Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Aquat Toxicol ; 260: 106584, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37267806

ABSTRACT

Estrogenic endocrine disrupting chemicals (EEDC) have been suspected to impact offspring in a transgenerational manner via modifications of the germline epigenome in the directly exposed generations. A holistic assessment of the concentration/ exposure duration-response, threshold level, and critical exposure windows (parental gametogenesis and embryogenesis) for the transgenerational evaluation of reproduction and immune compromise concomitantly will inform the overall EEDC exposure risk. We conducted a multigenerational study using the environmental estrogen, 17α-ethinylestradiol (EE2), and the marine laboratory model fish Oryzias melastigma (adult, F0) and their offspring (F1-F4) to identify transgenerationally altered offspring generations and phenotype persistence. Three exposure scenarios were used: short parental exposure, long parental exposure, and a combined parental and embryonic exposure using two concentrations of EE2 (33ng/L, 113ng/L). The reproductive fitness of fish was evaluated by assessing fecundity, fertilization rate, hatching success, and sex ratio. Immune competence was assessed in adults via a host-resistance assay. EE2 exposure during both parental gametogenesis and embryogenesis was found to induce concentration/ exposure duration-dependent transgenerational reproductive effects in the unexposed F4 offspring. Furthermore, embryonic exposure to 113 ng/L EE2 induced feminization of the directly exposed F1 generation, followed by subsequent masculinization of the F2 and F3 generations. A sex difference was found in the transgenerationally impaired reproductive output with F4 females being sensitive to the lowest concentration of EE2 (33 ng/L) upon long-term ancestral parent exposure (21 days). Conversely, F4 males were affected by ancestral embryonic EE2 exposure. No definitive transgenerational impacts on immune competence were identified in male or female offspring. In combination, these results indicate that EEDCs can be transgenerational toxicants that may negatively impact the reproductive success and population sustainability of fish populations.


Subject(s)
Oryzias , Water Pollutants, Chemical , Animals , Female , Male , Oryzias/physiology , Genetic Fitness , Water Pollutants, Chemical/toxicity , Reproduction , Fertility , Ethinyl Estradiol/toxicity
2.
Microsc Res Tech ; 79(7): 657-63, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27151371

ABSTRACT

The description and application of a modified Scanning Electron Microscope preparation technique using hexamethyldisilazane for small parasitic copepods was demonstrated though a high resolution depiction of individuals of Ergasilus labracis sampled from three spined stickleback (Gasterosteus aculeatus) in Bay D'Espoir, Newfoundland during summer 2015 and from archival samples retrieved from Atlantic salmon par (Salmo salar) stored at the Atlantic reference centre, St. Andrews, New Brunswick. The specimens were very well preserved showing high quality detail of important features and verifying those previously described using light microscopy by Hogans. Additionally the technique allowed excellent in situ demonstrations of mouth parts, swimming legs, and unusual and previously undescribed features of the second antenna including prominent striations and pore-like structures found to define the claw. It is thought that this technique will become a quick and efficient tool for describing important taxonomic features of small parasitic copepods like E. labracis or other similar small aquatic organisms. Microsc. Res. Tech. 79:657-663, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Copepoda/ultrastructure , Microscopy, Electron, Scanning/methods , Organosilicon Compounds/chemistry , Animals , Arthropod Antennae/ultrastructure , Female , Mouth/ultrastructure
3.
Exp Cell Res ; 314(8): 1823-30, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18394600

ABSTRACT

Mammalian stanniocalcin-2 (STC2) is a secreted glycoprotein hormone with a putative role in unfolded protein response and apoptosis. Here we reported that STC2 expression was sporadically abrogated in human cancer cells by transcriptional silencing associated with CpG island promoter hypermethylation. Direct sequencing of bisulfite-modified DNA from a panel of seven human cancer cell lines revealed that CpG dinucleotides in STC2 promoter was methylated in human ovarian epithelial cancer (SKOV3, OVCAR3 and CaOV3), pancreatic cancer (BxP3), colon adenoma (HT29), and leukemia (Jurkat cells). STC2 CpG island hypermethylation was accompanied with a low basal STC2 expression level. Treatment of these cancer cells with 5-aza-2'-deoxycytidine (5-aza-CdR), an inhibitor of DNA methylation significantly induced STC2 expression. Using SKOV3 cells as a model, the link between DNA demethylation and STC2 expression was consistently demonstrated with hydralazine treatment, which was shown to reduce the protein level of DNA methyltransferase 1 (DNMT1) but stimulated STC2 expression. Two human normal surface ovarian cell-lines (i.e. IOSE 29 and 398) showed no methylation at CpG dinucleotides in the examined promoter region and were accompanied with high basal STC2 levels. Hypoxia stimulated STC2 expression in SKOV3 cells was markedly increased in 5-aza-CdR pretreated cells, showing that DNA methylation may hinder the HIF-1 mediated activation. To elucidate this possibility, RNA interference studies confirmed that endogenous HIF-1 alpha was a key factor for STC2 gene activation as well as in the synergistic induction of STC2 expression in 5-aza-CdR pretreated cells. Chromatin immunoprecipitation (ChIP) assay demonstrated the binding of HIF-1 alpha to STC2 promoter. The binding was increased in 5-aza-CdR pretreated cells. Collectively, this is the first report to show that STC2 was aberrantly hypermethylated in human cancer cells. The findings demonstrated that STC2 epigenetic inactivation may interfere with HIF-1 mediated activation of STC2 expression.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Glycoproteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Hypoxia , Cell Line, Tumor , CpG Islands , DNA Methylation , Decitabine , Glycoproteins/metabolism , Humans , Hydralazine/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , RNA Interference , Transcriptional Activation
4.
Oncogene ; 27(29): 4065-74, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18332866

ABSTRACT

Genotoxic stress activates the phosphatidylinositol 3-kinase-like kinases (PIKKs) that phosphorylate proteins involved in cell cycle arrest, DNA repair and apoptosis. Previous work showed that the PIKK ataxia telangiectasia mutated (ATM) but not ATM and Rad3 related phosphorylates p53 (Ser15) during hyperoxia, a model of prolonged oxidative stress and DNA damage. Here, we show hSMG-1 is responsible for the rapid and early phosphorylation of p53 (Ser15) and that ATM helps maintain phosphorylation after 24 h. Despite reduced p53 phosphorylation and abundance in cells depleted of hSMG-1 or ATM, levels of the p53 target p21 were still elevated and the G(1) checkpoint remained intact. Conditional overexpression of p21 in p53-deficient cells revealed that hyperoxia also stimulates wortmannin-sensitive degradation of p21. siRNA depletion of hSMG-1 or ATM restored p21 stability and the G(1) checkpoint during hyperoxia. These findings establish hSMG-1 as a proximal regulator of DNA damage signaling and reveal that the G(1) checkpoint is tightly regulated during prolonged oxidative stress by both PIKK-dependent synthesis and proteolysis of p21.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , G1 Phase/physiology , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Androstadienes/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/physiology , DNA Repair/drug effects , DNA Repair/physiology , DNA-Binding Proteins/genetics , G1 Phase/drug effects , Humans , Hyperoxia/genetics , Hyperoxia/metabolism , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Wortmannin
5.
Bioresour Technol ; 99(14): 6097-104, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18243690

ABSTRACT

Lipase-catalyzed production of palm esters by alcoholysis of palm oil with oleyl alcohol in n-hexane was performed in 2L stirred-tank reactor (STR). Investigation on the performance of reactor operation was carried out in batch mode STR with single impeller mounted on the centrally located shaft. Rushton turbine (RT) impellers provide the highest reaction yield (95.8%) at lower agitation speed as compared to AL-hydrofoil (AL-H) and 2-bladed elephant ear (EE) impellers. Homogenous enzyme particles suspension was obtained at 250 rpm by using RT impeller. At higher impeller speed, the shear effect on the enzyme particles caused by agitation has decreased the reaction performance. Palm esters reaction mixture in STR follows Newtons' law due to the linear relation between the shear stress (tau) and shear rate (dupsilon/dy). High stability of Lipozyme RM IM was observed as shown by its ability to be repeatedly used to give high percentage yield (79%) of palm esters even after 15 cycles of reaction. The process was successfully scale-up to 75 L STR (50 L working volume) based on a constant impeller tip speed approach, which gave the yield of 97.2% after 5h reaction time.


Subject(s)
Esters/metabolism , Lipase/metabolism , Catalysis , Chromatography, Gas , Rheology , Viscosity
6.
Biochem Biophys Res Commun ; 356(4): 968-75, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17395153

ABSTRACT

There is growing evidence to suggest that altered patterns of STC1 gene expression relate to the process of human cancer development. Our previous study has demonstrated the involvement of HIF-1 in the regulation of STC1 expression in human cancer cells. Recently, STC1 has been implicated as a putative pro-apoptotic factor in regulating the cell-death mechanism. Thus it would be of interest to know if STC1 is regulated by a tumor suppressor protein, p53. In this study, we provide evidence to demonstrate that the induction of STC1 expression in apoptotic human nasopharyngeal cancer cells (CNE2) is mediated by the activation of p53. Our study indicated that the activation of STC1 and heat-shock protein (hsp70) accompanied iodoacetamide (IDAM)-induced apoptosis in CNE-2. In addition, cellular events such as GSH depletion, mitochondrial membrane depolarization, reduction of pAkt and procaspase-3, and the induction of total p53 protein, acetylated p53, and annexin V positive cells were observed. The activation of STC1 was found to be at the transcriptional level and was independent of prior protein synthesis. Co-treatment of IDAM exposed cells with N-acetyl cysteine (NAC) prevented cell death by restoring mitochondrial membrane potential and cellular levels of GSH. NAC co-treatment also suppressed STC1 expression but had no effect on IDAM-induced hsp70 expression. RNA interference studies demonstrated that endogenous p53 was involved in activating STC1 gene expression. Collectively, the present findings provide the first evidence of p53 regulation of STC1 expression in human cancer cells.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Signal Transduction
7.
Oncogene ; 25(4): 622-32, 2006 Jan 26.
Article in English | MEDLINE | ID: mdl-16247478

ABSTRACT

The p53 protein has been implicated in multiple cellular responses related to DNA damage. Alterations in any of these cellular responses could be related to increased genomic instability. Our previous study has shown that mutations in p53 lead to hypermutability to ionizing radiation. To investigate further how p53 is involved in regulating mutational processes, we used 8K cDNA microarrays to compare the patterns of gene expression among three closely related human cell lines with different p53 status including TK6 (wild-type p53), NH32 (p53-null), and WTK1 (mutant p53). Total RNA samples were collected at 1, 3, 6, 9, and 24 h after 10 Gy gamma-irradiation. Template-based clustering analysis of the gene expression over the time course showed that 464 genes are either up or downregulated by at least twofold following radiation treatment. In addition, cluster analyses of gene expression profiles among these three cell lines revealed distinct patterns. In TK6, 165 genes were upregulated, while 36 genes were downregulated. In contrast, in WTK1 75 genes were upregulated and 12 genes were downregulated. In NH32, only 54 genes were upregulated. Furthermore, we found several genes associated with DNA repair namely p53R2, DDB2, XPC, PCNA, BTG2, and MSH2 that were highly induced in TK6 compared to WTK1 and NH32. p53R2, which is regulated by the tumor suppressor p53, is a small subunit of ribonucleotide reductase. To determine whether it is involved in radiation-induced mutagenesis, p53R2 protein was inhibited by siRNA in TK6 cells and followed by 2 Gy radiation. The background mutation frequencies at the TK locus of siRNA-transfected TK6 cells were about three times higher than those seen in TK6 cells. The mutation frequencies of siRNA-transfected TK6 cells after 2 Gy radiation were significantly higher than the irradiated TK6 cells without p53R2 knock down. These results indicate that p53R2 was induced by p53 protein and is involved in protecting against radiation-induced mutagenesis.


Subject(s)
DNA Repair , Mutation , Transcription, Genetic/radiation effects , Tumor Suppressor Protein p53/physiology , Cells, Cultured , DNA Damage , Gene Expression/radiation effects , Gene Expression Profiling , Humans , NF-kappa B/metabolism , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
8.
Endocrinology ; 146(11): 4951-60, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16109785

ABSTRACT

Stanniocalcin-1 (STC1) is an endocrine hormone originally discovered in the corpuscles of Stannius, endocrine glands on kidneys of bony fishes, and also has been identified in mammals. The mammalian STC1 gene is widely expressed in various tissues and appears to be involved in diverse biological processes. There is growing evidence to suggest that altered patterns of gene expression have a role in human cancer development. Recently STC1 has been identified as a stimulator of mitochondrial respiration and has been hypothesized to be functionally related to the Warburg effect, of which hypoxia-inducible factor (HIF)-1 plays a key role in reprogramming tumor metabolism. This prompted us to examine the involvement of HIF-1 in the regulation of STC1 expression in tumor hypoxia. Our data reveal that hypoxia can stimulate STC1 gene expression in various human cancer cell lines, including those derived from colon carcinomas, nasopharyngeal cancer (CNE-2, HONE-1, HK-1), and ovarian cancer (CaOV3, OVCAR3, SKOV3). By far, the greatest response was observed in CNE-2 cells. In further studies on CNE-2 cells, desferrioxamine, cobalt chloride, and O(2) depletion all increased HIF-1alpha protein and STC1 mRNA levels. Desferrioxamine treatment, when coupled with Fe replenishment, abolished these effects. RNA interference studies further confirmed that endogenous HIF-1alpha was a key factor in hypoxia-induced STC1 expression. The ability of vascular endothelial growth factor to stimulate STC1 expression in CNE-2 cells was comparatively low. Collectively, the present findings provide the first evidence of HIF-1 regulation of STC1 expression in human cancer cells. The studies have implications as to the role of STC1 in hypoxia induced adaptive responses in tumor cells.


Subject(s)
Colonic Neoplasms/metabolism , Glycoproteins/metabolism , Nasopharyngeal Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Deferoxamine/pharmacology , Female , Ferric Compounds/pharmacology , Ferricyanides/pharmacology , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Humans , Hypoxia/chemically induced , Hypoxia/metabolism , Iron Chelating Agents/pharmacology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , RNA, Messenger/biosynthesis , RNA, Small Interfering , Transfection
9.
Am J Clin Oncol ; 24(5): 432-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11586091

ABSTRACT

The objective of this study was to apply preclinical research of paclitaxel radiosensitization to the treatment of thoracic malignancy. Human lung cancer cell line NCI520 and epidermoid cell line A431 were investigated in vitro for radiosensitizing effects of paclitaxel. Optimal schedule of paclitaxel treatment was applied to a clinical protocol as well as off-protocol treatment of thoracic malignancy. Pulsed paclitaxel with concurrent once-daily radiation was delivered every 48 hours during the week using doses of 15 mg/m2, 20 mg/m2, or 25 mg/m2 in a phase I clinical trial of dose escalation. Preclinical data support the finding that low-dose paclitaxel is sufficient for radiosensitization. Data also support that delaying radiation is better than immediate radiation after drug treatment. Twenty-three patients have enrolled in the phase I clinical trial. Seventeen patients completed treatment (6 at 15 mg/m2; 5 at 20 mg/m2; and 6 at 25 mg/m2). Mean tumor shrinkage at 4 to 6 weeks after therapy was 82%, 84%, and 84% for dose levels I, II, and III, respectively [average primary tumor shrinkage was 83% +/- 8% (95% C.I.)]. Locoregional tumor response rate was 100% [12% (2/17) complete response and 88% (15/17) partial response] with low rates of toxicity. It is concluded that pulsed low-dose paclitaxel and radiation is a very effective and well-tolerated regimen for thoracic malignancy.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Paclitaxel/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Adult , Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Drug Administration Schedule , Female , Humans , Male , Mesothelioma/drug therapy , Mesothelioma/radiotherapy , Middle Aged , Paclitaxel/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Radiotherapy Dosage , Tumor Cells, Cultured
10.
Am J Clin Oncol ; 24(5): 486-90, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11586101

ABSTRACT

Mutations in the p53 tumor suppressor gene have been found in more than 50% of human tumors including those in breast, colon, lung, and oral cavity. However, the significance of p53 mutation in radiation sensitivity and its underlying mechanisms still remains unclear. In this study, we have measured the effects of p53 mutation on cell cycle delay, apoptosis, and radiation sensitivity using mouse cells transfected with different forms of p53 mutations. Wild-type p53 and p53-Null mouse embryo fibroblast cells were used as positive and negative controls, respectively. Exponentially growing cells were irradiated with 0- to 9-Gy gamma rays and then assayed for cell survival, p53 expression, cell cycle checkpoint, and apoptosis. Cell survivals determined by clonogenic assay show that p53 mutant cells are generally more sensitive to ionizing radiation than cells with wild-type p53. Western blot analysis indicates that exposure to 6-Gy gamma rays increases the p53 expression levels by two- to threefold in wild-type p53 cells. However, the p53 level remains unchanged in cells with mutant p53 during the same postirradiation period. Irradiation with 6-Gy gamma rays produces G2/M arrest in all cell lines, indicating that p53 is probably not involved in the G2/M checkpoint. However, all mutant cells fail to show any significant G1/S arrest after irradiation, suggesting that G1/S arrest may be implicated in radiation sensitivity. Finally, there is very little apoptosis (<3% by Tat-mediated dUTP nick-end labeling [TUNNEL] and morphologic assays) detected in wild-type and p53 mutant cell lines after 6-Gy gamma rays. Our results suggest that mutant forms of p53 represent a phenotype that affects the radiation sensitivity and is not dependent on the apoptotic pathway.


Subject(s)
Apoptosis/genetics , Apoptosis/radiation effects , Cell Cycle/genetics , Cell Cycle/radiation effects , Genes, p53 , Mutation , Radiation Tolerance , Radiation, Ionizing , Animals , Cell Line , Cell Survival/genetics , Cell Survival/radiation effects , Fibroblasts , Mice , Transfection
11.
Cancer Genet Cytogenet ; 129(2): 131-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566343

ABSTRACT

To determine the pathogenic role of chromosomes 11 and 17 in the carcinogenesis of human ovarian cancers, neo(R)-tagged chromosome 11 or 17 was transferred from cell lines A9H11 or A9H17, respectively, into the ovarian carcinoma cell line SKOV-3 using microcell-mediated chromosome transfer. The chromosome transfer was verified by polymerase chain reaction detection of the neo(R) gene, fluorescence in situ hybridization detection of an extra chromosome 11, and microsatellite polymorphism detection of an exogeneous chromosome 11. Five SKOV-3/A9H11 hybrids and five SKOV-3/A9H17 hybrid clones were generated. For the chromosome 11 transfer, complete suppression of tumorigenicity was observed in four clones, (11)9-8 and 11(H)7-2, 11(H)8-3, and 11(H)7-2, 100 days post implantation. For the chromosome 17 transfer, no complete suppression of tumorigenicity was observed. However, an increased latency period ranging from 25 to 49 days in contrast to 7 days for the SKOV-3 parental line, and a significant reduction in tumor size was observed. There was no correlation between the in vitro growth rate and the tumorigenicity or length of latency period. Our results demonstrate functionally that chromosome 11 may carry a tumor suppressor gene(s) while chromosome 17 may carry a tumor growth-inhibitor gene(s) for the ovarian carcinoma cell line, SKOV-3.


Subject(s)
Carcinoma/genetics , Chromosomes, Human, Pair 11/genetics , Gene Transfer Techniques , Genes, Tumor Suppressor/genetics , Ovarian Neoplasms/genetics , Animals , Cell Division/genetics , Chromosomes, Human, Pair 17/genetics , Clone Cells , Disease Progression , Female , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, SCID , Neoplasm Transplantation , Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured
12.
Am J Respir Cell Mol Biol ; 24(6): 703-10, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11415935

ABSTRACT

The lung is a major target tissue for oxidative stress, including hyperoxia used to relieve tissue hypoxia. Unfortunately, severe hyperoxia damages DNA, inhibits proliferation, and kills cells, resulting in morbidity and mortality. Although hyperoxia induces the tumor suppressor p53 and its downstream target, the cyclin-dependent kinase inhibitor p21(Cip1/WAF1/Sdi1) (p21), their role in pulmonary injury remains unknown. Using p53- and p21-deficient mice we demonstrate that hyperoxia induces p21 in the absence of p53, suggesting that previous conclusions that p53 does not modify hyperoxic lung injury cannot be extrapolated to p21. In fact, mean survival of p21-deficient mice decreased by 40% and was associated with terminal deoxyribonucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick-end labeling staining of alveolar debris, indicative of DNA fragmentation and cell death. Ultrastructural analyses revealed that alveolar endothelial and type I epithelial cells died rapidly by necrosis. Although hyperoxia decreased DNA replication in p21-wild-type lungs, it had no effect on replication in p21-deficient lungs. Our findings suggest that p21 protects the lung from oxidative stress, in part, by inhibiting DNA replication and thereby allowing additional time to repair damaged DNA. Our findings have implications for patients suffering from the toxic effects of supplemental oxygen therapies.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Hyperoxia/metabolism , Lung/metabolism , Oxidative Stress/physiology , Animals , Cell Death , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Damage , DNA Fragmentation , DNA Replication , Male , Mice , Mice, Mutant Strains , Pulmonary Alveoli/metabolism , Respiratory Mucosa/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 280(4): L617-26, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238001

ABSTRACT

Previous studies have shown that hyperoxia inhibits proliferation and increases the expression of the tumor suppressor p53 and its downstream target, the cyclin-dependent kinase inhibitor p21(CIP1/WAF1), which inhibits proliferation in the G1 phase of the cell cycle. To determine whether growth arrest was mediated through activation of the p21-dependent G1 checkpoint, the kinetics of cell cycle movement during exposure to 95% O2 were assessed in the Mv1Lu and A549 pulmonary adenocarcinoma cell lines. Cell counts, 5-bromo-2'-deoxyuridine incorporation, and cell cycle analyses revealed that growth arrest of both cell lines occurred in S phase, with A549 cells also showing evidence of a G1 arrest. Hyperoxia increased p21 in A549 but not in Mv1Lu cells, consistent with the activation of the p21-dependent G1 checkpoint. The ability of p21 to exert the G1 arrest was confirmed by showing that hyperoxia inhibited proliferation of HCT 116 colon carcinoma cells predominantly in G1, whereas an isogenic line lacking p21 arrested in S phase. The cell cycle arrest in S phase appears to be a p21-independent process caused by a gradual reduction in the rate of DNA strand elongation. Our data reveal that hyperoxia inhibits proliferation in G1 and S phase and demonstrate that p53 and p21 retain their ability to affect G1 checkpoint control during exposure to elevated O2 levels.


Subject(s)
Cyclins/physiology , Hyperoxia/pathology , Hyperoxia/physiopathology , Adenosine Triphosphate/metabolism , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , G1 Phase/physiology , G1 Phase/radiation effects , Gamma Rays , Hyperoxia/genetics , Hyperoxia/metabolism , Phosphorylation , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
14.
Biochem Biophys Res Commun ; 276(2): 724-8, 2000 Sep 24.
Article in English | MEDLINE | ID: mdl-11027538

ABSTRACT

Although circadian variations in hematopoiesis have been well documented, the molecular mechanism of the circadian rhythms remains elusive. To determine if a clock system exists in bone marrow to mediate the circadian rhythms, we analyzed the expression of mPer1 and mPer2, both mouse homologues of the Drosophila period gene and known regulators of the clock system, in murine bone marrow by relative quantitative reverse transcriptase-polymerase chain reaction (RT-PCR). We demonstrated that both genes were expressed in bone marrow. Furthermore, the expression patterns of mPer1 and mPer2 in total bone marrow cells exhibited two peaks over a 24-h period. In contrast, the expression patterns of these two genes in the Gr-1-positive cells isolated from bone marrow mainly contributed to one of the two peaks. These results indicate that a clock system exists in bone marrow and suggest that the circadian rhythms in bone marrow are lineage- and/or differentiation stage-dependent.


Subject(s)
Biological Clocks/genetics , Bone Marrow/metabolism , Gene Expression Regulation , Nuclear Proteins/genetics , Analysis of Variance , Animals , Cell Cycle Proteins , Circadian Rhythm/genetics , Female , Male , Mice , Mice, Inbred BALB C , Nuclear Proteins/biosynthesis , Period Circadian Proteins , Transcription Factors
15.
Br J Cancer ; 82(6): 1223-32, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10735510

ABSTRACT

Over the past several years, our group has provided considerable evidence that the expression of sigma-2 (sigma2) receptors may serve as a biomarker of tumour cell proliferation. In these in vitro studies, sigma2 receptors were expressed 8-10 times more in proliferative (P) tumour cells than in quiescent (Q) tumour cells, and the extent and kinetics of their expression were independent of a number of biological, physiological and environmental factors often found in solid tumours. Moreover, the expression of sigma2 receptors followed both the population growth kinetics when Q-cells were recruited into the P-cell compartment and the proliferative status of human breast tumour cells treated with cytostatic concentrations of tamoxifen. However, these in vitro studies may or may not be indicative of what might occur in solid tumours. In the present study, the sigma2 receptor P:Q ratio was determined for the cells from subcutaneous 66 (diploid) and 67 (aneuploid) tumours grown in female nude mice. The sigma2 receptor P:Q ratio of the 66 tumours was 10.6 compared to the sigma2 receptor P:Q ratio of 9.5 measured for the 66 tissue culture model. The sigma2 receptor P:Q ratio of the 67 tumours was 4.5 compared to the sigma2 receptor P:Q ratio of approximately equal 8 measured for the 67 tissue culture model. The agreement between the solid tumour and tissue culture data indicates that: (1) the expression of sigma2 receptors may be a reliable biomarker of the proliferative status of solid tumours and (2) radioligands with both high affinity and high selectivity for sigma2 receptors may have the potential to non-invasively assess the proliferative status of human solid tumours using imaging techniques such as positron emission tomography or single-photon emission computerized tomography.


Subject(s)
Biomarkers, Tumor/analysis , Mammary Neoplasms, Animal/genetics , Receptors, sigma/genetics , Animals , Cell Division , Female , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/physiopathology , Mice , Neoplasm Invasiveness , Prognosis , Receptors, sigma/physiology , Tumor Cells, Cultured
16.
Br J Cancer ; 82(4): 937-44, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10732769

ABSTRACT

We have previously shown that BALB/c-derived EMT6 mammary tumours transfected with interleukin (IL)-2 have decreased hypoxia compared to parental tumours, due to increased vascularization. Since hypoxia is a critical factor in the response of tumours to radiation treatment, we compared the radiation response of IL-2-transfected tumours to that of parental EMT6 tumours. Because the IL-2 tumours have an altered host cell composition, which could affect the interpretation of radiation sensitivity as measured by clonogenic cells, we employed flow cytometric analysis to determine the proportion of tumour cells vs host cells in each tumour type. Using this approach, we were able to correct the plating efficiency based on the number of actual tumour cells derived from tumours, making the comparison of the two types of tumours possible. We also excluded the possibility that cytotoxic T-cells present in EMT6/IL-2 tumours could influence the outcome of the clonogenic cell survival assay, by demonstrating that the plating efficiency of cells derived from EMT6/IL-2 tumours remained unchanged after depletion of Thy-1+ cells. The in vivo radiation response results demonstrated that IL-2-transfected tumours were more sensitive to radiation than parental EMT6 tumours. The hypoxic fraction of the EMT6/IL-2 tumours growing in vivo was markedly decreased relative to parental EMT6 tumours thus the increased sensitivity results from the increased vascularity we have previously observed in these tumours. These results indicate the potential therapeutic benefit of combining radiation and immunotherapy in the treatment of tumours.


Subject(s)
Gene Transfer Techniques , Interleukin-2/genetics , Radiation Tolerance/genetics , Animals , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology
17.
Am J Physiol Lung Cell Mol Physiol ; 278(3): L552-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10710528

ABSTRACT

Previous studies have shown that lungs of adult mice exposed to >95% oxygen have increased terminal deoxyribonucleotidyltransferase dUTP nick end-label staining and accumulate p53, the expression of which increases in cells exposed to DNA-damaging agents. The present study was designed to determine whether hyperoxia also increased expression of the growth arrest and DNA damage (GADD) gene 45 and GADD153, which are induced by genotoxic stress through p53-dependent and -independent pathways. GADD proteins have been shown to inhibit proliferation and stimulate DNA repair and/or apoptosis. GADD45 and GADD153 mRNAs were not detected in lungs exposed to room air but were detected after 48 and 72 h of exposure to hyperoxia. In situ hybridization and immunohistochemistry revealed that hyperoxia increased GADD45 and GADD153 expression in the bronchiolar epithelium and GADD45 expression predominantly in alveolar cells that were morphologically consistent with type II cells. Hyperoxia also increased GADD expression in p53-deficient mice. Terminal deoxyribonucleotidyltransferase dUTP nick end-label staining of lung cells from p53 wild-type and p53-null mice exposed to hyperoxia for 48 h revealed that hyperoxia-induced DNA fragmentation was not modified by p53 deficiency. These studies are consistent with the hypothesis that hyperoxia-induced DNA fragmentation is associated with the expression of GADD genes that may participate in DNA repair and/or apoptosis.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/genetics , Gene Expression Regulation , Hyperoxia/genetics , Lung/physiology , Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/physiology , Animals , DNA Fragmentation , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Proteins/metabolism , RNA, Messenger/metabolism , Transcription Factor CHOP , GADD45 Proteins
18.
Am J Physiol ; 277(6): L1172-8, 1999 12.
Article in English | MEDLINE | ID: mdl-10600888

ABSTRACT

High concentrations of O(2) inhibit epithelial cell proliferation that resumes on recovery in room air. To determine whether growth arrest is mediated by transforming growth factor-beta (TGF-beta), changes in cell proliferation during exposure to hyperoxia were assessed in the mink lung epithelial cell line Mv1Lu and the clonal variant R1B, which is deficient for the type I TGF-beta receptor. Mv1Lu cells treated with TGF-beta accumulated in the G(1) phase of the cell cycle as determined by propidium iodide staining, whereas proliferation of R1B cells was unaffected by TGF-beta. In contrast, hyperoxia inhibited proliferation of both cell lines within 24 h of exposure through an accumulation in the S phase. Mv1Lu cells treated with TGF-beta and exposed to hyperoxia accumulated in the G(1) phase, suggesting that TGF-beta can inhibit the S phase accumulation observed with hyperoxia alone. Cyclin A was detected in cultures exposed to room air or growth arrested by hyperoxia while decreasing in cells growth arrested in the G(1) phase by TGF-beta. Finally, hyperoxia failed to activate a TGF-beta-dependent transcriptional reporter in both Mv1Lu and R1B cells. These findings reveal that simple growth arrest by hyperoxia involves a defect in S phase progression that is independent of TGF-beta signaling.


Subject(s)
Epithelial Cells/cytology , Lung/cytology , Oxygen/pharmacology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Adenocarcinoma , Animals , Cell Division/drug effects , Cell Division/physiology , Epithelial Cells/drug effects , Flow Cytometry , G1 Phase/drug effects , Gene Expression/drug effects , Genes, Reporter , Luciferases/genetics , Lung Neoplasms , Mink , S Phase/drug effects , S Phase/physiology , Transcription, Genetic/drug effects , Tumor Cells, Cultured
19.
Br J Cancer ; 81(6): 925-33, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10576647

ABSTRACT

Recently, we demonstrated that sigma-2 receptors may have the potential to be a biomarker of tumour cell proliferation (Mach et al (1997) Cancer Res 57: 156-161). If sigma-2 receptors were a biomarker of tumour cell proliferation, they would be amenable to detection by non-invasive imaging procedures, thus eliminating many of the problems associated with the flow cytometric measures of tumour cell proliferation presently used in the clinic. To be a good biomarker of tumour cell proliferation, the expression of sigma-2 receptors must be essentially independent of many of the biological, physiological, and/or environmental properties that are found in solid tumours. In the investigation reported here, the mouse mammary adenocarcinoma lines, 66 (diploid) and 67 (aneuploid), 9L rat brain tumour cells, and MCF-7 human breast tumour cells were used to study the extent and kinetics of expression of sigma-2 receptors in proliferative (P) and quiescent (Q) tumour cells as a function of species, cell type, ploidy, pH, nutrient depletion, metabolic state, recruitment from the Q-cell compartment to the P-cell compartment, and treatment with tamoxifen. In these experiments, the expression of sigma-2 receptors solely reflected the proliferative status of the tumour cells. None of the biological, physiological, or environmental properties that were investigated had a measurable effect on the expression of sigma-2 receptors in these model systems. Consequently, these data suggest that the proliferative status of tumours and normal tissues can be non-invasively assessed using radiolabelled ligands that selectively bind sigma-2 receptors.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Receptors, sigma/biosynthesis , Tamoxifen/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/analysis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Division , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Ploidies , Rats , Receptors, sigma/analysis , Tumor Cells, Cultured
20.
Br J Cancer ; 79(9-10): 1372-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10188878

ABSTRACT

We have studied the effects of hypoxia on aminolaevulinic acid (ALA)-induced protoporphyrin IX (PpIX) synthesis in EMT6 monolayer cultures characterized by different cell densities and proliferation rates. Specifically, after ALA incubation under hypoxic or normoxic conditions, we detected spectrofluorometrically the PpIX content of the following populations: (a) low-density exponentially growing cells; (b) high-density fed-plateau cells; and (c) high-density unfed-plateau cells. These populations were selected either for the purpose of comparison with other in vitro studies (low-density exponentially growing cells) or as representatives of tumour regions adjacent to (high-density fed-plateau cells) and further away from (high-density unfed-plateau cells) capillaries. The amount of PpIX per cell produced by each one of these populations was higher after normoxic ALA incubation. The magnitude of the effect of hypoxia on PpIX synthesis was dependent on cell density and proliferation rate. A 42-fold decrease in PpIX fluorescence was observed for the high-density unfed-plateau cells. PpIX production by the low-density exponential cells was affected the least by ALA incubation under hypoxic conditions (1.4-fold decrease), whereas the effect on the high-density fed-plateau population was intermediate (20-fold decrease).


Subject(s)
Aminolevulinic Acid/pharmacology , Cell Hypoxia/drug effects , Protoporphyrins/biosynthesis , Cell Count , Cell Division , Cell Hypoxia/physiology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...