Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Breast Cancer Res Treat ; 94(3): 213-23, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16258703

ABSTRACT

First line treatment of hormone dependent breast cancer initially causes tumor regression but later results in adaptive changes and tumor re-growth. Responses to second line treatments occur but tumors again begin to progress after a period of 12???18??months. In depth understanding of the adaptive process would allow the identification of targets to abrogate the development of hormonal resistance and prolong the efficacy of endocrine therapy. We have developed a model system to examine adaptive changes in human MCF-7 breast cancer cells. Upon deprivation of estradiol for a prolonged period of time, a maneuver analogous to surgical oophorectomy in pre-menopausal women and use of aromatase inhibitors in post-menopausal patients, tumor cells adapt and become hypersensitive to estradiol. We reasoned that the expression pattern of multiple genes would change in response to estradiol deprivation and that cDNA microarrays would provide an efficient means of assessing these changes. Accordingly, we examined the transcriptional responses to estradiol in long-term estradiol deprived (LTED) MCF-7 cells with a cDNA microarray containing 1901 known genes and ESTs. To assess the changes induced by long-term estradiol deprivation, we compared the effects of estradiol administration in LTED cells with those in MCF-7 cells, which we had previously reported, and confirmed with real time PCR using the parental and LTED cells. Seven genes and one EST were induced by estradiol in LTED but not in wild type MCF-7 cells, whereas ten genes were down-regulated by estradiol only in LTED cells. The expression of seven genes increased concurrently and five decreased in response to estradiol in both cell types. From these observations, we generated testable hypotheses regarding several genes including DKFZP, RAP-1, ribosomal protein S6, and TM4SF1. Based upon the known functions of these genes and the patterns of observed changes, we postulate that divergent regulation of these genes may contribute to the different biologic responses to estrogen in these cell lines. These results provide targets for further mechanistic studies in our experimental system. Our findings indicate that long-term estradiol deprivation causes expression changes in multiple genes and emphasizes the complexity of the process of cellular adaptation.


Subject(s)
Breast Neoplasms/pathology , Estradiol/physiology , Gene Expression Regulation, Neoplastic , Adaptation, Physiological , Aromatase Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Ovariectomy , Tumor Cells, Cultured
2.
Mol Hum Reprod ; 11(6): 441-50, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15879465

ABSTRACT

Many studies have implicated numerous hormones, growth factors, cytokines and other signal transduction molecules in the pathogenesis of uterine leiomyoma. Estrogen and estrogen-related genes are thought to play a key role in the growth of uterine leiomyomas, but the molecular mechanisms are unclear. In an attempt to investigate various pathways that might be involved in estrogen-regulated uterine leiomyoma growth as well as to identify any novel effector genes, microarray studies comparing estrogen-treated uterine leiomyoma cells (UtLM) and normal myometrial cells to untreated cells were performed. Several genes were differentially expressed in estrogen treated UtLM cells, including insulin-like growth factor-I (IGF-I) and others potentially involved in the IGF-I signalling pathway, specifically genes for A-myb, a transcription factor which promotes cell cycle progression and for MKP-1, a dual specificity phosphatase that dephosphorylates mitogen-activated protein kinase. IGF-I and A-myb were up-regulated in estrogen-treated cells while MKP-1 was down-regulated. Two other cell cycle promoting genes, c-fos and myc, were also down-regulated in estrogen treated UtLM cells. These genes are typically up-regulated in response to estrogen in some cells, notably breast epithelial cells, yet consistently have lower expression levels in uterine leiomyoma tissue when compared to autologous myometrium. Our results demonstrate some novel genes that may play a role in the growth of uterine leiomyoma, strengthen the case for involvement of the IGF-I pathway in the response of UtLM to estrogen and corroborate evidence that uterine smooth muscle cells respond to estrogen with a different gene expression pattern than that seen in epithelial cells.


Subject(s)
Estrogens/pharmacology , Insulin-Like Growth Factor I/genetics , Leiomyoma/genetics , MAP Kinase Signaling System/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Uterine Neoplasms/genetics , Cell Cycle Proteins/genetics , Dual Specificity Phosphatase 1 , Estrogens/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/genetics , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/metabolism , Leiomyoma/immunology , Leiomyoma/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/metabolism , Trans-Activators/analysis , Trans-Activators/metabolism , Tumor Cells, Cultured , Up-Regulation , Uterine Neoplasms/immunology , Uterine Neoplasms/metabolism , Uterus/cytology
3.
Physiol Genomics ; 13(3): 263-75, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12657712

ABSTRACT

Atherogenic stimuli trigger complex responses in vascular smooth muscle cells (VSMCs) that culminate in activation/repression of overlapping signal transduction cascades involving oxidative stress. In the case of benzo[a]pyrene (BaP), a polycyclic aromatic hydrocarbon present in tobacco smoke, the atherogenic response involves interference with redox homeostasis by oxidative intermediates of BaP metabolism. The present studies were conducted to define genomic profiles and predictive gene biological networks associated with the atherogenic response of murine (aortic) VSMCs to BaP. A combined oxidant-antioxidant treatment regimen was used to identify redox-sensitive targets during the early course of the atherogenic response. Gene expression profiles were defined using cDNA microarrays coupled to analysis of variance and several clustering methodologies. A predictor algorithm was then applied to gain insight into critical gene-gene interactions during atherogenesis. Supervised and nonsupervised analyses identified clones highly regulated by BaP, unaffected by antioxidant, and neutralized by combined chemical treatments. Lymphocyte antigen-6 complex, histocompatibility class I component factors, secreted phosphoprotein, and several interferon-inducible proteins were identified as novel redox-regulated targets of BaP. Predictor analysis confirmed these relationships and identified immune-related genes as critical molecular targets of BaP. Redox-dependent patterns of gene deregulation indicate that oxidative stress plays a prominent role during the early stages of BaP-induced atherogenesis.


Subject(s)
Arteriosclerosis/chemically induced , Arteriosclerosis/genetics , Benzo(a)pyrene/toxicity , Gene Expression Profiling/methods , Genomics/methods , Oligonucleotide Array Sequence Analysis/methods , Oxidants/toxicity , Algorithms , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Arteriosclerosis/metabolism , Benzo(a)pyrene/metabolism , Cells, Cultured , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling/statistics & numerical data , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Oxidants/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Predictive Value of Tests
4.
Neoplasia ; 4(3): 204-17, 2002.
Article in English | MEDLINE | ID: mdl-11988840

ABSTRACT

Activation of the insulin-like growth factor-1 receptor (IGF-1R) by IGF-1 is associated with the risk and progression of many types of cancer, although despite this it remains unclear how activated IGF-1R contributes to cancer progression. In this study, gene expression changes elicited by IGF-1 were profiled in breast epithelial cells. We noted that many genes are functionally linked to cancer progression and angiogenesis. To validate some of the changes observed, the RNA and/or protein was confirmed for c-fos, cytochrome P450 1A1, cytochrome P450 1B1, interleukin-1 beta, fas ligand, vascular endothelial growth factor, and urokinase plasminogen activator. Nuclear proteins were also temporally monitored to address how gene expression changes were regulated. We found that IGF-1 stimulated the nuclear translocation of phosphorylated AKT, hypoxic-inducible factor-1 alpha, and phosphorylated cAMP-responsive element-binding protein, which correlated with temporal changes in gene expression. Next, the promoter regions of IGF-1-regulated genes were searched in silico. The promoters of genes that clustered together had similar regulatory regions. In summary, IGF-1 inscribes a gene expression profile relevant to cancer progression, and this study provides insight into the mechanism(s) whereby some of these changes occur.


Subject(s)
Epithelium/pathology , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Neovascularization, Pathologic , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Progression , Endothelial Growth Factors/metabolism , Fas Ligand Protein , Humans , Lymphokines/metabolism , Membrane Glycoproteins/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Biotechniques ; 32(2): 322, 324, 326-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11848409

ABSTRACT

The use of DNA microarrays has spanned numerous disciplines of life science research. Despite the volume of studies utilizing this technology, no consensus exists on basic issues such as the determination of significantly altered genes in a given experiment, often leading to either false-negative or false-positive data. In this report, we study the effect of dilution of biological alterations on the detection level of gene expression differences using cDNA microarrays. We propose that subtle alterations in transcript levels of genes below the 2-fold level should be considered when replicate hybridizations are performed, because these subtle gene expression changes may be due to a robust response in few cells. We measured the effect of dilution of gene expression and found that differences in gene expression between the two cell lines assayed (HaCaT and MCF-7) were detected even after a 20-fold dilution factor. These results better our understanding of biological alterations that comprise a relatively small percentage of an assayed organ and help in the interpretation of gene expression data.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Cell Line , DNA, Complementary , Humans , Sensitivity and Specificity
6.
Environ Health Perspect ; 109(9): 881-91, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11673116

ABSTRACT

Microarray technology has been applied to a variety of different fields to address fundamental research questions. The use of microarrays, or DNA chips, to study the gene expression profiles of biologic samples began in 1995. Since that time, the fundamental concepts behind the chip, the technology required for making and using these chips, and the multitude of statistical tools for analyzing the data have been extensively reviewed. For this reason, the focus of this review will be not on the technology itself but on the application of microarrays as a research tool and the future challenges of the field.


Subject(s)
Environmental Health , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/trends , Humans , Infections , Medical Informatics , Neoplasms/genetics , Research Design
7.
Proc Natl Acad Sci U S A ; 98(17): 9587-92, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11493692

ABSTRACT

Mutational inactivation of BRCA1 confers a cumulative lifetime risk of breast and ovarian cancers. However, the underlying basis for the tissue-restricted tumor-suppressive properties of BRCA1 remains poorly defined. Here we show that BRCA1 mediates ligand-independent transcriptional repression of the estrogen receptor alpha (ERalpha), a principal determinant of the growth, differentiation, and normal functional status of breasts and ovaries. In Brca1-null mouse embryo fibroblasts and BRCA1-deficient human ovarian cancer cells, ERalpha exhibited ligand-independent transcriptional activity that was not observed in Brca1-proficient cells. Ectopic expression in Brca1-deficient cells of wild-type BRCA1, but not clinically validated BRCA1 missense mutants, restored ligand-independent repression of ERalpha in a manner dependent upon apparent histone deacetylase activity. In estrogen-dependent human breast cancer cells, chromatin immunoprecipitation analysis revealed the association of BRCA1 with ERalpha at endogenous estrogen-response elements before, but not after estrogen stimulation. Collectively, these results reveal BRCA1 to be a ligand-reversible barrier to transcriptional activation by unliganded promoter-bound ERalpha and suggest a possible mechanism by which functional inactivation of BRCA1 could promote tumorigenesis through inappropriate hormonal regulation of mammary and ovarian epithelial cell proliferation.


Subject(s)
BRCA1 Protein/physiology , Gene Silencing , Receptors, Estrogen/genetics , Saccharomyces cerevisiae Proteins , Transcriptional Activation , Adenocarcinoma/pathology , Animals , Breast Neoplasms/pathology , Cathepsin D/biosynthesis , Cathepsin D/genetics , Cells, Cultured , DNA-Binding Proteins , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogens , Female , Fibroblasts/metabolism , Fungal Proteins/genetics , Genes, BRCA1 , Genes, p53 , Histone Deacetylases/metabolism , Humans , Ligands , Mice , Mice, Knockout , Mutation, Missense , Neoplasms, Hormone-Dependent/pathology , Ovarian Neoplasms/pathology , Protein Biosynthesis , Proteins/genetics , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Trefoil Factor-1 , Tumor Suppressor Proteins
8.
Bioinformatics ; 17(6): 564-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395436

ABSTRACT

SUMMARY: MAPS is a MicroArray Project System for management and interpretation of microarray gene expression experiment information and data. Microarray project information is organized to track experiments and results that are: (1) validated by performing analysis on stored replicate gene expression data; and (2) queried according to the biological classifications of genes deposited on microarray chips.


Subject(s)
Database Management Systems , Oligonucleotide Array Sequence Analysis/standards , Computer Communication Networks , Data Display , Programming Languages
9.
Cancer Res ; 61(4): 1392-7, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245440

ABSTRACT

MLH1 is an integral part of the mismatch repair complex, and the loss of this protein is associated with the acquisition of a mutator phenotype, microsatellite instability, and a predisposition to cancer. Deficiencies in the mismatch repair complex, including the loss of MLH1, result in elevated resistance to specific inducers of DNA damage, yet the mechanisms involved in this DNA-damage resistance are largely unknown. Abnormal cellular responses to DNA damage can lead to the selection of cells with a greater propensity for neoplastic transformation and might also reduce the effectiveness of certain chemotherapeutic drugs. It is therefore important to identify agents that provide selective pressure for growth of MLH1-deficient cells and to characterize further the pathways involved. In this study, we show that both human epithelial and mouse embryo fibroblast cell lines lacking the MLH1 protein are more resistant to two inducers of oxidative stress, hydrogen peroxide and tert-butyl hydroperoxide. Our analyses suggest that the observed differences in cellular viability are mediated primarily through apoptotic pathways and not through deficiencies in cell cycle checkpoint controls. Additional characterization of the signaling pathways for hydrogen peroxide-induced apoptosis in MLH1-proficient cells demonstrates the involvement of increased mitochondrial permeability, the release of cytochrome c, and caspase 3 activation. Together, our data indicate that cells lacking MLH1 may possess a selective growth advantage under oxidatively stressed conditions via the disregulation of apoptosis, possibly involving the mitochondria.


Subject(s)
Apoptosis/physiology , Neoplasm Proteins/physiology , Oxidative Stress/physiology , Peroxides/toxicity , Adaptor Proteins, Signal Transducing , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Base Pair Mismatch , Carrier Proteins , Cell Cycle/drug effects , Cell Line , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Repair , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydrogen Peroxide/toxicity , Mice , MutL Protein Homolog 1 , Nuclear Proteins , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured , tert-Butylhydroperoxide/toxicity
10.
Toxicol Sci ; 59(2): 193-5, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158710

ABSTRACT

The article highlighted in this issue is "An Aryl Hydrocarbon Receptor Independent Mechanism of JP-8 Jet Fuel Immunotoxicity in Ah-Responsive and Ah-Nonresponsive Mice" by Andrew C. Dudley, Margie M. Peden-Adams, Jackie EuDaly, Richard S. Pollenz, and Deborah E. Keil (pp. 251-259).


Subject(s)
Genomics , Proteome/analysis , Toxicology/methods , Animals , DNA/analysis , Gene Expression Profiling , Humans , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Proteome/genetics , Receptors, Aryl Hydrocarbon/genetics
11.
Mol Cell Biol ; 21(3): 940-51, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11154280

ABSTRACT

Inactivation of DNA mismatch repair by mutation or by transcriptional silencing of the MLH1 gene results in genome instability and cancer predisposition. We recently found (P. V. Shcherbakova and T. A. Kunkel, Mol. Cell. Biol. 19:3177-3183, 1999) that an elevated spontaneous mutation rate can also result from increased expression of yeast MLH1. Here we investigate the mechanism of this mutator effect. Hybridization of poly(A)(+) mRNA to DNA microarrays containing 96.4% of yeast open reading frames revealed that MLH1 overexpression did not induce changes in expression of other genes involved in DNA replication or repair. MLH1 overexpression strongly enhanced spontaneous mutagenesis in yeast strains with defects in the 3'-->5' exonuclease activity of replicative DNA polymerases delta and epsilon but did not enhance the mutation rate in strains with deletions of MSH2, MLH1, or PMS1. This suggests that overexpression of MLH1 inactivates mismatch repair of replication errors. Overexpression of the PMS1 gene alone caused a moderate increase in the mutation rate and strongly suppressed the mutator effect caused by MLH1 overexpression. The mutator effect was also reduced by a missense mutation in the MLH1 gene that disrupted Mlh1p-Pms1p interaction. Analytical ultracentrifugation experiments showed that purified Mlh1p forms a homodimer in solution, albeit with a K(d) of 3.14 microM, 36-fold higher than that for Mlh1p-Pms1p heterodimerization. These observations suggest that the mismatch repair defect in cells overexpressing MLH1 results from an imbalance in the levels of Mlh1p and Pms1p and that this imbalance might lead to formation of nonfunctional mismatch repair complexes containing Mlh1p homodimers.


Subject(s)
Base Pair Mismatch , DNA Repair , Fungal Proteins/genetics , Fungal Proteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Primers/genetics , Dimerization , Fungal Proteins/chemistry , Gene Expression , Gene Silencing , Genes, Fungal , Genome, Fungal , Molecular Sequence Data , MutL Protein Homolog 1 , MutL Proteins , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , Protein Structure, Quaternary , Sequence Homology, Amino Acid , Suppression, Genetic
12.
J Biochem Mol Toxicol ; 15(5): 231-42, 2001.
Article in English | MEDLINE | ID: mdl-11835620

ABSTRACT

Toxicogenomics is a term that represents the merging of toxicology with novel genomics techniques. Data generated in the new-age era of toxicology is relatively complex, requires new bioinformatics tools for adequate interpretation, and allows for the rapid generation of testable hypotheses. Hazard identification and risk assessment processes will advance from the use of genomics techniques, which will lead to greater understanding of mechanism(s) of action of toxicants, development of novel biomarkers of exposure and effect, and better identification of sensitive subpopulations.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Toxicology , Animals , DNA, Complementary , Humans , Polymorphism, Genetic , Risk Assessment
13.
Breast Cancer Res ; 2(2): 139-48, 2000.
Article in English | MEDLINE | ID: mdl-11056686

ABSTRACT

STATEMENT OF FINDINGS: We tested the hypothesis that BRCA1 may play a role in the regulation of ovarian tumor cell death as well as the inhibition of ovarian cell proliferation. Introduction of BRCA1 antisense retroviral constructs into BG-1 estrogen-dependent ovarian adenocarcinoma cells resulted in reduced BRCA1 expression. BRCA1 antisense pooled populations and derived subclones were able to proliferate in monolayer culture without estrogen, whereas control cells began to die after 10 days of estrogen deprivation. In addition, both populations and subclones of BRCA1 antisense infected cells demonstrated a growth advantage in monolayer culture in the presence of estrogen and were able to proliferate in monolayer culture without estrogen, while control cells did not. Furthermore, clonal studies demonstrated that reduced levels of BRCA1 protein correlated with growth in soft agar and greater tumor formation in nude mice in the absence of estrogen. These data suggest that reduction of BRCA1 protein in BG-1 ovarian adenocarcinoma cells may have an effect on cell survival during estrogen deprivation both in vitro and in vivo.


Subject(s)
Adenocarcinoma/pathology , BRCA1 Protein/antagonists & inhibitors , Ovarian Neoplasms/pathology , RNA, Antisense/pharmacology , Receptors, Estrogen/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Survival , Cell Transformation, Neoplastic , Estrogens/pharmacology , Female , Genes, Tumor Suppressor , Humans , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Messenger/metabolism , Retroviridae , Transfection , Tumor Cells, Cultured/drug effects
14.
Gene ; 250(1-2): 201-8, 2000 May 30.
Article in English | MEDLINE | ID: mdl-10854793

ABSTRACT

The BRCA1 gene, mutations of which contribute significantly to hereditary breast cancer, was not identified in the existing YAC and BAC libraries. The gene is now available only as a set of overlapping fragments that form a contig. In this work we describe direct isolation of a genomic copy of BRCA1 from human DNA by transformation-associated recombination (TAR) cloning. Despite the presence of multiple repeats, most of the primary BRCA1 YAC isolates did not contain detectable deletions and could be stably propagated in a host strain with conditional RAD52. Similar to other circular YACs, approximately 90kb BRCA1 YACs were efficiently and accurately retrofitted into bacterial artificial chromosomes (BACs) with the Neo(R) mammalian selectable marker and transferred as circular BAC/YACs in E. coli cells. The BRCA1 BAC/YAC DNAs were isolated from bacterial cells and were used to transfect mouse cells using the Neo(R) gene as selectable marker. Western blot analysis of transfectants showed that BRCA1 YACs isolated by a TAR cloning contained a functional gene. The advantage of this expression vector is that the expression of BRCA1 is generated from its own regulatory elements and does not require additional promoter elements that may result in overexpression of the protein. In contrast to the results with cDNA expression vectors, the level of BRCA1 expression from this TAR vector is stable, does not induce cell death, maintains serum regulation, and approximates the level of endogenously expressed BRCA1 in human cells. The entire isolation procedure of BRCA1 described in this paper can be accomplished in approximately 10 days and can be applied to isolation of gene from clinical material. We propose that the opportunity to directly isolate normal and mutant forms of BRCA1 will greatly facilitate analysis of the gene and its contribution to breast cancer.


Subject(s)
BRCA1 Protein/genetics , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Animals , BRCA1 Protein/metabolism , Cell Line , Chromosomes, Artificial, Yeast , Chromosomes, Bacterial , Cloning, Molecular , Gene Expression , Genetic Vectors , Humans , Transfection , Transformation, Genetic , Tumor Cells, Cultured
15.
Mutat Res ; 462(2-3): 179-88, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10767629

ABSTRACT

The p53 tumor suppressor is a universal sensor of genotoxic stress that regulates the transcription of genes required for cell-cycle arrest and apoptosis. In response to DNA damage, the p53 protein is phosphorylated at its amino-terminus and becomes stabilized upon disruption of an interaction with its negative regulator, MDM2. Subsequent phosphorylation and acetylation of p53 promote different interactions with other proteins and with target gene regulatory elements to facilitate cell-cycle arrest, apoptosis, or adaptation in response to DNA damage. Downstream of p53, p21 is responsible for growth arrest in G1, but other p53 target genes are responsible for G2 cell-cycle arrest. In response to genotoxic insult, p53-induced apoptosis results from overlapping downstream pathways that both suppress mitogenic and survival signaling and promote pro-apoptotic signaling. Adaptation to DNA damage is manifested by p53-mediated expression of its negative regulator, MDM2. The frequency of observed mutations in p53 predicts that its inactivation is a requisite step in tumorigenesis, as p53 is mutated in approximately 50% of human tumors. Thus, it is likely that in the remaining tumors, genetic aberrations will occur in pathways that regulate p53 or in pathways directly downstream of p53. The advances in the understanding of p53 signaling over the past few years point to many potential overlapping signaling pathways, where mutations may occur as alternative modes to p53 mutation.


Subject(s)
Mutagens/pharmacology , Nuclear Proteins , Tumor Suppressor Protein p53/drug effects , DNA Damage , Gene Expression Regulation , Humans , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Gynecol Oncol ; 77(1): 121-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10739701

ABSTRACT

OBJECTIVE: The aim of this study was to examine whether cells containing the heterozygous form of a BRCA1 185delAG mutation would exhibit abnormal growth or an altered response to DNA damage. METHODS: A primary culture of human mammary epithelial cells (90P) was obtained from the nontumor breast tissue of a 35-year-old patient who had undergone a mastectomy for removal of a breast tumor. These cells were immortalized (90PE6E7) following retroviral infection with HPV-16 viral E6/E7. genes. Both the 90P cell strain and the cell line were characterized for their ability to grow in culture, form colonies in soft agar, and produce tumors in athymic nude mice compared to normal breast epithelial cells containing wild-type BRCA1. 90P cells were also analyzed for cellular response to gamma radiation and H(2)O(2). RESULTS: These cells were confirmed to contain a frameshift mutation, 185delAG, of the BRCA1 gene. Despite being heterozygous for wild-type BRCA1, the 220-kDa full-size BRCA1 protein was abundantly expressed. 90P and 90PE6E7 cells grew at a similar rate and were anchorage dependent. 90PE6E7 also failed to form tumors in athymic nude mice. Finally, 90P cells exhibited a survival response similar to that of normal mammary epithelial cells to radiation damage and exposure to oxidative stress. CONCLUSION: To our knowledge the 90P cells and the 90PE6E7 cells are the first characterized, non-tumor-derived breast epithelial cells that are heterozygous for the BRCA1 germline mutation 185delAG. Our conclusion is that these BRCA1 mutant cells appear to have growth and stress response characteristics similar to those of normal human breast cells, which is consistent with the hypothesis that loss of heterozygosity must occur to impair putative BRCA1 function.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Frameshift Mutation , Genes, BRCA1/genetics , Loss of Heterozygosity , Adult , Animals , Breast Neoplasms/pathology , Culture Media , DNA Damage , Female , Germ-Line Mutation , Humans , Mice , Mice, Nude , Tumor Cells, Cultured
17.
Cancer Res ; 59(19): 4759-60, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519378

ABSTRACT

One major challenge facing today's cancer researchers and toxicologists is the development of new approaches for the identification of carcinogens and other environmental hazards. Here, we describe the potential impact of emerging technologies for measuring gene expression profiles on carcinogen identification and on the general field of toxicology. An example of one of these technologies is the use of cDNA microarray chips. We provide an overview to the key questions that are confronting investigators charged with determining the relative safety of natural or synthetic chemicals to which humans are exposed, followed by a discussion of how cDNA microarray technology may be applied to these questions. Gene chip technology is still a relatively new technology, and only a handful of studies have demonstrated its utility. However, as the technical hurdles to development are passed, the use of this methodology in addressing the questions raised here will be critical to increase the sensitivity of detection of the potential toxic effects of environmental chemicals and to understand their risks to humans.


Subject(s)
Carcinogens/toxicity , Consumer Product Safety , DNA, Complementary , Carcinogens/classification , Gene Library , Humans , Oligonucleotide Array Sequence Analysis , Pharmacology, Clinical/methods , Toxicology/methods
18.
Mol Carcinog ; 24(3): 153-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10204799

ABSTRACT

The availability of genome-scale DNA sequence information and reagents has radically altered life-science research. This revolution has led to the development of a new scientific subdiscipline derived from a combination of the fields of toxicology and genomics. This subdiscipline, termed toxicogenomics, is concerned with the identification of potential human and environmental toxicants, and their putative mechanisms of action, through the use of genomics resources. One such resource is DNA microarrays or "chips," which allow the monitoring of the expression levels of thousands of genes simultaneously. Here we propose a general method by which gene expression, as measured by cDNA microarrays, can be used as a highly sensitive and informative marker for toxicity. Our purpose is to acquaint the reader with the development and current state of microarray technology and to present our view of the usefulness of microarrays to the field of toxicology.


Subject(s)
Gene Expression , Genome , Noxae/toxicity , Nucleic Acid Hybridization/methods , Toxicology/methods , Animal Testing Alternatives , Animals , Automation , Biological Assay , Cricetinae , DNA Damage , DNA, Complementary/genetics , Drug Evaluation/methods , Environmental Monitoring/methods , Fluorescent Dyes , Forecasting , Gene Expression Regulation/drug effects , Humans , Mice , Microchemistry , Noxae/analysis , Noxae/pharmacology , Oligonucleotides/chemical synthesis , Oligonucleotides/genetics , Photochemistry , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Rats , Toxicology/instrumentation
19.
Cancer Res ; 59(24): 6087-90, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10626795

ABSTRACT

The promoter of the hTERT gene encoding the catalytic subunit of telomerase was recently cloned and has a dense CG-rich CpG island, suggesting a role for methylation in regulation of hTERT expression. In this study, we have initiated the analysis of the regulation of hTERT expression by examining the methylation status of up to 72 CpG sites extending from 500 bases upstream of the transcriptional start site of the hTERT gene into the first exon in 37 cell lines. These cell lines represent a variety of cell and tissue types, including normal, immortalized, and cancer cell lines from lung, breast, and other tissues. Using bisulfite genomic sequencing, we did not find a generalized pattern of site-specific or region-specific methylation that correlated with expression of the hTERT gene: most of the hTERT-negative normal cells and about one-third of the hTERT-expressing cell lines had the unmethylated/hypomethylated promoter, whereas the other hTERT-expressing cell lines showed partial or total methylation of the promoter. The promoter of one hTERT-negative fibroblast cell line, SUSM-1, was methylated at all sites examined. Treatment of SUSM-1 cells with the demethylating agent 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor trichostatin A induced the cells to express hTERT, suggesting a potential role for DNA methylation and/or histone deacetylation in negative regulation of hTERT. This study indicates that there are multiple levels of regulation of hTERT expression in CpG island methylation-dependent and -independent manners.


Subject(s)
DNA Methylation , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , RNA , Telomerase/genetics , Cell Line , Chromatin/chemistry , Chromatin/physiology , DNA-Binding Proteins , Humans , Sulfites/chemistry , Telomerase/biosynthesis , Tumor Cells, Cultured
20.
Exp Cell Res ; 244(1): 33-42, 1998 Oct 10.
Article in English | MEDLINE | ID: mdl-9770346

ABSTRACT

We have observed that normal, diploid Syrian hamster embryo cells (SHE) express the enzyme telomerase but undergo senescence at the end of their replicative lifespan. After 20-30 population doublings (pd) these cells cease proliferating, enlarge in size, exhibit a pH 6.0 senescence-associated beta-galactosidase activity, and fail to phosphorylate the RB protein or enter into S-phase after serum stimulation. We have observed that SHE cells express telomerase throughout their replicative lifespan and that the average telomere length does not appear to decrease, remaining at about 23 kb in senescent cells. In addition, individual clones of SHE cells also have telomerase activity and telomeres that do not decrease in length, ruling out the possibility that there is a rare, immortal subpopulation of telomerase-expressing cells that is lost during passaging. Together, these data suggest that SHE cells are likely to senesce by a mechanism that does not involve telomere loss.


Subject(s)
Cellular Senescence , Telomerase/biosynthesis , Animals , Cell Line, Transformed , Cells, Cultured , Cricetinae , Embryo, Mammalian , Enzyme Activation , Humans , Mesocricetus , Telomerase/metabolism , Telomere/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...