Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nature ; 610(7931): 356-365, 2022 10.
Article in English | MEDLINE | ID: mdl-36198802

ABSTRACT

Hepatocellular carcinoma (HCC), the fourth leading cause of cancer mortality worldwide, develops almost exclusively in patients with chronic liver disease and advanced fibrosis1,2. Here we interrogated functions of hepatic stellate cells (HSCs), the main source of liver fibroblasts3, during hepatocarcinogenesis. Genetic depletion, activation or inhibition of HSCs in mouse models of HCC revealed their overall tumour-promoting role. HSCs were enriched in the preneoplastic environment, where they closely interacted with hepatocytes and modulated hepatocarcinogenesis by regulating hepatocyte proliferation and death. Analyses of mouse and human HSC subpopulations by single-cell RNA sequencing together with genetic ablation of subpopulation-enriched mediators revealed dual functions of HSCs in hepatocarcinogenesis. Hepatocyte growth factor, enriched in quiescent and cytokine-producing HSCs, protected against hepatocyte death and HCC development. By contrast, type I collagen, enriched in activated myofibroblastic HSCs, promoted proliferation and tumour development through increased stiffness and TAZ activation in pretumoural hepatocytes and through activation of discoidin domain receptor 1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.


Subject(s)
Carcinogenesis , Carcinoma, Hepatocellular , Hepatic Stellate Cells , Liver Neoplasms , Animals , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Collagen Type I/metabolism , Discoidin Domain Receptor 1/metabolism , Disease Progression , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocyte Growth Factor/metabolism , Hepatocytes , Humans , Liver Cirrhosis/complications , Liver Neoplasms/pathology , Mice , Myofibroblasts/pathology
2.
Cell Rep ; 39(12): 110987, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732131

ABSTRACT

The liver is one of the most-favored distant metastatic sites for solid tumors, and interactions between cancer cells and components of the hepatic microenvironment are essential for liver metastasis (LM). Although sex is one of the determinants for primary liver cancer, sexual dimorphism in LM (SDLM) and the underlying mechanisms remain unclear. We herein demonstrate a significant male-biased SDLM, which is attributed to host androgen/androgen receptor (Ar) signaling that promotes hepatic seeding of tumor cells and subsequent outgrowth in a neutrophil-dependent manner. Mechanistically, androgen/Ar signaling promotes hepatic accumulation of neutrophils by promoting proliferation and development of neutrophil precursors in the bone marrow, as well as modulating hepatic recruitment of neutrophils and their functions. Antagonizing the androgen/Ar/neutrophil axis significantly mitigates LM in males. Our data thus reveal an important role of androgen in LM and suggest that androgen/Ar modulation represents a promising target for LM therapy in men.


Subject(s)
Androgens , Liver Neoplasms , Neutrophils , Sex Characteristics , Humans , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Male , Neutrophils/cytology , Receptors, Androgen , Tumor Microenvironment
4.
Clin Transl Med ; 11(4): e379, 2021 04.
Article in English | MEDLINE | ID: mdl-33931972

ABSTRACT

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.


Subject(s)
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
5.
Cancer Cell ; 39(6): 866-882.e11, 2021 06 14.
Article in English | MEDLINE | ID: mdl-33930309

ABSTRACT

Cancer-associated fibroblasts (CAF) are a poorly characterized cell population in the context of liver cancer. Our study investigates CAF functions in intrahepatic cholangiocarcinoma (ICC), a highly desmoplastic liver tumor. Genetic tracing, single-cell RNA sequencing, and ligand-receptor analyses uncovered hepatic stellate cells (HSC) as the main source of CAF and HSC-derived CAF as the dominant population interacting with tumor cells. In mice, CAF promotes ICC progression, as revealed by HSC-selective CAF depletion. In patients, a high panCAF signature is associated with decreased survival and increased recurrence. Single-cell RNA sequencing segregates CAF into inflammatory and growth factor-enriched (iCAF) and myofibroblastic (myCAF) subpopulations, displaying distinct ligand-receptor interactions. myCAF-expressed hyaluronan synthase 2, but not type I collagen, promotes ICC. iCAF-expressed hepatocyte growth factor enhances ICC growth via tumor-expressed MET, thus directly linking CAF to tumor cells. In summary, our data demonstrate promotion of desmoplastic ICC growth by therapeutically targetable CAF subtype-specific mediators, but not by type I collagen.


Subject(s)
Bile Duct Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Cholangiocarcinoma/pathology , Aged , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/pathology , Cancer-Associated Fibroblasts/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Collagen Type I/metabolism , Female , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/pathology , Hepatocyte Growth Factor/metabolism , Humans , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Hyaluronic Acid/metabolism , Male , Mice, Transgenic , Middle Aged , Proto-Oncogene Proteins c-met/metabolism , Tumor Microenvironment
6.
Front Immunol ; 12: 625472, 2021.
Article in English | MEDLINE | ID: mdl-33763069

ABSTRACT

Neutrophils are the largest population of circulating leukocytes and the first responder against invading pathogens or other danger signals. Sophisticated machineries help them play critical roles in immunity and inflammation, including phagocytosis, superoxide production, cytokine and chemokine production, degranulation, and formation of neutrophil extracellular traps (NETs). After maturation and release from the bone marrow, neutrophils migrate to inflamed tissues in response to many stimuli. Increasing evidences indicate that neutrophils are critically involved in the pathogenesis of liver diseases, including liver cancer, thus making them promising target for the treatment of liver diseases. Here, we would like to provide the latest finding about the role of neutrophils in liver diseases and discuss the potentiality of neutrophils as target for liver diseases.


Subject(s)
Liver Diseases/immunology , Liver/immunology , Neutrophil Activation , Neutrophil Infiltration , Neutrophils/immunology , Animals , Chronic Disease , Cytokines/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
7.
Acta Pharmacol Sin ; 42(2): 282-289, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32555508

ABSTRACT

Identification of the functional impact of mutated and altered genes in cancer is critical for implementing precision oncology and drug repurposing. In recent years, the emergence of multiomics data from large, well-characterized patient cohorts has provided us with an unprecedented opportunity to address this problem. In this study, we investigated survival-associated genes across 26 cancer types and found that these genes tended to be hub genes and had higher K-core values in biological networks. Moreover, the genes associated with adverse outcomes were mainly enriched in pathways related to genetic information processing and cellular processes, while the genes with favorable outcomes were enriched in metabolism and immune regulation pathways. We proposed using the number of survival-related neighbors to assess the impact of mutations. In addition, by integrating other databases including the Human Protein Atlas and the DrugBank database, we predicted novel targets and anticancer drugs using the drug repurposing strategy. Our results illustrated the significance of multidimensional analysis of clinical data in important gene identification and drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Repositioning/methods , Neoplasms/genetics , Databases, Factual , Drug Development/methods , Humans , Molecular Targeted Therapy , Mutation , Neoplasms/pathology , Survival
8.
NPJ Precis Oncol ; 2(1): 6, 2018.
Article in English | MEDLINE | ID: mdl-29872724

ABSTRACT

Hepatocellular carcinoma (HCC) has become a leading cause of cancer-related death, making the elucidation of its underlying mechanisms an urgent priority. Inflammation is an adaptive response to infection and tissue injury under strict regulations. When the host regulatory machine runs out of control, nonresolving inflammation occurs. Nonresolving inflammation is a recognized hallmark of cancer that substantially contributes to the development and progression of HCC. The HCC-associated inflammation can be initiated and propagated by extrinsic pathways through activation of pattern-recognition receptors (PRRs) by pathogen-associated molecule patterns (PAMPs) derived from gut microflora or damage-associated molecule patterns (DAMPs) released from dying liver cells. The inflammation can also be orchestrated by the tumor itself through secreting factors that recruit inflammatory cells to the tumor favoring the buildup of a microenvironment. Accumulating datas from human and mouse models showed that inflammation promotes HCC development by promoting proliferative and survival signaling, inducing angiogenesis, evading immune surveillance, supporting cancer stem cells, activating invasion and metastasis as well as inducing genomic instability. Targeting inflammation may represent a promising avenue for the HCC treatment. Some inhibitors targeting inflammatory pathways have been developed and under different stages of clinical trials, and one (sorafenib) have been approved by FDA. However, as most of the data were obtained from animal models, and there is a big difference between human HCC and mouse HCC models, it is challenging on successful translation from bench to bedside.

9.
Nat Commun ; 9(1): 191, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335551

ABSTRACT

The communication between tumor-derived elements and stroma in the metastatic niche has a critical role in facilitating cancer metastasis. Yet, the mechanisms tumor cells use to control metastatic niche formation are not fully understood. Here we report that in the lung metastatic niche, high-metastatic hepatocellular carcinoma (HCC) cells exhibit a greater capacity to convert normal fibroblasts to cancer-associated fibroblasts (CAFs) than low-metastatic HCC cells. We show high-metastatic HCC cells secrete exosomal miR-1247-3p that directly targets B4GALT3, leading to activation of ß1-integrin-NF-κB signaling in fibroblasts. Activated CAFs further promote cancer progression by secreting pro-inflammatory cytokines, including IL-6 and IL-8. Clinical data show high serum exosomal miR-1247-3p levels correlate with lung metastasis in HCC patients. These results demonstrate intercellular crosstalk between tumor cells and fibroblasts is mediated by tumor-derived exosomes that control lung metastasis of HCC, providing potential targets for prevention and treatment of cancer metastasis.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Hepatocellular/metabolism , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , MicroRNAs/genetics , N-Acetyllactosamine Synthase/genetics , Animals , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Cell Communication , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Exosomes/chemistry , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , MicroRNAs/metabolism , N-Acetyllactosamine Synthase/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Signal Transduction
10.
Hepatology ; 67(6): 2414-2429, 2018 06.
Article in English | MEDLINE | ID: mdl-29091291

ABSTRACT

Fibrosis and cancer represent two major complications of chronic liver disease. MicroRNAs have been implicated in the development of fibrosis and cancer, thus constituting potential therapeutic targets. Here, we investigated the role of microRNA-21 (miR-21), a microRNA that has been implicated in the development of fibrosis in multiple organs and has also been suggested to act as an "oncomir." Accordingly, miR-21 was the microRNA that showed the strongest up-regulation in activated hepatic stellate cells (HSCs) in multiple models of fibrogenesis, with an 8-fold to 24-fold induction compared to quiescent HSCs. However, miR-21 antisense inhibition did not suppress the activation of murine or human HSCs in culture or in liver slices. Moreover, genetic deletion of miR-21 in two independently generated knockout mice or miR-21 antisense inhibition did not alter HSC activation or liver fibrosis in models of toxic and biliary liver injury. Despite a strong up-regulation of miR-21 in injury-associated hepatocellular carcinoma and in cholangiocarcinoma, miR-21 deletion or antisense inhibition did not reduce the development of liver tumors. As inhibition of the most up-regulated microRNA did not affect HSC activation, liver fibrosis, or fibrosis-associated liver cancer, we additionally tested the role of microRNAs in HSCs by HSC-specific Dicer deletion. Although Dicer deletion decreased microRNA expression in HSCs and altered the expression of select genes, it only exerted negligible effects on HSC activation and liver fibrosis. CONCLUSION: Genetic and pharmacologic manipulation of miR-21 does not inhibit the development of liver fibrosis and liver cancer. Moreover, suppression of microRNA synthesis does not significantly affect HSC phenotype and activation. (Hepatology 2018;67:2414-2429).


Subject(s)
DEAD-box RNA Helicases/physiology , Hepatic Stellate Cells/physiology , Liver Cirrhosis/etiology , MicroRNAs/physiology , Ribonuclease III/physiology , Animals , Female , Humans , Male , Mice, Knockout
11.
Nat Rev Gastroenterol Hepatol ; 14(9): 527-539, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28676707

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of worldwide cancer mortality. HCC almost exclusively develops in patients with chronic liver disease, driven by a vicious cycle of liver injury, inflammation and regeneration that typically spans decades. Increasing evidence points towards a key role of the bacterial microbiome in promoting the progression of liver disease and the development of HCC. Here, we will review mechanisms by which the gut microbiota promotes hepatocarcinogenesis, focusing on the leaky gut, bacterial dysbiosis, microbe-associated molecular patterns and bacterial metabolites as key pathways that drive cancer-promoting liver inflammation, fibrosis and genotoxicity. On the basis of accumulating evidence from preclinical studies, we propose the intestinal-microbiota-liver axis as a promising target for the simultaneous prevention of chronic liver disease progression and HCC development in patients with advanced liver disease. We will review in detail therapeutic modalities and discuss clinical settings in which targeting the gut-microbiota-liver axis for the prevention of disease progression and HCC development seems promising.


Subject(s)
Carcinoma, Hepatocellular/microbiology , Gastrointestinal Microbiome , Liver Neoplasms/microbiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Humans , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control
12.
Annu Rev Pathol ; 12: 153-186, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27959632

ABSTRACT

Liver cancer is the second leading cause of cancer mortality worldwide, causing more than 700,000 deaths annually. Because of the wide landscape of genomic alterations and limited therapeutic success of targeting tumor cells, a recent focus has been on better understanding and possibly targeting the microenvironment in which liver tumors develop. A unique feature of liver cancer is its close association with liver fibrosis. More than 80% of hepatocellular carcinomas (HCCs) develop in fibrotic or cirrhotic livers, suggesting an important role of liver fibrosis in the premalignant environment (PME) of the liver. Cholangiocarcinoma (CCA), in contrast, is characterized by a strong desmoplasia that typically occurs in response to the tumor, suggesting a key role of cancer-associated fibroblasts (CAFs) and fibrosis in its tumor microenvironment (TME). Here, we discuss the functional contributions of myofibroblasts, CAFs, and fibrosis to the development of HCC and CCA in the hepatic PME and TME, focusing on myofibroblast- and extracellular matrix-associated growth factors, fibrosis-associated immunosuppressive pathways, as well as mechanosensitive signaling cascades that are activated by increased tissue stiffness. Better understanding of the role of myofibroblasts in HCC and CCA development and progression may provide the basis to target these cells for tumor prevention or therapy.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Myofibroblasts/pathology , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/metabolism , Humans , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Myofibroblasts/metabolism , Signal Transduction
13.
Oncoimmunology ; 5(9): e1183850, 2016.
Article in English | MEDLINE | ID: mdl-27757296

ABSTRACT

Despite their central function in tumor immunity, dendritic cells (DCs) can respond to inhibitory signals and become tolerogenic, curtailing T cell responses in vivo. Here, we provide the evidence for an inhibitory function of signal regulatory protein (SIRP) α in DC survival and activation. In tumors from human liver cancer patients, infiltrative DCs expressed elevated levels of SIRPα, which is correlated with the induction of immune tolerance within the tumors. Silencing of SIRPα resulted in a significant increase in the longevity of antigen-pulsed DCs in the draining lymph nodes. In addition, SIRPα controls the activation and output of DCs. Silencing of DC-expressed SIRPα induced spontaneous and enhanced production of IL12 and costimulatory molecules, resulting in more potent cytotoxic T lymphocyte responses, including the eradication of previously established solid tumors. SIRPα exerted such effects, at least in part, via the association and sequestration of p85 subunit of PI3K. Thus, SIRPα is a critical regulator of DC lifespan and activity, and its inhibition might improve the clinical efficacy of DC-based tumor vaccines.

14.
J Hepatol ; 65(2): 314-24, 2016 08.
Article in English | MEDLINE | ID: mdl-27154061

ABSTRACT

BACKGROUND & AIMS: Considerable evidence suggests that adrenergic signaling played an essential role in tumor progression. However, its role in hepatocellular carcinoma (HCC) and the underlying mechanisms remain unknown. METHODS: The effect of adrenaline in hepatocarcinogenesis was observed in a classical diethylnitrosamine-induced HCC mouse model. Effects of ADRB2 signaling inhibition in HCC cell lines were analyzed in proliferation, apoptosis, colony formation assays. Autophagy regulation by ADRB2 was assessed in immunoblotting, immunofluorescence and immunoprecipitation assays. In vivo tumorigenic properties and anticancer effects of sorafenib were examined in nude mice. Expression levels of ADRB2 and hypoxia-inducible factor-1α (HIF1α) in 150 human HCC samples were evaluated by immunohistochemistry. RESULTS: We uncovered that adrenaline promoted DEN-induced hepatocarcinogenesis, which was reversed by the ADRB2 antagonist ICI118,551. ADRB2 signaling also played an essential role in sustaining HCC cell proliferation and survival. Notably, ADRB2 signaling negatively regulated autophagy by disrupting Beclin1/VPS34/Atg14 complex in an Akt-dependent manner, leading to HIF1α stabilization, reprogramming of HCC cells glucose metabolism, and the acquisition of resistance to sorafenib. Conversely, inhibition of ADRB2 signaling by ICI118,551, or knockdown ADRB2 expression, led to enhanced autophagy, HIF1α destabilization, tumor growth suppression, and improved anti-tumor activity of sorafenib. Consistently, ADRB2 expression correlated positively with HIF1α in HCC specimens and was associated with HCC outcomes. CONCLUSIONS: Our results uncover an important role of ADRB2 signaling in regulating HCC progression. Given the efficacy of ADRB2 modulation on HCC inhibition and sorafenib resistance, adrenoceptor antagonist appears to be a putative novel treatment for HCC and chemoresistance. LAY SUMMARY: ADRB2 signaling played an essential role in sustaining hepatocellular carcinoma cell proliferation and survival. ADRB2 signaling negatively regulated autophagy, leading to hypoxia-inducible factor-1α stabilization, reprogramming of hepatocellular carcinoma cells glucose metabolism, and the acquisition of resistance to sorafenib. Adrenoceptor antagonist appears to be a putative novel treatment for hepatocellular carcinoma and chemoresistance.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Autophagy , Disease Progression , Drug Resistance, Neoplasm , Humans , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Receptors, Adrenergic, beta-2 , Signal Transduction , Sorafenib
15.
Hepatology ; 61(2): 585-97, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25294684

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a prototype of inflammation-associated cancer. Oncoprotein Gankyrin, which mostly increases in HCC, plays a critical role in HCC development and metastasis. However, the exact mechanism of Gankyrin up-regulation in HCC remains unclear. A Gankyrin luciferase reporter was developed to screen a potential regulator for Gankyrin from a list of proinflammatory cytokines, and interleukin (IL)-1ß was found as one of its activators. In clinical premalignant and malignant liver disease samples, enhanced IL-1ß/interleukin-1 receptor-associated kinase 1 (IRAK-1) signaling accompanied by increased Gankyrin was observed. Lower expression of Gankyrin and phospho-IRAK-1 are favorable prognostic markers for HCC. A similar correlation was observed in the diethylnitrosamine (DEN) model of rat hepatocarcinogenesis. The results from Gankyrin reporter activity, real-time polymerase chain reaction, or immunoblotting further confirmed the up-regulation of Gankyrin by IL-1ß/IRAK-1 inflammatory signaling. Moreover, a series of Gankyrin's truncated reporters were constructed, and electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) were performed to analyze the properties of Gankyrin promoter. Mechanistically, the core promoter of Gankyrin contains the binding site of nuclear factor Y (NF-Y) family members, which can recruit histone acetyltransferase coactivator E1A-binding protein p300 (p300) or CREB-binding protein (CBP) to promote Gankyrin transcription. Conversely, knockdown of NF-Y, p300, or CBP inhibits Gankyrin expression. IL-1ß stimulation causes sequential phosphorylation of IRAK-1, c-Jun N-terminal kinase (JNK), and p300 and enhances recruitment of the p300/CBP/NF-Y complex to Gankyrin promoter. Inhibition of phospho-JNK impairs IL-1ß/IRAK-1 signaling-mediated up-regulation of Gankyrin. CONCLUSION: The finding of IL-1ß/IRAK-1 signaling promoting Gankyrin expression through JNK and NF-Y/p300/CBP complex provides a fresh view on inflammation-enhanced hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1beta/metabolism , Liver Neoplasms, Experimental/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/metabolism , Adult , Aged , Animals , CCAAT-Binding Factor/metabolism , CREB-Binding Protein/metabolism , Carcinoma, Hepatocellular/virology , Case-Control Studies , Cattle , Diethylnitrosamine , E1A-Associated p300 Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Hep G2 Cells , Hepatitis B/complications , Hepatitis B/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Liver Neoplasms, Experimental/virology , Male , Mice , Middle Aged , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins/genetics , Rats, Sprague-Dawley , Young Adult
16.
Nat Commun ; 5: 5256, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25348021

ABSTRACT

Increasing evidence suggests that TLR4 expression by tumour cells promotes tumour progression, but it is unclear whether TLR4 is involved in metastasis. Here we show that TLR4 deficiency significantly diminishes experimental lung metastasis without affecting primary tumour growth. Bone marrow transplantation experiment and application of antiplatelet agents in mice demonstrate that TLR4 on platelets plays an important role in metastasis. TLR4 is critical for platelet-tumour cell interaction in vitro. Furthermore, high-mobility group box1 (HMGB1) neutralization attenuates platelet-tumour cell interaction in vitro and metastasis in vivo in a TLR4-dependent manner, indicating that tumour cell-released HMGB1 is the key factor that interacts with TLR4 on platelets and mediates platelet-tumour cell interaction, which promotes metastasis. These findings demonstrate a mechanism by which platelets promote tumour cell metastasis and suggest TLR4, and its endogenous ligand HMGB1 as targets for antimetastatic therapies.


Subject(s)
Blood Platelets/metabolism , Carcinoma, Lewis Lung/pathology , Cell Communication , HMGB1 Protein/metabolism , Melanoma, Experimental/pathology , Toll-Like Receptor 4/metabolism , Animals , Cell Communication/drug effects , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Hematopoiesis/drug effects , Male , Mice, Inbred C57BL , Neoplasm Metastasis , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/pharmacology , Protein Binding/drug effects , Toll-Like Receptor 4/deficiency
17.
Clin Cancer Res ; 19(19): 5372-80, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23942093

ABSTRACT

PURPOSE: The AMP-activated protein kinase (AMPK) serves as an energy sensor in eukaryotic cells and occupies a central role in linking metabolism and cancer development. However, the phosphorylation status of AMPK and its therapeutic value in human hepatocellular carcinoma (HCC) remain unclear. EXPERIMENTAL DESIGN: The phosphorylation status of AMPK (Thr172) was determined by immunoblotting and immunostaining in specimens from 273 patients with HCC (including 253 patients with hepatitis B virus -related HCC). Kaplan-Meier survival analysis was used to determine the correlation with prognosis. The effects of therapeutic metformin/AMPK activation were assessed in cultured human HCC cell lines and primary HCC cells in vitro and in xenograft tumors model in vivo. To define the mechanisms of anticancer effects of metformin, we examined its influence on AMPK activation and NF-κB pathway. RESULTS: AMPK is dysfunctional in patients with HCC, and low p-AMPK staining is correlated with aggressive clinicopathologic features and poor prognosis. Activation of AMPK by metformin not only inhibited HCC cells growth in vitro and in vivo, but also augmented cisplatin-induced growth inhibition in HCC cells. Knockdown of AMPKα expression can greatly decrease the inhibitory effect of metformin, indicating that AMPK activation is required for the anticancer action of metformin. Mechanistically, metformin/AMPK activation inhibited NF-κB signaling through upregulation of IκBα. Activation of NF-κB signaling by ectopic expression of P65 or overexpression of an undegradable mutant form of IκBα attenuated the anticancer effects of metformin. CONCLUSIONS: These results present novel insight into a critical role of AMPK in HCC progression. Anticancer effects of therapeutic metformin/AMPK activation unravel metformin's potential in treatment of HCC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Metformin/pharmacology , Adult , Aged , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Female , Humans , Interleukin-6/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Metformin/administration & dosage , Mice , Middle Aged , NF-kappa B/metabolism , Neoplasm Staging , Phosphorylation/drug effects , Prognosis , Risk Factors , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects
18.
Cancer Lett ; 335(1): 160-7, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23410872

ABSTRACT

ß-Catenin plays many critical roles during various liver physiological and pathological processes. However, the role of ß-Catenin in acute liver failure remains unclear. Using hepatocyte specific ß-Catenin knockout mice, we found that loss of ß-Catenin in hepatocyte significantly reduced GalN/LPS-induced liver damage and hepatocyte apoptosis, but exacerbated Jo2-mediated liver injury. Mechanistically, the dual effects of ß-Catenin attributes on its function of inhibiting NF-κB signaling, which aggravates oxidative stress but decreases Fas expression under injury conditions. In conclusion, ß-Catenin plays an important role in regulating the balance between TNF-α and Fas-induced liver injury via its effect on NF-κB.


Subject(s)
Acute Lung Injury/metabolism , Tumor Necrosis Factor-alpha/physiology , beta Catenin/physiology , fas Receptor/physiology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Alanine Transaminase/blood , Animals , Apoptosis , Aspartate Aminotransferases/blood , Cell Line, Tumor , Gene Knockout Techniques , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Oxidative Stress
19.
J Hepatol ; 57(4): 803-12, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22727732

ABSTRACT

BACKGROUND & AIMS: Due to its anatomic connection, the liver is constantly exposed to gut-derived bacterial products or metabolites. Disruption of gut homeostasis is associated with many human diseases. The aim of this study was to determine the role of gut homeostasis in initiation and progression of hepatocellular carcinoma (HCC). METHODS: Disruption of intestinal homeostasis by penicillin or dextran sulfate sodium (DSS) and its restoration by probiotics were applied in a diethylnitrosamine (DEN) model of rat hepatocarcinogenesis. RESULTS: Patients with liver cirrhosis and HCC had significantly increased serum endotoxin levels. Chronic DEN treatment of rats was associated with an imbalance of subpopulations of the gut microflora including a significant suppression of Lactobacillus species, Bifidobacterium species and Enterococcus species as well as intestinal inflammation. Induction of enteric dysbacteriosis or intestinal inflammation by penicillin or DSS, respectively, significantly promoted tumor formation. Administration of probiotics dramatically mitigated enteric dysbacteriosis, ameliorated intestinal inflammation, and most importantly, decreased liver tumor growth and multiplicity. Interestingly, probiotics not only inhibited the translocation of endotoxin, which bears pathogen-associated molecular patterns (PAMPs) but also the activation of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1). As a result, the production of pro- and anti-inflammatory cytokines was skewed in favor of a reduced tumorigenic inflammation in the liver. CONCLUSIONS: The data highlights the importance of gut homeostasis in the pathogenesis of HCC. Modulation of the gut microbiota by probiotics may represent a new avenue for therapeutic intervention to treat or prevent HCC development.


Subject(s)
Carcinoma, Hepatocellular/pathology , Endotoxins/metabolism , Gastrointestinal Tract/microbiology , Homeostasis , Liver Neoplasms, Experimental/pathology , Probiotics/pharmacology , Alkylating Agents/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bifidobacterium/drug effects , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/etiology , Cytokines/biosynthesis , Dextran Sulfate/pharmacology , Diethylnitrosamine/pharmacology , Diethylnitrosamine/toxicity , Disease Progression , Endotoxins/blood , Enterococcus/drug effects , Gastroenteritis/chemically induced , Gastroenteritis/drug therapy , Gastroenteritis/metabolism , Gastrointestinal Tract/physiopathology , HMGB1 Protein/metabolism , Homeostasis/drug effects , Humans , Lactobacillus/drug effects , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/microbiology , Male , Penicillins/pharmacology , Probiotics/therapeutic use , Rats , Rats, Sprague-Dawley
20.
J Hepatol ; 57(3): 613-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22612999

ABSTRACT

BACKGROUND & AIMS: Accumulating evidence suggests the involvement of tumor-initiating cells (T-ICs) in cancer genesis, but whether liver T-ICs contribute to HCC invasion and metastasis remains unclear. METHODS: OV6(+) T-ICs were isolated from SMMC7721 and HuH7 cell lines by magnetic sorting. Characteristics of T-ICs were assessed by in vitro and mouse xenograft assays. Expression of OV6 was determined by immunostaining in specimens from 218 HCC patients, and Kaplan-Meier survival analysis was used to determine the correlation of OV6 expression with prognosis. RESULTS: OV6(+) T-ICs isolated from HCC cell lines not only possess a higher capacity to form tumor spheroids in vitro, but also had a greater potential to form tumors when implanted in non-obese diabetic/severe combined immunodeficient mice, suggesting their elevated self-renewal capacity and tumorigenicity. Moreover, OV6(+) T-ICs exhibited more invasive and metastatic potentials both in vitro and in vivo. Patients with more OV6(+) tumor cells were associated with aggressive clinicopathologic features and poor prognosis. CXCR4 is expressed at higher levels in OV6(+) cells. Recombinant stromal cell-derived factor-1 (SDF-1) treatment expanded the OV6(+) HCC T-ICs population, by sustaining the stem cell property of OV6(+) cells. The SDF-1 effect was blocked by a specific CXCR4 inhibitor, AMD3100, or transfection of siRNA targeting CXCR4. CONCLUSIONS: OV6(+) HCC cells may represent a subpopulation of T-ICs with augmented invasion and metastasis potential, which contribute to progression and metastasis of HCC. The SDF-1/CXCR4 axis also provides therapeutic targets for elimination of liver T-ICs.


Subject(s)
Antigens, Differentiation/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Benzylamines , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Cyclams , Disease Progression , Disease-Free Survival , Epithelial Cell Adhesion Molecule , Female , Gene Knockdown Techniques , Glycoproteins/metabolism , Heterocyclic Compounds/pharmacology , Humans , Kaplan-Meier Estimate , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Peptides/metabolism , Prognosis , RNA, Small Interfering , Receptors, CXCR4/drug effects , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Spheroids, Cellular
SELECTION OF CITATIONS
SEARCH DETAIL
...