Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Radiother Oncol ; 128(2): 283-300, 2018 08.
Article in English | MEDLINE | ID: mdl-29929859

ABSTRACT

Radiation therapy is used to treat cancer by radiation-induced DNA damage. Despite the best efforts to eliminate cancer, some cancer cells survive irradiation, resulting in cancer progression or recurrence. Alteration in DNA damage repair pathways is common in cancers, resulting in modulation of their response to radiation. This article focuses on the recent findings about molecules and pathways that potentially can be targeted to sensitize prostate cancer cells to ionizing radiation, thereby achieving an improved therapeutic outcome.


Subject(s)
DNA Damage/radiation effects , DNA Repair/radiation effects , Prostatic Neoplasms/radiotherapy , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/radiation effects , Aurora Kinases/radiation effects , Cell Cycle/radiation effects , Checkpoint Kinase 1/radiation effects , Cyclin-Dependent Kinases/radiation effects , Cyclins/radiation effects , HSP90 Heat-Shock Proteins/radiation effects , Histone Deacetylases/radiation effects , Humans , Hyaluronan Receptors/radiation effects , Hypoxia-Inducible Factor 1, alpha Subunit/radiation effects , Male , Mutation/radiation effects , NEDD8 Protein/radiation effects , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/radiotherapy , Neoplasm, Residual , Neoplastic Stem Cells/radiation effects , Phosphatidylinositol 3-Kinases/radiation effects , Poly(ADP-ribose) Polymerases/radiation effects , Proto-Oncogene Proteins c-met/radiation effects , Radiation Tolerance , Radiation, Ionizing , Receptors, Androgen/radiation effects , TOR Serine-Threonine Kinases/radiation effects , Zinc Finger Protein GLI1/radiation effects
2.
Cell Prolif ; 37(5): 337-49, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15377333

ABSTRACT

Low-frequency electromagnetic fields are suspected of being involved in carcinogenesis, particularly in processes that could be related to cancer promotion. Because development of cancer is associated with deregulated cell growth and we previously observed a magnetic field-induced decrease in DNA synthesis [Lange et al. (2002) Alterations in the cell cycle and in the protein level of cyclin D1p, 21CIP1, and p16INK4a after exposure to 50 HZ. MF in human cells. Radiat. Environ. Biophys.41, 131], this study aims to document the influence of 50 Hz, 1 mT magnetic fields (MF), with or without initial gamma-ionizing radiation (IR), on the following cell proliferation-relevant parameters in human amniotic fluid cells (AFC): cell cycle distribution, expression of the G1 phase-regulating proteins Cdk4, cyclin D1, p21CIP1 and p16INK4a, and Cdk4 activity. While IR induced a G1 delay and a dose-dependent G2 arrest, no discernible changes in cell cycle kinetics were observed due to MF exposure. However, a significant decrease in the protein expression of cyclin D1 and an increase in p21CIP1- and p16INK4a-expression could be detected after exposure to MF alone. IR-exposure caused an augmentation of p21CIP1- and p16INK4a- levels as well, but did not alter cyclin D1 expression. A slight diminution of Cdk4 activity was noticed after MF exposure only, indicating that Cdk4 appears not to act as a mediator of MF- or IR-induced changes in the cell cycle of AFC cells. Co-exposure to MF/IR affected neither cell cycle distribution nor protein expression or kinase activity additionally or synergistically, and therefore MF seems not to modify the mutagenic potency of IR.


Subject(s)
Cell Cycle Proteins/radiation effects , Cell Cycle/radiation effects , Electromagnetic Fields , Eukaryotic Cells/radiation effects , G1 Phase/radiation effects , Radiation, Ionizing , Amnion/cytology , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/radiation effects , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin D1/radiation effects , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/radiation effects , Cyclins/genetics , Cyclins/metabolism , Cyclins/radiation effects , Down-Regulation/genetics , Down-Regulation/radiation effects , G1 Phase/genetics , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/radiation effects
3.
Oncogene ; 23(20): 3670-80, 2004 Apr 29.
Article in English | MEDLINE | ID: mdl-15116098

ABSTRACT

Metastasis-promoting Mts1(S100A4) protein belongs to the S100 family of Ca(2+)-binding proteins. A mouse strain with a germ-line inactivation of the S100A4 gene was generated. The mice were viable and did not display developmental abnormalities in the postnatal period. However, an abnormal sex ratio was observed in the litters with the S100A4-/- genotype, raising the possibility of a certain level of embryonic lethality in this strain. In all, 10% of 10-14-month-old S100A4-null animals developed tumors. This is a characteristic feature of mouse strains with inactivated tumor suppressor genes. Spontaneous tumors of S100A4-/- mice were p53 positive. Recently, we have shown that S100A4 interacts with p53 tumor suppressor protein and induces apoptosis. We proposed that impairment of this interaction could affect the apoptosis-promoting function of p53 that is involved in its tumor suppressor activity. The frequency of apoptosis in the spleen of S100A4-/- animals after whole-body gamma-irradiation was reduced compared to the wild-type animals. The same was true for the transcriptional activation of the p53 target genes - waf/p21/cip1 and bax. Taken together, these observations indicate that spontaneous tumors in S100A4-/- mice are a result of functional destabilization of p53 tumor suppressor gene.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , Genetic Predisposition to Disease , Neoplasm Metastasis/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2 , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Cyclins/radiation effects , Gamma Rays , Mice , Neoplasms/etiology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/radiation effects , Spleen/radiation effects , Thymus Gland/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
4.
Health Phys ; 85(1): 36-42, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12852469

ABSTRACT

Advances in high throughput analysis of mRNA expression have made it possible to establish gene expression profiles for different cells, tissues, diseases and exposure states. For instance, recent studies have demonstrated the utility of such an approach to classify sub-types of cancers with more detail than was previously possible. In addition, gene expression studies of ionizing radiation exposure both in vitro and in vivo are affording insight into the molecular mechanisms of mammalian radiation response. We have demonstrated that radiation expression profiles are a good predictor of p53 function in cell lines, and such profiles also indicate a major role for p53-regulated genes in the in vivo radiation response. Gene expression can be a sensitive indicator of radiation response as we have shown linear dose-responses for induction of several genes down to doses as low as 2 cGy. As profiles are established from radiation studies, it is hoped that they may be useful for identifying individuals with specific exposures or predisposition to negative outcome of exposure. Although this technology holds great promise, some obstacles remain to be overcome before it can be successfully applied to population studies.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation/radiation effects , Oligonucleotide Array Sequence Analysis/methods , Radiometry/methods , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/radiation effects , DNA/radiation effects , Gamma Rays , Gene Expression/radiation effects , Genes, p53/radiation effects , Humans , Lymphocytes/metabolism , Lymphocytes/radiation effects , Myoblasts/metabolism , Myoblasts/radiation effects , Reproducibility of Results , Sensitivity and Specificity , Statistics as Topic
5.
J Neurosurg ; 98(2): 378-84, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12593626

ABSTRACT

OBJECT: Ionizing radiation is the gold-standard adjuvant treatment for glioblastoma multiforme (GBM), the most aggressive primary brain tumor. The mechanisms underlying neoplastic glial cell growth inhibition after administration of ionizing radiation, however, remain largely unknown. In this report, the authors characterize the response of GBM cells to ionizing radiation and elucidate factors that correlate with the radiosensitivity of these tumors. METHODS: Six human GBM cell lines were subjected to increasing doses of radiation. Each demonstrated a dose-dependent suppression of cell proliferation. In the most radiosensitive cell line, the authors demonstrated a transient increase in the expression of the cyclin-dependent kinase inhibitors (CDKIs) p21 and p27, which corresponded with a G1 cell-cycle arrest. In contrast, the most radioresistant cell line demonstrated a decrease in p21 and p27 expression levels, which correlated with a failure to arrest. Apoptosis did not occur in any cell line following irradiation. Instead, autophagic cell changes were observed following administration of radiation, regardless of the relative radiosensitivity of the cell line. CONCLUSIONS: These findings elucidate some of the molecular responses of GBMs to irradiation and suggest novel targets for future therapy.


Subject(s)
Autophagy/genetics , Autophagy/radiation effects , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Cycle Proteins/radiation effects , Cyclins/radiation effects , Enzyme Inhibitors/radiation effects , G1 Phase/genetics , G1 Phase/radiation effects , Glioblastoma/pathology , Glioblastoma/radiotherapy , Radiation, Ionizing , Tumor Suppressor Proteins/radiation effects , Brain Neoplasms/genetics , Cell Cycle Proteins/analysis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/analysis , Enzyme Inhibitors/analysis , Glioblastoma/genetics , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Organelles/genetics , Organelles/radiation effects , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Time Factors , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Proteins/analysis
6.
Genetika ; 38(7): 980-4, 2002 Jul.
Article in Russian | MEDLINE | ID: mdl-12174591

ABSTRACT

Synthesis of p53 and WAF1 (p21) proteins was studied in cells of patients with Nijmegen breakage syndrome (NBS) and of patients with ataxia telangiectasia (AT), as well as in normal cells with respect to their response to ionizing radiation (IR). In the NBS cells, the p53 protein was progressively accumulated with increasing radiation dose and reached the maximum 2 h after exposure to radiation at a dose of 5 Gy. The amount of p53 protein was consistently lower than that in normal cells, which was correlated with low content of the WAF1, the protein regulated by p53 at the level of transcription. Suboptimal induction of p53 observed in NBS cells was also characteristic of the AT cells, though the quantitative parameters of the protein synthesis in AT cells were intermediate relative to those in normal and NBS cells. In four NBS lines, the time schedule of p53 synthesis was similar to that observed in normal cells, whereas in AT cells, induction of p53 was significantly delayed as compared to control. In response to irradiation, the amount of p53 protein synthesized in patients with AT and NBS was significantly lower than that in normal cells. The results obtained, as well as the previously published medical and genetic evidence, suggest that the two diseases are of different origin and different genes are responsible for their development.


Subject(s)
Ataxia Telangiectasia/pathology , Chromosome Disorders/pathology , Cyclins/metabolism , Tumor Suppressor Protein p53/radiation effects , Ataxia Telangiectasia/etiology , Ataxia Telangiectasia/radiotherapy , Cells, Cultured , Chromosome Breakage , Chromosome Disorders/etiology , Chromosome Disorders/radiotherapy , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/radiation effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Radiation, Ionizing , Reference Values , Tumor Suppressor Protein p53/metabolism
7.
Eur J Neurosci ; 15(4): 627-36, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11886443

ABSTRACT

Radiation-induced death in the developing brain is p53-dependent. However, genetic studies indicate that the signalling pathways that couple irradiation to p53 expression can vary between different developing neural populations [Herzog et al. (1998) Science, 280, 1089-1091]. Here we establish that signalling downstream of p53 also exhibits brain region-specific differences that are associated with the relative vulnerability of some cell populations to radiation-induced killing in the mouse. Following gamma-irradiation, p53 and p21WAF1/cip1, but not Bax, protein levels increased in the developing cerebellum. In contrast, neither p21WAF1/cip1 nor Bax protein levels were elevated in the retina following irradiation, despite increased p53 expression. In the retina, p53 expression was associated with cells destined to die, whereas in the cerebellum, p53 was expressed in both radiation-sensitive and radiation-resistant neuroblasts of the external granule cell layer. Although p21WAF1/cip1 mRNA was expressed in all p53-positive neuroblasts after irradiation, p21WAF1/cip1 protein was only detected in radiation-resistant neuroblasts of the cerebellum. Thus, p21WAF1/cip1 was subject to post-transcriptional regulation with p21WAF1/cip1 protein only accumulating in cells destined to survive irradiation. Nevertheless, p21WAF1/cip1 function was not essential for radiation resistance, as postmitotic neuroblasts in the external granule cell layer were spared in p21WAF1/cip1 knockout mice.


Subject(s)
Cell Death/radiation effects , Central Nervous System/radiation effects , Cyclins/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Neurons/radiation effects , Proto-Oncogene Proteins c-bcl-2 , Transcription, Genetic/radiation effects , Animals , Animals, Newborn , Cell Death/genetics , Cell Division/genetics , Cell Division/radiation effects , Cell Survival/genetics , Cell Survival/radiation effects , Central Nervous System/growth & development , Central Nervous System/metabolism , Cerebellum/growth & development , Cerebellum/metabolism , Cerebellum/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/radiation effects , Retina/growth & development , Retina/metabolism , Retina/radiation effects , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/radiation effects , bcl-2-Associated X Protein
8.
Gene Ther ; 9(4): 263-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11896465

ABSTRACT

Nitric oxide (NO(*)) has many characteristics including cytotoxicity, radiosensitization and anti-angiogenesis, which make it an attractive molecule for use in cancer therapy. We have investigated the use of iNOS gene transfer, driven by both a constitutive (CMV) and X-ray inducible (WAF1) promoter, for generating high concentrations of NO(*) within tumour cells. We have combined this treatment with radiation to exploit the radiosensitizing properties of this molecule. Transfection of murine RIF-1 tumour cells in vitro with the iNOS constructs resulted in increased iNOS protein levels. Under hypoxic conditions cells were radiosensitized by delivery of both constructs so that these treatments effectively eliminated the radioresistance observed under hypoxic conditions. In vivo transfer of the CMV/iNOS construct by direct tumour injection resulted in a delay (4.2 days) in tumour growth compared with untreated controls. This was equivalent to the effect of 20 Gy X-rays alone. Combination of CMV/iNOS gene transfer with 20 Gy X-rays resulted in a dramatic 19.8 day growth delay compared with controls. Tumours treated with the CMV/iNOS showed large areas of necrosis and abundant apoptosis. We believe that iNOS gene transfer has the potential to be a highly effective treatment in combination with radiotherapy.


Subject(s)
Fibrosarcoma/therapy , Genetic Therapy/methods , Nitric Oxide Synthase/genetics , Radiation-Sensitizing Agents/metabolism , Animals , Apoptosis/radiation effects , Cell Division/radiation effects , Cell Survival/radiation effects , Combined Modality Therapy , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/radiation effects , Female , Fibrosarcoma/pathology , Fibrosarcoma/radiotherapy , Gene Transfer Techniques , Genetic Vectors/therapeutic use , Mice , Mice, Inbred C3H , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Promoter Regions, Genetic/radiation effects , Transfection , Tumor Cells, Cultured
9.
Int J Radiat Oncol Biol Phys ; 52(3): 720-8, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11849795

ABSTRACT

PURPOSE: In vitro, ionizing radiation of epithelial cells leads to upregulation of wild-type p53 and subsequent induction of p21(waf1). The effect of radiotherapy (RT) on the expression of these proteins in patients is unknown. We assessed the influence of RT on the expression of p53 and p21(waf1) in normal mucosa and rectal carcinomas in vivo. METHODS: Tumor and normal tissue samples were derived from rectal cancer patients randomized in a clinical trial in which the value of preoperative RT was evaluated. p53 and p21(waf1) expression was determined in 51 irradiated and 52 nonirradiated patients using immunohistochemistry. RESULTS: In normal mucosa, both p53 and p21(waf1) were strongly upregulated after RT compared with the expression in unirradiated normal tissue (p <0.001). In tumor cells, no significant difference in the expression of p53 or p21(waf1) was found in the irradiated vs. nonirradiated group. In the few rectal tumors with wild-type p53, induction of p53 after RT did not necessarily lead to upregulation of p21(waf1). CONCLUSION: These findings demonstrate that in normal mucosa, a functional p53-p21(waf1) pathway is present, whereas in tumor cells it is defective in almost all cases because of either p53 mutation or down- or upstream disruption in tumors with wild-type p53. Therefore, we believe that the role of p53 expression as a single prognostic marker in rectal cancer needs reconsideration.


Subject(s)
Cyclins/radiation effects , Rectal Neoplasms/radiotherapy , Rectum/radiation effects , Tumor Suppressor Protein p53/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Female , Gene Expression/radiation effects , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Male , Middle Aged , Rectal Neoplasms/metabolism , Rectum/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation
10.
Radiat Res ; 156(5 Pt 1): 558-63, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11604069

ABSTRACT

The spatial correlation between individual particle traversals and the nuclear CDKN1A (p21) response after high-LET irradiation of human fibroblasts was investigated. The experiments were based on a technique for the retrospective detection of particle traversals by means of nuclear track detectors, which were used as the cell substratum. This technique requires the precise repositioning of a sample at different steps of the experimental procedure and uses a computerized microscope stage control. The precision of the spatial correlation is further enhanced by means of reference marks in the track etch material that are produced by preirradiation of the plates with charged-particle beams at low fluences. The pattern of the CDKN1A foci that were induced by charged-particle traversals at 1 h postirradiation was found to coincide extremely well with the pattern of particle tracks. This represents direct evidence that CDKN1A foci are located at the sites of particle traversals and thus provides further evidence that the radiation-induced accumulation of the CDKN1A protein takes place at the sites of the primary damage.


Subject(s)
Cyclins/radiation effects , DNA Damage , Bystander Effect , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , Humans , Linear Energy Transfer
11.
Oncogene ; 20(3): 320-8, 2001 Jan 18.
Article in English | MEDLINE | ID: mdl-11313961

ABSTRACT

The high prevalence and great diversity of p53 tumor suppressor gene mutations in human tumors call for development of therapeutic molecules that rescue function of aberrant p53 protein. P53 mutations also offer new approaches to the study of the origins of mutations in human cancer. An experimental mouse model with a genetically modified but normal functioning p53 gene harboring the human rather than the murine core domain, would be of considerable benefit to research on both cancer therapeutics and etiology; however, it is uncertain whether such mice would permit biological functions of p53 to be retained. Using a Cre/lox P gene-targeting approach, we have constructed a human p53 knock-in (hupki) mouse strain in which exons 4-9 of the endogenous mouse p53 allele were replaced with the homologous, normal human p53 gene sequence. The chimeric p53 allele (p53(KI)) is properly spliced, transcribed in various tissues at levels equivalent to wild-type mice, and yields cDNA with the anticipated sequence, that is, with a core domain matching that of humans. The hupki p53 protein binds to p53 consensus sequences in gel mobility shift assays and accumulates in the nucleus of hupki fibroblasts in response to UV irradiation, as is characteristic of wild-type p53. Induction of various p53-regulated genes in spleen of gamma-irradiated homozygous hupki mice (p53(KI/KI)), and the kinetics of p53-dependent apoptosis in thymocytes are similar to results with wild-type (p53(+/+)) mice, further indicating normal p53 pathway function in the hupki strain. The mice are phenotypically normal and do not develop spontaneous tumors at an early age, in contrast to knock-out (p53(-/-)) strains with a defective p53 gene. The chimeric (p53(KI)) allele thus appears to provide a biological equivalent to the endogenous murine (p53(+)) gene. This strain is a unique tool for examining in vivo spontaneous and induced mutations in human p53 gene sequences for comparison with published human tumor p53 mutation spectra. In addition, the hupki strain paves the way for mouse models in pre-clinical testing of pharmaceuticals designed to modulate DNA-binding activity of human p53.


Subject(s)
DNA Damage/drug effects , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/genetics , Viral Proteins , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Cyclins/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , Dexamethasone/pharmacology , Embryonic and Fetal Development/genetics , Exons , Gamma Rays , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Homozygote , Humans , Integrases/genetics , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/radiation effects , Spleen/radiation effects , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/radiation effects , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays , bcl-2-Associated X Protein
13.
Mutagenesis ; 16(1): 17-23, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11139595

ABSTRACT

We evaluated F3 mouse offspring from paternal F0 attenuated 137Cs gamma-irradiation (1.0 Gy) for heritable effects on gene products that can modulate cell proliferation rate and that may be markers for genomic instability. The F3 generation was selected for evaluation as a stringent test for heritability of effects from paternal F0 germline irradiation. Male CD1 mice were bred 6 weeks after irradiation so that the fertilizing sperm were type B spermatogonia at the time of irradiation. The resulting F1 males were bred to CD1 females to produce F2 four-cell embryos. The F2 embryos with a radiation history were paired with 'control' CD1 four-cell embryos that were heterozygous for the neo transgene. These F2 XY-XY chimeras, consisting of cells derived from both an embryo with a paternal F0 radiation history and a control embryo, were transferred to foster mothers, raised to adulthood and bred to produce F3 offspring. F3 offspring were evaluated for hepatic activities of receptor tyrosine kinase, protein kinase C and MAP kinase and for protein levels of nuclear p53 and p21(waf1). All three protein kinase activities were altered and nuclear levels of p53 and p21(waf1) protein were higher in the group of offspring that included F3 offspring with a paternal F0 radiation history than in littermates in the neo-positive control group. To our knowledge, this is the first observation in the descendants of paternal germline irradiation of effects on signal protein kinase activities and downstream nuclear target proteins that can influence cell proliferation rates.


Subject(s)
Gamma Rays , Protein Kinases/genetics , Protein Kinases/radiation effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Animals , Body Weight/radiation effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Cyclins/radiation effects , Enzyme Activation/radiation effects , Female , Liver/radiation effects , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Protein Kinases/metabolism , Radiation Chimera , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/radiation effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects
14.
Oncogene ; 19(39): 4480-90, 2000 Sep 14.
Article in English | MEDLINE | ID: mdl-11002421

ABSTRACT

The activation of cell cycle checkpoints in response to genotoxic stressors is essential for the maintenance of genomic integrity. Although most prior studies of cell cycle effects of UV irradiation have used UVC, this UV range does not penetrate the earth's atmosphere. Thus, we have investigated the mechanisms of ultraviolet B (UVB) irradiation-induced cell cycle arrest in a biologically relevant target cell type, the early stage human melanoma cell line, WM35. Irradiation of WM35 cells with UVB resulted in arrests throughout the cell cycle: at the G1/S transition, in S phase and in G2. G1 arrest was accompanied by increased association of p21 with cyclin E/cdk2 and cyclin A/cdk2, increased binding of p27 to cyclin E/cdk2 and inhibition of these kinases. A loss of Cdc25A expression was associated with an increased inhibitory phosphotyrosine content of cyclin E- and cyclin A-associated cdk2 and may also contribute to G1 arrest following UVB irradiation. The association of Cdc25A with 14-3-3 was increased by UVB. Reduced cyclin D1 protein and increased binding of p21 and p27 to cyclin D1/cdk4 complexes were also observed. The loss of cyclin D1 could not be attributed to inhibition of either MAPK or PI3K/PKB pathways, since both were activated by UVB. Cdc25B levels fell and the remaining protein showed an increased association with 14-3-3 in response to UVB. Losses in cyclin B1 expression and an increased binding of p21 to cyclin B1/cdk1 complexes also contributed to inhibition of this kinase activity, and G2/M arrest. Oncogene (2000) 19, 4480 - 4490.


Subject(s)
Cell Cycle/radiation effects , Melanoma/pathology , Muscle Proteins , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/radiation effects , Cyclins/metabolism , Cyclins/radiation effects , Humans , Melanoma/metabolism , Melanoma/radiotherapy , Microfilament Proteins/metabolism , Microfilament Proteins/radiation effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/radiation effects , Protein Kinases/metabolism , Protein Kinases/radiation effects , Proteins/metabolism , Proteins/radiation effects , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/radiation effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays , cdc25 Phosphatases/metabolism , cdc25 Phosphatases/radiation effects
15.
Oncogene ; 19(34): 3858-65, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10951579

ABSTRACT

We studied the ability of F9 teratocarcinoma cells to arrest in G1/S and G2/M checkpoints after gamma-irradiation. Wild-type p53 protein was rapidly accumulated in F9 cells after gamma-irradiation, however, this was followed not by a G1/S arrest but by a short and reversible delay of the cell cycle in G2/M. In order to elucidate the reasons of the lack of G1/S arrest in F9 cells, we investigated the expression of p53 downstream target Cdk inhibitor p21WAF1/CIP1. In spite of p53-dependent activation of p21WAF1/CIP1 gene promoter and p21WAF1/CIP1 mRNA accumulation upon irradiation, the p21WAF1/CIP1 protein was not detected by either immunoblot or immunofluorescence techniques. However, the cells treated with a specific proteasome inhibitor lactacystin revealed the p21WAF1/CIP1 protein both in non-irradiated and irradiated cells. Therefore we suggest that p21WAF1/CIP1 protein is degraded by a proteasome-dependent mechanism in F9 cells and the lack of G1/S arrest after gamma-irradiation is due to this degradation. We also examined the expression and activity of cell cycle regulatory proteins: G1- and G2-cyclins and cyclin-dependent kinases. In the absence of functional p21WAF1/CIP1 inhibitor, the activity of G1 cyclin/Cdk complexes was insufficiently inhibited to cause a G1 arrest, whereas a decrease of cdc2 and cyclin B1-associated kinase activities was enough to contribute to a reversible G2 arrest following gamma-irradiation. After gamma-irradiation, the majority of F9 cells undergo apoptosis implying that wt-p53 likely triggers pro-apoptotic gene expression in DNA damaged cells. Elimination of defected cells might ensure maintenance of genome integrity in the remaining cell population.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/physiology , Cell Cycle/radiation effects , Cyclins/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Protein p53/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Embryonal/pathology , Carcinoma, Embryonal/radiotherapy , Cyclin B/metabolism , Cyclin B1 , Cyclin D1/metabolism , Cyclin D1/radiation effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/radiation effects , Cyclins/genetics , Cyclins/radiation effects , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , DNA Damage/radiation effects , G1 Phase/radiation effects , Gamma Rays , Mice , Multienzyme Complexes/drug effects , Nocodazole/pharmacology , Promoter Regions, Genetic , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , S Phase/radiation effects , Tumor Cells, Cultured
16.
Cancer Res ; 60(10): 2623-31, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10825133

ABSTRACT

The effects of exposure to high and very low fluence alpha-particles on the G1 checkpoint were investigated in human diploid fibroblasts irradiated and released from density-inhibited confluent cultures by the use of the cumulative labeling index method. Transient and permanent arrests in G1 occurred in fibroblast populations exposed to mean doses as low as 1 cGy, suggesting that nontraversed bystander cells may contribute to the low dose response. In cells exposed to high fluences, the G1 checkpoint is at least as extensive as in gamma-irradiated cells. In contrast to gamma-irradiated cells, neither repair of potentially lethal damage nor a reduction in the fraction of cells transiently or permanently arrested in G1 were observed in cells held in confluence for 6 h after alpha-particle irradiation. Studies with isogenic wild-type, p53-/-, and p21Waf1-/- mouse embryo fibroblasts exposed to either gamma or alpha-particle radiation revealed a total lack of G1 arrest in either p53-/- or p21waf1-/- cells, indicating that the G1 checkpoint in wild-type cells is p53-dependent and that p21Wf1 fully mediates the role of p53 in its induction. In contrast to human cells, mouse embryo fibroblasts do not undergo a permanent G1 arrest. Except under conditions favoring potentially lethal damage repair, a comparable expression pattern of p53, p21Waf1, and other cell cycle-regulated proteins (pRb, p34cdc2, and cyclin B1) was observed in alpha-particle or gamma-irradiated human fibroblasts.


Subject(s)
Alpha Particles , Fibroblasts/radiation effects , G1 Phase/radiation effects , Animals , CDC2 Protein Kinase/biosynthesis , CDC2 Protein Kinase/radiation effects , Cell Line , Cell Survival , Cyclin B/biosynthesis , Cyclin B/radiation effects , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/radiation effects , DNA Replication/radiation effects , Fibroblasts/cytology , Gamma Rays , Gene Expression Regulation/radiation effects , Humans , Mice , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/radiation effects , Up-Regulation/radiation effects
17.
Oncogene ; 19(5): 649-60, 2000 Feb 03.
Article in English | MEDLINE | ID: mdl-10698510

ABSTRACT

The mechanisms by which the p53 tumour suppressor protein would, in vivo, co-ordinate the adaptive response to genotoxic stress is poorly understood. p53 has been shown to transactivate several genes that could be involved in two main cellular responses, growth arrest and apoptosis. To get further insight into the tissue-specific regulation of p53 transcriptional activity, we performed an extensive study looking at the expression of four well characterized p53-responsive genes, before and after gamma-irradiation in p53 wild-type (p53+/+) and p53-deficient (p53-/-) mice. The waf1, bax, fas and mdm2 genes were chosen for their different potential roles in the cellular response to stress. Our data demonstrate the strict p53-dependence of mRNA up-regulation for bax, fas and mdm2 in irradiated tissues and confirm such findings for waf1. They further highlight complex levels of regulatory mechanisms that could lead, in vivo, to selective transcriptional activation of genes by p53. In addition, our results provide arguments for the involvement of p53 in the basal mRNA expression of the four genes in some organs. Finally, in situ expression of Bax and p21Waf-1 protein suggests, at least in lymphoid organs, a direct correlation between selective p53-target gene expression and a particular response of a cell to ionising radiation.


Subject(s)
Cyclins/genetics , Gamma Rays , Nuclear Proteins , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/physiology , fas Receptor/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/radiation effects , Immunohistochemistry , Lymphoid Tissue/metabolism , Lymphoid Tissue/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/radiation effects , Oxidative Stress/radiation effects , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/radiation effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/radiation effects , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/biosynthesis , RNA, Messenger/radiation effects , Tumor Suppressor Protein p53/deficiency , bcl-2-Associated X Protein , fas Receptor/radiation effects
18.
Mol Carcinog ; 27(2): 141-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10657906

ABSTRACT

In order to investigate the hypothesis that aberrant expression of cell-cycle regulatory proteins may represent early events in the process of carcinogenesis, levels of expression of the negative regulators p21(waf1/cip1) (p21), p27(kip1) (p27), and p16(ink4a) (p16) and/or the positive regulators cyclin D(1) and cyclin E were examined by western blot analysis in cells transformed in vitro by ionizing radiation. The levels of these proteins in 12 independently derived mouse 10T(1/2) cell clones transformed by 1.5 Gy of alpha radiation were compared with those in nine similarly derived nontransformed control clones. Constitutive levels of p21 were very low in all control clones, whereas p21 expression was significantly elevated in nine of 12 transformed clones. Two of the three transformed clones displaying low levels of p21 expressed increased levels of p53. p21 regulation was also altered in response to radiation in transformed clones as compared with controls, only minimal induction was observed 4 h following gamma irradiation. Western blot analysis indicated a constant expression of p27 protein but slightly decreased levels of p16 in these transformed clones. Cyclin D(1) was overexpressed in 11 of 12 transformed clones; in only two of these were the levels of cyclin E elevated. Overall, the results suggest that alterations in the expression of cell cycle regulatory proteins may represent important events in radiation-induced oncogenic transformation in vitro. Although the specific alterations vary among different transformed clones, overexpression and aberrant regulation of p21 appear to be the most frequent ones.


Subject(s)
Alpha Particles , Cell Cycle Proteins , Cell Transformation, Neoplastic/radiation effects , Clone Cells/radiation effects , Cyclins/biosynthesis , Tumor Suppressor Proteins , Animals , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Clone Cells/metabolism , Cyclin D1/biosynthesis , Cyclin D1/radiation effects , Cyclin E/biosynthesis , Cyclin E/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/radiation effects , Embryo, Mammalian , Fibroblasts , Mice , Mice, Inbred C3H , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/radiation effects
19.
Oncogene ; 18(42): 5795-805, 1999 Oct 14.
Article in English | MEDLINE | ID: mdl-10523860

ABSTRACT

The tumor suppressor protein, p53, plays a critical role as a transcriptional activator of downstream target genes involved in the cellular response to DNA damaging agents. We examined the cell cycle checkpoint response of human mammary epithelial cells (HMEC) and their isogenic fibroblast counterparts to ionizing (IR) and ultraviolet (UV) radiation, two genotoxic agents whose DNA damage response pathways involve p53. Using flow cytometric analysis, we found that both mortal and immortalized HMEC, which contain wild-type p53 sequence, do not exhibit a G1 arrest in response to IR, but show an intact G2 checkpoint. Supportive evidence from Western analyses revealed that there was neither an increase in p53 nor one of its downstream targets, p21WAF1, in HMEC exposed to IR. In contrast, isogenic mammary fibroblasts arrest at the G1 checkpoint and induce the p53 and p21WAF1 proteins following IR. By comparison, HMEC exposed to UV displayed an S phase arrest and induced the expression of p53 and p21WAF1. Our results show that the cellular response to DNA damage depends on both the type of damage introduced into the DNA and the specific cell type.


Subject(s)
Breast/cytology , Epithelial Cells/radiation effects , Gamma Rays , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays , Breast/radiation effects , Cell Cycle/radiation effects , Cell Line, Transformed , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/physiology , Cyclins/radiation effects , Female , Fibroblasts/radiation effects , G1 Phase/radiation effects , Humans , Radiation Tolerance , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...