Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nat Commun ; 10(1): 83, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622257

ABSTRACT

Mitochondrial topoisomerase IB (TOP1MT) is a nuclear-encoded topoisomerase, exclusively localized to mitochondria, which resolves topological stress generated during mtDNA replication and transcription. Here, we report that TOP1MT is overexpressed in cancer tissues and demonstrate that TOP1MT deficiency attenuates tumor growth in human and mouse models of colon and liver cancer. Due to their mitochondrial dysfunction, TOP1MT-KO cells become addicted to glycolysis, which limits synthetic building blocks and energy supply required for the proliferation of cancer cells in a nutrient-deprived tumor microenvironment. Mechanistically, we show that TOP1MT associates with mitoribosomal subunits, ensuring optimal mitochondrial translation and assembly of oxidative phosphorylation complexes that are critical for sustaining tumor growth. The TOP1MT genomic signature profile, based on Top1mt-KO liver cancers, is correlated with enhanced survival of hepatocellular carcinoma patients. Our results highlight the importance of TOP1MT for tumor development, providing a potential rationale to develop TOP1MT-targeted drugs as anticancer therapies.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , DNA Topoisomerases, Type I/metabolism , Liver Neoplasms, Experimental/pathology , Liver Neoplasms/pathology , Mitochondria/metabolism , Protein Biosynthesis , Animals , Carcinogens/toxicity , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Nucleus/metabolism , Cell Proliferation , DNA Topoisomerases, Type I/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/isolation & purification , Datasets as Topic , Energy Metabolism , Female , Fibroblasts , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glycolysis , HCT116 Cells , Humans , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Knockout , Mice, Nude , Mitochondria/pathology , Prognosis , Survival Analysis , Xenograft Model Antitumor Assays
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Biol Chem ; 276(5): 3106-14, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11060283

ABSTRACT

Animal thioredoxin reductases (TRs) are selenocysteine-containing flavoenzymes that utilize NADPH for reduction of thioredoxins and other protein and nonprotein substrates. Three types of mammalian TRs are known, with TR1 being a cytosolic enzyme, and TR3, a mitochondrial enzyme. Previously characterized TR1 and TR3 occurred as homodimers of 55-57-kDa subunits. We report here that TR1 isolated from mouse liver, mouse liver tumor, and a human T-cell line exhibited extensive heterogeneity as detected by electrophoretic, immunoblot, and mass spectrometry analyses. In particular, a 67-kDa band of TR1 was detected. Furthermore, a novel form of mouse TR1 cDNA encoding a 67-kDa selenoprotein subunit with an additional N-terminal sequence was identified. Subsequent homology analyses revealed three distinct isoforms of mouse and rat TR1 mRNA. These forms differed in 5' sequences that resulted from the alternative use of the first three exons but had common downstream sequences. Similarly, expression of multiple mRNA forms was observed for human TR3 and Drosophila TR. In these genes, alternative first exon splicing resulted in the formation of predicted mitochondrial and cytosolic proteins. In addition, a human TR3 gene overlapped with the gene for catechol-O-methyltransferase (COMT) on a complementary DNA strand, such that mitochondrial TR3 and membrane-bound COMT mRNAs had common first exon sequences; however, transcription start sites for predicted cytosolic TR3 and soluble COMT forms were separated by approximately 30 kilobases. Thus, this study demonstrates a remarkable heterogeneity within TRs, which, at least in part, results from evolutionary conserved genetic mechanisms employing alternative first exon splicing. Multiple transcription start sites within TR genes may be relevant to complex regulation of expression and/or organelle- and cell type-specific location of animal thioredoxin reductases.


Subject(s)
Alternative Splicing , Genetic Variation , Thioredoxin-Disulfide Reductase/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/analysis , Drosophila/enzymology , Drosophila/genetics , Exons , Humans , Introns , Male , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Rats , Sequence Homology, Amino Acid , Thioredoxin-Disulfide Reductase/isolation & purification
11.
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
12.
Gene ; 256(1-2): 59-67, 2000 Oct 03.
Article in English | MEDLINE | ID: mdl-11054536

ABSTRACT

Cyclins control cell-cycle progression by regulating the activity of cyclin-dependent kinases. Cyclin I was recently added to the cyclin family of proteins because of the presence of a cyclin box motif in the deduced amino-acid sequence. Cyclin I may share functional roles with cyclin G1 and G2 because of the high structural similarity between their deduced amino-acid sequences. However, the biological and functional roles of this subclass of cyclins remain obscure. The mouse cyclin G1 and G2 genes have previously been cloned and characterized. In this report, we describe the cloning of the mouse homolog of cyclin I. The cyclin I cDNA sequence was used to determine the genomic organization of the mouse cyclin I gene which co-localizes with cyclin G2 to chromosome 5E3.3-F1.3. Cyclin I was transcribed from seven exons distributed over more than 19kb of genomic sequence. The expression of cyclin I was determined in various tissues, but no clear correlation with the proliferative state was found. Furthermore, in contrast to cyclin G1, cyclin I expression was stable during cell-cycle progression after partial hepatectomy in both the absence and presence of DNA damage. Transient expression of cyclin I-green fluorescent protein (GFP) fusion proteins in cell lines showed that cyclin I was distributed throughout the cell in contrast with the mainly cytoplasmic localization of cyclin G2 and nuclear localization of cyclin G1. Our results indicate that despite the close structural similarity between cyclin G1, G2 and I, these three proteins are likely to have distinct biological roles.


Subject(s)
Cyclins/genetics , Genes/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Cloning, Molecular , Cyclin I , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Exons , Gene Expression , Introns , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Tissue Distribution
13.
Cancer Res ; 60(16): 4531-7, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969803

ABSTRACT

Transforming growth factor (TGF)-beta1 functions as a tumor suppressor in vivo. Using transgenic mice, we show that hepatic TGF-beta1 overexpression inhibits abundance of the cyclin-dependent kinase activating tyrosine phosphatase cdc25A protein. The reduction in cdc25A protein levels was associated with increased binding of histone deacetylase 1 to p130 in the hepatic extracts. In cultured cells, HDAC1/p130 overexpression inhibited activity of the cdc25A promoter through an E2F site. TGF-beta1 treatment enhanced p130 binding to the cdc25A promoter E2F site assessed in chromatin immunoprecipitation assays. Hepatic proliferation induced by partial hepatectomy was associated with a decrease in the amount of HDAC1 bound to p130, without a significant decrease in p130 abundance, suggesting that HDAC1 binding to p130 may be regulated by proliferative stimuli. The induction of cdc25A abundance induced by partial hepatectomy correlated with the induction of DNA synthesis. These studies suggest that TGF-beta1 may enhance HDAC1 binding to p130 in vivo, thereby inhibiting cdc25A gene expression. TGF-beta1 regulation of HDAC1/pocket protein associations may provide a link between chromatin remodeling proteins and cdk inhibition through induction of cdc25A in vivo.


Subject(s)
Histone Deacetylases/metabolism , Proteins , Transforming Growth Factor beta/physiology , Viral Structural Proteins/metabolism , Animals , Cell Division/physiology , HeLa Cells , Hepatectomy , Histone Deacetylase 1 , Humans , Liver/metabolism , Liver Regeneration/physiology , Male , Mice , Mice, Transgenic , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation , Precipitin Tests , Retinoblastoma-Like Protein p130 , Transfection , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Viral Structural Proteins/biosynthesis , cdc25 Phosphatases/antagonists & inhibitors , cdc25 Phosphatases/metabolism
14.
Am J Pathol ; 156(4): 1253-61, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751351

ABSTRACT

Polypeptide growth factors stimulate mammalian cell proliferation by binding to specific cell surface receptors. This interaction triggers numerous biochemical responses including the activation of protein phosphorylation cascades and the enhanced expression of specific genes. We have identified several fibroblast growth factor (FGF)-inducible genes in murine NIH 3T3 cells and recently reported that one of them, the FGF-inducible 14 (Fn14) immediate-early response gene, is predicted to encode a novel, cell surface-localized type Ia transmembrane protein. Here, we report that the human Fn14 homolog is located on chromosome 16p13.3 and encodes a 129-amino acid protein with approximately 82% sequence identity to the murine protein. The human Fn14 gene, like the murine Fn14 gene, is expressed at elevated levels after FGF, calf serum or phorbol ester treatment of fibroblasts in vitro and is expressed at relatively high levels in heart and kidney in vivo. We also report that the human Fn14 gene is expressed at relatively low levels in normal liver tissue but at high levels in liver cancer cell lines and in hepatocellular carcinoma specimens. Furthermore, the murine Fn14 gene is rapidly induced during liver regeneration in vivo and is expressed at high levels in the hepatocellular carcinoma nodules that develop in the c-myc/transforming growth factor-alpha-driven and the hepatitis B virus X protein-driven transgenic mouse models of hepatocarcinogenesis. These results indicate that Fn14 may play a role in hepatocyte growth control and liver neoplasia.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation , Gene Expression , Genes, Immediate-Early , Liver Neoplasms/genetics , Liver Regeneration/genetics , Membrane Proteins/genetics , Receptors, Tumor Necrosis Factor , 3T3 Cells , Amino Acid Sequence/genetics , Animals , Cells, Cultured , Chromosome Mapping , DNA, Complementary/genetics , Humans , Liver/metabolism , Mice , Mice, Inbred Strains , Molecular Sequence Data , RNA, Messenger/metabolism , TWEAK Receptor
15.
Toxicol Lett ; 112-113: 553-5, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10720779

ABSTRACT

Double transgenic mice bearing fusion genes consisting of mouse albumin enhancer/promoter-mouse c-myc cDNA and mouse metallothionein 1 promoter-human TGF-alpha cDNA were generated to investigate the interaction of these genes in hepatic oncogenesis and to provide a general paradigm for characterizing both the interaction of nuclear oncogenes and growth factors in tumorigenesis. In addition, these mice provide an experimental model to test how environmental chemicals might interact with the c-myc and TGF-alpha transgenes during the neoplastic process. We show experimental evidence that co-expression of TGF-alpha and c-myc transgenes in mouse liver promotes overproduction of ROS and thus creates an oxidative stress environment. This phenomenon may account for the massive DNA damage and acceleration of hepatocarcinogenesis observed in the TGF-alpha/c-myc mouse model. Also, the role of mutagenesis in hepatocarcinogenesis induced by 2-amino-3,8-dimethylimidazo(4,5-f)-quinoxaline (MeIQx) was demonstrated in C57BL/lacZ (Muta Mice) and double transgenic c-myc/lacZ mice that carry the lacZ mutation reporter gene. The MeLQx hepatocarcinogenicity was associated with an increase in in vivo mutagenicity as scored by mutations in the lacZ reporter gene. These results suggest that transgenic mouse models may provide important tools for testing both the carcinogenic potential of environmental chemicals and the interaction/cooperation of these compounds with specific genes during the neoplastic process.


Subject(s)
Disease Models, Animal , Liver Neoplasms, Experimental/genetics , Mice, Transgenic , Animals , Mice , Proto-Oncogene Proteins c-myc/genetics , Research , Transforming Growth Factor alpha/genetics
16.
Proc Natl Acad Sci U S A ; 97(5): 2196-201, 2000 Feb 29.
Article in English | MEDLINE | ID: mdl-10681450

ABSTRACT

We have previously shown that chronic activation of mitogenic signaling induced by over-expression of c-myc and transforming growth factor-alpha (TGFalpha) transgenes in mouse liver induces a state of oxidative stress. We therefore proposed that increased reactive oxygen species (ROS) generation might be responsible for the extensive chromosomal damage and acceleration of hepatocarcinogenesis characteristic for TGFalpha/c-myc mice. In this study, we show that vitamin E (VE), a potent free radical scavenging antioxidant, is able to protect liver tissue against oxidative stress and suppress tumorigenic potential of c-myc oncogene. Dietary supplementation with VE, starting from weaning, decreased ROS generation coincident with a marked inhibition of hepatocyte proliferation while increasing the chromosomal as well as mtDNA stability in the liver. Similarly, dietary VE reduced liver dysplasia and increased viability of hepatocytes. At 6 mo of age, VE treatment decreased the incidence of adenomas by 65% and prevented malignant conversion. These results indicate that ROS generated by over-expression of c-myc and TGFalpha in the liver are the primary carcinogenic agents in this animal model. Furthermore, the data demonstrate that dietary supplementation of VE can effectively inhibit liver cancer development.


Subject(s)
Antioxidants/metabolism , Antioxidants/pharmacology , Liver Neoplasms/prevention & control , Vitamin E/analogs & derivatives , alpha-Tocopherol/analogs & derivatives , Animals , DNA Damage , DNA, Mitochondrial , Dietary Supplements , Disease Models, Animal , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reactive Oxygen Species/metabolism , Tocopherols , Vitamin E/metabolism , Vitamin E/pharmacology
17.
Am J Pathol ; 154(6): 1693-700, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362794

ABSTRACT

We have previously shown in transgenic mice that transforming growth factor (TGF)-alpha dramatically enhances c-myc-induced hepatocarcinogenesis by promoting proliferation and survival of hepatocellular carcinoma (HCC) cells. As transgenic livers display increased levels of mature TGF-beta1 from the early stages of hepatocarcinogenesis, we have now assessed whether impairment of TGF-beta1 signaling contributes to the deregulation of cell cycle progression and apoptosis observed during this process. Focal preneoplastic lesions lacking expression of TGF-beta receptor type II (TbetaRII) were detected in c-myc/TGF-alpha but not in c-myc livers. In c-myc/TGF-alpha mice, 40% (2/5) of adenomas and 90% (27/30) of HCCs showed down-regulation of TbetaRII expression in comparison with 11% (2/18) of adenomas and 47% (14/30) of HCCs in c-myc mice. Down-regulation of the TGF-beta1-inducible p15(INK4B) mRNA and reduced apoptotic rates in TbetaRII-negative HCCs further indicated the disruption of TGF-beta1 signaling. Furthermore, both TbetaRII-negative and -positive c-myc TGF-alpha HCCs, but not c-myc HCCs, were characterized by decreased levels of the cell cycle inhibitor p27. These results suggest 1) an inverse correlation of decreased p27 expression with the particularly strong expression of TGF-alpha in these lesions, consistent with the capacity of TGF-alpha signaling to post-transcriptionally regulate p27, and 2) the presence of alternative, downstream defects of TGF-beta1 signaling in c-myc/TGF-alpha HCCs that may impair the growth-inhibitory response to TGF-beta1. Thus, the accelerated neoplastic development in c-myc/TGF-alpha mice is associated with an early and frequent occurrence of TbetaRII-negative lesions and with reduced levels of p27 in HCC cells, indicating that disruption of TGF-beta1 responsiveness may play a crucial role in the enhancement of c-myc-induced hepatocarcinogenesis by TGF-alpha.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16 , Liver Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Signal Transduction/genetics , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins , Animals , Blotting, Northern , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carrier Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Down-Regulation , Enzyme Inhibitors/metabolism , Immunohistochemistry , Liver Neoplasms, Experimental/genetics , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , RNA, Messenger/biosynthesis , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/physiology
18.
Lab Invest ; 79(2): 103-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10068199

ABSTRACT

Many acute and chronic liver diseases are often associated with atypical ductular proliferation (ADP). These ADPs have gained increasing interest since a number of recent observations suggest that ADPs may represent progenies of the putative liver stem cell compartment. In this study, we show that feeding mice with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) results in persistent proliferation of primitive ductules with poorly defined lumens. Similar to oval cell proliferation in other rodent models as well as in various human liver diseases, DDC-induced ADP originated from the portal tract, spread into the hepatic lobule, and was associated closely with appearance of hepatocytes harboring an antigen (A6), which normally is expressed in biliary epithelium. Furthermore, DDC treatment severely inhibited the regenerative capacity of mice after partial hepatectomy. The development of ADP was selectively blocked in DDC-fed TGF-beta1 transgenic mice producing active TGF-beta1 in the liver and no accumulation of new hepatocytes expressing the A6 antigen was observed. Moreover, the transforming growth factor beta1 (TGF-beta1) transgenic mice did not survive beyond 3 weeks from starting the DDC-containing diet. The results suggest that persistent activation of the hepatic stem cell compartment is essential for liver regeneration in the DDC model and that active TGF-beta1 may negatively control activation of stem cells in the liver. These data further emphasize the relevance of the DDC model as an experimental tool for studying chronic liver diseases.


Subject(s)
Bile Ducts, Intrahepatic/drug effects , Bile Ducts, Intrahepatic/pathology , Liver Diseases, Alcoholic/pathology , Transforming Growth Factor beta/pharmacology , Animals , Cell Division/drug effects , Chemical and Drug Induced Liver Injury , Chronic Disease , Common Bile Duct , Dicarbethoxydihydrocollidine , Disease Models, Animal , Epithelial Cells/pathology , Hepatectomy/methods , Ligation , Liver/drug effects , Liver/pathology , Liver Regeneration/drug effects , Mice , Mice, Transgenic/genetics , Transforming Growth Factor beta/genetics
19.
Biochem Biophys Res Commun ; 251(2): 488-93, 1998 Oct 20.
Article in English | MEDLINE | ID: mdl-9792801

ABSTRACT

There is strong evidence that selenium protects against certain human cancers, but the underlying mechanism is unknown. Glutathione peroxidase (GPX1) and thioredoxin reductase (TR), the most abundant antioxidant selenium-containing proteins in mammals, have been implicated in this protection. We analyzed the expression of TR and GPX1 in the following model cancer systems: (1) liver tumors in TGFalpha/c-myc transgenic mice; (2) human prostate cell lines from normal and cancer tissues; and (3) p53-induced apoptosis in a human colon cancer cell line. TR was induced while GPX1 was repressed in malignancies relative to controls in transgenic mice and prostate cell lines. In the colon cell line, p53 expression resulted in elevated GPX1, but repressed TR. The data indicate that TR and GPX1 are regulated in a contrasting manner in the cancer systems tested and reveal the p53-dependent regulation of selenoprotein expression. The data suggest that additional studies on selenoprotein regulation in different cancers are required to evaluate future implementation of selenium as a dietary supplement in individuals at risk for developing certain cancers.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Colonic Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glutathione Peroxidase/genetics , Liver Neoplasms/enzymology , Prostatic Neoplasms/enzymology , Thioredoxin-Disulfide Reductase/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Cell Line , Colonic Neoplasms/genetics , Enzyme Induction , Epithelial Cells/enzymology , Genes, myc , Glutathione Peroxidase/biosynthesis , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Prostate/enzymology , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Thioredoxin-Disulfide Reductase/biosynthesis , Transcription, Genetic , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
20.
Biochem Biophys Res Commun ; 249(1): 1-5, 1998 Aug 10.
Article in English | MEDLINE | ID: mdl-9705820

ABSTRACT

To further elucidate the role of CCAAT/Enhancer Binding Protein alpha (C/EBP alpha) in hepatocyte differentiation, we investigated fetal and newborn C/EBP alpha-deficient (C/EBP alpha -/-) mice using confocal microscopy and markers specific for hepatocyte (AFP) and biliary epithelial cell (A6) differentiation. Histologically, in fetal liver of C/EBP alpha -/- mice, pseudoglandular structures appeared starting at 16.5 days of gestation. In newborn livers, the diameters of these structures greatly increased. They were randomly distributed between portal and central veins and interfered with the establishment of normal hepatic plates. However, the portal bile ducts developed normally. The pseudoglandular structures were lined with small hepatocytes with round nuclei and were positive for both AFP and A6 antigens. These data show that C/EBP alpha -/- hepatocytes exhibit biliary epithelial cell characters and suggest an involvement of C/EBP alpha in the control of the switch in the differentiation of bi-potential hepatoblasts along the hepatocyte lineage.


Subject(s)
Bile Ducts/cytology , Bile Ducts/physiology , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Liver/cytology , Liver/physiology , Nuclear Proteins/genetics , Animals , CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Nuclear Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...