Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Oncol Res ; 19(6): 265-74, 2011.
Article in English | MEDLINE | ID: mdl-21776822

ABSTRACT

The ability to image gene expression using 18F-labeled antisense oligonucleotides (asODNs) directed to specific mRNA transcripts during, or immediately following, radio- or chemotherapy would be a valuable clinical tool to monitor the early tumor response to treatment. Imaging of upregulated p21 mRNA postirradiation using 18F-labeled asODNs could offer insights into early tumor responses by detecting signs of accelerated cellular senescence. Thus, the aim of this work was to evaluate the uptake and distribution of a (radio)-fluorinated asODN in vitro in HCT116 p21(+/+) human colon carcinoma cells, asODN and a random sequence oligonucleotide (rsODN) were conjugated with a (radio)fluorine prosthetic group. Irradiated HCT116 cells were treated with naked or liposome-transfected ODNs. Cell fractionation, confocal microscopy, immunofluorescence, and Western blot studies were performed to observe uptake, distribution, and antisense activity of the probes. [F]asODN demonstrated similar antisense binding ability as the unlabeled asODN to p21 mRNA. Liposomal-transfected 18F-labeled asODNs and rsODNs exhibited a three-to fivefold increase in uptake at 2.5 h compared to the naked [18F]ODNs. Distribution of transfected [18F]asODN in the cytoplasm and endosomes increased over time whereas no change in intracellular distribution was observed with transfected [18F]rsODN or naked ODNs. Antisense activity was not compromised with the addition of a fluorine moiety on asODN. The cellular accumulation and distribution of the (radio)fluorinated ODNs was not altered by the addition of the prosthetic group. Radiolabeled ODNs were able to penetrate the cell with preferential uptake observed with the liposome-transfected probes. Increased distribution of [18F]asODN in the cytoplasm suggests the probe is available for targeting its transcript mRNA. This warrants further investigations into the potential of [18F]asODN to image accelerated senescence postirradiation.


Subject(s)
Colonic Neoplasms/diagnostic imaging , Cyclin-Dependent Kinase Inhibitor p21/genetics , Fluorine Radioisotopes , Oligonucleotide Probes , Oligonucleotides, Antisense , Blotting, Western , Cellular Senescence , Cobalt Radioisotopes , Colonic Neoplasms/genetics , Drug Therapy, Combination , Gene Expression , Humans , Microscopy, Confocal , Radionuclide Imaging , Radiopharmaceuticals , Tissue Distribution , Transfection , Tumor Cells, Cultured
2.
Phys Med Biol ; 54(21): 6623-33, 2009 Nov 07.
Article in English | MEDLINE | ID: mdl-19826205

ABSTRACT

Scattered radiation in the penumbra of a megavoltage radiation therapy beam can deposit a non-negligible dose in the healthy tissue around a target volume. The lower energy of the radiation in this region suggests that its biological effectiveness might not be the same as that of the open beam. In this work, we determined the relative biological damage in normal human fibroblasts after megavoltage irradiation in two geometries. The first was an open-beam irradiation and the second was a blocked configuration in which only scattered radiation could reach the target cells. The biological damage was evaluated by the gamma-H2AX immunofluorescence assay, which is capable of detecting DNA double-strand breaks in individual cells. We report that the scattered radiation is more effective at producing biological damage than the open beam radiation. We found a 27% enhancement in the net mean nuclear gamma-H2AX fluorescence intensity at 2 Gy and a 48% enhancement at 4 Gy. These findings are of interest due to the increased doses of penumbral radiation close to target volumes both in dose escalation studies and in IMRT treatment deliveries where high dose gradients exist for the purpose of conformal avoidance of healthy tissues.


Subject(s)
Fibroblasts/radiation effects , Photons , Cell Nucleus/metabolism , Dose-Response Relationship, Radiation , Electrons , Histones/metabolism , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence/methods , Models, Statistical , Monte Carlo Method , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted , Radiotherapy, Intensity-Modulated , Scattering, Radiation
3.
Planta Med ; 73(8): 748-54, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17611930

ABSTRACT

Alpinia galanga, or galangal, has been a popular condiment used in Thai and Asian cuisine for many years. However, relatively little is known of the potential beneficial or adverse health effects of this spice. This study was conducted to analyze the capacity of galangal extract to induce cytotoxicity and DNA damage in six different human cell lines including normal and p53-inactive fibroblasts, normal epithelial and tumour mammary cells and a lung adenocarcinoma cell line. We deliberately focused on treatment with the crude aqueous extract of galangal rhizomes, rather than compounds extracted into an organic solvent, to more closely reflect the mode of dietary consumption of galangal. The cell lines displayed a broad range of cytotoxicity. There was no evidence for preferential cytotoxicity of tumour cells, but there was an indication that p53-active cell lines may be more sensitive than their p53-inactive counterparts. The contribution of apoptosis to total cell killing was only appreciable after exposure to 300 microg/mL of extract. Apoptosis appeared to be independent of p53 expression. Exposure to as little as 100 microg/mL galangal extract generated a significant level of DNA single-strand breaks as judged by the single-cell gel electrophoresis technique (comet assay). The three major UV-absorbing compounds in the aqueous extract were identified by mass spectrometry as 1'-acetoxychavicol acetate and its deacetylated derivatives. However, when tested in A549 human lung adenocarcinoma cells, these compounds were not responsible for the cytotoxicity induced by the complete aqueous extract.


Subject(s)
Alpinia , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , DNA Damage/drug effects , Phytotherapy , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Breast/cytology , Cell Line, Tumor/drug effects , Epithelial Cells/drug effects , Fibroblasts/drug effects , Humans , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Rhizome
4.
Environ Mol Mutagen ; 38(2-3): 191-9, 2001.
Article in English | MEDLINE | ID: mdl-11746754

ABSTRACT

Cultured cells from patients inheriting the rare cancer-prone and radiotherapy-sensitive disorder ataxia telangiectasia (AT) exhibit defects in the activation of cell-cycle checkpoints after exposure to ionizing radiation. In particular, the failure of AT cells to arrest transiently the DNA de novo replication machinery immediately after irradiation--so-called radioresistant DNA synthesis (RDS)--is often taken as a molecular hallmark of the disease. Recently we reported that: (i) the radiation-responsive S-phase checkpoint operating in normal human cells is mediated by a signal transduction pathway involving Ca2+/calmodulin-dependent protein kinase II (CaMKII); and (ii) the RDS phenotype of AT cells is associated with failure to mobilize Ca2+ from intracellular stores, which is required for activation of the CaMKII-dependent S-phase arrest. In the present study, we demonstrate that the RDS phenotype of AT dermal fibroblasts can be rectified in the absence of ectopic expression of functional ATM, the 350-kDa protein kinase encoded by the gene mutated in AT. Correction of RDS was observed when AT fibroblasts were coincubated with normal fibroblasts under conditions in which the 2 different cell cultures shared the same medium but were completely separated physically. The RDS trait was also rectified when AT fibroblasts were briefly incubated with prostaglandin E2 in the absence of normal feeder cells, signifying that this ubiquitous eicosanoid can serve as the diffusible "RDS-correction factor" for AT cells in the aforementioned cocultivation studies. It would therefore appear that prostaglandin E2 can assume the role of an extracellular signaling modulator of the S-phase checkpoint in AT cells exposed to ionizing radiation, inducing DNA synthesis shutdown via an alternative, ATM-independent signal transduction pathway.


Subject(s)
Ataxia Telangiectasia/drug therapy , DNA Replication/drug effects , Dinoprostone/pharmacology , Oxytocics/pharmacology , Radiation Tolerance , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cells, Cultured , DNA Replication/radiation effects , DNA-Binding Proteins , Dinoprostone/therapeutic use , Fibroblasts/drug effects , Fibroblasts/physiology , Fibroblasts/radiation effects , Humans , Oxytocics/therapeutic use , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Suppressor Proteins
5.
Photochem Photobiol ; 72(4): 562-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11045730

ABSTRACT

The contribution of DNA strand breaks accumulating in the course of nucleotide excision repair to upregulation of the p53 tumor suppressor protein was investigated in human dermal fibroblast strains after treatment with 254 nm ultraviolet (UV) light. For this purpose, fibroblast cultures were exposed to UV and incubated for 3 h in the presence or absence of l-beta-D-arabinofuranosylcytosine (araC) and/or hydroxyurea (HU), and then assayed for DNA strand breakage and p53 protein levels. As expected from previous studies, incubation of normal and ataxia telangiectasia (AT) fibroblasts with araC and HU after UV irradiation resulted in an accumulation of DNA strand breaks. Such araC/HU-accumulated strand breaks (reflecting nonligated repair-incision events) following UV irradiation were not detected in xeroderma pigmentosum (XP) fibroblast strains belonging to complementation groups A and G. Western blot analysis revealed that normal fibroblasts exhibited little upregulation of p53 (approximately 1.2-fold) when incubated without araC after 5 J/m2 irradiation, but showed significant (three-fold) upregulation of p53 when incubated with araC after irradiation. AraC is known to inhibit nucleotide excision repair at both the damage removal and repair resynthesis steps. Therefore, the potentiation of UV-induced upregulation of p53 evoked by araC in normal cells may be a consequence of either persistent bulky DNA lesions or persistent incision-associated DNA strand breaks. To distinguish between these two possibilities, we determined p53 induction in AT fibroblasts (which do not upregulate p53 in response to DNA strand breakage) and in XP fibroblasts (which do not exhibit incision-associated breaks after UV irradiation). The p53 response after treatment with 5 J/m2 UV and incubation with araC was similar in AT, XPA, XPG and normal fibroblasts. In addition, exposure of XPA and XPG fibroblasts to UV (5, 10 or 20 J/m2) followed by incubation without araC resulted in a strong upregulation of p53. We further demonstrated that HU, an inhibitor of replicative DNA synthesis (but not of nucleotide excision repair), had no significant impact on p53 protein levels in UV irradiated and unirradiated human fibroblasts. We conclude that upregulation of p53 at early times after exposure of diploid human fibroblasts to UV light is triggered by persistent bulky DNA lesions, and that incision-associated DNA strand breaks accumulating in the course of nucleotide excision repair and breaks arising as a result of inhibition of DNA replication contribute little (if anything) to upregulation of p53.


Subject(s)
DNA Damage , Fibroblasts/radiation effects , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia/pathology , Cells, Cultured , DNA/metabolism , DNA/radiation effects , DNA Repair , Fibroblasts/metabolism , Humans , Tumor Suppressor Protein p53/biosynthesis , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology
6.
Biochem J ; 345 Pt 3: 583-93, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10642517

ABSTRACT

A novel N-glycosylated, mannose-rich protein has been purified approx. 4000-fold from human liver in a seven-step procedure including ion-exchange chromatography and fractionation on concanavalin A-Sepharose, Sephadex G-75 and oligo(dT)-cellulose matrices. The molecular mass of the protein is 46 kDa when measured by gel filtration (i.e. under non-denaturing conditions) and 60 kDa by SDS/PAGE (i.e. under denaturing conditions). The protein possesses two DNA backbone-incising activities, namely, the random introduction of single-strand breaks in native DNA and the rupture of the phosphodiester linkage internal to cyclobutyl pyrimidine dimers, the major class of DNA lesions induced by solar UV rays. Both activities are optimal at pH 5.0 in vitro, although the non-specific nuclease displays appreciable activity at neutral pH, depending on the buffer composition. The protein has been named acidic nuclease/intra-cyclobutyl-pyrimidine-dimer-DNA phosphodiesterase (AN/IDP). As a nuclease, the protein 'prefers' a linear DNA structure over a covalently closed circular molecule and is more proficient at digesting single-stranded than double-stranded DNA. The polynucleotide cleavage products of the nuclease contain 5'-OH and 3'-PO(4) termini, which are refractory to direct rejoining by DNA ligases. Depending on the substrate, the nuclease activity exhibits a temperature optimum of 50 degrees C or greater, and is neither stimulated by Mg(2+) or Ca(2+) nor inhibited by Zn(2+). AN/IDP is present in human liver and in cultured human cells of both fibroblastic and lymphocytic origins. Intracellularly, the protein can be readily detected in both the cytosolic and nuclear fractions, although much more (approx. 3-fold) is found in the latter fraction. We propose that this bifunctional enzyme may be involved in both apoptotic DNA digestion and metabolism of cyclobutyl pyrimidine dimers in UV-irradiated human cells.


Subject(s)
Deoxyribonucleases/isolation & purification , Deoxyribonucleases/metabolism , Phosphoric Diester Hydrolases/isolation & purification , Phosphoric Diester Hydrolases/metabolism , Animals , Calcium/metabolism , Calcium/pharmacology , Cattle , Cells, Cultured , DNA/metabolism , DNA/radiation effects , Dimerization , Fibroblasts/enzymology , Glycosylation , Humans , Hydrogen-Ion Concentration , Liver/enzymology , Lymphocytes/enzymology , Magnesium/metabolism , Magnesium/pharmacology , Pyrimidines/chemistry , Pyrimidines/metabolism , Temperature , Ultraviolet Rays
7.
Br J Cancer ; 81(6): 959-65, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10576651

ABSTRACT

Wortmannin is a potent inhibitor of phosphatidylinositol (PI) 3-kinase and PI 3-kinase-related proteins (e.g. ATM), but it does not inhibit the activity of purified calmodulin-dependent protein kinase II (CaMKII). In the present study, we compared the effects of wortmannin and the CaMKII inhibitor KN62 on the response of normal human dermal fibroblast cultures to gamma radiation. We demonstrate that wortmannin confers a phenotype on normal fibroblasts remarkably similar to that characteristic of cells homozygous for the ATM mutation. Thus wortmannin-treated normal fibroblasts exhibit increased sensitivity to radiation-induced cell killing, lack of temporary block in transition from G1 to S phase following irradiation (i.e. impaired G1/S checkpoint), and radioresistant DNA synthesis (i.e. impaired S phase checkpoint). Wortmannin-treated cultures display a diminished capacity for radiation-induced up-regulation of p53 protein and expression of p21WAF1, a p53-regulated gene involved in cell cycle arrest at the G1/S border; the treated cultures also exhibit decreased capacity for enhancement of CaMKII activity post-irradiation, known to be necessary for triggering the S phase checkpoint. We further demonstrate that KN62 confers a radioresistant DNA synthesis phenotype on normal fibroblasts and moderately potentiates their sensitivity to killing by gamma rays, without modulating G1/S checkpoint, p53 up-regulation and p21WAF1 expression following radiation exposure. We conclude that CaMKII is involved in the radiation responsive signalling pathway mediating S phase checkpoint but not in the p53-dependent pathway controlling G1/S checkpoint, and that a wortmannin-sensitive kinase functions upstream in both pathways.


Subject(s)
Androstadienes/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/pharmacology , Fibroblasts/physiology , Protein Kinase Inhibitors , Radiation-Sensitizing Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Humans , Radiation, Ionizing , S Phase/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Wortmannin
8.
Carcinogenesis ; 20(6): 941-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10357771

ABSTRACT

Ionizing radiation-induced stabilization and the resultant transient accumulation of the p53 tumor suppressor protein is impaired in cells from ataxia telangiectasia (AT) patients, indicating a key role for ATM, the gene mutated in AT, upstream in the radiation-responsive p53 signaling pathway. Activation of this pathway is generally assumed to be triggered by DNA strand breaks produced directly following genotoxic stress or indirectly during excision repair of DNA lesions. The aim of this study was to identify the triggering signal for induction of p53 in diploid human dermal fibroblasts treated with 4-nitroquinoline 1-oxide (4NQO), a model environmental carcinogen that produces both DNA strand breaks (like ionizing radiation) and alkali-stable bulky DNA lesions (like UV light). 4NQO treatment of fibroblasts cultured from normal and AT donors and those from patients with the UV-hypersensitivity disorder xeroderma pigmentosum (XP, complementation groups A, E and G) resulted in up-regulation of p53 protein. In normal fibroblasts, there was no temporal relationship between the incidence of DNA strand breaks and levels of p53 protein; >90% of strand breaks and alkali-labile sites were repaired over 2 h following treatment with 1 microM 4NQO, whereas approximately 3 h of post-treatment incubation was required to demonstrate a significant rise in p53 protein. In contrast, exposure of normal fibroblasts to gamma-rays resulted in a rapid up-regulation of p53 and the level peaked at 2 h post-irradiation. XP cells with a severe deficiency in the nucleotide excision repair pathway showed abnormally high levels of p53 protein in response to 4NQO treatment, indicating that lesions other than incision-associated DNA strand breaks trigger p53 up-regulation. We observed a consistent, inverse correlation between the ability of the various fibroblast cultures to induce p53 following 4NQO treatment and their DNA repair efficiencies. Treatment with 0.12 microM 4NQO, for example, caused a >2-fold up-regulation of p53 in excision repair-deficient (AT, XPA and XPG) strains without eliciting any effect on p53 levels in repair-proficient (normal and XPE) strains. We conclude that up-regulation of p53 by 4NQO is mediated solely by an ATM-independent mechanism and that the p53 response is primarily triggered by persistent alkali-stable 4NQO-DNA adducts.


Subject(s)
4-Nitroquinoline-1-oxide/toxicity , DNA Damage , DNA Repair , Mutagens/toxicity , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , RNA/biosynthesis , Tumor Suppressor Protein p53/biosynthesis
9.
Oncogene ; 17(5): 533-43, 1998 Aug 06.
Article in English | MEDLINE | ID: mdl-9704919

ABSTRACT

The purpose of this study is to better understand the roles of the p53 tumor suppressor protein and the product of the p53-regulated gene p21WAF1 in the response of diploid human dermal fibroblast cultures to 254 nm ultraviolet (UV) light. We report that Li-Fraumeni syndrome (LFS) fibroblast strains heterozygous for TP53 mutation at either codon 245 or 234 exhibit markedly reduced or no expression of p21WAF1 following UV irradiation, respectively. These strains also exhibit defective nucleotide excision repair and pronounced inhibition of RNA synthesis following UV exposure, both of which are molecular hallmarks of cells derived from patients with the UV-sensitive syndrome xeroderma pigmentosum. In sharp contrast to xeroderma pigmentosum cells, however, the repair-deficient LFS cells show abnormal resistance, rather than hypersensitivity, to the killing effect of UV light. We further demonstrate that exposure of normal human fibroblasts to biologically relevant fluences (< or = 15 J/m2) of UV does not induce apoptotic cell death, indicating that UV resistant phenotype displayed by LFS strains is not associated with deregulated apoptosis. In normal fibroblasts, such treatment results in a moderate ( threefold) up-regulation of p53 protein, induction of the p21WAF1 gene, and a senescence-like growth arrest. On the other hand, exposure to > or = 20 J/m2 UV results in a striking up-regulation of p53, inhibition of p21WAF1 expression, and activation of an apoptotic pathway. We conclude that: (i) p21WAF1-mediated senescence is the principal mode of cell death induced by < or = 15 J/m2 UV light in normal human fibroblasts; (ii) there is a threshold effect for p53-dependent apoptosis and that, in normal human cells, this threshold level is induced upon expsoure to 20 J/m2 UV; (iii) the p53 signaling pathway is malfunctional in the TP53 heterozygous LFS strains examined; and (iv) the enhanced resistance to UV-induced cell killing displayed by these LFS strains is a consequence of diminished growth arrest, which is presumably mediated by p21WAF1 and not abnormalities in an apoptotic pathway.


Subject(s)
Cyclins/metabolism , Fibroblasts/radiation effects , Li-Fraumeni Syndrome , Tumor Suppressor Protein p53/physiology , Ultraviolet Rays , Apoptosis , Cell Division , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , DNA Damage , Fibroblasts/cytology , Heterozygote , Humans , Mutation , Pyrimidine Dimers , RNA/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation
10.
Radiat Res ; 150(1): 11-6, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9650596

ABSTRACT

Ataxia telangiectasia (AT) is an autosomal recessive human disorder featuring diverse clinical abnormalities including proneness to cancer and extreme sensitivity to ionizing radiation. Although cells from AT patients exhibit faulty activation of the p53 signal transduction pathway at early times after radiation exposure, it has been proposed that high levels of DNA damage persisting in AT cells may up-regulate p53 through an ATM-independent mechanism at late times after irradiation, leading to cell death by apoptosis. In this study we demonstrate that diploid skin fibroblast strains homozygous for the AT mutation fail to up-regulate p53 protein at late times (< or = 48 h) after irradiation with 60Co gamma rays. Moreover, exposure of normal and AT fibroblasts to a dose of 8 Gy does not result in a significant increase in the fraction of apoptotic cells. Since this treatment reduces the clonogenic potential of human cells by at least two orders of magnitude, we conclude that apoptosis is not the primary mechanism of cell death induced by ionizing radiation in human normal and AT fibroblast cultures. Therefore, our results are not in accordance with the current hypothesis suggesting that increased radiosensitivity of AT cells is associated with deregulated apoptosis.


Subject(s)
Apoptosis/radiation effects , Ataxia Telangiectasia/pathology , Fibroblasts/pathology , Fibroblasts/radiation effects , Apoptosis/physiology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , DNA/radiation effects , DNA Damage , Fibroblasts/metabolism , HL-60 Cells/pathology , HL-60 Cells/radiation effects , Humans , Radiation Tolerance , Skin/cytology , Skin/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Up-Regulation/radiation effects
12.
Radiat Res ; 147(1): 13-21, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8989364

ABSTRACT

Proliferating human cells exposed to ionizing radiation show complex cellular responses including a delay in progression through various phases in the cell cycle. These cell cycle checkpoints are regulated by mitogenic signaling pathways which transduce the extracellular signals to the cell cycle control machinery. In this study we demonstrate that microinjection of a cellular extract, prepared from gamma-irradiated (40 Gy) HeLa cells, into the cytoplasm of normal human fibroblasts results in suppression of DNA replicative synthesis, indicating the presence of a trans-acting DNA synthesis-inhibiting factor(s). The addition of this same extract to the culture medium for a short time (< or = 2 h) also inhibits DNA synthesis in human fibroblasts, affecting both replicon initiation and DNA chain elongation processes. Moreover, a 2-h incubation of the fibroblast cultures with the extract causes a transient delay in cell progression from G1 to S phase coupled with up-regulation of the p53 tumor suppressor protein. Both the DNA synthesis-inhibiting and G1-phase-blocking activities are reduced markedly when the extract is heated (80 degrees C; 10 min) prior to its addition to the culture medium. On the other hand, pretreatment of the fibroblast cultures with KN62, an inhibitor of calmodulin-dependent kinase II (CaMKII), serves to abrogate the inhibitory effect of the extract on DNA synthesis without influencing its ability to induce the G1-phase block. These results are compatible with the presence in HeLa cell extracts of a heat-labile trans-acting factor that triggers, in normal human cells, the activation of (1) a CaMKII-dependent signal transduction pathway mediating suppression of DNA synthesis and (2) a p53-dependent pathway mediating G1-phase checkpoint control.


Subject(s)
Cell Cycle , DNA Replication , HeLa Cells/radiation effects , Cobalt Radioisotopes , DNA/biosynthesis , Dose-Response Relationship, Radiation , Fibroblasts/cytology , Fibroblasts/physiology , G1 Phase/drug effects , Gamma Rays , Humans , Kinetics , Microinjections , S Phase/drug effects , Thymidine/metabolism , Time Factors , Tissue Extracts/administration & dosage , Tissue Extracts/pharmacology
13.
Carcinogenesis ; 17(4): 691-8, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8625479

ABSTRACT

Dermal fibroblast strains cultured from affected members of a cancer-prone family with Li-Fraumeni syndrome (LFS) harbor a point mutation in one allele of the p53 tumor suppressor gene, resulting in loss of normal p53 function. In this study we have examined the ability of these p53-deficient strains to carry out the long-patch mode of excision repair, mediated by DNA polymerases delta and epsilon, after exposure to 60Co gamma radiation or far ultraviolet (UV) (chiefly 254 nm) light. Repair was monitored by incubation of the irradiated cultures in the presence of aphidicolin (apc) or 1-beta-D-arabinofuranosylcytosine (araC), each a specific inhibitor of long-patch repair, followed by measurement of drug-induced DNA strand breaks (reflecting non-ligated strand incision events) by alkaline sucrose velocity sedimentation. The LFS strains displayed deficient repair capacity in response to both gamma rays and UV light. The repair anomaly in UV-irradiated LFS cultures was manifested not only in the overall genome, but also in the transcriptionally active, preferentially repaired c-myc gene. Using autoradiography we also assessed unscheduled DNA synthesis (UDS) after UV irradiation and found this conventional measure of repair replication to be deficient in LFS strains. Moreover, both apc and araC decreased the level of UV-induced UDS by approximately 75% in normal cells, but each had only a marginal effect on LFS cells. We further demonstrated that the LFS strains are impaired in the recovery of both RNA and replicative DNA syntheses after UV treatment, two molecular anomalies of the DNA repair deficiency disorders xeroderma pigmentosum and Cockayne's syndrome. Together these results imply a critical role for wild-type p53 protein in DNA polymerase delta/epsilon-mediated excision repair, both the mechanism operating on the entire genome and that acting on expressed genes.


Subject(s)
DNA Repair , DNA-Directed DNA Polymerase/metabolism , DNA/radiation effects , Genes, p53 , Li-Fraumeni Syndrome/genetics , Cytarabine/pharmacology , DNA/drug effects , DNA Polymerase II , DNA Polymerase III , Deoxycytidine Kinase/metabolism , Fibroblasts/enzymology , Gamma Rays , Genes, APC , Genes, myc , Humans , Li-Fraumeni Syndrome/pathology , Phosphorylation , Ultraviolet Rays
14.
Radiat Res ; 144(3): 288-93, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7494872

ABSTRACT

The induction and repair of DNA double-strand breaks were studied in cells of two isogenic human malignant glioma cell lines which vary in their SF2 values by a factor of approximately 30. M059J cells are radiosensitive (SF2 = 0.02) and lack the p350 component of DNA-dependent protein kinase (DNA-PK); M059K cells are radioresistant (SF2 = 0.64) and express normal levels of DNA-PK. Zero integrated field gel electrophoresis and alkaline sucrose gradient experiments indicated that equivalent numbers of DNA lesions were produced by ionizing radiation in M059J and M059K cells. To compare the capacity of both lines to repair sublethal damage, the split-dose recovery experiment after exposure to equitoxic doses of radiation was carried out. Significant sublethal damage repair was shown for M059K cells, with a 5.8-fold increase in relative survival peaking at 4 h, whereas M059J cells showed little repair activity. Electrophoresis studies indicated that more double-strand breaks were repaired by 30 min in M059K cells than in M059J cells. These results suggest that deficient DNA repair processes may be a major determinant of radiosensitivity in M059J cells.


Subject(s)
DNA Damage , DNA Repair , DNA, Neoplasm/radiation effects , Cell Line , DNA, Neoplasm/isolation & purification , Dose-Response Relationship, Radiation , Glioma , Humans , Kinetics , Time Factors , Tumor Cells, Cultured
15.
Oncogene ; 11(8): 1597-605, 1995 Oct 19.
Article in English | MEDLINE | ID: mdl-7478584

ABSTRACT

Cultured cells from patients inheriting the rare cancer-prone and radiotherapy-sensitive disorder ataxia-telangiectasia (A-T) exhibit anomalies in cell cycle control and protein kinase C (PKC)-mediated upregulation of p53 protein following exposure to ionizing radiation. It remains unclear, however, as to whether this irregularity in a p53-dependent signal transduction pathway controlling the G1/S checkpoint is causally linked to the most consistent molecular hallmark of A-T-namely, marked attenuation in the inhibition of replicative DNA synthesis at early times (< or = 2 h) after irradiation [radioresistant DNA synthesis (RDS)]. We report here that treatment of normal human fibroblast strains with inhibitors of calmodulin (CaM) (i.e. W7 and W13) and CaM-dependent protein kinases II and IV (i.e. KN62) prior to radiation exposure elicits an 'A-T-like' RDS phenotype, whereas treatment with PKC inhibitors (e.g. staurosporine) does not produce this response. Moreover, at 1 h post-gamma irradiation A-T fibroblasts undergo normal induction of p53 protein while exhibiting the RDS trait. At later times (e.g. 4 h) following irradiation, however, these A-T cells contain abnormally low levels of p53 protein, as do their lymphoblastoid cell line counterparts during the entire post-gamma ray incubation period. On the other hand, human cells which either lack the p53 gene completely (i.e. HL60 leukemia cells) or harbor a germline mutation in the gene (i.e. Li-Fraumeni syndrome cells) shut down their DNA replication machinery normally upon sustaining radiation damage. We thus conclude that the transitory delay in DNA synthesis routinely experienced by human cells in the face of radiation injury is mediated through a CaM-dependent regulatory cascade which involves neither PKC nor p53 protein. Accordingly, A-T cells appear to be malfunctional in at least two distinct radiation-responsive signalling pathways, one regulating the G1/S checkpoint and governed by p53 and PKC and another controlling passage through S phase and requiring CaM.


Subject(s)
Calmodulin/physiology , DNA Repair/radiation effects , DNA Replication/radiation effects , Protein Kinase C/physiology , Tumor Suppressor Protein p53/physiology , Ataxia Telangiectasia/genetics , Calmodulin/antagonists & inhibitors , Cells, Cultured , Dose-Response Relationship, Radiation , Gamma Rays , HL-60 Cells , Humans , Signal Transduction
16.
Br J Cancer ; 71(6): 1221-30, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7779715

ABSTRACT

Non-malignant dermal fibroblast strains, cultured from affected members of a Li-Fraumeni syndrome (LFS) family with diverse neoplasms associated with radiation exposure, display a unique increased resistance to the lethal effects of gamma-radiation. In the studies reported here, this radioresistance (RR) trait has been found to correlate strongly with an abnormal pattern of post-gamma-ray DNA replicative synthesis, as monitored by radiolabelled thymidine incorporation and S-phase cell autoradiography. In particular, the time interval between the gamma-ray-induced shutdown of DNA synthesis and its subsequent recovery was greater in all four RR strains examined and the post-recovery replication rate was much higher and was maintained longer than in normal and spousal controls. Alkaline sucrose sedimentation profiles of pulse-labelled cellular DNA indicated that the unusual pattern of DNA replication in irradiated RR strains may be ascribed to anomalies in both replicon initiation and DNA chain elongation processes. Moreover, the RR strain which had previously displayed the highest post-gamma-ray clonogenic survival was found to harbour a somatic (codon 234) mutation (presumably acquired during culture in vitro) in the same conserved region of the p53 tumour-suppressor gene as the germline (codon 245) mutation in the remaining three RR strains from other family members, thus coupling the RR phenotype and abnormal post-gamma-ray DNA synthesis pattern with faulty p53 expression. Significantly, these two aberrant radioresponse end points, along with documented anomalies in c-myc and c-raf-1 proto-oncogenes, are unprecedented among other LFS families carrying p53 germline mutations. We thus speculate that this peculiar cancer-prone family may possess in its germ line a second, as yet unidentified, genetic defect in addition to the p53 mutation.


Subject(s)
DNA Damage , DNA Repair , DNA Replication/radiation effects , Li-Fraumeni Syndrome/genetics , Skin/radiation effects , Adolescent , Adult , Aged , Cells, Cultured , Child , Child, Preschool , Cobalt Radioisotopes , Female , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gamma Rays , Humans , Kinetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Male , Middle Aged , Skin/metabolism , Skin/pathology , Time Factors
17.
Carcinogenesis ; 15(10): 2319-24, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7525096

ABSTRACT

We have investigated the genotoxicity of two 3'-derivatives of cytidine, 2,3'-O-cyclocytidine (3'-cycloC) and beta-xylocytidine (xyloC), in human leukemia and solid tumor cell lines. Both derivatives were found to be cytotoxic at micromolar concentrations. For example, in the alveolar tumor cell line A549 which was included in all experiments as a reference, drug concentrations required to induce 50% inhibition of cell growth (D50 values) equalled 55 microM for 3'-cycloC and 80 microM for xyloC. Compared with the response of this reference cell line, none of the solid tumor cell lines tested--representing five different malignancies--displayed significant hypersensitivity to these drugs, while the acute lymphoblastic leukemia cell lines proved to be hypersensitive (range of D50 values, 5-13 microM). To gain insight into the modes of cytotoxic action of xyloC and 3'-cycloC, we compared the effect on DNA metabolism of these compounds with that of 1-beta-D-arabinofuranosylcytosine (araC), a potent inhibitor of semi-conservative DNA replication and long-patch excision repair. As seen with araC, the xylo compound strongly inhibited both DNA replicative synthesis and the repair of DNA damage induced by UV light and 60Co gamma-radiation. In gamma-irradiated A549 cells, the extent of repair inhibition by 1 mM xyloC was approximately 40% of that inhibited by araC, and concomitant exposure of the irradiated cultures to xyloC plus araC gave rise to a synergistic response. Since araC was employed at a concentration (0.1 mM) which produced a maximal effect on DNA repair when applied alone, the observed synergistic response implies that the mode of action of xyloC on DNA repair is different from that of araC. In contrast to that observed with xyloC, 3'-cycloC proved to be a very weak inhibitor of DNA replication and repair, strongly suggesting that the genotoxic action of the latter analog may be through a mechanism other than inhibition of DNA synthesis.


Subject(s)
Ancitabine/toxicity , Cytarabine/toxicity , Cytidine/analogs & derivatives , Leukemia/drug therapy , Leukemia/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cell Division/drug effects , Cell Survival/drug effects , Child , Cytarabine/administration & dosage , Cytidine/toxicity , DNA Damage , DNA Repair/drug effects , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Leukemia/pathology , Neoplasms/pathology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
18.
Biochim Biophys Acta ; 1227(1-2): 92-100, 1994 Oct 21.
Article in English | MEDLINE | ID: mdl-7918688

ABSTRACT

The mammalian DNA polymerase inhibitors aphidicolin and 1-beta-D-arabinofuranosylcytosine (araC), when used in combination, inhibit the repair of DNA damage induced by gamma rays or 4-nitroquinoline 1-oxide in normal human fibroblasts to an extent 2- to 4-fold greater than that seen with each inhibitor alone. Thus either aphidicolin modulates the rate of intracellular accumulation of araC 5'-triphosphate (araCTP), the presumed rate-limiting step in the genotoxic action of araC, or aphidicolin and araC inhibit repair by different mechanisms. To explore these possibilities, we compared the effects of aphidicolin, araC, araCTP, and 2',3'-dideoxythymidine triphosphate (ddTTP) on repair of DNA damage induced by 60Co gamma radiation in intact versus permeable human fibroblasts. Both aphidicolin and araC strongly inhibited repair in permeable cells, as indicated by the accumulation of DNA strand breaks in irradiated cultures that were subsequently treated with saponin (25 micrograms/ml; 10 min) and incubated for 2 h with either chemical. The extent of repair inhibition by each drug was comparable in intact and permeable cells, amounting to approximately 1.1 sites/10(8) daltons/2 h upon exposure to 150 Gy. The active metabolite of araC, araCTP, did not inhibit repair in intact cells, but did so in permeable cells to an extent within the range of that seen with araC or aphidicolin alone. The incidence of DNA strand breaks accumulating in gamma-irradiated permeable cultures as a result of incubation with araCTP plus aphidicolin, or araC plus aphidicolin, was approximately 2-fold greater than that arising in parallel cultures which had been incubated with optimal concentrations of each of the three drugs alone. Although the resolution of our assays compelled us to monitor repair events in moribund cell populations, we have reason to be confident that within the short post-irradiation period considered here, the observed drug-accumulated breaks truly represent functional repair inhibition and not merely abortive pathological responses. We thus conclude that (1) the accumulation of araCTP in intact cells is not limiting the ability of araC to inhibit DNA repair; and (2) the mode of the inhibitory action of araC/araCTP on gamma ray repair is different from that of aphidicolin. In contrast to the observations with these chemicals, ddTTP (20 microM), a potent inhibitor of DNA polymerase beta, did not produce any measurable effect on DNA repair in gamma-irradiated permeable fibroblasts, nor did it enhance the efficacy of araC, araCTP or aphidicolin to inhibit repair. These results strongly suggest that DNA polymerase beta plays no significant role in the repair of gamma radioproducts in human fibroblasts.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Aphidicolin/pharmacology , Cytarabine/pharmacology , DNA Repair/drug effects , Nucleic Acid Synthesis Inhibitors , Arabinofuranosylcytosine Triphosphate/pharmacology , Cells, Cultured/drug effects , Child , Drug Combinations , Fibroblasts/drug effects , Fibroblasts/radiation effects , Humans , Saponins/pharmacology
19.
Mutat Res ; 314(2): 99-113, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7510369

ABSTRACT

O6-Methylguanine (m6G) is an altered base produced in DNA by SN1 methylating agents such as N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). This lesion is repaired by the protein O6-methylguanine-DNA methyltransferase (MGMT) in normal human cell lines, but is not repaired in certain human tumor lines that are termed Mex- or Mer-. Compared with repair-proficient cell lines, such repair-deficient tumor lines are hypersensitive to the production by MNNG of sister-chromatid exchanges (SCE), mutations and lethality. We report here that MNNG treatment produces 1 SCE for every 42 +/- 10 m6G formed in the genome of Mer- tumor cells, 1 6TG-resistant mutant for every 8 (range of 5-14) m6G produced statistically in the coding region of the hypoxanthine phosphoribosyltransferase gene, and 1 lethal event per 6650 +/- 1200 m6G. In addition, in vitro base mismatch incision at m6G: BrU pairs was similar to that at m6G: T pairs, the lesions that likely initiate SCE production. We conclude that m6G residues in genomic DNA are very recombinogenic as well as highly mutagenic in Mer- human tumor cells. The results are interpreted in terms of the relationship between methylation-induced SCE and G: T mismatch recognition.


Subject(s)
DNA/chemistry , Guanine/analogs & derivatives , Mutation , Sister Chromatid Exchange , Base Sequence , Cell Death , DNA/drug effects , DNA/genetics , DNA Repair , Guanine/analysis , Humans , Methylnitronitrosoguanidine/toxicity , Methyltransferases/metabolism , Molecular Sequence Data , Mutagens/toxicity , Nucleic Acid Heteroduplexes , O(6)-Methylguanine-DNA Methyltransferase , Thioguanine/pharmacology , Tumor Cells, Cultured
20.
Mol Biotechnol ; 1(1): 29-48, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7859152

ABSTRACT

Polybrene/DMSO-assisted gene transfer is a simple and versatile transfection strategy capable of producing high numbers of stable transfectants from adherent monolayer cultures with low (nanogram) quantities of exogenous DNA. The procedure involves two stages: adsorption and internalization. The former is mediated by polybrene (a polycation polymer) and favors the uniform coating of target cells with polybrene-DNA complexes. Following adsorption, the cells are permeabilized by a brief exposure to dimethyl sulfoxide (DMSO) to facilitate the uptake of DNA complexes. Diverse cell types can be exposed to a wide range of polybrene concentrations without adverse effects. By contrast, the key determinant of success is the DMSO permeabilization regime, which must be configured independently for each cell line. Protocols optimized for gene transfer in murine and human fibroblasts are presented along with a guide for the rapid optimization of the method. The advantages and limitations of the method are also discussed.


Subject(s)
Gene Transfer Techniques , Genetic Techniques , 3T3 Cells , Adsorption , Animals , Biotechnology , Cells, Cultured , DNA/administration & dosage , DNA/genetics , DNA/metabolism , Dimethyl Sulfoxide , Hexadimethrine Bromide , Humans , Mice , Permeability , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...