Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Mol Carcinog ; 58(5): 643-653, 2019 05.
Article in English | MEDLINE | ID: mdl-30575099

ABSTRACT

Cytidine deaminase APOBEC3B (A3B) is known to play important roles in creating de novo genomic C-to-T mutations in cancers and contribute to induction of genomic instability. Our study evaluated the roles of A3B in the progression and metastasis of human hepatocellular carcinoma (HCC). Using whole-transcriptome and whole-exome sequencing, and quantitative PCR, we found that A3B was overexpressed in human HCCs and A3B expression was significantly correlated with the proportion of genomic C-to-A and G-to-T mutations. Upon clinicopathological correlation, higher A3B expression was associated with more aggressive tumor behavior. Wild-type A3B (wt-A3B) overexpression in HCC cells promoted cell proliferation, and cell migratory and invasive abilities in vitro, and tumorigenicity and metastasis in vivo. On the other hand, knockdown of A3B suppressed cell proliferation, migratory, and invasive abilities of HCC cells with high endogenous A3B level. However, to our surprise, overexpression of A3B deaminase-dead double mutant (E68A/E255Q) led to similar results as wt-A3B in HCC. Furthermore, overexpression of wt-A3B and mutant A3B both enhanced cell cycle progression in HCC cells. Altogether, our data demonstrated a novel deaminase-independent role of A3B in contributing to HCC tumorigenesis and metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Cytidine Deaminase/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Minor Histocompatibility Antigens/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Cytidine Deaminase/genetics , Deamination , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Minor Histocompatibility Antigens/genetics , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Hepatology ; 66(1): 152-166, 2017 07.
Article in English | MEDLINE | ID: mdl-28295457

ABSTRACT

Adjuvant interferon-α (IFN-α) therapy is used to control certain types of cancer in clinics. For hepatocellular carcinoma (HCC), IFN-α therapy is effective in only a subgroup of patients; therefore, identifying biomarkers to predict the response to IFN-α therapy is of high significance and clinical utility. As the induced IFN-stimulated gene expression following IFN-α treatment plays pivotal roles in IFN-α effects, we screened IFN-stimulated gene expression in HCC tissues and found that several IFN-stimulated genes were significantly decreased in HCC. Interestingly, expression of IFN-induced protein with tetratricopeptide repeats (IFIT) family members, including IFIT1, IFIT2, IFIT3, and IFIT5, was decreased in HCC tissues. We further analyzed the expression of IFIT family members in HCC and their roles in patients' responses to IFN-α therapy in two independent randomized controlled IFN-α therapy clinical trials of HCC patients. We found that higher expression of IFIT3, but not other IFITs, in HCC tissues predicts better response to IFN-α therapy, suggesting that IFIT3 may be a useful predictor of the response to IFN-α therapy in HCC patients. Mechanistically, IFIT3 enhanced the antitumor effects of IFN-α by promoting IFN-α effector responses both in vitro and in vivo. IFIT3 could bind signal transducer and activator of transcription 1 (STAT1) and STAT2 to enhance STAT1-STAT2 heterodimerization and nuclear translocation upon IFN-α treatment, thus promoting IFN-α effector signaling. CONCLUSION: Higher IFIT3 expression in HCC tissues predicts better response to IFN-α therapy in HCC patients; IFIT3 promotes IFN-α effector responses and therapeutic effects by strengthening IFN-α effector signaling in HCC. (Hepatology 2017;66:152-166).


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/drug therapy , Interferon-alpha/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/drug therapy , Adult , Aged , Analysis of Variance , Biopsy, Needle , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cohort Studies , Disease-Free Survival , Female , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Risk Assessment , Survival Rate , Treatment Outcome
3.
Gut ; 66(2): 342-351, 2017 02.
Article in English | MEDLINE | ID: mdl-26669617

ABSTRACT

OBJECTIVE: The nature of the tumour-infiltrating leucocytes (TILs) is known to impact clinical outcome in carcinomas, including hepatocellular carcinoma (HCC). However, the role of tumour-infiltrating B cells (TIBs) remains controversial. Here, we investigate the impact of TIBs and their interaction with T cells on HCC patient prognosis. DESIGN: Tissue samples were obtained from 112 patients with HCC from Singapore, Hong Kong and Zurich and analysed using immunohistochemistry and immunofluorescence. RNA expression of CD19, CD8A, IFNG was analysed using quantitative PCR. The phenotype of freshly isolated TILs was analysed using flow cytometry. A mouse model depleted of mature B cells was used for functional study. RESULTS: Tumour-infiltrating T cells and B cells were observed in close contact with each other and their densities are correlated with superior survival in patients with HCC. Furthermore, the density of TIBs was correlated with an enhanced expression of granzyme B and IFN-γ, as well as with reduced tumour viability defined by low expression of Ki-67, and an enhanced expression of activated caspase-3 on tumour cells. CD27 and CD40 costimulatory molecules and TILs expressing activation marker CD38 in the tumour were also correlated with patient survival. Mice depleted of mature B cells and transplanted with murine hepatoma cells showed reduced tumour control and decreased local T cell activation, further indicating the important role of B cells. CONCLUSIONS: The close proximity of tumour-infiltrating T cells and B cells indicates a functional interaction between them that is linked to an enhanced local immune activation and contributes to better prognosis for patients with HCC.


Subject(s)
Antigens, CD/analysis , B-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating , T-Lymphocytes/immunology , ADP-ribosyl Cyclase 1/analysis , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD19/genetics , Antigens, CD20/analysis , B-Lymphocytes/chemistry , B-Lymphocytes/pathology , CD3 Complex/analysis , CD40 Antigens/analysis , CD8 Antigens/analysis , CD8 Antigens/genetics , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/pathology , Caspase 3/analysis , Disease Progression , Female , Gene Expression , Granzymes/analysis , Humans , Interferon-gamma/genetics , Ki-67 Antigen/analysis , Liver Neoplasms/chemistry , Liver Neoplasms/pathology , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype , Retrospective Studies , Survival Rate , T-Lymphocytes/chemistry , T-Lymphocytes/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 7/analysis , Young Adult
4.
Gut ; 66(8): 1496-1506, 2017 08.
Article in English | MEDLINE | ID: mdl-27974549

ABSTRACT

OBJECTIVE: We investigated the mutational landscape of mammalian target of rapamycin (mTOR) signalling cascade in hepatocellular carcinomas (HCCs) with chronic HBV background, aiming to evaluate and delineate mutation-dependent mechanism of mTOR hyperactivation in hepatocarcinogenesis. DESIGN: We performed next-generation sequencing on human HCC samples and cell line panel. Systematic mutational screening of mTOR pathway-related genes was undertaken and mutant genes were evaluated based on their recurrence. Protein expressions of tuberous sclerosis complex (TSC)1, TSC2 and pRPS6 were assessed by immunohistochemistry in human HCC samples. Rapamycin sensitivity was estimated by colony-formation assay in HCC cell lines and the treatment was further tested using our patient-derived tumour xenograft (PDTX) models. RESULTS: We identified and confirmed multiple mTOR components as recurrently mutated in HBV-associated HCCs. Of significance, we detected frequent (16.2%, n=18/111) mutations of TSC1 and TSC2 genes in the HCC samples. The spectrum of TSC1/2 mutations likely disrupts the endogenous gene functions in suppressing the downstream mTOR activity through different mechanisms and leads to more aggressive tumour behaviour. Mutational disruption of TSC1 and TSC2 was also observed in HCC cell lines and our PDTX models. TSC-mutant cells exhibited reduced colony-forming ability on rapamycin treatment. With the use of biologically relevant TSC2-mutant PDTXs, we demonstrated the therapeutic benefits of the hypersensitivity towards rapamycin treatment. CONCLUSIONS: Taken together, our findings suggest the significance of previously undocumented mutation-dependent mTOR hyperactivation and frequent TSC1/2 mutations in HBV-associated HCCs. They define a molecular subset of HCC having genetic aberrations in mTOR signalling, with potential significance of effective specific drug therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Adult , Aged , Animals , Antibiotics, Antineoplastic/pharmacology , Axin Protein/genetics , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Mutational Analysis , DNA-Binding Proteins , Female , Humans , Liver Neoplasms/chemistry , Liver Neoplasms/metabolism , Male , Mice , Middle Aged , Mutation Rate , Neoplasm Transplantation , Nuclear Proteins/genetics , Signal Transduction , Sirolimus/pharmacology , Transcription Factors/genetics , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/analysis , Young Adult , beta Catenin/genetics
5.
J Hepatol ; 64(6): 1256-64, 2016 06.
Article in English | MEDLINE | ID: mdl-26867494

ABSTRACT

BACKGROUND & AIMS: Hepatitis B virus (HBV) integration is common in HBV-associated hepatocellular carcinoma (HCC) and may play an important pathogenic role through the production of chimeric HBV-human transcripts. We aimed to screen the transcriptome for HBV integrations in HCCs. METHODS: Transcriptome sequencing was performed on paired HBV-associated HCCs and corresponding non-tumorous liver tissues to identify viral-human chimeric sites. Validation was further performed in an expanded cohort of human HCCs. RESULTS: Here we report the discovery of a novel pre-mRNA splicing mechanism in generating HBV-human chimeric protein. This mechanism was exemplified by the formation of a recurrent HBV-cyclin A2 (CCNA2) chimeric transcript (A2S), as detected in 12.5% (6 of 48) of HCC patients, but in none of the 22 non-HCC HBV-associated cirrhotic liver samples examined. Upon the integration of HBV into the intron of the CCNA2 gene, the mammalian splicing machinery utilized the foreign splice sites at 282nt. and 458nt. of the HBV genome to generate a pseudo-exon, forming an in-frame chimeric fusion with CCNA2. The A2S chimeric protein gained a non-degradable property and promoted cell cycle progression, demonstrating its potential oncogenic functions. CONCLUSIONS: A pre-mRNA splicing mechanism is involved in the formation of HBV-human chimeric proteins. This represents a novel and possibly common mechanism underlying the formation of HBV-human chimeric transcripts from intronically integrated HBV genome with functional impact. LAY SUMMARY: HBV is involved in the mammalian pre-mRNA splicing machinery in the generation of potential tumorigenic HBV-human chimeras. This study also provided insight on the impact of intronic HBV integration with the gain of splice sites in the development of HBV-associated HCC.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B virus , Liver Neoplasms , Oncogene Proteins, Fusion , RNA Precursors , RNA Splicing , Virus Integration , Adult , Aged , Female , Humans , Male , Middle Aged , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Cyclin A2/genetics , Hepatitis B virus/genetics , Introns , Liver Neoplasms/genetics , Liver Neoplasms/virology , Oncogene Proteins, Fusion/biosynthesis , RNA Precursors/genetics , Transcriptome
6.
Liver Int ; 35(5): 1597-606, 2015 May.
Article in English | MEDLINE | ID: mdl-25424744

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is one of the most common human cancers. Recently, emerging evidence has suggested the role of long non-coding RNAs (lncRNAs) in human carcinogenesis. In this study, we aimed to investigate the expression and functional implications of lncRNAs in human HCC. METHODS: Eighty-eight well-annotated lncRNAs were profiled in primary HCC by quantitative RT-PCR. Functional relevance of lncRNAs was elucidated in HCC cell lines and nude mice models. The regulatory relationship between miRNA and lncRNA was predicted in silico and further validated by luciferase reporter assay and expression analysis. RESULTS: In our profiling study, HOTTIP was identified as the most significantly up-regulated lncRNA in human HCCs, even in early stage of HCC formation. Functionally, knock-down of HOTTIP attenuated HCC cell proliferation in vitro and markedly abrogated tumourigenicity in vivo. In addition, knock-down of HOTTIP also inhibited migratory ability of HCC cells and significantly abrogated lung metastasis in orthotopic implantation model in nude mice. HOTTIP is an antisense lncRNA mapped to the distal end of the HOXA gene cluster. Knock-down of HOTTIP significantly suppressed the expression of a number of HOXA genes. Furthermore, we identified miR-125b as a post-transcriptional regulator of HOTTIP. Ectopic expression of miR-125b reduced HOTTIP-coupled luciferase activity and suppressed the endogenous level of HOTTIP. Moreover, in human HCCs, HOTTIP expression negatively correlated with that of miR-125b. CONCLUSIONS: HOTTIP is a novel oncogenic lncRNA, which negatively regulated by miR-125b. Overexpression of HOTTIP contributes to hepatocarcinogenesis by regulating the expression of its neighbouring protein-coding genes.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Transcriptional Activation , Up-Regulation
7.
Hepatology ; 57(1): 131-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22821423

ABSTRACT

UNLABELLED: Random integration of hepatitis B virus (HBV) DNA into the host genome is frequent in human hepatocellular carcinoma (HCC) and this leads to truncation of the HBV DNA, particularly at the C-terminal end of the HBV X protein (HBx). In this study, we investigated the frequency of this natural C-terminal truncation of HBx in human HCCs and its functional significance. In 50 HBV-positive patients with HCC, full-length HBx was detected in all nontumorous livers. However, full-length HBx was found in only 27 (54%) of the HCC tumors, whereas natural carboxylic acid (COOH)-truncated HBx was found in the remaining 23 (46%) tumors. Upon clinicopathological analysis, the presence of natural COOH-truncated HBx significantly correlated with the presence of venous invasion, a hallmark of metastasis (P = 0.005). Inducible stable expression of the COOH-truncated HBx protein (with 24 amino acids truncated at the C-terminal end) enhanced the cell-invasive ability of HepG2 cells, as compared to full-length HBx, using the Matrigel cell-invasion assay. It also resulted in increased C-Jun transcriptional activity and enhanced transcription of matrix metalloproteinase 10 (MMP10), whereas activation of the MMP10 promoter by COOH-truncated HBx was abolished when the activator protein 1-binding sites on the MMP10 promoter were mutated. Furthermore, silencing of MMP10 by short interfering RNA in HBxΔC1-expressing HepG2 cells resulted in significant reduction of cell invasiveness. CONCLUSIONS: Our data suggest that COOH truncation of HBx, particularly with 24 amino acids truncated at the C-terminal end, plays a role in enhancing cell invasiveness and metastasis in HCC by activating MMP10 through C-Jun.


Subject(s)
Carcinoma, Hepatocellular/virology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/virology , Matrix Metalloproteinase 10/metabolism , Trans-Activators/metabolism , Adult , Aged , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Enzyme Activation , Female , Gene Expression , Hep G2 Cells , Humans , Liver/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Viral Regulatory and Accessory Proteins
8.
Hepatology ; 55(3): 807-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21994122

ABSTRACT

UNLABELLED: A novel theory in the field of tumor biology postulates that cancer growth is driven by a population of stem-like cells, called tumor-initiating cells (TICs). We previously identified a TIC population derived from hepatocellular carcinoma (HCC) that is characterized by membrane expression of CD133. Here, we describe a novel mechanism by which these cells mediate tumor growth and angiogenesis by systematic comparison of the gene expression profiles between sorted CD133 liver subpopulations through genome-wide microarray analysis. A significantly dysregulated interleukin-8 (IL-8) signaling network was identified in CD133(+) liver TICs obtained from HCC clinical samples and cell lines. IL-8 was found to be overexpressed at both the genomic and proteomic levels in CD133(+) cells isolated from HCC cell lines or clinical samples. Functional studies found enhanced IL-8 secretion in CD133(+) liver TICs to exhibit a greater ability to self-renew, induce tumor angiogenesis, and initiate tumors. In further support of these observations, IL-8 repression in CD133(+) liver TICs by knockdown or neutralizing antibody abolished these effects. Subsequent studies of the IL-8 functional network identified neurotensin (NTS) and CXCL1 to be preferentially expressed in CD133(+) liver TICs. Addition of exogenous NTS resulted in concomitant up-regulation of IL-8 and CXCL1 with simultaneous activation of p-ERK1/2 and RAF-1, both key components of the mitogen-activated protein kinase (MAPK) pathway. Enhanced IL-8 secretion by CD133(+) liver TICs can in turn activate an IL-8-dependent feedback loop that signals through the MAPK pathway. Further, in its role as a liver TIC marker CD133 also plays a functional part in regulating tumorigenesis of liver TICs by way of regulating NTS, IL-8, CXCL1, and MAPK signaling. CONCLUSION: CD133(+) liver TICs promote angiogenesis, tumorigenesis, and self-renewal through NTS-induced activation of the IL-8 signaling cascade.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Chemokine CXCL1/physiology , Glycoproteins/metabolism , Interleukin-8/physiology , Liver Neoplasms/pathology , Neoplastic Stem Cells/physiology , Neovascularization, Pathologic/physiopathology , Neurotensin/physiology , Peptides/metabolism , Signal Transduction/physiology , AC133 Antigen , Animals , Antigens, CD/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Cells, Cultured , Feedback, Physiological , Glycoproteins/deficiency , Glycoproteins/genetics , Hepatectomy , Humans , Interleukin-8/deficiency , Interleukin-8/genetics , Liver/blood supply , Liver/pathology , Liver/surgery , Liver Neoplasms/blood supply , Liver Neoplasms/surgery , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases , Neoplastic Stem Cells/immunology , Neurotensin/pharmacology , Peptides/deficiency , Peptides/genetics , Xenograft Model Antitumor Assays
9.
Gut ; 61(3): 427-38, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21930732

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is a heterogeneous disease with poor prognosis and limited methods for predicting patient survival. The nature of the immune cells that infiltrate tumours is known to impact clinical outcome. However, the molecular events that regulate this infiltration require further understanding. Here the ability of immune genes expressed in the tumour microenvironment to predict disease progression was investigated. METHODS: Using quantitative PCR, the expression of 14 immune genes in resected tumour tissues from 57 Singaporean patients was analysed. The nearest-template prediction method was used to derive and test a prognostic signature from this training cohort. The signature was then validated in an independent cohort of 98 patients from Hong Kong and Zurich. Intratumoural components expressing these critical immune genes were identified by in situ labelling. Regulation of these genes was analysed in vitro using the HCC cell line SNU-182. RESULTS: The identified 14 immune-gene signature predicts patient survival in both the training cohort (p=0.0004 and HR=5.2) and the validation cohort (p=0.0051 and HR=2.5) irrespective of patient ethnicity and disease aetiology. Importantly, it predicts the survival of patients with early disease (stages I and II), for whom classical clinical parameters provide limited information. The lack of predictive power in late disease stages III and IV emphasises that a protective immune microenvironment has to be established early in order to impact disease progression significantly. This signature includes the chemokine genes CXCL10, CCL5 and CCL2, whose expression correlates with markers of T helper 1 (Th1), CD8(+) T and natural killer (NK) cells. Inflammatory cytokines (tumour necrosis factor α, interferon γ) and Toll-like receptor 3 ligands stimulate intratumoural production of these chemokines which drive tumour infiltration by T and NK cells, leading to enhanced cancer cell death. CONCLUSION: A 14 immune-gene signature, which identifies molecular cues driving tumour infiltration by lymphocytes, accurately predicts survival of patients with HCC especially in early disease.


Subject(s)
Carcinoma, Hepatocellular/immunology , Chemokines/immunology , Liver Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Chemokine CXCL10/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Killer Cells, Natural/immunology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Neoplasm Staging , Prognosis , Th1 Cells/immunology , Toll-Like Receptor 3/immunology , Young Adult
10.
N Engl J Med ; 361(15): 1437-47, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19812400

ABSTRACT

BACKGROUND: Hepatocellular carcinoma is a common and aggressive cancer that occurs mainly in men. We examined microRNA expression patterns, survival, and response to interferon alfa in both men and women with the disease. METHODS: We analyzed three independent cohorts that included a total of 455 patients with hepatocellular carcinoma who had undergone radical tumor resection between 1999 and 2003. MicroRNA-expression profiling was performed in a cohort of 241 patients with hepatocellular carcinoma to identify tumor-related microRNAs and determine their association with survival in men and women. In addition, to validate our findings, we used quantitative reverse-transcriptase-polymerase-chain-reaction assays to measure microRNAs and assess their association with survival and response to therapy with interferon alfa in 214 patients from two independent, prospective, randomized, controlled trials of adjuvant interferon therapy. RESULTS: In patients with hepatocellular carcinoma, the expression of miR-26a and miR-26b in nontumor liver tissue was higher in women than in men. Tumors had reduced levels of miR-26 expression, as compared with paired noncancerous tissues, which indicated that the level of miR-26 expression was also associated with hepatocellular carcinoma. Moreover, tumors with reduced miR-26 expression had a distinct transcriptomic pattern, and analyses of gene networks revealed that activation of signaling pathways between nuclear factor kappaB and interleukin-6 might play a role in tumor development. Patients whose tumors had low miR-26 expression had shorter overall survival but a better response to interferon therapy than did patients whose tumors had high expression of the microRNA. CONCLUSIONS: The expression patterns of microRNAs in liver tissue differ between men and women with hepatocellular carcinoma. The miR-26 expression status of such patients is associated with survival and response to adjuvant therapy with interferon alfa.


Subject(s)
Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/genetics , Gene Expression , Interferon-alpha/therapeutic use , Liver Neoplasms/genetics , MicroRNAs/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Cohort Studies , Female , Gene Expression Profiling , Gene Regulatory Networks , Hepatitis B/complications , Humans , Liver Cirrhosis/complications , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Male , Middle Aged , Pharmacogenetics , Prognosis , Proportional Hazards Models , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Survival Analysis , Young Adult
11.
Neurosignals ; 20(2): 112-26, 2012.
Article in English | MEDLINE | ID: mdl-22204965

ABSTRACT

Deleted in liver cancer 2 (DLC2) is a novel Rho GTPase-activating protein that regulates RhoA activity. DLC2 is ubiquitously expressed in most tissues, including the brain, spinal cord and peripheral nerves, and is thought to be involved in actin cytoskeletal reorganization. Unlike DLC1-deficient mice, DLC2-deficient mice (DLC2(-/-)) are viable and without gross anatomical abnormalities. Interestingly, DLC2(-/-) mice exhibit hyperalgesia to noxious thermal stimuli and inflammation-inducing chemicals, such as formalin and acetic acid. There was no difference in the structure or morphology of cutaneous or sural nerves between DLC2(+/+) and DLC2(-/-) mice. However, sensory nerve conduction velocity in DLC2(-/-) mice was significantly higher than that in DLC2(+/+) mice, whereas motor nerve conduction velocity was not affected. After formalin injection, DLC2(-/-) mice showed increased RhoA activity in the spinal cord and an increased number of phosphorylated ERK1/2-positive cells. The inflammatory hyperalgesia in DLC2(-/-) mice appeared to be mediated through the activation of RhoA and ERK1/2. Taken together, DLC2 plays a key role in pain modulation during inflammation by suppressing the activation of RhoA and ERK to prevent an exaggerated pain response, and DLC2(-/-) mice provide a valuable tool for further understanding the regulation of inflammatory pain.


Subject(s)
Hyperalgesia/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Brain/metabolism , Brain/pathology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/pathology , Inflammation/physiopathology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Spinal Cord/metabolism , Spinal Cord/pathology , Sural Nerve/ultrastructure
12.
Biochim Biophys Acta ; 1806(2): 138-45, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20580775

ABSTRACT

Cadherin is an important cell adhesion molecule that plays paramount roles in organ development and the maintenance of tissue integrity. Dysregulation of cadherin expression is often associated with disease pathology including tissue dysplasia, tumor formation, and metastasis. Cadherin-17 (CDH17), belonging to a subclass of 7D-cadherin superfamily, is present in fetal liver and gastrointestinal tract during embryogenesis, but the gene becomes silenced in healthy adult liver and stomach tissues. It functions as a peptide transporter and a cell adhesion molecule to maintain tissue integrity in epithelia. However, recent findings from our group and others have reported aberrant expression of CDH17 in major gastrointestinal malignancies including hepatocellular carcinoma (HCC), stomach and colorectal cancers, and its clinical association with tumor metastasis and advanced tumor stages. Furthermore, alternative splice isoforms and genetic polymorphisms of CDH17 gene have been identified in HCC and linked to an increased risk of HCC. CDH17 is an attractive target for HCC therapy. Targeting CDH17 in HCC can inhibit tumor growth and inactivate Wnt signaling pathway in concomitance with activation of tumor suppressor genes. Further investigation on CDH17-mediated oncogenic signaling and cognate molecular mechanisms would shed light on new targeting therapy on HCC and potentially other gastrointestinal malignancies.


Subject(s)
Cadherins/physiology , Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Animals , Biomarkers, Tumor/analysis , Cadherins/analysis , Cadherins/chemistry , Cadherins/genetics , Carcinoma, Hepatocellular/drug therapy , Epigenesis, Genetic , Humans , Liver Neoplasms/drug therapy , Protein Isoforms , Signal Transduction
13.
Liver Transpl ; 17(2): 115-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21280183

ABSTRACT

The middle hepatic vein may be included in right liver living donor liver transplantation (LDLT) to optimize hepatic venous outflow. We studied the graft's ability to relieve portal hypertension and accommodate portal hyperperfusion with portal manometry and ultrasonic flowmetry. Surgical outcomes with respect to portal hemodynamometry were also investigated. The ages of the recipients and donors for 46 consecutive LDLT procedures were 50 (range, 16-66 years) and 31 years (range, 18-54 years), respectively. The graft to standard liver volume ratio was 47.4% (range, 32.4%-69.0%). The hospital mortality rate was 4.4% as 2 recipients died from a subarachnoid hemorrhage and sepsis. The portal pressure dropped by 8 mm Hg (range, -7 to 19 mm Hg) from 23 (range, 8-37 mm Hg) to 14 mm Hg (range, 10-26 mm Hg) after graft implantation. The portal inflow positively correlated with the portal pressure before native liver hepatectomy (R(2) = 0.305, P = 0.001) and not with the graft size. The portal inflow increased from 81 mL/minute/100 g (range, 35-210 mL/minute/100 g) before donor right hepatectomy to 318 mL/minute/100 g (range, 102-754 mL/minute/100 g) after graft implantation. The graft portal inflow had a positive linear correlation with the recipient portal pressure before native liver total hepatectomy (R(2) = 0.261, P = 0.001) but not after graft implantation, and it had a negative correlation with the graft to standard liver volume ratio (R(2) = 0.247, P = 0.001). Only 1 of the graft biopsies showed moderate sinusoidal congestion. Twelve recipients had Clavien grade 2+ complications that were not related to the portal inflow and pressure or graft size. Right liver LDLT including the middle hepatic vein effectively lowered the recipient portal pressure by allowing unimpeded venous outflow.


Subject(s)
Hepatectomy , Hepatic Veins/surgery , Hypertension, Portal/surgery , Liver Transplantation , Living Donors , Portal Pressure , Portal Vein/surgery , Adolescent , Adult , Biopsy , Blood Flow Velocity , Female , Hepatic Veins/physiopathology , Hong Kong , Hospital Mortality , Humans , Hypertension, Portal/mortality , Hypertension, Portal/physiopathology , Linear Models , Liver Transplantation/adverse effects , Liver Transplantation/mortality , Male , Manometry , Middle Aged , Portal Vein/physiopathology , Prospective Studies , Regional Blood Flow , Rheology , Time Factors , Treatment Outcome , Young Adult
14.
BMC Cancer ; 11: 481, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22070665

ABSTRACT

BACKGROUND: The prognosis of hepatocellular carcinoma (HCC) varies following surgical resection and the large variation remains largely unexplained. Studies have revealed the ability of clinicopathologic parameters and gene expression to predict HCC prognosis. However, there has been little systematic effort to compare the performance of these two types of predictors or combine them in a comprehensive model. METHODS: Tumor and adjacent non-tumor liver tissues were collected from 272 ethnic Chinese HCC patients who received curative surgery. We combined clinicopathologic parameters and gene expression data (from both tissue types) in predicting HCC prognosis. Cross-validation and independent studies were employed to assess prediction. RESULTS: HCC prognosis was significantly associated with six clinicopathologic parameters, which can partition the patients into good- and poor-prognosis groups. Within each group, gene expression data further divide patients into distinct prognostic subgroups. Our predictive genes significantly overlap with previously published gene sets predictive of prognosis. Moreover, the predictive genes were enriched for genes that underwent normal-to-tumor gene network transformation. Previously documented liver eSNPs underlying the HCC predictive gene signatures were enriched for SNPs that associated with HCC prognosis, providing support that these genes are involved in key processes of tumorigenesis. CONCLUSION: When applied individually, clinicopathologic parameters and gene expression offered similar predictive power for HCC prognosis. In contrast, a combination of the two types of data dramatically improved the power to predict HCC prognosis. Our results also provided a framework for understanding the impact of gene expression on the processes of tumorigenesis and clinical outcome.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/surgery , Cell Transformation, Neoplastic/genetics , Disease-Free Survival , Female , Gene Expression , Gene Expression Profiling , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/surgery , Male , Middle Aged , Predictive Value of Tests , Prognosis
15.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33445170

ABSTRACT

The RNA-binding protein Apobec1 complementation factor (A1CF) regulates posttranscriptional ApoB mRNA editing, but the range of RNA targets and the long-term effect of altered A1CF expression on liver function are unknown. Here we studied hepatocyte-specific A1cf-transgenic (A1cf+/Tg), A1cf+/Tg Apobec1-/-, and A1cf-/- mice fed chow or high-fat/high-fructose diets using RNA-Seq, RNA CLIP-Seq, and tissue microarrays from human hepatocellular cancer (HCC). A1cf+/Tg mice exhibited increased hepatic proliferation and steatosis, with increased lipogenic gene expression (Mogat1, Mogat2, Cidea, Cd36) associated with shifts in polysomal RNA distribution. Aged A1cf+/Tg mice developed spontaneous fibrosis, dysplasia, and HCC, and this development was accelerated on a high-fat/high-fructose diet and was independent of Apobec1. RNA-Seq revealed increased expression of mRNAs involved in oxidative stress (Gstm3, Gpx3, Cbr3), inflammatory response (Il19, Cxcl14, Tnfα, Ly6c), extracellular matrix organization (Mmp2, Col1a1, Col4a1), and proliferation (Kif20a, Mcm2, Mcm4, Mcm6), and a subset of mRNAs (including Sox4, Sox9, Cdh1) were identified in RNA CLIP-Seq. Increased A1CF expression in human HCC correlated with advanced fibrosis and with reduced survival in a subset with nonalcoholic fatty liver disease. In conclusion, we show that hepatic A1CF overexpression selectively alters polysomal distribution and mRNA expression, promoting lipogenic, proliferative, and inflammatory pathways leading to HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Fatty Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Fatty Liver/genetics , Fatty Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics
16.
J Proteome Res ; 9(4): 1923-30, 2010 Apr 05.
Article in English | MEDLINE | ID: mdl-20121168

ABSTRACT

Small hepatocellular carcinomas (HCCs) can be effectively cured by surgery with good clinical outcomes. However, the conventional AFP marker is ineffective in detecting small tumors. Here we employed a proteomic profiling approach to identify a candidate marker for HCC (

Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/blood , Liver Neoplasms/blood , Proteomics/methods , Vimentin/blood , Adult , Aged , Area Under Curve , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cohort Studies , Early Diagnosis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Male , Middle Aged , Sensitivity and Specificity
17.
Hepatology ; 50(5): 1453-63, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19676131

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a lethal malignancy for which there are no effective therapies. To develop rational therapeutic approaches for treating this disease, we are performing proof-of-principle studies targeting molecules crucial for the development of HCC. Here, we show that cadherin-17 (CDH17) adhesion molecule is up-regulated in human liver cancers and can transform premalignant liver progenitor cells to produce liver carcinomas in mice. RNA interference-mediated knockdown of CDH17 inhibited proliferation of both primary and highly metastatic HCC cell lines in vitro and in vivo. The antitumor mechanisms underlying CDH17 inhibition involve inactivation of Wnt signaling, because growth inhibition and cell death were accompanied by relocalization of beta-catenin to the cytoplasm and a concomitant reduction in cyclin D1 and an increase in retinoblastoma. CONCLUSION: Our results identify CDH17 as a novel oncogene in HCC and suggest that CDH17 is a biomarker and attractive therapeutic target for this aggressive malignancy.


Subject(s)
Cadherins/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Liver Neoplasms/pathology , Signal Transduction/physiology , Wnt Proteins/metabolism , Adult , Aged , Animals , Cadherins/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Middle Aged , Phenotype , Transplantation, Heterologous , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
18.
BMC Cancer ; 9: 389, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19886989

ABSTRACT

BACKGROUND: Surgical resection is one important curative treatment for hepatocellular carcinoma (HCC), but the prognosis following surgery differs substantially and such large variation is mainly unexplained. A review of the literature yields a number of clinicopathologic parameters associated with HCC prognosis. However, the results are not consistent due to lack of systemic approach to establish a prediction model incorporating all these parameters. METHODS: We conducted a retrospective analysis on the common clinicopathologic parameters from a cohort of 572 ethnic Chinese HCC patients who received curative surgery. The cases were randomly divided into training (n = 272) and validation (n = 300) sets. Each parameter was individually tested and the significant parameters were entered into a linear classifier for model building, and the prediction accuracy was assessed in the validation set RESULTS: Our findings based on the training set data reveal 6 common clinicopathologic parameters (tumor size, number of tumor nodules, tumor stage, venous infiltration status, and serum alpha-fetoprotein and total albumin levels) that were significantly associated with the overall HCC survival and disease-free survival (time to recurrence). We next built a linear classifier model by multivariate Cox regression to predict prognostic outcomes of HCC patients after curative surgery This analysis detected a considerable fraction of variance in HCC prognosis and the area under the ROC curve was about 70%. We further evaluated the model using two other protocols; leave-one-out procedure (n = 264) and independent validation (n = 300). Both were found to have excellent prediction power. The predicted score could separate patients into distinct groups with respect to survival (p-value = 1.8e-12) and disease free survival (p-value = 3.2e-7). CONCLUSION: This described model will provide valuable guidance on prognosis after curative surgery for HCC in clinical practice. The adaptive nature allows easy accommodation for future new biomarker inputs, and it may serve as the foundation for future modeling and prediction for HCC prognosis after surgical treatment.


Subject(s)
Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/surgery , Liver Neoplasms/mortality , Liver Neoplasms/surgery , Area Under Curve , Carcinoma, Hepatocellular/pathology , Disease-Free Survival , Humans , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Neoplasm Staging , Prognosis , ROC Curve , Retrospective Studies , Treatment Outcome
19.
Int J Cancer ; 123(5): 1043-52, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18553387

ABSTRACT

T-cadherin is an atypical cadherin and growing evidence has indicated that T-cadherin exerts tumor-suppressive effects on cancers of epithelial cell type and also causes positive effects on tumor angiogenesis. Human hepatocellular carcinoma (HCC) is a hypervascular tumor and T-cadherin has been shown to be overexpressed in intratumoral endothelial cells of HCCs. However, the expression status and functions of T-cadherin in hepatocytes or HCC cells remain unclear. Here, we demonstrated that T-cadherin was underexpressed in HCC cells (26.5%, 13/49 cases), but was frequently (77.6%, 38/49) overexpressed in intratumoral endothelial cells immunohistochemically. Semiquantitative RT-PCR analysis also showed that the T-cadherin gene was underexpressed in 7 of 11 HCC cell lines. Loss of heterozygosity analysis revealed that 32-38% of the 42 human HCC samples had allelic losses at this locus. Upon pharmacological treatment with demethylating agent 5-aza-2'-deoxycytidine or histone deacetylase inhibitor trichostatin A, T-cadherin promoter hypermethylation and/or histone deacetylation was frequently observed in HCC samples and cell lines. Functionally, enforced expression of T-cadherin induced G(2)/M cell cycle arrest, reduced cell proliferation in low serum medium, suppressed anchorage-independent growth in soft agar and increased sensitivity to TNFalpha-mediated apoptosis in HCC cells. Intriguingly, we found that T-cadherin significantly suppressed the activity of c-Jun, a crucial oncoprotein constitutively activated in HCC cells. To conclude, T-cadherin was differentially expressed in human HCCs. The underexpression of T-cadherin in HCC cells suggests it may be another critical event in addition to T-cadherin-mediated angiogenesis during HCC development.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Hepatocellular/genetics , Gene Silencing/drug effects , Hydroxamic Acids/pharmacology , Liver Neoplasms/genetics , Acetylation/drug effects , Adult , Aged , Anticarcinogenic Agents/metabolism , Apoptosis/drug effects , Azacitidine/pharmacology , Blotting, Western , Cadherins/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Down-Regulation , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Immunohistochemistry , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Loss of Heterozygosity , Male , Middle Aged , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
20.
Ann Surg ; 247(6): 1049-57, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18520234

ABSTRACT

OBJECTIVE: To test the hypothesis that acute phase small-for-size graft injury may promote late phase tumor recurrence after liver transplantation. SUMMARY BACKGROUND DATA: Living donor liver transplantation may provide the substantial intention-to-treat survival advantage for liver cancer patients. However, liver grafts from live donors are almost always small for size for adult recipients. Besides, tumor recurrence and metastasis after living donor liver transplantation have been reported. METHOD: An orthotopic Buffalo rat liver transplantation model using whole (100%, group W) and small-for-size grafts (50%, group S) was applied. Hepatoma cells were injected into the grafts after reperfusion. Comparison was made as regards acute phase graft injury and tumor growth together with cell proliferation (Ki67), angiogenesis (vascular endothelial growth factor), stellate cell activation (alpha-smooth muscle actin), and cell signaling pathway related to migration and invasion (Rac, rho-associated, coiled-coil containing protein kinase, and proline-rich tyrosine kinase 2). Invasiveness of the tumors developed was further assessed after their direct implantation into livers of nude mice. RESULTS: Liver tumors developed earlier and faster in group S with significantly greater tumor burden [hepatic replacement area: 61%; range, 47%-72%; vs. 18%; 12%-27%; P = 0.001] and tumor cell proliferation (92% vs. 59%; P = 0.0021) in a more invasive growth pattern with a higher incidence of venous invasion (91.7% vs. 25%; P = 0.003) and more frequent hepatic stellate cell activation. There was upregulation of protein expression of Rac/rho-associated, coiled-coil containing protein kinase/proline-rich tyrosine kinase 2/vascular endothelial growth factor signaling in group S. When implanted into livers of nude mice, tumors from group S had a higher incidence of local (70% vs. 0%; P = 0.003) and lung metastasis (50% vs. 0%; P = 0.033). This phenotype was consistent with their ultrastructural features linking to angiogenesis and invasiveness. CONCLUSION: Significant activation of cell signaling pathways leading to tumor invasion and migration in small-for-size liver grafts promotes tumor growth and metastasis after liver transplantation.


Subject(s)
Acute-Phase Reaction/metabolism , Acute-Phase Reaction/pathology , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Liver Transplantation , Animals , Enzyme-Linked Immunosorbent Assay , Graft Survival , Intercellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Organ Size , Rats , Signal Transduction , Statistics, Nonparametric , Vascular Endothelial Growth Factor A/blood
SELECTION OF CITATIONS
SEARCH DETAIL