Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Prenat Diagn ; 43(10): 1366-1369, 2023 09.
Article in English | MEDLINE | ID: mdl-37529930

ABSTRACT

Prenatal testing was performed in a 39-year-old Chinese pregnant woman referred for increased nuchal translucency measuring 5.7 mm. Non-invasive prenatal testing and SNP array study on amniotic fluid samples were normal. Whole exome sequencing (WES) was initiated further as the fetus had pericardial effusion of 1.2 mm, thickened myocardium over the right ventricular lateral wall and aberrant right subclavian artery. A detailed fetal echocardiogram also revealed persistent left superior vena cava and dilated coronary sinus at 20 weeks. From whole exome sequencing of the trio, a de novo heterozygous variant NM_005359.5(SMAD4): c.1499T>C (p.Ile500Thr) was detected. This pathogenic variant has been reported in the postnatal case cohort of Myhre syndrome. This condition is characterized by facial dysmorphism, intellectual disability, hearing loss, skeletal abnormalities and potential life threatening respiratory or cardiovascular manifestations. Termination of pregnancy was performed at 23 weeks. Small chins, pre-axial polydactyly, brachydactyly and clinodactyly were noted in the abortus. Ultrasound findings of increased nuchal translucency, thickened myocardium and pericardial effusion prompted further genetic evaluation for the prenatal diagnosis of Myhre syndrome by whole exome sequencing.


Subject(s)
Heart Defects, Congenital , Intellectual Disability , Pericardial Effusion , Pregnancy , Female , Humans , Adult , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Nuchal Translucency Measurement , Vena Cava, Superior , Prenatal Diagnosis , Ultrasonography, Prenatal , Smad4 Protein/genetics
2.
Int J Pediatr Endocrinol ; 2020: 13, 2020.
Article in English | MEDLINE | ID: mdl-32670376

ABSTRACT

BACKGROUND: Beckwith-Wiedemann syndrome (BWS) is an overgrowth syndrome with variable clinical phenotype and complex molecular aetiology. It is mainly caused by dysregulation of the chromosome 11p15 imprinted region, which results in overgrowth in multiple tissues, often in a mosaic manner. CASE PRESENTATION: A large-for-gestational-age infant without any other somatic features of BWS presented with medically refractory hyperinsulinism (HI) requiring 80% pancreatectomy. Next generation sequencing with congenital HI sequencing panel identified a pathogenic ABCC8:c.1792C > T (p.Arg598Ter) variant of paternal origin, suggestive of focal HI. However, pancreatic histology revealed atypical findings of coalescing nests and trabeculae of adenomatosis scattered with islets with isolated enlarged, hyperchromatic nuclei scattered throughout the pancreas. Methylation analysis, SNP-based chromosomal microarray and short tandem repeat markers analysis revealed mosaic segmental paternal uniparental disomy (UPD) 11p15.5-p15.1 in the pancreatic tissue, but not the peripheral blood, suggestive of BWS/BW-spectrum HI. CONCLUSIONS: This case highlights the importance of integrating the clinical presentation and subsequent clinical course, together with radiological, genetic and histological findings in the definitive diagnosis of this rare yet clinically important entity. In addition, this is the first report that demonstrated the level of paternal inherited c.1792 T pathogenic variant in the pancreatic tissue being directly correlated to the mosaic level of pUPD.

3.
Front Genet ; 11: 620162, 2020.
Article in English | MEDLINE | ID: mdl-33584815

ABSTRACT

Balanced chromosomal abnormalities (BCAs) are changes in the localization or orientation of a chromosomal segment without visible gain or loss of genetic material. BCAs occur at a frequency of 1 in 500 newborns and are associated with an increased risk of multiple congenital anomalies and/or neurodevelopmental disorders, especially if it is a de novo mutation. In this pilot project, we used short read genome sequencing (GS) to retrospectively re-sequence ten prenatal subjects with de novo BCAs and compared the performance of GS with the original karyotyping. GS characterized all BCAs found by conventional karyotyping with the added benefit of precise sub-band delineation. By identifying BCA breakpoints at the nucleotide level using GS, we found disruption of OMIM genes in three cases and identified cryptic gain/loss at the breakpoints in two cases. Of these five cases, four cases reached a definitive genetic diagnosis while the other one case had a BCA interpreted as unknown clinical significance. The additional information gained from GS can change the interpretation of the BCAs and has the potential to improve the genetic counseling and perinatal management by providing a more specific genetic diagnosis. This demonstrates the added clinical utility of using GS for the diagnosis of BCAs.

4.
J Hepatol ; 67(4): 758-769, 2017 10.
Article in English | MEDLINE | ID: mdl-28532996

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is a major leading cause of cancer mortality worldwide. Epigenetic deregulation is a common trait of human HCC. G9s is an important epigenetics regulator however, its role in liver carcinogenesis remains to be investigated. METHODS: Gene expressions were determined by RNA-Seq and qRT-PCR. G9a knockdown and knockout cell lines were established by lentiviral-based shRNA and CRISPR/Cas9 gene editing system. Tumor-promoting functions of G9a was studied in both HCC cell lines and nude mice model. The downstream targets of G9a were identified by RNA-Seq and confirmed by ChIP assay. The therapeutic value of G9a inhibitors was evaluated both in vitro and in vivo. RESULTS: We identified G9a as a frequently upregulated histone methyltransferase in human HCCs. Upregulation of G9a was significantly associated with HCC progression and aggressive clinicopathological features. Functionally, we demonstrated that inactivation of G9a by RNAi knockdown, CRISPR/Cas9 knockout, and pharmacological inhibition remarkably abolished H3K9 di-methylation and suppressed HCC cell proliferation and metastasis in both in vitro and in vivo models. Mechanistically, we showed that the frequent upregulation of G9a in human HCCs was attributed to gene copy number gain at chromosome 6p21. In addition, we identified miR-1 as a negative regulator of G9a. Loss of miR-1 relieved the post-transcriptional repression on G9a and contributed to its upregulation in human HCC. Utilizing RNA sequencing, we identified the tumor suppressor RARRES3 as a critical target of G9a. Epigenetic silencing of RARRES3 contributed to the tumor-promoting function of G9a. CONCLUSION: This study shows a frequent deregulation of miR-1/G9a/RARRES3 axis in liver carcinogenesis, highlighting the pathological significance of G9a and its therapeutic potential in HCC treatment. Lay summary: In this study, we identified G9a histone methyltransferase was frequently upregulated in human HCC and contributes to epigenetic silencing of tumor suppressor gene RARRES3 in liver cancer. Targeting G9a may be a novel approach for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Receptors, Retinoic Acid/genetics , 3' Untranslated Regions , Animals , Carcinoma, Hepatocellular/etiology , Cell Line, Tumor , Epigenesis, Genetic , Gene Dosage , Gene Knockdown Techniques , Gene Knockout Techniques , Gene Silencing , Genes, Tumor Suppressor , Humans , Liver Neoplasms/etiology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/genetics , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Up-Regulation
5.
Hepatology ; 63(2): 474-87, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481868

ABSTRACT

UNLABELLED: Epigenetic deregulation plays an important role in liver carcinogenesis. Using transcriptome sequencing, we examined the expression of 591 epigenetic regulators in hepatitis B-associated human hepatocellular carcinoma (HCC). We found that aberrant expression of epigenetic regulators was a common event in HCC. We further identified SETDB1 (SET domain, bifurcated 1), an H3K9-specific histone methyltransferase, as the most significantly up-regulated epigenetic regulator in human HCCs. Up-regulation of SETDB1 was significantly associated with HCC disease progression, cancer aggressiveness, and poorer prognosis of HCC patients. Functionally, we showed that knockdown of SETDB1 reduced HCC cell proliferation in vitro and suppressed orthotopic tumorigenicity in vivo. Inactivation of SETDB1 also impeded HCC cell migration and abolished lung metastasis in nude mice. Interestingly, SETDB1 protein was consistently up-regulated in all metastatic foci found in different organs, suggesting that SETDB1 was essential for HCC metastatic progression. Mechanistically, we showed that the frequent up-regulation of SETDB1 in human HCC was attributed to the recurrent SETDB1 gene copy gain at chromosome 1q21. In addition, hyperactivation of specificity protein 1 transcription factor in HCC enhanced SETDB1 expression at the transcriptional level. Furthermore, we identified miR-29 as a negative regulator of SETDB1. Down-regulation of miR-29 expression in human HCC contributed to SETDB1 up-regulation by relieving its post-transcriptional regulation. CONCLUSION: SETDB1 is an oncogene that is frequently up-regulated in human HCCs; the multiplicity of SETDB1 activating mechanisms at the chromosomal, transcriptional, and posttranscriptional levels together facilitates SETDB1 up-regulation in human HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Protein Methyltransferases/genetics , Up-Regulation , Animals , Cells, Cultured , Disease Progression , Epigenesis, Genetic , Histone Methyltransferases , Humans , Male , Mice , Mice, Nude
6.
Front Med ; 9(3): 331-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26293610

ABSTRACT

MicroRNAs (miRNAs), an important class of small non-coding RNAs, regulate gene expression at the post-transcriptional level. miRNAs are involved in a wide range of biological processes and implicated in different diseases, including cancers. In this study, miRNA profiling and qRT-PCR validation revealed that miR-142-3p and miR-142-5p were significantly downregulated in hepatocellular carcinoma (HCC) and their expression levels decreased as the disease progressed. The ectopic expression of miR-142 significantly reduced HCC cell migration and invasion. Overexpression of either miR-142-3p or miR-142-5p suppressed HCC cell migration, and overexpression of both synergistically inhibited cell migration, which indicated that miR-142-3p and miR-142-5p may cooperatively regulate cell movement. miR-142-3p and miR-142-5p, which are mature miRNAs derived from the 3'- and 5'-strands of the precursor miR-142, target distinct pools of genes because of their different seed sequences. Pathway enrichment analysis showed a strong association of the putative gene targets of miR-142-3p and miR-142-5p with several cell motility-associated pathways, including those regulating actin cytoskeleton, adherens junctions, and focal adhesion. Importantly, a number of the putative gene targets were also significantly upregulated in human HCC cells. Moreover, overexpression of miR-142 significantly abrogated stress fiber formation in HCC cells and led to cell shrinkage. This study shows that mature miR-142 pairs collaboratively regulate different components of distinct signaling cascades and therefore affects the motility of HCC cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Movement/genetics , Liver Neoplasms/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans
7.
Oncotarget ; 6(15): 13658-70, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25909223

ABSTRACT

MiR-200 family is an important regulator of epithelial-mesenchymal transition and has been implicated in human carcinogenesis. However, their expression and functions in human cancers remain controversial. In the work presented here, we showed that miR-200 family members were frequently down-regulated in hepatocellular carcinoma (HCC). Although all five members of miR-200 family inhibited ZEB1/2 expression in HCC cell lines, we showed that overexpression only of the miR-200b/200c/429 subfamily, but not the miR-200a/141 subfamily, resulted in impeded HCC cell migration. Further investigations led to the identification of RhoA and ROCK2 as specific down-stream targets of the miR-200b/200c/429 subfamily. We demonstrated that the miR-200b/200c/429 subfamily inhibited HCC cell migration through modulating Rho/ROCK mediated cell cytoskeletal reorganization and cell-substratum adhesion. Re-expression of miR-200b significantly suppressed lung metastasis of HCC cells in an orthotopic liver implantation model in vivo. In conclusion, our findings identified the miR-200b/200c/429 subfamily as metastasis suppressor microRNAs in human HCC and highlighted the functional discrepancy among miR-200 family members.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Metastasis , Signal Transduction , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
8.
PLoS One ; 9(12): e115036, 2014.
Article in English | MEDLINE | ID: mdl-25541689

ABSTRACT

Hepatocellular carcinoma (HCC) is an aggressive tumor, with a high mortality rate due to late symptom presentation and frequent tumor recurrences and metastasis. It is also a rapidly growing tumor supported by different metabolic mechanisms; nevertheless, the biological and molecular mechanisms involved in the metabolic reprogramming in HCC are unclear. In this study, we found that pyruvate kinase M2 (PKM2) was frequently over-expressed in human HCCs and its over-expression was associated with aggressive clinicopathological features and poor prognosis of HCC patients. Furthermore, knockdown of PKM2 suppressed aerobic glycolysis and cell proliferation in HCC cell lines in vitro. Importantly, knockdown of PKM2 hampered HCC growth in both subcutaneous injection and orthotopic liver implantation models, and reduced lung metastasis in vivo. Of significance, PKM2 over-expression in human HCCs was associated with a down-regulation of a liver-specific microRNA, miR-122. We further showed that miR-122 interacted with the 3UTR of the PKM2 gene. Re-expression of miR-122 in HCC cell lines reduced PKM2 expression, decreased glucose uptake in vitro, and suppressed HCC tumor growth in vivo. Our clinical data and functional studies have revealed a novel biological mechanism involved in HCC metabolic reprogramming.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Membrane Proteins/metabolism , MicroRNAs/genetics , Thyroid Hormones/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Glycolysis , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental , Lung Neoplasms/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Nude , MicroRNAs/metabolism , Prognosis , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins
9.
Hepatology ; 60(5): 1645-58, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25048396

ABSTRACT

UNLABELLED: Poor prognosis of cancers, including hepatocellular carcinoma (HCC), is mainly associated with metastasis; however, the underlying mechanisms remain poorly understood. This article investigates the role of lysyl oxidase-like 2 (LOXL-2) in the biology of HCC metastasis. First, we showed that HCC metastasis relies on a collagen-modifying enzyme, LOXL2, which was significantly overexpressed in tumorous tissues and sera of HCC patients, indicating that LOXL2 may be a good diagnostic marker for HCC patients. Second, we delineated a complex, interlinked signaling network that involves multiple regulators, including hypoxia, transforming growth factor beta (TGF-ß), and microRNAs (miRNAs), converging to control the expression of LOXL2. We found not only that LOXL2 was regulated by hypoxia/hypoxia-inducible factor 1 alpha (HIF-1α), but also that TGF-ß activated LOXL2 transcription through mothers against decapentaplegic homolog 4 (Smad4), whereas two frequently underexpressed miRNA families, miR-26 and miR-29, cooperatively suppressed LOXL2 transcription through interacting with the 3' untranslated region of LOXL2. Third, we demonstrated the imperative roles of LOXL2 in modifying the extracellular matrix components in the tumor microenvironment and metastatic niche of HCC. LOXL2 promoted intrahepatic metastasis by increasing tissue stiffness, thereby enhancing the cytoskeletal reorganization of HCC cells. Furthermore, LOXL2 facilitated extrahepatic metastasis by enhancing recruitment of bone-marrow-derived cells to the metastatic site. CONCLUSION: These findings integrate the clinical relevance, molecular regulation, and functional implications of LOXL2 in HCC metastasis.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms, Experimental/enzymology , Animals , Case-Control Studies , Cell Adhesion , Cell Line, Tumor , Collagen/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neoplasm Metastasis , Smad4 Protein/metabolism , Tumor Microenvironment
10.
PLoS One ; 8(6): e68226, 2013.
Article in English | MEDLINE | ID: mdl-23826380

ABSTRACT

Enhancer of zeste homolog 2 (EZH2), the histone methyltransferase of the Polycomb Repressive complex 2 catalyzing histone H3 lysine 27 tri-methylation (H3K27me3), is frequently up-regulated in human cancers. In this study, we identified the tumor suppressor Deleted in liver cancer 1 (DLC1) as a target of repression by EZH2-mediated H3K27me3. DLC1 is a GTPase-activating protein for Rho family proteins. Inactivation of DLC1 results in hyper-activated Rho/ROCK signaling and is implicated in actin cytoskeleton reorganization to promote cancer metastasis. By chromatin immunoprecipitation assay, we demonstrated that H3K27me3 was significantly enriched at the DLC1 promoter region of a DLC1-nonexpressing HCC cell line, MHCC97L. Depletion of EZH2 in MHCC97L by shRNA reduced H3K27me3 level at DLC1 promoter and induced DLC1 gene re-expression. Conversely, transient overexpression of GFP-EZH2 in DLC1-expressing Huh7 cells reduced DLC1 mRNA level with a concomitant enrichment of EZH2 on DLC1 promoter. An inverse relation between EZH2 and DLC1 expression was observed in the liver, lung, breast, prostate, and ovarian cancer tissues. Treating cancer cells with the EZH2 small molecular inhibitor, 3-Deazaneplanocin A (DZNep), restored DLC1 expression in different cancer cell lines, indicating that EZH2-mediated H3K27me3 epigenetic regulation of DLC1 was a common mechanism in human cancers. Importantly, we found that DZNep treatment inhibited HCC cell migration through disrupting actin cytoskeleton network, suggesting the therapeutic potential of DZNep in targeting cancer metastasis. Taken together, our study has shed mechanistic insight into EZH2-H3K27me3 epigenetic repression of DLC1 and advocated the significant pro-metastatic role of EZH2 via repressing tumor and metastasis suppressors.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenetic Repression , GTPase-Activating Proteins/metabolism , Histones/metabolism , Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Methylation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Tumor Suppressor Proteins/genetics
11.
Front Med ; 7(2): 231-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23620257

ABSTRACT

Hepatocellular carcinoma (HCC) development is characterized by the presence of epigenetic alterations, including promoter DNA hypermethylation and post-translational modifications of histone, which profoundly affect expression of a wide repertoire of genes critical for cancer development. Emerging data suggest that deregulation of polycomb group (PcG) proteins, which are key chromatin modifiers repressing gene transcription during developmental stage, plays a causative role in oncogenesis. PcG proteins assemble into polycomb repressive complex 1 (PRC1) and polycomb repressive complex 2 (PRC2) to impose the histone H3 lysine 27 trimethylation (H3K27me3) modification for repression. In this review, we will first recapitulate the mechanisms of two key epigenetic pathways: DNA methylation and histone modifications. Specifically, we will focus our discussion on the molecular roles of PcG proteins. Next, we will highlight recent findings on PcG proteins, their clinicopathological implication and their downstream molecular consequence in hepatocarcinogenesis. Last but not least, we will consider the therapeutic potential of targeting enhancer of zeste homolog 2 (EZH2) as a possible treatment for HCC. Improving our understanding on the roles of PcG proteins in hepatocarcinogenesis can benefit the development of epigenetic-based therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Epigenomics/methods , Liver Neoplasms/genetics , Polycomb-Group Proteins/genetics , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Polycomb-Group Proteins/metabolism
12.
Hepatology ; 57(2): 637-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22991213

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a major liver malignancy. We previously demonstrated that deregulation of epigenetic regulators is a common event in human HCC. Suppressor of variegation 3-9 homolog 1 (SUV39H1), the prototype of histone methyltransferase, is the major enzyme responsible for histone H3 lysine 9 trimethylation, which, essentially, is involved in heterochromatin formation, chromosome segregation, and mitotic progression. However, the implication of SUV39H1 in hepatocarcinogenesis remains elusive. In this study, we found that SUV39H1 was frequently up-regulated in human HCCs and was significantly associated with increased Ki67 expression (P < 0.001) and the presence of venous invasion (P = 0.017). To investigate the role of SUV39H1 in HCC development, both gain- and loss-of-function models were established. SUV39H1 overexpression remarkably enhanced HCC cell clonogenicity, whereas knockdown of SUV39H1 substantially suppressed HCC cell proliferation and induced cell senescence. In addition, ectopic expression of SUV39H1 increased the migratory ability of HCC cells, whereas a reduced migration rate was observed in SUV39H1 knockdown cells. The significance of SUV39H1 in HCC was further demonstrated in a nude mice model; SUV39H1 knockdown drastically inhibited in vivo tumorigenicity and abolished pulmonary metastasis of HCC cells. We also identified microRNA-125b (miR-125b) as a post-transcriptional regulator of SUV39H1. Ectopic expression of miR-125b inhibited SUV39H1 3'-untranslated-region-coupled luciferase activity and suppressed endogenous SUV39H1 expression at both messenger RNA and protein levels. We have previously reported frequent down-regulation of miR-125b in HCC. Interestingly, miR-125b level was found to be inversely correlated with SUV39H1 expression (P = 0.001) in clinical specimens. Our observations suggested that miR-125b down-regulation may account for the aberrant SUV39H1 level in HCC. CONCLUSION: Our study demonstrated that SUV39H1 up-regulation contributed to HCC development and metastasis. The tumor-suppressive miR-125b served as a negative regulator of SUV39H1.


Subject(s)
Methyltransferases/physiology , MicroRNAs/physiology , Repressor Proteins/physiology , Animals , Carcinoma, Hepatocellular/genetics , Cell Movement/genetics , Cell Proliferation , Disease Progression , Histone-Lysine N-Methyltransferase , Humans , Liver Neoplasms/genetics , Mice , Mice, Nude , Up-Regulation
13.
Hepatology ; 56(2): 622-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22370893

ABSTRACT

UNLABELLED: Epigenetic alterations and microRNA (miRNA) deregulation are common in hepatocellular carcinoma (HCC). The histone H3 lysine 27 (H3K27) tri-methylating enzyme, enhancer of zeste homolog 2 (EZH2) mediates epigenetic silencing of gene expression and is frequently up-regulated in human cancers. In this study we aimed to delineate the implications of EZH2 up-regulation in miRNA deregulation and HCC metastasis. Expressions of a total of 90 epigenetic regulators were first determined in 38 pairs of primary HCCs and their corresponding nontumorous livers. We identified EZH2 and its associated polycomb repressive complex 2 (PRC2) as one of the most significantly deregulated epigenetic regulators in primary HCC samples. Up-regulation of EZH2 was next confirmed in 69.5% (41/59) of primary HCCs. Clinicopathologically, EZH2 up-regulation was associated with HCC progression and multiple HCC metastatic features, including venous invasion (P = 0.043), direct liver invasion (P = 0.014), and absence of tumor encapsulation (P = 0.043). We further demonstrated that knockdown of EZH2 in HCC cell lines reduced the global levels of tri-methylated H3K27, and suppressed HCC motility in vitro and pulmonary metastasis in a nude mouse model. By interrogating the miRNA expression profile in EZH2-knockdown cell lines and primary HCC samples, we identified a subset of miRNA that was epigenetically suppressed by EZH2 in human HCC. These included well-characterized tumor-suppressor miRNAs, such as miR-139-5p, miR-125b, miR-101, let-7c, and miR-200b. Pathway enrichment analysis revealed a common regulatory role of these EZH2-silenced miRNAs in modulating cell motility and metastasis-related pathways. Our findings suggest that EZH2 exerts its prometastatic function by way of epigenetic silencing of multiple tumor suppressor miRNAs. CONCLUSION: Our study demonstrated that EZH2 epigenetically silenced multiple miRNAs that negatively regulate HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , DNA-Binding Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , Transcription Factors/genetics , Animals , Cell Movement/physiology , Computer Simulation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/genetics , Gene Knockdown Techniques , Gene Silencing/physiology , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/genetics , Transplantation, Heterologous
14.
Hepatology ; 55(5): 1453-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22135159

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a prevalent cancer with an extremely high mortality rate attributed to HCC metastasis, which is the major cause of tumor recurrence and organ failure. Presence of tumor thrombi in the portal veins (venous metastases) is a clinicopathological feature of metastatic HCCs. In this study, we analyzed the microRNA (miRNA) expression profiles of nontumorous livers, primary HCCs, and venous metastases in the same livers from 20 HCC patients by way of TaqMan low-density array (TLDA) and identified the precise alterations of miRNA expression from nontumorous livers to primary HCCs and venous metastases globally. By unsupervised clustering analysis, nontumorous livers were distinctly segregated from primary HCCs and venous metastases, whereas no discernible difference in the expression pattern could be found between primary HCCs and venous metastases. However, a marked global reduction of miRNA expression levels was detected in venous metastases, as compared with primary HCCs. These data suggest that miRNA deregulation is an early event in liver carcinogenesis and the later global miRNA down-regulation aggravates the preexisting miRNA deregulation to further promote HCC metastasis. CONCLUSION: Our study has enriched the current understanding of the deregulation of miRNAs in HCC progression and highlighted the sequential and distinctive alterations of miRNA expression in primary HCC and venous metastasis formation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Vascular Neoplasms/secondary , Biopsy, Needle , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cluster Analysis , Cohort Studies , Databases, Factual , Disease Progression , Down-Regulation , Female , Humans , Immunohistochemistry , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , MicroRNAs/metabolism , Neoplasm Staging , Neoplastic Cells, Circulating/pathology , Paraffin Embedding , Prognosis , Real-Time Polymerase Chain Reaction , Reference Values , Retrospective Studies , Risk Assessment , Survival Analysis , Vascular Neoplasms/genetics , Vascular Neoplasms/mortality , Veins/pathology
15.
PLoS One ; 6(2): e16702, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21347439

ABSTRACT

DNA methylation and histone modifications are two major epigenetic events regulating gene expression and chromatin structure, and their alterations are linked to human carcinogenesis. DNA methylation plays an important role in tumor suppressor gene inactivation, and can be revised by DNA methylation inhibitors. The reversible nature of DNA methylation forms the basis of epigenetic cancer therapy. However, it has been reported that DNA re-methylation and gene re-silencing could occur after removal of demethylation treatment and this may significantly hamper the therapeutic value of DNA methylation inhibitors. In this study we have provided detailed evidence demonstrating that mammalian cells possess a bona fide DNA methylation recovery system. We have also shown that DNA methylation recovery was mediated by the major human DNA methyltransferase, DNMT1. In addition, we found that H3K9-tri-methylation and H3K27-tri-methylation were closely associated with this DNA methylation recovery. These persistent transcriptional repressive histone modifications may have a crucial role in regulating DNMT1-mediated DNA methylation recovery. Our findings may have important implications towards a better understanding of epigenetic regulation and future development of epigenetic therapeutic intervention.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Gene Silencing , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Transcription, Genetic/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation/drug effects , Decitabine , GTPase-Activating Proteins/genetics , Gene Silencing/drug effects , Genetic Loci/genetics , HCT116 Cells , HeLa Cells , Humans , Stochastic Processes , Transcription, Genetic/drug effects , Tumor Suppressor Proteins/genetics
16.
Gastroenterology ; 140(1): 322-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20951699

ABSTRACT

BACKGROUND & AIMS: We investigated mechanisms of hepatocellular carcinoma (HCC) metastasis and identified an antimetastatic microRNA (miRNA), miR-139, that is down-regulated in human HCC samples. METHODS: Effects of stable and transient expression of miRNA-139 and its inhibitors were studied in the human HCC cell lines SMMC-7721 and BEL7402; cells were analyzed for migration and invasion. Liver samples from patients with metastatic HCC were analyzed for levels of miRNA-139; data were compared with survival data using the Kaplan-Meier method and compared between groups by the log-rank test. Tumor formation and metastasis from human HCC MHCC97L cells that did or did not express miR-139 were analyzed in mice. RESULTS: Down-regulation of miR-139 in HCC was associated significantly with poor prognosis of patients and features of metastatic tumors, including venous invasion, microsatellite formation, absence of tumor encapsulation, and reduced differentiation. miR-139 expression was reduced in metastatic HCC tumors compared with primary tumors. Overexpression of miR-139 in HCC cells significantly reduced cell migration and invasion in vitro and the incidence and severity of lung metastasis from orthotopic liver tumors in mice. miR-139 interacted with the 3' untranslated region of Rho-kinase 2 (ROCK2) and reduced its expression in HCC cells. Levels of miR-139 were correlated inversely with ROCK2 protein in human HCC samples. Overexpression of miR-139 did not inhibit HCC cell motility when ROCK2 was knocked down. CONCLUSIONS: The microRNA miR-139 interacts with ROCK2 and reduces its expression in HCC cells. Down-regulation of miR-139 increased the invasive abilities of HCC cells in vitro and HCC metastasis in vivo. Expression of miR-139 is reduced in human metastatic HCC samples and correlates with prognosis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , rho-Associated Kinases/metabolism , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Colorectal Neoplasms/secondary , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Transplantation , rho-Associated Kinases/analysis
17.
Liver Int ; 30(5): 642-56, 2010 May.
Article in English | MEDLINE | ID: mdl-20726051

ABSTRACT

Hepatocellular carcinoma (HCC) is an intractable disease with an extremely high mortality rate. Metastasis is the major factor of liver failure, tumour recurrence and death in HCC patients. Unfortunately, no promising curative therapy for HCC metastasis is available as yet; therefore, treatment for advanced HCC still remains a formidable challenge. A large body of evidence has demonstrated that the RhoGTPases/Rho-effector pathway plays important roles in mediating HCC metastasis based on their foremost functions in orchestrating the cell cytoskeletal reorganization. This review will first discuss the general principles of cancer metastasis and cancer cell movement with a particular focus on HCC.We will then summarize the implications of various members in the RhoGTPases/Rho-effectors signalling cascade including the upstream RhoGTPase regulators RhoGTPases and Rho-effectors and their downstream targets in HCC metastasis. Finally, we will discuss the therapeutic insight of targeting the RhoGTPases/Rho-effector pathway in HCC. Taken together, the literature demonstrates the importance of the RhoGTPases/Rho-effector signalling pathway in HCC metastasis and marks the necessity to have a more thorough knowledge of this complicated signalling network in order to develop novel therapeutic strategies for HCC patients.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , rho GTP-Binding Proteins/metabolism , Cell Movement , Humans , Neoplasm Metastasis , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...