Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Oncogene ; 28(34): 3022-32, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19543317

ABSTRACT

B-lymphoma Moloney murine leukaemia virus insertion region-1 (BMI1) is a member of the polycomb group of transcription repressors, which functions in stem cell maintenance and oncogenesis through the inhibition of the INK4A/ARF tumour suppressor locus. Overexpression of BMI1 is associated with poor prognosis in several human cancers, including breast cancer. We have previously shown that BMI1 collaborates with H-RAS to induce transformation of MCF10A human mammary epithelial cells through dysregulation of multiple growth pathways independent of the INK4A/ARF locus. In this study, we show that BMI1 collaborates with H-RAS to promote increased proliferation, invasion and resistance to apoptosis in vitro, and an increased rate of spontaneous metastases from mammary fat pad xenografts including novel metastases to the brain. Furthermore, in collaboration with H-RAS, BMI1 induced fulminant metastatic disease in the lung using a tail vein model of haematogenous spread through accelerated cellular proliferation and inhibition of apoptosis. Finally, we show that knockdown of BMI1 in several established breast cancer cell lines leads to decreased oncogenic behaviour in vitro and in vivo. In summary, BMI1 collaborates with H-RAS to induce an aggressive and metastatic phenotype with the unusual occurrence of brain metastasis, making it an important target for diagnosis and treatment of aggressive breast cancer.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Nuclear Proteins/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Humans , Lung Neoplasms/secondary , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Phenotype , Polycomb Repressive Complex 1 , Transplantation, Heterologous
2.
Br J Cancer ; 96(1): 126-33, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17179983

ABSTRACT

Bmi-1 is a polycomb group protein that was identified as c-myc cooperating oncogene in murine lymphomagenesis. The current study was undertaken to determine the role of Bmi-1 in human oral carcinogenesis. Bmi-1 protein and RNA expression levels were markedly enhanced in the cells of oral squamous cell carcinomas (OSCC) compared with that of normal human oral keratinocytes (NHOK). Enhanced-Bmi-1 expression was also detected in situ in the archived oral mucosal tissues with cancerous and precancerous histopathology, including that of mild epithelial dysplasia. Thus, Bmi-1 expression occurs at a very early stage in oral carcinogenesis. To determine the biological role of Bmi-1 in cell proliferation, endogenous Bmi-1 was knocked down in actively proliferating SCC4 cells and NHOK by RNA interference. After Bmi-1 knockdown, cell replication was severely retarded. However, the expression of p16(INK4A), a known cellular target of Bmi-1, was not changed in cells with or without Bmi-1 knockdown. Furthermore, Bmi-1 knockdown in HOK-16B-BaP-T cells, in which the p16(INK4A)/pRb pathway was abrogated, led to immediate arrest of replication and loss of viable cells. Thus, our data suggest that Bmi-1 may act through p16(INK4A)-independent pathways to regulate cellular proliferation during oral cancer progression.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Amplification , Humans , Immunohistochemistry , Keratinocytes/cytology , Keratinocytes/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Staining and Labeling , Tumor Cells, Cultured
3.
Cancer Res ; 61(21): 8014-21, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691827

ABSTRACT

The normal epithelial cell-specific-1 (NES1)/kallikrein 10 gene is expressed in normal mammary epithelial cells, but its expression is dramatically decreased in breast cancer cell lines. Now, we have cloned and characterized the active promoter region of NES1. Using a luciferase reporter system, we demonstrate that most tumor cell lines are able to support full or partial transcription from the NES1 promoter, suggesting a role for promoter-independent cis-acting mechanisms of loss of NES1 expression. We show that hypermethylation of the NES1 gene represents one such mechanism. Using methylation-specific PCR and sequence analysis of sodium bisulfite-treated genomic DNA, we demonstrate a strong correlation between exon 3 hypermethylation and loss of NES1 mRNA expression in a panel of breast cancer cell lines and in primary tumors. Treatment of NES1-nonexpressing cells with a demethylating agent led to reexpression of NES1, suggesting an important role of hypermethylation in the loss of NES1 expression. We suggest that hypermethylation is responsible for tumor-specific loss of NES1 gene expression. Our results also suggest that hypermethylation of the NES1 gene may serve as a potential marker for breast cancer.


Subject(s)
Azacitidine/analogs & derivatives , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , DNA Methylation , Kallikreins/genetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , CpG Islands , DNA Methylation/drug effects , Decitabine , Exons , Gene Expression Regulation, Neoplastic , Humans , Introns , Kallikreins/biosynthesis , Luciferases/genetics , Luciferases/metabolism , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
4.
Eur J Biochem ; 268(10): 2784-91, 2001 May.
Article in English | MEDLINE | ID: mdl-11358493

ABSTRACT

Many normal cells respond to potentially oncogenic stimuli by undergoing cellular senescence, a state of irreversibly arrested proliferation and altered differentiated function. Cellular senescence very likely evolved to suppress tumorigenesis. In support of this idea, it is regulated by several tumor suppressor genes. At the heart of this regulation is p53. p53 is essential for the senescence response to short telomeres, DNA damage, oncogenes and supraphysiological mitogenic signals, and overexpression of certain tumor suppressor genes. Despite the well-documented central role for p53 in the senescence response, many questions remain regarding how p53 senses senescence-inducing stimuli and how it elicits the senescent phenotype.


Subject(s)
Aging , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/physiology , Animals , Cell Division , DNA Damage , Humans , Models, Biological , Neoplasms/metabolism , Phenotype , Signal Transduction , Telomere/metabolism
5.
Mol Cell Biol ; 20(1): 273-85, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10594030

ABSTRACT

Normal cells do not divide indefinitely due to a process known as replicative senescence. Human cells arrest growth with a senescent phenotype when they acquire one or more critically short telomeres as a consequence of cell division. Recent evidence suggests that certain types of DNA damage, chromatin remodeling, and oncogenic forms of Ras or Raf can also elicit a senescence response. We show here that E2F1, a multifunctional transcription factor that binds the retinoblastoma (pRb) tumor suppressor and that can either promote or suppress tumorigenesis, induces a senescent phenotype when overexpressed in normal human fibroblasts. Normal human cells stably arrested proliferation and expressed several markers of replicative senescence in response to E2F1. This activity of E2F1 was independent of its pRb binding activity but dependent on its ability to stimulate gene expression. The E2F1 target gene critical for the senescence response appeared to be the p14(ARF) tumor suppressor. Replicatively senescent human fibroblasts overexpressed p14(ARF), and ectopic expression of p14(ARF) in presenescent cells induced a phenotype similar to that induced by E2F1. Consistent with a critical role for p14(ARF), cells with compromised p53 function were immune to senescence induction by E2F1, as were cells deficient in p14(ARF). Our findings support the idea that the senescence response is a critical tumor-suppressive mechanism, provide an explanation for the apparently paradoxical roles of E2F1 in oncogenesis, and identify p14(ARF) as a potentially important mediator of the senescent phenotype.


Subject(s)
Carrier Proteins , Cellular Senescence/genetics , DNA-Binding Proteins , Proteins/genetics , Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Cell Division/genetics , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Tumor Suppressor Protein p14ARF
6.
Aviat Space Environ Med ; 70(12): 1161-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10596768

ABSTRACT

BACKGROUND: Research has focused mainly on the relationship of zinc and copper contents and physical stresses like running, cycling, etc. It has also been reported that other forms of stresses change the concentration of these trace elements in humans. However,there are no reports on the effects of high altitude induced hypoxic stress on the plasma levels of these metals. Since hypoxia is one of the important stresses, we considered it appropriate to observe the changes in the levels of zinc and copper concentrations and in certain related zinc and copper enzymes and hormones in the plasma of human volunteers on acute induction to high altitude. From these findings, we intended to ascertain whether supplementation of these trace elements would be required for optimal health under such conditions. HYPOTHESIS: On acute induction to hypoxia, contents of these trace elements may change as the requirements of stressed organs and tissue may increase. Hence, further supplementation may be beneficial under hypoxic stress for better adaptability. METHOD: Volunteers were divided into two groups: with and without zinc and copper salt supplementation. Blood samples were collected at sea level and on induction to acute hypoxia on days 3 and 10. Trace mineral contents and their related enzyme (alkaline phosphatase) and hormone (ceruloplasmin) levels were determined in plasma samples. RESULTS: Plasma zinc contents were significantly reduced upon induction to high altitude in the non-supplemented group, but not in the zinc-supplemented group. Alkaline phosphatase activity increased significantly upon induction to the high altitude stress. The enzyme activity remained elevated up to day 10 of the stress. Plasma copper contents and ceruloplasmin activity did not change upon induction to high altitude. CONCLUSION: Under hypoxic stress, circulating levels of zinc and alkaline phosphatase in plasma changed appreciably as plasma zinc was transported into the organs and tissues. However, circulating levels of copper and ceruloplasmin in plasma did not change, indicating no extra supplementation of copper is required under hypoxic stress.


Subject(s)
Altitude Sickness/blood , Altitude Sickness/drug therapy , Copper/blood , Copper/therapeutic use , Zinc/blood , Zinc/therapeutic use , Acute Disease , Adaptation, Physiological , Adult , Alkaline Phosphatase/blood , Altitude Sickness/enzymology , Ceruloplasmin/analysis , Copper/deficiency , Copper/metabolism , Humans , Male , Nutritional Requirements , Time Factors , Tissue Distribution , Zinc/deficiency , Zinc/metabolism
7.
J Cell Physiol ; 168(3): 580-8, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8816912

ABSTRACT

The induction of dihydrofolate reductase (DHFR), a key enzyme in DNA biosynthesis that is induced just before the onset of S phase, is markedly attenuated in senescent human fibroblasts (Pang and Chen, 1994, J. Cell. Physiol., 160:531-538). Footprinting analysis of the 365 bp promoter region of the human DHFR gene (-381 to -17) indicated that nuclear proteins bind to a cluster of cis-elements, including two overlapping E2F binding sequences, two Sp1 sites, and one Yi sequence. Gel mobility shift assays were performed to assess the role of each cis-element in the regulation of DHFR gene expression. We found that 1) Sp1 binding activity was constitutively expressed throughout the cell cycle in early passage and senescent cells; 2) Yi binding activity was undetectable in both early passage and senescent cells; and 3) E2F binding activity was serum-inducible, senescence-dependent, and prominent in presenescent cells but strikingly diminished in senescent cells. Northern blot analysis of the expression of E2F and DP family members showed that the E2F-1, E2F-4, and E2F-5 mRNA was growth- and senescence-dependent, whereas E2F-3, DP-1, and DP-2 expression was constitutive and senescence-independent. In contrast, E2F-2 mRNA was not detectable in IMR-90 or WI-38 human fibroblasts. Western blot analysis showed that among the E2F-associated proteins, the expression of E2F-1, cyclin A, and cyclin B but not p107 was cell cycle- and senescence-dependent. A nuclear extract mixing experiment suggested that an inhibitory factor may further reduce E2F binding activity in senescent cells.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Gene Expression Regulation, Enzymologic , Tetrahydrofolate Dehydrogenase/genetics , Base Sequence , Cells, Cultured , Cellular Senescence , DNA Primers/chemistry , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , E2F4 Transcription Factor , E2F5 Transcription Factor , HeLa Cells , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 1 , Sp1 Transcription Factor/metabolism , Transcription Factor DP1 , Transcription Factors/genetics
8.
Mol Cell Biol ; 16(6): 2987-97, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8649410

ABSTRACT

p21Sdi1/WAF1/Cip1 inhibits cyclin-dependent protein kinases and cell proliferation. p21 is presumed to inhibit growth by preventing the phosphorylation of growth-regulatory proteins, including the retinoblastoma tumor suppressor protein (pRb). The ultimate effector(s) of p21 growth inhibition, however, is largely a matter of conjecture. We show that p21 inhibits the activity of E2F, an essential growth-stimulatory transcription factor that is negatively regulated by unphosphorylated pRb. p21 suppressed the activity of E2F-responsive promoters (dihydrofolate reductase and cdc2), but E2F-unresponsive promoters (c-fos and simian virus 40 early) were unaffected. Moreover, the simian virus 40 early promoter was rendered p21 suppressible by introducing wild-type, but not mutant, E2F binding sites; p21 deletion mutants showed good agreement in their abilities to inhibit E2F transactivation and DNA synthesis; and E2F-1 (which binds pRb), but not E2F-4 (which does not), reversed both inhibitory effects of p21. Despite the central role for pRb in regulating E2F, p21 suppressed growth and E2F activity in cells lacking a functional pRb. Moreover, p21 protein (wild type but not mutant) specifically disrupted an E2F-cyclin-dependent protein kinase 2-p107 DNA binding complex in nuclear extracts of proliferating cells, whether or not they expressed normal pRb. Thus, E2F is a critical target and ultimate effector of p21 action, and pRb is not essential for the inhibition of growth or E2F-dependent transcription.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/pharmacology , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Retinoblastoma Protein/metabolism , Transcription Factors/antagonists & inhibitors , 3T3 Cells , Animals , Base Sequence , Binding Sites/genetics , Cell Division/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , DNA/biosynthesis , DNA/genetics , DNA/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Humans , Mice , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription, Genetic
9.
Biol Signals ; 5(3): 154-62, 1996.
Article in English | MEDLINE | ID: mdl-8864060

ABSTRACT

Normal somatic cells invariably enter a state of permanent growth arrest and altered function after a finite number of divisions. This phenomenon is termed cellular or replicative senescence. Replicative senescence is thought to be a tumor-suppressive mechanism, and a contributing factor in aging. Three features distinguish senescent from presenescent cells: an irreversible block to cell proliferation, increased resistance to apoptotic death, and changes in differentiated functions. Senescence entails an altered pattern of gene expression, much of which is due to altered transcription. At least three growth regulatory transcriptional modulators are repressed in senescent cells: the c-fos component of the AP1 transcription factor, the Id1 and Id2 helix-loop-helix (HLH) proteins that negatively regulate basic HLH transcription factors, and the E2F-1 component of the E2F transcription factor. Failure to express any one of these modulators is very likely sufficient to arrest cell proliferation. Loss of these modulators may also explain many of the functional changes shown by senescent cells. In the case of c-fos repression, the resulting decline in AP-1 activity may be exacerbated by an altered ratio of AP-1 components to a protein known as QM or Jif. QM interacts with the c-jun component of AP-1 and suppresses AP-1 activity. We cloned QM from a senescent fibroblast cDNA library, and found that it was neither cell cycle- nor senescence-regulated. However, QM suppressed the growth of murine and human fibroblasts when overexpressed. Thus, an altered balance between positive factors (e.g., AP-1 components) and negative factors (e.g., QM) may lead to the growth arrest, as well as the changes in differentiated gene expression, that are a hallmark of senescent cells.


Subject(s)
Cell Cycle Proteins , Cell Physiological Phenomena , DNA-Binding Proteins , Repressor Proteins , Ribosomal Proteins , Transcription Factors/physiology , Carrier Proteins/metabolism , Cell Cycle , Cell Division/physiology , Cell Line , Cells/cytology , Cells/metabolism , Cellular Senescence/physiology , DNA/antagonists & inhibitors , DNA/biosynthesis , E2F Transcription Factors , E2F1 Transcription Factor , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Inhibitor of Differentiation Protein 1 , Retinoblastoma-Binding Protein 1 , Ribosomal Protein L10 , Transcription Factor AP-1/metabolism , Transcription Factor DP1 , Transcription Factors/metabolism
10.
Dev Genet ; 18(2): 161-72, 1996.
Article in English | MEDLINE | ID: mdl-8934878

ABSTRACT

Normal somatic cells of higher organisms do not divide indefinitely. After a finite number of divisions, normal cells irreversibly cease proliferation by a process termed replicative or cellular senescence. Replicative senescence is controlled by multiple, dominant-acting genes about which very little is known. The only genes known to reactivate DNA synthesis in senescent cells are viral oncogenes encoding proteins that bind and inactivate the p53 and retinoblastoma (pRb) tumor suppressor proteins. SV40 T antigen is the best studied of these viral oncoproteins. T[K1] is a T antigen point mutant that selectively is defective in binding pRb and the pRb-related proteins p107 and p130. We show that T[K1] stimulated quiescent human fibroblasts to synthesize DNA nearly as well as wild-type T but was incapable of stimulating senescent cells. We tested several growth regulatory genes that are repressed in senescent cells for ability to restore activity to T[K1]. These included c-fos, c-jun, Id-1, Id-2, E2F-1, and cdc2. Only the helix-loop-helix (HLH) protein, Id-1, restored the ability of T[K1] to reactivate DNA synthesis in senescent cells. This activity of Id-1 was not shared by Id-2, a related protein, and depended on an intact HLH domain. It did not appear that Id-1 interacted directly with pRb or p107. Constitutive Id-1 expression failed to rescue proliferating cells from growth inhibition by pRb, p107, or p130, and failed to interact with pRb in the yeast two hybrid system. Because Id proteins negatively regulate basic-HLH (bHLH) transcription factors, we suggest that senescent cells express one or more bHLH factor that cooperates with pRb, or pRb-related proteins, to suppress proliferation.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , DNA/biosynthesis , Fibroblasts/physiology , Repressor Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Cell Division , Cell Line , Cellular Senescence , E2F Transcription Factors , E2F1 Transcription Factor , Fibroblasts/cytology , Helix-Loop-Helix Motifs , Humans , Inhibitor of Differentiation Protein 1 , Mutation , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
11.
Exp Gerontol ; 31(1-2): 7-12, 1996.
Article in English | MEDLINE | ID: mdl-8706807

ABSTRACT

Replicative senescence is a fundamental feature of most, if not all, somatic higher eukaryotic cells. The phenomenon has been studied for more than three decades, during which time the genetics and cell biology of senescent cells were characterized. In recent years, progress has been made on understanding the molecular basis for replicative senescence. At present, we now have a good, albeit still incomplete, understanding of some of the immediate causes for the growth arrest of senescent cells. The challenges for the future will be to understand the molecular bases for the prime causes of senescent phenotype, including the growth arrest and the altered differentiation.


Subject(s)
Cellular Senescence , Animals , Cell Division , Cells, Cultured , Genes, Regulator , Humans , Phenotype , Proto-Oncogenes
12.
Proc Natl Acad Sci U S A ; 92(20): 9363-7, 1995 Sep 26.
Article in English | MEDLINE | ID: mdl-7568133

ABSTRACT

Normal somatic cells invariably enter a state of irreversibly arrested growth and altered function after a finite number of divisions. This process, termed replicative senescence, is thought to be a tumor-suppressive mechanism and an underlying cause of aging. There is ample evidence that escape from senescence, or immortality, is important for malignant transformation. By contrast, the role of replicative senescence in organismic aging is controversial. Studies on cells cultured from donors of different ages, genetic backgrounds, or species suggest that senescence occurs in vivo and that organismic lifespan and cell replicative lifespan are under common genetic control. However, senescent cells cannot be distinguished from quiescent or terminally differentiated cells in tissues. Thus, evidence that senescent cells exist and accumulate with age in vivo is lacking. We show that several human cells express a beta-galactosidase, histochemically detectable at pH 6, upon senescence in culture. This marker was expressed by senescent, but not presenescent, fibroblasts and keratinocytes but was absent from quiescent fibroblasts and terminally differentiated keratinocytes. It was also absent from immortal cells but was induced by genetic manipulations that reversed immortality. In skin samples from human donors of different age, there was an age-dependent increase in this marker in dermal fibroblasts and epidermal keratinocytes. This marker provides in situ evidence that senescent cells may exist and accumulate with age in vivo.


Subject(s)
Cellular Senescence , Skin Aging , Skin/enzymology , beta-Galactosidase/biosynthesis , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Cell Differentiation , Cell Division , Cell Line , Cell Transformation, Neoplastic , Cells, Cultured , DNA/biosynthesis , Epidermis/enzymology , Female , Fibroblasts/cytology , Fibroblasts/enzymology , HeLa Cells , Humans , Keratinocytes/cytology , Keratinocytes/enzymology , Male , Skin/cytology , Skin/pathology , Skin Neoplasms/surgery , Thymidine/metabolism , beta-Galactosidase/analysis
13.
J Biol Chem ; 269(23): 16180-6, 1994 Jun 10.
Article in English | MEDLINE | ID: mdl-8206919

ABSTRACT

Several mammalian genes expressed in late G1 are positively regulated by E2F, a heterodimeric transcription factor. Genes encoding two E2F proteins, E2F-1 and DP-1, were regulated differently during the cell cycle and replicative senescence of normal human fibroblasts. In presenescent cells, E2F-1 mRNA was cell-cycle regulated, appearing a few hours before S phase. By contrast, DP-1 mRNA was constitutively expressed, independent of position in the cell cycle. After a finite number of divisions, normal cells enter a state of irreversible growth arrest termed senescence. Many genes remain mitogen-inducible in senescent cells; there are, however, exceptions, including several late G1 genes potentially regulated by E2F. Senescent cells expressed DP-1 at the presenescent level, but did not express E2F-1 mRNA. Senescent cells were also markedly deficient in E2F binding activity associated with the dihydrofolate reductase promoter. E2F-1 and DP-1 expression vectors only weakly induced DNA synthesis in quiescent or senescent human cells and immortal murine NIH3T3 cells, although the E2F-1 vector stimulated DNA synthesis in immortal murine A31 cells, and transactivated E2F-responsive promoters in NIH3T3 cells. The results suggest that senescent cells may fail to express late G1 genes due to repression of E2F-1, leading to a deficiency of E2F activity. Furthermore, although E2F-1 stimulates DNA synthesis in some cells, other cells, including normal human fibroblasts, require additional factors.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle/physiology , DNA-Binding Proteins , Gene Expression Regulation , Transcription Factors/metabolism , Animals , Antigens, Viral, Tumor/genetics , Base Sequence , Cell Nucleus/metabolism , Cellular Senescence/physiology , E2F Transcription Factors , E2F1 Transcription Factor , Fibroblasts/metabolism , Humans , Mice , Molecular Sequence Data , Protein Binding , RNA, Messenger/analysis , Resting Phase, Cell Cycle/physiology , Retinoblastoma-Binding Protein 1 , S Phase/physiology , Sp1 Transcription Factor/metabolism , Transcription Factor DP1 , Transcription Factors/genetics , Transcriptional Activation , Transfection
14.
Exp Cell Res ; 212(1): 132-40, 1994 May.
Article in English | MEDLINE | ID: mdl-8174635

ABSTRACT

Normal eukaryotic cells divide a limited number of times, after which they enter a state of irreversible growth arrest and altered function termed senescence. Cell senescence entails changes in the expression of growth- and differentiation-specific genes, suggesting that senescent cells express an altered profile of transcription factors. Nuclear extracts were prepared from presenescent (quiescent and growing) and senescent human fibroblasts (WI-38) and from SV40-immortalized WI-38 cells and Y79 human retinoblastoma tumor cells--both of which have escaped senescence. The extracts were assayed for ability to form specific protein-DNA complexes with oligonucleotides containing binding sites for the general transcription factors CTF (CAAT-binding transcription factor), SP1 (promotor-specific transcription factor-1), and TFIID (transcription factor-IID) and the more gene-specific factors AP1 (activator protein factor-1), CREBP (cAMP response element-binding protein), GREBP (glucocorticoid response element-binding protein), NF-kappa B (nuclear factor kappa B) and OctBP (octamer-binding protein). Two TFIID complexes and the GREBP, NF-kappa B, and SP1 complexes were similar in presenescent and senescent cells. AP1, CREBP, and CTF complexes were reduced in senescent cells. Two activities were more abundant in senescent cells: OctBP and one TFIID complex. This TFIID complex was present in quiescent cells, but absent from four human cell lines that lack a functional retinoblastoma protein (pRb); both pRb-specific and TFIID-specific antibodies selectively disrupted it. The data suggest that an altered profile of transcription factors may specify the senescent phenotype and that pRb may interact with TFIID or a TFIID-associated protein(s).


Subject(s)
Cell Nucleus/metabolism , Fibroblasts/metabolism , Oligonucleotide Probes/metabolism , Transcription Factors/metabolism , Base Sequence , Cells, Cultured , Cellular Senescence/physiology , Down-Regulation , Humans , Lung/cytology , Molecular Sequence Data , Retinoblastoma Protein/metabolism , Transcription, Genetic
16.
J Bacteriol ; 174(14): 4583-93, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1624447

ABSTRACT

Repetitive extragenic palindromic (REP) sequences are highly conserved inverted repeat sequences originally discovered in Escherichia coli and Salmonella typhimurium. We have physically mapped these sequences in the E. coli genome by using Southern hybridization of an ordered phage bank of E. coli (Y. Kohara, K. Akiyama, and K. Isono, Cell 50:495-508, 1987) with generic REP probes derived from the REP consensus sequence. The set of REP probe-hybridizing clones was correlated with a set of clones expected to contain REP sequences on the basis of computer searches. We also show that a generic REP probe can be used in Southern hybridization to analyze genomic DNA digested with restriction enzymes to determine genetic relatedness among natural isolates of E. coli. A search for these sequences in other members of the family Enterobacteriaceae shows a consistent correlation between both the number of occurrences and the hybridization strength and genealogical relationship.


Subject(s)
Chromosome Mapping/methods , Enterobacteriaceae/genetics , Escherichia coli/genetics , Repetitive Sequences, Nucleic Acid/genetics , Base Sequence , Escherichia coli/classification , Genetic Variation , Genome, Bacterial , Molecular Sequence Data , Phylogeny , Species Specificity
18.
Nucleic Acids Res ; 18(1): 151-6, 1990 Jan 11.
Article in English | MEDLINE | ID: mdl-2155395

ABSTRACT

The gene gyrA encoding the DNA gyrase A subunit of Klebsiella pneumoniae has been cloned in the plasmid pBR322. Bases of about 3.5 Kb DNA have been sequenced to locate the gyrA gene. An open reading frame of 2628 nucleotides coding for a 97 KD protein has been identified. Homology to the extent of about 85% was detected at the nucleotide level and about 90% at the amino acid level, when the sequences were compared with that of Escherichia coli gyrA. Some very interesting differences have, however, been found in the promoter region.


Subject(s)
DNA Topoisomerases, Type II/genetics , Genes, Bacterial , Klebsiella pneumoniae/genetics , Promoter Regions, Genetic , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Hydrogen Bonding , Molecular Sequence Data , Nucleic Acid Conformation , Regulatory Sequences, Nucleic Acid , Restriction Mapping
19.
Appl Opt ; 28(1): 17, 1989 Jan 01.
Article in English | MEDLINE | ID: mdl-20548411
SELECTION OF CITATIONS
SEARCH DETAIL
...