Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Hum Genet ; 130(5): 623-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21442329

ABSTRACT

Human trisomy 21, the chromosomal basis of Down syndrome (DS), is the most common genetic cause of heart defects. Regions on human chromosome 21 (Hsa21) are syntenically conserved with three regions located on mouse chromosome 10 (Mmu10), Mmu16 and Mmu17. In this study, we have analyzed the impact of duplications of each syntenic region on cardiovascular development in mice and have found that only the duplication on Mmu16, i.e., Dp(16)1Yey, is associated with heart defects. Furthermore, we generated two novel mouse models carrying a 5.43-Mb duplication and a reciprocal deletion between Tiam1 and Kcnj6 using chromosome engineering, Dp(16Tiam1-Kcnj6)Yey/+ and Df(16Tiam1-Kcnj6)Yey/+, respectively, within the 22.9-Mb syntenic region on Mmu16. We found that Dp(16Tiam1-Kcnj6)Yey/+, but not Dp(16)1Yey/Df(16Tiam1-Kcnj6)Yey, resulted in heart defects, indicating that triplication of the Tiam1-Knj6 region is necessary and sufficient to cause DS-associated heart defects. Our transcriptional analysis of Dp(16Tiam1-Kcnj6)Yey/+ embryos confirmed elevated expression levels for the genes located in the Tiam-Kcnj6 region. Therefore, we established the smallest critical genomic region for DS-associated heart defects to lay the foundation for identifying the causative gene(s) for this phenotype.


Subject(s)
Down Syndrome/genetics , Heart Defects, Congenital/genetics , Animals , Disease Models, Animal , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Gene Duplication/genetics , Guanine Nucleotide Exchange Factors/genetics , Male , Mice , Mice, Mutant Strains , Sequence Deletion/genetics , Synteny/genetics , T-Lymphoma Invasion and Metastasis-inducing Protein 1
2.
Kidney Int ; 79(1): 46-56, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20861821

ABSTRACT

Pyrrole-imidazole (PI) polyamides are small synthetic molecules that recognize and attach to the minor groove of DNA, thereby inhibiting gene transcription by blocking transcription factor binding. These derivatives can act as gene silencers inhibiting target gene expression under stimulatory conditions such as disease. To evaluate PI polyamides as treatments for the progression of renal diseases, we examined morphological effects, pharmacological properties, and the specificity of PI polyamides targeted to the transforming growth factor (TGF)-ß1 promoter during salt-induced hypertensive nephrosclerosis in Dahl salt-sensitive rats. The targeted PI polyamide markedly reduced glomerulosclerosis and interstitial fibrosis without side effects. PI polyamide significantly decreased expression of TGF-ß1 and extracellular matrix in the renal cortex. Microarray analysis found that only 3% of the transcripts were affected by PI polyamide, but this included decreased expression of extracellular matrix, TGF-ß1-related cytokines, angiogenic, and cell stabilizing factors, proteinases, and renal injury-related factors. Thus, targeted PI polyamides are potential gene silencers for diseases not treatable by current remedies.


Subject(s)
Amino Acids/pharmacology , Gene Expression Regulation , Gene Silencing , Imidazoles/pharmacology , Kidney Glomerulus/pathology , Nylons/pharmacology , Pyrroles/pharmacology , Transcription, Genetic/drug effects , Transforming Growth Factor beta1/genetics , Animals , Fibrosis/genetics , Fibrosis/pathology , Fibrosis/prevention & control , Genetic Therapy/methods , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/prevention & control , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Male , Promoter Regions, Genetic/drug effects , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl
3.
Cell Cycle ; 9(23): 4656-65, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21099353

ABSTRACT

A subset of AKAPs (A Kinase Anchoring Proteins) regulate signaling and cytoskeletal pathways through the spaciotemporal scaffolding of multiple protein kinases (PK) such as PKC and PKA, and associations with the plasma membrane and the actin-based cytoskeleton. SSeCKS/Gravin/Akap12 expression is severely downregulated in many advanced cancers and exhibits tumor- and metastasis-suppressing activity. akap12-null (KO) mice develop prostatic hyperplasia with focal dysplasia, but the precise mechanism how Akap12 prevents oncogenic progression remains unclear. Here, we show that KO mouse embryonic fibroblasts (MEF) exhibit premature senescence marked by polyploidy and multinucleation, and by increased susceptibility to oncogenic transformation. Although p53 and Rb pathways are activated in the absence of Akap12, senescence is dependent on Rb. Senescence is driven by the activation of PKCα, which induces p16(Ink4a)/Rb through a MEK-dependent downregulation of Id1, and PKCδ, which downregulates Lats1/Warts, a mitotic exit network kinase required for cytokinesis. Our data strongly suggest that Akap12 controls Rb-mediated cell aging and oncogenic progression by directly scaffolding and attenuating PKCα/δ.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic , Cellular Senescence , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Retinoblastoma Protein/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation , Fibroblasts/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Mice , Mice, Knockout , Polyploidy , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism
4.
Mamm Genome ; 21(5-6): 258-67, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20512340

ABSTRACT

Copy-number variation in the human genome can be disease-causing or phenotypically neutral. This type of genetic rearrangement associated with human chromosome 21 (Hsa21) underlies partial Monosomy 21 and Trisomy 21. Mental retardation is a major clinical manifestation of partial Monosomy 21. To model this human chromosomal deletion disorder, we have generated novel mouse mutants carrying heterozygous deletions of the 2.3- and 1.1-Mb segments on mouse chromosome 10 (Mmu10) and Mmu17, respectively, which are orthologous to the regions on human 21q22.3, using Cre/loxP-mediated chromosome engineering. Alterations of the transcriptional levels of genes within the deleted intervals reflect gene-dosage effects in the mutant mice. The analysis of cognitive behaviors shows that the mutant mice carrying the deletion on either Mmu10 or Mmu17 are impaired in learning and memory. Therefore, these mutants represent mouse models for Monosomy 21-associated mental retardation, which can serve as a powerful tool to study the molecular mechanism underlying the clinical phenotype and should facilitate efforts to identify the haploinsufficient causative genes.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Cognition Disorders/genetics , Cognition , Animals , Chromosome Deletion , Disease Models, Animal , Down Syndrome , Gene Dosage , Humans , Learning , Memory , Mice , Phenotype , Synteny
5.
Hum Mol Genet ; 19(14): 2780-91, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20442137

ABSTRACT

Down syndrome (DS) is caused by the presence of an extra copy of human chromosome 21 (Hsa21) and is the most common genetic cause for developmental cognitive disability. The regions on Hsa21 are syntenically conserved with three regions located on mouse chromosome 10 (Mmu10), Mmu16 and Mmu17. In this report, we describe a new mouse model for DS that carries duplications spanning the entire Hsa21 syntenic regions on all three mouse chromosomes. This mouse mutant exhibits DS-related neurological defects, including impaired cognitive behaviors, reduced hippocampal long-term potentiation and hydrocephalus. These results suggest that when all the mouse orthologs of the Hsa21 genes are triplicated, an abnormal cognitively relevant phenotype is the final outcome of the elevated expressions of these orthologs as well as all the possible functional interactions among themselves and/or with other mouse genes. Because of its desirable genotype and phenotype, this mutant may have the potential to serve as one of the reference models for further understanding the developmental cognitive disability associated with DS and may also be used for developing novel therapeutic interventions for this clinical manifestation of the disorder.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/pathology , Mice, Transgenic , Animals , Cells, Cultured , Down Syndrome/physiopathology , Female , Hand Strength/physiology , Hippocampus/physiopathology , Humans , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Models, Biological , Physical Conditioning, Animal , Pregnancy , Synteny/genetics
6.
BMC Biol ; 8: 6, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-20092614

ABSTRACT

BACKGROUND: Folate (vitamin B9) is essential for cellular proliferation as it is involved in the biosynthesis of deoxythymidine monophosphate (dTMP) and s-adenosylmethionine (AdoMet). The link between folate depletion and the genesis and progression of cancers of epithelial origin is of high clinical relevance, but still unclear. We recently demonstrated that sensitivity to low folate availability is affected by the rate of polyamine biosynthesis, which is prominent in prostate cells. We, therefore, hypothesized that prostate cells might be highly susceptible to genetic, epigenetic and phenotypic changes consequent to folate restriction. RESULTS: We studied the consequences of long-term, mild folate depletion in a model comprised of three syngenic cell lines derived from the transgenic adenoma of the mouse prostate (TRAMP) model, recapitulating different stages of prostate cancer; benign, transformed and metastatic. High-performance liquid chromatography analysis demonstrated that mild folate depletion (100 nM) sufficed to induce imbalance in both the nucleotide and AdoMet pools in all prostate cell lines. Random oligonucleotide-primed synthesis (ROPS) revealed a significant increase in uracil misincorporation and DNA single strand breaks, while spectral karyotype analysis (SKY) identified five novel chromosomal rearrangements in cells grown with mild folate depletion. Using global approaches, we identified an increase in CpG island and histone methylation upon folate depletion despite unchanged levels of total 5-methylcytosine, indicating a broad effect of folate depletion on epigenetic regulation. These genomic changes coincided with phenotype changes in the prostate cells including increased anchorage-independent growth and reduced sensitivity to folate depletion. CONCLUSIONS: This study demonstrates that prostate cells are highly susceptible to genetic and epigenetic changes consequent to mild folate depletion as compared to cells grown with supraphysiological amounts of folate (2 microM) routinely used in tissue culture. In addition, we elucidate for the first time the contribution of these aspects to consequent phenotype changes in epithelial cells. These results provide a strong rationale for studying the effects of folate manipulation on the prostate in vivo, where cells might be more sensitive to changes in folate status resulting from folate supplementation or antifolate therapeutic approaches.


Subject(s)
Epigenesis, Genetic/genetics , Folic Acid Deficiency/genetics , Folic Acid Deficiency/physiopathology , Prostatic Neoplasms/genetics , 5-Methylcytosine/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Chromatography, High Pressure Liquid , CpG Islands/genetics , DNA Damage/genetics , DNA Methylation/genetics , Male , Mice , Prostatic Neoplasms/metabolism
7.
PLoS One ; 4(11): e7784, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19907659

ABSTRACT

Transcriptional silencing of the p73 gene through methylation has been demonstrated in human leukemias and lymphomas. However, the role of p73 in the malignant process remains to be explored. We show here that p73 acts as a T cell-specific tumor suppressor in a genetically defined mouse model, and that concomitant ablation of p53 and p73 predisposes mice to an increased incidence of thymic lymphomas compared to the loss of p53 alone. Our results demonstrate a causal role for loss of p73 in progression of T cell lymphomas to the stage of aggressive, disseminated disease. We provide evidence that tumorigenesis in mice lacking p53 and p73 proceeds through mechanisms involving altered patterns of gene expression, defects in early T cell development, impaired apoptosis, and the ensuing accumulation of chromosomal aberrations. Collectively, our data imply that tumor suppressive properties of p73 are highly dependent on cellular context, wherein p73 plays a major role in T cell development and neoplasia.


Subject(s)
DNA-Binding Proteins/genetics , Gene Deletion , Lymphoma/metabolism , Nuclear Proteins/genetics , T-Lymphocytes/cytology , Tumor Suppressor Proteins/genetics , Animals , Apoptosis , Chromosome Aberrations , DNA Methylation , Gene Silencing , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Mice , Mice, Knockout , Nucleic Acid Hybridization , T-Lymphocytes/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics
8.
Cancer Res ; 69(14): 5946-53, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19584277

ABSTRACT

We used a combination of spectral karyotyping, array comparative genomic hybridization, and cDNA microarrays to gain insights into the structural and functional changes of the genome in the MCF10 human breast cancer progression model cell lines. Spectral karyotyping data showed several chromosomal aberrations and array comparative genomic hybridization analysis identified numerous genomic gains and losses that might be involved in the progression toward cancer. Analysis of the expression levels of genes located within these genomic regions revealed a lack of correlation between chromosomal gains and losses and corresponding up-regulation or down-regulation for the majority of the approximately 1,000 genes analyzed in this study. We conclude that other mechanisms of gene regulation that are not directly related to chromosomal gains and losses play a major role in breast cancer progression.


Subject(s)
Cytogenetics/methods , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Breast/cytology , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Chromosome Aberrations , Comparative Genomic Hybridization/methods , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Spectral Karyotyping/methods , Up-Regulation
9.
Chromosome Res ; 16(8): 1177-92, 2008.
Article in English | MEDLINE | ID: mdl-19005637

ABSTRACT

The organization of the type I interferon (IFN) gene cluster (9p21.3) was studied in a human osteosarcoma cell line (MG63). Array comparative genomic hybridization (aCGH) showed an amplification of approximately 6-fold which ended at both ends of the gene cluster with a deletion that extended throughout the 9p21.3 band. Spectral karyotyping (SKY) combined with fluorescence in-situ hybridization (FISH) identified an arrangement of the gene cluster in a ladder-like array of 5-7 'bands' spanning a single chromosome termed the 'IFN chromosome'. Chromosome painting revealed that the IFN chromosome is derived from components of chromosomes 4, 8 and 9. Labelling with centromeric probes demonstrated a ladder-like amplification of centromeric 4 and 9 sequences that co-localized with each other and a similar banding pattern of chromosome 4, as well as alternating with the IFN gene clusters. In contrast, centromere 8 was not detected on the IFN chromosome. One of the amplified centromeric 9 bands was identified as the functional centromere based on its location at the chromosome constriction and immunolocalization of the CENP-C protein. A model is presented for the generation of the IFN chromosome that involves breakage-fusion-bridge events.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Interferon Type I/genetics , Multigene Family/genetics , Nucleic Acid Amplification Techniques/methods , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Chromosome Painting , Comparative Genomic Hybridization , Humans , In Situ Hybridization, Fluorescence , Karyotyping
10.
Mol Cancer ; 7: 51, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18534021

ABSTRACT

BACKGROUND: Widely accepted somatic mutation theory of carcinogenesis states that mutations in oncogenes and tumor suppressor genes in genomes of somatic cells is the cause of neoplastic transformation. Identifying frequent mutations in cancer cells suggests the involvement of mutant genes in carcinogenesis. RESULTS: To develop an in vitro model for the analysis of genetic alterations associated with breast carcinogenesis, we used random mutagenesis and selection of human non-tumorigenic immortalized breast epithelial cells MCF-10A in tissue-culture conditions that mimic tumor environment. Random mutations were generated in MCF-10A cells by cultivating them in a tissue-culture medium containing the frameshift-inducing agent ICR191. The first selective condition we used to transform MCF1-10A cells was cultivation in a medium containing mutagen at a concentration that allowed cell replication despite p53 protein accumulation induced by mutagen treatment. The second step of selection was either cell cultivation in a medium with reduced growth-factor supply or in a medium that mimics a hypoxia condition or growing in soft agar. Using mutagenesis and selection, we have generated several independently derived cultures with various degrees of transformation. Gene Identification by Nonsense-mediated mRNA decay Inhibition (GINI) analysis has identified the ICR191-induced frameshift mutations in the TP53, smoothelin, Ras association (RalGDS/AF-6) domain family 6 (RASSF6) and other genes in the transformed MCF-10A cells. The TP53 gene mutations resulting in the loss of protein expression had been found in all independently transformed MCF-10A cultures, which form large progressively growing tumors with sustained angiogenesis in nude mice. CONCLUSION: Identifying genes containing bi-allelic ICR191-induced frameshift mutations in the transformed MCF-10A cells generated by random mutagenesis and selection indicates putative breast-tumor suppressors. This can provide a model for studying the role of mutant genes in breast carcinogenesis.


Subject(s)
Aminacrine/analogs & derivatives , Breast Neoplasms/genetics , Frameshift Mutation/genetics , Genes, Tumor Suppressor , Models, Biological , Mutagenesis/drug effects , Nitrogen Mustard Compounds/toxicity , Aminacrine/toxicity , Animals , Cell Line, Transformed , Cell Line, Tumor , Chromosomal Instability/drug effects , Female , Frameshift Mutation/drug effects , Humans , Mammary Neoplasms, Experimental/genetics , Mice , Neoplasm Transplantation , Nucleic Acid Hybridization , RNA Stability/drug effects , Spectral Karyotyping
11.
Cancer Biol Ther ; 7(11): 1732-43, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19151587

ABSTRACT

Human mitochondrial DNA (mtDNA) encodes 13 proteins involved in oxidative phosphorylation (OXPHOS). In order to investigate the role of mitochondrial OXPHOS genes in breast tumorigenesis, we have developed a breast epithelial cell line devoid of mtDNA (rho(0) cells). Our analysis revealed that depletion of mtDNA in breast epithelial cells results in in vitro tumorigenic phenotype as well as breast tumorigenesis in a xenograft model. We identified two major gene networks which were differentially regulated between parental and rho(0) epithelial cells. The focal proteins in these networks include (i) FN1 (fibronectin) and (ii) p53. Bioinformatic analyses of FN1 network identified laminin, integrin and 3 of 6 members of peroxiredoxin whose expression were altered in rho(0) epithelial cells. In the p53 network, we identified SMC4 and WRN whose changes in expression suggest that this network may affect chromosomal stability. Consistent with above finding our study revealed an increase in DNA double strand breaks and unique chromosomal rearrangements in rho(0) breast epithelial cells. Additionally, we identified tight junction proteins claudin-1 and claudin-7 in p53 network. To determine the functional relevance of altered gene expression, we focused on detailed analyses of claudin-1 and -7 proteins in breast tumorigenesis. Our study determined that (i) claudin-1 and 7 were indeed downregulated in rho(0) breast epithelial cells, (ii) downregulation of claudin-1 or -7 led to neoplastic transformation of breast epithelial cells, and (iii) claudin-1 and -7 were also downregulated in primary breast tumors. Together, our study suggest that mtDNA encoded OXPHOS genes play a key role in transformation of breast epithelial cells and that multiple pathway involved in mitochondria-to-nucleus retrograde regulation contribute to transformation of breast epithelial cells.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , DNA, Mitochondrial/metabolism , Gene Expression Regulation, Neoplastic , Mitochondria/pathology , Animals , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Epithelium/pathology , Female , Humans , Mice , Mitochondria/metabolism , Neoplasm Transplantation , Oxygen/metabolism , Phosphorylation , Tumor Suppressor Protein p53/metabolism
12.
Mutat Res ; 625(1-2): 112-24, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17658559

ABSTRACT

Mitochondrial function plays an important role in multiple human diseases and mutations in the mitochondrial genome have been detected in nearly every type of cancer investigated to date. However, the mechanism underlying the interrelation is unknown. We used human cell lines depleted of mitochondrial DNA as models and analyzed the outcome of mitochondrial dysfunction on major cellular repair activities. We show that the deoxyribonucleoside triphosphate (dNTP) pools are affected, most prominently we detect a 3-fold reduction of the dTTP pool when normalized to the number of cells in S-phase. It is known that imbalanced dNTP pools are mutagenic and in accordance, we show that mitochondrial dysfunction results in chromosomal instability, which can explain its role in tumor development. We did not find any straightforward correlation between ATP levels and dNTP pools in cells with defective mitochondrial activity. Our results suggest that mitochondria are central players in maintaining genomic stability and in controlling essential nuclear processes such as upholding a balanced supply of nucleotides.


Subject(s)
Chromosomal Instability/physiology , Deoxyribonucleotides/metabolism , Mitochondria/metabolism , Chromosomal Instability/genetics , Comet Assay , DNA Repair/genetics , DNA Repair/physiology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , HeLa Cells , Humans , Micronucleus Tests , Mitochondria/genetics , Thymine Nucleotides/metabolism
13.
Mamm Genome ; 18(5): 328-37, 2007 May.
Article in English | MEDLINE | ID: mdl-17565425

ABSTRACT

The LGI1 gene has been implicated in the development of epilepsy and the invasion phenotype of glial cells. Controversy over the specific tissue expression pattern of this gene has stemmed from conflicting reports generated using immunohistochemistry and the polymerase chain reaction. LGI1 is one of a four-member family of secreted proteins with high homology and here we demonstrate, using GFP-tagged constructs from the four LGI1family members, that commonly used antibodies against LGI1 cross-react with different family members. With the uncertainty surrounding the use of commercially available antibodies to truly establish the expression pattern of LGI1, we generated transgenic mice carrying the LGI1-containing BAC, RP23-127G7, which had been modified to express the GFP reporter gene under the control of the endogenous regulatory elements required for LGI1 expression. Three founder mice were generated, and immunohistochemistry was used to determine the tissue-specific pattern of expression. In the brain, distinct regions of glial and neuronal cell expression were identified, as well as the choriod plexus, which is largely pia-derived. In addition, strong expression levels were identified in glandular regions of the prostate, individual tubules in the kidney, sympathetic ganglia in the kidney, sebaceous glands in the skin, the islets of Langerhans, the endometrium, and the ovary and testes. All other major organs analyzed were negative. The pattern of reporter gene expression was identical in three individual founder mice, arguing against a position effect altering expression profile due to the integration site of the BAC.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Proteins/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Transfection
14.
Arch Otolaryngol Head Neck Surg ; 133(5): 457-63, 2007 May.
Article in English | MEDLINE | ID: mdl-17515504

ABSTRACT

OBJECTIVES: To assess the forms and extent of genomic instability in thyroid cancers and colorectal neoplasms and to determine if such measurements could explain the generally excellent prognosis of thyroid malignant neoplasms compared with colon carcinoma. DESIGN: Tumor genome analyses. Genomic instability was measured by the following 4 methods, listed in ascending order based on the size of events detected: inter-simple sequence repeat polymerase chain reaction (ISSR-PCR), fractional allelic loss (FAL) analysis, array-based comparative genomic hybridization (aCGH), and spectral karyotyping (SKY). RESULTS: The genomic instability index of 32 thyroid carcinomas, 59 colon carcinomas, and 11 colon polyps was determined by ISSR-PCR; no difference was seen among the 3 groups by this method. Fractional allelic loss rates were comparable in thyroid cancers and colon polyps and lower than FAL rates in colorectal cancers. Indolent papillary thyroid carcinomas were essentially diploid with no large-scale alterations in chromosome number or structure when evaluated by aCGH or SKY. In anaplastic thyroid cancers, aCGH revealed abundant chromosome alterations. Colorectal carcinomas showed extensive copy number changes and chromosomal rearrangements when analyzed by aCGH and SKY. CONCLUSIONS: Genomic alterations in papillary thyroid carcinoma, such as in benign colon polyps, are principally smaller events detected by ISSR-PCR. With the more aggressive tumor types (ie, anaplastic thyroid and colorectal carcinomas), larger events detected by FAL analysis, aCGH, and SKY were revealed. We hypothesize that mutations caused by smaller genomic alterations enable thyroid cells to achieve a minimal malignant phenotype. Mutations for aggressive biological behavior appear with larger genomic events.


Subject(s)
Carcinoma, Papillary/genetics , Carcinoma/genetics , Colonic Neoplasms/genetics , Genomic Instability/genetics , Thyroid Neoplasms/genetics , Alleles , Biomarkers, Tumor , Chromosomes, Human, Pair 8/genetics , Humans , Karyotyping , Loss of Heterozygosity/genetics , Point Mutation/genetics , Polymerase Chain Reaction
15.
Hum Mol Genet ; 16(11): 1359-66, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17412756

ABSTRACT

Down syndrome is caused by a genomic imbalance of human chromosome 21 which is mainly observed as trisomy 21. The regions on human chromosome 21 are syntenically conserved in three regions on mouse chromosomes 10, 16 and 17. Ts65Dn mice, the most widely used model for Down syndrome, are trisomic for approximately 56.5% of the human chromosome 21 syntenic region on mouse chromosome 16. To generate a more complete trisomic mouse model of Down syndrome, we have established a 22.9 Mb duplication spanning the entire human chromosome 21 syntenic region on mouse chromosome 16 in mice using Cre/loxP-mediated long-range chromosome engineering. The presence of the intact duplication in mice was confirmed by fluorescent in situ hybridization and BAC-based array comparative genomic hybridization. The expression levels of the genes within the duplication interval reflect gene-dosage effects in the mutant mice. The cardiovascular and gastrointestinal phenotypes of the mouse model were similar to those of patients with Down syndrome. This new mouse model represents a powerful tool to further understand the molecular and cellular mechanisms of Down syndrome.


Subject(s)
Cardiovascular Abnormalities/genetics , Chromosomes, Human, Pair 21/genetics , Digestive System Abnormalities/genetics , Gene Duplication , Synteny , Animals , Humans , Mice , Translocation, Genetic
16.
BMC Cancer ; 5: 145, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16271139

ABSTRACT

BACKGROUND: Inflammation is commonly associated with lung tumors. Since inflammatory mediators, including members of the interleukin-6 (IL-6) cytokine family, suppress proliferation of normal epithelial cells, we hypothesized that epithelial cells must develop mechanisms to evade this inhibition during the tumorigenesis. This study compared the cytokine responses of normal epithelial cells to that of premalignant cells. METHODS: Short-term primary cultures of epithelial cells were established from bronchial brushings. Paired sets of brushings were obtained from areas of normal bronchial epithelium and from areas of metaplastic or dysplastic epithelium, or areas of frank endobronchial carcinoma. In 43 paired cultures, the signalling through the signal transducer and activator of transcription (STAT) and extracellular regulated kinase (ERK) pathways and growth regulation by IL-6, leukemia inhibitory factor (LIF), oncostatin M (OSM), interferon-gamma (IFNgamma) or epidermal growth factor (EGF) were determined. Inducible expression and function of the leukemia inhibitory factor receptor was assessed by treatment with the histone deacetylase inhibitor depsipeptide. RESULTS: Normal epithelial cells respond strongly to OSM, IFNgamma and EGF, and respond moderately to IL-6, and do not exhibit a detectable response to LIF. In preneoplastic cells, the aberrant signaling that was detected most frequently was an elevated activation of ERK, a reduced or increased IL-6 and EGF response, and an increased LIF response. Some of these changes in preneoplastic cell signaling approach those observed in established lung cancer cell lines. Epigenetic control of LIF receptor expression by histone acetylation can account for the gain of LIF responsiveness. OSM and macrophage-derived cytokines suppressed proliferation of normal epithelial cells, but reduced inhibition or even stimulated proliferation was noted for preneoplastic cells. These alterations likely contribute to the supporting effects that inflammation has on lung tumor progression. CONCLUSION: This study indicates that during the earliest stage of premalignant transformation, a modified response to cytokines and EGF is evident. Some of the altered cytokine responses in primary premalignant cells are comparable to those seen in established lung cancer cell lines.


Subject(s)
Cell Transformation, Neoplastic , Cytokines/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Densitometry , Epidermal Growth Factor/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Histones/metabolism , Humans , Inflammation , Interferon-gamma/metabolism , Interleukin-6/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Macrophages/metabolism , Oncostatin M , Signal Transduction , Time Factors , Treatment Outcome
17.
Cancer Genet Cytogenet ; 163(1): 23-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16271952

ABSTRACT

The t(3;9)(p14;p21) in the MCF10A human mammary gland epithelial cell line was the single cytogenetic event that accompanied the transition from primary culture to immortalized cell line, suggesting that it is related to the development of the immortalization phenotype. To study the molecular consequences of the breakpoints in this rearrangement, we used a combination of fluorescence in situ hybridization (FISH) and array comparative genomic hybridization (CGH). The 3p14 translocation breakpoint occurs within BAC RP11-795e22, which accommodates only the TAFA1 gene, a novel cysteine-rich secreted protein thought to be involved in cytokine signaling. TAFA1 is expressed in normal breast tissue, not in MCF10A, and shows differential expression in a range of breast cancer cell lines. The 9p translocation breakpoint results in a deletion of approximately 4 megabases on the derivative chromosome 9, which includes the CDKN2A (p16) gene. Array CGH and FISH analysis demonstrated that BAC 149i22, which contains the CDKN2A/B genes, is also deleted specifically on the apparently normal copy of chromosome 9, making MCF10A null for the p16/p15 genes. The exact extent of gains and losses of chromosome regions resulting from rearrangements involving chromosomes 1q, 5q, and 8q have also been characterized using the BAC arrays.


Subject(s)
Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 9 , Mammary Glands, Human/physiology , Translocation, Genetic , Cell Line, Tumor , Chromosome Mapping , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mammary Glands, Human/cytology , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
18.
Cancer Res ; 65(19): 8635-9, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16204030

ABSTRACT

DNA methyltransferase 1 (DNMT1)-deficient mice are tumor-prone, and this has been proposed to result from the induction of genomic instability. To address whether loss of DNMT1, or the related protein DNMT3b, results in genomic instability in human cancer cells, we used a near-diploid human colorectal cancer cell line, HCT116, in which one or both DNMT genes were disrupted by homologous recombination. Array-based comparative genomic hybridization analyses indicated that double, but not single, DNMT knock-out cells display two specific alterations in regional DNA copy number, suggesting that DNMT deficiency and genomic DNA hypomethylation are not associated with widespread genomic amplifications or deletions in human cancer cells. However, spectral karyotype analyses revealed that DNMT-deficient HCT116 cells are highly unstable with respect to large-scale chromosomal alterations; furthermore, this effect is characterized by a high degree of individual cell heterogeneity. The induction of chromosomal alterations in DNMT-deficient cells was evidenced both by aneuploidy and by large increases in the number of novel chromosomal translocations. Studies of double knock-out cells indicated that the generation of chromosomal alterations is spontaneous and persistent in vitro, meeting the formal definition of genomic instability. In summary, we show that DNMT deficiency in human cancer cells results in constitutive genomic instability manifested by chromosomal translocations.


Subject(s)
Chromosomal Instability/genetics , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Aneuploidy , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/deficiency , DNA, Neoplasm/genetics , Gene Dosage , HCT116 Cells , Humans , DNA Methyltransferase 3B
19.
Nat Genet ; 37(10): 1099-103, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142235

ABSTRACT

Cultured human embryonic stem cell (hESC) lines are an invaluable resource because they provide a uniform and stable genetic system for functional analyses and therapeutic applications. Nevertheless, these dividing cells, like other cells, probably undergo spontaneous mutation at a rate of 10(-9) per nucleotide. Because each mutant has only a few progeny, the overall biological properties of the cell culture are not altered unless a mutation provides a survival or growth advantage. Clonal evolution that leads to emergence of a dominant mutant genotype may potentially affect cellular phenotype as well. We assessed the genomic fidelity of paired early- and late-passage hESC lines in the course of tissue culture. Relative to early-passage lines, eight of nine late-passage hESC lines had one or more genomic alterations commonly observed in human cancers, including aberrations in copy number (45%), mitochondrial DNA sequence (22%) and gene promoter methylation (90%), although the latter was essentially restricted to 2 of 14 promoters examined. The observation that hESC lines maintained in vitro develop genetic and epigenetic alterations implies that periodic monitoring of these lines will be required before they are used in in vivo applications and that some late-passage hESC lines may be unusable for therapeutic purposes.


Subject(s)
Embryo, Mammalian/cytology , Genome, Human/genetics , Mutation , Stem Cells/metabolism , Cell Culture Techniques , Cell Line , DNA/genetics , DNA/metabolism , DNA Methylation , DNA, Mitochondrial/chemistry , Humans , Promoter Regions, Genetic
20.
Genes Chromosomes Cancer ; 44(4): 392-404, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16110500

ABSTRACT

Amplification of a defined chromosome segment on the short arm of chromosome 7 has frequently been reported in glioblastoma multiforme (GBM), where it is generally assumed that it is the result of over expression of the epidermal growth factor receptor (EGFR) gene that provides the selective pressure to maintain the amplification event. We have used high resolution array comparative genomic hybridization (aCGH) to analyze amplification events on chromosome 7p in GBM, which demonstrates that, in fact, several other regions distinct from EGFR can be amplified. To determine the changes in gene expression levels associated with these amplification events, we used oligonucleotide expression arrays to investigate which of the genes in the amplified regions were also over expressed. These analyses demonstrated that not all genes in the amplicons showed increased expression, and we have defined a series of over expressed genes on 7p that could potentially contribute to the development of the malignant phenotype in these tumors. The global analysis of amplification afforded by aCGH analysis has improved our ability to define numerical chromosome abnormalities in cancer cells and has raised the possibility that genes other than EGFR may be important.


Subject(s)
Chromosomes, Human, Pair 7 , Genes, Neoplasm , Genes, erbB-1 , Glioblastoma/genetics , Nucleic Acid Hybridization , Chromosomes, Artificial, Bacterial , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Microarray Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...