Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
J Control Release ; 330: 1071-1079, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33188826

ABSTRACT

We report the synthesis of a series of symmetrical lipids composed of dihydroxyacetone and even­carbon fatty acids (eight to sixteen carbons), both components of the human metabolome, and characterize their formulation into porous microparticles through spontaneous emulsification without the use of additional porogens. Lipid hydrolysis products were identified by 1H NMR to validate lipid degradation into the parent metabolic synthons. Microparticle architecture, as determined by scanning electron microscopy, was lipid-length dependent, with shorter alkyl chains forming tight structures and longer alkyl chains forming larger pores with plate-like lipid architectures. In all cases, the lipids formed organized patterns, not irregular shapes. As a demonstration of the potential use of these solid lipid-based microparticles, the release kinetics of a model drug (piroxicam) was quantified showing that release was more greatly influenced by microparticle porosity, and hence surface area, than by hydrophobicity of the lipids.


Subject(s)
Dihydroxyacetone , Lipids , Humans , Microscopy, Electron, Scanning , Particle Size , Porosity
2.
Int J Mol Sci ; 19(8)2018 Jul 30.
Article in English | MEDLINE | ID: mdl-30061540

ABSTRACT

It has been well established that hypoxia significantly increases both cellular and tumor resistance to ionizing radiation. Hypoxia associated radiation resistance has been known for some time but there has been limited success in sensitizing cells to radiation under hypoxic conditions. These studies show that, when irradiated with low linear energy transfer (LET) gamma-rays, poly (ADP-ribose), polymerase (PARP), Fanconi Anemia (FANC), and mutant Chinese Hamster Ovary (CHO) cells respond similarly to the non-homologous end joining (NHEJ) and the homologous recombination (HR) repair mutant CHO cells. Comparable results were observed in cells exposed to 13 keV/µm carbon ions. However, when irradiated with higher LET spread out Bragg peak (SOBP) carbon ions, we observed a decrease in the oxygen enhancement ratio (OER) in all the DNA of repair mutant cell lines. Interestingly, PARP mutant cells were observed as having the largest decrease in OER. Finally, these studies show a significant increase in the relative biological effectiveness (RBE) of high LET SOBP carbon and iron ions in HR and PARP mutants. There was also an increase in the RBE of NHEJ mutants when irradiated to SOBP carbon and iron ions. However, this increase was lower than in other mutant cell lines. These findings indicate that high LET radiation produces unique types of DNA damage under hypoxic conditions and PARP and HR repair pathways play a role in repairing this damage.


Subject(s)
DNA Damage/radiation effects , Ovary/cytology , Ovary/radiation effects , Animals , CHO Cells , Cell Hypoxia/radiation effects , Cell Survival/radiation effects , Cricetinae , Cricetulus , DNA Repair/radiation effects , Female , Gamma Rays/adverse effects , Linear Energy Transfer , Micronucleus Tests , Ovary/metabolism , Oxygen/metabolism , Radiation, Ionizing
3.
Int J Mol Sci ; 19(2)2018 Feb 07.
Article in English | MEDLINE | ID: mdl-29414878

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase inhibitor, which has been widely utilized throughout the cancer research field. SAHA-induced radiosensitization in normal human fibroblasts AG1522 and lung carcinoma cells A549 were evaluated with a combination of γ-rays, proton, and carbon ion exposure. Growth delay was observed in both cell lines during SAHA treatment; 2 µM SAHA treatment decreased clonogenicity and induced cell cycle block in G1 phase but 0.2 µM SAHA treatment did not show either of them. Low LET (Linear Energy Transfer) irradiated A549 cells showed radiosensitization effects on cell killing in cycling and G1 phase with 0.2 or 2 µM SAHA pretreatment. In contrast, minimal sensitization was observed in normal human cells after low and high LET radiation exposure. The potentially lethal damage repair was not affected by SAHA treatment. SAHA treatment reduced the rate of γ-H2AX foci disappearance and suppressed RAD51 and RPA (Replication Protein A) focus formation. Suppression of DNA double strand break repair by SAHA did not result in the differences of SAHA-induced radiosensitization between human cancer cells and normal cells. In conclusion, our results suggest SAHA treatment will sensitize cancer cells to low and high LET radiation with minimum effects to normal cells.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , A549 Cells , Biomarkers, Tumor/analysis , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Elementary Particles/therapeutic use , Fibroblasts/drug effects , Gamma Rays/therapeutic use , Histone Deacetylase Inhibitors/adverse effects , Humans , Hydroxamic Acids/adverse effects , Photons/therapeutic use , Radiation-Sensitizing Agents/adverse effects , Vorinostat
4.
Methods Mol Biol ; 1599: 277-285, 2017.
Article in English | MEDLINE | ID: mdl-28477126

ABSTRACT

The combination of live cell imaging and laser micro-irradiation is an important technique to investigate the recruitment and kinetics of DNA repair molecules to DNA damage sites. In this chapter, we describe the detailed methods to study the dynamics of fluorescently tagged ATM protein kinase at laser-induced DNA double strand break (DSB) sites. The same protocol can be applied to analyze the recruitment and kinetics of other potential or known DNA repair proteins to DSB sites.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Damage/physiology , Humans , Phosphorylation/genetics , Phosphorylation/physiology
5.
Biochem Biophys Res Commun ; 477(2): 235-40, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27297111

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a serine/threonine kinase that plays an essential role in the repair of DNA double-strand breaks (DSBs) in the non-homologous end-joining (NHEJ) pathway. The DNA-PK holoenzyme consists of a catalytic subunit (DNA-PKcs) and DNA-binding subunit (Ku70/80, Ku). Ku is a molecular sensor for double-stranded DNA and once bound to DSB ends it recruits DNA-PKcs to the DSB site. Subsequently, DNA-PKcs is activated and heavily phosphorylated, with these phosphorylations modulating DNA-PKcs. Although phosphorylation of DNA-PKcs is well studied, other post-translational modifications of DNA-PKcs are not. In this study, we aimed to determine if acetylation of DNA-PKcs regulates DNA-PKcs-dependent DSB repair. We report that DNA-PKcs is acetylated in vivo and identified two putative acetylation sites, lysine residues 3241 and 3260. Mutating these sites to block potential acetylation results in increased radiosensitive, a slight decrease in DSB repair capacity as assessed by γH2AX resolution, and increased chromosomal aberrations, especially quadriradial chromosomes. Together, our results provide evidence that acetylation potentially regulates DNA-PKcs.


Subject(s)
DNA Damage/genetics , DNA-Activated Protein Kinase/genetics , DNA/genetics , Genomic Instability/genetics , Genomic Instability/radiation effects , Nuclear Proteins/genetics , Radiation Tolerance/genetics , Animals , CHO Cells , Cricetulus , DNA/radiation effects , DNA-Activated Protein Kinase/radiation effects , Dose-Response Relationship, Radiation , Endodeoxyribonucleases , Escherichia coli Proteins , Lysine/genetics , Lysine/radiation effects , Nuclear Proteins/radiation effects , Radiation Dosage , Structure-Activity Relationship
6.
Oncotarget ; 7(1): 46-65, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26695548

ABSTRACT

Faithful and complete genome replication in human cells is essential for preventing the accumulation of cancer-promoting mutations. WRN, the protein defective in Werner syndrome, plays critical roles in preventing replication stress, chromosome instability, and tumorigenesis. Herein, we report that ATR-mediated WRN phosphorylation is needed for DNA replication and repair upon replication stress. A serine residue, S1141, in WRN is phosphorylated in vivo by the ATR kinase in response to replication stress. ATR-mediated WRN S1141 phosphorylation leads to ubiquitination of WRN, facilitating the reversible interaction of WRN with perturbed replication forks and subsequent degradation of WRN. The dynamic interaction between WRN and DNA is required for the suppression of new origin firing and Rad51-dependent double-stranded DNA break repair. Significantly, ATR-mediated WRN phosphorylation is critical for the suppression of chromosome breakage during replication stress. These findings reveal a unique role for WRN as a modulator of DNA repair, replication, and recombination, and link ATR-WRN signaling to the maintenance of genome stability.


Subject(s)
DNA Replication , Exodeoxyribonucleases/metabolism , Proteasome Endopeptidase Complex/metabolism , RecQ Helicases/metabolism , Signal Transduction , Ubiquitins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Binding Sites/genetics , Blotting, Western , Cell Line, Tumor , Cells, Cultured , DNA Damage , DNA Repair , Exodeoxyribonucleases/genetics , Fluorescence Recovery After Photobleaching , HeLa Cells , Humans , Microscopy, Confocal , Phosphorylation , RecQ Helicases/genetics , Serine/genetics , Serine/metabolism , Werner Syndrome Helicase
7.
Nucleic Acids Res ; 44(4): 1732-45, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26712563

ABSTRACT

Multiple DNA double-strand break (DSB) repair pathways are active in S phase of the cell cycle; however, DSBs are primarily repaired by homologous recombination (HR) in this cell cycle phase. As the non-homologous end-joining (NHEJ) factor, Ku70/80 (Ku), is quickly recruited to DSBs in S phase, we hypothesized that an orchestrated mechanism modulates pathway choice between HR and NHEJ via displacement of the Ku heterodimer from DSBs to allow HR. Here, we provide evidence that phosphorylation at a cluster of sites in the junction of the pillar and bridge regions of Ku70 mediates the dissociation of Ku from DSBs. Mimicking phosphorylation at these sites reduces Ku's affinity for DSB ends, suggesting that phosphorylation of Ku70 induces a conformational change responsible for the dissociation of the Ku heterodimer from DNA ends. Ablating phosphorylation of Ku70 leads to the sustained retention of Ku at DSBs, resulting in a significant decrease in DNA end resection and HR, specifically in S phase. This decrease in HR is specific as these phosphorylation sites are not required for NHEJ. Our results demonstrate that the phosphorylation-mediated dissociation of Ku70/80 from DSBs frees DNA ends, allowing the initiation of HR in S phase and providing a mechanism of DSB repair pathway choice in mammalian cells.


Subject(s)
Antigens, Nuclear/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , S Phase/genetics , Animals , DNA Damage/genetics , DNA Repair/genetics , Fibroblasts/metabolism , HCT116 Cells , Homologous Recombination , Humans , Ku Autoantigen , Mice , Signal Transduction
8.
J Fluoresc ; 25(6): 1775-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26411799

ABSTRACT

Photobleaching is a key limitation in two-photon imaging of fluorescent proteins with femtosecond pulsed excitation. We present measurements of the dependence of eGFP photobleaching on the spectral amplitude and phase of the pulses used. A strong dependence on the excitation wavelength was confirmed and measured over a 800-950 nm range. A fiber continuum light source and pulse shaping techniques were used to investigate photobleaching with broadband, 15 fs transform limited, pulses with differing spectral amplitude and phase. Narrow band pulses, >150 fs transform limited, typical of femtosecond laser sources used in two-photon imaging applications, were also investigated for their photobleaching dependence on pulse dispersion and bandwidth. The bleach rate for broadband pulses was found to be primarily determined by the second harmonic spectrum of the excitation light. On the other hand, for narrow band excitation pulses with similar center wavelengths improvement in bleach rate was found to be mostly dependent on reducing the pulse length. A simple model to predict the relative bleach rates for broadband pulses is presented and compared to the experimental data.


Subject(s)
Green Fluorescent Proteins/chemistry , Photobleaching , Photons , Kinetics , Lasers , Spectrometry, Fluorescence
9.
Chem Biol ; 22(7): 849-61, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26119999

ABSTRACT

Regulatory mechanisms underlying γH2AX induction and the associated cell fate decision during DNA damage response (DDR) remain obscure. Here, we discover a bromodomain (BRD)-like module in DNA-PKcs (DNA-PKcs-BRD) that specifically recognizes H2AX acetyl-lysine 5 (K5ac) for sequential induction of γH2AX and concurrent cell fate decision(s). First, top-down mass spectrometry of radiation-phenotypic, full-length H2AX revealed a radiation-inducible, K5ac-dependent induction of γH2AX. Combined approaches of sequence-structure modeling/docking, site-directed mutagenesis, and biochemical experiments illustrated that through docking on H2AX K5ac, this non-canonical BRD determines not only the H2AX-targeting activity of DNA-PKcs but also the over-activation of DNA-PKcs in radioresistant tumor cells, whereas a Kac antagonist, JQ1, was able to bind to DNA-PKcs-BRD, leading to re-sensitization of tumor cells to radiation. This study elucidates the mechanism underlying the H2AX-dependent regulation of DNA-PKcs in ionizing radiation-induced, differential DDR, and derives an unconventional, non-catalytic domain target in DNA-PKs for overcoming resistance during cancer radiotherapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/chemistry , DNA-Activated Protein Kinase/metabolism , Histones/chemistry , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Sequence , Cell Line, Tumor , DNA Repair , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Lysine/metabolism , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Radiation Tolerance
10.
J Allergy Clin Immunol ; 135(6): 1578-88.e5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25842288

ABSTRACT

BACKGROUND: PRKDC encodes for DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a kinase that forms part of a complex (DNA-dependent protein kinase [DNA-PK]) crucial for DNA double-strand break repair and V(D)J recombination. In mice DNA-PK also interacts with the transcription factor autoimmune regulator (AIRE) to promote central T-cell tolerance. OBJECTIVE: We sought to understand the causes of an inflammatory disease with granuloma and autoimmunity associated with decreasing T- and B-cell counts over time that had been diagnosed in 2 unrelated patients. METHODS: Genetic, molecular, and functional analyses were performed to characterize an inflammatory disease evocative of a combined immunodeficiency. RESULTS: We identified PRKDC mutations in both patients. These patients exhibited a defect in DNA double-strand break repair and V(D)J recombination. Whole-blood mRNA analysis revealed a strong interferon signature. On activation, memory T cells displayed a skewed cytokine response typical of TH2 and TH1 but not TH17. Moreover, mutated DNA-PKcs did not promote AIRE-dependent transcription of peripheral tissue antigens in vitro. The latter defect correlated in vivo with production of anti-calcium-sensing receptor autoantibodies, which are typically found in AIRE-deficient patients. In addition, 9 months after bone marrow transplantation, patient 1 had Hashimoto thyroiditis, suggesting that organ-specific autoimmunity might be linked to nonhematopoietic cells, such as AIRE-expressing thymic epithelial cells. CONCLUSION: Deficiency of DNA-PKcs, a key AIRE partner, can present as an inflammatory disease with organ-specific autoimmunity, suggesting a role for DNA-PKcs in regulating autoimmune responses and maintaining AIRE-dependent tolerance in human subjects.


Subject(s)
DNA-Activated Protein Kinase/genetics , Granuloma/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , Nuclear Proteins/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics , Adolescent , Animals , Autoantibodies/biosynthesis , Autoimmunity/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , DNA End-Joining Repair/immunology , DNA-Activated Protein Kinase/deficiency , DNA-Activated Protein Kinase/immunology , Female , Gene Expression Regulation , Granuloma/immunology , Granuloma/metabolism , Granuloma/pathology , Humans , Immune Tolerance , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Male , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , Transcription Factors/immunology , V(D)J Recombination/immunology , Young Adult , AIRE Protein
11.
Oncol Lett ; 9(4): 1621-1627, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25789011

ABSTRACT

The present study investigated the effect of targeted mutations in the DNA-dependent protein kinase catalytic subunit and phosphorylation domains on the survival of cells in response to different qualities of ionizing radiation. Mutated Chinese hamster ovary V3 cells were exposed to 500 MeV/nucleon initial energy and 200 keV/µm monoenergetic Fe ions; 290 MeV/nucleon initial energy and average 50 keV/µm spread-out Bragg peak C ions; 70 MeV/nucleon initial energy and 1 keV/µm monoenergetic protons; and 0.663 MeV initial energy and 0.3 keV/µm Cs137 γ radiation. The results demonstrated that sensitivity to high linear energy transfer radiation is increased when both S2056 and T2609 clusters each contain a point mutation or multiple mutations are present in either cluster, whereas the phosphoinositide 3 kinase cluster only requires a single mutation to induce the sensitized phenotype of V3 cells. Additionally, the present study demonstrated that sensitivity to DNA cross-linking damage by cisplatin only requires a single mutation in one of the three clusters and that additional point mutations do not increase cell sensitivity.

12.
Radiat Res ; 183(3): 345-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25738894

ABSTRACT

The purpose of this study was to identify the roles of non-homologous end-joining (NHEJ) or homologous recombination (HR) pathways in repairing DNA double-strand breaks (DSBs) induced by exposure to high-energy protons and carbon ions (C ions) versus gamma rays in Chinese hamster cells. Two Chinese hamster cell lines, ovary AA8 and lung fibroblast V79, as well as various mutant sublines lacking DNA-PKcs (V3), X-ray repair cross-complementing protein-4 [XRCC4 (XR1), XRCC3 (irs1SF) and XRCC2 (irs1)] were exposed to gamma rays ((137)Cs), protons (200 MeV; 2.2 keV/µm) and C ions (290 MeV; 50 keV/µm). V3 and XR1 cells lack the NHEJ pathway, whereas irs1 and irs1SF cells lack the HR pathway. After each exposure, survival was measured using a clonogenic survival assay, in situ DSB induction was evaluated by immunocytochemical analysis of histone H2AX phosphorylation at serine 139 (γ-H2AX foci) and chromosome aberrations were examined using solid staining. The findings from this study showed that clonogenic survival clearly depended on the NHEJ and HR pathway statuses, and that the DNA-PKcs(-/-) cells (V3) were the most sensitive to all radiation types. While protons and γ rays yielded almost the same biological effects, C-ion exposure greatly enhanced the sensitivity of wild-type and HR-deficient cells. However, no significant enhancement of sensitivity in cell killing was seen after C-ion irradiation of NHEJ deficient cells. Decreases in the number of γ-H2AX foci after irradiation occurred more slowly in the NHEJ deficient cells. In particular, V3 cells had the highest number of residual γ-H2AX foci at 24 h after C-ion irradiation. Chromosomal aberrations were significantly higher in both the NHEJ- and HR-deficient cell lines than in wild-type cell lines in response to all radiation types. Protons and gamma rays induced the same aberration levels in each cell line, whereas C ions introduced higher but not significantly different aberration levels. Our results suggest that the NHEJ pathway plays an important role in repairing DSBs induced by both clinical proton and C-ion beams. Furthermore, in C ions the HR pathway appears to be involved in the repair of DSBs to a greater extent compared to gamma rays and protons.


Subject(s)
DNA Damage/radiation effects , DNA End-Joining Repair/radiation effects , DNA Repair/radiation effects , Recombination, Genetic/radiation effects , Animals , Cell Cycle/radiation effects , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heavy Ion Radiotherapy/adverse effects , Humans , X-Rays
14.
Cell Rep ; 9(4): 1387-401, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456133

ABSTRACT

WRN, the protein defective in Werner syndrome (WS), is a multifunctional nuclease involved in DNA damage repair, replication, and genome stability maintenance. It was assumed that the nuclease activities of WRN were critical for these functions. Here, we report a nonenzymatic role for WRN in preserving nascent DNA strands following replication stress. We found that lack of WRN led to shortening of nascent DNA strands after replication stress. Furthermore, we discovered that the exonuclease activity of MRE11 was responsible for the shortening of newly replicated DNA in the absence of WRN. Mechanistically, the N-terminal FHA domain of NBS1 recruits WRN to replication-associated DNA double-stranded breaks to stabilize Rad51 and to limit the nuclease activity of its C-terminal binding partner MRE11. Thus, this previously unrecognized nonenzymatic function of WRN in the stabilization of nascent DNA strands sheds light on the molecular reason for the origin of genome instability in WS individuals.


Subject(s)
DNA Replication , DNA/metabolism , Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Stress, Physiological , Animals , CHO Cells , Camptothecin/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded/drug effects , DNA Replication/drug effects , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/chemistry , Genomic Instability/drug effects , Humans , MRE11 Homologue Protein , Mice , Models, Biological , Nuclear Proteins/metabolism , Protein Transport/drug effects , Rad51 Recombinase/metabolism , RecQ Helicases/chemistry , Replication Protein A/metabolism , Stress, Physiological/drug effects , Structure-Activity Relationship , Werner Syndrome Helicase
15.
J Thorac Oncol ; 9(9 Suppl 2): S88-96, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25396317

ABSTRACT

Although the presence of nodal disease is prognostic in thymic malignancy, the significance of the extent of nodal disease has yet to be defined. Lymph node dissection has not been routinely performed, and there is currently no node map defined for thymic malignancy. To establish a universal language for reporting as well as characterize the staging of this disease more accurately, an empiric node map is proposed here. This was developed using prior classification systems, series reporting specifics of nodal involvement, anatomical studies of lymphatic drainage, and preexisting node maps of the chest as defined by the International Association for the Study of Lung Cancer and the neck as defined by the American Academy of Otolaryngology-Head and Neck Surgery and the American Society for Head and Neck Surgery. The development of this node map was a joint effort by the International Thymic Malignancy Interest Group and the Thymic Domain of the IASLC Staging and Prognostic Factors Committee. It was reviewed and subsequently approved by the members of ITMIG. This map will be used as an adjunct to define node staging as part of a universal stage classification for thymic malignancy. As more data are gathered using definitions set forth by this node map, a revision may be undertaken in the future.


Subject(s)
Lymph Nodes/pathology , Neoplasms, Glandular and Epithelial/classification , Thymus Neoplasms/classification , Humans , Lymphatic Metastasis , Neoplasms, Glandular and Epithelial/pathology , Thymus Neoplasms/pathology
16.
Radiother Oncol ; 113(1): 146-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25284063

ABSTRACT

BACKGROUND AND PURPOSE: The causes of tumor response variation to radiation remain obscure, thus hampering the development of predictive assays and strategies to decrease resistance. The present study evaluates the impact of host tumor stromal elements and the in vivo environment on tumor cell kill, and relationship between tumor cell radiosensitivity and the tumor control dose. MATERIAL AND METHODS: Five endpoints were evaluated and compared in a radiosensitive DNA double-strand break repair-defective (DNA-PKcs(-/-)) tumor line, and its DNA-PKcs repair competent transfected counterpart. In vitro colony formation assays were performed on in vitro cultured cells, on cells obtained directly from tumors, and on cells irradiated in situ. Permanent local control was assessed by the TCD50 assay. Vascular effects were evaluated by functional vascular density assays. RESULTS: The fraction of repair competent and repair deficient tumor cells surviving radiation did not substantially differ whether irradiated in vitro, i.e., in the absence of host stromal elements and factors, from the fraction of cells killed following in vivo irradiation. Additionally, the altered tumor cell sensitivity resulted in a proportional change in the dose required to achieve permanent local control. The estimated number of tumor cells per tumor, their cloning efficiency and radiosensitivity, all assessed by in vitro assays, were used to predict successfully, the measured tumor control doses. CONCLUSION: The number of clonogens per tumor and their radiosensitivity govern the permanent local control dose.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA-Activated Protein Kinase/metabolism , Neoplasms/radiotherapy , Radiation Tolerance/radiation effects , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Lethal Dose 50 , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/genetics , Radiation Tolerance/genetics , Radiotherapy Dosage , Stromal Cells/radiation effects , Transfection , Tumor Cells, Cultured
17.
Nucleic Acids Res ; 42(18): 11487-501, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25223785

ABSTRACT

Non-homologous end-joining (NHEJ) and homologous recombination (HR) are the two prominent pathways responsible for the repair of DNA double-strand breaks (DSBs). NHEJ is not restricted to a cell-cycle stage, whereas HR is active primarily in the S/G2 phases suggesting there are cell cycle-specific mechanisms that play a role in the choice between NHEJ and HR. Here we show NHEJ is attenuated in S phase via modulation of the autophosphorylation status of the NHEJ factor DNA-PKcs at serine 2056 by the pro-HR factor BRCA1. BRCA1 interacts with DNA-PKcs in a cell cycle-regulated manner and this interaction is mediated by the tandem BRCT domain of BRCA1, but surprisingly in a phospho-independent manner. BRCA1 attenuates DNA-PKcs autophosphorylation via directly blocking the ability of DNA-PKcs to autophosphorylate. Subsequently, blocking autophosphorylation of DNA-PKcs at the serine 2056 phosphorylation cluster promotes HR-required DNA end processing and loading of HR factors to DSBs and is a possible mechanism by which BRCA1 promotes HR.


Subject(s)
BRCA1 Protein/metabolism , DNA-Activated Protein Kinase/metabolism , S Phase , BRCA1 Protein/chemistry , Cell Line , DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/chemistry , HeLa Cells , Humans , Phosphorylation , Protein Structure, Tertiary , Radiation Tolerance , Recombinational DNA Repair , S Phase/genetics , Serine/metabolism
18.
Genes Dev ; 28(13): 1472-84, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24990965

ABSTRACT

The ATR (ATM [ataxia telangiectasia-mutated]- and Rad3-related) checkpoint is a crucial DNA damage signaling pathway. While the ATR pathway is known to transmit DNA damage signals through the ATR-Chk1 kinase cascade, whether post-translational modifications other than phosphorylation are important for this pathway remains largely unknown. Here, we show that protein SUMOylation plays a key role in the ATR pathway. ATRIP, the regulatory partner of ATR, is modified by SUMO2/3 at K234 and K289. An ATRIP mutant lacking the SUMOylation sites fails to localize to DNA damage and support ATR activation efficiently. Surprisingly, the ATRIP SUMOylation mutant is compromised in the interaction with a protein group, rather than a single protein, in the ATR pathway. Multiple ATRIP-interacting proteins, including ATR, RPA70, TopBP1, and the MRE11-RAD50-NBS1 complex, exhibit reduced binding to the ATRIP SUMOylation mutant in cells and display affinity for SUMO2 chains in vitro, suggesting that they bind not only ATRIP but also SUMO. Fusion of a SUMO2 chain to the ATRIP SUMOylation mutant enhances its interaction with the protein group and partially suppresses its localization and functional defects, revealing that ATRIP SUMOylation promotes ATR activation by providing a unique type of protein glue that boosts multiple protein interactions along the ATR pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Signal Transduction , Sumoylation , Adaptor Proteins, Signal Transducing/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/genetics , Enzyme Activation , HEK293 Cells , HeLa Cells , Humans , Protein Binding/genetics , Protein Transport , Ubiquitin-Conjugating Enzymes/metabolism
19.
PLoS Genet ; 10(6): e1004419, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24922507

ABSTRACT

Translesion synthesis (TLS) enables DNA replication through damaged bases, increases cellular DNA damage tolerance, and maintains genomic stability. The sliding clamp PCNA and the adaptor polymerase Rev1 coordinate polymerase switching during TLS. The polymerases Pol η, ι, and κ insert nucleotides opposite damaged bases. Pol ζ, consisting of the catalytic subunit Rev3 and the regulatory subunit Rev7, then extends DNA synthesis past the lesion. Here, we show that Rev7 binds to the transcription factor TFII-I in human cells. TFII-I is required for TLS and DNA damage tolerance. The TLS function of TFII-I appears to be independent of its role in transcription, but requires homodimerization and binding to PCNA. We propose that TFII-I bridges PCNA and Pol ζ to promote TLS. Our findings extend the general principle of component sharing among divergent nuclear processes and implicate TLS deficiency as a possible contributing factor in Williams-Beuren syndrome.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , Mad2 Proteins/metabolism , Transcription Factors, TFII/genetics , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , DNA-Directed DNA Polymerase/biosynthesis , Genomic Instability , HEK293 Cells , HeLa Cells , Humans , Mad2 Proteins/biosynthesis , Mad2 Proteins/genetics , Nuclear Proteins/biosynthesis , Nucleotidyltransferases/biosynthesis , Proliferating Cell Nuclear Antigen/biosynthesis , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factors, TFII/biosynthesis , Transcription Factors, TFII/metabolism
20.
Cell ; 157(3): 565-79, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766806

ABSTRACT

The mammalian heart has a remarkable regenerative capacity for a short period of time after birth, after which the majority of cardiomyocytes permanently exit cell cycle. We sought to determine the primary postnatal event that results in cardiomyocyte cell-cycle arrest. We hypothesized that transition to the oxygen-rich postnatal environment is the upstream signal that results in cell-cycle arrest of cardiomyocytes. Here, we show that reactive oxygen species (ROS), oxidative DNA damage, and DNA damage response (DDR) markers significantly increase in the heart during the first postnatal week. Intriguingly, postnatal hypoxemia, ROS scavenging, or inhibition of DDR all prolong the postnatal proliferative window of cardiomyocytes, whereas hyperoxemia and ROS generators shorten it. These findings uncover a protective mechanism that mediates cardiomyocyte cell-cycle arrest in exchange for utilization of oxygen-dependent aerobic metabolism. Reduction of mitochondrial-dependent oxidative stress should be an important component of cardiomyocyte proliferation-based therapeutic approaches.


Subject(s)
Cell Cycle Checkpoints , Myocytes, Cardiac/cytology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Cell Proliferation/drug effects , DNA Damage , Free Radical Scavengers/pharmacology , Mice , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...