233 results on '"Popescu, NC"'
Search Results
2. DLC1 tumor suppressor gene inhibits migration and invasion of multiple myeloma cells through RhoA GTPase pathway
- Author
-
Ullmannova-Benson, V, Guan, M, Zhou, X, Tripathi, V, Yang, X-Y, Zimonjic, DB, and Popescu, NC
- Published
- 2009
3. Human uteroglobin gene: Structure, subchromosomal localization, and polymorphism
- Author
-
Zhang, ZJ, Zimonjic, DB, Popescu, NC, Gerhard, DS, Stone, EM, Arbour, NC, DeVries, HG, Scheffer, H, Gerritsen, J, Collee, JM, TenKate, LP, Mukherjee, AB, and Faculteit Medische Wetenschappen/UMCG
- Subjects
ANTIINFLAMMATORY PEPTIDES ANTIFLAMMINS ,MOLECULAR-CLONING ,PHOSPHOLIPASE-A2 ACTIVITY ,RABBIT UTEROGLOBIN ,LIPOCORTIN-I ,BASOPHILIC LEUKEMIA-CELLS ,IN-SITU HYBRIDIZATION ,10-KD PROTEIN ,TISSUE-SPECIFIC EXPRESSION ,PLATELET-ACTIVATING-FACTOR - Abstract
Human uteroglobin (hUG) or Clara cell 10-kD protein (cc10 kDa) is a steroid-dependent, immunomodulatory, cytokine-like protein, It is secreted by mucosal epithelial cells of all vertebrates studied, The cDNA encoding hUG and the 5' promoter region of the gene have been characterized previously, Here, we report that the structure of the entire hUG gene is virtually identical to those of rabbit, rat, and mouse, It is localized on human chromosome 11q12.3-13.1, a region in which several important candidate disease genes have been mapped by linkage analyses, Our data indicate that candidate genes for atopic (allergic) asthma and Best's vitelliform macular dystrophy are in closest proximity to the hUG gene, To determine whether hUG gene mutation may be involved in the pathogenesis of these diseases, we studied two isolated groups of patients, each afflicted with either atopy or Best's disease, respectively, We detected a single base-pair change in the hUG gene in Best's disease patients and normal controls but no such change was detected in atopy patients, This alteration in hUG gene-sequence in Best disease family appears to be a polymorphism, Although the results of our investigation did not uncover mutations in hUG gene that could be causally related to the pathogenesis of either of these diseases, its conservation throughout vertebrate phyla implies that this gene is of physiological importance, Moreover, the close proximity of this gene to several candidate disease genes makes it an important chromosomal marker in cloning and characterization of those genes.
- Published
- 1997
4. Expression of alternative forms of Ras exchange factors GRF and SOS1 in different human tissues and cell lines
- Author
-
Guerrero C, Rojas JM, Chedid M, Esteban LM, Zimonjic DB, Popescu NC, Font de Mora J, and Santos E
- Subjects
Adult ,ras-GRF1 ,Brain ,Proteins ,3T3 Cells ,Blotting, Northern ,Kidney ,Cell Line ,Fungal Proteins ,Repressor Proteins ,Alternative Splicing ,Mice ,Fetus ,Gene Expression Regulation ,Tumor Cells, Cultured ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,RNA ,ras Guanine Nucleotide Exchange Factors ,SOS1 Protein ,Lung ,Pancreas ,In Situ Hybridization - Abstract
DNA probes and antibodies specific for different coding regions of human SOS1 and GRF genes were used to screen expression of these genes in a variety of adult and fetal human tissues and cell lines. Despite previous reports of the exclusive expression of hGRF RNA in brain, we also observed expression of this gene in various other tissues including lung and pancreas, as well as several tumor cell lines. At least three different hGRF mRNA transcripts were observed depending on the probe used, with the larger transcripts being detected by probes corresponding to the 5' end of the gene while smaller transcripts were detected by probes corresponding to the 3' end. Expression of hSOS1-related transcripts was more ubiquitous and homogeneous than with hGRF, with similar levels of specific transcripts being detected in most tissues and cell fines tested. Three to five different transcripts were detected in human tissues when using probes for the 5' end and middle regions of this gene, whereas only two were detected with probes corresponding to the 3' end. Screening of multiple human tumor cell lines showed ubiquitous expression of three specific transcripts, although the level and ratio of each of these transcripts varied widely among individual cell lines. Consistent with the variety of transcripts detected, several protein forms were also identified in Western immunoblots with antisera raised against specific domains of hSOS1 and human Ras-GRF gene products. Fluorescence in situ chromosomal hybridization suggested that, in both cases, the multiple forms arise from single chromosomal loci. The heterogeneity of hGRF and hSOS1 gene products detected (which appear to retain in most cases a functional catalytic domain), suggests that differentially expressed, alternatively spliced hSOS1 and hGRF forms may contribute to fine regulation of Ras activation in different tissues or at different stages of development.
- Published
- 1996
5. CUMULATIVE GENE AND CHROMOSOME ALTERATIONS ASSOCIATED WITH IN-VITRO NEOPLASTIC TRANSFORMATION OF HUMAN CERVICAL CELLS
- Author
-
POPESCU, NC, primary, ZIMONJIC, DB, additional, SIMPSON, S, additional, and DIPAOLO, JA, additional
- Published
- 1995
- Full Text
- View/download PDF
6. Fragile sites and cancer genes on the short arm of chromosome 8.
- Author
-
Popescu NC, Birnbaum D, and Chaffanet M
- Published
- 2004
- Full Text
- View/download PDF
7. Cooperative antiproliferative effect of coordinated ectopic expression of DLC1 tumor suppressor protein and silencing of MYC oncogene expression in liver cancer cells: Therapeutic implications.
- Author
-
Yang X, Zhou X, Tone P, Durkin ME, and Popescu NC
- 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.
- Published
- 2016
- Full Text
- View/download PDF
8. Deleted in liver cancer-1 (DLC1): an emerging metastasis suppressor gene.
- Author
-
Popescu NC and Goodison S
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Humans, Neoplasm Metastasis genetics, Neoplasms pathology, GTPase-Activating Proteins genetics, GTPase-Activating Proteins metabolism, Neoplasm Metastasis pathology, Neoplasms genetics, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism
- 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.
- Published
- 2014
- Full Text
- View/download PDF
9. DLC1 induces expression of E-cadherin in prostate cancer cells through Rho pathway and suppresses invasion.
- Author
-
Tripathi V, Popescu NC, and Zimonjic DB
- Subjects
- Cell Aggregation physiology, Cell Differentiation physiology, Cell Line, Tumor, GTPase-Activating Proteins genetics, GTPase-Activating Proteins physiology, Gene Knockdown Techniques, Humans, Male, Neoplasm Invasiveness, Prostatic Neoplasms genetics, Transfection, Tumor Suppressor Proteins genetics, rho-Associated Kinases genetics, Cadherins biosynthesis, GTPase-Activating Proteins metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Tumor Suppressor Proteins metabolism, rho-Associated Kinases metabolism
- Abstract
E-cadherin is a cell-cell adhesion molecule that acts as a suppressor of cancer cell invasion and its expression is downregulated in many advanced, poorly differentiated, human cancers. In this study, we found that the expression of DLC1 (deleted in liver cancer 1) tumor-suppressor gene in metastatic prostate carcinoma (PCA) cells increased the expression of E-cadherin and resulted in an elevated rate of cell-cell aggregation as measured by aggregation assay. DLC1-mediated increase in E-cadherin expression was not dependent on α-catenin, a DLC1-binding protein associated with E-cadherin, and/or cellular density. The increase of E-cadherin expression occurred at mRNA level and relied on DLC1 RhoGAP function, leading to suppression of high level of RhoA-GTP and RhoC-GTP activity in metastatic PCA cells. Application of Rho/ROCK inhibitors produced the same effect as introduction of DLC1. Knocking down of RhoA produced a moderate increase in E-cadherin whereas knocking down of RhoC resulted in a significant increase of E-cadherin. Downregulation of E-cadherin caused by constitutively active RhoA(V14) and RhoC(V14) could not be reversed by expression of DLC1 in DLC1-negative cell line. DLC1-mediated suppression of metastatic PCA cells invasion was comparable with the one associated with ectopic E-cadherin expression, or caused by suppression of Rho pathway either by Rho/ROCK inhibitors, or by shRNA repression. This study demonstrates that DLC1 expression positively regulates E-cadherin and suppresses highly metastatic PCA cell invasion by modulating Rho pathway, which appears as a feasible therapeutic target in cancers with high activity of RhoGTPases.
- Published
- 2014
- Full Text
- View/download PDF
10. DLC1 suppresses NF-κB activity in prostate cancer cells due to its stabilizing effect on adherens junctions.
- Author
-
Tripathi V, Popescu NC, and Zimonjic DB
- 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.
- Published
- 2014
- Full Text
- View/download PDF
11. Isolation of Mouse Embryo Fibroblasts.
- Author
-
Durkin ME, Qian X, Popescu NC, and Lowy DR
- 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.
- Published
- 2013
- Full Text
- View/download PDF
12. In vitro and in vivo effects of geranylgeranyltransferase I inhibitor P61A6 on non-small cell lung cancer cells.
- Author
-
Zimonjic DB, Chan LN, Tripathi V, Lu J, Kwon O, Popescu NC, Lowy DR, and Tamanoi F
- Subjects
- 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, 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
- 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.
- Published
- 2013
- Full Text
- View/download PDF
13. Inactivation of the Dlc1 gene cooperates with downregulation of p15INK4b and p16Ink4a, leading to neoplastic transformation and poor prognosis in human cancer.
- Author
-
Qian X, Durkin ME, Wang D, Tripathi BK, Olson L, Yang XY, Vass WC, Popescu NC, and Lowy DR
- Subjects
- 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, Cell Transformation, Neoplastic genetics, Cyclin-Dependent Kinase Inhibitor p15 genetics, GTPase-Activating Proteins genetics, Neoplasms genetics, Tumor Suppressor Proteins genetics
- 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., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
14. Role of DLC1 tumor suppressor gene and MYC oncogene in pathogenesis of human hepatocellular carcinoma: potential prospects for combined targeted therapeutics (review).
- Author
-
Zimonjic DB and Popescu NC
- Subjects
- 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, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Hepatocellular genetics, GTPase-Activating Proteins physiology, Genes, myc, Liver Neoplasms genetics, Tumor Suppressor Proteins physiology
- 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.
- Published
- 2012
- Full Text
- View/download PDF
15. DLC1 interaction with α-catenin stabilizes adherens junctions and enhances DLC1 antioncogenic activity.
- Author
-
Tripathi V, Popescu NC, and Zimonjic DB
- Subjects
- Binding Sites, Cadherins metabolism, Cell Adhesion, Cell Line, Tumor, Cell Movement, Humans, Male, Neoplasm Metastasis, Protein Binding, rho GTP-Binding Proteins metabolism, Adherens Junctions metabolism, GTPase-Activating Proteins metabolism, Tumor Suppressor Proteins metabolism, alpha Catenin metabolism
- 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 β-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.
- Published
- 2012
- Full Text
- View/download PDF
16. Preclinical evaluation of combined antineoplastic effect of DLC1 tumor suppressor protein and suberoylanilide hydroxamic acid on prostate cancer cells.
- Author
-
Zhou X, Yang XY, and Popescu NC
- Subjects
- 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, Antineoplastic Agents therapeutic use, GTPase-Activating Proteins genetics, Histone Deacetylase Inhibitors therapeutic use, Hydroxamic Acids therapeutic use, Prostatic Neoplasms therapy, Tumor Suppressor Proteins genetics
- 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., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
17. Restoration of senescence in breast and ovarian cancer cells following the transfer of the YAC carrying SEN6A gene located at 6q16.3.
- Author
-
Rane NS, Sandhu AK, Zhawar VS, Kaur G, Popescu NC, Kandpal RP, Jhanwar-Uniyal M, and Athwal RS
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Chromosome Mapping, Chromosomes, Artificial, Bacterial, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Ovarian Neoplasms metabolism, Rats, Breast Neoplasms genetics, Cellular Senescence genetics, Chromosomes, Artificial, Yeast, Chromosomes, Human, Pair 6, Genetic Loci genetics, Ovarian Neoplasms genetics
- 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.
- Published
- 2011
18. A novel isoform of the 8p22 tumor suppressor gene DLC1 suppresses tumor growth and is frequently silenced in multiple common tumors.
- Author
-
Low JS, Tao Q, Ng KM, Goh HK, Shu XS, Woo WL, Ambinder RF, Srivastava G, Shamay M, Chan AT, Popescu NC, and Hsieh WS
- Subjects
- Base Sequence, DNA Methylation, DNA Primers, Gene Silencing, Humans, Molecular Sequence Data, Neoplasms genetics, Chromosomes, Human, Pair 8, GTPase-Activating Proteins genetics, Genes, Tumor Suppressor, Neoplasms pathology, Tumor Suppressor Proteins genetics
- Abstract
The critical 8p22 tumor suppressor deleted in liver cancer 1 (DLC1) is frequently inactivated by aberrant CpG methylation and/or genetic deletion and implicated in tumorigeneses of multiple tumor types. Here, we report the identification and characterization of its new isoform, DLC1 isoform 4 (DLC1-i4). This novel isoform encodes an 1125-aa (amino acid) protein with distinct N-terminus as compared with other known DLC1 isoforms. Similar to other isoforms, DLC1-i4 is expressed ubiquitously in normal tissues and immortalized normal epithelial cells, suggesting a role as a major DLC1 transcript. However, differential expression of the four DLC1 isoforms is found in tumor cell lines: Isoform 1 (longest) and 3 (short thus probably nonfunctional) share a promoter and are silenced in almost all cancer and immortalized cell lines, whereas isoform 2 and 4 utilize different promoters and are frequently downregulated. DLC1-i4 is significantly downregulated in multiple carcinoma cell lines, including 2/4 nasopharyngeal, 8/16 (50%) esophageal, 4/16 (25%) gastric, 6/9 (67%) breast, 3/4 colorectal, 4/4 cervical and 2/8(25%) lung carcinoma cell lines. The functional DLC1-i4 promoter is within a CpG island and is activated by wild-type p53. CpG methylation of the DLC1-i4 promoter is associated with its silencing in tumor cells and was detected in 38-100% of multiple primary tumors. Treatment with 5-aza-2'-deoxycytidine or genetic double knockout of DNMT1 and DNMT3B led to demethylation of the promoter and reactivation of its expression, indicating a predominantly epigenetic mechanism of silencing. Ectopic expression of DLC1-i4 in silenced tumor cells strongly inhibited their growth and colony formation. Thus, we identified a new isoform of DLC1 with tumor suppressive function. The differential expression of various DLC1 isoforms suggests interplay in modulating the complex activities of DLC1 during carcinogenesis.
- Published
- 2011
- Full Text
- View/download PDF
19. DLC1 interaction with S100A10 mediates inhibition of in vitro cell invasion and tumorigenicity of lung cancer cells through a RhoGAP-independent mechanism.
- Author
-
Yang X, Popescu NC, and Zimonjic DB
- Subjects
- 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, Annexin A2 metabolism, Carcinoma, Non-Small-Cell Lung metabolism, GTPase-Activating Proteins metabolism, Lung Neoplasms metabolism, S100 Proteins metabolism, Tumor Suppressor Proteins metabolism
- 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., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
20. Synergistic antineoplastic effect of DLC1 tumor suppressor protein and histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), on prostate and liver cancer cells: perspectives for therapeutics.
- Author
-
Zhou X, Yang XY, and Popescu NC
- Subjects
- 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, Genetic Therapy, Histone Deacetylase Inhibitors pharmacology, Hydroxamic Acids pharmacology, Liver Neoplasms therapy, Prostatic Neoplasms therapy, Tumor Suppressor Proteins genetics
- 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.
- Published
- 2010
- Full Text
- View/download PDF
21. Role of SV40 integration site at chromosomal interval 1q21.1 in immortalized CRL2504 cells.
- Author
-
Liu J, Kaur G, Zhawar VK, Zimonjic DB, Popescu NC, Kandpal RP, and Athwal RS
- Subjects
- 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, Antigens, Polyomavirus Transforming genetics, Bronchi physiology, Cell Transformation, Viral genetics, Chromosomes, Human, Pair 1, Simian virus 40 genetics, Virus Integration
- 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.
- Published
- 2009
- Full Text
- View/download PDF
22. Acquired genetic and functional alterations associated with transforming growth factor beta type I resistance in Hep3B human hepatocellular carcinoma cell line.
- Author
-
Zimonjic DB, Zhou X, Lee JS, Ullmannova-Benson V, Tripathi V, Thorgeirsson SS, and Popescu NC
- Subjects
- 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, Carcinoma, Hepatocellular genetics, Liver Neoplasms genetics, Transforming Growth Factor beta1 genetics
- 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.
- Published
- 2009
- Full Text
- View/download PDF
23. Recurrent and nonrandom DNA copy number and chromosome alterations in Myc transgenic mouse model for hepatocellular carcinogenesis: implications for human disease.
- Author
-
Zimonjic DB, Ullmannova-Benson V, Factor VM, Thorgeirsson SS, and Popescu NC
- Subjects
- 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, 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
- 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.
- Published
- 2009
- Full Text
- View/download PDF
24. p120Ras-GAP binds the DLC1 Rho-GAP tumor suppressor protein and inhibits its RhoA GTPase and growth-suppressing activities.
- Author
-
Yang XY, Guan M, Vigil D, Der CJ, Lowy DR, and Popescu NC
- Subjects
- Catalytic Domain, Cell Line, Tumor, Cell Proliferation, GTPase-Activating Proteins, Humans, Tumor Suppressor Proteins analysis, Tumor Suppressor Proteins antagonists & inhibitors, Tumor Suppressor Proteins chemistry, p120 GTPase Activating Protein analysis, p120 GTPase Activating Protein chemistry, src Homology Domains, Neoplasms pathology, Tumor Suppressor Proteins physiology, p120 GTPase Activating Protein physiology, rhoA GTP-Binding Protein metabolism
- Abstract
DLC1 (deleted in liver cancer 1), which encodes a Rho GTPase-activating protein (Rho-GAP), is a potent tumor suppressor gene that is frequently inactivated in several human cancers. DLC1 is a multidomain protein that has been shown previously to bind members of the tensin gene family. Here we show that p120Ras-GAP (Ras-GAP; also known as RASA1) interacts and extensively colocalizes with DLC1 in focal adhesions. The binding was mapped to the SH3 domain located in the N terminus of Ras-GAP and to the Rho-GAP catalytic domain located in the C terminus of the DLC1. In vitro analyses with purified proteins determined that the isolated Ras-GAP SH3 domain inhibits DLC1 Rho-GAP activity, suggesting that Ras-GAP is a negative regulator of DLC1 Rho-GAP activity. Consistent with this possibility, we found that ectopic overexpression of Ras-GAP in a Ras-GAP-insensitive tumor line impaired the growth-suppressing activity of DLC1 and increased RhoA activity in vivo. Our observations expand the complexity of proteins that regulate DLC1 function and define a novel mechanism of the cross talk between Ras and Rho GTPases.1R01CA129610
- Published
- 2009
- Full Text
- View/download PDF
25. DLC1 suppresses distant dissemination of human hepatocellular carcinoma cells in nude mice through reduction of RhoA GTPase activity, actin cytoskeletal disruption and down-regulation of genes involved in metastasis.
- Author
-
Zhou X, Zimonjic DB, Park SW, Yang XY, Durkin ME, and Popescu NC
- Subjects
- 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, 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
- 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.
- Published
- 2008
- Full Text
- View/download PDF
26. Adenovirus-mediated restoration of expression of the tumor suppressor gene DLC1 inhibits the proliferation and tumorigenicity of aggressive, androgen-independent human prostate cancer cell lines: prospects for gene therapy.
- Author
-
Guan M, Tripathi V, Zhou X, and Popescu NC
- Subjects
- Androgens metabolism, Animals, Cell Line, Tumor, GTPase-Activating Proteins, Humans, Male, Mice, Mice, Nude, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Tumor Suppressor Proteins genetics, Xenograft Model Antitumor Assays, Adenoviridae genetics, Cell Proliferation, Genetic Therapy methods, Prostatic Neoplasms therapy, Tumor Suppressor Proteins physiology
- Abstract
Our recent study showing highly recurrent loss of function of DLC1 (deleted in liver cancer 1), a tumor suppressor gene in primary prostate carcinoma (PCA), implicates this gene in the pathogenesis of this disease. To evaluate the response of PCA to oncosuppressive activity of DLC1, we examined now the effects of adenoviral vector for human DLC1 transduction into the DLC1-deficient, androgen-independent (AI) and aggressive human PCA cell lines PC-3 and C4-2-B2. Adenovirus-mediated restoration of DLC1 expression inhibited the proliferation, invasiveness and anchorage-independent growth of PC-3 and C4-2-B2 cells in vitro as well as the tumorigenicity of PC-3 cells in nude mice. It also induced cell-cycle arrest, inhibited the activation of RhoA and the formation of actin stress fibers. DLC1 induced apoptosis in C4-2-B2 cells, whereas it did not elicit such an effect in PC-3 cells. The abundance of the antiapoptotic protein Bcl-2 was greater in PC-3 cells than in C4-2-B2 cells, and PC-3 cells were rendered sensitive to DLC1-induced apoptosis by treatment with the Bcl-2 inhibitor HA14-1. These results suggest that adenovirus-mediated DLC1 transfer, alone or together with other agents, such as inhibitors of Bcl-2 or histone deacetylase, might prove effective in the treatment of aggressive, AI-PCA.
- Published
- 2008
- Full Text
- View/download PDF
27. Delayed kinetics of DNA double-strand break processing in normal and pathological aging.
- Author
-
Sedelnikova OA, Horikawa I, Redon C, Nakamura A, Zimonjic DB, Popescu NC, and Bonner WM
- Subjects
- 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, Aging pathology, Cellular Senescence physiology, DNA Repair
- 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.
- Published
- 2008
- Full Text
- View/download PDF
28. Morphological changes and nuclear translocation of DLC1 tumor suppressor protein precede apoptosis in human non-small cell lung carcinoma cells.
- Author
-
Yuan BZ, Jefferson AM, Millecchia L, Popescu NC, and Reynolds SH
- Subjects
- Actins metabolism, Amino Acid Sequence, Cell Line, Tumor, Cell Movement, Cell Proliferation, Cell Surface Extensions metabolism, Cytoskeleton metabolism, Epithelial Cells cytology, Epithelial Cells metabolism, GTPase-Activating Proteins, Humans, Lung Neoplasms metabolism, Molecular Sequence Data, Mutant Proteins metabolism, Nuclear Localization Signals metabolism, Protein Structure, Tertiary, Protein Transport, Serine, Structure-Activity Relationship, Tumor Suppressor Proteins chemistry, Apoptosis, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Nucleus metabolism, Cell Shape, Lung Neoplasms pathology, Tumor Suppressor Proteins metabolism
- Abstract
We have previously shown that reactivation of DLC1, a RhoGAP containing tumor suppressor gene, inhibits tumorigenicity of human non-small cell lung carcinoma cells (NSCLC). After transfection of NSCLC cells with wild type (WT) DLC1, changes in cell morphology were observed. To determine whether such changes have functional implications, we generated several DLC1 mutants and examined their effects on cell morphology, proliferation, migration and apoptosis in a DLC1 deficient NSCLC cell line. We show that WT DLC1 caused actin cytoskeleton-based morphological alterations manifested as cytoplasmic extensions and membrane blebbings in most cells. Subsequently, a fraction of cells exhibiting DLC1 protein nuclear translocation (PNT) underwent caspase 3-dependent apoptosis. We also show that the RhoGAP domain is essential for the occurrence of morphological alterations, PNT and apoptosis, and the inhibition of cell migration. DLC1 PNT is dependent on a bipartite nuclear localizing sequence and most likely is regulated by a serine-rich domain at N-terminal part of the DLC1 protein. Also, we found that DLC1 functions in the cytoplasm as an inhibitor of tumor cell proliferation and migration, but in the nucleus as an inducer of apoptosis. Our analyses provide evidence for a possible link between morphological alterations, PNT and proapoptotic and anti-oncogenic activities of DLC1 in lung cancer.
- Published
- 2007
- Full Text
- View/download PDF
29. Nonclassic functions of human topoisomerase I: genome-wide and pharmacologic analyses.
- Author
-
Miao ZH, Player A, Shankavaram U, Wang YH, Zimonjic DB, Lorenzi PL, Liao ZY, Liu H, Shimura T, Zhang HL, Meng LH, Zhang YW, Kawasaki ES, Popescu NC, Aladjem MI, Goldstein DJ, Weinstein JN, and Pommier Y
- Subjects
- 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, Breast Neoplasms enzymology, Colonic Neoplasms enzymology, DNA Topoisomerases, Type I physiology
- 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.
- Published
- 2007
- Full Text
- View/download PDF
30. DLC-1:a Rho GTPase-activating protein and tumour suppressor.
- Author
-
Durkin ME, Yuan BZ, Zhou X, Zimonjic DB, Lowy DR, Thorgeirsson SS, and Popescu NC
- Subjects
- 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, GTPase-Activating Proteins metabolism, Tumor Suppressor Proteins metabolism
- 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.
- Published
- 2007
- Full Text
- View/download PDF
31. Enrichment of a population of mammary gland cells that form mammospheres and have in vivo repopulating activity.
- Author
-
Liao MJ, Zhang CC, Zhou B, Zimonjic DB, Mani SA, Kaba M, Gifford A, Reinhardt F, Popescu NC, Guo W, Eaton EN, Lodish HF, and Weinberg RA
- Subjects
- Animals, Cell Culture Techniques, Cell Differentiation, Cell Shape, Female, Mice, Mice, Inbred C57BL, Mice, Knockout, Organoids cytology, Adult Stem Cells cytology, Adult Stem Cells transplantation, Cell Proliferation, Mammary Glands, Animal cytology, Mammary Glands, Animal transplantation
- 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.
- Published
- 2007
- Full Text
- View/download PDF
32. The interferon-alpha responsive gene TMEM7 suppresses cell proliferation and is downregulated in human hepatocellular carcinoma.
- Author
-
Zhou X, Popescu NC, Klein G, and Imreh S
- Subjects
- Amino Acid Sequence, Animals, Azacitidine analogs & derivatives, Azacitidine pharmacology, Cell Line, Tumor, DNA Methylation, Decitabine, Epigenesis, Genetic, Histone Deacetylase Inhibitors, Humans, Hydroxamic Acids pharmacology, Immunohistochemistry, Male, Mice, Mice, Nude, Molecular Sequence Data, Neoplasm Invasiveness prevention & control, Promoter Regions, Genetic, Sequence Homology, Amino Acid, Carcinoma, Hepatocellular genetics, Cell Proliferation, Gene Silencing, Genes, Tumor Suppressor physiology, Interferon-alpha pharmacology, Liver Neoplasms genetics
- Abstract
Multiple regions on the chromosome arm 3p are frequently affected by loss of heterozygosity in human cancers. A candidate tumor suppressor gene is TMEM7, at 3p21.3, which encodes a transmembrane protein. TMEM7 is expressed specifically in the liver, and the encoded protein shares substantial sequence homology with human and mouse 28-kDa interferon-alpha (IFN-alpha) responsive protein. In investigation of the possible role of TMEM7 in development of hepatocellular carcinoma (HCC), we examined TMEM7 expression in 20 primary HCC and 18 HCC cell lines and found recurrent functional alterations. Although TMEM7 mRNA was expressed in normal hepatic cells, downregulation or inactivation of the gene was detected in 85% of primary HCC and 33% of HCC cell lines. To identify the mechanisms responsible, we examined genomic deletion and mutation, and also the effect of inhibitors of DNA methyltransferase and histone deacetylase on cells with low or no endogenous TMEM7 expression. Homozygous deletion of TMEM7 was not detected in 17 pairs of human HCC and corresponding noncancerous liver tissues, nor in any of the 18 HCC cell lines. TMEM7 mutation was not detected in the 18 HCC cell lines (low or normal TMEM7 expression). Treatment of two of six cell lines exhibiting downregulation or loss of TMEM7 with 5-aza-2'-deoxycytidine and trichostatin A yielded additive increase in TMEM7 expression, implicating aberrant DNA methylation and histone deacetylation in transcriptional silencing of this gene. Ectopic expression of TMEM7 in two TMEM7-deficient HCC lines suppressed cell proliferation, colony formation, and cell migration in vitro and reduced tumor formation in nude mice. Treatment of two highly invasive HCC cell lines with IFN-alpha for 7 days significantly increased TMEM7 expression and inhibited cell migration. These findings implicate loss of TMEM7 expression in hepatocarcinogenesis and suggest that modification of TMEM7 expression by IFN-alpha may have therapeutic relevance in a subset of HCC.
- Published
- 2007
- Full Text
- View/download PDF
33. Deleted in liver cancer 3 (DLC-3), a novel Rho GTPase-activating protein, is downregulated in cancer and inhibits tumor cell growth.
- Author
-
Durkin ME, Ullmannova V, Guan M, and Popescu NC
- Subjects
- Amino Acid Sequence, Breast metabolism, Breast Neoplasms metabolism, Cell Proliferation, Down-Regulation, Female, Genes, Tumor Suppressor, Humans, Male, Molecular Sequence Data, Prostate metabolism, Prostatic Neoplasms metabolism, Reverse Transcriptase Polymerase Chain Reaction, GTPase-Activating Proteins metabolism, Neoplasms metabolism, Tumor Suppressor Proteins genetics
- Abstract
Two related Rho GTPase-activating proteins, DLC-1 (deleted in liver cancer 1) and DLC-2, are emerging as bona fide tumor suppressor genes that inhibit cancer cell growth. In this report, we characterized a gene on chromosome Xq13 that encodes DLC-3 (also known as KIAA0189 and STARD8), a third member of the DLC family. The DLC-3 gene has transcripts with alternative 5' ends, one of which, DLC-3alpha, encodes an 1103-amino acid polypeptide highly similar to DLC-1 and DLC-2. A second isoform (DLC-3beta) would yield a protein lacking the N-terminal sterile alpha motif domain. The DLC-3 gene is widely expressed in normal tissues, but DLC-3 mRNA levels were low or absent in a significant number of breast, ovarian, liver and prostate cancer cell lines. Using a cancer profiling array to compare matched tumor and normal human tissues, downregulation of DLC-3 mRNA was observed in kidney, lung, ovarian, uterine and breast cancer samples. By quantitative reverse transcriptase-polymerase chain reaction, DLC-3 expression was reduced in primary prostate carcinomas relative to normal prostate tissue. Transfection of human breast and prostate cancer cells with a DLC-3alpha expression vector inhibited cell proliferation, colony formation and growth in soft agar. These results indicate that deregulation of DLC-3 may contribute to breast and prostate tumorigenesis.
- Published
- 2007
- Full Text
- View/download PDF
34. Oncogenic inhibition by a deleted in liver cancer gene requires cooperation between tensin binding and Rho-specific GTPase-activating protein activities.
- Author
-
Qian X, Li G, Asmussen HK, Asnaghi L, Vass WC, Braverman R, Yamada KM, Popescu NC, Papageorge AG, and Lowy DR
- Subjects
- Animals, Cell Line, Humans, Integrins metabolism, Mice, Mutation genetics, Oncogene Proteins genetics, Phosphotyrosine metabolism, Protein Binding, Tensins, Tumor Suppressor Proteins genetics, Tyrosine genetics, Tyrosine metabolism, src Homology Domains, GTPase-Activating Proteins metabolism, Microfilament Proteins metabolism, Oncogene Proteins metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The three deleted in liver cancer genes (DLC1-3) encode Rho-GTPase-activating proteins (RhoGAPs) whose expression is frequently down-regulated or silenced in a variety of human malignancies. The RhoGAP activity is required for full DLC-dependent tumor suppressor activity. Here we report that DLC1 and DLC3 bind to human tensin1 and its chicken homolog. The binding has been mapped to the tensin Src homology 2 (SH2) and phosphotyrosine binding (PTB) domains at the C terminus of tensin proteins. Distinct DLC1 sequences are required for SH2 and PTB binding. DCL binding to both domains is constitutive under basal conditions. The SH2 binding depends on a tyrosine in DCL1 (Y442) but is phosphotyrosine-independent, a highly unusual feature for SH2 binding. DLC1 competed with the binding of other proteins to the tensin C terminus, including beta 3-integrin binding to the PTB domain. Point mutation of a critical tyrosine residue (Y442F) in DLC1 rendered the protein deficient for binding the tensin SH2 domain and binding full-length tensin. The Y442F protein was diffusely cytoplasmic, in contrast to the localization of wild-type DLC1 to focal adhesions, but it retained the ability to reduce the intracellular levels of Rho-GTP. The Y442F mutant displayed markedly reduced biological activity, as did a mutant that was RhoGAP-deficient. The results suggest that DLC1 is a multifunctional protein whose biological activity depends on cooperation between its tensin binding and RhoGAP activities, although neither activity depends on the other.
- Published
- 2007
- Full Text
- View/download PDF
35. Inhibition of cell proliferation, induction of apoptosis, reactivation of DLC1, and modulation of other gene expression by dietary flavone in breast cancer cell lines.
- Author
-
Ullmannova V and Popescu NC
- Subjects
- Biomarkers, Tumor metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Caspase 3 metabolism, Cell Cycle, Cyclin-Dependent Kinase Inhibitor p21 metabolism, DNA Methylation, Female, Flow Cytometry, GTPase-Activating Proteins, Gene Expression Profiling, Genes, Tumor Suppressor, Humans, Immunoblotting, Male, Oligonucleotide Array Sequence Analysis, Proto-Oncogene Mas, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Tumor Suppressor Proteins metabolism, Apoptosis drug effects, Breast Neoplasms pathology, Cell Proliferation drug effects, Flavones pharmacology, Gene Expression Regulation, Neoplastic drug effects, Tumor Suppressor Proteins genetics
- Abstract
Background: Dietary flavone was previously shown to increase the expression of deleted in liver cancer-1 gene (DLC-1) in HT-29 colon carcinoma cell line [Herzog A, Kindermann B, Doring F, Daniel H, Wenzel U. Pleiotropic molecular effects of the pro-apoptotic dietary constituent flavone in human colon cancer cells identified by protein and mRNA expression profiling. Proteomics 2004;4:2455-64]. DLC-1 that encodes a Rho GTPase-activating protein, functions as a tumor suppressor gene and is frequently inactivated or down-regulated in several common cancers. Restoration of DLC-1 expression suppresses in vitro tumor cells proliferation and tumorigenicity in vivo., Methods: Here, the effect of flavone was examined in several DLC-1-deficient cell lines derived from different types human cancer using assays for cell proliferation, gene expression and transfer., Results: We show that exposure to 150 microM flavone increased DLC1 expression in breast but not in liver or prostate carcinoma cells or a nonmalignant breast epithelial cell line. Flavone restored the expression of DLC1 in the breast carcinoma cell lines MDA-MB-468, MDA-MB-361, and BT20 as well as in the colon carcinoma cell line HT-29 all of which are DLC-1-negative due to promoter hypermethylation. We further show that flavone inhibited cell proliferation, induced cell cycle arrest at G(2)-M, increased p21(Waf1) gene expression, and caused apoptosis. Microarray analysis of these aggressive and metastatic breast carcinoma cells revealed 29 flavone-responsive genes, among which the DNA damage-inducible GADD genes were up-regulated and the proto-oncogene STMN1 and IGFBP3 were down-regulated., Conclusions: Flavone-mediated alterations of genes that regulate tumor cell proliferation, cell cycle, and apoptosis contribute to chemopreventive and antitumoral effects of flavone. Alone or in combination with demethylating agents, flavone may be an effective adjunct to chemotherapy in preventing breast cancer metastasis.
- Published
- 2007
- Full Text
- View/download PDF
36. Expression profile of the tumor suppressor genes DLC-1 and DLC-2 in solid tumors.
- Author
-
Ullmannova V and Popescu NC
- Subjects
- Cell Line, Tumor, GTPase-Activating Proteins, Humans, Oligonucleotide Array Sequence Analysis, Gene Expression Profiling, Genes, Tumor Suppressor, Neoplasms genetics, Tumor Suppressor Proteins genetics
- Abstract
Several years after the isolation of deleted in liver cancer 1 (DLC-1), a gene that encodes a Rho GTPase activating protein, the closely related DLC-2 gene was identified. DLC-1 and DLC-2 are approximately 50% identical and share the same SAM-RhoGAP-START domain organization. Since DLC-1 and -2 are located at chromosome regions that are commonly deleted in cancer cells and have been found to function as tumor suppressor genes, we sought to compare their expression profiles in several common types of cancer and to determine whether dlc1 and dlc2 proteins cooperate in tumor development. Using cancer-profiling arrays, we detected for the first time down-regulation of DLC-1 expression in renal, uterine and rectal cancers and down-regulation of DLC-2 expression in lung, ovarian, renal, breast, uterine, gastric, colon and rectal tumors. Since DLC-1 also functions as a metastasis suppressor gene in breast cancer, DLC-1 and DLC-2 expression were examined in a series of primary ductal carcinomas derived from patients with regional lymph node metastases. Using quantitative RT-PCR we detected a significantly lower expression of DLC-1 and DLC-2 in high percentage of tumors, suggesting that deficiency of either DLC gene facilitates dissemination of breast carcinoma cells to secondary sites. We examined DLC-2 expression in DLC-1-negative cell lines derived from human breast, non-small cell lung, and hepatocellular carcinomas, that could be rendered less or non-tumorigenic by ectopic expression of DLC-1. DLC-2 transcripts were detected in all cell lines, indicating that none of the cells were deficient in both members of the DLC family. This comparative expression analysis of DLC-1 and -2 identifies down-regulation of the two emerging bona fide tumor suppressor genes in additional types of solid tumors. The large spectrum of cancers with dysregulated DLC genes underlines the involvement of this family of genes in cancer development.
- Published
- 2006
37. Aberrant methylation and deacetylation of deleted in liver cancer-1 gene in prostate cancer: potential clinical applications.
- Author
-
Guan M, Zhou X, Soulitzis N, Spandidos DA, and Popescu NC
- Subjects
- Acetylation, Adult, Aged, Aged, 80 and over, Azacitidine analogs & derivatives, Azacitidine pharmacology, Base Sequence, Blotting, Southern, Blotting, Western, Chromatin Immunoprecipitation, Decitabine, Down-Regulation, Enzyme Inhibitors pharmacology, GTPase-Activating Proteins, Humans, Male, Middle Aged, Molecular Sequence Data, Promoter Regions, Genetic genetics, Prostate metabolism, Prostatic Hyperplasia genetics, Prostatic Hyperplasia metabolism, Prostatic Hyperplasia pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Tumor Cells, Cultured, Tumor Suppressor Proteins metabolism, DNA Methylation, Gene Deletion, Prostatic Neoplasms genetics, Tumor Suppressor Proteins genetics
- Abstract
Purpose: The deleted in liver cancer-1 (DLC-1) gene that encodes a Rho GTPase-activating protein with tumor suppressor function is located on chromosome 8p21-22, a region frequently deleted in prostate carcinomas. This study was designed to determine whether DLC-1 is deregulated in prostate carcinomas and to assess the contribution of DLC-1 alterations to prostate carcinogenesis., Experimental Design: Primary prostate carcinomas, prostate carcinoma cell lines, benign prostatic hyperplasias, and normal prostatic tissues were examined for detection of functional and structural alterations of the DLC-1 gene by real-time PCR, methylation-specific PCR, and Southern and Western blots., Results: Down-regulation or loss of DCL-1 mRNA expression was detected in 10 of 27 (37%) prostate carcinomas, 3 of 5 (60%) prostate carcinoma cell lines, and 5 of 21 (24%) benign prostatic hyperplasias. DLC-1 promoter methylation was identified in 13 of 27 (48%) prostate carcinomas and 2 matching normal tissues and in 15 of 21 (71%) benign prostatic hyperplasias but was absent in 10 normal prostatic tissues from noncancerous individuals. Genomic deletions were found in only 3 prostate carcinomas and 1 benign prostatic hyperplasia. DLC-1 protein was not detected in 8 of 27 (30%) prostate carcinomas and 11 of 21 (52%) benign prostatic hyperplasias. Methylation of DLC-1 correlated with age in prostate carcinoma patients (P = 0.006) and with prostate-specific antigen blood levels in benign prostatic hyperplasia patients (P = 0.029). Treatment of the three prostate carcinoma cell lines (PC-3, LNCaP, and 22Rv1) expressing a low level of DLC-1 transcripts with inhibitors of DNA methyltransferase or histone deacetylase increased DLC-1 expression., Conclusions: These results show that the transcriptional silencing of DLC-1 by two epigenetic mechanisms is common and may be involved in the pathogenesis of prostate carcinomas and benign prostatic hyperplasias and could have potential clinical application in the early detection and gene therapy of prostate cancer.
- Published
- 2006
- Full Text
- View/download PDF
38. POTE paralogs are induced and differentially expressed in many cancers.
- Author
-
Bera TK, Saint Fleur A, Lee Y, Kydd A, Hahn Y, Popescu NC, Zimonjic DB, Lee B, and Pastan I
- Subjects
- Base Sequence, Cell Line, Tumor, Female, Humans, In Situ Hybridization, Fluorescence, Male, Molecular Sequence Data, Neoplasm Proteins genetics, Neoplasms genetics, Neoplasms pathology, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Placenta metabolism, Placenta pathology, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Reverse Transcriptase Polymerase Chain Reaction, Testicular Neoplasms genetics, Testicular Neoplasms metabolism, Testicular Neoplasms pathology, Neoplasm Proteins biosynthesis, Neoplasms metabolism
- Abstract
To identify new antigens that are targets for the immunotherapy of prostate and breast cancer, we used expressed sequence tag and genomic databases and discovered POTE, a new primate-specific gene family. Each POTE gene encodes a protein that contains three domains, although the proteins vary greatly in size. The NH2-terminal domain is novel and has properties of an extracellular domain but does not contain a signal sequence. The second and third domains are rich in ankyrin repeats and spectrin-like helices, respectively. The protein encoded by POTE-21, the first family member discovered, is localized on the plasma membrane of the cell. In humans, 13 highly homologous paralogs are dispersed among eight chromosomes. The expression of POTE genes in normal tissues is restricted to prostate, ovary, testis, and placenta. A survey of several cancer samples showed that POTE was expressed in 6 of 6 prostate, 12 of 13 breast, 5 of 5 colon, 5 of 6 lung, and 4 of 5 ovarian cancers. To determine the relative expression of each POTE paralog in cancer and normal samples, we employed a PCR-based cloning and analysis method. We found that POTE-2alpha, POTE-2beta, POTE-2gamma, and POTE-22 are predominantly expressed in cancers whereas POTE expression in normal tissues is somewhat more diverse. Because POTE is primate specific and is expressed in testis and many cancers but only in a few normal tissues, we conclude POTE is a new primate-specific member of the cancer-testis antigen family. It is likely that POTE has a unique role in primate biology.
- Published
- 2006
- Full Text
- View/download PDF
39. Effect of hepatitis C virus core protein on the molecular profiling of human B lymphocytes.
- Author
-
Wu CG, Budhu A, Chen S, Zhou X, Popescu NC, Valerie K, and Wang XW
- Subjects
- Adenoviridae genetics, Apoptosis, B-Lymphocytes cytology, B-Lymphocytes immunology, Blotting, Western, Gene Expression Regulation, Hepacivirus physiology, Humans, Microarray Analysis, Multigene Family, RNA, Messenger genetics, RNA, Messenger metabolism, Reproducibility of Results, Viral Core Proteins immunology, beta-Galactosidase metabolism, B-Lymphocytes metabolism, B-Lymphocytes virology, Gene Expression Profiling, Viral Core Proteins metabolism
- Abstract
Hepatitis C virus (HCV) core protein features many intriguing properties and plays a pivotal role in cellular immunity, cell growth, apoptosis, cell transformation, and eventually in tumor development. However, the role of B cells, the primary players in the humoral immune response, during HCV infection is largely unknown. To explore the molecular effects of HCV core on human B cells, we conducted gene expression profiling of serial RNA samples from B cells that were infected with adenovirus harboring full-length HCV core protein and beta-galactosidase as a reference using a microarray platform containing 22,149 human oligo probes. The entire experiment was performed in duplicate in B lymphocytes that were isolated from two individual donors and incubated for up to 3 days after infection with adenovirus expressing HCV core protein to identify dynamic gene expression patterns. Differential expression of representative genes was validated by quantitative RT-PCR. We found that HCV core significantly inhibited B-lymphocyte apoptosis. We showed a dramatic downregulation of MHC class II molecules in B cells expressing HCV core, whereas the expression of immunoglobulin genes was not significantly altered. Moreover, genes associated with leukemia and B-lymphoma were consistently upregulated by HCV core. In contrast, downregulation of caspase-1 and caspase-4 was found to be associated with core's ability to prevent B-lymphocyte apoptosis. In summary, we have identified several clusters of genes that are differentially expressed in human B lymphocytes expressing HCV core, suggesting a potential impairment of antigen processing and presentation, which may provide more insights into HCV infection in B lymphocytes.
- Published
- 2006
- Full Text
- View/download PDF
40. GTF2IRD1 in craniofacial development of humans and mice.
- Author
-
Tassabehji M, Hammond P, Karmiloff-Smith A, Thompson P, Thorgeirsson SS, Durkin ME, Popescu NC, Hutton T, Metcalfe K, Rucka A, Stewart H, Read AP, Maconochie M, and Donnai D
- Subjects
- Adolescent, Adult, Animals, Cell Line, Child, Child, Preschool, Chromosomes, Human, Pair 7, Female, Gene Deletion, Goosecoid Protein genetics, Goosecoid Protein physiology, Homozygote, Humans, Infant, Infant, Newborn, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Transcription Factors, TFII physiology, Craniofacial Abnormalities genetics, Face embryology, Muscle Proteins physiology, Nuclear Proteins physiology, Skull embryology, Trans-Activators physiology, Williams Syndrome genetics
- Abstract
Craniofacial abnormalities account for about one-third of all human congenital defects, but our understanding of the genetic mechanisms governing craniofacial development is incomplete. We show that GTF2IRD1 is a genetic determinant of mammalian craniofacial and cognitive development, and we implicate another member of the TFII-I transcription factor family, GTF2I, in both aspects. Gtf2ird1-null mice exhibit phenotypic abnormalities reminiscent of the human microdeletion disorder Williams-Beuren syndrome (WBS); craniofacial imaging reveals abnormalities in both skull and jaws that may arise through misregulation of goosecoid, a downstream target of Gtf2ird1. In humans, a rare WBS individual with an atypical deletion, including GTF2IRD1, shows facial dysmorphism and cognitive deficits that differ from those of classic WBS cases. We propose a mechanism of cumulative dosage effects of duplicated and diverged genes applicable to other human chromosomal disorders.
- Published
- 2005
- Full Text
- View/download PDF
41. Chromosomal aberrations in cell lines derived from thyroid tumors spontaneously developed in TRbetaPV/PV mice.
- Author
-
Zimonjic DB, Kato Y, Ying H, Popescu NC, and Cheng SY
- Subjects
- Adenocarcinoma, Follicular metabolism, Animals, Cell Line, Tumor, Mice, Mice, Mutant Strains, Mutation, Spectral Karyotyping, Thyroglobulin analysis, Thyroid Neoplasms metabolism, Adenocarcinoma, Follicular genetics, Chromosome Aberrations, Thyroid Hormone Receptors beta genetics, Thyroid Neoplasms genetics
- Abstract
The etiology and genetic alterations of follicular thyroid carcinoma are not well understood. By targeting a mutation (PV) into the thyroid hormone receptor beta gene (TRbetaPV mouse), we created a knock-in mutant TRbeta(PV/PV) mouse that spontaneously develop follicular thyroid carcinoma with progression to metastasis similar to human follicular thyroid carcinoma. This mouse model provides a valuable tool to ascertain the nature and the extent of genomic rearrangements that occur during carcinogenesis of the thyroid. Spectral karyotyping analysis (SKY) of seven cell lines derived from thyroid tumors developed in TRbeta(PV/PV) mice showed that all of them had abnormal karyotypes, with chromosome number ranging from near-diploid (39-42 chromosomes) to hypotetraploid (63-79 chromosomes). These seven cell lines also exhibited a variety of structural chromosomal aberrations, including common recurrent translocations and deletions. This SKY analysis shows that the development and progression of follicular thyroid carcinoma in knock-in TRbeta(PV/PV) mutant mice comprise recurrent structural and numerical genomic changes, some of which mimic those described in human thyroid cancer.
- Published
- 2005
- Full Text
- View/download PDF
42. The RhoGAP protein DLC-1 functions as a metastasis suppressor in breast cancer cells.
- Author
-
Goodison S, Yuan J, Sloan D, Kim R, Li C, Popescu NC, and Urquidi V
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Movement genetics, DNA, Complementary genetics, Female, GTPase-Activating Proteins, Gene Expression Regulation, Neoplastic, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Neoplasm Metastasis, Neoplasm Transplantation, Oligonucleotide Array Sequence Analysis, Transduction, Genetic, Transplantation, Heterologous, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins genetics, Up-Regulation, Breast Neoplasms pathology, Tumor Suppressor Proteins physiology
- Abstract
The identification of molecular signatures characteristic of tumor cells that are capable of metastatic spread is required for the development of therapeutic interventions to abrogate this lethal process. To facilitate this, we have previously characterized an experimental system in which the role of candidate metastasis-related genes can be screened and tested. Monoclonal cell lines M4A4 and NM2C5 are spontaneously occurring sublines of the MDA-MB-435 cell breast tumor cell line that exhibit phenotypic differences in growth, invasion, and metastatic efficiency in athymic mice. In this study, transcriptional profiles of these cell lines were created using oligonucleotide microarrays representing over 12,000 genes. Intensity modeling and hierarchical clustering analysis identified a 171-gene expression signature that correlated with metastatic phenotype and highlighted several GTPase signaling components. Restoration of one of these GTPases, deleted in liver cancer-1 (DLC-1), in metastatic M4A4 cells to levels observed in the nonmetastatic NM2C5 cell line resulted in the inhibition of migration and invasion in vitro and a significant reduction in the ability of these cells to form pulmonary metastases in athymic mice. These studies show the utility of expression profiling, in an appropriate experimental system, to identify genetic determinants of metastatic sufficiency. The finding that DLC-1 can act as a metastasis-suppressor gene supports an influential role for GTPase signaling in tumor progression.
- Published
- 2005
- Full Text
- View/download PDF
43. The human AKNA gene expresses multiple transcripts and protein isoforms as a result of alternative promoter usage, splicing, and polyadenylation.
- Author
-
Sims-Mourtada JC, Bruce S, McKeller MR, Rangel R, Guzman-Rojas L, Cain K, Lopez C, Zimonjic DB, Popescu NC, Gordon J, Wilkinson MF, and Martinez-Valdez H
- Subjects
- B-Lymphocytes metabolism, Blotting, Western, CD40 Antigens metabolism, Cell Nucleus metabolism, Chromosomes, Human, Pair 9, DNA-Binding Proteins, Electrophoretic Mobility Shift Assay, Exons, Flow Cytometry, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Lymphoid Tissue metabolism, Nuclear Proteins, Protein Isoforms metabolism, RNA, Messenger metabolism, Restriction Mapping, T-Lymphocytes metabolism, Alternative Splicing, Polyadenylation, Promoter Regions, Genetic, Protein Isoforms genetics, Transcription Factors genetics
- Abstract
We previously showed that the human AKNA gene encodes an AT-hook transcription factor that regulates the expression of costimulatory cell surface molecules on lymphocytes. However, AKNA cDNA probes hybridize with multiple transcripts, suggesting either the existence of other homologous genes or a complex regulation operating on a single gene. Here we report evidence for the latter, as we find that AKNA is encoded by a single gene that spans a 61-kb locus of 24 exons on the fragile FRA9E region of human chromosome 9q32. This gene gives rise to at least nine distinct transcripts, most of which are expressed in a tissue-specific manner in lymphoid organs. Many of the AKNA transcripts originate from alternative splicing; others appear to derive from differential polyadenylation and promoter usage. The alternative AKNA transcripts are predicted to encode overlapping protein isoforms, some of which (p70 and p100) are readily detectable using a polyclonal anti-AKNA antisera that we generated. We also find that AKNA PEST-dependent cleavage into p50 polypeptides is targeted to mature B cells and appears to be required for CD40 upregulation. The unusual capacity of the AKNA gene to generate multiple transcripts and proteins may reflect its functional diversity, and it may also provide a fail-safe mechanism that preserves AKNA expression.
- Published
- 2005
- Full Text
- View/download PDF
44. DLC-1, a Rho GTPase-activating protein with tumor suppressor function, is essential for embryonic development.
- Author
-
Durkin ME, Avner MR, Huh CG, Yuan BZ, Thorgeirsson SS, and Popescu NC
- Subjects
- Animals, Cells, Cultured, Cytoskeleton genetics, Cytoskeleton metabolism, Cytoskeleton pathology, Fibroblasts, GTPase-Activating Proteins deficiency, GTPase-Activating Proteins genetics, Gene Deletion, Gene Expression Regulation, Developmental, Heterozygote, Mice, Mice, Knockout, Mutation genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Transcription, Genetic genetics, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, GTPase-Activating Proteins metabolism, Tumor Suppressor Proteins metabolism
- Abstract
DLC-1 (deleted in liver cancer 1) is a Rho GTPase-activating protein that is able to inhibit cell growth and suppress tumorigenesis. We have used homologous recombination to inactivate the mouse DLC-1 gene (Arhgap7). Mice heterozygous for the targeted allele were phenotypically normal, but homozygous mutant embryos did not survive beyond 10.5 days post coitum. Histological analysis revealed that DLC-1-/- embryos had defects in the neural tube, brain, heart, and placenta. Cultured fibroblasts from DLC-1-deficient embryos displayed alterations in the organization of actin filaments and focal adhesions.
- Published
- 2005
- Full Text
- View/download PDF
45. Assignment of murine placental cathepsin R to mouse chromosome bands 13B2-B3 by fluorescence in situ hybridization.
- Author
-
Zimonjic DB, Liu J, Xu WH, Zhou X, Popescu NC, and Shi GP
- Subjects
- Animals, Base Sequence, Chromosome Banding, Chromosomes, Artificial, Bacterial, Chromosomes, Mammalian, Cloning, Molecular, DNA Primers, Female, In Situ Hybridization, Fluorescence, Mice, Pregnancy, Placenta enzymology, Serine Endopeptidases genetics
- Published
- 2005
- Full Text
- View/download PDF
46. Frequent downregulation and loss of WWOX gene expression in human hepatocellular carcinoma.
- Author
-
Park SW, Ludes-Meyers J, Zimonjic DB, Durkin ME, Popescu NC, and Aldaz CM
- Subjects
- Apoptosis, Carcinoma, Hepatocellular pathology, Chromosome Fragile Sites, Down-Regulation, Humans, Liver Neoplasms pathology, Loss of Heterozygosity, Microsatellite Repeats, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Tumor Suppressor Proteins, WW Domain-Containing Oxidoreductase, Carcinoma, Hepatocellular genetics, Chromosomes, Human, Pair 16 genetics, Gene Deletion, Gene Expression Regulation, Neoplastic, Liver Neoplasms genetics, Oxidoreductases biosynthesis
- Abstract
The WWOX (WW-domain containing oxidoreductase) is a candidate tumour suppressor gene spanning the same chromosome region, 16q23, as the second most common fragile site (FS), FRA16D. Deletions detected by comparative genomic hybridisation (CGH) and loss of heterozygosity at microsatellite markers on chromosome 16q are common in many human cancers including hepatocellular carcinoma (HCC). The development of human HCC is closely associated with exposure to oncogenic viruses and chemical carcinogens, agents known to frequently target common FS. We examined the status of WWOX genomic DNA, RNA and protein in 18 cell lines derived from human HCC and found recurrent alterations of the gene. Loss of DNA copy-number confined to band 16q23 was detected by CGH in several cell lines. Although homozygous deletions of the WWOX gene were not detected, WWOX mRNA expression was absent or lower in 60% of cell lines. The occurrence of aberrant WWOX reverse transcription-PCR products with deletion of exons 6-8 correlated significantly with altered WWOX expression. All of the cell lines showing mRNA downregulation had a decreased or undetectable level of WWOX protein as demonstrated by Western blotting with antibody to WWOX. Furthermore, 13 out of the 18 cell lines expressed decreased levels or no WWOX protein when compared with normal liver. These results show that WWOX gene is frequently altered in HCC and raise the possibility that this gene is implicated in hepatocarcinogenesis.
- Published
- 2004
- Full Text
- View/download PDF
47. Aberrant gene expression in human non small cell lung carcinoma cells exposed to demethylating agent 5-aza-2'-deoxycytidine.
- Author
-
Yuan BZ, Jefferson AM, Popescu NC, and Reynolds SH
- Subjects
- Antimetabolites, Antineoplastic toxicity, Apoptosis drug effects, Carcinoma, Non-Small-Cell Lung, Cell Division drug effects, Cell Line, Tumor, DNA Methylation, Decitabine, Humans, Kinetics, Lung Neoplasms, Oligonucleotide Array Sequence Analysis, Transfection, Azacitidine analogs & derivatives, Azacitidine toxicity, Gene Expression Regulation, Neoplastic drug effects
- Abstract
The identification of genes undergoing genetic or epigenetic alterations and contributing to the development of cancer is critical to our understanding of the molecular mechanisms of carcinogenesis. A new approach in identifying alterations of genes that might be relevant to the process of tumor development was used in this study by examining the gene expression profile in human lung cancer cells exposed to 5-aza-2'-deoxycytidine (5-aza-dC). A cDNA array analysis was carried out on 5-aza-dC-treated and untreated non small cell lung cancer (NSCLC) cell line NCI-H522. Sixteen and 14 genes were upregulated and downregulated, respectively, by 5-aza-dC treatment. Among them, downregulation of tyrosine protein kinase ABL2 (ABL2) gene and upregulation of hint/protein kinase C inhibitor 1 (Hint/PKCI-1), DVL1, TIMP-1, and TRP-1 genes were found in expanded observations in two or three of five 5-aza-dC-treated NSCLC cell lines. Among these genes, we found that cDNA transfer of Hint/PKCI-1 resulted in a significant in vitro growth inhibition in two cell lines exhibiting 5-aza-dC-induced upregulation of Hint/PKCI-1 and significantly reduced in vivo tumorigenicity of one NSCLC cell line. Hint/PKCI-1, which is the only other characterized human histidine triad (HIT) nucleotide-binding protein in addition to tumor-suppressor gene FHIT, might be involved in lung carcinogenesis., (Copyright 2004 Neoplasia Press, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
48. Thirteen-exon-motif signature for vertebrate nuclear and mitochondrial type IB topoisomerases.
- Author
-
Zhang H, Meng LH, Zimonjic DB, Popescu NC, and Pommier Y
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, Cluster Analysis, Conserved Sequence genetics, Gene Duplication, Humans, In Situ Hybridization, Fluorescence, Mitochondria genetics, Molecular Sequence Data, Vertebrates genetics, Cell Nucleus enzymology, DNA Topoisomerases, Type I genetics, Evolution, Molecular, Exons genetics, Mitochondria enzymology
- Abstract
DNA topoisomerases contribute to various cellular activities that involve DNA. We previously identified a human nuclear gene that encodes a mitochondrial DNA topoisomerase. Here we show that genes for mitochondrial DNA topoisomerases (type IB) exist only in vertebrates. A 13-exon topoisomerase motif was identified as a characteristic of genes for both nuclear and mitochondrial type IB topoisomerases. The presence of this signature motif is thus an indicator of the coexistence of nuclear and mitochondrial type IB DNA topoisomerases. We hypothesize that the prototype topoisomerase IB with the 13-exon structure formed first, and then duplicated. One topoisomerase specialized for nuclear DNA and the other for mitochondrial DNA.
- Published
- 2004
- Full Text
- View/download PDF
49. Rho GTPase activating protein cDNA on chromosome 13q12 is the deleted in liver cancer (DLC2) gene.
- Author
-
Popescu NC and Durkin ME
- Subjects
- DNA, Complementary genetics, Gene Deletion, Humans, Chromosomes, Human, Pair 13, GTPase-Activating Proteins genetics, Liver Neoplasms genetics
- Published
- 2004
- Full Text
- View/download PDF
50. DLC-1 operates as a tumor suppressor gene in human non-small cell lung carcinomas.
- Author
-
Yuan BZ, Jefferson AM, Baldwin KT, Thorgeirsson SS, Popescu NC, and Reynolds SH
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung metabolism, DNA Methylation, GTPase-Activating Proteins, Humans, Immunoblotting, Mice, Mice, Nude, Promoter Regions, Genetic, Tumor Cells, Cultured transplantation, Tumor Suppressor Proteins metabolism, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms, Tumor Suppressor Proteins genetics
- Abstract
The deleted in liver cancer (DLC-1) gene at chromosome 8p21-22 is altered mainly by genomic deletion or aberrant promoter methylation in a large number of human cancers such as breast, liver, colon and prostate and is known to have an inhibitory effect on breast and liver tumor cell growth. Given the high frequency of deletion involving region 8p21-22 in human non-small cell lung carcinoma (NSCLC), we examined alterations of DLC-1 in a series of primary tumors and tumor cell lines and tested effects of DLC-1 on tumor cell growth. A significant decrease or absence of the DLC-1 mRNA expression was found in 95% of primary NSCLC (20/21) and 58% of NSCLC cell lines (11/19). Transcriptional silencing of DLC-1 was primarily associated with aberrant DNA methylation, rather than genomic deletion as 5-aza-2'-deoxycytidine induced reactivation of DLC-1 expression in 82% (9/11) NSCLC cell lines showing downregulated DLC-1. It was further evidenced by an aberrant DLC-1 promoter methylation pattern, which was detected by Southern blotting in 73% (8/11) of NSCLC cell lines with downregulation of the gene. The transfer of DLC-1 into three DLC-1 negative cell lines caused a significant inhibition in cell proliferation and/or a decrease in colony formation. Furthermore, stable transfer of DLC-1 abolished tumorigenicity in nude mice of two cell lines, suggesting that DLC-1 plays a role in NSCLC by acting as a bona fide new tumor suppressor gene.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.