Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Genes Chromosomes Cancer ; 60(6): 434-446, 2021 06.
Article in English | MEDLINE | ID: mdl-33527590

ABSTRACT

Renal cell carcinoma (RCC) is not a single disease but is made up of several different histologically defined subtypes that are associated with distinct genetic alterations which require subtype specific management and treatment. Papillary renal cell carcinoma (pRCC) is the second most common subtype after conventional/clear cell RCC (ccRCC), representing ~20% of cases, and is subcategorized into type 1 and type 2 pRCC. It is important for preclinical studies to have cell lines that accurately represent each specific RCC subtype. This study characterizes seven cell lines derived from both primary and metastatic sites of type 1 pRCC, including the first cell line derived from a hereditary papillary renal carcinoma (HPRC)-associated tumor. Complete or partial gain of chromosome 7 was observed in all cell lines and other common gains of chromosomes 16, 17, or 20 were seen in several cell lines. Activating mutations of MET were present in three cell lines that all demonstrated increased MET phosphorylation in response to HGF and abrogation of MET phosphorylation in response to MET inhibitors. CDKN2A loss due to mutation or gene deletion, associated with poor outcomes in type 1 pRCC patients, was observed in all cell line models. Six cell lines formed tumor xenografts in athymic nude mice and thus provide in vivo models of type 1 pRCC. These type 1 pRCC cell lines provide a comprehensive representation of the genetic alterations associated with pRCC that will give insight into the biology of this disease and be ideal preclinical models for therapeutic studies.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Line Authentication/methods , Kidney Neoplasms/genetics , Xenograft Model Antitumor Assays/methods , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Chromosomal Instability , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Kidney Neoplasms/pathology , Mice , Mutation , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism
2.
Carcinogenesis ; 39(8): 993-1005, 2018 07 30.
Article in English | MEDLINE | ID: mdl-29800151

ABSTRACT

Intratumor heterogeneity is a major challenge in cancer treatment. To decipher patterns of chromosomal heterogeneity, we analyzed six colorectal cancer cell lines by multiplex interphase FISH (miFISH). The mismatch-repair-deficient cell lines DLD-1 and HCT116 had the most stable copy numbers, whereas aneuploid cell lines (HT-29, SW480, SW620 and H508) displayed a higher degree of instability. We subsequently assessed the clonal evolution of single cells in two colorectal carcinoma cell lines, SW480 and HT-29, which both have aneuploid karyotypes but different degrees of chromosomal instability. The clonal compositions of the single cell-derived daughter lines, as assessed by miFISH, differed for HT-29 and SW480. Daughters of HT-29 were stable, clonal, with little heterogeneity. Daughters of SW480 were more heterogeneous, with the single cell-derived daughter lines separating into two distinct populations with different ploidy (hyper-diploid and near-triploid), morphology, gene expression and tumorigenicity. To better understand the evolutionary trajectory for the two SW480 populations, we constructed phylogenetic trees which showed ongoing instability in the daughter lines. When analyzing the evolutionary development over time, most single cell-derived daughter lines maintained their major clonal pattern, with the exception of one daughter line that showed a switch involving a loss of APC. Our meticulous analysis of the clonal evolution and composition of these colorectal cancer models shows that all chromosomes are subject to segregation errors, however, specific net genomic imbalances are maintained. Karyotype evolution is driven by the necessity to arrive at and maintain a specific plateau of chromosomal copy numbers as the drivers of carcinogenesis.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Evolution, Molecular , Cell Line, Tumor , Chromosomal Instability , Chromosome Aberrations , Clonal Evolution , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Karyotype , Phylogeny
3.
Genes Chromosomes Cancer ; 56(3): 199-213, 2017 03.
Article in English | MEDLINE | ID: mdl-27750367

ABSTRACT

Human colorectal carcinomas are defined by a nonrandom distribution of genomic imbalances that are characteristic for this disease. Often, these imbalances affect entire chromosomes. Understanding the role of these aneuploidies for carcinogenesis is of utmost importance. Currently, established transgenic mice do not recapitulate the pathognonomic genome aberration profile of human colorectal carcinomas. We have developed a novel model based on the spontaneous transformation of murine colon epithelial cells. During this process, cells progress through stages of pre-immortalization, immortalization and, finally, transformation, and result in tumors when injected into immunocompromised mice. We analyzed our model for genome and transcriptome alterations using ArrayCGH, spectral karyotyping (SKY), and array based gene expression profiling. ArrayCGH revealed a recurrent pattern of genomic imbalances. These results were confirmed by SKY. Comparing these imbalances with orthologous maps of human chromosomes revealed a remarkable overlap. We observed focal deletions of the tumor suppressor genes Trp53 and Cdkn2a/p16. High-level focal genomic amplification included the locus harboring the oncogene Mdm2, which was confirmed by FISH in the form of double minute chromosomes. Array-based global gene expression revealed distinct differences between the sequential steps of spontaneous transformation. Gene expression changes showed significant similarities with human colorectal carcinomas. Pathways most prominently affected included genes involved in chromosomal instability and in epithelial to mesenchymal transition. Our novel mouse model therefore recapitulates the most prominent genome and transcriptome alterations in human colorectal cancer, and might serve as a valuable tool for understanding the dynamic process of tumorigenesis, and for preclinical drug testing. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , Disease Models, Animal , Genome , Transcriptome/genetics , Animals , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Spectral Karyotyping
4.
Genes Chromosomes Cancer ; 56(10): 719-729, 2017 10.
Article in English | MEDLINE | ID: mdl-28736828

ABSTRACT

Chromophobe renal cell carcinoma (ChRCC) represents 5% of all RCC cases and frequently demonstrates multiple chromosomal losses and an indolent pattern of local growth, but can demonstrate aggressive features and resistance to treatment in a metastatic setting. Cell line models are an important tool for the investigation of tumor biology and therapeutic drug efficacy. Currently, there are few ChRCC-derived cell lines and none is well characterized. This study characterizes a novel ChRCC-derived cell line model, UOK276. A large ChRCC tumor with regions of sarcomatoid differentiation was used to establish a spontaneously immortal cell line, UOK276. UOK276 was evaluated for chromosomal, mutational, and metabolic aberrations. The UOK276 cell line is hyperdiploid with a modal number of 49 chromosomes per cell, and evidence of copy-neutral loss of heterozygosity, as opposed to the classic pattern of ChRCC chromosomal losses. UOK276 demonstrated a TP53 missense mutation, expressed mutant TP53 protein, and responded to treatment with a small-molecule therapeutic agent, NSC319726, designed to reactivate mutated TP53. Xenograft tumors grew in nude mice and provide an in vivo animal model for the investigation of potential therapeutic regimes. The xenograft pathology and genetic analysis suggested that UOK276 was derived from the sarcomatoid region of the original tumor. In summary, UOK276 represents a novel in vitro and in vivo cell line model for aggressive, sarcomatoid-differentiated, TP53 mutant ChRCC. This preclinical model system could be used to investigate the novel biology of aggressive, sarcomatoid ChRCC and evaluate the new therapeutic regimes.


Subject(s)
Carcinoma, Renal Cell/genetics , Karyotype , Kidney Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Mutation, Missense , Tumor Suppressor Protein p53/genetics
5.
Genome Res ; 24(2): 185-99, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24201445

ABSTRACT

Genomic instability is a hallmark of human cancers, including the 5% caused by human papillomavirus (HPV). Here we report a striking association between HPV integration and adjacent host genomic structural variation in human cancer cell lines and primary tumors. Whole-genome sequencing revealed HPV integrants flanking and bridging extensive host genomic amplifications and rearrangements, including deletions, inversions, and chromosomal translocations. We present a model of "looping" by which HPV integrant-mediated DNA replication and recombination may result in viral-host DNA concatemers, frequently disrupting genes involved in oncogenesis and amplifying HPV oncogenes E6 and E7. Our high-resolution results shed new light on a catastrophic process, distinct from chromothripsis and other mutational processes, by which HPV directly promotes genomic instability.


Subject(s)
DNA Replication/genetics , Genomic Instability , Human papillomavirus 16/genetics , Neoplasms/genetics , DNA, Viral/genetics , Female , Human papillomavirus 16/growth & development , Humans , Male , Neoplasms/classification , Neoplasms/pathology , Neoplasms/virology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Virus Integration/genetics
6.
Blood ; 126(20): 2291-301, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26400962

ABSTRACT

The serine-threonine kinase ataxia-telangiectasia mutated (ATM) plays a central role in maintaining genomic integrity. In mice, ATM deficiency is exclusively associated with T-cell lymphoma development, whereas B-cell tumors predominate in human ataxia-telangiectasia patients. We demonstrate in this study that when T cells are removed as targets for lymphomagenesis and as mediators of immune surveillance, ATM-deficient mice exclusively develop early-onset immunoglobulin M(+) B-cell lymphomas that do not transplant to immunocompetent mice and that histologically and genetically resemble the activated B cell-like (ABC) subset of human diffuse large B-cell lymphoma (DLBCL). These B-cell lymphomas show considerable chromosomal instability and a recurrent genomic amplification of a 4.48-Mb region on chromosome 18 that contains Malt1 and is orthologous to a region similarly amplified in human ABC DLBCL. Of importance, amplification of Malt1 in these lymphomas correlates with their dependence on nuclear factor (NF)-κB, MALT1, and B-cell receptor (BCR) signaling for survival, paralleling human ABC DLBCL. Further, like some human ABC DLBCLs, these mouse B-cell lymphomas also exhibit constitutive BCR-dependent NF-κB activation. This study reveals that ATM protects against development of B-cell lymphomas that model human ABC DLBCL and identifies a potential role for T cells in preventing the emergence of these tumors.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/deficiency , Immunologic Surveillance , Lymphoma, Large B-Cell, Diffuse/immunology , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins/immunology , Caspases/genetics , Caspases/immunology , Cell Line, Tumor , Chromosomal Instability/immunology , Genetic Loci/immunology , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/genetics , NF-kappa B/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Proteins/immunology
7.
Nature ; 452(7186): 492-6, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18337721

ABSTRACT

The Sir2 deacetylase regulates chromatin silencing and lifespan in Saccharomyces cerevisiae. In mice, deficiency for the Sir2 family member SIRT6 leads to a shortened lifespan and a premature ageing-like phenotype. However, the molecular mechanisms of SIRT6 function are unclear. SIRT6 is a chromatin-associated protein, but no enzymatic activity of SIRT6 at chromatin has yet been detected, and the identity of physiological SIRT6 substrates is unknown. Here we show that the human SIRT6 protein is an NAD+-dependent, histone H3 lysine 9 (H3K9) deacetylase that modulates telomeric chromatin. SIRT6 associates specifically with telomeres, and SIRT6 depletion leads to telomere dysfunction with end-to-end chromosomal fusions and premature cellular senescence. Moreover, SIRT6-depleted cells exhibit abnormal telomere structures that resemble defects observed in Werner syndrome, a premature ageing disorder. At telomeric chromatin, SIRT6 deacetylates H3K9 and is required for the stable association of WRN, the factor that is mutated in Werner syndrome. We propose that SIRT6 contributes to the propagation of a specialized chromatin state at mammalian telomeres, which in turn is required for proper telomere metabolism and function. Our findings constitute the first identification of a physiological enzymatic activity of SIRT6, and link chromatin regulation by SIRT6 to telomere maintenance and a human premature ageing syndrome.


Subject(s)
Chromatin/metabolism , Histone Deacetylases/metabolism , Sirtuins/metabolism , Telomere/metabolism , Acetylation , Cell Line , Cellular Senescence/genetics , Chromatin/genetics , DNA Replication , Exodeoxyribonucleases/metabolism , Fibroblasts , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Histones/chemistry , Histones/metabolism , Humans , Lysine/metabolism , Phenotype , Protein Binding , RecQ Helicases/metabolism , Sirtuins/deficiency , Sirtuins/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Werner Syndrome/genetics , Werner Syndrome Helicase
8.
Carcinogenesis ; 34(8): 1929-39, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23619298

ABSTRACT

Human epithelial cancers are defined by a recurrent distribution of specific chromosomal aneuploidies, a trait less typical for murine cancer models induced by an oncogenic stimulus. After prolonged culture, mouse epithelial cells spontaneously immortalize, transform and become tumorigenic. We assessed genome and transcriptome alterations in cultures derived from bladder and kidney utilizing spectral karyotyping, array CGH, FISH and gene expression profiling. The results show widespread aneuploidy, yet a recurrent and tissue-specific distribution of genomic imbalances, just as in human cancers. Losses of chromosome 4 and gains of chromosome 15 are common and occur early during the transformation process. Global gene expression profiling revealed early and significant transcriptional deregulation. Chromosomal aneuploidy resulted in expression changes of resident genes and consequently in a massive deregulation of the cellular transcriptome. Pathway interrogation of expression changes during the sequential steps of transformation revealed enrichment of genes associated with DNA repair, centrosome regulation, stem cell characteristics and aneuploidy. Genes that modulate the epithelial to mesenchymal transition and genes that define the chromosomal instability phenotype played a dominant role and were changed in a directionality consistent with loss of cell adhesion, invasiveness and proliferation. Comparison with gene expression changes during human bladder and kidney tumorigenesis revealed remarkable overlap with changes observed in the spontaneously transformed murine cultures. Therefore, our novel mouse models faithfully recapitulate the sequence of genomic and transcriptomic events that define human tumorigenesis, hence validating them for both basic and preclinical research.


Subject(s)
Carcinogenesis/genetics , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/genetics , Gene Amplification , Oncogenes , Aneuploidy , Animals , Carcinogenesis/metabolism , Chromosomal Instability , Chromosome Aberrations , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Spectral Karyotyping/methods , Transcription, Genetic , Transcriptome , Urinary Bladder/cytology
9.
Genes Chromosomes Cancer ; 51(4): 353-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22161874

ABSTRACT

Human carcinomas are defined by recurrent chromosomal aneuploidies, which result in a tissue-specific distribution of genomic imbalances. In order to develop models for these genome mutations and to determine their role in tumorigenesis, we generated 45 spontaneously transformed murine cell lines from normal epithelial cells derived from bladder, cervix, colon, kidney, lung, and mammary gland. Phenotypic changes, chromosomal aberrations, centrosome number, and telomerase activity were assayed in control uncultured cells and in three subsequent stages of transformation. Supernumerary centrosomes, binucleate cells, and tetraploidy were observed as early as 48 hr after explantation. In addition, telomerase activity increased throughout progression. Live-cell imaging revealed that failure of cytokinesis, not cell fusion, promoted genome duplication. Spectral karyotyping demonstrated that aneuploidy preceded immortalization, consisting predominantly of whole chromosome losses (4, 9, 12, 13, 16, and Y) and gains (1, 10, 15, and 19). After transformation, focal amplifications of the oncogenes Myc and Mdm2 were frequently detected. Fifty percent of the transformed lines resulted in tumors on injection into immunocompromised mice. The phenotypic and genomic alterations observed in spontaneously transformed murine epithelial cells recapitulated the aberration pattern observed during human carcinogenesis. The dominant aberration of these cell lines was the presence of specific chromosomal aneuploidies. We propose that our newly derived cancer models will be useful tools to dissect the sequential steps of genome mutations during malignant transformation, and also to identify cancer-specific genes, signaling pathways, and the role of chromosomal instability in this process.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Epithelial Cells/pathology , Animals , Cell Line, Transformed , Epithelial Cells/metabolism , Female , Genes, myc , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Phenotype , Proto-Oncogene Proteins c-mdm2/genetics
10.
Transfusion ; 52(3): 537-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21883270

ABSTRACT

BACKGROUND: Reports of Monosomy 7 in patients receiving granulocyte-colony-stimulating factor (G-CSF) have raised concerns that this cytokine may promote genomic instability. However, there are no studies addressing whether repeated administration of G-CSF produces Monosomy 7 aneuploidy in healthy donors. STUDY DESIGN AND METHODS: We examined Chromosomes 7 and 8 by fluorescent in situ hybridization (FISH) in CD34+ cells from 35 healthy hematopoietic stem cell transplant (HSCT) donors after G-CSF administration for 5 days and by spectral karyotyping analysis (SKY) in four individuals to assess chromosomal integrity. We also studied 38 granulocyte donors who received up to 42 doses of G-CSF and dexamethasone (Dex) using FISH for Chromosomes 7 and 8. RESULTS: We found no abnormalities in Chromosomes 7 and 8 in G-CSF-mobilized CD34+ cells when assessed by FISH or SKY, nor did we detect aneuploidy in G-CSF- and Dex-treated donors. CONCLUSION: G-CSF does not promote clinically detectable Monosomy 7 or Trisomy 8 aneuploidy in HSCT or granulocyte donors. These findings should be reassuring to healthy HSCT and granulocyte donors.


Subject(s)
Genomic Instability/drug effects , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocytes/drug effects , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Aneuploidy , Chromosome Deletion , Chromosomes, Human, Pair 7/drug effects , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Granulocytes/physiology , Granulocytes/transplantation , Hematopoietic Stem Cell Mobilization/adverse effects , Hematopoietic Stem Cell Mobilization/standards , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Humans , Tissue Donors
11.
Genes Chromosomes Cancer ; 49(3): 204-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19927377

ABSTRACT

In defining the genetic profiles in cancer, cytogenetically aberrant cell lines derived from primary tumors are important tools for the study of carcinogenesis. Here, we present the results of a comprehensive investigation of 15 established colorectal cancer cell lines using spectral karyotyping (SKY), fluorescence in situ hybridization, and comparative genomic hybridization (CGH). Detailed karyotypic analysis by SKY on five of the lines (P53HCT116, T84, NCI-H508, NCI-H716, and SK-CO-1) is described here for the first time. The five lines with karyotypes in the diploid range and that are characterized by defects in DNA mismatch repair had a mean of 4.8 chromosomal abnormalities per line, whereas the 10 aneuploid lines exhibited complex karyotypes and a mean of 30 chromosomal abnormalities. Of the 150 clonal translocations, only eight were balanced and none were recurrent among the lines. We also reviewed the karyotypes of 345 cases of adenocarcinoma of the large intestine listed in the Mitelman Database of Chromosome Aberrations in Cancer. The types of abnormalities observed in the cell lines reflected those seen in primary tumors: there were no recurrent translocations in either tumors or cell lines; isochromosomes were the most common recurrent abnormalities; and breakpoints occurred most frequently at the centromeric/pericentromeric and telomere regions. Of the genomic imbalances detected by array CGH, 87% correlated with chromosome aberrations observed in the SKY studies. The fact that chromosome abnormalities predominantly result in copy number changes rather than specific chromosome or gene fusions suggests that this may be the major mechanism leading to carcinogenesis in colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Cytogenetic Analysis/methods , Cell Line, Tumor , Chromosome Aberrations/statistics & numerical data , Chromosome Banding , Comparative Genomic Hybridization/methods , DNA Mismatch Repair , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Translocation, Genetic
12.
Curr Biol ; 17(5): 431-7, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17320392

ABSTRACT

Chromosomal instability (CIN) underlies malignant properties of many solid cancers and their ability to escape therapy, and it might itself cause cancer [1, 2]. CIN is sustained by deficiencies in proteins, such as the tumor suppressor p53 [3-5], that police genome integrity, but the primary cause of CIN in sporadic cancers remains uncertain [6, 7]. The primary suspects are mutations that deregulate telomere maintenance, or mitosis, yet such mutations have not been identified in the majority of sporadic cancers [6]. Alternatively, CIN could be caused by a transient event that destabilizes the genome without permanently affecting mechanisms of mitosis or proliferation [5, 8]. Here, we show that an otherwise harmless virus rapidly causes massive chromosomal instability by fusing cells whose cell cycle is deregulated by oncogenes. This synergy between fusion and oncogenes "randomizes" normal diploid human fibroblasts so extensively that each analyzed cell has a unique karyotype, and some produce aggressive, highly aneuploid, heterogeneous, and transplantable epithelial cancers in mice. Because many viruses are fusogenic, this study suggests that viruses, including those that have not been linked to carcinogenesis, can cause chromosomal instability and, consequently, cancer by fusing cells.


Subject(s)
Carcinoma/virology , Cell Fusion , Chromosomal Instability , Fibroblasts/virology , Mason-Pfizer monkey virus/pathogenicity , Neoplasms/virology , Animals , Carcinoma/physiopathology , Cell Transformation, Neoplastic , Cell Transformation, Viral , Female , Fibroblasts/metabolism , Humans , Mice , Mice, Nude , Neoplasms/physiopathology , Oncogenes/genetics , Transduction, Genetic
13.
Genes Chromosomes Cancer ; 48(11): 1002-17, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19691111

ABSTRACT

To evaluate the mechanisms and consequences of chromosomal aberrations in colorectal cancer (CRC), we used a combination of spectral karyotyping, array comparative genomic hybridization (aCGH), and array-based global gene expression profiling on 31 primary carcinomas and 15 established cell lines. Importantly, aCGH showed that the genomic profiles of primary tumors are recapitulated in the cell lines. We revealed a preponderance of chromosome breakpoints at sites of copy number variants (CNVs) in the CRC cell lines, a novel mechanism of DNA breakage in cancer. The integration of gene expression and aCGH led to the identification of 157 genes localized within high-level copy number changes whose transcriptional deregulation was significantly affected across all of the samples, thereby suggesting that these genes play a functional role in CRC. Genomic amplification at 8q24 was the most recurrent event and led to the overexpression of MYC and FAM84B. Copy number dependent gene expression resulted in deregulation of known cancer genes such as APC, FGFR2, and ERBB2. The identification of only 36 genes whose localization near a breakpoint could account for their observed deregulated expression demonstrates that the major mechanism for transcriptional deregulation in CRC is genomic copy number changes resulting from chromosomal aberrations.


Subject(s)
Colorectal Neoplasms/genetics , DNA Copy Number Variations , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Chromosome Breakpoints , Chromosomes, Human, Pair 8 , Colorectal Neoplasms/metabolism , Comparative Genomic Hybridization/methods , Gene Deletion , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Spectral Karyotyping/methods , Transcription, Genetic
14.
Cancer Res ; 67(19): 9077-83, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909011

ABSTRACT

Cisplatin, an anticancer drug, forms DNA interstrand cross-links (ICL) that interfere with replication, whereas TREX2 is a 3'-->5' exonuclease that removes 3' mismatched nucleotides and promotes cellular proliferation. Here, we show that TREX2 is depleted in human cells derived from cancer after exposure to cisplatin but not other genotoxins including another cross-linking agent, mitomycin C (MMC), indicating a potential role for TREX2 depletion in cisplatin-induced cytotoxicity. To better understand TREX2 cellular function, we deleted TREX2 in mouse embryonic stem (ES) cells by gene targeting and find these cells exhibit reduced proliferation and gross chromosomal rearrangements including Robertsonian translocations (RbT). Quite interestingly, ES cells exposed to cisplatin also exhibit RbTs. By contrast, RbTs are not observed for ES cells exposed to MMC, indicating that RbTs are not caused by ICLs but instead TREX2 depletion by either cisplatin exposure or mutation. Taken together, our results show that cisplatin depletes TREX2 and causes genomic instability that is similarly observed in TREX2-mutant cells. Thus, cisplatin has two potential cytotoxic activities: (a) the generation of ICLs and (b) the depletion of TREX2.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA Damage , Exodeoxyribonucleases/deficiency , Phosphoproteins/deficiency , Translocation, Genetic/drug effects , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Dose-Response Relationship, Drug , Exodeoxyribonucleases/metabolism , HeLa Cells , Humans , Mitomycin/pharmacology , Phosphoproteins/metabolism
15.
Cancer Genet Cytogenet ; 180(2): 100-9, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18206534

ABSTRACT

The establishment, characterization, and tumorigenicity of a new epithelial cell line (UOK 257) derived from human renal carcinoma of an individual with Birt-Hogg-Dubé (BHD) syndrome are reported. Unlike other established renal tumor cell lines from sporadic renal cell carcinoma, this is the first established renal tumor cell line of BHD, an inheritable neoplastic syndrome. The isolated tumor cells display loss of contact inhibition in vitro, and produce subcutaneous tumors in mouse xenografts. Histopathologic, ultrastructural, and cytogenetic characterizations of the established tumor cells are reported. Cytogenetic analysis using spectral karyotyping on UOK 257 cells revealed 17p loss and a near-triploid and aneuploid karyotype with multiple fluorescence in situ hybridization analysis using a locus-specific gene probe for MYC. The result demonstrates that the established tumor cells consist of two cell populations, one containing four and one containing five copies of the MYC oncogene.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Models, Biological , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Carcinoma, Renal Cell/pathology , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Chromosome Aberrations , Chromosomes, Human, Pair 17 , Gene Duplication , Genes, myc , Humans , Karyotyping , Kidney Neoplasms/pathology , Male , Middle Aged , Ploidies , Proto-Oncogene Proteins/physiology , Sequence Analysis, DNA , Signal Transduction/genetics , Syndrome , Tumor Suppressor Proteins/physiology
16.
J Hered ; 99(3): 241-53, 2008.
Article in English | MEDLINE | ID: mdl-18339652

ABSTRACT

Chromosome painting was used to investigate the conservation of high-resolution longitudinal 4',6-diamidino-2-phenylindole (DAPI)/G bands in Carnivore chromosomes. Cat (Felis catus) and raccoon dog (Nyctereutes procyonoides) painting probes were hybridized to the ringtail (Bassaricus astutus), dwarf mongoose (Helogale parvula), and Malagasy civet (Fossa fossa) to identify homologous chromosome elements. The patterns of chromosome segment homology among Carnivore species allowed us to reconstruct and propose the disposition of a high-resolution banded ancestral carnivore karyotype (ACK). Three bi-armed chromosomes consistently found among Caniformia species are represented as 6 homologous acrocentric chromosomes among Feliformia species of Carnivora. However, reexamination of the most basal of Feliformia species, the African palm civet Nandinia, revealed the presence of the 3 heretofore Caniformia bi-armed chromosomes. Because these 3 bi-armed chromosomes are found in both Caniformia and Feliformia lineages, they are presumed ancestral for all Carnivora, suggesting that the ACK chromosome number would be 38, rather than the previously supposed 42. Banded chromosomes of the ACK are used to evaluate the consistency between recently determined molecular phylogenetic relationships and postulated cytogenetic dynamics in the same Carnivore species.


Subject(s)
Carnivora/genetics , Chromosomes, Mammalian , Phylogeny , Procyonidae/genetics , Animals , Cats , Chromosome Banding , Chromosomes, Mammalian/classification , Eupleridae/genetics , Herpestidae/genetics , Raccoon Dogs/genetics , Ursidae/genetics
17.
Cancer Res ; 65(20): 9388-97, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230402

ABSTRACT

The origin of drug-resistant cells in human cancers has been a fundamental problem of cancer pharmacology. Two major contrasting hypotheses (genetics versus epigenetics) have been proposed to elucidate the mechanisms of acquired drug resistance. In this study, we answer these fundamental questions through investigation of the genetic and epigenetic pathways that control the origin of ABCB1 (MDR1) gene activation with acquired multidrug resistance in drug-sensitive human sarcoma (MES-SA cells). The genetic and epigenetic bases of this selected activation involve the initiation of transcription at a site 112 kb upstream of the ABCB1 proximal promoter (P1) in the drug-resistant cells. This activation was associated with a chromatin-remodeling process characterized by an increase in acetylated histone H3 within a 968-bp region 5' of the ABCB1 upstream promoter. These alterations provide both genetic and epigenetic susceptibility for ABCB1 expression in drug-resistant cells. Complete activation of the ABCB1 gene through the coding region was proposed by interactions of selected trans-alterations or epigenetic changes on the ABCB1 proximal promoter, which occurred during initial drug exposure. Thus, our data provide evidence for a major genomic alteration that changes the chromatin structure of the ABCB1 upstream promoter via acetylation of histone H3 initiating ABCB1 activation, further elucidating the genetic and epigenetic bases that determine chemotherapeutic response in drug-resistant derivatives of MES-SA cells.


Subject(s)
Drug Resistance, Multiple/genetics , Genes, MDR/genetics , Sarcoma/drug therapy , Sarcoma/genetics , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Acetylation , Base Sequence , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Chromatin/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Expression Regulation , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma/metabolism , Transcriptional Activation , Uterine Neoplasms/metabolism
18.
Cancer Res ; 77(23): 6729-6745, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28951459

ABSTRACT

Epigenetic regulation of chromatin states is thought to control gene expression programs during lineage specification. However, the roles of repressive histone modifications, such as trimethylated histone lysine 20 (H4K20me3), in development and genome stability are largely unknown. Here, we show that depletion of SET and MYND domain-containing protein 5 (SMYD5), which mediates H4K20me3, leads to genome-wide decreases in H4K20me3 and H3K9me3 levels and derepression of endogenous LTR- and LINE-repetitive DNA elements during differentiation of mouse embryonic stem cells. SMYD5 depletion resulted in chromosomal aberrations and the formation of transformed cells that exhibited decreased H4K20me3 and H3K9me3 levels and an expression signature consistent with multiple human cancers. Moreover, dysregulated gene expression in SMYD5 cancer cells was associated with LTR and endogenous retrovirus elements and decreased H4K20me3. In addition, depletion of SMYD5 in human colon and lung cancer cells results in increased tumor growth and upregulation of genes overexpressed in colon and lung cancers, respectively. These findings implicate an important role for SMYD5 in maintaining chromosome integrity by regulating heterochromatin and repressing endogenous repetitive DNA elements during differentiation. Cancer Res; 77(23); 6729-45. ©2017 AACR.


Subject(s)
Cell Differentiation/genetics , Chromosomes/physiology , Colonic Neoplasms/genetics , Embryonic Stem Cells/cytology , Heterochromatin/physiology , Lung Neoplasms/genetics , Methyltransferases/genetics , A549 Cells , Animals , Base Sequence , Cell Line, Tumor , Colonic Neoplasms/pathology , DNA Copy Number Variations/genetics , Epigenesis, Genetic , HCT116 Cells , Histone Code/genetics , Histones/metabolism , Humans , Lung Neoplasms/pathology , MCF-7 Cells , Methylation , Mice , RNA Interference , RNA, Small Interfering/genetics , Sequence Analysis, DNA , Xenograft Model Antitumor Assays
19.
Cancer Genet Cytogenet ; 170(1): 1-8, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16965948

ABSTRACT

Ovarian cancer is one of the most common types of malignancy in women throughout the developed world. Despite recent therapeutic advances, long-term survival is poor because ovarian cancer is largely asymptomatic in its early stages. Comparative genomic hybridization (CGH) was applied to a series of 8 benign, 8 borderline, and 17 malignant ovarian to establish genomic imbalances associated with tumor progression. Benign and borderline tumors were characterized by losses at 1p32 approximately p11, 2q14 approximately q34, 4q13 approximately q34, 5q11 approximately q23, and 6q12 approximately q24, as well as gains of 6p and chromosome 12. Similar chromosomal changes were also detected in malignant tumors but included additional chromosomal changes: gains at 1q21 approximately q31, 2p, 3q, 5p, 7, 10p, 12p, 16p, 17, 19q, 20q, and 22q, as well as losses at X, 3p, 8p, 9, 11p, 13, 14, and 18. Some individual cases of benign and borderline tumors revealed no genetic alterations detectable by CGH, suggesting that these tumors may represent a subset of tumors that originate by an alternative mechanism of tumorigenesis. Furthermore, our findings reveal that borderline tumors are more similar to benign tumors than to malignant tumors with respect to their genetic profiles.


Subject(s)
Nucleic Acid Hybridization , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Chromosome Aberrations , Female , Genome, Human , Humans
20.
Oncogene ; 21(6): 890-8, 2002 Jan 31.
Article in English | MEDLINE | ID: mdl-11840334

ABSTRACT

The conditional expression of activated HER2/neu gene under its endogenous promoter in the mammary epithelium of the mouse results in accelerated lobular development and focal mammary tumors. Carcinogenesis, however, requires amplification and considerably increased expression levels of oncogenic neu. Deducing from the multiple genetic aberrations required for human breast cancer to develop, we hypothesized that in addition to the over-expression of an activated HER2/neu, secondary aberrations would occur. We have therefore conducted a genomic screen for chromosomal imbalances and translocations using comparative genomic hybridization and spectral karyotyping. The results reveal a moderate degree of chromosomal instability and micronuclei formation in short-term cultures established from primary tumors. Genomic instability appears to be linked to the amplification of functional centrosomes, a phenomenon that we frequently observed in other tumor types. Seventy per cent of the tumors revealed genomic amplification of HER2/neu, often in the form of double minute chromosomes, which correlated with recurring loss of mouse chromosome 4D-E, a region that is orthologous to distal human chromosome 1p. It is likely that this region contains putative tumor suppressor genes whose inactivation is required for tumor formation in this model of human breast cancer.


Subject(s)
Adenocarcinoma/genetics , Cell Transformation, Neoplastic/genetics , Chromosome Aberrations , Chromosome Deletion , Gene Amplification , Genes, Tumor Suppressor , Genes, erbB-2 , Mammary Neoplasms, Experimental/genetics , Animals , Centrosome/ultrastructure , Chromosome Mapping , Female , Flow Cytometry , Gene Silencing , In Situ Hybridization, Fluorescence , Karyotyping/methods , Mice , Models, Animal , Nucleic Acid Hybridization , Receptor, ErbB-2/physiology
SELECTION OF CITATIONS
SEARCH DETAIL