Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Gastrointest Surg ; 26(8): 1-9, 2022 08.
Article in English | MEDLINE | ID: mdl-35508683

ABSTRACT

BACKGROUND AND PURPOSE: The present study aimed to establish a γ-glutamyl transpeptidase-to-albumin ratio (GAR)-based nomogram model to predict early recurrence of hepatocellular carcinoma (HCC) after radical surgery. METHODS: Patients enrolled in this study were randomly allocated into a train and validation cohort in a ratio of 7:3. The Least Absolute Shrinkage and Selection Operator (LASSO) proportional hazards model and cox regression model were combined to identify independent risk factors related to HCC recurrence. Based on these risk factors, a predictive nomogram was constructed and validated in both inner and outer test cohorts. The performance of the nomogram was evaluated by C-index, the area under the receiver operating characteristic curve (AUC), the calibration curve and decision curve analysis. RESULTS: The tumor size, tumor number, BCLC stage, microvascular invasion (MVI) and GAR value were identified as independent risk factors related to HCC recurrence and used to construct the predictive nomogram. AUC of the nomogram showed satisfactory accuracy in predicting 1-, 3- and 5-year disease-free survival. The calibration curve showed agreement between the ideal and predicted values. The risk score more than 72 as calculated by the nomogram was related to early recurrence of HCC after radical surgery. DCA plots showed better clinical usability of the nomogram as compared with the BCLC staging system in all three included cohorts. CONCLUSION: The nomogram based on the GAR value may provide a new option for screening of the target HCC cohort of patients who need anti-recurrence therapy after surgery.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Albumins , Carcinoma, Hepatocellular/pathology , Hepatectomy , Humans , Liver Neoplasms/pathology , Nomograms , Retrospective Studies , gamma-Glutamyltransferase
2.
Cancer Manag Res ; 14: 969-980, 2022.
Article in English | MEDLINE | ID: mdl-35283645

ABSTRACT

Background: RBM10's function in hepatocellular carcinoma (HCC) has rarely been addressed. We intend to explore the prognostic significance and therapeutic meaning of RBM10 in HCC in this study. Methods: Multiple common databases were integrated to analyze the expression status and prognostic meaning of RBM10 in HCC. The relationship between RBM10 mRNA level and clinical features was also assessed. Multiple enrichment analyses of the differentially expressed genes between RBM10 high- and low- transcription groups were constructed by using R software (version 4.0.2). A Search Tool for Retrieval of Interacting Genes database was used to construct the protein-protein interaction network between RBM10 and other proteins. A tumor immune estimation resource database was employed to identify the relationship between RBM10 expression and immune cell infiltrates. The prognostic value of RBM10 expression was validated in our HCC cohort by immunohistochemistry test. Results: The transcription of RBM10 mRNA was positively correlated with tumor histologic grade (p < 0.001), T classification (p < 0.001), and tumor stage (p < 0.001). High transcription of RBM10 in HCC predicted a dismal overall survival (p = 0.0037) and recurrence-free survival (p < 0.001). Kyoto Encyclopedia of Genes and Genomes, Gene Ontology, and Gene Set Enrichment Analysis all revealed that RBM10 was involved in the regulation of cell cycle, DNA replication, and immune-related pathways. Tumor immune estimation analysis revealed that RBM10 transcription was positively related to multiple immune cell infiltrates and the expressions of PD-1 and PD-L1. Conclusion: RBM10 was demonstrated to be a dismal prognostic factor and a potential biomarker for immune therapy in HCC in that it may be involved in the immune-related signaling pathways.

3.
J Gastroenterol ; 55(1): 51-66, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31562534

ABSTRACT

BACKGROUND: Gallbladder carcinoma (GBC) is one of the most aggressive and lethal tumors, with extremely high metastatic activity and poor prognosis. Previously we have studied miRNAs that promote metastasis and progression of GBC, the aim of present study was to systematically elucidate the metastasis suppressor miRNAs in GBC. METHODS: A novel designed high-throughput screening method that combined high content screening (HCS) and miRNA microarray analysis was conducted to filter out anti-metastatic miRNAs of GBC. Frozen samples were analyzed for the expression of goal miRNAs by real-time PCR. The biological functions of miRNAs were studied by transwell, immunoblot. Liver metastasis model via spleen injection was further examined in nude mice. Kaplan-Meier and Cox regression analyses were used to analyze the effect of goal miRNAs on overall survival. The target genes and interaction network of goal miRNAs were determined by whole transcriptome genome sequencing. RESULTS: Out of the miRNAs library, a series of prominent metastatic suppressor miRNA candidates were filtered out. Among them, miR-7-2-3p and miR-29c-3p were discovered downregulated in GBC, and upregulation of them could reverse epithelial-mesenchymal transition and decrease the metastasis ability of GBC cells in vitro and in vivo, which was dominated by the miRNA-mRNA-lncRNA co-expression network. And DCLK1 and SLC36A1 are the direct target genes of miR-7-2-3p and miR-29c-3p. Moreover, the deficiency of miR-7-2-3p and miR-29c-3p was closely associated with poor prognosis of GBC patients. CONCLUSIONS: Our findings indicate that miR-7-2-3p and miR-29c-3p play crucial roles in the pathogenesis and worse prognosis of GBCs, which may serve as prognosis biomarkers and promise potential therapeutic targets in the future.


Subject(s)
Biomarkers, Tumor/metabolism , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , MicroRNAs/metabolism , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/genetics , Blotting, Western , Cell Line, Tumor , Down-Regulation , Female , Follow-Up Studies , Gallbladder Neoplasms/diagnosis , Gallbladder Neoplasms/mortality , High-Throughput Screening Assays , Humans , Immunoblotting , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Prognosis , Real-Time Polymerase Chain Reaction , Retrospective Studies , Survival Analysis , Up-Regulation , Exome Sequencing
4.
Article in English | MEDLINE | ID: mdl-31824932

ABSTRACT

MicroRNAs (miRNA) have been identified as oncogenic drivers and tumor suppressors in every major cancer type. In this work, we design an artificial intelligent signal amplification (AISA) system including double-stranded SQ (S, signal strand; Q, quencher strand) and FP (F, fuel strand; P, protect strand) according to thermodynamics principle for sensitive detection of miRNA in vitro and in vivo. In this AISA system for miRNA detection, strand S carries a quenched imaging marker inside the SQ. Target miRNA is constantly replaced by a reaction intermediate and circulatively participates in the reaction, similar to enzyme. Therefore, abundant fluorescent substances from S and SP are dissociated from excessive SQ for in vitro and in vivo visualization. The versatility and feasibility for disease diagnosis using this system were demonstrated by constructing two types of AISA system to detect Hsa-miR-484 and Hsa-miR-100, respectively. The minimum target concentration detected by the system in vitro (10 min after mixing) was 1/10th that of the control group. The precancerous lesions of liver cancer were diagnosed, and the detection accuracy were larger than 94% both in terms of location and concentration. The ability to establish this design framework for AISA system with high specificity provides a new way to monitor tumor progression and to assess therapeutic responses.

5.
Biochem Biophys Res Commun ; 516(3): 851-857, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31266633

ABSTRACT

A number of homeobox genes are implicated in the malignancy of various cancers. Here, we investigated the role of the homeobox gene SIX4 in non-small-cell lung cancer (NSCLC). The sine oculis homeobox (SIX4) gene was found to be highly expressed at both mRNA and protein levels in NSCLC tumor tissues as compared with matching normal counterparts. In this study, the SIX4 gene of two human NSCLC cell lines (A549 and PC9) was overexpressed or silenced using the lentiviral system. We evaluated the malignancy-associated phenotype of transfected cells, which demonstrated that exogenous expression of the SIX4 gene greatly enhanced the proliferation, migration, and invasion of NSCLC cells. The opposite was true in the SIX4-silenced cells. Transcriptomic profiling analysis revealed that the SIX4 gene modulated the expression of hundreds of downstream target genes in a cell context-dependent manner. Most notably, the SIX4 gene controls the expression of crucial genes with evidently oncogenic function. We conclude that SIX4 plays an oncogenic role and may be potentially utilized as a diagnostic and therapeutic marker for NSCLC.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Trans-Activators/genetics , A549 Cells , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Profiling , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Homeodomain Proteins/agonists , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Trans-Activators/agonists , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism
6.
PeerJ ; 7: e6629, 2019.
Article in English | MEDLINE | ID: mdl-30993034

ABSTRACT

BACKGROUND: Identification of specific biomarkers is important for the diagnosis and treatment of non-small cell lung cancer (NSCLC). HOXC6 is a homeodomain-containing transcription factor that is highly expressed in several human cancers; however, its role in NSCLC remains unknown. METHODS: The expression and protein levels of HOXC6 were assessed in NSCLC tissue samples by Quantitative real-time PCR (qRT-PCR) and immunohistochemistry, respectively. HOXC6 was transfected into the NSCLC cell lines A549 and PC9, and used to investigate its effect on proliferation, migration, and invasion using CFSE, wound healing, and Matrigel invasion assays. Next-generation sequencing was also used to identify downstream targets of HOXC6 and to gain insights into the molecular mechanisms underlying its biological function. RESULTS: HOXC6 expression was significantly increased in 66.6% (20/30) of NSCLC tumor samples in comparison to normal controls. HOXC6 promoted proliferation, migration, and invasion of NSCLC cells in vitro. RNA-seq analysis demonstrated the upregulation of 310 and 112 genes in A549-HOXC6 and PC9-HOXC6 cells, respectively, and the downregulation of 665 and 385 genes in A549-HOXC6 and PC9-HOXC6 cells, respectively. HOXC6 was also found to regulate the expression of genes such as CEACAM6, SPARC, WNT6, CST1, MMP2, and KRT13, which have documented pro-tumorigenic functions. DISCUSSION: HOXC6 is highly expressed in NSCLC, and it may enhance lung cancer progression by regulating the expression of pro-tumorigenic genes involved in proliferation, migration, and invasion. Our study highlighted the oncogenic potential of HOXC6, and suggests that it may be a novel biomarker for the diagnosis and treatment of NSCLC.

7.
J Cell Biochem ; 119(8): 6842-6856, 2018 08.
Article in English | MEDLINE | ID: mdl-29693272

ABSTRACT

Baicalein, a natural flavonoid, has fascinating anti-cancer properties in breast cancer. Long noncoding RNAs (lncRNAs), a class of transcripts with no protein-coding potential, also exhibit critical roles in breast cancer. However, the molecular mechanisms mediating the anti-cancer properties of baicalein and whether lncRNAs are involved in the anti-cancer effects are still unclear. In this study, we identified a novel isoform of lncRNA PAX8-AS1 (PAX8-AS1-N), which is activated by baicalein in a dose- and time-dependent manner. Functional assays showed that PAX8-AS1-N reduced cell viability, inhibited cell-cycle progression, and induced apoptosis of breast cancer cells in vitro. Depletion of PAX8-AS1-N promoted breast xenograft tumor growth in vivo. Furthermore, depletion of PAX8-AS1-N attenuated the suppressive roles of baicalein on cell viability, the apoptosis induced by baicalein, and also the suppressive roles of baicalein on tumor growth in vivo. Mechanistically, PAX8-AS1-N bound to miR-17-5p, and up-regulated miR-17-5p targets, such as PTEN, CDKN1A, and ZBTB4. In addition, PAX8-AS1-N was down-regulated in breast cancer and reduced expression of PAX8-AS1-N indicated poor survival of breast cancer patients. In conclusion, our results demonstrated that PAX8-AS1-N activation mediated the anti-cancer effects of baicalein via regulating miR-17-5p, and suggested that baicalein and enhancing PAX8-AS1-N would be potential therapeutic strategies against breast cancer.


Subject(s)
Breast Neoplasms , Flavanones/pharmacology , RNA Isoforms , RNA, Long Noncoding , RNA, Neoplasm , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA Isoforms/genetics , RNA Isoforms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Xenograft Model Antitumor Assays
8.
Chem Biol Interact ; 285: 48-58, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29481769

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancer and leading cause of cancer-related death worldwide. Baicalein, a principle flavonoid, has shown attractive anti-cancer effects on HCC. However, the underlying molecular mechanisms and influencing factors contributing to the anti-cancer effects of baicalein on HCC are still largely unknown. Long noncoding RNAs (lncRNAs) have been revealed to be fascinating therapeutic targets for cancers. The roles of NF-κB Interacting LncRNA (NKILA) are recently explored in several cancers. However, the expressions, clinical significances, roles and action mechanisms of NKILA in the anti-cancer effects of baicalein on HCC are unknown. In this study, we found that NKILA is down-regulated in HCC and reduced expression of NKILA indicts poor survival of HCC patients. Functional assays showed that overexpression of NKILA enhances the roles of baicalein on HCC cell proliferation inhibition, apoptosis induction, and migration inhibition in vitro and tumor growth suppression in vivo. Conversely, knockdown of NKILA suppresses the effects of baicalein. Mechanistically, we found that NKILA inhibits IκBα phosphorylation, NF-κB nuclear translocation, and NF-κB activity. NKILA also enhances the inhibitory effects of baicalein on NF-κB signaling. Furthermore, the effects of NKILA on baicalein-induced NF-κB activity inhibition, cell growth inhibition, apoptosis induction, and migration inhibition are reversed by NF-κB nuclear translocation inhibitor JSH-23. Collectively, our data demonstrated that NKILA enhances the anti-cancer effects of baicalein on HCC in vitro and in vivo via the regulation of NF-κB signaling, and implied that the combination of NKILA and baicalein would be potential therapeutic strategies for HCC.


Subject(s)
Drug Synergism , Flavanones/pharmacology , Liver Neoplasms/therapy , NF-kappa B/metabolism , RNA, Long Noncoding/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/physiopathology , Flavanones/therapeutic use , Humans , Liver Neoplasms/physiopathology , Mice , RNA, Long Noncoding/therapeutic use , Tumor Burden/drug effects
9.
Hepatology ; 67(3): 1071-1087, 2018 03.
Article in English | MEDLINE | ID: mdl-28960380

ABSTRACT

It is urgent that the means to improve liver regeneration (LR) be found, while mitigating the concurrent risk of hepatocarcinogenesis (HCG). Nuclear receptor corepressor 1 (NCoR1) is a co-repressor of nuclear receptors, which regulates the expression level of metabolic genes; however, little is known about its potential contribution for LR and HCG. Here, we found that liver-specific NCoR1 knockout in mice (NCoR1Δhep ) dramatically enhances LR after partial hepatectomy and, surprisingly, blocks the process of diethylnitrosamine (DEN)-induced HCG. Both RNA-sequencing and metabolic assay results revealed improved expression of Fasn and Acc2 in NCoR1Δhep mice, suggesting the critical role of de novo fatty acid synthesis (FAS) in LR. Continual enhanced de novo FAS in NCoR1Δhep mice resulted in overwhelmed adenosine triphosphate ATP and nicotinamide adenine dinucleotide phosphate (NADPH) consumption and increased mitochondrial reactive oxygen species production, which subsequently attenuated HCG through inducing apoptosis of hepatocytes at an early stage after DEN administration. CONCLUSION: NCoR1 functions as a negative modulator for hepatic de novo FAS and mitochondria energy adaptation, playing distinct roles in regeneration or carcinogenesis. (Hepatology 2018;67:1071-1087).


Subject(s)
Carcinogenesis/metabolism , Lipogenesis/genetics , Liver Regeneration/genetics , Liver/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Animals , Apoptosis , Cell Proliferation/genetics , Fatty Acids/biosynthesis , Hepatocytes/metabolism , Liver/pathology , Mice , Mice, Knockout
10.
Int J Biochem Cell Biol ; 94: 107-118, 2018 01.
Article in English | MEDLINE | ID: mdl-29175387

ABSTRACT

Baicalein, an active flavonoid extracted from the root of Scutellaria baicalensis Georgi, has fascinating anti-cancer effects on many cancers. Our previous study also found that baicalein inhibited cervical cancer cell proliferation and migration, and induced cervical cancer cell apoptosis and cell cycle arrest. However, the molecular mechanisms underlying the anti-cancer effects of baicalein are largely unknown. In this study, we identified a novel long noncoding RNA (lncRNA), which is downregulated by baicalein in a dose- and time-dependent manner in cervical cancer. We named this lncRNA as baicalein down-regulated long noncoding RNA (BDLNR). Gain-of- and loss-of-function assays showed that BDLNR was required for baicalein-induced cell proliferation inhibition, cell death induction, migration inhibition, and in vivo tumor growth inhibition of cervical cancer. Mechanistically, BDLNR physically bound to YBX1, recruited YBX1 to PIK3CA promoter, activated PIK3CA expression and PI3K/Akt pathway. Furthermore, BDLNR was upregulated in cervical cancer and associated with poor prognosis of cervical cancer patients. Collectively, our data demonstrated that BDLNR mediated the anti-cancer effects of baicalein in cervical cancer via activating PI3K/Akt pathway, and implied that BDLNR would be potential therapeutic target for enhancing the anti-cancer effects of baicalein in cervical cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cervix Uteri/drug effects , Down-Regulation/drug effects , Flavanones/therapeutic use , RNA, Long Noncoding/antagonists & inhibitors , RNA, Neoplasm/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Cell Line , Cell Line, Tumor , Cell Proliferation , Cervix Uteri/metabolism , Cervix Uteri/pathology , Female , Flavanones/adverse effects , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Signal Transduction/drug effects , Survival Analysis , Tumor Burden/drug effects , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
J Mol Cell Biol ; 9(6): 489-503, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28655161

ABSTRACT

Little is known about how chronic inflammation contributes to the progression of hepatocellular carcinoma (HCC), especially the initiation of cancer. To uncover the critical transition from chronic inflammation to HCC and the molecular mechanisms at a network level, we analyzed the time-series proteomic data of woodchuck hepatitis virus/c-myc mice and age-matched wt-C57BL/6 mice using our dynamical network biomarker (DNB) model. DNB analysis indicated that the 5th month after birth of transgenic mice was the critical period of cancer initiation, just before the critical transition, which is consistent with clinical symptoms. Meanwhile, the DNB-associated network showed a drastic inversion of protein expression and coexpression levels before and after the critical transition. Two members of DNB, PLA2G6 and CYP2C44, along with their associated differentially expressed proteins, were found to induce dysfunction of arachidonic acid metabolism, further activate inflammatory responses through inflammatory mediator regulation of transient receptor potential channels, and finally lead to impairments of liver detoxification and malignant transition to cancer. As a c-Myc target, PLA2G6 positively correlated with c-Myc in expression, showing a trend from decreasing to increasing during carcinogenesis, with the minimal point at the critical transition or tipping point. Such trend of homologous PLA2G6 and c-Myc was also observed during human hepatocarcinogenesis, with the minimal point at high-grade dysplastic nodules (a stage just before the carcinogenesis). Our study implies that PLA2G6 might function as an oncogene like famous c-Myc during hepatocarcinogenesis, while downregulation of PLA2G6 and c-Myc could be a warning signal indicating imminent carcinogenesis.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Cytochrome P450 Family 2/genetics , Gene Regulatory Networks , Group VI Phospholipases A2/genetics , Inflammation/pathology , Liver Neoplasms/genetics , Signal Transduction , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Chronic Disease , Cytochrome P450 Family 2/metabolism , Down-Regulation , Group VI Phospholipases A2/metabolism , Humans , Inflammation/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Proteomics , Reproducibility of Results
12.
Genes Dev ; 31(3): 247-259, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28223311

ABSTRACT

Tumor infiltrated type II (M2) macrophages promote tumorigenesis by suppressing immune clearance, promoting proliferation, and stimulating angiogenesis. Interestingly, macrophages were also found to enrich in small foci of altered hepatocytes containing liver tumor-initiating cells (TICs). However, whether and how TICs specifically recruit macrophages and the function of these macrophages in tumor initiation remain unknown due to technical difficulties. In this study, by generating genetically defined liver TICs, we demonstrate that TICs actively recruit M2 macrophages from as early as the single-cell stage. Elimination of TIC-associated macrophages (TICAMs) abolishes tumorigenesis in a manner dependent on the immune system. Mechanistically, activation of the Hippo pathway effector Yes-associated protein (YAP) underlies macrophage recruitment by TICs. These results demonstrate for the first time that macrophages play a decisive role in the survival of single TICs in vivo and provide a proof of principle for TIC elimination by targeting YAP or M2 macrophages.


Subject(s)
Carcinoma, Hepatocellular/immunology , Cell Transformation, Neoplastic/immunology , Hepatocytes/immunology , Liver Neoplasms/immunology , Macrophages/immunology , Neoplastic Stem Cells/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Communication/immunology , Cell Cycle Proteins , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Hepatocyte Growth Factor/physiology , Hepatocytes/metabolism , Hepatocytes/pathology , Homeodomain Proteins/physiology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Macrophages/cytology , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Neoplastic Stem Cells/cytology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Serine-Threonine Kinase 3 , Tumor Suppressor Protein p53/physiology , YAP-Signaling Proteins
13.
Gut ; 65(7): 1186-201, 2016 07.
Article in English | MEDLINE | ID: mdl-26860770

ABSTRACT

OBJECTIVE: Precancerous lesion, a well-established histopathologically premalignant tissue with the highest risk for tumourigenesis, develops preferentially from activation of DNA damage checkpoint and persistent inflammation. However, little is known about the mechanisms by which precancerous lesions are initiated and their physiological significance. DESIGN: Laser capture microdissection was used to acquire matched normal liver, precancerous lesion and tumour tissues. miR-484(-/-), Ifnar1(-/-) and Tgfbr2(△hep) mice were employed to determine the critical role of the interferon (IFN)-microRNA pathway in precancerous lesion formation and tumourigenesis. RNA immunoprecipitation (RIP), pull-down and chromatin immunoprecipitation (ChIP) assays were applied to explore the underlying mechanisms. RESULTS: miR-484 is highly expressed in over 88% liver samples clinically. DEN-induced precancerous lesions and hepatocellular carcinoma were dramatically impaired in miR-484(-/-) mice. Mechanistically, ectopic expression of miR-484 initiates tumourigenesis and cell malignant transformation through synergistic activation of the transforming growth factor-ß/Gli and nuclear factor-κB/type I IFN pathways. Specific acetylation of H3K27 is indispensable for basal IFN-induced continuous transcription of miR-484 and cell transformation. Convincingly, formation of precancerous lesions were significantly attenuated in both Tgfbr2(△hep) and Ifnar1(-/-) mice. CONCLUSIONS: These findings demonstrate a new protumourigenic axis involving type I IFN-microRNA signalling, providing a potential therapeutic strategy to manipulate or reverse liver precancerous lesions and tumourigenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Precancerous Conditions/genetics , Signal Transduction , Acetylation , Animals , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/chemistry , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Hepatocytes , Humans , Interferon Type I/metabolism , Liver/chemistry , Liver Neoplasms/chemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Knockout , MicroRNAs/analysis , NF-kappa B/metabolism , NIH 3T3 Cells , Pentanones , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Receptor, Interferon alpha-beta/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1/metabolism
14.
Cancer Lett ; 364(1): 33-43, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25931210

ABSTRACT

Epigenetic modification of miR-429 can manipulate liver T-ICs via targeting the RBBP4/E2F1/Oct4 axis, which might be crucial for hepatocarcinogenesis. However, whether miR-429 plays a role in regulating metastasis of hepatocellular carcinoma is still unclear. Using quantitative methylation analysis and real-time PCR, we have identified the hypomethylated status and upregulation of miR-429 in portal vein metastasis samples in comparison with their matched primary tumor. The ectopic expression of miR-429 dramatically induced the expression of MMP2/7/9 and enhanced HCC migration and invasion in vitro and in vivo in an EMT-independent manner. Both bioinformatics and functional studies elucidated the direct regulation of miR-429 on the 3'UTR of the PTEN gene, which leads to the activation of PI3K/AKT signaling and the nuclear translocation of ß-catenin, eventually. Conversely, the knockdown of miR-429 efficiently recovered the expression of PTEN and attenuated PI3K/AKT/ß-catenin-mediated cell metastasis. Clinically, the higher expression of miR-429 and nucleus relocation of ß-catenin were identified as the adverse prognosis factors for recurrence-free survival (RFS) and overall survival (OS). In summary, our results here defined miR-429 as a key inducer for HCC pathogenesis and metastasis with potential utility for tumor intervention.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition , Liver Neoplasms/pathology , MicroRNAs/physiology , Neoplasm Metastasis , Wnt Proteins/metabolism , Base Sequence , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Molecular Sequence Data , PTEN Phosphohydrolase/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction
15.
Cancer Res ; 75(7): 1470-81, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25649767

ABSTRACT

CYP3A5 is a cytochrome P450 protein that functions in the liver metabolism of many carcinogens and cancer drugs. However, it has not been thought to directly affect cancer progression. In this study, we challenge this perspective by demonstrating that CYP3A5 is downregulated in many hepatocellular carcinomas (HCC), where it has an important role as a tumor suppressor that antagonizes the malignant phenotype. CYP3A5 was downregulated in multiple cohorts of human HCC examined. Lower CYP3A5 levels were associated with more aggressive vascular invasion, poor differentiation, shorter time to disease recurrence after treatment, and worse overall patient survival. Mechanistic investigations showed that CYP3A5 overexpression limited MMP2/9 function and suppressed HCC migration and invasion in vitro and in vivo by inhibiting AKT signaling. Notably, AKT phosphorylation at Ser473 was inhibited in CYP3A5-overexpressing HCC cells, an event requiring mTORC2 but not Rictor/mTOR complex formation. CYP3A5-induced ROS accumulation was found to be a critical upstream regulator of mTORC2 activity, consistent with evidence of reduced GSH redox activity in most clinical HCC specimens with reduced metastatic capacity. Taken together, our results defined CYP3A5 as a suppressor of HCC pathogenesis and metastasis with potential utility a prognostic biomarker.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP3A/physiology , Liver Neoplasms/enzymology , Lung Neoplasms/enzymology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Movement , Down-Regulation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mechanistic Target of Rapamycin Complex 2 , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , Proportional Hazards Models , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Transcriptional Activation
16.
Gut ; 64(1): 156-67, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24572141

ABSTRACT

OBJECTIVE: Liver tumour-initiating cells (T-ICs) are critical for hepatocarcinogenesis. However, the underlying mechanism regulating the function of liver T-ICs remains unclear. METHODS: Tissue microarrays containing 242 hepatocellular carcinoma (HCC) samples were used for prognostic analysis. Magnetically activated cell sorting was used to isolate epithelial cell adhesion molecule (EPCAM)-positive cells. The gene expressions affected by miR-429 were determined by arrays. Co-immunoprecipitation was used to study interactions among retinoblastoma protein (RB1), Rb binding protein 4 (RBBP4) and E2F transcription factor 1 (E2F1). The DNA methylation status in CpG islands was detected by quantitative methylation analysis. miRNAs in microvesicles were isolated by a syringe filter system. RESULTS: The significant prognosis factor miR-429 was upregulated in HCC tissues and also in primary liver T-ICs isolated from clinical samples. The enrichment of miR-429 in EPCAM+ T-ICs contributed to hepatocyte self-renewal, malignant proliferation, chemoresistance and tumorigenicity. A novel functional axis involving miR-429, RBBP4, E2F1 and POU class 5 homeobox 1 (POU5F1 or OCT4) governing the regulation of liver EPCAM+ T-ICs was established in vitro and in vivo. The molecular mechanism regulating miR-429 expression, involving four abnormal hypomethylated sites upstream of the miR-200b/miR-200a/miR-429 cluster, was first defined in both EPCAM+ liver T-ICs and very early-stage HCC tissues. miR-429 secreted by high-expressing cells has the potential to become a proactive signalling molecule to mediate intercellular communication. CONCLUSIONS: Epigenetic modification of miR-429 can manipulate liver T-ICs by targeting the RBBP4/E2F1/OCT4 axis. This miRNA might be targeted to inactivate T-ICs, thus providing a novel strategy for HCC prevention and treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Epigenesis, Genetic , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , Neoplastic Stem Cells , Retinoblastoma Protein/physiology , Humans , Prognosis , Tumor Cells, Cultured
17.
Gastroenterology ; 145(6): 1436-48.e1-12, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23933603

ABSTRACT

BACKGROUND & AIMS: Aberrant expression of MUC15 correlates with development of colorectal adenocarcinoma, and MUC15 has been reported to prevent trophoblast invasion of human placenta. However, little is known about the role of MUC15 in pathogenesis of hepatocellular carcinoma (HCC). METHODS: We analyzed HCC samples and matched nontumor liver tissues (controls) collected from 313 patients who underwent hepatectomy in Shanghai, China, from January 2006 through September 2009. Levels of messenger RNAs and proteins were determined by immunohistochemical, quantitative reverse transcription polymerase chain reaction, and immunoblot analyses. Statistical analyses were used to associate levels of MUC15 with tumor features and patient outcomes. RESULTS: Levels of MUC15 messenger RNA and protein were reduced in a greater percentage of HCC samples than control tissues. Tumors with reduced levels of MUC15 were more likely to have aggressive characteristics (eg, high levels of α-fetoprotein, vascular invasion, lack of encapsulation, and poor differentiation) than those with low levels. Patients whose tumors had reduced levels of MUC15 had shorter overall survival times (24 months vs 46 months for patients with tumors with high levels of MUC15) and time to disease recurrence. Stable expression of MUC15 in HCC cell lines (SMMC-7721 and HCC-LM3) reduced their proliferation and invasive features in vitro, and ability to form metastatic tumors in mice. MUC15 reduced transcription of the matrix metalloproteinases 2 and 7 increased expression of tissue inhibitor of metalloproteinase-2, which required phosphoinositide 3-kinase-v-akt murine thymoma viral oncogene homolog signaling. Physical interaction between MUC15 and epidermal growth factor receptor led to its relocation and degradation within early endosomes and was required for inactivation of phosphoinositide 3-kinase-v-akt murine thymoma viral oncogene homolog signaling. CONCLUSIONS: Reduced levels of MUC15 in HCCs are associated with shorter survival times of patients and reduced time to disease recurrence. Expression of MUC15 in HCC cells reduces their aggressive behavior in vitro and in mice by inducing dimerization of epidermal growth factor receptor and decreasing phosphoinositide 3-kinase signaling via v-akt murine thymoma viral oncogene homolog.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Dimerization , ErbB Receptors/physiology , Liver Neoplasms/physiopathology , Mucins/physiology , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Female , Humans , In Vitro Techniques , Liver/pathology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , Neoplasm Invasiveness/physiopathology , Predictive Value of Tests , Prognosis , Survival Rate
18.
J Hepatol ; 59(3): 518-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23665284

ABSTRACT

BACKGROUND & AIMS: The dysfunction of miRNAs has been demonstrated to participate in the development of various tumors. However, whether miRNAs are involved in metastasis and progression of gallbladder carcinoma (GBC) remains unknown. METHODS: A new designed gain-of-function miRNA screening technology was applied to filter out pro-metastatic miRNAs in GBC. Their expression in GBC tissues was validated by real-time PCR. The biological functions of miRNAs were intensively studied by transwell, immunoblot, immunohistochemical, and in situ hybridization assays. Tumorigenicity and liver metastasis were further examined in nude mice. RESULTS: Of 880 miRNAs, 17 were filtered out as the prominent metastatic inducers of GBCs. Among them, the upregulation of pro-metastatic miR-20a was closely associated with local invasion, distant metastasis, and poor prognosis of 67 followed-up GBC patients, clinically. Patients with higher miR-20a expression exhibited worse overall survival (OS and median OS time was 5 and 20 months, respectively) than the lower expression group. A dramatically increased TGF-ß1 level was found in GBC patients, which was responsible for the elevation of miR-20a. The ectopic expression of miR-20a could induce epithelial-mesenchymal transition and enhance metastasis of GBC cells in vitro and in vivo, by directly targeting the 3' UTR of Smad7, and subsequently promoting nuclear translocation of ß-catenin. Conversely, the blockage of miR-20a by specific antagomir effectively restored the expression of Smad7 and attenuated TGF-ß-induced cell metastasis. CONCLUSIONS: TGF-ß1-mediated activation of the miR-20a/Smad7/ß-catenin axis plays a pivotal role in the pathogenesis and worse prognosis of GBCs and may serve as a potential therapeutic target in the future.


Subject(s)
Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Metastasis/genetics , Animals , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gallbladder Neoplasms/pathology , Heterografts , Humans , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/secondary , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transforming Growth Factor beta1/metabolism , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...