Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 135(5): 852-64, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-19012953

ABSTRACT

Cancers are highly heterogeneous and contain many passenger and driver mutations. To functionally identify tumor suppressor genes relevant to human cancer, we compiled pools of short hairpin RNAs (shRNAs) targeting the mouse orthologs of genes recurrently deleted in a series of human hepatocellular carcinomas and tested their ability to promote tumorigenesis in a mosaic mouse model. In contrast to randomly selected shRNA pools, many deletion-specific pools accelerated hepatocarcinogenesis in mice. Through further analysis, we identified and validated 13 tumor suppressor genes, 12 of which had not been linked to cancer before. One gene, XPO4, encodes a nuclear export protein whose substrate, EIF5A2, is amplified in human tumors, is required for proliferation of XPO4-deficient tumor cells, and promotes hepatocellular carcinoma in mice. Our results establish the feasibility of in vivo RNAi screens and illustrate how combining cancer genomics, RNA interference, and mosaic mouse models can facilitate the functional annotation of the cancer genome.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genes, Tumor Suppressor , Genomics , Liver Neoplasms/genetics , RNA Interference , Animals , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice , Peptide Initiation Factors/genetics , RNA, Untranslated/genetics , RNA-Binding Proteins/genetics , Smad3 Protein/metabolism , Eukaryotic Translation Initiation Factor 5A
2.
J Surg Oncol ; 123(5): 1253-1262, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33524213

ABSTRACT

BACKGROUND AND OBJECTIVES: In this retrospective study, we examined the CA17 tissue expression and analyzed its clinical significance in cholangiocarcinoma (CCA). MATERIALS AND METHODS: Immunohistochemistry was performed to assess CA17 expression on tissue microarrays in a training cohort enrolling 120 CCA patients and a validation cohort comprising 60 CCA patients. Image pro plus was applied to score the staining intensity and expression level of CA17 marker. Kaplan-Meier analysis, Cox's proportional hazards regression, and nomogram were applied to evaluate the prognostic significance of CA17. RESULTS: CA17 cancer biomarker over-expression was significantly observed in CCA compared to their non-tumor counterparts, and positively correlated with aggressive tumor phenotypes, like lymph node metastasis. Meanwhile, patients with high expression of CA17 correlated with worse postoperative overall survival (OS) and recurrence-free survival. Besides, multivariate analysis identified that CA17 expression was an independent prognostic factor for cholangiocarcinoma patients, which indicated that the CA17 could be more efficient than serum CA19-9 in predicting the OS of CCA patients. Notably, the nomogram integrating CA17 expression had better prognostic performance as compared with current TNM staging systems. CONCLUSION: CA17 was an independent adverse prognostic factor for CCA patients' survival, which may serve as a promising prognostic biomarker for CCA patients.


Subject(s)
Bile Duct Neoplasms/pathology , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Cholangiocarcinoma/pathology , Neoplasm Recurrence, Local/pathology , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/surgery , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/surgery , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/surgery , Prognosis , Retrospective Studies , Survival Rate
3.
Mol Pharm ; 17(10): 3941-3951, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32931292

ABSTRACT

In cancer photodynamic therapy (PDT), a photosensitizer taken up by cancer cells can generate reactive oxygen species upon near-infrared light activation to induce cancer cell death. To increase PDT potency and decrease its adverse effect, one approach is to conjugate the photosensitizer with an antibody that specifically targets cancer cells. In the present study, IR700, a hydrophilic phthalocyanine photosensitizer, was conjugated to the humanized monoclonal antibody ARB102, which binds specifically cadherin-17 (CDH17 aka CA17), a cell surface marker highly expressed in gastrointestinal cancer to produce ARB102-IR700. Photoimmunotherapy (PIT) of gastrointestinal cancer cell lines was conducted by ARB102-IR700 treatment and near-infrared light irradiation. The results showed that ARB102-IR700 PIT could induce cell death in CDH17-positive cancer cells with high potency. In a co-culture model, CDH17-negative and CDH17-overexpressing SW480 cells were labeled with distinct fluorescent dyes and cultured together prior to PIT treatment. The results confirmed that ARB102-IR700 PIT could kill CDH17-positive cells specifically, while leaving the adjacent CDH17-negative cells unaffected. An in vivo efficacy study was conducted using a pancreatic adenocarcinoma AsPC-1 xenograft tumor model in nude mice. Fluorescence scanning indicated that ARB102-IR700 accumulated specifically in the tumor sites. To perform PIT, at 24 and 48 h postinjection, mice were irradiated with a 680 nm laser at the tumor site to activate the photosensitizer. It was shown that ARB102-IR700 PIT could inhibit tumor growth significantly. In summary, this study demonstrated that the novel ARB102-IR700 is a promising agent for PIT in gastrointestinal cancers.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Cadherins/antagonists & inhibitors , Gastrointestinal Neoplasms/drug therapy , Photochemotherapy/methods , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cadherins/metabolism , Cell Culture Techniques , Cell Line, Tumor , Coculture Techniques , Female , Gastrointestinal Neoplasms/pathology , Humans , Infrared Rays , Injections, Intravenous , Mice , Tissue Distribution , Xenograft Model Antitumor Assays
4.
N Engl J Med ; 368(24): 2266-76, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23758232

ABSTRACT

BACKGROUND: Hepatocellular carcinoma is the third leading cause of cancer-related deaths worldwide. In the heterogeneous group of hepatocellular carcinomas, those with characteristics of embryonic stem-cell and progenitor-cell gene expression are associated with the worst prognosis. The oncofetal gene SALL4, a marker of a subtype of hepatocellular carcinoma with progenitor-like features, is associated with a poor prognosis and is a potential target for treatment. METHODS: We screened specimens obtained from patients with primary hepatocellular carcinoma for the expression of SALL4 and carried out a clinicopathological analysis. Loss-of-function studies were then performed to evaluate the role of SALL4 in hepatocarcinogenesis and its potential as a molecular target for therapy. To assess the therapeutic effects of a peptide that targets SALL4, we used in vitro functional and in vivo xenograft assays. RESULTS: SALL4 is an oncofetal protein that is expressed in the human fetal liver and silenced in the adult liver, but it is reexpressed in a subgroup of patients who have hepatocellular carcinoma and an unfavorable prognosis. Gene-expression analysis showed the enrichment of progenitor-like gene signatures with overexpression of proliferative and metastatic genes in SALL4-positive hepatocellular carcinomas. Loss-of-function studies confirmed the critical role of SALL4 in cell survival and tumorigenicity. Blocking SALL4-corepressor interactions released suppression of PTEN (the phosphatase and tensin homologue protein) and inhibited tumor formation in xenograft models in vivo. CONCLUSIONS: SALL4 is a marker for a progenitor subclass of hepatocellular carcinoma with an aggressive phenotype. The absence of SALL4 expression in the healthy adult liver enhances the potential of SALL4 as a treatment target in hepatocellular carcinoma. (Funded by the Singapore National Medical Research Council and others.).


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Transcription Factors/genetics , Adult , Animals , Carcinoma, Hepatocellular/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Metabolic Networks and Pathways/physiology , Mice , Mice, Inbred Strains , PTEN Phosphohydrolase/metabolism , Prognosis , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Genome Res ; 23(9): 1422-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23788652

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most deadly cancers worldwide and has no effective treatment, yet the molecular basis of hepatocarcinogenesis remains largely unknown. Here we report findings from a whole-genome sequencing (WGS) study of 88 matched HCC tumor/normal pairs, 81 of which are Hepatitis B virus (HBV) positive, seeking to identify genetically altered genes and pathways implicated in HBV-associated HCC. We find beta-catenin to be the most frequently mutated oncogene (15.9%) and TP53 the most frequently mutated tumor suppressor (35.2%). The Wnt/beta-catenin and JAK/STAT pathways, altered in 62.5% and 45.5% of cases, respectively, are likely to act as two major oncogenic drivers in HCC. This study also identifies several prevalent and potentially actionable mutations, including activating mutations of Janus kinase 1 (JAK1), in 9.1% of patients and provides a path toward therapeutic intervention of the disease.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genome, Human , Liver Neoplasms/genetics , Mutation , Amino Acid Sequence , Carcinoma, Hepatocellular/virology , DNA, Viral/genetics , Female , Hepatitis B virus/genetics , Humans , Janus Kinase 1/genetics , Liver Neoplasms/virology , Male , Molecular Sequence Data , STAT Transcription Factors/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics , Virus Integration , Wnt Signaling Pathway/genetics , beta Catenin/genetics
6.
FASEB J ; 29(2): 724-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25384421

ABSTRACT

Hippo signaling pathway is emerging as a novel target for anticancer therapy because it plays key roles in organ size control and tumorigenesis. As the downstream effectors, Yes-associated protein (YAP)-transcriptional enhancer activation domain family member (TEAD) association is essential for YAP-driven oncogenic activity, while TEAD is largely dispensable for normal tissue growth. We present the design of YAP-like peptides (17mer) to occupy the interface 3 on TEAD. Introducing cysteines at YAP sites 87 and 96 can induce disulfide formation, as confirmed by crystallography. The engineered peptide significantly improves the potency in disrupting YAP-TEAD interaction in vitro. To confirm that blocking YAP-TEAD complex formation by directly targeting on TEAD is a valid approach, we report a significant reduction in tumor growth rate in a hepatocellular carcinoma xenograft model after introducing the dominant-negative mutation (Y406H) of TEAD1 to abolish YAP-TEAD interaction. Our results suggest that targeting TEAD is a promising strategy against YAP-induced oncogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Binding, Competitive , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cloning, Molecular , Crystallography, X-Ray , Cysteine/chemistry , Disulfides , Female , Glutathione Transferase/metabolism , Hippo Signaling Pathway , Humans , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasm Transplantation , Peptides/chemistry , Peptides, Cyclic/chemistry , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Surface Plasmon Resonance , TEA Domain Transcription Factors , YAP-Signaling Proteins
7.
Mol Cancer ; 13: 66, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24655440

ABSTRACT

BACKGROUND: Constitutive activation of signal transducer and activator of transcription 3 (STAT3) has been linked with proliferation, survival, invasion and angiogenesis of a variety of human cancer cells, including hepatocellular carcinoma (HCC). Thus, novel agents that can suppress STAT3 activation have potential for both prevention and treatment of HCC. Here we report, garcinol, a polyisoprenylated benzophenone, could suppress STAT3 activation in HCC cell lines and in xenografted tumor of HCC in nude mice model. EXPERIMENTAL DESIGN: Different HCC cell lines have been treated with garcinol and the inhibition of STAT3 activation, dimerization and acetylation have been checked by immunoblotting, immuno-fluorescence, and DNA binding assays. Xenografted tumor model has been generated in nude mice using HCC cell line and effect of garcinol in the inhibition of tumor growth has been investigated. RESULTS: Garcinol could inhibit both constitutive and interleukin (IL-6) inducible STAT3 activation in HCC cells. Computational modeling showed that garcinol could bind to the SH2 domain of STAT3 and suppress its dimerization in vitro. Being an acetyltransferase inhibitor, garcinol also inhibits STAT3 acetylation and thus impairs its DNA binding ability. The inhibition of STAT3 activation by garcinol led to the suppression of expression of various genes involved in proliferation, survival, and angiogenesis. It also suppressed proliferation and induced substantial apoptosis in HCC cells. Remarkably, garcinol inhibited the growth of human HCC xenograft tumors in athymic nu/nu mice, through the inhibition of STAT3 activation. CONCLUSION: Overall, our results suggest that garcinol exerts its anti-proliferative and pro-apoptotic effects through suppression of STAT3 signaling in HCC both in vitro and in vivo.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , STAT3 Transcription Factor/biosynthesis , Terpenes/administration & dosage , Acetylation/drug effects , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dimerization , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors
8.
PLoS Med ; 11(12): e1001770, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25536056

ABSTRACT

BACKGROUND: Typically observed at 2 y after surgical resection, late recurrence is a major challenge in the management of hepatocellular carcinoma (HCC). We aimed to develop a genomic predictor that can identify patients at high risk for late recurrence and assess its clinical implications. METHODS AND FINDINGS: Systematic analysis of gene expression data from human liver undergoing hepatic injury and regeneration revealed a 233-gene signature that was significantly associated with late recurrence of HCC. Using this signature, we developed a prognostic predictor that can identify patients at high risk of late recurrence, and tested and validated the robustness of the predictor in patients (n = 396) who underwent surgery between 1990 and 2011 at four centers (210 recurrences during a median of 3.7 y of follow-up). In multivariate analysis, this signature was the strongest risk factor for late recurrence (hazard ratio, 2.2; 95% confidence interval, 1.3-3.7; p = 0.002). In contrast, our previously developed tumor-derived 65-gene risk score was significantly associated with early recurrence (p = 0.005) but not with late recurrence (p = 0.7). In multivariate analysis, the 65-gene risk score was the strongest risk factor for very early recurrence (<1 y after surgical resection) (hazard ratio, 1.7; 95% confidence interval, 1.1-2.6; p = 0.01). The potential significance of STAT3 activation in late recurrence was predicted by gene network analysis and validated later. We also developed and validated 4- and 20-gene predictors from the full 233-gene predictor. The main limitation of the study is that most of the patients in our study were hepatitis B virus-positive. Further investigations are needed to test our prediction models in patients with different etiologies of HCC, such as hepatitis C virus. CONCLUSIONS: Two independently developed predictors reflected well the differences between early and late recurrence of HCC at the molecular level and provided new biomarkers for risk stratification. Please see later in the article for the Editors' Summary.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Adult , Aged , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/genetics , Risk Factors , STAT3 Transcription Factor/genetics , Young Adult
9.
Dig Dis Sci ; 59(10): 2477-87, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24811246

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) belong to a group of small non-coding RNA with differential expression in tumors, including hepatocellular carcinoma (HCC). AIM: This study investigates the involvement of miR-125b in HCC. METHODS: Clinical analysis of miR-125b was performed using data derived from miRNA profiling and qPCR. Phenotypic changes of liver cell lines were examined after ectopic miR-125b expression. Lastly, bioinformatics analysis coupled with luciferase reporter assay was used to reveal the cellular target of miR-125b. RESULTS: A down-regulation of miR-125b was found in HCC tumors and cultured cells. Patients having tumors with ≥twofold reduction in miR-125b compared to adjacent non-tumor tissues contributed to 23 out of 49 HCC cases (46.9 %), while this down-regulation was usually found in patients with tumor venous infiltration and recurrence. miR-125b expression was also negatively correlated with increased serum AFP level and poor overall survival of patients. Ectopic expression of miR-125b led to alleviated tumor phenotypes of HCC cells. Among the 110 bioinformatically predicated candidates, 31 of them negatively correlated with miR-125b in HCC tumors for which one of them named eukaryotic translation initiation factor 5A2 (eIF5A2), known also as a liver oncofetal molecule, was validated to be a direct target of miR-125b in HCC. CONCLUSIONS: This study has evidenced for the negative correlation of tumor miR-125b expression with poor prognosis of HCC patients. Expression of miR-125b can reverse the tumorigenic properties of cultured HCC cells via suppressing the tumorigenic molecule eIF5A2, thus postulating restoration of miR-125b level as a way to counteract liver tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Peptide Initiation Factors/metabolism , Cell Line, Tumor , Down-Regulation , Humans , MicroRNAs/genetics , Peptide Initiation Factors/genetics , Transcriptome
10.
Biochim Biophys Acta ; 1826(2): 357-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22683405

ABSTRACT

Hippo pathway, originally discovered in Drosophila, is responsible for organ size control. The pathway is conserved in mammals and has a significant role in restraining cancer development. Regulating the Hippo pathway thus represents a potential therapeutic approach to treat cancer, which however requires deep understanding of the targeted pathway. Despite our limited knowledge on the pathway, there are increasing discoveries of new molecules that regulate and modulate the Hippo downstream signaling particularly in various solid malignancies, from extracellular stimuli or via pathway crosstalk. Herein, we discuss the roles of newly identified and key regulators that connect with core components (MST1/2, LATS1/2, SAV1, and MOB1) and downstream effector (YAP) in the Hippo pathway having an important role in cancer development and progression. Understanding of the mammalian Hippo pathway regulation may shed new insights to allow us selecting the right oncogenic targets and designing effective drugs for cancer treatments.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Neoplasms/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Cycle Proteins/physiology , Humans , Protein Serine-Threonine Kinases/physiology , Serine-Threonine Kinase 3 , Tumor Suppressor Proteins/physiology
11.
Expert Rev Mol Diagn ; 23(2): 171-179, 2023 02.
Article in English | MEDLINE | ID: mdl-36744385

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of cancer deaths in Hong Kong. We tested the hypothesis that circulating tumor cell (CTC) analysis by ARB101 antibody could be used as a tool for CRC detection, progression, and therapy response. RESEARCH METHODS: ARB101 antibody was used for investigation of CDH17 expression in formalin-fixed, paraffin-embedded (FFPE) tissue sections and circulating tumor cells (CTCs) of CRC patients. RESULTS: Using ARB101, highest sensitivity was observed in 98/100 (98%) colorectal cancer tissue compared to 72/100 gastric cancer (72%) and 27/32 pancreatic cancer (84%). Immunoreactivity of CDH17 was significantly higher in distant metastatic (tumor-node-metastasis [TNM] stage IV) than non-distant metastatic (TNM stage I to III) CRC. ARB101 antibody also manifested the higher sensitivity than c-erbB2 (8%) and epidermal growth factor receptor (EGFR)-targeting antibodies (37%) with the significance (p < 0.0001). ARB101 positive CTCs were detected in 64/83 (77%) TNM stage I to IV CRC patients. Furthermore, ARB101 positive CTCs detected in TNM stage I to III CRC patients before and after surgical operation are statistically significant (p < 0.0001). CONCLUSIONS: CTC detection by ARB101 antibody could serve as a potential non-invasive approach for CRC detection, progression, and monitoring of treatment response.


Subject(s)
Colorectal Neoplasms , Neoplastic Cells, Circulating , Pancreatic Neoplasms , Stomach Neoplasms , Humans , Neoplastic Cells, Circulating/pathology , Colorectal Neoplasms/metabolism , Hong Kong , Biomarkers, Tumor/metabolism , Cadherins
12.
J Biol Chem ; 286(35): 30706-30713, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21737452

ABSTRACT

Hepatocellular carcinoma (HCC) is a heterogeneous and highly aggressive malignancy, for which there are no effective cures. Identification of a malignant stemlike subtype of HCC may offer patients with a dismal prognosis a potential targeted therapy using c-MET and Wnt pathway inhibitors. MicroRNAs (miRNAs) show promise as diagnostic and prognostic tools for cancer detection and stratification. Using a TRE-c-Met-driven transgenic HCC mouse model, we identified a cluster of 23 miRNAs that is encoded within the Dlk1-Gtl2 imprinted region on chromosome 12qF1 overexpressed in all of the isolated liver tumors. Interestingly, this region is conserved among mammalian species and maps to the human DLK1-DIO3 region on chromosome 14q32.2. We thus examined the expression of the DLK1-DIO3 miRNA cluster in a cohort of 97 hepatitis B virus-associated HCC patients and identified a subgroup (n = 18) of patients showing strong coordinate overexpression of miRNAs in this cluster but not in other cancer types (breast, lung, kidney, stomach, and colon) that were tested. Expression levels of imprinted gene transcripts from neighboring loci in this 14q32.2 region and from a subset of other imprinted sites were concomitantly elevated in human HCC. Interestingly, overexpression of the DLK1-DIO3 miRNA cluster was positively correlated with HCC stem cell markers (CD133, CD90, EpCAM, Nestin) and associated with a high level of serum α-fetoprotein, a conventional biomarker for liver cancer, and poor survival rate in HCC patients. In conclusion, our findings suggest that coordinate up-regulation of the DLK1-DIO3 miRNA cluster at 14q32.2 may define a novel molecular (stem cell-like) subtype of HCC associated with poor prognosis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Chromosomes, Human, Pair 14/genetics , Intercellular Signaling Peptides and Proteins/genetics , Iodide Peroxidase/genetics , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Membrane Proteins/genetics , MicroRNAs/genetics , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins , Cohort Studies , Humans , Liver/metabolism , MicroRNAs/metabolism , Multigene Family , Prognosis , Tissue Distribution , Treatment Outcome , Up-Regulation
13.
J Biol Chem ; 286(20): 18066-78, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21402708

ABSTRACT

MicroRNAs function as important regulators of gene expression and are commonly linked to development, differentiation, and diseases such as cancer. To better understand their roles in various biological processes, identification of genes targeted by microRNAs is necessary. Although prediction tools have significantly helped with this task, experimental approaches are ultimately required for extensive target search and validation. We employed two independent yet complementary high throughput approaches to map a large set of mRNAs regulated by miR-122, a liver-specific microRNA implicated in regulation of fatty acid and cholesterol metabolism, hepatitis C infection, and hepatocellular carcinoma. The combination of luciferase reporter-based screening and shotgun proteomics resulted in the identification of 260 proteins significantly down-regulated in response to miR-122 in at least one method, 113 of which contain predicted miR-122 target sites. These proteins are enriched for functions associated with the cell cycle, differentiation, proliferation, and apoptosis. Among these miR-122-sensitive proteins, we identified a large group with strong connections to liver metabolism, diseases, and hepatocellular carcinoma. Additional analyses, including examination of consensus binding motifs for both miR-122 and target sequences, provide further insight into miR-122 function.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Genes, Neoplasm , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/metabolism , Carcinoma, Hepatocellular/genetics , Cell Line , Humans , Liver Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics
14.
Cancer ; 118(22): 5489-96, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22517484

ABSTRACT

BACKGROUND: Excessive reactive oxygen species (ROS) accumulation is a common phenomenon in carcinogenesis. However, the rationale behind ROS involvement in gastric cancer is unclear. In this study, the authors investigated the clinical significance of the single nucleotide polymorphisms (SNPs) of 2 ROS metabolic process-related genes: superoxide dismutase 2 (SOD2) and glutathione S-transferase π (GSTP1). METHODS: In total of 929 patients with gastric cancer who had definitive clinicopathologic and follow-up data were collected. SOD2 reference SNP 4880 (rs4880) and GSTP1 rs1695 genotyping were examined in DNA samples extracted from paraffin-embedded tumor tissue. Association of the 2 SNPs with each clinicopathologic feature was analyzed using the Pearson chi-square test and the independent Student t test. Gastric cancer-specific overall survival was analyzed using Kaplan-Meier curves and log-rank tests. Multivariate Cox regression analyses of these SNPs also were performed. RESULTS: The SOD2 rs4880 CT + CC genotypes were significantly associated with a high level of lymph node metastasis (P = .023), whereas the GSTP1 rs1695 GA + GG genotypes were significantly associated with larger tumor size (>5 cm long; P = .048). Kaplan-Meier and Cox regression data indicated that the SOD2 rs4880 CT + CC genotypes alone (hazard ratio, 1.299; 95% confidence interval, 1.053-1.603; P = .015) and the GSTP1 rs1695 GA + GG combined genotypes (hazard ratio, 1.496; 95% CI, 1.078-2.074; P = .016) were independent predictors for overall survival. CONCLUSIONS: The current data, based on a large cohort (n = 929) of Chinese patients with gastric cancer, suggested that the presence of SOD2 rs4880 and GSTP1 rs1695 genotypes may contribute to cancer progression as well as tumor aggressiveness. The components of ROS metabolism pathways may be potential therapeutic targets for this aggressive malignancy.


Subject(s)
Glutathione S-Transferase pi/genetics , Lymphatic Metastasis/genetics , Stomach Neoplasms/genetics , Superoxide Dismutase/genetics , Adult , Aged , Aged, 80 and over , China , Disease Progression , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Reactive Oxygen Species/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Survival Rate , Treatment Outcome
15.
World J Surg ; 36(1): 90-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21976009

ABSTRACT

UNLABELLED: Esophageal cancer is a deadly cancer with esophageal squamous cell carcinoma (ESCC) as the major type. Until now there has been a lack of reliable prognostic markers for this malignancy. This study aims to investigate the clinical correlation between Forkhead box M1 (FoxM1)and patients' parameters in ESCC. METHODS: Immunohistochemistry was performed to investigate the expression and localization of FoxM1 in 64ESCC tissues and 10 nontumor esophageal tissues randomly selected from 64 patients before these data were used for clinical correlations. RESULTS: Cytoplasmic and nuclear expressions of FoxM1 were found in 63 and 16 of the 64 ESCC tissues, respectively.Low cytoplasmic expression of FoxM1 was correlated with early pathological stage in ESCC (P = 0.018),while patients with nuclear FoxM1 were younger in age than those without nuclear expression (P\0.001).Upregulation of FoxM1 mRNA was found in five ESCCcell lines (HKESC-1, HKESC-2, HKESC-3, HKESC-4,and SLMT-1) when compared to non-neoplastic esophageal squamous cell line NE-1 using quantitative polymerase chain reaction (qPCR). Except for HKESC-3, all studied ESCC cell lines demonstrated a high expression of FoxM1 protein using immunoblot. A high mRNA level of FoxM1 was observed in all of the ESCC tissues examined when compared to their adjacent nontumor tissues using qPCR. CONCLUSION: Cytoplasmic FoxM1 was correlated with pathological stage and might be a biomarker for advanced ESCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/surgery , Case-Control Studies , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/surgery , Esophagectomy , Female , Forkhead Box Protein M1 , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
16.
Clin Exp Pharmacol Physiol ; 39(3): 311-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21834865

ABSTRACT

1. Bioactive compounds from medicinal plants with anticancer and anti-inflammatory effects have become key resources in drug discovery fields for the treatment of various malignancies and immunological disorders. 2. Tripterygium wilfordii (TW) is a medicinal plant that exhibits profound immunosuppressive effects and has been used in herbal regimens for the treatment of immunological diseases for thousand years in China. 3. Procedures for the isolation and characterization of TW bioactive compounds have been well established. Over the past three decades, more than 46 diterpenoids, 20 new triterpenoids, 26 alkaloids and other small molecules have been identified from TW. 4. Triptolide, celastrol and tripchlorolide are among the bioactive compounds conferring the immunosuppressive and anticancer activities of TW. Accumulated evidence suggests that the TW bioactive compounds exert their pharmacological actions by modulating the transcriptional activity of nuclear factor-κB signalling molecule. 5. Triptolide derivatives with improved water solubility have emerged as promising drug candidates. Clinical trials are being conducted on the derivatives to provide encyclopaedic knowledge on triptolide pharmacokinetics in patients.


Subject(s)
Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/chemistry , Biological Products/chemistry , Plant Extracts/chemistry , Tripterygium/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/therapeutic use , Biological Products/isolation & purification , Biological Products/therapeutic use , Humans , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Tripterygium/physiology
17.
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
18.
Int J Cancer ; 129(8): 1806-14, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21165951

ABSTRACT

Stress protein mortalin (mtHSP70) is highly expressed in cancer cells. It was shown to contribute to carcinogenesis by sequestrating the wild type p53, a key tumor suppressor protein, in the cytoplasm resulting in an abrogation of its transcriptional activation function. We have found that the level of mortalin expression has significant correlation with human hepatocellular carcinoma (HCC) malignancy and therefore investigated whether it interacts with and influences the activities of mutant p53, frequently associated with HCC development. We have detected mortalin-p53 interactions in liver tumor and five HCC cell lines that harbored mutant p53. The data was in contrast to the normal liver and immortalized normal hepatocytes that lacked mortalin-p53 interaction. Furthermore, we have found that the shRNA-mediated mortalin silencing could induce mutant p53-mediated tumor-specific apoptosis in HCC. Such allotment of apoptotic function to mutant p53 by targeting mortalin-p53 interaction in cancer cells is a promising strategy for HCC therapy.


Subject(s)
Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Genes, p53 , HSP70 Heat-Shock Proteins/physiology , Liver Neoplasms/genetics , Cell Line, Tumor , HSP70 Heat-Shock Proteins/genetics , Humans , Mutation , RNA, Small Interfering/pharmacology
19.
Mol Syst Biol ; 6: 402, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20739924

ABSTRACT

Tumorigenesis involves multistep genetic alterations. To elucidate the microRNA (miRNA)-gene interaction network in carcinogenesis, we examined their genome-wide expression profiles in 96 pairs of tumor/non-tumor tissues from hepatocellular carcinoma (HCC). Comprehensive analysis of the coordinate expression of miRNAs and mRNAs reveals that miR-122 is under-expressed in HCC and that increased expression of miR-122 seed-matched genes leads to a loss of mitochondrial metabolic function. Furthermore, the miR-122 secondary targets, which decrease in expression, are good prognostic markers for HCC. Transcriptome profiling data from additional 180 HCC and 40 liver cirrhotic patients in the same cohort were used to confirm the anti-correlation of miR-122 primary and secondary target gene sets. The HCC findings can be recapitulated in mouse liver by silencing miR-122 with antagomir treatment followed by gene-expression microarray analysis. In vitro miR-122 data further provided a direct link between induction of miR-122-controlled genes and impairment of mitochondrial metabolism. In conclusion, miR-122 regulates mitochondrial metabolism and its loss may be detrimental to sustaining critical liver function and contribute to morbidity and mortality of liver cancer patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/genetics , Liver Neoplasms/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Animals , Cell Line, Tumor , Down-Regulation/genetics , Energy Metabolism/genetics , Gene Expression Profiling , Genes, Mitochondrial/genetics , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Survival Analysis , Up-Regulation/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL