdl-24892551

ABSTRACT

We sought to identify a secreted biomarker for ß-catenin activation commonly seen in hepatocellular carcinoma (HCC). By examination of our previously published genearray of hepatocyte-specific ß-catenin knockout (KO) livers, we identified secreted factors whose expression may be ß-catenin-dependent. We verified expression and secretion of the leading factor in HCC cells transfected with mutated (Hep3BS33Y)-ß-catenin. Serum levels of biomarker were next investigated in a mouse model of HCC with ß-catenin gene (Ctnnb1) mutations and eventually in HCC patients. Leukocyte cell-derived chemotaxin-2 (LECT2) expression was decreased in KO livers. Hep3BS33Y expressed and secreted more LECT2 in media as compared to Hep3BWT. Mice developing HCC with Ctnnb1 mutations showed significantly higher serum LECT2 levels. However patients with CTNNB1 mutations showed LECT2 levels of 54.28 ± 22.32 ng/mL (Mean ± SD; n = 8) that were insignificantly different from patients with non-neoplastic chronic liver disease (32.8 ± 21.1 ng/mL; n = 15) or healthy volunteers (33.2 ± 7.2 ng/mL; n = 11). Intriguingly, patients without ß-catenin mutations showed significantly higher serum LECT2 levels (54.26 ± 22.25 ng/mL; n = 46). While ß-catenin activation was evident in a subset of non-mutant ß-catenin HCC group with high LECT2 expression, serum LECT2 was unequivocally similar between ß-catenin-active and -normal group. Further analysis showed that LECT2 levels greater than 50 ng/ml diagnosed HCC in patients irrespective of ß-catenin mutations with specificity of 96.1% and positive predictive value of 97.0%. Thus, LECT2 is regulated by ß-catenin in HCC in both mice and men, but serum LECT2 reflects ß-catenin activity only in mice. Serum LECT2 could be a potential biomarker of HCC in patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Animals , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Mice , Mice, Knockout , beta Catenin/genetics , beta Catenin/metabolism
17.
Am J Pathol ; 184(7): 2111-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24819961

ABSTRACT

Hepatocellular carcinoma (HCC), the third most common cause of cancer-related deaths worldwide, lacks effective medical therapy. Large subsets of HCC demonstrate Wnt/ß-catenin activation, making this an attractive therapeutic target. We report strategy and characterization of a novel small-molecule inhibitor, ICG-001, known to affect Wnt signaling by disrupting ß-catenin-CREB binding protein interactions. We queried the ZINC online database for structural similarity to ICG-001 and identified PMED-1 as the lead compound, with ≥70% similarity to ICG-001. PMED-1 significantly reduced ß-catenin activity in hepatoblastoma and several HCC cells, as determined by TOPflash reporter assay, with an IC50 ranging from 4.87 to 32 µmol/L. Although no toxicity was observed in primary human hepatocytes, PMED-1 inhibited Wnt target expression in HCC cells, including those with CTNNB1 mutations, and impaired cell proliferation and viability. PMED-1 treatment decreased ß-catenin-CREB binding protein interactions without affecting total ß-catenin levels or activity of other common kinases. PMED-1 treatment of Tg(OTM:d2EGFP) zebrafish expressing GFP under the ß-catenin/Tcf reporter led to a notable decrease in ß-catenin activity. The PMED effect on ß-catenin signaling lasted from 12 to 24 hours in vitro and 6 to 15 hours in vivo. Thus, using a rapid and cost-effective computational methodology, we have identified a novel and specific small-molecule inhibitor of Wnt signaling that may have implications for HCC treatment.


Subject(s)
Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CREB-Binding Protein/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Drug Discovery , Humans , Inhibitory Concentration 50 , Liver Neoplasms/metabolism , Pyrimidinones/pharmacology , Structure-Activity Relationship , Zebrafish , beta Catenin/metabolism
18.
Curr Cancer Drug Targets ; 13(8): 867-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23822752

ABSTRACT

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.


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