Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Oncol Lett ; 12(2): 1591-1596, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27446476

ABSTRACT

Human hepatocellular carcinoma (HCC) is one of the most common types of cancer and has a very poor prognosis; thus, the development of effective therapies for the treatment of advanced HCC is of high clinical priority. In the present study, the anti-oncogenic effect of combined knockdown of c-Myc expression and ectopic restoration of deleted in liver cancer 1 (DLC1) expression was investigated in human liver cancer cells. Expression of c-Myc in human HCC cells was knocked down by stable transfection with a Myc-specific short hairpin (sh) RNA vector. DLC1 expression in Huh7 cells was restored by adenovirus transduction, and the effects of DLC1 expression and c-Myc knockdown on Ras homolog gene family, member A (RhoA) levels, cell proliferation, soft agar colony formation and cell invasion were measured. Downregulation of c-Myc or re-expression of DLC1 led to a marked reduction in RhoA levels, which was associated with decreases in cell proliferation, soft agar colony formation and invasiveness; this inhibitory effect was augmented with a combination of DLC1 transduction and c-Myc suppression. To determine whether liver cell-specific delivery of DLC1 was able to enhance the inhibitory effect of c-Myc knockdown on tumor growth in vivo, DLC1 vector DNA complexed with galactosylated polyethylene glycol-linear polyethyleneimine was administered by tail vein injection to mice bearing subcutaneous xenografts of Huh7 cells transfected with shMyc or control shRNA. A cooperative inhibitory effect of DLC1 expression and c-Myc knockdown on the growth of Huh7-derived tumors was observed, suggesting that targeted liver cell delivery of DLC1 and c-Myc shRNA may serve as a possible gene therapy modality for the treatment of human HCC.

2.
Springerplus ; 3: 27, 2014.
Article in English | MEDLINE | ID: mdl-24683532

ABSTRACT

DLC1 (Deleted in Liver Cancer 1) gene encodes a RhoGTPase-activating protein (RhoGAP), which exerts most of its tumor suppressor functions through suppression of small Rho GTPases proteins RhoA, RhoB, RhoC and to some degree Cdc42, but not Rac. RhoGTPases are implicated in NF-κB activation in highly invasive prostate carcinoma (PCA), with consequences on cell proliferation, survival and metastatic capacity. Here we demonstrate that DLC1 transduction in two androgen-independent (AI) and highly metastatic PCA cell lines negatively regulates NF-κB activity in a GAP- and α-catenin-dependent manner. Expressed DLC1 protein suppresses the phosphorylation of NF-κB inhibitor, IκBα, causes its relocation from membrane ruffles into cytoplasm and attenuates its ubiquitination and subsequent degradation. DLC1-mediated NF-kB suppression and its effects are comparable to NF-κB inhibition using either shRNA knockdown or peptide inhibitor. Expression of transduced DLC1 suppressed the expression of NF-κB mediated genes. Such effects were found to be reliant on presence of calcium, indicating that the observed modifications are dependent on, and enabled by DLC-mediated stabilization of adherens junctions. These results expand the multitude of DLC1 interactions with other genes that modulate its oncosuppressive function, and may have potential therapeutic implications.

3.
Mol Diagn Ther ; 18(3): 293-302, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24519699

ABSTRACT

While significant progress continues to be made in the early detection and therapeutic management of primary tumors, the incidence of metastatic disease remains the major cause of mortality. Accordingly, the development of novel effective therapies that can ameliorate dissemination and secondary tumor growth are a clinical priority. The identification of genetic and functional alterations in cancer cells that affect factors implicated in the metastatic process is critical for designing preventive and therapeutic strategies. Evidence implicating the protein deleted in liver cancer-1 (DLC1), a Rho GTPase activator, in metastasis has accumulated to a point where DLC1 may be considered as a metastasis suppressor gene. This review presents evidence supporting an anti-metastatic role for DLC1 in several human cancers and discusses the mechanisms contributing to its inhibitory effects. In addition, promising opportunities for therapeutic interventions based on DLC1 function and downstream pathways involved in the metastatic process are considered.


Subject(s)
GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Neoplasm Metastasis/pathology , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Neoplasm Metastasis/genetics , Neoplasms/pathology
4.
BMC Cancer ; 13: 198, 2013 Apr 22.
Article in English | MEDLINE | ID: mdl-23607551

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer-related mortality. Therapies against non-small cell lung cancer (NSCLC) are particularly needed, as this type of cancer is relatively insensitive to chemotherapy and radiation therapy. We recently identified GGTI compounds that are designed to block geranylgeranylation and membrane association of signaling proteins including the Rho family G-proteins. One of the GGTIs is P61A6 which inhibits proliferation of human cancer cells, causes cell cycle effects with G1 accumulation and exhibits tumor-suppressing effects with human pancreatic cancer xenografts. In this paper, we investigated effects of P61A6 on non-small cell lung cancer (NSCLC) cells in vitro and in vivo. METHODS: Three non-small cell lung cancer cell lines were used to test the ability of P61A6 to inhibit cell proliferation. Further characterization involved analyses of geranylgeranylation, membrane association and activation of RhoA, and anchorage-dependent and -independent growth, as well as cell cycle effects and examination of cell cycle regulators. We also generated stable cells expressing RhoA-F, which bypasses the geranylgeranylation requirement of wild type RhoA, and examined whether the proliferation inhibition by P61A6 is suppressed in these cells. Tumor xenografts of NSCLC cells growing in nude mice were also used to test P61A6's tumor-suppressing ability. RESULTS: P61A6 was shown to inhibit proliferation of NSCLC lines H358, H23 and H1507. Detailed analysis of P61A6 effects on H358 cells showed that P61A6 inhibited geranylgeranylation, membrane association of RhoA and caused G1 accumulation associated with decreased cyclin D1/2. The effects of P61A6 to inhibit proliferation could mainly be ascribed to RhoA, as expression of the RhoA-F geranylgeranylation bypass mutant rendered the cells resistant to inhibition by P61A6. We also found that P61A6 treatment of H358 tumor xenografts growing in nude mice reduced their growth as well as the membrane association of RhoA in the tumors. CONCLUSION: Thus, P61A6 inhibits proliferation of NSCLC cells and causes G1 accumulation associated with decreased cyclin D1/2. The result with the RhoA-F mutant suggests that the effect of P61A6 to inhibit proliferation is mainly through the inhibition of RhoA. P61A6 also shows efficacy to inhibit growth of xenograft tumor.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Phenylalanine/analogs & derivatives , Sulfonamides/pharmacology , rhoA GTP-Binding Protein/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin D2/metabolism , Enzyme Inhibitors/therapeutic use , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Lung Neoplasms/enzymology , Mice , Mice, Inbred BALB C , Mice, Nude , Phenylalanine/pharmacology , Phenylalanine/therapeutic use , Prenylation/drug effects , Sulfonamides/therapeutic use
5.
Bio Protoc ; 3(18)2013 Sep 20.
Article in English | MEDLINE | ID: mdl-27376106

ABSTRACT

Preparation of primary cultures of embryo fibroblasts from genetically engineered mouse strains can provide a valuable resource for analyzing the consequences of genetic alterations at the cellular level. Mouse embryo fibroblasts (MEFs) have been particularly useful in cancer research, as they have facilitated the identification of the genetic changes that allow cells to overcome senescence and proliferate indefinitely in culture. The immortalized MEFs can then acquire additional mutations that lead to anchorage-independent growth and the ability to form tumors in mice. Recently we developed an MEF model system for analysis of the role of the tumor suppressor gene DLC1 in cellular transformation (Qian et al., 2012). In this communication we describe a protocol for the isolation of MEFs from day 13.5-day 14.5 mouse embryos. The MEFs obtained by this procedure are suitable for use in biochemical assays and for further genetic manipulations.

6.
Cancer Res ; 72(22): 5900-11, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23010077

ABSTRACT

The tumor suppressor gene deleted in liver cancer-1 (DLC1), which encodes a protein with strong RhoGAP (GTPase activating protein) activity and weak Cdc42GAP activity, is inactivated in various human malignancies. Following Dlc1 inactivation, mouse embryo fibroblasts (MEF) with a conditional Dlc1 knockout allele reproducibly underwent neoplastic transformation. In addition to inactivation of Dlc1 and increased activity of Rho and Cdc42, transformation depended on the subsequent decreased expression of the Cdk4/6 inhibitors p15(Ink4b) and p16(Ink4a) together with increased expression and activation of Cdk4/6. The level of expression of these cell-cycle regulatory genes was relevant to human tumors with low DLC1 expression. Analysis of publicly available annotated datasets of lung and colon cancer with gene expression microarray profiles indicated that, in pairwise comparisons, low DLC1 expression occurred frequently together (P < 0.01) with downregulation of p15(Ink4b) or p16(Ink4a) or upregulation of CDK4 or CDK6. In addition, an unfavorable prognosis (P < 0.05) was associated with low DLC1 and low p15(Ink4b) in lung cancer and colon cancer, low DLC1 and low p16(Ink4a) in lung cancer, low DLC1 and high CDK4 in lung cancer, and low DLC1 and high CDK6 in colon cancer. Thus, several genes and biochemical activities collaborate with the inactivation of DLC1 to give rise to cell transformation in MEFs, and the identified genes are relevant to human tumors with low DLC1 expression.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , GTPase-Activating Proteins/genetics , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Down-Regulation , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, p16 , Genes, ras , Humans , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , rho-Associated Kinases/metabolism
7.
Int J Oncol ; 41(2): 393-406, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22580498

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer death, and its incidence is increasing worldwide in an alarming manner. The development of curative therapy for advanced and metastatic HCC is a high clinical priority. The HCC genome is complex and heterogeneous; therefore, the identification of recurrent genomic and related gene alterations is critical for developing clinical applications for diagnosis, prognosis and targeted therapy of the disease. This article focuses on recent research progress and our contribution in identifying and deciphering the role of defined genetic alterations in the pathogenesis of HCC. A significant number of genes that promote or suppress HCC cell growth have been identified at the sites of genomic reorganization. Notwithstanding the accumulation of multiple genetic alterations, highly recurrent changes on a single chromosome can alter the expression of oncogenes and tumor suppressor genes (TSGs) whose deregulation may be sufficient to drive the progression of normal hepatocytes to malignancy. A distinct and highly recurrent pattern of genomic imbalances in HCC includes the loss of DNA copy number (associated with loss of heterozygosity) of TSG-containing chromosome 8p and gain of DNA copy number or regional amplification of protooncogenes on chromosome 8q. Even though 8p is relatively small, it carries an unusually large number of TSGs, while, on the other side, several oncogenes are dispersed along 8q. Compelling evidence demonstrates that DLC1, a potent TSG on 8p, and MYC oncogene on 8q play a critical role in the pathogenesis of human HCC. Direct evidence for their role in the genesis of HCC has been obtained in a mosaic mouse model. Knockdown of DLC1 helps MYC in the induction of hepatoblast transformation in vitro, and in the development of HCC in vivo. Therapeutic interventions, which would simultaneously target signaling pathways governing both DLC1 and MYC functions in hepatocarcinogenesis, could result in progress in the treatment of liver cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/genetics , GTPase-Activating Proteins/physiology , Genes, myc , Liver Neoplasms/genetics , Tumor Suppressor Proteins/physiology , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Molecular Targeted Therapy , Mutation , Tumor Suppressor Proteins/genetics
8.
Mol Cell Biol ; 32(11): 2145-59, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22473989

ABSTRACT

The DLC1 (for deleted in liver cancer 1) tumor suppressor gene encodes a RhoGAP protein that inactivates Rho GTPases, which are implicated in regulation of the cytoskeleton and adherens junctions (AJs), a cell-cell adhesion protein complex associated with the actin cytoskeleton. Malignant transformation and tumor progression to metastasis are often associated with changes in cytoskeletal organization and cell-cell adhesion. Here we have established in human cells that the AJ-associated protein α-catenin is a new binding partner of DLC1. Their binding was mediated by the N-terminal amino acids 340 to 435 of DLC1 and the N-terminal amino acids 117 to 161 of α-catenin. These proteins colocalized in the cytosol and in the plasma membrane, where together they associated with E-cadherin and ß-catenin, constitutive AJ proteins. Binding of DLC1 to α-catenin led to their accumulation at the plasma membrane and required DLC1 GAP activity. Knocking down α-catenin in DLC1-positive cells diminished DLC1 localization at the membrane. The DLC1-α-catenin complex reduced the Rho GTP level at the plasma membrane, increased E-cadherin's mobility, affected actin organization, and stabilized AJs. This process eventually contributed to a robust oncosuppressive effect of DLC1 in metastatic prostate carcinoma cells. Together, these results unravel a new mechanism through which DLC1 exerts its strong oncosuppressive function by positively influencing AJ stability.


Subject(s)
Adherens Junctions/metabolism , GTPase-Activating Proteins/metabolism , Tumor Suppressor Proteins/metabolism , alpha Catenin/metabolism , Binding Sites , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Humans , Male , Neoplasm Metastasis , Protein Binding , rho GTP-Binding Proteins/metabolism
9.
Biochem Biophys Res Commun ; 420(2): 325-30, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22425986

ABSTRACT

Deleted in liver cancer (DLC1), a tumor suppressor gene in multiple cancers, is recurrently down regulated or inactivated by epigenetic mechanisms in primary prostate carcinomas (PCAs). In this study the methylation and acetylation profile of the DLC1 promoter region was examined in three PCA cell lines with low or undetectable DLC1 expression: LNCaP, its derivative C4-2B-2, and 22Rv1. Two histone deacetylase inhibitors (HDAC), suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA) induced histone acetylation of the DLC1 promoter in all three lines. DLC1 promoter methylation and deacetylation were detected in LNCaP and C4-2B-2 cells while in 22Rv1 cells DLC1 is silenced by deacetylation. Treatment with SAHA or TSA efficiently increased DLC1 expression in all lines, particularly in 22Rv1 cells, and activated the DLC1 promoter through the same Sp1 sites. The 22Rv1 cell line was selected to evaluate the efficacy of combined DLC1 transduction and SAHA treatment on tumor growth in athymic mice. Individually, DLC1 transduction and SAHA exposure reduced the tumor size by 75-80% compared to controls and in combination almost completely inhibited tumor growth. The antitumor effect was associated with the induction of apoptosis and inhibition of RhoA activity. SAHA alone significantly reduced RhoA activity, showing that this RhoGTPase is a target for SAHA. These results, obtained with a reliable preclinical in vivo test, predict that combined therapeutic agents targeting the pathways governing DLC1 function and HDAC inhibitors may be beneficial in management of prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , GTPase-Activating Proteins/genetics , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Prostatic Neoplasms/therapy , Tumor Suppressor Proteins/genetics , Animals , BALB 3T3 Cells , Cell Line, Tumor , Combined Modality Therapy , DNA Methylation/drug effects , Drug Evaluation, Preclinical , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Transduction, Genetic , Vorinostat
10.
Cancer Genomics Proteomics ; 8(5): 227-33, 2011.
Article in English | MEDLINE | ID: mdl-21980037

ABSTRACT

We previously located a senescence gene locus (SEN6A), at chromosome 6q14-21 by a functional strategy using chromosome transfer into immortal ovarian tumor cells. To further elucidate the SEN6A locus, intact chromosome 6 or 6q was transferred into rat ovarian tumor cells and a panel of immortal revertant clones of senescent cells was generated. The panel of independent colonies as well as mixed populations of revertant cells was analyzed for the presence or absence of chromosome 6 specific markers. These investigations led to the identification of a fine deletion of approximately 1cM at chromosomal interval 6q16.3. A contiguous stretch containing five yeast artificial chromosome (YAC) clones was constructed across the deleted region. The non-chimeric YAC clones were retrofitted and transferred into mouse A9 cells by spheroplast fusion to generate YAC/A9 hybrids. YAC DNA present in YAC/A9 hybrids was subsequently transferred by microcell fusion into immortal tumor cells, and the hybrid cells were characterized for their senescence phenotype. Using this functional strategy, the transfer of YAC clone 966b10 was shown to restore senescence in both rat and human ovarian and breast tumor cells. Our results demonstrate that the SEN6A gene is carried on a 1 Mb YAC, 966b10, which maps at 6q16.3.


Subject(s)
Breast Neoplasms/genetics , Cellular Senescence/genetics , Chromosomes, Artificial, Yeast , Chromosomes, Human, Pair 6 , Genetic Loci/genetics , Ovarian Neoplasms/genetics , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromosome Mapping , Chromosomes, Artificial, Bacterial , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Ovarian Neoplasms/metabolism , Rats
11.
Cancer Res ; 71(8): 2916-25, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21372205

ABSTRACT

The DLC1 gene encodes a Rho GTPase-activating protein (RhoGAP) that functions as a tumor suppressor in several common human cancers. The multidomain structure of DLC1 enables interaction with a number of other proteins. Here we report that the proinflammatory protein S100A10 (also known as p11), a key cell surface receptor for plasminogen which regulates pericellular proteolysis and tumor cell invasion, is a new binding partner of DLC1 in human cells. We determined that the 2 proteins colocalize in the cell cytoplasm and that their binding is mediated by central sequences in the central domain of DLC1 and the C-terminus of S100A10. Because the same S100A10 sequence also mediates binding to Annexin 2, we found that DLC1 competed with Annexin 2 for interaction with S100A10. DLC1 binding to S100A10 did not affect DLC1's RhoGAP activity, but it decreased the steady-state level of S100A10 expression in a dose-dependent manner by displacing it from Annexin 2 and making it accessible to ubiquitin-dependent degradation. This process attenuated plasminogen activation and resulted in inhibition of in vitro cell migration, invasion, colony formation, and anchorage-independent growth of aggressive lung cancer cells. These results suggest that a novel GAP-independent mechanism contributes to the tumor suppressive activity of DLC1, and highlight the importance and complexity of protein-protein interactions involving DLC1 in certain cancers.


Subject(s)
Annexin A2/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , GTPase-Activating Proteins/metabolism , Lung Neoplasms/metabolism , S100 Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Down-Regulation , HEK293 Cells , Humans , Lung Neoplasms/pathology , Plasminogen/metabolism , Protein Binding , Ubiquitination
12.
Int J Oncol ; 36(4): 999-1005, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20198346

ABSTRACT

Inactivation of tumor suppressor genes is a major contributing alteration in the initiation or progression of cancer. The human tumor suppressor gene DLC1 (deleted in liver cancer 1) is frequently downregulated or silenced in multiple cancers, predominantly by epigenetic mechanisms. With the current considerable interest and progress in epigenetic therapy, a number of promising antineoplastic agents, particularly histone deacetylase (HDAC) inhibitors, have been developed and used successfully in clinical trials. Both DLC1 and HDAC inhibitors exert antineoplastic functions, and their combined action could be exploited for a more effective cancer therapy. To evaluate the potential benefits of this approach, we examined the antineoplastic effects of adenoviral (Ad)-DLC1-mediated transduction and exposure to suberoylanilide hydroxamic acid (SAHA), a powerful HDAC inhibitor, in two human cancer cell lines that lack intrinsic DLC1 expression, 22Rv1 prostate cancer cells and 7703K human hepatocellular carcinoma cells. Consistent with the oncosuppressive function of DLC1 in several cancers, including prostate and liver cancer, transduction of 22Rv1 and 7703K cells with an Ad-DLC1 expression vector resulted in alterations of cell morphology, induction of apoptosis, and inhibition of cell proliferation, migration, and anchorage-independent growth. A low concentration of SAHA (5 microM) efficiently restored the expression of DLC1 in 22Rv1 cells that lack DLC1 expression due to histone deacetylation but had a minimal effect in 7703K cells in which silencing of the DLC1 gene is due mainly to promoter hypermethylation. Regardless of the epigenetic mechanism of DLC1 inactivation, SAHA treatment of DLC1-transduced cells had a synergistic inhibitory effect on tumor cell proliferation and tumorigenesis in both cell lines. In 22Rv1 cells, this combination regimen nearly abolished the formation of colonies in semisolid media as a measure of tumorigenicity in vitro. Current in vitro results validate this protocol as a potentially new therapeutic option in certain cancers.


Subject(s)
Genetic Therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Liver Neoplasms/therapy , Prostatic Neoplasms/therapy , Tumor Suppressor Proteins/genetics , Adenoviridae/genetics , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Enzyme Activation , GTPase-Activating Proteins , Genetic Vectors , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Time Factors , Transduction, Genetic , Tumor Suppressor Proteins/biosynthesis , Vorinostat
13.
Cancer Res ; 69(19): 7819-25, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789346

ABSTRACT

We have applied a functional gene transfer strategy to show the importance of viral integration site in cellular immortalization. The large tumor antigen of SV40 is capable of extending the cellular life span by sequestering tumor suppressor proteins pRB and p53 in virus-transformed human cells. Although SV40 large T antigen is essential, it is not sufficient for cellular immortalization, suggesting that additional alterations in cellular genes are required to attain infinite proliferation. We show here that the disruption of human chromosomal interval at 1q21.1 by SV40 integration can be an essential step for cellular immortalization. The transfer of a 150-kb bacterial artificial chromosome (BAC) clone, RP364B14, corresponding to viral integration site in CRL2504 cells, reverted their immortal phenotype. Interestingly, the BAC transfer clones of CRL2504 cells displayed characteristics of either senescence as shown by beta-galactosidase activity or apoptosis as revealed by positive staining with M30 CytoDEATH antibody. The SV40 integration at 1q21.1, in the vicinity of epidermal differentiation complex (EDC) genes, resulted in the down-regulation of the filaggrin (FLG) gene that is part of the EDC. FLG gene expression was increased in BAC transfer senescent and apoptotic clones. Our results suggest that the disruption of native genomic sequence by SV40 may alter expression of genes involved in senescence and apoptosis by modulating chromatin structure. These studies imply that identification of genes located in the vicinity of viral integration sites in human cancers may be helpful in developing new diagnostic and therapeutic strategies.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Bronchi/physiology , Cell Transformation, Viral/genetics , Chromosomes, Human, Pair 1 , Simian virus 40/genetics , Virus Integration , Apoptosis/genetics , Base Sequence , Bronchi/virology , Cell Line, Transformed , Cellular Senescence/genetics , Chromosomes, Artificial, Bacterial , Cloning, Molecular , Epithelial Cells/physiology , Epithelial Cells/virology , Filaggrin Proteins , Gene Transfer Techniques , Humans , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Molecular Sequence Data
14.
J Cell Mol Med ; 13(9B): 3985-92, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19426152

ABSTRACT

During the neoplastic process tumour cells frequently acquire resistance to the antiproliferative signals of transforming growth factor-beta (TGF-beta). Here we examined a human hepatocellular carcinoma cell line (Hep3B-TS) sensitive to TGF-beta signalling, and a derivative line (Hep3B-TR) rendered resistant to TGF-beta by stepwise exposure to TGF-beta(1). Comprehensive molecular cytogenetic analysis revealed that the acquisition of TGF-beta-resistance by Hep3B-TR cells was due to loss of TGF-beta receptor 2 (TGFbetaRII) gene. As demonstrated by spectral karyotyping and array-based comparative genomic hybridization, and in difference to Hep3B-TS cells, which have three rearranged and two normal copies of chromosome 3 that harbour the TGFbetaRII gene, Hep3B-TR cells have four rearranged and one apparently normal chromosome 3, which nonetheless underwent a critical microdeletion at the site of TGFbetaRII gene. Gene expression analysis using an oligonucleotide microarray of 21,397 genes showed that Hep3B-TR differentially expressed 307 genes, out of which 197 and 110 were up- and down-regulated, respectively, compared to Hep3B-TS. Six of differentially expressed genes were identified as downstream targets of the tumour necrosis factor (TNF) gene, suggesting that loss of TGFbetaRII triggered activation of the TNF pathway known to be regulated by TGF-beta(1) network. On the functional level, the TGF-beta-resistant Hep3B-TR cells displayed significantly enhanced capacity for anchorage independent growth and cell migration in vitro, and also increased tumorigenicity in vivo and in vitro and in vivo tumorigenicity compared with parental sensitive cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Transforming Growth Factor beta1/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Comparative Genomic Hybridization , Gene Deletion , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Models, Genetic , Neoplasm Invasiveness , Wound Healing
15.
Cancer Genet Cytogenet ; 191(1): 17-26, 2009 May.
Article in English | MEDLINE | ID: mdl-19389504

ABSTRACT

Mouse models for hepatocellular carcinoma (HCC) provide an experimental ground for dissecting the genetic and biological complexities of human liver cancer and contribute to our ability to gain insights into the relevance of candidate cancer genes. We examined, using spectral karyotyping (SKY) and array-based CGH (aCGH), seven cell lines derived from HCC spontaneously developed in transgenic Myc mice (Myc), and four cell lines established from tumors induced in nude mice by inoculation with the original Myc cells (nuMyc). All the cell lines exhibited gain of material from chromosomes 5, 6, 8, 10, 11, 15, and 19 and DNA copy-number loss from chromosomes 2, 4, 7, 9, 12, 14, and X. In addition, several recurrent chromosome reorganizations were found, including del(3), t(3;8), del(4), t(4;11), t(6;5), del(7), del(8), del(9), t(10;14), del(11), and del(16). Chromosome breakpoints underlying rearrangements clustered in the regions previously identified as important for the early stages of Myc-induced hepatocarcinogenesis. The results strongly suggest the importance of recurrent breakage and loss of chromosomes 4, 9, and 14 and gain of chromosomes 15 and 19 in mouse liver neoplasia. Genomic changes observed in Myc HCC cell lines are also recurrent in HCC developed in other transgenic mouse models, in mouse spontaneous HCC and derivative cell lines, and in preneoplastic liver lesions induced with chemical carcinogens. Overall, the present results document selective, nonrandom genomic changes involving chromosomal regions homologous to those implicated in human HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Chromosome Aberrations , DNA, Neoplasm/genetics , Gene Dosage , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chromosomes, Mammalian/genetics , Comparative Genomic Hybridization , Disease Models, Animal , Gene Rearrangement , Humans , Injections, Subcutaneous , Liver Neoplasms/pathology , Mice , Mice, Transgenic , Spectral Karyotyping
16.
Int J Oncol ; 32(6): 1285-91, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497990

ABSTRACT

The process of cell dissemination from the primary tumors to distant sites is the most harmful event during cancer progression, and the leading cause of cancer death. We have previously demonstrated that restoration of DLC1 tumor suppressor gene expression in the DLC1-negative Focus and 7703K human hepatocellular carcinoma (HCC) cell lines induced caspase-3 mediated apoptosis, reduced cell growth in vitro and tumorigenicity in vivo and diminished the ability to migrate through Matrigel, a property suggestive of metastatic potential in vivo. We now show that subcutaneous tumors developing after inoculation of Focus and 7703K cells into nude mice disseminate cells to liver and lung, and this process is markedly suppressed by restoration of DLC1 expression. Inhibition of tumor cell dissemination was associated with lower levels of RhoA activity, an increase in rounded cells and a reduction in actin stress fibers and focal adhesion molecules that are of critical importance in cancer cell invasion and metastasis. In addition, DLC1 down-regulated the expression of osteopontin and matrix metalloproteinase-9, which are highly up-regulated in most primary HCC with associated metastases. These observations implicate the DLC1 gene in suppression of HCC cell dissemination and identify novel cellular and genetic alterations that contribute to prevention of metastasis, a life-threatening event in cancer progression.


Subject(s)
Actins/metabolism , Carcinoma, Hepatocellular/prevention & control , Liver Neoplasms/prevention & control , Lung Neoplasms/prevention & control , Skin Neoplasms/pathology , Tumor Suppressor Proteins/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Cell Proliferation , Collagen/metabolism , Cytoskeleton/metabolism , Down-Regulation , Drug Combinations , GTPase-Activating Proteins , Humans , Laminin/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Mice, Nude , Neoplasm Invasiveness , Osteopontin/antagonists & inhibitors , Osteopontin/metabolism , Proteoglycans/metabolism , Tumor Cells, Cultured , rhoA GTP-Binding Protein/antagonists & inhibitors
17.
Aging Cell ; 7(1): 89-100, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18005250

ABSTRACT

Accumulation of DNA damage may play an essential role in both cellular senescence and organismal aging. The ability of cells to sense and repair DNA damage declines with age. However, the underlying molecular mechanism for this age-dependent decline is still elusive. To understand quantitative and qualitative changes in the DNA damage response during human aging, DNA damage-induced foci of phosphorylated histone H2AX (gamma-H2AX), which occurs specifically at sites of DNA double-strand breaks (DSBs) and eroded telomeres, were examined in human young and senescing fibroblasts, and in lymphocytes of peripheral blood. Here, we show that the incidence of endogenous gamma-H2AX foci increases with age. Fibroblasts taken from patients with Werner syndrome, a disorder associated with premature aging, genomic instability and increased incidence of cancer, exhibited considerably higher incidence of gamma-H2AX foci than those taken from normal donors of comparable age. Further increases in gamma-H2AX focal incidence occurred in culture as both normal and Werner syndrome fibroblasts progressed toward senescence. The rates of recruitment of DSB repair proteins to gamma-H2AX foci correlated inversely with age for both normal and Werner syndrome donors, perhaps due in part to the slower growth of gamma-H2AX foci in older donors. Because genomic stability may depend on the efficient processing of DSBs, and hence the rapid formation of gamma-H2AX foci and the rapid accumulation of DSB repair proteins on these foci at sites of nascent DSBs, our findings suggest that decreasing efficiency in these processes may contribute to genome instability associated with normal and pathological aging.


Subject(s)
Aging/pathology , Cellular Senescence/physiology , DNA Repair , Adult , Aging/genetics , Aging/radiation effects , Cells, Cultured , Cellular Senescence/genetics , Cellular Senescence/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/physiology , Gamma Rays , Genomic Instability , Histones/metabolism , Humans , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Lymphocytes/physiology , MRE11 Homologue Protein , Microscopy, Confocal , Middle Aged , Telomere , Tumor Suppressor p53-Binding Protein 1 , Werner Syndrome/pathology
18.
J Cell Mol Med ; 11(5): 1185-207, 2007.
Article in English | MEDLINE | ID: mdl-17979893

ABSTRACT

The deleted in liver cancer 1 (DLC-1) gene encodes a GTPase activating protein that acts as a negative regulator of the Rho family of small GTPases. Rho proteins transduce signals that influence cell morphology and physiology, and their aberrant up-regulation is a key factor in the neoplastic process, including metastasis. Since its discovery, compelling evidence has accumulated that demonstrates a role for DLC-1 as a bona fide tumour suppressor gene in different types of human cancer. Loss of DLC-1 expression mediated by genetic and epigenetic mechanisms has been associated with the development of many human cancers, and restoration of DLC-1 expression inhibited the growth of tumour cells in vivo and in vitro. Two closely related genes, DLC-2 and DLC-3, may also be tumour suppressors. This review presents the current status of progress in understanding the biological functions of DLC-1 and its relatives and their roles in neoplasia.


Subject(s)
GTPase-Activating Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Humans , Molecular Sequence Data , Neoplasms/metabolism , Neoplasms/pathology , Protein Structure, Tertiary , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
19.
Cancer Res ; 67(18): 8752-61, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875716

ABSTRACT

The biological functions of nuclear topoisomerase I (Top1) have been difficult to study because knocking out TOP1 is lethal in metazoans. To reveal the functions of human Top1, we have generated stable Top1 small interfering RNA (siRNA) cell lines from colon and breast carcinomas (HCT116-siTop1 and MCF-7-siTop1, respectively). In those clones, Top1 is reduced approximately 5-fold and Top2alpha compensates for Top1 deficiency. A prominent feature of the siTop1 cells is genomic instability, with chromosomal aberrations and histone gamma-H2AX foci associated with replication defects. siTop1 cells also show rDNA and nucleolar alterations and increased nuclear volume. Genome-wide transcription profiling revealed 55 genes with consistent changes in siTop1 cells. Among them, asparagine synthetase (ASNS) expression was reduced in siTop1 cells and in cells with transient Top1 down-regulation. Conversely, Top1 complementation increased ASNS, indicating a causal link between Top1 and ASNS expression. Correspondingly, pharmacologic profiling showed L-asparaginase hypersensitivity in the siTop1 cells. Resistance to camptothecin, indenoisoquinoline, aphidicolin, hydroxyurea, and staurosporine and hypersensitivity to etoposide and actinomycin D show that Top1, in addition to being the target of camptothecins, also regulates DNA replication, rDNA stability, and apoptosis. Overall, our studies show the pleiotropic nature of human Top1 activities. In addition to its classic DNA nicking-closing functions, Top1 plays critical nonclassic roles in genomic stability, gene-specific transcription, and response to various anticancer agents. The reported cell lines and approaches described in this article provide new tools to perform detailed functional analyses related to Top1 function.


Subject(s)
Breast Neoplasms/enzymology , Colonic Neoplasms/enzymology , DNA Topoisomerases, Type I/physiology , Aspartate-Ammonia Ligase/biosynthesis , Aspartate-Ammonia Ligase/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Chromosome Aberrations , Colonic Neoplasms/genetics , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genome, Human , Genomic Instability , HCT116 Cells , Histones/biosynthesis , Histones/genetics , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Transfection
20.
Cancer Res ; 67(17): 8131-8, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804725

ABSTRACT

The identification of mammary gland stem cells (MGSC) or progenitors is important for the study of normal breast development and tumorigenesis. Based on their immunophenotype, we have isolated a population of mouse mammary gland cells that are capable of forming "mammospheres" in vitro. Importantly, mammospheres are enriched for cells that regenerate an entire mammary gland on implantation into a mammary fat pad. We also undertook cytogenetic analyses of mammosphere-forming cells after prolonged culture, which provided preliminary insight into the genomic stability of these cells. Our identification of new cell surface markers for enriching mammosphere-initiating cells, including endoglin and prion protein, will facilitate the elucidation of the cell biology of MGSC.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/transplantation , Cell Proliferation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/transplantation , Animals , Cell Culture Techniques , Cell Differentiation , Cell Shape , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoids/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...