Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 331
Filter
1.
Oncogene ; 34(39): 5095-104, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-25597408

ABSTRACT

Identification of key drivers and new therapeutic targets is important given the poor prognosis for hepatocellular carcinoma (HCC) patients, particularly those ineligible for surgical resection or liver transplant. However, the approach to identify such driver genes is facing significant challenges due to the genomically heterogenous nature of HCC. Here we tested whether the integrative genomic profiling of a well-defined HCC subset that is classified by an extreme EpCAM(+) AFP(+) gene expression signature and associated with poor prognosis, all attributes of a stem cell-like phenotype, could uncover survival-related driver genes in HCC. Following transcriptomic analysis of the well-defined HCC cases, a Gene Set Enrichment Analysis coupled with genomic copy number alteration assessment revealed that YY1-associated protein 1 (YY1AP1) is a critical oncoprotein specifically activated in EpCAM(+) AFP(+) HCC. YY1AP1 silencing eliminates oncogene addiction by altering the chromatin landscape and triggering massive apoptosis in vitro and tumor suppression in vivo. YY1AP1 expression promotes HCC proliferation and is required for the maintenance of stem cell features. We revealed that YY1AP1 cooperates with YY1 to alter the chromatin landscape and activate transcription of stemness regulators. Thus YY1AP1 may serve as a key molecular target for EpCAM(+) AFP(+) HCC subtype. Our results demonstrate the feasibility and power of a new strategy by utilizing well-defined patient samples and integrative genomics to uncover critical pathways linked to HCC subtypes with prognostic impact.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/metabolism , Genomics , Liver Neoplasms/metabolism , Nuclear Proteins/physiology , Transcription Factors/physiology , alpha-Fetoproteins/metabolism , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Cycle Proteins , Chromatin/metabolism , Epithelial Cell Adhesion Molecule , Humans , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcriptome
2.
Oncogene ; 32(25): 3091-100, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-22824796

ABSTRACT

Mutations in the genes encoding isocitrate dehydrogenase, IDH1 and IDH2, have been reported in gliomas, myeloid leukemias, chondrosarcomas and thyroid cancer. We discovered IDH1 and IDH2 mutations in 34 of 326 (10%) intrahepatic cholangiocarcinomas. Tumor with mutations in IDH1 or IDH2 had lower 5-hydroxymethylcytosine and higher 5-methylcytosine levels, as well as increased dimethylation of histone H3 lysine 79 (H3K79). Mutations in IDH1 or IDH2 were associated with longer overall survival (P=0.028) and were independently associated with a longer time to tumor recurrence after intrahepatic cholangiocarcinoma resection in multivariate analysis (P=0.021). IDH1 and IDH2 mutations were significantly associated with increased levels of p53 in intrahepatic cholangiocarcinomas, but no mutations in the p53 gene were found, suggesting that mutations in IDH1 and IDH2 may cause a stress that leads to p53 activation. We identified 2309 genes that were significantly hypermethylated in 19 cholangiocarcinomas with mutations in IDH1 or IDH2, compared with cholangiocarcinomas without these mutations. Hypermethylated CpG sites were significantly enriched in CpG shores and upstream of transcription start sites, suggesting a global regulation of transcriptional potential. Half of the hypermethylated genes overlapped with DNA hypermethylation in IDH1-mutant gliobastomas, suggesting the existence of a common set of genes whose expression may be affected by mutations in IDH1 or IDH2 in different types of tumors.


Subject(s)
Cholangiocarcinoma/genetics , Glioblastoma/genetics , Isocitrate Dehydrogenase/genetics , Liver Neoplasms/genetics , Base Sequence , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cholangiocarcinoma/metabolism , CpG Islands , DNA Methylation , Glioblastoma/metabolism , Histones/genetics , Humans , Liver Neoplasms/metabolism , Mutation , Neoplasm Recurrence, Local/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/metabolism
3.
Oncogene ; 32(37): 4397-405, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23085761

ABSTRACT

NANOG is a stem cell transcription factor that is essential for embryonic development, reprogramming normal adult cells and malignant transformation and progression. The nearly identical retrogene NANOGP8 is expressed in multiple cancers, but generally not in normal tissues and its function is not well defined. Our postulate is that NANOGP8 directly modulates the stemness of individual human colorectal carcinoma (CRC) cells. Stemness was measured in vitro as the spherogenicity of single CRC cells in serum-free medium and the size of the side population (SP) and in vivo as tumorigenicity and experimental metastatic potential in NOD/SCID mice. We found that 80% of clinical liver metastases express a NANOG with 75% of the positive metastases containing NANOGP8 transcripts. In all, 3-62% of single cells within six CRC lines form spheroids in serum-free medium in suspension. NANOGP8 is translated into protein. The relative expression of a NANOG gene increased 8- to 122-fold during spheroid formation, more than the increase in OCT4 or SOX2 transcripts with NANOGP8 the more prevalent family member. Short hairpin RNA (shRNA) to NANOG not only inhibits spherogenicity but also reduces expression of OCT4 and SOX2, the size of the SP and tumor growth in vivo. Inhibition of NANOG gene expression is associated with inhibition of proliferation and decreased phosphorylation of G2-related cell-cycle proteins. Overexpression of NANOGP8 rescues single-cell spherogenicity when NANOG gene expression is inhibited and increases the SP in CRC. Thus, NANOGP8 can substitute for NANOG in directly promoting stemness in CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nanog Homeobox Protein , Neoplasm Metastasis , Spheroids, Cellular , Tumor Cells, Cultured
5.
Oncogene ; 30(40): 4175-84, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21499307

ABSTRACT

Development of targeted therapy for hepatocellular carcinoma (HCC) remains a major challenge. We have recently identified an elevated expression of the fifth subunit of COP9 signalosome (CSN5) in early HCC as compared with dysplastic stage. In the present study, we explored the possibility of CSN5 being a potential therapeutic target for HCC. Our results show that CSN5 knockdown by small-interfering (si) RNA caused a strong induction of apoptosis and inhibition of cell-cycle progression in HCC cells in vitro. The down-regulation of CSN5 was sufficient to interfere with CSN function as evidenced by the accumulation of neddylated Cullin 1 and changes in the protein levels of CSN-controlled substrates SKP2, p53, p27 and nuclear factor-κB, albeit to a different degree depending on the HCC cell line, which could account for the CSN5 knockdown phenotype. The transcriptomic analysis of CSN5 knockdown signature showed that the anti-proliferative effect was driven by a common subset of molecular alterations including down-regulation of cyclin-dependent kinase 6 (CDK6) and integrin ß1 (ITGB1), which were functionally interconnected with key oncogenic regulators MYC and TGFß1 involved in the control of proliferation, apoptotic cell death and HCC progression. Consistent with microarray analysis, western blotting revealed that CSN5 depletion increased phosphorylation of Smad 2/3, key mediators of TGFß1 signaling, decreased the protein levels of ITGB1, CDK6 and cyclin D1 and caused reduced expression of anti-apoptotic Bcl-2, while elevating the levels of pro-apoptotic Bak. A chemically modified variant of CSN5 siRNA was then selected for in vivo application based on the growth inhibitory effect and minimal induction of unwanted immune response. Systemic delivery of the CSN5 3/8 variant by stable-nucleic-acid-lipid particles significantly suppressed the tumor growth in Huh7-luc+ orthotopic xenograft model. Taken together, these results indicate that CSN5 has a pivotal role in HCC pathogenesis and maybe an attractive molecular target for systemic HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/drug therapy , Peptide Hydrolases/metabolism , COP9 Signalosome Complex , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Division , Cell Line, Tumor , Down-Regulation , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Peptide Hydrolases/genetics , RNA, Small Interfering/genetics
6.
J Hepatol ; 53(3): 568-77, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20646772

ABSTRACT

The two dominant models of carcinogenesis postulate stochastic (clonal evolution) or hierarchic organization of tumor (cancer stem cell model). According to the latter, at the germinal center of tumor evolution is a cancer stem cell (CSC) which, similar to normal adult stem cells, possesses the capacity of self-renewal and a differentiation potential. Over the past few years, compelling evidence has emerged in support of the hierarchic cancer model for many solid tumors including hepatocellular cancers. The CSCs are posited to be responsible not only for tumor initiation but also for the generation of distant metastasis and relapse after therapy. These characteristics are particularly relevant for a multi-resistant tumor entity like human hepatocellular carcinoma and may herald a paradigm shift in the management of this deadly disease. Identification and detailed characterization of liver CSCs is therefore imperative for improving prevention approaches, enhancing early detection, and extending the limited treatment options. Despite the current progress in understanding the contribution of CSCs to the generation of heterogeneity of tumors, the molecular complexity and exact regulation of CSCs is poorly understood. This review focuses on the genetic and epigenetic mechanisms that regulate and define the unique CSC properties with an emphasis on key regulatory pathways of liver CSCs and their clinical significance.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Epigenesis, Genetic , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , AC133 Antigen , Aldehyde Dehydrogenase/metabolism , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Cell Adhesion Molecules/metabolism , Cell Division , Cell Separation/methods , Epithelial Cell Adhesion Molecule , Genes, myc , Glycoproteins/metabolism , Hedgehog Proteins/metabolism , Humans , Liver Neoplasms/metabolism , MicroRNAs/genetics , Models, Biological , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Peptides/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , RNA, Neoplasm/genetics , Receptors, Notch/metabolism , Repressor Proteins/genetics , Signal Transduction , Thy-1 Antigens/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
7.
Oncogene ; 28(40): 3526-36, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19617899

ABSTRACT

Growing evidence indicates that microRNAs have a significant role in tumor development and may constitute robust biomarkers for cancer diagnosis and prognosis. In this study, we evaluated the clinical and functional relevance of microRNA-122 (miR-122) expression in human hepatocellular carcinoma (HCC). We report that miR-122 is specifically repressed in a subset of primary tumors that are characterized by poor prognosis. We further show that the loss of miR-122 expression in tumor cells segregates with specific gene expression profiles linked to cancer progression, namely the suppression of hepatic phenotype and the acquisition of invasive properties. We identify liver-enriched transcription factors as central regulatory molecules in the gene networks associated with loss of miR-122, and provide evidence suggesting that miR-122 is under the transcriptional control of HNF1A, HNF3A and HNF3B. We further show that loss of miR-122 results in an increase of cell migration and invasion and that restoration of miR-122 reverses this phenotype. In conclusion, miR-122 is a marker of hepatocyte-specific differentiation and an important determinant in the control of cell migration and invasion. From a clinical point of view, our study emphasizes miR-122 as a diagnostic and prognostic marker for HCC progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/physiology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Expression Profiling , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , MicroRNAs/analysis , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Prognosis
8.
Neurobiol Dis ; 26(1): 112-24, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17270452

ABSTRACT

Williams Syndrome (WS, [MIM 194050]) is a disorder caused by a hemizygous deletion of 25-30 genes on chromosome 7q11.23. Several of these genes including those encoding cytoplasmic linker protein-115 (CYLN2) and general transcription factors (GTF2I and GTF2IRD1) are expressed in the brain and may contribute to the distinct neurological and cognitive deficits in WS patients. Recent studies of patients with partial deletions indicate that hemizygosity of GTF2I probably contributes to mental retardation in WS. Here we investigate whether CYLN2 and GTF2IRD1 contribute to the motoric and cognitive deficits in WS. Behavioral assessment of a new patient in which STX1A and LIMK1, but not CYLN2 and GTF2IRD1, are deleted showed that his cognitive and motor coordination functions were significantly better than in typical WS patients. Comparative analyses of gene specific CYLN2 and GTF2IRD1 knockout mice showed that a reduced size of the corpus callosum as well as deficits in motor coordination and hippocampal memory formation may be attributed to a deletion of CYLN2, while increased ventricle volume can be attributed to both CYLN2 and GTF2IRD1. We conclude that the motor and cognitive deficits in Williams Syndrome are caused by a variety of genes and that heterozygous deletion of CYLN2 is one of the major causes responsible for such dysfunctions.


Subject(s)
Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Muscle Proteins/genetics , Muscle Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Williams Syndrome/pathology , Williams Syndrome/psychology , Animals , Cognition/physiology , Conditioning, Operant/physiology , DNA/genetics , Eye Movements/physiology , Fear/psychology , In Situ Hybridization, Fluorescence , Intelligence Tests , Magnetic Resonance Imaging , Mice , Mice, Knockout , Motor Activity/physiology , Neuropsychological Tests , Postural Balance/physiology , Psychomotor Performance/physiology , Williams Syndrome/genetics
9.
Oncogene ; 25(27): 3801-9, 2006 Jun 26.
Article in English | MEDLINE | ID: mdl-16799621

ABSTRACT

Global gene expression profiling of hepatocellular carcinoma (HCC) is a promising new technology that has already refined the diagnosis and prognostic predictions of HCC patients. This has been accomplished by identifying genes whose expression pattern is associated with clinicopathological features of HCC tumors. Molecular characterization of HCC from gene expression profiling studies will undoubtedly improve the prediction of treatment responses, selection of treatments for specific molecular subtypes of HCC and ultimately the clinical outcome of HCC patients. The research focus is now shifting toward the identification of genetic determinants that are components of the specific regulatory pathways altered in cancers, and that may constitute novel therapeutic targets. Here we review the recent advances in gene expression profiling of HCC and discuss the future strategies for analysing large and complicated data sets from microarray studies and how to integrate these with diverse genomic data.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Genomics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Gene Expression Profiling/methods , Gene Expression Profiling/trends , Humans , Liver Neoplasms/pathology
10.
Eur J Cancer ; 39(14): 2093-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12957465

ABSTRACT

The signal transducer and activator of transcription (STAT)-3 regulates basic biological processes and it has been reported to be constitutively active in different types of malignant tumours. STAT-3 is active during the regenerative growth of the liver, but there are hardly any data about its presence in liver tumours. We investigated and found a high activity of STAT-3 using an electrophoretic mobility shift assay (EMSA) in chemically-induced rat hepatocellular carcinomas (HCCs). Dexamethasone treatment downregulated both STAT-3 activity and cell proliferation in the tumours. Therefore, the activity of the STAT-3 signal transduction pathway seems to be required for the growth of HCCs and could be a potential new target for therapeutic trials of this tumour type.


Subject(s)
Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Trans-Activators/metabolism , 2-Acetylaminofluorene/adverse effects , Alkylating Agents/adverse effects , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Blotting, Northern , Carcinogens/adverse effects , Carcinoma, Hepatocellular/chemically induced , Dexamethasone/therapeutic use , Diethylnitrosamine/adverse effects , Electrophoretic Mobility Shift Assay , Interleukin-6/metabolism , Liver Neoplasms/chemically induced , Male , RNA/metabolism , Rats , STAT3 Transcription Factor , Tumor Necrosis Factor-alpha/metabolism
11.
Cancer Res ; 61(18): 6665-8, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559531

ABSTRACT

The transforming growth factor-beta (TGF-beta) receptor complex and its downstream signaling intermediates constitute a tumor suppressor pathway. In many cancers, expression of TGF-beta type II receptor (TbetaR-II) is markedly decreased. In the present study, we show that the hepatocytes isolated from 15-day-old, but not 9-month-old, mice heterozygous for the deletion of the TbetaR-II gene are slightly less sensitive to the growth-inhibitory effect of TGF-beta when compared with wild-type littermates of same age. In addition, the proliferation index of hepatocytes as indicated by bromodeoxyuridine incorporation is mildly increased in the heterozygous mice. These subtle changes in cellular phenotype did not result in either gross or microscopic abnormality of the liver. The treatment of these mice with the chemical carcinogen, diethylnitrosamine, results in a significantly enhanced tumorigenesis in the liver when compared with the wild-type littermates. Our results demonstrate the gene-dosage effect of TbetaR-II and indicate that the reduced expression of TbetaR-II in mice increases susceptibility to tumorigenesis in the liver.


Subject(s)
Cell Transformation, Neoplastic/genetics , Liver Neoplasms, Experimental/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Carcinogens , Diethylnitrosamine , Female , Gene Dosage , Genes, cdc/physiology , Genetic Predisposition to Disease , Heterozygote , Liver/drug effects , Liver/metabolism , Liver/physiology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , Phenobarbital/pharmacology , Pregnancy , Protein Serine-Threonine Kinases , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
12.
Cytogenet Cell Genet ; 93(1-2): 114-6, 2001.
Article in English | MEDLINE | ID: mdl-11474192

ABSTRACT

DNA amplification is associated with genomic instability, the main characteristic of cancer cells, and it frequently involves protooncogenes. Double minute chromosomes (DM) and homogeneously stained regions (HSR) are cytological manifestations of DNA amplification. Gain of chromosome 19 is a recurrent alteration in mouse hepatocellular carcinoma (HCC). In one tumor cell line established from HCC developed in myc transgenic mice, DM derived from chromosome 19 were identified by spectral karyotyping and confirmed by fluorescence in situ hybridization (FISH). A probe generated by PCR from microdissected DM was localized by FISH on normal and HCC-derived cell lines on DM and chromosome 19 at two sites separated by several medium size G-bands. This organization of DM containing amplified sequences from separate loci of the same chromosome, indicates a complex mechanism of DNA amplification, possibly involving more than one gene. DM or HSR were not previously identified in mouse HCC and adult human HCC. The recognition of these loci could lead to the cloning of new genes or identification of known genes important in development or progression of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Chromosomes/genetics , Gene Amplification/genetics , Liver Neoplasms, Experimental/genetics , Mutation/genetics , Animals , Chromosome Banding , DNA Probes , Disease Progression , Genes, myc/genetics , In Situ Hybridization, Fluorescence , Karyotyping , Mice , Mice, Transgenic , Tumor Cells, Cultured
13.
Hepatology ; 34(1): 32-41, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11431731

ABSTRACT

NF-kappaB regulates liver cell death during development, regeneration, and neoplastic transformation. For example, we showed that oncogenic Ras- or Raf-mediated transformation of rat liver epithelial cells (RLEs) led to altered NF-kappaB regulation through IKK complex activation, which rendered these cells more resistant to TGF-beta1-induced apoptosis. Thus, based on these findings, we sought to determine whether NF-kappaB could also be involved in tumor growth of liver cells in vivo. Hepatocellular carcinomas (HCCs) derived from bitransgenic mice harboring TGF-alpha and c-myc transgenes targeted specifically to the liver were compared with HCCs from c-myc single transgenic mice. Tumors from bitransgenic mice are characterized by a higher frequency of appearance, lower apoptotic index, and a higher rate of cell proliferation. Here we show that NF-kappaB is activated in HCCs of double TGF-alpha/c-myc transgenic mice, but not of c-myc single transgenic mice, suggesting that TGF-alpha mediates induction of NF-kappaB. Activation of the IKK complex was observed in the HCCs of double TGF-alpha/c-myc transgenic mice, implicating this pathway in NF-kappaB induction. Lastly, activation of the Akt/protein kinase B (PKB), which has recently been implicated in NF-kappaB activation by PDGF, TNF-alpha, and Ras, was also observed. Importantly, human HCC cell lines similarly displayed NF-kappaB activation. Thus, these studies elucidate an anti-apoptotic mechanism by a TGF-alpha-Akt/PKB-IKK pathway, which likely contributes to survival and proliferation, thereby accelerating c-myc-induced liver neoplastic development in vivo.


Subject(s)
Carrier Proteins/physiology , Liver Neoplasms, Experimental/metabolism , NF-kappa B/biosynthesis , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins/physiology , Transforming Growth Factor alpha/genetics , Animals , Apoptosis , Cell Division , Enzyme Activation , Gene Expression , Intracellular Signaling Peptides and Proteins , Liver/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc/physiology , RNA-Binding Proteins , Transforming Growth Factor alpha/physiology
14.
Genomics ; 73(1): 20-7, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11352562

ABSTRACT

Transgenic mice expressing c-myc under the control of the albumin promoter and enhancer develop liver tumors and have served as a useful model for studying the progression of hepatocarcinogenesis. The chromosomes of one line of c-myc transgenic mice carry the reciprocal translocation t(5;6)(G1;F2) adjacent to the transgene insertion site on the 5G1-ter segment translocated to chromosome 6. To characterize the genomic alterations in the c-myc transgenic animals, we have cloned the mouse DNA flanking the transgene array. By linkage mapping, the transgene integration site was localized to the region of distal chromosome 5 syntenic to the region on human chromosome 7q11.23 that is hemizgygously deleted in Williams-Beuren syndrome, a multisystemic developmental disorder. Comparison of the genomic DNA structure in wildtype and transgenic mice revealed that the transgene integration had induced an approximately 40-kb deletion, starting downstream of the Cyln2 gene and including the first exon of the Gtf2ird1 gene. Gtf2ird1 encodes a polypeptide related to general transcription factor TFII-I, and it is the mouse orthologue of GTF2IRD1 (WBSCR11), one of the genes commonly deleted in Williams-Beuren syndrome patients. Loss of the 5' end of the Gtf2ird1 gene resulted in greatly reduced expression of Gtf2ird1 mRNA in mice homozygous for the transgene.


Subject(s)
Genes, myc , Transcription Factors/genetics , Williams Syndrome/genetics , Animals , Base Sequence , Chromosome Mapping/methods , Chromosomes, Human, Pair 7 , DNA , Exons , Gene Deletion , Gene Expression , Genetic Linkage , Helix-Loop-Helix Motifs , Humans , Mice , Mice, Transgenic , Models, Animal , Molecular Sequence Data , RNA, Messenger/genetics , Transgenes , Translocation, Genetic
15.
Carcinogenesis ; 22(6): 975-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375907

ABSTRACT

The mechanisms underlying peroxisome proliferator-induced hepatocarcinogenesis are not understood. Because of the uncertainty of human cancer risk associated with peroxisome proliferators, delineating the mechanisms of carcinogenesis by these agents is of great interest. Alterations in liver growth factors were postulated to contribute to the carcinogenic effect of peroxisome proliferators. Administration of these compounds to rodents results in down-regulation of hepatocyte growth factor (HGF) and supplementing culture medium with HGF is reported to suppress cell proliferation of preneoplastic and neoplastic cells from WY-14,643-treated livers. Combined, these observations suggest that reduced levels of hepatic HGF contribute to the mechanisms underlying peroxisome proliferator-induced hepatocarcinogenesis. To determine if HGF can prevent the effects of peroxisome proliferators in liver, the short-term influence of WY-14,643 in two different lines of HGF transgenic mice was examined. Mice were fed either a control diet or one containing 0.1% WY-14-643 for one week. Hepatomegaly was found in both HGF transgenic mouse lines fed WY-14,643 compared with controls. Additionally, hepatic expression of typical mRNA markers of peroxisome proliferation including those encoding peroxisomal fatty acid metabolizing enzymes and cell cycle control proteins were all significantly elevated in HGF transgenic mice fed WY-14,643 compared with controls. Down-regulation of HGF was found to be dependent on PPARalpha since lower levels of HGF mRNA and protein were observed in wild-type mice fed WY-14,643 for 1 week and not in similarly treated PPARalpha-null mice. These results demonstrate that the early increase in hepatic mRNAs associated with peroxisome and cell proliferation induced by WY-14,643 treatment can not be prevented by overexpression of HGF in vivo.


Subject(s)
Hepatocyte Growth Factor/physiology , Hepatocytes/drug effects , Peroxisome Proliferators/toxicity , Animals , Carcinogens/toxicity , Cell Division/drug effects , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hepatocytes/cytology , Humans , Liver Neoplasms, Experimental/chemically induced , Mice , Mice, Transgenic , Pyrimidines/toxicity , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Am J Pathol ; 158(4): 1313-23, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11290549

ABSTRACT

We have analyzed the histological changes in rat liver after 2-acetylaminofluorene (AAF) administration. The data demonstrate that AAF-induced oval cells were preferentially generated by proliferation of the terminal biliary ductules that we suggest constitute the primary hepatic stem cell niche. The oval cells formed ductular structures, representing an extension of the canals of Hering. This histological organization provides continuous bile drainage of the hepatocytes and uninterrupted blood flow in the sinusoids. The oval cell ductules are surrounded by a continuous basement membrane that is intermittently disrupted by processes of stellate cells that form direct cell-cell contact with the oval cells. Although both AAF treatment and bile duct ligation results in proliferation of biliary epithelial cells, the mechanism(s) responsible for the proliferation of the biliary epithelium seems to differ in the two models. In contrast to the biliary proliferation stimulated by bile ligation, AAF-induced oval cell proliferation as well as the capacity of these cells to differentiate into hepatocytes, bile epithelial cells and possibly other cell lineages can be blocked by administration of dexamethasone.


Subject(s)
Liver/cytology , 2-Acetylaminofluorene/pharmacology , Animals , Bromodeoxyuridine/metabolism , Cell Division , Cell Size , Common Bile Duct , Hepatectomy/methods , Immunohistochemistry , Ligation , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Rats , Rats, Inbred F344 , Time Factors
17.
Cancer Res ; 61(5): 2085-91, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280770

ABSTRACT

Mutations affecting phosphorylation sites in the beta-catenin gene have been implicated in the development of human and rodent hepatocellular carcinomas (HCCs). To further investigate the involvement of this gene in hepatocarcinogenesis, we used several transgenic mouse models of hepatic tumors induced by overexpression of c-myc in the liver either alone or in combination with transforming growth factor (TGF) alpha or TGF-beta1. Activation of beta-catenin, as judged by the presence of mutations and/or nuclear translocation of the protein, was most frequent in liver tumors from c-myc (4/17; 23.5%) and c-myc/TGF-beta1 (6/18; 33.3%) transgenic mice. However, it was very rare in faster growing and histologically more aggressive HCCs developed in c-myc/TGF-alpha mice (1/20; 5%). Administration of diethylnitrosamine, phenobarbital, or 2-amino-3,8-diethylimidazo[4,5-f]quinoxaline did not significantly affect the occurrence of beta-catenin mutations. Notably, nuclear accumulation of beta-catenin was observed only in adenomas and highly differentiated carcinomas with eosinophilic phenotype. Furthermore, preneoplastic lesions with eosinophilic phenotype frequently displayed focal nuclear positivity, colocalized with areas of high proliferation. In contrast, basophilic and clear-cell foci, as well as pseudo-glandular and poorly differentiated HCCs, exhibited a normal or reduced membranous immunoreactivity for beta-catenin. These studies suggest that nuclear translocation of beta-catenin and activation of Wingless/Wnt signaling may represent an early event in liver carcinogenesis, providing a growth advantage in a subset of hepatic tumors with a more differentiated phenotype.


Subject(s)
Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms, Experimental/genetics , Trans-Activators , Animals , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/metabolism , Genes, myc/genetics , Humans , Immunohistochemistry , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-myc/biosynthesis , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor alpha/genetics , Tumor Cells, Cultured , beta Catenin
18.
Hepatology ; 33(2): 339-45, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11172335

ABSTRACT

The liver has an extremely effective regenerative capacity. When 70% of a rat liver is removed by surgery, the liver mass regrows in 7 to 10 days by the compensatory hyperplasia of the remnant part. In case of damage to the surviving hepatocytes, the facultative stem-cell compartment is activated and the liver regenerates by means of oval-cell proliferation/differentiation. In the present study, we demonstrate that when both hepatocyte proliferation and stem-cell activation were prevented by dexamethasone (Dex) administration, the liver mass was restored in the absence of DNA synthesis. The restoration of the liver was accomplished by the preferential enlargement/hypertrophy of the periportal hepatocytes. A similar response was observed when cell proliferation was arrested by 5-fluorouracil (FU) following partial hepatectomy. Therefore, the hepatocytic hypertrophy appears to provide an alternative mechanism of liver-mass restoration. This hypertrophic condition of the liver is not stable, because following the withdrawal of Dex, the enlarged hepatocytes entered in the cell cycle and the normal liver structure and DNA content was re-established.


Subject(s)
Hepatocytes/pathology , Liver Regeneration/physiology , Liver/pathology , Animals , Bromodeoxyuridine/metabolism , Cell Division/drug effects , DNA/biosynthesis , Dexamethasone/pharmacology , Fluorouracil/pharmacology , Glucocorticoids/pharmacology , Hepatectomy/methods , Hypertrophy , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Inbred F344
19.
Hepatology ; 32(6): 1248-54, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093731

ABSTRACT

Tissue inhibitor of metalloproteinases-1 (TIMP-1) has been shown to be increased in liver fibrosis development both in murine experimental models and human samples. However, the direct role of TIMP-1 during liver fibrosis development has not been defined. To address this issue, we developed transgenic mice overexpressing human TIMP-1 (hTIMP-1) in the liver under control of the albumin promoter/ enhancer. A model of CCl(4)-induced hepatic fibrosis was used to assess the extent of fibrosis development in TIMP-1 transgenic (TIMP-Tg) mice and control hybrid (Cont) mice. Without any treatment, overexpression of TIMP-1 itself did not induce liver fibrosis. There were no significant differences of pro-(alpha1)-collagen-I, (alpha2)-collagen-IV, and alpha-smooth muscle actin (alpha-SMA) mRNA expression in the liver between TIMP-Tg and Cont-mice, suggesting that overexpression of TIMP-1 itself did not cause hepatic stellate cell (HSC) activation. After 4-week treatment with CCl(4), however, densitometric analysis revealed that TIMP-Tg-mice had a seven-fold increase in liver fibrosis compared with the Cont-mice. The hepatic hydroxyproline content and serum hyaluronic acid were also significantly increased in TIMP-Tg-mice, whereas CCl(4)-induced liver dysfunction was not altered. An active form of matrix metalloproteinases-2 (MMP-2) level in the liver of TIMP-Tg-mice was decreased relative to that in Cont-mice because of the transgenic TIMP-1. Immunohistochemical analysis revealed that collagen-I and collagen-IV accumulation was markedly increased in the liver of CCl(4)-treated TIMP-Tg-mice with a pattern similar to that of alpha-SMA positive cells. These results suggest that TIMP-1 does not by itself result in liver fibrosis, but strongly promotes liver fibrosis development.


Subject(s)
Liver Cirrhosis/chemically induced , Tissue Inhibitor of Metalloproteinase-1 , Actins/genetics , Actins/metabolism , Animals , Carbon Tetrachloride/pharmacology , Collagen/genetics , Humans , Immunohistochemistry , Liver/drug effects , Liver/enzymology , Liver/metabolism , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic/genetics , Muscle, Smooth/metabolism , RNA, Messenger/metabolism , Reference Values , Tissue Inhibitor of Metalloproteinase-1/genetics
20.
Oncogene ; 19(44): 5054-62, 2000 Oct 19.
Article in English | MEDLINE | ID: mdl-11042693

ABSTRACT

Deregulation of E2F transcriptional control has been implicated in oncogenic transformation. Consistent with this idea, we recently demonstrated that during hepatocarcinogenesis in c-myc/TGFalpha double transgenic mice, there is increased expression of E2F-1 and E2F-2, as well as induction of putative E2F target genes. Therefore, we generated transgenic mice expressing E2F-1 under the control of the albumin enhancer/promoter to test the hypothesis that E2F family members may contribute to liver tumor development. Overexpression of E2F-1 resulted in mild but persistent increases in cell proliferation and death during postnatal liver growth, and no increases in hepatic regenerative growth in response to partial hepatectomy. Nevertheless, from 2 months postnatally E2F-1 transgenic mice exhibited prominent hepatic histological abnormalities including preneoplastic foci adjacent to portal tracts and pericentral large cell dysplasia. From 6 to 8 months onward, there was an abrupt increase in the number of neoplastic nodules ('adenomas') with 100% incidence by 10 months. Some adenomas showed evidence of malignant transformation, and two of six mice killed at 12 months showed trabecular hepatocellular carcinoma. Endogenous c-myc was up-regulated in the early stages of E2F-1 hepatocarcinogenesis, whereas p53 was overexpressed in the tumors, suggesting that both E2F-1-mediated proliferation and apoptosis are operative but at different stages of hepatocarcinogenesis. In conclusion, E2F-1 overexpression in the liver causes dysplasia and tumors and suggests a cooperation between E2F-1 and c-myc oncogenes during liver oncogenesis.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins , Liver Neoplasms, Experimental/genetics , Transcription Factors/physiology , Albumins/genetics , Animals , Apoptosis/physiology , Cell Division/physiology , Cell Transformation, Neoplastic/metabolism , Crosses, Genetic , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic , Genes, myc/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/physiology , Humans , Liver/metabolism , Liver/pathology , Liver/physiology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Liver Regeneration/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...