Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Bioorg Med Chem ; 27(2): 278-284, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30552005

ABSTRACT

Electron transfer through π-stacked arrays of double-stranded DNA contributes to the redox chemistry of bases, including guanine oxidation and thymine-thymine dimer repair by photolyase. 5-Bromouracil is an attractive photoreactive thymine analogue that can be used to investigate electron transfer in DNA, and is a useful probe for protein-DNA interaction analysis. In the present study using BrU we found that UV irradiation facilitated electron injection from mitochondrial transcription factor A into DNA. We also observed that this electron injection could lead to repair of a thymine-thymine dimer.


Subject(s)
DNA Repair/radiation effects , DNA-Binding Proteins/chemistry , DNA/chemistry , Electrons , Mitochondrial Proteins/chemistry , Pyrimidine Dimers/chemistry , Transcription Factors/chemistry , Base Sequence , Bromouracil/chemistry , Bromouracil/radiation effects , DNA/genetics , DNA/metabolism , DNA/radiation effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/radiation effects , Humans , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/radiation effects , Promoter Regions, Genetic/radiation effects , Protein Binding , Pyrimidine Dimers/radiation effects , Transcription Factors/metabolism , Transcription Factors/radiation effects , Ultraviolet Rays
2.
Angew Chem Int Ed Engl ; 58(23): 7626-7630, 2019 06 03.
Article in English | MEDLINE | ID: mdl-30908862

ABSTRACT

Herein, the direct visualization of the dynamic interaction between a photoresponsive transcription factor fusion, GAL4-VVD, and DNA using high-speed atomic force microscopy (HS-AFM) is reported. A series of different GAL4-VVD movements, such as binding, sliding, stalling, and dissociation, was observed. Inter-strand jumping on two double-stranded (ds) DNAs was also observed. Detailed analysis using a long substrate DNA strand containing five GAL4-binding sites revealed that GAL4-VVD randomly moved on the dsDNA using sliding and hopping to rapidly find specific binding sites, and then stalled to the specific sites to form a stable complex formation. These results suggest the existence of different conformations of the protein to enable sliding and stalling. This single-molecule imaging system with nanoscale resolution provides an insight into the searching mechanism used by DNA-binding proteins.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Fungal Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Binding Sites , DNA/chemistry , DNA/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/radiation effects , Fungal Proteins/genetics , Fungal Proteins/radiation effects , Light , Microscopy, Atomic Force , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/radiation effects , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/radiation effects , Transcription Factors/genetics , Transcription Factors/radiation effects
3.
Photochem Photobiol Sci ; 14(11): 1998-2006, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26441326

ABSTRACT

Aureochrome-1 (AUREO1) has been identified as a blue light (BL) receptor responsible for the BL-induced blanching of a stramenopile alga, Vaucheria frigida. BL induces the dimerization of monomeric AUREO1, which subsequently increases its affinity for the target sequence. We made a synthetic gene encoding N-terminally truncated monomeric AUREO1 (Photozipper protein) containing a basic region/leucine zipper (bZIP) domain and a light-oxygen-voltage-sensing domain. In the present study, yellow fluorescent protein or mCherry protein was fused with the Photozipper (PZ) protein, and their oligomeric structures and DNA-binding were compared in the dark and light states. Dynamic light scattering and size exclusion chromatography demonstrated that the hydrodynamic radii and molecular masses of the fusion proteins increased upon BL illumination, suggesting that fusion PZs underwent BL-induced dimerization. Moreover, BL-induced dimerization enhanced their affinities for the target sequence. Taken together, PZ likely functions as a BL-regulated bZIP module in fusion proteins, and can possibly provide a new approach for controlling bZIP transcription factors.


Subject(s)
Algal Proteins/chemistry , Algal Proteins/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Leucine Zippers/radiation effects , Light , Algal Proteins/radiation effects , Base Sequence , Binding Sites , DNA/chemistry , DNA/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/radiation effects , Protein Binding/radiation effects
4.
Mol Biol (Mosk) ; 49(6): 1035-40, 2015.
Article in Russian | MEDLINE | ID: mdl-26710787

ABSTRACT

The bioluminescence induced by luciferases of marine bacteria promotes repair of UV damaged DNA of Escherichia coli AB1886 uvrA6. It is shown that bacterial photolyase that implements photoreactivation activity is the major contributor to DNA repair. However, the intensity of bioluminescence increasing induced by UV-irradiation (SOS-induction) in bacterial cells is not enough for efficient photoreactivation.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Luciferases, Bacterial/metabolism , SOS Response, Genetics , Ultraviolet Rays , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/radiation effects , Escherichia coli/radiation effects , Escherichia coli Proteins/genetics , Escherichia coli Proteins/radiation effects , Luciferases, Bacterial/genetics , Luciferases, Bacterial/radiation effects , Photobacterium/enzymology
5.
Biochemistry ; 52(38): 6653-61, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23992349

ABSTRACT

With their utilization of light-driven allostery to control biochemical activities, photosensory proteins are of great interest as model systems and novel reagents for use by the basic science and engineering communities. One such protein, the light-activated EL222 transcription factor, from the marine bacterium Erythrobacter litoralis HTCC2594, is appealing for such studies, as it harnesses blue light to drive the reorientation of light-oxygen-voltage (LOV) sensory and helix-turn-helix (HTH) effector domains to allow photoactivation of gene transcription in natural and artificial systems. The protein conformational changes required for this process are not well understood, in part because of the relatively short lifetime of the EL222 photoexcited state (τ ∼ 29 s), which complicates its characterization via certain biophysical methods. Here we report how we have circumvented this limitation by creating an EL222 variant harboring V41I, L52I, A79Q, and V121I point mutations (AQTrip) that stabilizes the photoactivated state. Using the wild-type and AQTrip EL222 proteins, we have probed EL222 activation using a combination of solution scattering, nuclear magnetic resonance (NMR), and electromobility shift assays. Size-exclusion chromatography and light scattering indicate that AQTrip oligomerizes in the absence of DNA and selects for an EL222 dimer-DNA complex in the presence of DNA substrates. These results are confirmed in wild-type EL222 with a high-affinity DNA-binding site that stabilizes the complex. NMR analyses of the EL222-DNA complex confirm a 2:1 stoichiometry in the presence of a previously characterized DNA substrate. Combined, these novel approaches have validated a key mechanistic step, whereby blue light induces EL222 dimerization through LOV and HTH interfaces.


Subject(s)
Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , Transcription Factors/chemistry , Alphaproteobacteria/chemistry , Alphaproteobacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/radiation effects , Catalytic Domain , Chromatography, Gel , DNA-Binding Proteins/genetics , DNA-Binding Proteins/radiation effects , Light , Nuclear Magnetic Resonance, Biomolecular , Protein Multimerization , Protein Structure, Tertiary , Scattering, Radiation , Transcription Factors/genetics , Transcription Factors/radiation effects
6.
Biochem Biophys Res Commun ; 433(1): 36-9, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23454122

ABSTRACT

In the present study, the protein microarray was used to investigate the protein expression in human B-cell lymphoblastoid cells intermittently exposed to 1.8-GHz GSM radiofrequency radiation (RFR) at the specific absorption rate (SAR) of 2.0 W/kg for 24 h. The differential expression of 27 proteins was found, which were related to DNA damage repair, apoptosis, oncogenesis, cell cycle and proliferation (ratio >1.5-fold, P<0.05). The results validated with Western blot assay indicated that the expression of RPA32 was significantly down-regulated (P<0.05) while the expression of p73 was significantly up-regulated in RFR exposure group (P<0.05). Because of the crucial roles of those proteins in DNA repair and cell apoptosis, the results of present investigation may explain the biological effects of RFR on DNA damage/repair and cell apoptosis.


Subject(s)
Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/radiation effects , Proteins/metabolism , Proteins/radiation effects , Radio Waves/adverse effects , Apoptosis/radiation effects , Cell Cycle/radiation effects , Cell Line , Cell Proliferation/radiation effects , Cell Transformation, Neoplastic/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/radiation effects , Down-Regulation/radiation effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/radiation effects , Nuclear Proteins/metabolism , Nuclear Proteins/radiation effects , Protein Array Analysis , Replication Protein A/metabolism , Replication Protein A/radiation effects , Tumor Protein p73 , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/radiation effects , Up-Regulation/radiation effects
7.
J Immunol ; 186(1): 255-63, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21131418

ABSTRACT

Th2-type inflammation spontaneously shown in Bcl6-knockout (KO) mice is mainly caused by bone marrow (BM)-derived nonlymphoid cells. However, the function of dendritic cells (DCs) in Bcl6-KO mice has not been reported. We show in this article that the numbers of CD4(+) conventional DCs (cDCs) and CD8α(+) cDCs, but not of plasmacytoid DCs, were markedly reduced in the spleen of Bcl6-KO mice. Generation of cDCs from DC progenitors in BM cells was perturbed in the spleen of irradiated wild-type (WT) mice transferred with Bcl6-KO BM cells, indicating an intrinsic effect of Bcl6 in cDC precursors. Although cDC precursors were developed in a Bcl6-KO BM culture with Fms-like tyrosine kinase 3 ligand, the cDC precursors were more apoptotic than WT ones. Also p53, one of the molecular targets of Bcl6, was overexpressed in the precursors. The addition of a p53 inhibitor to Bcl6-KO BM culture protected apoptosis, suggesting that Bcl6 is required by cDC precursors for survival by controlling p53 expression. Furthermore, large numbers of T1/ST2(+) Th2 cells were naturally developed in the spleen of Bcl6-KO mice. Th2 skewing was accelerated in the culture of WT CD4 T cells stimulated with Ags and LPS-activated Bcl6-KO BM-derived DCs, which produced more IL-6 and less IL-12 than did WT DCs; the addition of anti-IL-6 Abs to the culture partially abrogated the Th2 skewing. These results suggest that Bcl6 is required in cDC precursors for survival and in activated DCs for modulating the cytokine profile.


Subject(s)
CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Differentiation/immunology , DNA-Binding Proteins/physiology , Dendritic Cells/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Cells/radiation effects , CD4 Antigens/radiation effects , CD8 Antigens/radiation effects , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/radiation effects , Dendritic Cells/radiation effects , Dendritic Cells/transplantation , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6 , Spleen/immunology , Spleen/pathology , Spleen/radiation effects
8.
Proc Natl Acad Sci U S A ; 107(32): 14205-10, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660770

ABSTRACT

Ionizing radiation (IR) induces a variety of DNA lesions among which DNA double-strand breaks (DSBs) are the biologically most significant. It is currently unclear if DSB repair is equally efficient after low and high doses. Here, we use gamma-H2AX, phospho-ATM (pATM), and 53BP1 foci analysis to monitor DSB repair. We show, consistent with a previous study, that the kinetics of gamma-H2AX and pATM foci loss in confluent primary human fibroblasts are substantially compromised after doses of 10 mGy and lower. Following 2.5 mGy, cells fail to show any foci loss. Strikingly, cells pretreated with 10 microM H(2)O(2) efficiently remove all gamma-H2AX foci induced by 10 mGy. At the concentration used, H(2)O(2) produces single-strand breaks and base damages via the generation of oxygen radicals but no DSBs. Moreover, 10 microM H(2)O(2) up-regulates a set of genes that is also up-regulated after high (200 mGy) but not after low (10 mGy) radiation doses. This suggests that low radical levels induce a response that is required for the repair of radiation-induced DSBs when the radiation damage is too low to cause the induction itself. To address the in vivo significance of this finding, we established gamma-H2AX and 53BP1 foci analysis in various mouse tissues. Although mice irradiated with 100 mGy or 1 Gy show efficient gamma-H2AX and 53BP1 foci removal during 24 h post-IR, barely any foci loss was observed after 10 mGy. Our data suggest that the cellular response to DSBs is substantially different for low vs. high radiation doses.


Subject(s)
DNA Repair , Dose-Response Relationship, Radiation , Fibroblasts/radiation effects , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/radiation effects , Cells, Cultured , DNA Breaks, Double-Stranded , DNA-Binding Proteins/radiation effects , Fibroblasts/cytology , Histones/radiation effects , Humans , Kinetics , Mice , Protein Serine-Threonine Kinases/radiation effects , Tumor Suppressor Proteins/radiation effects
9.
Acta Med Okayama ; 66(2): 83-92, 2012.
Article in English | MEDLINE | ID: mdl-22525466

ABSTRACT

Radiotherapy plays a central part in cancer treatment, and use of radiosensitizing agents can greatly enhance this modality. Although studies have shown that several chemotherapeutic agents have the potential to increase the radiosensitivity of tumor cells, investigators have also studied a number of molecularly targeted agents as radiosensitizers in clinical trials based on reasonably promising preclinical data. Recent intense research into the DNA damage-signaling pathway revealed that ataxia-telangiectasia mutated (ATM) and the Mre11-Rad50-NBS1 (MRN) complex play central roles in DNA repair and cell cycle checkpoints and that these molecules are promising targets for radiosensitization. Researchers recently developed three ATM inhibitors (KU-55933, CGK733, and CP466722) and an MRN complex inhibitor (mirin) and showed that they have great potential as radiosensitizers of tumors in preclinical studies. Additionally, we showed that a telomerase-dependent oncolytic adenovirus that we developed (OBP-301 [telomelysin]) produces profound radiosensitizing effects by inhibiting the MRN complex via the adenoviral E1B55kDa protein. A recent Phase I trial in the United States determined that telomelysin was safe and well tolerated in humans, and this agent is about to be tested in combination with radiotherapy in a clinical trial based on intriguing preclinical data demonstrating that telomelysin and ionizing radiation can potentiate each other. In this review, we highlight the great potential of ATM and MRN complex inhibitors, including telomelysin, as radiosensitizing agents.


Subject(s)
Ataxia Telangiectasia/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Radiation-Sensitizing Agents/therapeutic use , Tumor Suppressor Proteins/genetics , Viral Proteins/therapeutic use , Ataxia Telangiectasia/radiotherapy , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/radiation effects , DNA Damage , DNA Repair , DNA-Binding Proteins/radiation effects , Humans , Protein Serine-Threonine Kinases/radiation effects , Signal Transduction , Tumor Suppressor Proteins/radiation effects
10.
Nat Cell Biol ; 5(6): 572-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766777

ABSTRACT

DNA double-strand break repair (DSBR) is an essential process for preserving genomic integrity in all organisms. To investigate this process at the cellular level, we engineered a system of fluorescently marked DNA double-strand breaks (DSBs) in the yeast Saccharomyces cerevisiae to visualize in vivo DSBR in single cells. Using this system, we demonstrate for the first time that Rad52 DNA repair foci and DSBs colocalize. Time-lapse microscopy reveals that the relocalization of Rad52 protein into a focal assembly is a rapid and reversible process. In addition, analysis of DNA damage checkpoint-deficient cells provides direct evidence for coordination between DNA repair and subsequent release from checkpoint arrest. Finally, analyses of cells experiencing multiple DSBs demonstrate that Rad52 foci are centres of DNA repair capable of simultaneously recruiting more than one DSB.


Subject(s)
Bacterial Proteins , DNA Damage , DNA Repair , DNA, Fungal/genetics , DNA-Binding Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , DNA, Fungal/radiation effects , DNA-Binding Proteins/radiation effects , Deoxyribonucleases, Type II Site-Specific/metabolism , Escherichia coli Proteins/metabolism , G1 Phase/physiology , G1 Phase/radiation effects , G2 Phase/physiology , G2 Phase/radiation effects , Gamma Rays/adverse effects , Genes, Insect , Haploidy , Lac Repressors , Mitosis , Rad52 DNA Repair and Recombination Protein , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/radiation effects , Time Factors
11.
Mutat Res ; 708(1-2): 28-36, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21315088

ABSTRACT

Ataxia-telangiectasia mutated (ATM) encodes a nuclear serine/threonine protein kinase whose activity is increased in cells exposed to low doses of ionizing radiation (IR). Here we examine ATM kinase activation in cells exposed to either (32)P- or (33)P-orthophosphate under conditions typically employed in metabolic labelling experiments. We calculate that the absorbed dose of IR delivered to a 5cm×5cm monolayer of cells incubated in 2ml media containing 1mCi of the high-energy (1.70MeV) ß-particle emitter (32)P-orthophosphate for 30min is ∼1Gy IR. The absorbed dose of IR following an otherwise identical exposure to the low-energy (0.24MeV) ß-particle emitter (33)P-orthophosphate is ∼0.18Gy IR. We show that low-energy ß-particles emitted by (33)P induce a greater number of ionizing radiation-induced foci (IRIF) and greater ATM kinase signaling than energetic ß-particles emitted by (32)P. Hence, we demonstrate that it is inappropriate to use (33)P-orthophosphate as a negative control for (32)P-orthophosphate in experiments investigating DNA damage responses to DNA double-strand breaks (DSBs). Significantly, we show that ATM accumulates in the chromatin fraction when ATM kinase activity is inhibited during exposure to either radionuclide. Finally, we also show that chromosome aberrations accumulate in cells when ATM kinase activity is inhibited during exposure to ∼0.36Gy ß-particles emitted by (33)P. We therefore propose that direct cellular exposure to (33)P-orthophosphate is an excellent means to induce and label the IR-induced, ATM kinase-dependent phosphoproteome.


Subject(s)
Beta Particles , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/radiation effects , Chromosome Aberrations/radiation effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/radiation effects , Phosphorus Radioisotopes/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/radiation effects , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/radiation effects , Ataxia Telangiectasia Mutated Proteins , Chromatin/metabolism , DNA Damage/radiation effects , Enzyme Activation/radiation effects , Signal Transduction/radiation effects
12.
J Exp Med ; 176(3): 787-92, 1992 Sep 01.
Article in English | MEDLINE | ID: mdl-1512542

ABSTRACT

Interleukin 1 (IL-1) is a pluripotent cytokine involved in mediating a variety of physiological processes, including induction of cell proliferation upon wound healing. Treatment of quiescent FS-4 human dermal fibroblast cells with IL-1 activates c-myc gene transcription, and nuclear localization of NF-kappa B. Previously, we have noted that the murine c-myc gene contains two functional NF-kappa B sites located at -1101 to -1081 bp (upstream regulatory element [URE]) and +440 to +459 bp (internal regulatory element [IRE]) relative to the P1 promoter. Here we have demonstrated that IL-1 treatment induced binding of NF-kappa B-like proteins (p50/p65) to these c-myc elements. Heterologous promoter-CAT constructs driven by multiple copies of either the URE or IRE were IL-1 inducible when transfected into FS-4 cells. In contrast, constructs harboring elements with two G to C residue conversions, such that they were no longer able to bind NF-kappa B, were not responsive to IL-1. Mutation of these two base pairs at both NF-kappa B sites within a c-myc promoter/exon I-CAT construct, resulted in loss of inducibility with IL-1 upon transfection into quiescent FS-4 cells. Thus, IL-1 significantly induces c-myc expression through positive regulation by NF-kappa B, suggesting a role for this family of factors in activation of proliferation associated with wound healing.


Subject(s)
Gene Expression Regulation , Genes, myc , Interleukin-1/physiology , NF-kappa B/physiology , Transcription, Genetic , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/radiation effects , Fibroblasts , Forkhead Transcription Factors , Humans , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Nuclear Proteins/radiation effects , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcriptional Activation , Ultraviolet Rays
13.
Biochem Biophys Res Commun ; 397(2): 350-4, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20513357

ABSTRACT

Proteins are folded properly in the endoplasmic reticulum (ER). Various stress such as hypoxia, ischemia and starvation interfere with the ER function, causing ER stress, which is defined by the accumulation of unfolded protein (UP) in the ER. ER stress is prevented by the UP response (UPR) and ER-associated degradation (ERAD). These signaling pathways are activated by three major ER molecules, ATF6, IRE-1 and PERK. Using HaCaT cells, we investigated ER signaling in human keratinocytes irradiated by environmental doses of ultraviolet B (UVB). The expression of Ero1-L(alpha), an upstream signaling molecule of ER stress, decreased at 1-4h after 10 mJ/cm(2) irradiation, indicating that the environmental dose of UVB-induced ER stress in HaCaT cells, without growth retardation. Furthermore, expression of intact ATF6 was decreased and it was translocated to the nuclei. The expression of XBP-1, a downstream molecule of IRE-1, which is an ER chaperone whose expression is regulated by XBP-1, and UP ubiquitination were induced by 10 mJ/cm(2) UVB at 4h. PERK, which regulates apoptosis, was not phosphorylated. Our results demonstrate that UVB irradiation generates UP in HaCaT cells and that the UPR and ERAD systems are activated to protect cells from UVB-induced ER stress. This is the first report to show ER signaling in UVB-irradiated keratinocytes.


Subject(s)
Endoplasmic Reticulum/radiation effects , Environmental Exposure , Keratinocytes/radiation effects , Protein Folding/radiation effects , Stress, Physiological , Ultraviolet Rays , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/radiation effects , Active Transport, Cell Nucleus/drug effects , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/radiation effects , Endoplasmic Reticulum/metabolism , Endoribonucleases/metabolism , Endoribonucleases/radiation effects , Humans , Keratinocytes/metabolism , Membrane Proteins/metabolism , Membrane Proteins/radiation effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Regulatory Factor X Transcription Factors , Signal Transduction/radiation effects , Transcription Factors/metabolism , Transcription Factors/radiation effects , Ubiquitination , X-Box Binding Protein 1 , eIF-2 Kinase/metabolism
14.
Eur Biophys J ; 39(10): 1375-84, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20349312

ABSTRACT

The function of the E. coli lactose operon requires the binding of the tetrameric repressor protein to the operator DNA. We have previously shown that gamma-irradiation destabilises the repressor-operator complex because the repressor gradually loses its DNA-binding ability (Radiat Res 170:604-612, 2008). It was suggested that the observed oxidation of tyrosine residues and the concomitant structural changes of irradiated headpieces (DNA-binding domains of repressor monomers) could be responsible for the inactivation. To unravel the mechanisms that lead to repressor-operator complex destabilisation when tyrosine oxidation occurs, we have compared by molecular dynamic simulations two complexes: (1) the native complex formed by two headpieces and the operator DNA, and (2) the damaged complex, in which all tyrosines are replaced by their oxidation product 3,4-dihydroxyphenylalanine (DOPA). On a 20 ns time scale, MD results show effects consistent with complex destabilisation: increased flexibility, increased DNA bending, modification of the hydrogen bond network, and decrease of the positive electrostatic potential at the protein surface and of the global energy of DNA-protein interactions.


Subject(s)
DNA, Bacterial/radiation effects , DNA-Binding Proteins/radiation effects , Escherichia coli Proteins/radiation effects , Gamma Rays , Lac Repressors/radiation effects , Molecular Dynamics Simulation , Amino Acid Sequence , Base Sequence , Binding Sites/physiology , Binding Sites/radiation effects , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Dihydroxyphenylalanine/radiation effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Hydrogen Bonding , Lac Repressors/chemistry , Lac Repressors/metabolism , Models, Molecular , Molecular Sequence Data , Operator Regions, Genetic , Oxidation-Reduction , Static Electricity
15.
Dev Cell ; 4(4): 599-605, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12689597

ABSTRACT

Cell death in Drosophila is regulated by many of the same signals that control apoptosis in mammalian systems. For all the three major cell death pathways that have been described in humans, homologous components have been identified in Drosophila. Here we report that distinct pathways mediate UV-induced apoptosis at different developmental stages in the Drosophila embryo. In midstage embryos, UVC irradiation induces reaper expression and cell death through a mei-41(dATM)-dependent pathway; UVB does not have the same effect. In contrast, in pregastrulation embryos, both UVB and UVC promote apoptosis via transcriptional induction of the Drosophila Apaf-1/ced-4 homolog. This early UV response requires E2F but not mei-41 function and appears to be independent of DNA damage.


Subject(s)
Apoptosis/radiation effects , Drosophila melanogaster/embryology , Drosophila melanogaster/radiation effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/radiation effects , Signal Transduction/radiation effects , Ultraviolet Rays/adverse effects , Animals , Apoptosis/genetics , Caspases/genetics , Caspases/metabolism , Caspases/radiation effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/radiation effects , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/radiation effects , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/radiation effects , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Gastrula/cytology , Gastrula/metabolism , Gastrula/radiation effects , Inhibitor of Apoptosis Proteins , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Up-Regulation/genetics , Up-Regulation/radiation effects
16.
J Cell Biol ; 143(3): 563-75, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9813080

ABSTRACT

The subcellular distribution and posttranslational modification of human chromatin assembly factor 1 (CAF-1) have been investigated after UV irradiation of HeLa cells. In an asynchronous cell population only a subfraction of the two large CAF-1 subunits, p150 and p60, were found to exist in a chromatin-associated fraction. This fraction is most abundant during S phase in nonirradiated cells and is much reduced in G2 cells. After UV irradiation, the chromatin-associated form of CAF-1 dramatically increased in all cells irrespective of their position in the cell cycle. Such chromatin recruitment resembles that seen for PCNA, a DNA replication and repair factor. The chromatin-associated fraction of p60 was predominantly hypophosphorylated in nonirradiated G2 cells. UV irradiation resulted in the rapid recruitment to chromatin of phosphorylated forms of the p60 subunit. Furthermore, the amount of the p60 and p150 subunits of CAF-1 associated with chromatin was a function of the dose of UV irradiation. Consistent with these in vivo observations, we found that the amount of CAF-1 required to stimulate nucleosome assembly during the repair of UV photoproducts in vitro depended upon both the number of lesions and the phosphorylation state of CAF-1. The recruitment of CAF-1 to chromatin in response to UV irradiation of human cells described here supports a physiological role for CAF-1 in linking chromatin assembly to DNA repair.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/metabolism , Ultraviolet Rays , Animals , Cell Fractionation , Cell Nucleus/metabolism , Chromatin/radiation effects , Chromatin Assembly Factor-1 , DNA-Binding Proteins/physiology , DNA-Binding Proteins/radiation effects , G2 Phase , HeLa Cells , Humans , Octoxynol , Phosphorylation , Proliferating Cell Nuclear Antigen/physiology , Rabbits , Transcription Factors
17.
Mol Cell Biol ; 26(6): 2360-72, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508011

ABSTRACT

ARF encodes a potent tumor suppressor that antagonizes MDM2, a negative regulator of p53. ARF also suppresses the proliferation of cells lacking p53, and loss of ARF in p53-null mice, compared with ARF or p53 singly null mice, results in a broadened tumor spectrum and decreased tumor latency. To investigate the mechanism of p53-independent tumor suppression by ARF, potential interacting proteins were identified by yeast two-hybrid screen. The antiapoptotic transcriptional corepressor C-terminal binding protein 2 (CtBP2) was identified, and ARF interactions with both CtBP1 and CtBP2 were confirmed in vitro and in vivo. Interaction with ARF resulted in proteasome-dependent CtBP degradation. Both ARF-induced CtBP degradation and CtBP small interfering RNA led to p53-independent apoptosis in colon cancer cells. ARF induction of apoptosis was dependent on its ability to interact with CtBP, and reversal of ARF-induced CtBP depletion by CtBP overexpression abrogated ARF-induced apoptosis. CtBP proteins represent putative targets for p53-independent tumor suppression by ARF.


Subject(s)
ADP-Ribosylation Factors/metabolism , Apoptosis/physiology , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Protein p53/metabolism , ADP-Ribosylation Factors/genetics , Alcohol Oxidoreductases , Animals , Cell Nucleolus/metabolism , Cells, Cultured , Co-Repressor Proteins , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/radiation effects , Humans , Mice , Phosphoproteins/genetics , Phosphoproteins/radiation effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/radiation effects , Protein Transport , RNA, Small Interfering , Tumor Suppressor Protein p53/genetics , Two-Hybrid System Techniques , Ubiquitin/metabolism , Ultraviolet Rays
18.
Mutat Res ; 680(1-2): 17-24, 2009.
Article in English | MEDLINE | ID: mdl-19733688

ABSTRACT

The micronucleus-centromere assay using a pan-centromeric probe was used to assess chromosomal damage in lymphocytes of 47 industrial radiographers occupationally exposed to low dose ionizing radiation and 47 controls. The influence of genotype of DNA repair genes (XRCC1(399), XRCC3(241) and XPD(751)) on micronuclei (MN) frequency was also investigated. Centromere negative micronuclei (MNC-) frequency was significantly higher in radiographers than in controls, whereas similar centromere positive micronuclei (MNC+) frequency was observed in both groups. Poisson regression analyses revealed that the MNC- frequency was significantly associated with radiation occupational exposure and with cumulative-radiation doses in radiographers, after adjusting for confounding variables such as age, smoking, alcohol intake and genotypes. Compared to homozygous wild-type subjects, MNC- frequency in radiographers with variant XRCC3 genotype was significantly higher using univariate analysis. There were no differences in MNC- or MNC+ frequencies by genotype in controls. In conclusion, scoring of MNC- is a useful cytogenetic biomonitoring method for radiographers. Polymorphisms in XRCC3 might contribute to the increased genetic damage in individuals occupationally exposed to chronic ionizing radiation.


Subject(s)
Centromere/radiation effects , DNA-Binding Proteins/radiation effects , Gamma Rays/adverse effects , Micronuclei, Chromosome-Defective/radiation effects , Occupational Exposure/adverse effects , Polymorphism, Restriction Fragment Length/genetics , Adult , DNA Repair/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , Dose-Response Relationship, Radiation , Humans , Lymphocytes/radiation effects , Male , Micronucleus Tests , Radiation Monitoring , Radiography
19.
Cancer Res ; 67(12): 5649-57, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575131

ABSTRACT

Despite an extensive body of evidence linking UV radiation and melanoma tumorigenesis, a clear mechanistic understanding of this process is still lacking. Because heritable mutations in both INK4a and the nucleotide excision repair (NER) pathway predispose individuals to melanoma development, we set out to test the hypothesis that abrogation of NER, by deletion of the xeroderma pigmentosum C (Xpc) gene, will heighten melanoma photocarcinogenesis in an Ink4a-Arf-deficient background. Experimentally, we generated a strain of mice doubly deficient in Xpc and Ink4a-Arf and subjected wild-type, Xpc-/-Ink4a-Arf+/+, Xpc-/-Ink4a-Arf-/-, and Xpc+/+Ink4a-Arf-/- mice to a single neonatal (day P3) dose of UVB without additional chemical promotion. Indeed, there was a significant increase in the development of dermal spindle/epithelioid cell melanomas in Xpc-/-Ink4a-Arf-/- mice when compared with Xpc+/+Ink4a-Arf-/- mice (P = 0.005); wild-type and Xpc-/-Ink4a-Arf+/+ mice failed to develop tumors. These neoplasms bore a striking histologic resemblance to melanomas that arise in the Tyr-vHRAS/Ink4a-Arf-/- context and often expressed melanocyte differentiation marker Tyrp1, thus supporting their melanocytic origination. All strains, except wild-type mice, developed pigmented and non-pigmented epidermal-derived keratinocytic cysts, whereas Xpc+/+Ink4a-Arf-/- mice exhibited the greatest propensity for squamous cell carcinoma development. We then screened for NRas, HRas, Kras, and BRaf mutations in tumor tissue and detected a higher frequency of rare Kras(Q61) alterations in tumors from Xpc-/-Ink4a-Arf-/- mice compared with Xpc+/+Ink4a-Arf-/- mice (50% versus 7%, P = 0.033). Taken together, results from this novel UV-inducible melanoma model suggest that NER loss, in conjunction with Ink4a-Arf inactivation, can drive melanoma photocarcinogenesis possibly through signature Kras mutagenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , DNA Repair/radiation effects , DNA-Binding Proteins/radiation effects , Endonucleases/radiation effects , Melanoma, Experimental/genetics , Nuclear Proteins/radiation effects , Skin Neoplasms/genetics , Transcription Factors/radiation effects , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Epidermal Cyst/pathology , Melanoma, Experimental/pathology , Mice , Mice, Mutant Strains , Neoplasms, Radiation-Induced/genetics , Nuclear Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/pathology , Transcription Factors/metabolism , Ultraviolet Rays/adverse effects , ras Proteins/genetics , ras Proteins/radiation effects
20.
Trends Biochem Sci ; 16(9): 323-6, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1835191

ABSTRACT

Ultraviolet (UV) irradiation can initiate complex formation between proteins and DNA or RNA and so can be used to study such interactions. However, crosslink formation by standard UV light sources can take up to several hours. More recently, a beam of monochromatic UV light from a laser has been used to initiate crosslinking in nano- and picosecond time intervals. As noted in an earlier TIBS article 'the advantages of short pulse times and high-energy fluxes should make this a valuable technique in the future'. In this review we characterize laser-induced crosslinking and explore the applications of this method.


Subject(s)
DNA-Binding Proteins/radiation effects , Lasers , Ultraviolet Rays , Adenosine Triphosphatases/metabolism , Animals , Methods , Nucleic Acids/metabolism , Oligonucleotides/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL