Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 537
Filter
1.
Arch Toxicol ; 94(12): 4085-4097, 2020 12.
Article in English | MEDLINE | ID: mdl-32833043

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a recognized risk factor for liver fibrosis and malignancies, and is associated with features of metabolic syndrome, such as obesity and insulin resistance (IR). We previously demonstrated that the disturbance of connexin 32 (Cx32), a gap junctional protein of hepatocytes, exacerbated NASH in Cx32 dominant-negative transgenic (Cx32ΔTg) rats fed methionine choline-deficient diet (MCDD). MCDD is well-established means of inducing NASH in rodents; however, the Cx32ΔTg-MCDD NASH model does not reproduce obesity and IR. In this study, we aimed to establish an improved NASH model. Eight-week-old male Cx32ΔTg and wild-type (Wt) rats received a high-fat diet (HFD) with dimethylnitrosamine (DMN) for 12 weeks. The HFD with DMN led to gains in body, liver, and visceral fat weights in both genotypes. IR was significantly greater in Cx32ΔTg than in Wt rats. Elevation of serum hepatic enzymes (AST, ALT), inflammatory cytokine expressions (Tnfα, Il-6, Tgf-ß1, Il-1ß, Timp2, and Col1a1), steatohepatitis, and fibrosis were significantly greater in Cx32ΔTg as compared with Wt rats. Regarding carcinogenesis, the number and area of glutathione S-transferase placental form (GST-P)-positive preneoplastic hepatic foci were significantly increased in Cx32ΔTg versus Wt rats. Moreover, activation of NF-κB and JNK contributed to the progression of NASH in Cx32ΔTg rats. These results suggest that Cx32 dysfunction promoted the progression of NASH, metabolic syndrome, and carcinogenesis. Therefore, the novel Cx32ΔTg-HFD-DMN NASH model may be a rapid and useful tool for evaluating the progression of NASH.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Connexins/metabolism , Liver Cirrhosis, Experimental/etiology , Liver Neoplasms, Experimental/etiology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Connexins/genetics , Cytokines/metabolism , Diet, High-Fat , Dimethylnitrosamine , Disease Progression , Inflammation Mediators/metabolism , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Rats, Transgenic , Signal Transduction , Gap Junction beta-1 Protein
2.
Cancer Res ; 80(16): 3215-3221, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32606000

ABSTRACT

Type 2 diabetes, which is mainly linked to obesity, is associated with increased incidence of liver cancer. We have previously found that in various models of obesity/diabetes, hyperinsulinemia maintains heightened hepatic expression of cyclin D1, suggesting a plausible mechanism linking diabetes and liver cancer progression. Here we show that cyclin D1 is greatly elevated in human livers with diabetes and is among the most significantly upregulated genes in obese/diabetic liver tumors. Liver-specific cyclin D1 deficiency protected obese/diabetic mice against hepatic tumorigenesis, whereas lean/nondiabetic mice developed tumors irrespective of cyclin D1 status. Cyclin D1 dependency positively correlated with liver cancer sensitivity to palbociclib, an FDA-approved CDK4 inhibitor, which was effective in treating orthotopic liver tumors under obese/diabetic conditions. The antidiabetic drug metformin suppressed insulin-induced hepatic cyclin D1 expression and protected against obese/diabetic hepatocarcinogenesis. These results indicate that the cyclin D1-CDK4 complex represents a potential selective therapeutic vulnerability for liver tumors in obese/diabetic patients. SIGNIFICANCE: Obesity/diabetes-associated liver tumors are specifically vulnerable to cyclin D1 deficiency and CDK4 inhibition, suggesting that the obese/diabetic environment confers cancer-selective dependencies that can be therapeutically exploited.


Subject(s)
Cyclin D1/deficiency , Diabetes Mellitus, Type 2/complications , Liver Neoplasms, Experimental/etiology , Obesity/complications , Animals , Antineoplastic Agents/pharmacology , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Hyperinsulinism/metabolism , Hypoglycemic Agents/pharmacology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/prevention & control , Male , Metformin/pharmacology , Mice , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Up-Regulation
3.
BMC Genomics ; 21(1): 453, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32611366

ABSTRACT

BACKGROUND: One of the health risks posed to astronauts during deep space flights is exposure to high charge, high-energy (HZE) ions (Z > 13), which can lead to the induction of hepatocellular carcinoma (HCC). However, little is known on the molecular mechanisms of HZE irradiation-induced HCC. RESULTS: We performed comparative RNA-Seq transcriptomic analyses to assess the carcinogenic effects of 600 MeV/n 56Fe (0.2 Gy), 1 GeV/n 16O (0.2 Gy), and 350 MeV/n 28Si (0.2 Gy) ions in a mouse model for irradiation-induced HCC. C3H/HeNCrl mice were subjected to total body irradiation to simulate space environment HZE-irradiation, and liver tissues were extracted at five different time points post-irradiation to investigate the time-dependent carcinogenic response at the transcriptomic level. Our data demonstrated a clear difference in the biological effects of these HZE ions, particularly immunological, such as Acute Phase Response Signaling, B Cell Receptor Signaling, IL-8 Signaling, and ROS Production in Macrophages. Also seen in this study were novel unannotated transcripts that were significantly affected by HZE. To investigate the biological functions of these novel transcripts, we used a machine learning technique known as self-organizing maps (SOMs) to characterize the transcriptome expression profiles of 60 samples (45 HZE-irradiated, 15 non-irradiated control) from liver tissues. A handful of localized modules in the maps emerged as groups of co-regulated and co-expressed transcripts. The functional context of these modules was discovered using overrepresentation analysis. We found that these spots typically contained enriched populations of transcripts related to specific immunological molecular processes (e.g., Acute Phase Response Signaling, B Cell Receptor Signaling, IL-3 Signaling), and RNA Transcription/Expression. CONCLUSIONS: A large number of transcripts were found differentially expressed post-HZE irradiation. These results provide valuable information for uncovering the differences in molecular mechanisms underlying HZE specific induced HCC carcinogenesis. Additionally, a handful of novel differentially expressed unannotated transcripts were discovered for each HZE ion. Taken together, these findings may provide a better understanding of biological mechanisms underlying risks for HCC after HZE irradiation and may also have important implications for the discovery of potential countermeasures against and identification of biomarkers for HZE-induced HCC.


Subject(s)
Iron/toxicity , Liver Neoplasms, Experimental/etiology , Oxygen/toxicity , Silicon/toxicity , Animals , Hepatitis/etiology , Hepatitis/genetics , Hepatitis/metabolism , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Machine Learning , Male , Mice , RNA-Seq , Time Factors
4.
Metabolism ; 107: 154220, 2020 06.
Article in English | MEDLINE | ID: mdl-32243868

ABSTRACT

BACKGROUND AND AIMS: Obesity, diabetes and associated non-alcoholic steatohepatitis (NASH) are rising risk factors for hepatocellular carcinoma (HCC). Macrophages are important immune cells involved in inflammation and tumour development. Macrophage inositol-requiring enzyme 1 alpha (IRE1α), an ER-stress protein, has been shown to be involved in macrophage cytokine production, and myeloid-specific IRE1α knock-out (myeloid IRE1α-KO) mice showed reduced weight gain during high-fat diet feeding. However, the effect of myeloid IRE1α on NASH and subsequent HCC development has not been examined. Here, we characterized the transcriptional profile of the hepatic macrophage population in a diabetes-NASH-HCC mouse model, and investigated the effect of myeloid-specific IRE1α deletion on the phenotype of hepatic macrophage subsets and experimental NASH-HCC development. METHODS: Mice with non-functional myeloid IRE1α were created by crossing Ire1a floxed mice with Lysm-Cre mice. Two-day old myeloid IRE1α-KO and wild type (WT) mice were subcutaneously injected with streptozotocin (STZ), and male mice were fed a high-fat, -sucrose, -cholesterol diet (Western diet, WD) from the age of 4 weeks until 21 weeks. Control myeloid IRE1α-KO and WT mice received a PBS injection and were fed a matched control diet. These mice were evaluated for obesity, diabetes, NASH and HCC. The hepatic macrophage population was evaluated by flow cytometry and RNA sequencing on FACS-isolated macrophage subsets. RESULTS: STZ-injection and WD feeding resulted in an impaired glucose tolerance, advanced NASH with fibrosis, and HCC development. Myeloid IRE1α-KO STZ mice showed lower fasting glucose levels at the start of WD feeding, and an improved glucose tolerance and attenuated HCC development after 17 weeks of WD feeding despite a similar degree of liver steatosis and inflammation compared to WT mice. Transcriptomic analysis of WT liver Kupffer cells, macrophages and monocytes revealed phenotypical changes in those cell subsets during NASH-HCC development. Isolated liver Kupffer cells and macrophages from mice with a myeloid IRE1α deletion showed downregulated pathways involved in immune system activation and metabolic pathways (only in Kupffer cells), whereas pathways involved in cell division and metabolism were upregulated in monocytes. These transcriptional differences were attenuated during NASH-HCC development. CONCLUSION: Our results show that myeloid-specific IRE1α deletion results in an altered transcriptional profile of hepatic macrophages and dampens diabetes-induced NASH-HCC development, possibly by attenuated diabetes induction.


Subject(s)
Diabetes Mellitus, Experimental/complications , Endoribonucleases/genetics , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms/etiology , Liver Neoplasms/prevention & control , Non-alcoholic Fatty Liver Disease/complications , Protein Serine-Threonine Kinases/genetics , Animals , Blood Glucose/metabolism , Diet, Western , Glucose Intolerance/prevention & control , Kupffer Cells/pathology , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms, Experimental/genetics , Macrophages/pathology , Mice , Mice, Knockout , Mice, Transgenic , Transcriptional Activation
5.
Int Immunopharmacol ; 83: 106420, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32203905

ABSTRACT

The spleen is the largest secondary immune organ and plays a critical role in the progression of tumor. Psychological stress promotes tumor progression through inhibiting antitumor immune. However, the role of spleen in tumor progression induced by stress is unclear. Here, we showed that restraint stress promoted tumor growth, increased the percentage of CD11b+Gr-1+ MDSC while decreased the percentages of CD3-NK1.1+ NK and CD3+NK1.1+ NKT in the tumor tissues. Restraint stress decreased the percentages of CD3+CD4+ T lymphocytes and CD3+CD8+ T lymphocytes while increased the percentage of CD11b+Gr-1+ MDSC in the blood of tumor-bearing mice. Restraint stress increased the percentages of CD3+CD4+ T lymphocytes, CD3+CD8+ T lymphocytes, CD4+PD1+ T lymphocytes and CD8+PD1+ T lymphocytes while decreased the percentage of CD11b+Gr-1+ MDSC in the spleen of tumor-bearing mice. Interestingly, splenectomy inhibited tumor growth and attenuated the changes of CD3+CD4+ T lymphocytes, CD3+CD8+ T lymphocytes, and CD11b+Gr-1+ MDSC in blood induced by chronic restraint stress. Finally, splenectomy blocked the increases of CD11b+Gr-1+ MDSC but did not attenuate the decreases of CD3-NK1.1+ NK and CD3+NK1.1+ NKT in tumor tissue induced by chronic stress. Together, these data indicate that chronic restraint stress promotes hepatocellular carcinoma growth and suppresses the antitumor immunity of tumor-bearing mice. Splenectomy could inhibit tumor growth and partly block the decrease of antitumor immune activity induced by stress.


Subject(s)
Disease Progression , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/metabolism , Spleen/immunology , Stress, Psychological/immunology , Stress, Psychological/metabolism , Animals , Bone Marrow/immunology , Cell Line, Tumor , Immune Tolerance , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/psychology , Male , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Restraint, Physical/adverse effects , Splenectomy , Stress, Psychological/complications , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
FASEB J ; 34(4): 5590-5609, 2020 04.
Article in English | MEDLINE | ID: mdl-32100354

ABSTRACT

Hypoxia inactivates hypoxia-inducible factor (HIF) prolyl 4-hydroxylases (HIF-P4Hs), which stabilize HIF and upregulate genes to restore tissue oxygenation. HIF-P4Hs can also be inhibited by small molecules studied in clinical trials for renal anemia. Knowledge of systemic long-term inactivation of HIF-P4Hs is limited but crucial, since HIF overexpression is associated with cancers. We aimed to determine the effects of systemic genetic inhibition of the most abundant isoenzyme HIF prolyl 4-hydroxylase-2 (HIF-P4H-2)/PHD2/EglN1 on life span and tissue homeostasis in aged mice. Our data showed no difference between wild-type and HIF-P4H-2-deficient mice in the average age reached. There were several differences, however, in the primary causes of death and comorbidities, the HIF-P4H-2-deficient mice having less inflammation, liver diseases, including cancer, and myocardial infarctions, and not developing anemia. No increased cancer incidence was observed due to HIF-P4H-2-deficiency. These data suggest that chronic inactivation of HIF-P4H-2 is not harmful but rather improves the quality of life in senescence.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Inflammation/prevention & control , Kidney Diseases/prevention & control , Liver Diseases/prevention & control , Liver Neoplasms, Experimental/prevention & control , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Female , Inflammation/etiology , Inflammation/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Liver Diseases/etiology , Liver Diseases/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/pathology , Longevity , Male , Mice , Mice, Knockout
7.
Hepatology ; 72(4): 1430-1443, 2020 10.
Article in English | MEDLINE | ID: mdl-31965581

ABSTRACT

BACKGROUND AND AIMS: Activation of MYC and catenin beta-1 (CTNNB1, encoding ß-catenin) can co-occur in liver cancer, but how these oncogenes cooperate in tumorigenesis remains unclear. APPROACH AND RESULTS: We generated a mouse model allowing conditional activation of MYC and WNT/ß-catenin signaling (through either ß-catenin activation or loss of APC - adenomatous polyposis coli) upon expression of CRE recombinase in the liver and monitored their effects on hepatocyte proliferation, apoptosis, gene expression profiles, and tumorigenesis. Activation of WNT/ß-catenin signaling strongly accelerated MYC-driven carcinogenesis in the liver. Both pathways also cooperated in promoting cellular transformation in vitro, demonstrating their cell-autonomous action. Short-term induction of MYC and ß-catenin in hepatocytes, followed by RNA-sequencing profiling, allowed the identification of a "Myc/ß-catenin signature," composed of a discrete set of Myc-activated genes whose expression increased in the presence of active ß-catenin. Notably, this signature enriched for targets of Yes-associated protein (Yap) and transcriptional coactivator with PDZ-binding motif (Taz), two transcriptional coactivators known to be activated by WNT/ß-catenin signaling and to cooperate with MYC in mitogenic activation and liver transformation. Consistent with these regulatory connections, Yap/Taz accumulated upon Myc/ß-catenin activation and were required not only for the ensuing proliferative response, but also for tumor cell growth and survival. Finally, the Myc/ß-catenin signature was enriched in a subset of human hepatocellular carcinomas characterized by comparatively poor prognosis. CONCLUSIONS: Myc and ß-catenin show a strong cooperative action in liver carcinogenesis, with Yap and Taz serving as mediators of this effect. These findings warrant efforts toward therapeutic targeting of Yap/Taz in aggressive liver tumors marked by elevated Myc/ß-catenin activity.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Cycle Proteins/physiology , Liver Neoplasms, Experimental/etiology , Proto-Oncogene Proteins c-myc/physiology , Trans-Activators/physiology , beta Catenin/physiology , Animals , Mice , Mice, Inbred C57BL , Wnt Signaling Pathway/physiology , YAP-Signaling Proteins
8.
Gut ; 69(2): 365-379, 2020 02.
Article in English | MEDLINE | ID: mdl-31076403

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC), mostly developed in fibrotic/cirrhotic liver, exhibits relatively low responsiveness to immune checkpoint blockade (ICB) therapy. As myeloid-derived suppressor cell (MDSC) is pivotal for immunosuppression, we investigated its role and regulation in the fibrotic microenvironment with an aim of developing mechanism-based combination immunotherapy. DESIGN: Functional significance of MDSCs was evaluated by flow cytometry using two orthotopic HCC models in fibrotic liver setting via carbon tetrachloride or high-fat high-carbohydrate diet and verified by clinical specimens. Mechanistic studies were conducted in human hepatic stellate cell (HSC)-peripheral blood mononuclear cell culture systems and fibrotic-HCC patient-derived MDSCs. The efficacy of single or combined therapy with anti-programmed death-1-ligand-1 (anti-PD-L1) and a clinically trialled BET bromodomain inhibitor i-BET762 was determined. RESULTS: Accumulation of monocytic MDSCs (M-MDSCs), but not polymorphonuclear MDSCs, in fibrotic livers significantly correlated with reduced tumour-infiltrating lymphocytes (TILs) and increased tumorigenicity in both mouse models. In human HCCs, the tumour-surrounding fibrotic livers were markedly enriched with M-MDSC, with its surrogate marker CD33 significantly associated with aggressive tumour phenotypes and poor survival rates. Mechanistically, activated HSCs induced monocyte-intrinsic p38 MAPK signalling to trigger enhancer reprogramming for M-MDSC development and immunosuppression. Treatment with p38 MAPK inhibitor abrogated HSC-M-MDSC crosstalk to prevent HCC growth. Concomitant with patient-derived M-MDSC suppression by i-BET762, combined treatment with anti-PD-L1 synergistically enhanced TILs, resulting in tumour eradication and prolonged survival in the fibrotic-HCC mouse model. CONCLUSION: Our results signify how non-tumour-intrinsic properties in the desmoplastic microenvironment can be exploited to reinstate immunosurveillance, providing readily translatable combination strategies to empower HCC immunotherapy.


Subject(s)
Carcinoma, Hepatocellular/therapy , Immunotherapy/methods , Liver Neoplasms/therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/immunology , Cellular Reprogramming/immunology , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Hepatic Stellate Cells/immunology , Humans , Immune Tolerance , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Liver Neoplasms/etiology , Liver Neoplasms/immunology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/therapy , Male , Mice, Inbred C57BL , Monocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Signal Transduction/physiology , Tumor Cells, Cultured , Tumor Microenvironment , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/physiology
9.
Cell Death Differ ; 27(1): 85-101, 2020 01.
Article in English | MEDLINE | ID: mdl-31065105

ABSTRACT

Hepatocellular carcinoma (HCC) generally occurs in the presence of chronic liver injury, often as a sequela of liver fibrosis. Hepatic progenitor cells (HPCs) are known to be capable of forming both hepatocytes and cholangiocytes in chronic liver injury, which are also considered a source of myofibroblasts and tumor-initiating cells, under carcinogenic circumstances. However, the underlying mechanisms that activate HPCs to give rise to HCC are still unclear. In current study, the correlation between HPCs activation and liver fibrosis and carcinogenesis was investigated in rats and human specimens. We analyzed the role of HPCs in tumorigenesis, by transplanting exogenous HPCs in a diethylnitrosamine-induced rat HCC model. Our data indicated that HPC activation correlated with hepatic fibrosis and hepatocarcinogenesis. We further found that exogenous HPC infusion promoted liver fibrosis and hepatocarcinogenesis, while lipopolysaccharides (LPS) played an important role in this process. However, results of our study indicated that LPS did not induce HPCs to form tumor in nude mice directly. Rather, LPS induced myofibroblast-like morphology in HPCs, which enhanced the tumorigenic potential of HPCs. Further experiments showed that LPS/Toll-like receptor 4 (TLR4) signaling mediated the differentiation of HPCs into myofibroblasts and enhanced the production of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), which led to the aberrant expression of Ras and p53 signaling pathways in HPCs, and finally, promoted the proliferation and malignant transformation of HPCs, by long non-coding RNA regulation. Besides, examination of HCC clinical samples demonstrated that IL-6 and TNF-α production correlated with HPC activation, hepatic fibrosis, and HCC recurrence. Our study indicates that both myofibroblasts and tumor cells are derived from HPCs. HPC-derived myofibroblasts create tumor microenvironment and contribute to the proliferation and malignant transformation of HPCs. Furthermore, LPS present in the chronic liver inflammation microenvironment might play an important role in hepatocarcinogenesis, by regulating the plastic potential of HPCs.


Subject(s)
Lipopolysaccharides/physiology , Liver Neoplasms, Experimental/etiology , Myofibroblasts/metabolism , Stem Cells/metabolism , Tumor Microenvironment , Adult , Aged , Animals , Carcinogenesis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Female , Humans , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/pathology , Male , Mice, Nude , Middle Aged , Myofibroblasts/cytology , RNA, Long Noncoding/metabolism , Rats, Inbred F344 , Signal Transduction , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects , Toll-Like Receptor 4/metabolism , Young Adult
10.
Life Sci ; 241: 117121, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31794772

ABSTRACT

AIMS: This study addressed whether endogenous hepatic stem/progenitor (HSP) cells survival were related to the injured signals during liver cirrhosis. MATERIAL AND METHODS: Liver cirrhosis was induced in C57BL/6 mice by administering diethylnitrosamine (DEN) in drinking water. Hematoxylin-eosin staining and Masson's trichrome staining were used to identify infiltrative cells and connective tissues, respectively. The inflammatory activity grade and fibrotic stage, represented as G and S respectively, and evaluated by the International Simplified Grading and Staging System (ISGSS). Endogenous HSP cells (Ng2+HSP) were identified by immunofluorescence staining with an anti-neuro/glial antigen 2 (Ng2) antibody. All data were analyzed using SPSS 22.0 and GraphPad Prism 6 and Student's t-test was performed to determine statistical significance. p-values < 0.05 were considered statistically significant. KEY FINDINGS: All mice administered oral DEN developed liver fibrosis, liver cirrhosis and hepatocellular carcinoma (HCC). During the fibrosis period, observed a positive correlation of survival of endogenous HSP (Ng2+HSP) cells with inflammatory activity. As the disease developed into the cirrhotic stage, when lost correlation of endogenous HSP cells with inflammatory activity, revealed a dramatically reduced survival rate of endogenous HSP (Ng2+HSP) cells. SIGNIFICANCE: The DEN-induced liver cirrhosis could develop into three time zone of fibrosis, cirrhosis and cancer, and the histological patterns in the model are similar to those described in humans. Dramatic survival and less apoptosis of endogenous HSP (Ng2+HSP) cells was within fibrotic state, where inflammation signals is strong, indicating such time zone (1-6 weeks) during liver cirrhosis is important for mobilizing endogenous HSP-based regeneration.


Subject(s)
Carcinoma, Hepatocellular/therapy , Inflammation/therapy , Liver Cirrhosis/therapy , Liver Neoplasms, Experimental/therapy , Liver/cytology , Stem Cells/cytology , Animals , Carcinoma, Hepatocellular/etiology , Diethylnitrosamine/toxicity , Disease Models, Animal , Inflammation/etiology , Liver/immunology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Neoplasms, Experimental/etiology , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Stem Cells/physiology
11.
J Nutr ; 150(4): 775-783, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31851339

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death in the world. Choline deficiency has been well studied in the context of liver disease; however, less is known about the effects of choline supplementation in HCC. OBJECTIVE: The objective of this study was to test whether choline supplementation could influence the progression of HCC in a high-fat-diet (HFD)-driven mouse model. METHODS: Four-day-old male C57BL/6J mice were treated with the chemical carcinogen, 7,12-dimethylbenz[a]anthracene, and were randomly assigned at weaning to a cohort fed an HFD (60% kcal fat) or an HFD with supplemental choline (60% kcal fat, 1.2% choline; HFD+C) for 30 wk. Blood was isolated at 15 and 30 wk to measure immune cells by flow cytometry, and glucose-tolerance tests were performed 2 wk prior to killing. Overall tumor burden was quantified, hepatic lipids were measured enzymatically, and phosphatidylcholine species were measured by targeted MS methods. Gene expression and mitochondrial DNA were quantified by quantitative PCR. RESULTS: HFD+C mice exhibited a 50-90% increase in both circulating choline and betaine concentrations in the fed state (P ≤ 0.05). Choline supplementation resulted in a 55% decrease in total tumor numbers, a 67% decrease in tumor surface area, and a 50% decrease in hepatic steatosis after 30 wk of diet (P ≤ 0.05). Choline supplementation increased the abundance of mitochondria and the relative expression of ß-oxidation genes by 21% and ∼75-100%, respectively, in the liver. HFD+C attenuated circulating myeloid-derived suppressor cells at 15 wk of feeding (P ≤ 0.05). CONCLUSIONS: Choline supplementation attenuated HFD-induced HCC and hepatic steatosis in male C57BL/6J mice. These results suggest a therapeutic benefit of choline supplementation in blunting HCC progression.


Subject(s)
Choline/administration & dosage , Diet, High-Fat/adverse effects , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms/prevention & control , Animals , Betaine/blood , Choline/blood , DNA, Mitochondrial/analysis , Dietary Supplements , Fatty Liver/prevention & control , Gene Expression/drug effects , Lipid Metabolism/genetics , Liver/chemistry , Liver/pathology , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/pathology , Organ Size/drug effects
12.
Sci Rep ; 9(1): 10338, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31316146

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a major risk factor for the development of hepatocellular carcinoma (HCC), a leading cause of cancer mortality worldwide. Hepatitis B X protein (HBx) and pre-S2 mutant have been proposed as the two most important HBV oncoproteins that play key roles in HCC pathogenesis. Curcumin is a botanical constituent displaying potent anti-inflammatory and anti-cancer properties without toxic side effects. Phytosomal formulation of curcumin has been shown to exhibit enhanced bioavailability, improved pharmacokinetics, and excellent efficacy against many human diseases. However, effectiveness of phytosomal curcumin for HCC treatment remains to be clarified. In this study, we evaluated chemopreventive effect of phytosomal curcumin on HBV-related HCC by using a transgenic mouse model specifically expressing both HBx and pre-S2 mutant in liver. Compared with unformulated curcumin, phytosomal curcumin exhibited significantly greater effects on suppression of HCC formation, improvement of liver histopathology, decrease of lipid accumulation and leukocyte infiltration, and reduction of total tumor volume in transgenic mice. Moreover, phytosomal curcumin exerted considerably stronger effects on activation of anti-inflammatory PPARγ as well as inhibition of pro-inflammatory NF-κB than unformulated curcumin. Furthermore, phytosomal curcumin showed a comparable effect on suppression of oncogenic mTOR activation to unformulated curcumin. Our data demonstrated that phytosomal curcumin has promise for HCC chemoprevention in patients with chronic HBV infection.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Curcumin/administration & dosage , Hepatitis B virus/pathogenicity , Liver Neoplasms, Experimental/prevention & control , Animals , Chemoprevention , Drug Compounding , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/complications , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/virology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Protein Precursors/genetics , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins/genetics
13.
Am J Transplant ; 19(12): 3250-3262, 2019 12.
Article in English | MEDLINE | ID: mdl-31162867

ABSTRACT

A recurrence of hepatocellular carcinoma (HCC) after living donor liver transplantation (LDLT) is one of the major concerns reflecting the higher mortality of HCC. This study aimed to explore the impact of circulating exosomes on HCC development and recurrence. One-shot transfusion of hepatoma serum to naïve rats induced liver cancer development with gradual elevation of alpha-fetoprotein (AFP), but exosome-free hepatoma serum failed to induce AFP elevation. The microarray analysis revealed miR-92b as one of the highly expressing microribonucleic acids in hepatoma serum exosomes. Overexpression of miR-92b enhanced the migration ability of liver cancer cell lines with active release of exosomal miR-92b. The hepatoma-derived exosomal miR-92b transferred to natural killer (NK) cells, resulting in the downregulation of CD69 and NK cell-mediated cytotoxicity. Furthermore, higher expression of miR-92b in serum exosomes was confirmed in HCC patients before LDLT, and its value at 1 month after LDLT was maintained at a higher level in the patients with posttransplant HCC recurrence. In summary, we demonstrated the impact of circulating exosomes on liver cancer development, partly through the suppression of CD69 on NK cells by hepatoma-derived exosomal miR-92b. The value of circulating exosomal miR-92b may predict the risk of posttransplant HCC recurrence.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/diagnosis , Liver Transplantation/adverse effects , MicroRNAs/genetics , Neoplasm Recurrence, Local/diagnosis , Postoperative Complications/diagnosis , Animals , Carcinoma, Hepatocellular/etiology , Cell Proliferation , Exosomes , Female , Gene Expression Regulation, Neoplastic , Liver Neoplasms, Experimental/diagnosis , Liver Neoplasms, Experimental/etiology , Male , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local/etiology , Postoperative Complications/etiology , Rats
14.
Transplant Proc ; 51(3): 925-935, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30979485

ABSTRACT

OBJECTIVES: Liver transplantation remains the only curative therapy for decompensated liver cirrhosis. However, it has several limitations, and not all patients can receive liver transplants. Therefore, liver regenerative therapy without liver transplantation is considered necessary. In this study, we attempted minimally invasive liver regenerative therapy by peripheral vein infusion of bone marrow-derived mesenchymal stem cells (BMSCs) cultured from a small amount of autologous bone marrow fluid and evaluated the effects of BMSCs on hepatocarcinogenesis in a mouse model. METHODS: C57BL/6 male mice were injected intraperitoneally with N-nitrosodiethylamine once at 2 weeks of age, followed by carbon tetrachloride twice a week from 6 weeks of age onwards, to create a mouse model of highly oncogenic liver cirrhosis. From 10 weeks of age, mouse isogenic green fluorescent protein-positive BMSCs (1.0 × 106/body weight) were infused once every 2 weeks, for a total of 5 times, and the effects of frequent BMSC infusion on hepatocarcinogenesis were evaluated. RESULTS: In the histologic evaluation, no significant differences were observed between the controls and BMSC-administered mice in terms of incidence rate, number, or average size of foci and tumors. However, significant suppression of fibrosis and liver injury was confirmed in the group that received BMSC infusions. DISCUSSION: Considering that BMSC infusion did not promote carcinogenesis, even in the state of highly oncogenic liver cirrhosis, autologous BMSC infusion might be a safe and effective therapy for human decompensated liver cirrhosis.


Subject(s)
Bone Marrow Transplantation/methods , Liver Cirrhosis/surgery , Liver Neoplasms, Experimental/prevention & control , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Animals , Carcinogenesis , Cells, Cultured , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred C57BL
15.
FASEB J ; 33(7): 7995-8007, 2019 07.
Article in English | MEDLINE | ID: mdl-30897344

ABSTRACT

Chronic liver inflammation (CLI) is a risk factor for development of hepatocellular carcinoma (HCC). Galectin-1 (Gal1) is involved in the regulation of inflammation, angiogenesis, and tumorigenesis, exhibiting multiple anti-inflammatory and protumorigenic activities. We aimed to explore its regulatory role in CLI and HCC progression using an established model of CLI-mediated HCC development, Abcb4 [multidrug-resistance 2 (Mdr2)]-knockout (KO) mice, which express high levels of Gal1 in the liver. We generated double-KO (dKO) Gal1-KO/Mdr2-KO mice on C57BL/6 and FVB/N genetic backgrounds and compared HCC development in the generated strains with their parental Mdr2-KO strains. Loss of Gal1 increased liver injury, inflammation, fibrosis, and ductular reaction in dKO mice of both strains starting from an early age. Aged dKO mutants displayed earlier hepatocarcinogenesis and increased tumor size compared with control Mdr2-KO mice. We found that osteopontin, a well-known modulator of HCC development, and oncogenic proteins Ntrk2 (TrkB) and S100A4 were overexpressed in dKO compared with Mdr2-KO livers. Our results demonstrate that in Mdr2-KO mice, a model of CLI-mediated HCC, Gal1-mediated protection from hepatitis, liver fibrosis, and HCC initiation dominates over its known procarcinogenic activities at later stages of HCC development. These findings suggest that anti-Gal1 treatments may not be applicable at all stages of CLI-mediated HCC.-Potikha, T., Pappo, O., Mizrahi, L., Olam, D., Maller, S. M., Rabinovich, G. A., Galun, E., Goldenberg, D. S. Lack of galectin-1 exacerbates chronic hepatitis, liver fibrosis, and carcinogenesis in murine hepatocellular carcinoma model.


Subject(s)
Galectin 1/physiology , Hepatitis/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Precancerous Conditions/metabolism , Alternative Splicing , Animals , Cell Division , Chronic Disease , Cocarcinogenesis , Female , Galectin 1/deficiency , Galectin 1/genetics , Hep G2 Cells , Hepatitis/genetics , Hepatitis/pathology , Hepatocytes/pathology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Neoplasm Proteins/genetics , Osteopontin/biosynthesis , Osteopontin/deficiency , Osteopontin/genetics , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Specific Pathogen-Free Organisms
16.
Chem Biol Interact ; 303: 70-78, 2019 Apr 25.
Article in English | MEDLINE | ID: mdl-30826251

ABSTRACT

Obesity has been implicated in the genesis of metabolic syndromes including insulin resistance and Type 2 Diabetes Mellitus (T2DM). Given the association between T2DM and the risk of hepatocellular carcinoma (HCC), our specific goal was to determine whether the liver of HFD-induced T2DM mice is more sensitive to the carcinogen diethylnitrosamine (DEN), due to a modification of the molecular pathways implicated in the early stages of HCC pathogenesis. C57BL/6 male mice (five-week-old) were divided into 4 groups: C, C + DEN, HFD and HFD + DEN. Mice were euthanized twenty-five weeks after DEN-injection. Livers of HDF-fed mice showed a higher proliferative index than Control groups. In line with this, HFD groups showed an increase of nuclear ß-catenin, and interestingly, DEN treatment led to a slight increase in the expression of this protein in HFD group. Based on these results, and to confirm this effect, we analyzed ß-catenin target genes, finding that DEN treatment in HFD group led to a significant increase of Vegf, c-myc, c-jun and cyclin D1 expression levels. According to our results, the expression of TCF4 showed to be significantly increased in HFD + DEN vs. HFD. In this regard, the ß-catenin/TCF4 complex enhanced its association with pSmads 2/3, as we observed an increase of nuclear Smads expression in HFD + DEN, suggesting a possible role of TGF-ß1/Smads signaling pathway in this phenomenon. Our results show that the liver of HFD fed model that resembles early T2DM pathology in mice, is more sensitive to DEN, by inducing both Wnt/ß-catenin and TGF ß1/Smads tumorigenic pathways.


Subject(s)
Carcinogenesis/genetics , Diet, High-Fat/adverse effects , Diethylnitrosamine/adverse effects , Liver Neoplasms, Experimental/etiology , Alkylating Agents/adverse effects , Animals , Carcinogenesis/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Wnt Signaling Pathway/drug effects
17.
Hepatology ; 70(6): 2003-2017, 2019 12.
Article in English | MEDLINE | ID: mdl-30737831

ABSTRACT

Inactivating mutations of axis inhibition protein 1 (AXIN1), a negative regulator of the Wnt/ß-Catenin cascade, are among the common genetic events in human hepatocellular carcinoma (HCC), affecting approximately 10% of cases. In the present manuscript, we sought to define the genetic crosstalk between Axin1 mutants and Wnt/ß-catenin as well as Notch signaling cascades along hepatocarcinogenesis. We discovered that c-MET activation and AXIN1 mutations occur concomitantly in ~3%-5% of human HCC samples. Subsequently, we generated a murine HCC model by means of CRISPR/Cas9-based gene deletion of Axin1 (sgAxin1) in combination with transposon-based expression of c-Met in the mouse liver (c-Met/sgAxin1). Global gene expression analysis of mouse normal liver, HCCs induced by c-Met/sgAxin1, and HCCs induced by c-Met/∆N90-ß-Catenin revealed activation of the Wnt/ß-Catenin and Notch signaling in c-Met/sgAxin1 HCCs. However, only a few of the canonical Wnt/ß-Catenin target genes were induced in c-Met/sgAxin1 HCC when compared with corresponding lesions from c-Met/∆N90-ß-Catenin mice. To study whether endogenous ß-Catenin is required for c-Met/sgAxin1-driven HCC development, we expressed c-Met/sgAxin1 in liver-specific Ctnnb1 null mice, which completely prevented HCC development. Consistently, in AXIN1 mutant or null human HCC cell lines, silencing of ß-Catenin strongly inhibited cell proliferation. In striking contrast, blocking the Notch cascade through expression of either the dominant negative form of the recombinant signal-binding protein for immunoglobulin kappa J region (RBP-J) or the ablation of Notch2 did not significantly affect c-Met/sgAxin1-driven hepatocarcinogenesis. Conclusion: We demonstrated here that loss of Axin1 cooperates with c-Met to induce HCC in mice, in a ß-Catenin signaling-dependent but Notch cascade-independent way.


Subject(s)
Axin Protein/physiology , Carcinoma, Hepatocellular/etiology , Liver Neoplasms, Experimental/etiology , Receptors, Notch/physiology , beta Catenin/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-met/physiology , Wnt Signaling Pathway/physiology
18.
Hepatology ; 69(4): 1549-1563, 2019 04.
Article in English | MEDLINE | ID: mdl-30014520

ABSTRACT

Functions of transforming growth factor-ß (TGF-ß) in the liver vary depending on specific cell types and their temporal response to TGF-ß during different stages of hepatocarcinogenesis (HCG). Through analysis of tumor tissues from hepatocellular carcinoma (HCC) patients, we were able to cluster hepatic epithelial cell-derived TGF-ß gene signatures in association with distinct clinical prognoses. To delineate the role of hepatic epithelial TGF-ß signaling in HCC development, we used an experimental system in which tumor-initiating hepatocytes (TICs) were isolated from TGF-ß receptor II floxed mice (Tgfbr2fl/fl ) and transplanted into syngeneic C57BL/6J mice by splenic injection. Recipient mice were then administered Cre-expressing adenovirus (Ad-Cre) to inactivate Tgfbr2 in transplanted TICs. After latency, Tgfbr2-inactivated TICs formed larger and more tumor nodules in recipient livers compared to TICs without Tgfbr2 inactivation. In vitro analyses revealed that treatment of cultured TICs with TGF-ß inhibited expression of progenitor cell factors (including SRY (sex determining region Y)-box 2 [Sox2]). RNA sequencing (RNA-seq) analysis identified H19 as one of the most up-regulated long noncoding RNA (lncRNA) in association with Tgfbr2 inactivation in TICs. Tgfbr2 inactivation by Ad-Cre led to a 5-fold increase of H19 expression in TICs. Accordingly, TGF-ß treatment reduced H19 expression. We observed that forced overexpression of Sox2 in TICs increased transcription of H19, whereas knockdown of Sox2 decreased it. Furthermore, depletion of H19 reduced the progenitor property of TICs in vitro and decreased their tumorigenic potential in vivo. Finally, we observed a low level of H19 mRNA expression in human HCC tissues from patients with the epithelial TGF-ß gene signature in association with favorable prognosis. Conclusion: Our findings describe a TGF-ß and H19 signaling axis by Sox2 in TICs that importantly regulates HCG.


Subject(s)
Liver Neoplasms, Experimental/etiology , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Animals , Female , Hepatocytes/metabolism , Humans , Liver Neoplasms, Experimental/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Receptor, Transforming Growth Factor-beta Type II/metabolism , SOXB1 Transcription Factors/metabolism , Young Adult
19.
Hepatology ; 69(4): 1735-1750, 2019 04.
Article in English | MEDLINE | ID: mdl-30329167

ABSTRACT

Hepatitis B virus (HBV) is a major risk factor for development of hepatocellular carcinoma (HCC), at least partially due to dysfunctional anti-HBV adaptive immunity; however, the role of innate immune response to HBV in this process is not well understood. In this study, low-dose polyinosinic:polycytidylic acid (poly [I:C]), a natural killer (NK) cell activator (3 µg/g body weight, twice/week for 8 weeks), induced HCC in HBV transgenic (HBs-Tg) mice, with an incidence of 100% after 6 months, while HBs-Tg mice without treatment only had HCC with an incidence of 16.7%. In HBs-Tg mice, poly (I:C) induced liver inflammation with markedly increased infiltrating lymphocytes, along with the concurrently increased apoptosis and proliferation of hepatocytes, leading to the accelerated epithelial-to-mesenchymal transition (EMT) of hepatocytes shown by increased expression of the typical transcriptional factors (Slug, Twist, and mothers against decapentaplegic-interacting protein 1) and phenotypic proteins (vimentin and chemokine [C-X-C motif] receptor 4). The EMT and tumorigenesis in this model depended on the presence of NK cells because depletion of these cells significantly reduced the HCC rate to 28.6%. Further, intrahepatic NK cells highly expressed interferon-gamma (IFN-γ), anti-IFN-γ neutralizing monoclonal antibody might obviously alleviate the hepatitis, and hepatocyte-specific IFN-γ overexpression promoted HCC. Moreover, IFN-γ deficiency in HBs-Tg mice prevented HCC occurring, though hepatic NK cells existed and could be activated, suggesting the critical role of IFN-γ in NK cell-mediated tumorigenesis. In an in vitro experiment, IFN-γ up-regulated epithelial cell adhesion molecule (EpCAM) expression through phosphorylated signal transducer and activator of transcription (p-STAT1) pathway, which was followed by EMT, and p-STAT1 inhibitor might absolutely abolish the expression of EpCAM and EMT in HBV surface antigen-positive hepatocytes. Conclusion: This work demonstrates that NK cell-derived IFN-γ promotes HCC through the EpCAM-EMT axis in HBs-Tg mice, revealing the importance of innate immunity in pathogenesis of HBV-associated HCC.


Subject(s)
Carcinoma, Hepatocellular/etiology , Epithelial-Mesenchymal Transition , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Liver Neoplasms, Experimental/etiology , Animals , Epithelial Cell Adhesion Molecule/metabolism , Hepatitis B/complications , Hepatitis B/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C , STAT1 Transcription Factor/metabolism
20.
Hepatology ; 69(2): 604-621, 2019 02.
Article in English | MEDLINE | ID: mdl-30053328

ABSTRACT

Mitochondria lie at the heart of innate immunity, and aberrant mitochondrial activity contributes to immune activation and chronic inflammatory diseases, including liver cancers. Mitophagy is a selective process for removing dysfunctional mitochondria. The link between mitophagy and inflammation in tumorigenesis remains largely unexplored. We observed that FUN14 domain-containing 1 (FUNDC1), a previously characterized mitophagy receptor, accumulates in most human hepatocellular carcinomas (HCCs), and we thus explored the role of FUNDC1-mediated mitophagy in HCC initiation and progression in a mouse model in which HCC is induced by the chemical carcinogen, diethylnitrosamine (DEN). We showed that specific knockout of FUNDC1 in hepatocytes promotes the initiation and progression of DEN-induced HCC, whereas FUNDC1 transgenic hepatocytes protect against development of HCC. Hepatocyte-specific FUNDC1 ablation results in the accumulation of dysfunctional mitochondria and triggers a cascade of events involving inflammasome activation and hyperactivation of Janus kinase/signal transducer and activator of transcription signaling. Specifically, cytosolic mitochondrial DNA (mtDNA) release and caspase-1 activation are increased in FUNDC1-depleted hepatocytes. This subsequently results in the elevated release of proinflammatory cytokines, such as interleukin-1ß (IL1ß) and hyperproliferation of hepatocytes. Conclusion: Our results suggest that FUNDC1 suppresses HCC initiation by reducing inflammasome activation and inflammatory responses in hepatocytes, whereas up-regulation of FUNDC1 expression at the late stage of tumor development may benefit tumor growth. Our study thus describes a mechanistic link between mitophagic modulation of inflammatory response and tumorigenesis, and further implies that FUNDC1-mediated mitophagy and its related inflammatory response may represent a therapeutic target for liver cancer.


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms, Experimental/etiology , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Animals , Carcinoma, Hepatocellular/metabolism , Caspase 1/metabolism , Diethylnitrosamine , Hepatocytes/metabolism , Humans , Inflammasomes/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Neoplasms, Experimental/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...