Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Front Biosci (Landmark Ed) ; 29(3): 120, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38538251

ABSTRACT

BACKGROUND: Osteosarcoma cells are prone to metastasis, and the mechanism of N6-methyladenosine (m6A) methylation modification in this process is still unclear. Methylation modification of m6A plays an important role in the development of osteosarcoma, which is mainly due to abnormal expression of enzymes related to methylation modification of m6A, which in turn leads to changes in the methylation level of downstream target genes messenger RNA (mRNA) leading to tumor development. METHODS: We analyzed the expression levels of m6A methylation modification-related enzyme genes in GSE12865 whole-genome sequencing data. And we used shRNA (short hairpin RNA) lentiviral interference to interfere with METTL3 (Methyltransferase 3) expression in osteosarcoma cells. We studied the cytological function of METTL3 by Cell Counting Kit-8 (CCK8), flow cytometry, migration and other experiments, and the molecular mechanism of METTL3 by RIP (RNA binding protein immunoprecipitation), Western blot and other experiments. RESULTS: We found that METTL3 is abnormally highly expressed in osteosarcoma and interferes with METTL3 expression in osteosarcoma cells to inhibit metastasis, proliferation, and apoptosis of osteosarcoma cells. We subsequently found that METTL3 binds to the mRNA of CBX4 (chromobox homolog 4), a very important regulatory protein in osteosarcoma metastasis, and METTL3 regulates the mRNA and protein expression of CBX4. Further studies revealed that METTL3 inhibited metastasis of osteosarcoma cells by regulating CBX4. METTL3 has been found to be involved in osteosarcoma cells metastasis by CBX4 affecting the protein expression of matrix metalloproteinase 2 (MMP2), MMP9, E-Cadherin and N-Cadherin associated with osteosarcoma cells metastasis. CONCLUSIONS: These results suggest that the combined action of METTL3 and CBX4 plays an important role in the regulation of metastasis of osteosarcoma, and therefore, the METTL3-CBX4 axis pathway may be a new potential therapeutic target for osteosarcoma.


Subject(s)
Adenine , Bone Neoplasms , Matrix Metalloproteinase 2 , Osteosarcoma , Humans , Adenine/analogs & derivatives , Epigenesis, Genetic , Ligases/genetics , Matrix Metalloproteinase 2/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Osteosarcoma/genetics , Osteosarcoma/secondary , Polycomb-Group Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering , Bone Neoplasms/pathology
2.
Mol Cancer ; 16(1): 136, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28810927

ABSTRACT

BACKGROUND: Several of the thousands of human long noncoding RNAs (lncRNAs) have been functionally characterized, yet their potential involvement in hepatocellular carcinoma (HCC) remains poorly understood. METHODS: LncRNA-HOXD-AS1 was identified by microarray and validated by real-time PCR. The clinicopathological significance of HOXD-AS1 was analyzed by Kaplan-Meier method. Chromatin immunoprecipitation was conducted to examine the mechanism of HOXD-AS1 upregulation. The role of HOXD-AS1 in HCC cells was assessed both in vitro and in vivo. ceRNA function of HOXD-AS1 was evaluated by RNA immunoprecipitation and biotin-coupled miRNA pull down assays. RESULTS: In this study, we found that HOXD-AS1 was significantly upregulated in HCC tissues. Clinical investigation demonstrated high expression level of HOXD-AS1 was associated with poor prognosis and high tumor node metastasis stage of HCC patients, and was an independent risk factor for survival. Moreover, our results revealed that STAT3 could specifically interact with the promoter of HOXD-AS1 and activate HOXD-AS1 transcription. Knockdown of HOXD-AS1 significantly inhibited migration and invasion of HCC cells in vitro and distant lung metastasis in vivo. Additionally, HOXD-AS1 was enriched in the cytoplasm, and shared miRNA response elements with SOX4. Overexpression of HOXD-AS1 competitively bound to miR-130a-3p that prevented SOX4 from miRNA-mediated degradation, thus activated the expression of EZH2 and MMP2 and facilitated HCC metastasis. CONCLUSIONS: In summary, HOXD-AS1 is a prognostic marker for HCC patients and it may play a pro-metastatic role in hepatocarcinogenesis.


Subject(s)
Liver Neoplasms/metabolism , RNA, Long Noncoding/metabolism , RNA/metabolism , SOXC Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis/genetics , RNA/genetics , RNA, Long Noncoding/analysis , RNA, Long Noncoding/genetics , SOXC Transcription Factors/genetics , STAT3 Transcription Factor/genetics , Up-Regulation/genetics
3.
Cancer Sci ; 108(4): 653-662, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28196303

ABSTRACT

Emerging evidence has indicated that deregulation of long non-coding RNAs (lncRNAs) can contribute to the progression and metastasis of human cancer, including hepatocellular carcinoma (HCC). However, the roles of most lncRNAs in HCC remain largely unknown. Here we found a long noncoding RNA termed SchLAH (seven chromosome locus associated with HCC; also called BC035072) was generally downregulated in HCC. Low expression of SchLAH was significantly correlated with shorter overall survival of HCC patients. In vitro and in vivo assays indicated that overexpression of SchLAH inhibited the migration and lung metastasis of HCC cells. Knockdown of SchLAH by siRNA pool promoted the migration of HCC cells. RNA pull-down and RNA immunoprecipitation assays demonstrated SchLAH physically interacted with fused in sarcoma (FUS). PCR array analysis showed that RhoA and Rac1 were the downstream effector molecules of SchLAH during HCC metastasis. Knockdown of FUS rescued the mRNA levels of RhoA and Rac1 that were repressed by SchLAH. These results suggest that SchLAH may suppress the metastasis of HCC cells by interacting with FUS, which indicates potential of SchLAH for the prognosis and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA-Binding Protein FUS/genetics , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Prognosis , Protein Binding , RNA Interference , RNA, Long Noncoding/metabolism , RNA-Binding Protein FUS/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
4.
Med Oncol ; 33(7): 70, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27262566

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers, and its incidence is increasing worldwide. Neuropeptide Y (NPY) broadly expressed in the central and peripheral nervous system. It participates in multiple physiological and pathological processes through specific receptors. Evidences are accumulating that NPY is involved in development and progression in neuro- or endocrine-related cancers. However, little is known about the potential roles and underlying mechanisms of NPY receptors in HCC. In this study, we analyzed the expression of NPY receptors by real-time polymerase chain reaction, Western blot, and immunohistochemical staining. Correlation between NPY1R levels and clinicopathological characteristics, and survival of HCC patients were explored, respectively. Cell proliferation was researched by CCK-8 in vitro, and tumor growth was studied by nude mice xenografts in vivo. We found that mRNA and protein level of NPY receptor Y1 subtype (NPY1R) significantly decreased in HCC tissues. Low expression of NPY1R closely correlated with poor prognosis in HCC patients. Proliferation of HCC cells was significantly inhibited by recombinant NPY protein in vitro. This inhibitory effect could be blocked by selected NPY1R antagonist BIBP3226. Furthermore, overexpression of NPY1R could significantly inhibit HCC cell proliferation. Knockdown of NPY1R promoted cell multiplication in vitro and increased tumorigenicity and tumor growth in vivo. NPY1R was found to participate in the inhibition of cell proliferation via inactivating mitogen-activated protein kinase signal pathway in HCC cells. Collectively, NPY1R plays an inhibitory role in tumor growth and may be a promising therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MAP Kinase Signaling System/physiology , Receptors, Neuropeptide Y/biosynthesis , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Proliferation/physiology , Down-Regulation , Female , Heterografts , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Real-Time Polymerase Chain Reaction
5.
Cancer Biomark ; 16(1): 161-9, 2016.
Article in English | MEDLINE | ID: mdl-26600397

ABSTRACT

BACKGROUND: It has been shown that circular RNA (circRNA) is associated with human cancers, however, few studies have been reported in hepatocellular carcinoma (HCC). OBJECTIVE: To estimate clinical values of a circular RNA, Hsa_circ_0001649, in HCC. METHODS: Expression level of hsa_circ_0001649 was detected in HCC and paired adjacent liver tissues by real-time quantitative reverse transcription-polymerase chain reactions (qRT-PCRs). Differences in expression level of hsa_circ_0001649 were analyzed using the paired t-test. Tests were performed between clinical information and hsa_circ_0001649 expression level by analysis of variance (ANOVA) or welch t-test and a receiver operating characteristics (ROC) curve was established to estimate the value of hsa_circ_0001649 expression as a biomarker in HCC. RESULTS: hsa_circ_0001649 expression was significantly downregulated in HCC tissues (p = 0.0014) based on an analysis of 89 paired samples of HCC and adjacent liver tissues and the area under the ROC curve (AUC) was 0.63. Furthermore, hsa_circ_0001649 expression was correlated with tumor size (p = 0.045) and the occurrence of tumor embolus (p = 0.017) in HCC. CONCLUSIONS: We first found hsa_circ_0001649 was significantly downregulated in HCC. Our findings indicate hsa_circ_0001649 might serve as a novel potential biomarker for HCC and may function in tumorigenesis and metastasis of HCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA/genetics , Adult , Aged , Biomarkers , Carcinoma, Hepatocellular/diagnosis , Cell Line, Tumor , DNA Helicases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/diagnosis , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , RNA/chemistry , RNA, Circular , ROC Curve , Sequence Analysis, DNA , Ubiquitin-Protein Ligases/genetics
6.
Cell Physiol Biochem ; 37(1): 201-13, 2015.
Article in English | MEDLINE | ID: mdl-26303949

ABSTRACT

BACKGROUND/AIMS: Non-small cell lung carcinoma (NSCLC) is the most common type of lung cancer and the cause of most cancer-related deaths. The molecular mechanisms that are involved in NSCLC development are currently not well understood. Accumulating evidence shows that histone demethylases play important roles in the regulation of pathological developmental processes in many diseases, including various types of cancers. METHODS: Mitochondrial membrane potential assays, migration and invasion assays, caspase-3 and caspase-9 activity assays and western blot analysis were used in this research. RESULTS: We found that overexpression of KDM6B, a demethylase that acts on histone H3 at lysine 27 (H3K27), inhibited cell growth by initiating mitochondria-dependent apoptosis and by attenuating the invasion-metastasis cascade in NSCLC cells. Moreover, our results showed that KDM6B directly interacted with FOXO1 and that overexpression of KDM6B promoted nuclear accumulation of FOXO1. The effects of KDM6B on cell apoptosis and metastasis were weakened by knockdown of FOXO1 expression. On the contrary, knocking down expression of KDM6B inhibited cell apoptosis and promoted cell growth by mitigating the nuclear translocation of FOXO1 in NSCLC cells. CONCLUSIONS: These findings suggest that KDM6B may act in a pro-apoptotic role in NSCLC by causing the nuclear translocation of FOXO1.


Subject(s)
Apoptosis/physiology , Carcinoma, Non-Small-Cell Lung/metabolism , Forkhead Transcription Factors/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lung Neoplasms/metabolism , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Forkhead Box Protein O1 , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Histone Demethylases/metabolism , Histones/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mitochondria/genetics , Mitochondria/metabolism , Protein Transport/genetics
7.
Carcinogenesis ; 36(10): 1201-12, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26249360

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes for cancer related mortality worldwide. Poor prognosis of HCC patients is mainly due to frequent metastasis and recurrence. Deregulation of metastasis suppressors in malignant cells plays critical roles during cancer metastasis. Thus, novel metastasis suppressors are urgently needed to be uncovered to shed new light on molecular mechanisms driving HCC metastasis. In the present study, decreased expression of leukemia inhibitory factor receptor (LIFR) was demonstrated in HCC, and its expression levels were even lower in HCC with metastasis. Downregulated LIFR expression predicted poor prognosis in HCC patients. LIFR was an independent and significant risk factor for their recurrence and survival. Silencing LIFR resulted in forced metastasis of HCC cells, whereas ectopic overexpression of LIFR attenuated migration and invasion of HCC cells in vitro and in vivo. Moreover, LIFR knockdown could activate phosphoinositide 3-kinase/V-akt Murine Thymoma Viral Oncogene Homolog (PI3K/AKT) signaling through enhancing phosphorylation of Janus kinase 1 (JAK1), which successively promoted matrix metalloproteinase 13 (MMP13) expression and HCC metastasis. Combination of LIFR and p-AKT or MMP13 was a more powerful predictor of poor prognosis for HCC patients. Together, these findings conclude that LIFR functions as a novel metastasis suppressor in HCC and may serve as a prognostic biomarker for HCC patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/biosynthesis , Liver Neoplasms/genetics , Matrix Metalloproteinase 13/biosynthesis , Oncogene Protein v-akt/genetics , Adult , Aged , Biomarkers, Tumor , Carcinoma, Hepatocellular/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Janus Kinase 1/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Prognosis , Signal Transduction
8.
Sci Rep ; 5: 10466, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26099564

ABSTRACT

Kynurenine 3-monooxygenase (KMO) is a pivotal enzyme in the kynurenine pathway of tryptophan degradation and plays a critical role in Huntington's and Alzheimer's diseases. This study aimed to examine the expression of KMO in human hepatocellular carcinoma (HCC) and investigate the relationship between its expression and prognosis of HCC patients. We first analyzed KMO expression in 120 paired HCC samples (HCC tissues vs matched adjacent non-cancerous liver tissues), and 205 clinical HCC specimens using immunohistochemistry (IHC). Kaplan-Meier survival and Cox regression analyses were executed to evaluate the prognosis of HCC. The results of IHC analysis showed that KMO expression was significantly higher in HCC tissues than that in normal liver tissues (all p < 0.05). Survival and recurrence analyses showed that KMO was an independent prognostic factor for overall survival (OS) and time to recurrence (TTR) (both p<0.01). And in vitro studies revealed that KMO positively regulated proliferation, migration, and invasion of HCC cells. These results suggest that KMO exhibits tumor-promoting effects towards HCC and it may serve as a novel prognostic marker in HCC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Movement , Kynurenine 3-Monooxygenase/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Aged , Cell Line, Tumor , Cell Proliferation , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/pathology , Prognosis , Proportional Hazards Models , RNA, Small Interfering/metabolism , Up-Regulation
9.
Oncotarget ; 6(9): 6989-99, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25749520

ABSTRACT

Differential diagnosis of well-differentiated hepatocellular carcinoma (WD-HCC) and high-grade dysplastic nodules (HGDNs) represents a challenge for pathologists. Several immunohistochemistry markers have been identified to distinguish hepatocellular carcinoma (HCC) from HGDNs. However, sensitivity or specificity of the individual marker is still limited. In this study, we analyzed dynamic alteration of leukemia inhibitory factor receptor (LIFR) and CD34 during hepatocarcinogenesis from dysplastic nodules to small HCC. The diagnostic performance of LIFR and CD34 combination in WD-HCC and HGDNs was investigated by logistic regression models and validated in an independent validation cohort. LIFR was decreased and CD34 was increased along with stepwise progression of hepatocarcinogenesis from low-grade dysplastic nodules (LGDNs) to small HCC. The sensitivity and specificity of the LIFR and CD34 combination for WD-HCC detection were 93.5% and 90.5%, respectively. In addition, colony formation assay was used to explore the role of LIFR in tumorigenesis. Silencing of LIFR could significantly promote colony formation of HCC cells, whereas ectopic overexpression of LIFR resulted in impaired ability of colony formation of HCC cells. These findings indicate that LIFR and CD34 combination may be used as an available differential diagnostic model for WD-HCC from HGDNs in clinical practice.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Receptors, OSM-LIF/metabolism , Adult , Antigens, CD34/metabolism , Biomarkers, Tumor/metabolism , Carcinogenesis , Cell Differentiation , Cell Line, Tumor , Cell Transformation, Neoplastic , Cohort Studies , Data Mining , Disease Progression , Female , Gene Expression Profiling , Hep G2 Cells , Humans , Male , Middle Aged , Regression Analysis , Sensitivity and Specificity
10.
Oncotarget ; 6(5): 2903-16, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25609201

ABSTRACT

Clusterin (CLU) is a stress-induced chaperone that confers proliferative and survival advantages to cancer cells. However, effects and molecular mechanisms of CLU in hepatocellular carcinoma (HCC) metastasis are still unknown. In this study, HCC tissue array (n = 198) was utilized to investigate correlation between CLU expression and clinicopathological features. Overexpression of CLU in HCC tissues was correlated with shorter overall survival and higher tumor recurrence. In vitro and in vivo assays demonstrated that silencing CLU attenuated the invasion and metastasis of HCC cells, whereas ectopic overexpression of CLU resulted in the forced metastasis of HCC cells. We also revealed that CLU activated Akt signaling through complexing with eukaryotic translation initiation factor 3 subunit I (EIF3I), which in turn promoted matrix metalloproteinase 13 (MMP13) expression and HCC metastasis. Positive correlations between CLU and MMP13, p-Akt, or EIF3I were found in HCC tissues. We further observed that CLU knockdown using the CLU inhibitor OGX-011 significantly suppressed HCC metastasis in two metastatic models through inhibiting EIF3I/Akt/MMP13 signaling. These findings indicate that CLU is an independent predictive factor for prognosis of HCC and it facilitates metastasis through EIF3I/Akt/MMP13 signaling. CLU suppression using OGX-011 may represent a promising therapeutic option for suppressing HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Movement , Clusterin/metabolism , Eukaryotic Initiation Factor-3/metabolism , Liver Neoplasms/enzymology , Matrix Metalloproteinase 13/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Carcinoma, Hepatocellular/therapy , Clusterin/genetics , Eukaryotic Initiation Factor-3/genetics , Female , Hep G2 Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Matrix Metalloproteinase 13/genetics , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Prognosis , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Thionucleotides/genetics , Thionucleotides/metabolism , Time Factors , Transfection , Xenograft Model Antitumor Assays
11.
Mol Cell ; 49(6): 1083-96, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23395002

ABSTRACT

Recently, long noncoding RNAs (lncRNAs) were found to be dysregulated in a variety of tumors. However, it remains unknown how and through what molecular mechanisms the expression of lncRNAs is controlled. In this study, we found that the lncRNA Low Expression in Tumor (lncRNA-LET) was generally downregulated in hepatocellular carcinomas, colorectal cancers, and squamous-cell lung carcinomas. We demonstrated that hypoxia-induced histone deacetylase 3 repressed lncRNA-LET by reducing the histone acetylation-mediated modulation of the lncRNA-LET promoter region. Interestingly, the downregulation of lncRNA-LET was found to be a key step in the stabilization of nuclear factor 90 protein, which leads to hypoxia-induced cancer cell invasion. Moreover, the relationship among hypoxia, histone acetylation disorder, low lncRNA-LET expression level, and metastasis was found in clinical hepatocellular carcinoma samples. These results advance our understanding of the role of lncRNA-LET as a regulator of hypoxia signaling and offer new avenues for therapeutic intervention against cancer progression.


Subject(s)
Carcinoma, Hepatocellular/secondary , Gene Expression Regulation, Neoplastic , Histone Deacetylases/physiology , Liver Neoplasms/pathology , Lung Neoplasms/secondary , RNA, Long Noncoding/genetics , Acetylation , Animals , Base Sequence , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Down-Regulation , Female , Gene Expression , Histones/metabolism , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Male , Mice , Mice, Nude , Middle Aged , Molecular Sequence Data , Neoplasm Transplantation , Nuclear Factor 90 Proteins/genetics , Nuclear Factor 90 Proteins/metabolism , Protein Processing, Post-Translational , RNA, Long Noncoding/metabolism
12.
Carcinogenesis ; 34(3): 577-86, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23222811

ABSTRACT

Although numerous long non-coding RNAs (lncRNAs) have been identified in mammals, many of their biological roles remain to be characterized. Early reports suggest that H19 contributes to carcinogenesis, including hepatocellular carcinoma (HCC). Examination of the Oncomine resource showed that most HCC cases express H19 at a level that is comparable with the liver, with a tendency toward lower expression. This is consistent with our previous microarray data and indicates a more complicated role of H19 in HCC that needs to be characterized. In this study, the expression level of H19 was assessed in different regions of HCC patients' liver samples. Loss- and gain-of-function studies on this lncRNA in the HCC cell lines, SMMC7721 and HCCLM3, were used to characterize its effects on gene expression and to assess its effect on HCC metastasis both in vitro and in vivo. In this study, we show that H19 was underexpressed in intratumoral HCC tissues (T), as compared with peritumoral tissues (L). Additionally, low T/L ratio of H19 predicted poor prognosis. H19 suppressed HCC progression metastasis and the expression of markers of epithelial-to-mesenchymal transition. Furthermore, H19 associated with the protein complex hnRNP U/PCAF/RNAPol II, activating miR-200 family by increasing histone acetylation. The results demonstrate that H19 can alter the miR-200 pathway, thus contributing to mesenchymal-to-epithelial transition and to the suppression of tumor metastasis. These data provide an explanation for the hitherto puzzling literature on the relationship between H19 and cancer, and could suggest the development of combination therapies that target H19 and the miR-200 family.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Epigenesis, Genetic , Liver Neoplasms/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/metabolism , Acetylation , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Proliferation , Disease-Free Survival , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Histones/metabolism , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , Protein Processing, Post-Translational , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Tumor Burden , Up-Regulation
13.
Hepatology ; 56(6): 2231-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22706893

ABSTRACT

UNLABELLED: Survival of patients with hepatocellular carcinoma (HCC) remains poor, which is largely attributed to active angiogenesis. However, the mechanisms underlying angiogenesis in HCC remain to be discovered. In this study, we found that long noncoding RNA associated with microvascular invasion in HCC (lncRNA MVIH) (lncRNA associated with microvascular invasion in HCC) was generally overexpressed in HCC. In a cohort of 215 HCC patients, the overexpression of MVIH was associated with frequent microvascular invasion (P = 0.016) and a higher tumor node metastasis stage (P = 0.009) as well as decreased recurrence-free survival (RFS) (P < 0.001) and overall survival (P = 0.007). Moreover, the up-regulation of MVIH served as an independent risk factor to predict poor RFS. We also found that MVIH could promote tumor growth and intrahepatic metastasis by activating angiogenesis in mouse models. Subsequent investigations indicated that MVIH could activate tumor-inducing angiogenesis by inhibiting the secretion of phosphoglycerate kinase 1 (PGK1). Additionally, in 65 HCC samples, MVIH expression was inversely correlated with the serum level of PGK1 and positively correlated with the microvessel density. CONCLUSION: Deregulation of lncRNA MVIH is a predictor for poor RFS of HCC patients after hepatectomy and could be utilized as a potential target for new adjuvant therapies against active angiogenesis.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , Phosphoglycerate Kinase/metabolism , RNA, Long Noncoding/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Gene Expression , Hepatectomy , Humans , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Mice , Microvessels/pathology , Middle Aged , Neoplasm Invasiveness , Phosphoglycerate Kinase/blood , Predictive Value of Tests , Proportional Hazards Models , Ribosomal Proteins/genetics , Risk Factors , Up-Regulation , alpha-Fetoproteins/metabolism
14.
Hepatology ; 54(6): 2025-35, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21837748

ABSTRACT

UNLABELLED: As an important epigenetic mechanism, histone acetylation modulates the transcription of many genes and plays important roles in hepatocellular carcinoma (HCC). Aberrations in histone acetylation have been observed in HCC, but the factors that contribute to the aberrations have not been fully elucidated. MicroRNAs (miRNAs), which are noncoding RNAs that regulate gene expression, are involved in important epigenetic mechanisms. In this study, we determined that miR-200a and the level of histone H3 acetylation at its promoter were reduced in human HCC tissues in comparison with adjacent noncancerous hepatic tissues. Furthermore, our results suggested that the histone deacetylase 4 (HDAC4) inhibited the expression of miR-200a and its promoter activity and reduced the histone H3 acetylation level at the mir-200a promoter through a Sp1-dependent pathway. Interestingly, we observed that the miR-200a directly targeted the 3'-untranslated region of the HDAC4 messenger RNA and repressed expression of HDAC4. Therefore, miR-200a ultimately induced its own transcription and increased the histone H3 acetylation level at its own promoter. Through targeting HDAC4, miR-200a also induced the up-regulation of total acetyl-histone H3 levels and increased the histone H3 acetylation level at the p21(WAF/Cip1) promoter. Finally, we determined that miR-200a inhibited the proliferation and migration of HCC cells in vivo and in vitro. CONCLUSION: Our findings suggest that the HDAC4/Sp1/miR-200a regulatory network induces the down-regulation of miR-200a and the up-regulation of HDAC4 in HCC. As a result, down-regulation of miR-200a enhances the proliferation and migration of HCC cells and induces aberrant histone acetylation in HCC. These findings highlight a potential therapeutic approach in targeting the HDAC4/Sp1/miR-200a regulatory network for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Histone Deacetylases/physiology , Liver Neoplasms/metabolism , MicroRNAs/physiology , Repressor Proteins/physiology , Sp1 Transcription Factor/physiology , Acetylation , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation , Histones/metabolism , Humans , Liver/metabolism , RNA, Messenger/metabolism , Up-Regulation
15.
Hepatology ; 54(5): 1679-89, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21769904

ABSTRACT

UNLABELLED: In recent years, long noncoding RNAs (lncRNAs) have been shown to have critical regulatory roles in cancer biology. However, the contributions of lncRNAs to hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) remain largely unknown. Differentially expressed lncRNAs between HBV-related HCC and paired peritumoral tissues were identified by microarray and validated using quantitative real-time polymerase chain reaction. Liver samples from patients with HBV-related HCC were analyzed for levels of a specific differentially expressed lncRNA High Expression In HCC (termed lncRNA-HEIH); data were compared with survival data using the Kaplan-Meier method and compared between groups by the log-rank test. The effects of lncRNA-HEIH were assessed by silencing and overexpressing the lncRNA in vitro and in vivo. The expression level of lncRNA-HEIH in HBV-related HCC is significantly associated with recurrence and is an independent prognostic factor for survival. We also found that lncRNA-HEIH plays a key role in G(0) /G(1) arrest, and further demonstrated that lncRNA-HEIH was associated with enhancer of zeste homolog 2 (EZH2) and that this association was required for the repression of EZH2 target genes. CONCLUSIONS: Together, these results indicate that lncRNA-HEIH is an oncogenic lncRNA that promotes tumor progression and leads us to propose that lncRNAs may serve as key regulatory hubs in HCC progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , Liver Neoplasms/genetics , RNA, Untranslated/genetics , Transcription Factors/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Cycle/genetics , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Prognosis , Repressor Proteins/genetics
16.
Neoplasia ; 13(7): 590-600, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21750653

ABSTRACT

The loss of microRNA-122 (miR-122) expression correlates to many characteristic properties of hepatocellular carcinoma (HCC) cells, including clonogenic survival, anchorage-independent growth, migration, invasion, epithelial-mesenchymal transition, and tumorigenesis. However, all of these findings do not sufficiently explain the oncogenic potential of miR-122. In the current study, we used two-dimensional differential in-gel electrophoresis to measure changes in the expression of thousands of proteins in response to the inhibition of miR-122 in human hepatoma cells. Several proteins that were upregulated on miR-122 inhibition were involved in the unfolded protein response (UPR) pathway. The overexpression of miR-122 resulted in the repression of UPR pathway activation. Therefore, miR-122 may act as an inhibitor of the chaperone gene expression and negatively regulate the UPR pathway in HCC. We further showed that the miR-122 inhibitor enhanced the stability of the 26S proteasome non-ATPase regulatory subunit 10 (PSMD10) through the up-regulation of its target gene cyclin-dependent kinase 4 (CDK4). This process may activate the UPR pathway to prevent chemotherapy-mediated tumor cell apoptosis. The current study suggests that miR-122 negatively regulates the UPR through the CDK4-PSMD10 pathway. The down-regulation of miR-122 activated the CDK4-PSMD10-UPR pathway to decrease tumor cell anticancer drug-mediated apoptosis. We identified a new HCC therapeutic target and proclaimed the potential risk of the therapeutic use of miR-122 silencing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular , Drug Resistance, Neoplasm/genetics , Liver Neoplasms , MicroRNAs/physiology , Unfolded Protein Response/physiology , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...