59 results on '"Burkhard Jakob"'
Search Results
2. Targeting AKT-Dependent Regulation of Antioxidant Defense Sensitizes AKT-E17K Expressing Cancer Cells to Ionizing Radiation
- Author
-
Isabell Goetting, Safa Larafa, Katharina Eul, Mikhail Kunin, Burkhard Jakob, Johann Matschke, and Verena Jendrossek
- Subjects
AKT ,radioresistance ,antioxidant defense ,glycolysis ,hexokinase 2 ,glutathione synthase ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Aberrant activation of the phosphatidyl-inositol-3-kinase/protein kinase B (AKT) pathway has clinical relevance to radiation resistance, but the underlying mechanisms are incompletely understood. Protection against reactive oxygen species (ROS) plays an emerging role in the regulation of cell survival upon irradiation. AKT-dependent signaling participates in the regulation of cellular antioxidant defense. Here, we were interested to explore a yet unknown role of aberrant activation of AKT in regulating antioxidant defense in response to IR and associated radiation resistance.We combined genetic and pharmacologic approaches to study how aberrant activation of AKT impacts cell metabolism, antioxidant defense, and radiosensitivity. Therefore, we used TRAMPC1 (TrC1) prostate cancer cells overexpressing the clinically relevant AKT-variant AKT-E17K with increased AKT activity or wildtype AKT (AKT-WT) and analyzed the consequences of direct AKT inhibition (MK2206) and inhibition of AKT-dependent metabolic enzymes on the levels of cellular ROS, antioxidant capacity, metabolic state, short-term and long-term survival without and with irradiation.TrC1 cells expressing the clinically relevant AKT1-E17K variant were characterized by improved antioxidant defense compared to TrC1 AKT-WT cells and this was associated with increased radiation resistance. The underlying mechanisms involved AKT-dependent direct and indirect regulation of cellular levels of reduced glutathione (GSH). Pharmacologic inhibition of specific AKT-dependent metabolic enzymes supporting defense against oxidative stress, e.g., inhibition of glutathione synthase and glutathione reductase, improved eradication of clonogenic tumor cells, particularly of TrC1 cells overexpressing AKT-E17K.We conclude that improved capacity of TrC1 AKT-E17K cells to balance antioxidant defense with provision of energy and other metabolites upon irradiation compared to TrC1 AKT-WT cells contributes to their increased radiation resistance. Our findings on the importance of glutathione de novo synthesis and glutathione regeneration for radiation resistance of TrC1 AKT-E17K cells offer novel perspectives for improving radiosensitivity in cancer cells with aberrant AKT activity by combining IR with inhibitors targeting AKT-dependent regulation of GSH provision.
- Published
- 2022
- Full Text
- View/download PDF
3. The Ubiquitin Ligase RNF138 Cooperates with CtIP to Stimulate Resection of Complex DNA Double-Strand Breaks in Human G1-Phase Cells
- Author
-
Nicole B. Averbeck, Carina Barent, Burkhard Jakob, Tatyana Syzonenko, Marco Durante, and Gisela Taucher-Scholz
- Subjects
DNA double-strand break (DSB) ,complex DSBs ,DSB resection ,ubiquitination ,RNF138 ,CtIP ,Cytology ,QH573-671 - Abstract
DNA double-strand breaks (DSBs) represent the molecular origin of ionizing-radiation inflicted biological effects. An increase in the ionization density causes more complex, clustered DSBs that can be processed by resection also in G1 phase, where repair of resected DSBs is considered erroneous and may contribute to the increased biological effectiveness of heavy ions in radiotherapy. To investigate the resection regulation of complex DSBs, we exposed G1 cells depleted for different candidate factors to heavy ions or α-particle radiation. Immunofluorescence microscopy was used to monitor the resection marker RPA, the DSB marker γH2AX and the cell-cycle markers CENP-F and geminin. The Fucci system allowed to select G1 cells, cell survival was measured by clonogenic assay. We show that in G1 phase the ubiquitin ligase RNF138 functions in resection regulation. RNF138 ubiquitinates the resection factor CtIP in a radiation-dependent manner to allow its DSB recruitment in G1 cells. At complex DSBs, RNF138′s participation becomes more relevant, consistent with the observation that also resection is more frequent at these DSBs. Furthermore, deficiency of RNF138 affects both DSB repair and cell survival upon induction of complex DSBs. We conclude that RNF138 is a regulator of resection that is influenced by DSB complexity and can affect the quality of DSB repair in G1 cells.
- Published
- 2022
- Full Text
- View/download PDF
4. O-GlcNAcylation Affects the Pathway Choice of DNA Double-Strand Break Repair
- Author
-
Sera Averbek, Burkhard Jakob, Marco Durante, and Nicole B. Averbeck
- Subjects
O-GlcNAcylation ,DNA-DSB repair ,chromatin remodeling ,high LET ,particle irradiation ,ionizing radiation ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Exposing cells to DNA damaging agents, such as ionizing radiation (IR) or cytotoxic chemicals, can cause DNA double-strand breaks (DSBs), which are crucial to repair to maintain genetic integrity. O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) is a post-translational modification (PTM), which has been reported to be involved in the DNA damage response (DDR) and chromatin remodeling. Here, we investigated the impact of O-GlcNAcylation on the DDR, DSB repair and chromatin status in more detail. We also applied charged particle irradiation to analyze differences of O-GlcNAcylation and its impact on DSB repair in respect of spatial dose deposition and radiation quality. Various techniques were used, such as the γH2AX foci assay, live cell microscopy and Fluorescence Lifetime Microscopy (FLIM) to detect DSB rejoining, protein accumulation and chromatin states after treating the cells with O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA) inhibitors. We confirmed that O-GlcNAcylation of MDC1 is increased upon irradiation and identified additional repair factors related to Homologous Recombination (HR), CtIP and BRCA1, which were increasingly O-GlcNAcyated upon irradiation. This is consistent with our findings that the function of HR is affected by OGT inhibition. Besides, we found that OGT and OGA activity modulate chromatin compaction states, providing a potential additional level of DNA-repair regulation.
- Published
- 2021
- Full Text
- View/download PDF
5. Correlative Light and Electron Microscopy (CLEM) Analysis of Nuclear Reorganization Induced by Clustered DNA Damage Upon Charged Particle Irradiation
- Author
-
Susanne Tonnemacher, Mikhail Eltsov, and Burkhard Jakob
- Subjects
dna repair ,carbon ions ,radiation-induced damage ,chromatin structure ,electron microscopy ,clem ,dna and rna cytochemistry ,osmium ammine b ,chromemt ,dna-specific staining ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Chromatin architecture plays major roles in gene regulation as well as in the repair of DNA damaged by endogenous or exogenous factors, such as after radiation. Opening up the chromatin might provide the necessary accessibility for the recruitment and binding of repair factors, thus facilitating timely and correct repair. The observed formation of ionizing radiation-induced foci (IRIF) of factors, such as 53BP1, upon induction of DNA double-strand breaks have been recently linked to local chromatin decompaction. Using correlative light and electron microscopy (CLEM) in combination with DNA-specific contrasting for transmission electron microscopy or tomography, we are able to show that at the ultrastructural level, these DNA damage domains reveal a chromatin compaction and organization not distinguishable from regular euchromatin upon irradiation with carbon or iron ions. Low Density Areas (LDAs) at sites of particle-induced DNA damage, as observed after unspecific uranyl acetate (UA)-staining, are thus unlikely to represent pure chromatin decompaction. RNA-specific terbium-citrate (Tb) staining suggests rather a reduced RNA density contributing to the LDA phenotype. Our observations are discussed in the view of liquid-like phase separation as one of the mechanisms of regulating DNA repair.
- Published
- 2020
- Full Text
- View/download PDF
6. Application of Fluorescence Lifetime Imaging Microscopy of DNA Binding Dyes to Assess Radiation-Induced Chromatin Compaction Changes
- Author
-
Elham Abdollahi, Gisela Taucher-Scholz, and Burkhard Jakob
- Subjects
FLIM microcopy ,Hoechst 34580 ,Syto 13 ,chromatin compaction ,histone deacetylation inhibitor (HDACi) ,irradiation ,pile-up ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
In recent years several approaches have been developed to address the chromatin status and its changes in eukaryotic cells under different conditions—but only few are applicable in living cells. Fluorescence lifetime imaging microscopy (FLIM) is a functional tool that can be used for the inspection of the molecular environment of fluorophores in living cells. Here, we present the use of single organic minor groove DNA binder dyes in FLIM for measuring chromatin changes following modulation of chromatin structure in living cells. Treatment with histone deacetylase inhibitors led to an increased fluorescence lifetime indicating global chromatin decompaction, whereas hyperosmolarity decreased the lifetime of the used dyes, thus reflecting the expected compaction. In addition, we demonstrate that time domain FLIM data based on single photon counting should be optimized using pile-up and counting loss correction, which affect the readout even at moderate average detector count rates in inhomogeneous samples. Using these corrections and utilizing Hoechst 34580 as chromatin compaction probe, we measured a pan nuclear increase in the lifetime following irradiation with X-rays in living NIH/3T3 cells thus providing a method to measure radiation-induced chromatin decompaction.
- Published
- 2018
- Full Text
- View/download PDF
7. Induction and Processing of the Radiation-Induced Gamma-H2AX Signal and Its Link to the Underlying Pattern of DSB: A Combined Experimental and Modelling Study.
- Author
-
Francesco Tommasino, Thomas Friedrich, Burkhard Jakob, Barbara Meyer, Marco Durante, and Michael Scholz
- Subjects
Medicine ,Science - Abstract
We present here an analysis of DSB induction and processing after irradiation with X-rays in an extended dose range based on the use of the γH2AX assay. The study was performed by quantitative flow cytometry measurements, since the use of foci counting would result in reasonable accuracy only in a limited dose range of a few Gy. The experimental data are complemented by a theoretical analysis based on the GLOBLE model. In fact, original aim of the study was to test GLOBLE predictions against new experimental data, in order to contribute to the validation of the model. Specifically, the γH2AX signal kinetics has been investigated up to 24 h after exposure to increasing photon doses between 2 and 500 Gy. The prolonged persistence of the signal at high doses strongly suggests dose dependence in DSB processing after low LET irradiation. Importantly, in the framework of our modelling analysis, this is related to a gradually increased fraction of DSB clustering at the micrometre scale. The parallel study of γH2AX dose response curves shows the onset of a pronounced saturation in two cell lines at a dose of about 20 Gy. This dose is much lower than expected according to model predictions based on the values usually adopted for the DSB induction yield (≈ 30 DSB/Gy) and for the γH2AX foci extension of approximately 2 Mbp around the DSB. We show and discuss how theoretical predictions and experimental findings can be in principle reconciled by combining an increased DSB induction yield with the assumption of a larger genomic extension for the single phosphorylated regions. As an alternative approach, we also considered in our model the possibility of a 3D spreading-mechanism of the H2AX phosphorylation around the induced DSB, and applied it to the analysis of both the aspects considered. Our results are found to be supportive for the basic assumptions on which GLOBLE is built. Apart from giving new insights into the H2AX phosphorylation process, experiments performed at high doses are of relevance in the context of radiation therapy, where hypo-fractionated schemes become increasingly popular.
- Published
- 2015
- Full Text
- View/download PDF
8. ATM alters the otherwise robust chromatin mobility at sites of DNA double-strand breaks (DSBs) in human cells.
- Author
-
Annabelle Becker, Marco Durante, Gisela Taucher-Scholz, and Burkhard Jakob
- Subjects
Medicine ,Science - Abstract
Ionizing radiation induces DNA double strand breaks (DSBs) which can lead to the formation of chromosome rearrangements through error prone repair. In mammalian cells the positional stability of chromatin contributes to the maintenance of genome integrity. DSBs exhibit only a small, submicron scale diffusive mobility, but a slight increase in the mobility of chromatin domains by the induction of DSBs might influence repair fidelity and the formation of translocations. The radiation-induced local DNA decondensation in the vicinity of DSBs is one factor potentially enhancing the mobility of DSB-containing chromatin domains. Therefore in this study we focus on the influence of different chromatin modifying proteins, known to be activated by the DNA damage response, on the mobility of DSBs. IRIF (ionizing radiation induced foci) in U2OS cells stably expressing 53BP1-GFP were used as a surrogate marker of DSBs. Low angle charged particle irradiation, known to trigger a pronounced DNA decondensation, was used for the defined induction of linear tracks of IRIF. Our results show that movement of IRIF is independent of the investigated chromatin modifying proteins like ACF1 or PARP1 and PARG. Also depletion of proteins that tether DNA strands like MRE11 and cohesin did not alter IRIF dynamics significantly. Inhibition of ATM, a key component of DNA damage response signaling, resulted in a pronounced confinement of DSB mobility, which might be attributed to a diminished radiation induced decondensation. This confinement following ATM inhibition was confirmed using X-rays, proving that this effect is not restricted to densely ionizing radiation. In conclusion, repair sites of DSBs exhibit a limited mobility on a small spatial scale that is mainly unaffected by depletion of single remodeling or DNA tethering proteins. However, it relies on functional ATM kinase which is considered to influence the chromatin structure after irradiation.
- Published
- 2014
- Full Text
- View/download PDF
9. Spatiotemporal dynamics of early DNA damage response proteins on complex DNA lesions.
- Author
-
Frank Tobias, Daniel Löb, Nicor Lengert, Marco Durante, Barbara Drossel, Gisela Taucher-Scholz, and Burkhard Jakob
- Subjects
Medicine ,Science - Abstract
The response of cells to ionizing radiation-induced DNA double-strand breaks (DSB) is determined by the activation of multiple pathways aimed at repairing the injury and maintaining genomic integrity. Densely ionizing radiation induces complex damage consisting of different types of DNA lesions in close proximity that are difficult to repair and may promote carcinogenesis. Little is known about the dynamic behavior of repair proteins on complex lesions. In this study we use live-cell imaging for the spatio-temporal characterization of early protein interactions at damage sites of increasing complexity. Beamline microscopy was used to image living cells expressing fluorescently-tagged proteins during and immediately after charged particle irradiation to reveal protein accumulation at damaged sites in real time. Information on the mobility and binding rates of the recruited proteins was obtained from fluorescence recovery after photobleaching (FRAP). Recruitment of the DNA damage sensor protein NBS1 accelerates with increasing lesion density and saturates at very high damage levels. FRAP measurements revealed two different binding modalities of NBS1 to damage sites and a direct impact of lesion complexity on the binding. Faster recruitment with increasing lesion complexity was also observed for the mediator MDC1, but mobility was limited at very high damage densities due to nuclear-wide binding. We constructed a minimal computer model of the initial response to DSB based on known protein interactions only. By fitting all measured data using the same set of parameters, we can reproduce the experimentally characterized steps of the DNA damage response over a wide range of damage densities. The model suggests that the influence of increasing lesion density accelerating NBS1 recruitment is only dependent on the different binding modes of NBS1, directly to DSB and to the surrounding chromatin via MDC1. This elucidates an impact of damage clustering on repair without the need of invoking extra processing steps.
- Published
- 2013
- Full Text
- View/download PDF
10. Cryo-electron tomography and deep learning denoising reveal native chromatin landscapes of interphase nuclei
- Author
-
Fadwa Fatmaoui, Pascal Carrivain, Diana Grewe, Burkhard Jakob, Jean-Marc Victor, Amélie Leforestier, and Mikhail Eltsov
- Abstract
The folding of nucleosome chains influences DNA availability for functional interactions necessary to the regulation of transcription, DNA replication and repair. Despite models based on in vitro studies, the nucleosome chain geometry within the crowded cell nucleus remains elusive. Using cryo-electron tomography and deep learning-based denoising, we directly observed the path of nucleosomal and linker DNA in situ in unstained flash-frozen Drosophila embryos. We quantified linker length and curvature characterizing a disordered zig-zag chromatin folding motif, with a low degree of linker bending. Additionally, nucleosome conformational variability with non-canonical structures and sub-nucleosomal particles were seen as individual objects, without structure averaging, highlighting the high structural heterogeneity of native chromatin.One-Sentence SummaryCryo-ET reveals local zig-zag motifs in interphase chromatin, a range of nucleosome conformations, and sub-nucleosomal particles.
- Published
- 2022
11. X-ray irradiation triggers immune response in human T-lymphocytes via store-operated $Ca^{2+}$ entry and NFAT activation
- Author
-
Dominique Tandl, Tim Sponagel, Dalia Alansary, Sebastian Fuck, Timo Smit, Stephanie Hehlgans, Burkhard Jakob, Claudia Fournier, Barbara A. Niemeyer, Franz Rödel, Bastian Roth, Anna Moroni, and Gerhard Thiel
- Subjects
ORAI1 Protein ,Physiology ,T-Lymphocytes ,X-Rays ,Immunity ,Leukocytes, Mononuclear ,Humans ,Calcium ,Calcium Signaling ,Stromal Interaction Molecule 1 ,ddc:610 - Abstract
The journal of general physiology 154(5), e202112865 (2022). doi:10.1085/jgp.202112865, Published by Rockefeller Univ. Press, New York, NY
- Published
- 2022
12. DNA double-strand breaks in heterochromatin elicit fast repair protein recruitment, histone H2AX phosphorylation and relocation to euchromatin
- Author
-
Sandro Conrad, Burkhard Jakob, Kay-Obbe Voss, Marco Durante, Markus Löbrich, Jörn Splinter, Daniele Zink, Gisela Taucher-Scholz, B., Jakob, J., Splinter, S., Conrad, K., Vo, D., Zink, Durante, Marco, M., Löbrich, and G., Taucher Scholz
- Subjects
DNA Repair ,Euchromatin ,Chromosomal Proteins, Non-Histone ,cells ,metabolism, DNA Break ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Genome Integrity, Repair and Replication ,Animals, Cell Cycle Protein ,Histones ,Mice ,chemistry.chemical_compound ,Heterochromatin ,metabolism, HeLa Cells, Heterochromatin ,DNA Breaks, Double-Stranded ,Phosphorylation ,Cells, Cultured ,Histone H2AX ,metabolism, Tumor Suppressor Protein ,metabolism, Cell ,Cell biology ,Cultured, Chromosomal Protein ,DNA-Binding Proteins ,Histone ,biological phenomena, cell phenomena, and immunity ,DNA repair ,DNA damage ,Protein Serine-Threonine Kinases ,Biology ,Genetics ,metabolism, Histone ,Animals ,Humans ,Double-Stranded, DNA Repair, DNA-Binding Protein ,metabolism, Humans, Kinetics, Mice, Phosphorylation, Protein-Serine-Threonine Kinase ,XRCC1 Gene ,Tumor Suppressor Proteins ,fungi ,Non-Histone ,Molecular biology ,metabolism, Euchromatin ,Kinetics ,enzymes and coenzymes (carbohydrates) ,X-ray Repair Cross Complementing Protein 1 ,chemistry ,Chromobox Protein Homolog 5 ,biology.protein ,metabolism ,DNA ,HeLa Cells - Abstract
DNA double-strand breaks (DSBs) can induce chromosomal aberrations and carcinogenesis and their correct repair is crucial for genetic stability. The cellular response to DSBs depends on damage signaling including the phosphorylation of the histone H2AX (��H2AX). However, a lack of ��H2AX formation in heterochromatin (HC) is generally observed after DNA damage induction. Here, we examine ��H2AX and repair protein foci along linear ion tracks traversing heterochromatic regions in human or murine cells and find the DSBs and damage signal streaks bending around highly compacted DNA. Given the linear particle path, such bending indicates a relocation of damage from the initial induction site to the periphery of HC. Real-time imaging of the repair protein GFP-XRCC1 confirms fast recruitment to heterochromatic lesions inside murine chromocenters. Using single-ion microirradiation to induce localized DSBs directly within chromocenters, we demonstrate that H2AX is early phosphorylated within HC, but the damage site is subsequently expelled from the center to the periphery of chromocenters within ���20 min. While this process can occur in the absence of ATM kinase, the repair of DSBs bordering HC requires the protein. Finally, we describe a local decondensation of HC at the sites of ion hits, potentially allowing for DSB movement via physical forces.
- Published
- 2022
- Full Text
- View/download PDF
13. O-GlcNAcylation Affects the Pathway Choice of DNA Double-Strand Break Repair
- Author
-
Nicole B. Averbeck, Sera Averbek, Marco Durante, Burkhard Jakob, Averbek, S., Jakob, B., Durante, M., and Averbeck, N. B.
- Subjects
DNA Repair ,QH301-705.5 ,DNA damage ,Cell ,N-Acetylglucosaminyltransferases ,Catalysis ,Chromatin remodeling ,Article ,chromatin remodeling ,Inorganic Chemistry ,DNA-DSB repair ,chemistry.chemical_compound ,O-GlcNAcylation ,Radiation, Ionizing ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Linear Energy Transfer ,Biology (General) ,Physical and Theoretical Chemistry ,Homologous Recombination ,QD1-999 ,Molecular Biology ,Spectroscopy ,high LET ,particle irradiation ,Organic Chemistry ,General Medicine ,Chromatin Assembly and Disassembly ,Double Strand Break Repair ,Chromatin ,Computer Science Applications ,MDC1 ,Cell biology ,DNA-Binding Proteins ,Chemistry ,medicine.anatomical_structure ,chemistry ,ddc:540 ,Homologous recombination ,ionizing radiation ,Protein Processing, Post-Translational ,DNA ,HeLa Cells - Abstract
Exposing cells to DNA damaging agents, such as ionizing radiation (IR) or cytotoxic chemicals, can cause DNA double-strand breaks (DSBs), which are crucial to repair to maintain genetic integrity. O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) is a post-translational modification (PTM), which has been reported to be involved in the DNA damage response (DDR) and chromatin remodeling. Here, we investigated the impact of O-GlcNAcylation on the DDR, DSB repair and chromatin status in more detail. We also applied charged particle irradiation to analyze differences of O-GlcNAcylation and its impact on DSB repair in respect of spatial dose deposition and radiation quality. Various techniques were used, such as the γH2AX foci assay, live cell microscopy and Fluorescence Lifetime Microscopy (FLIM) to detect DSB rejoining, protein accumulation and chromatin states after treating the cells with O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA) inhibitors. We confirmed that O-GlcNAcylation of MDC1 is increased upon irradiation and identified additional repair factors related to Homologous Recombination (HR), CtIP and BRCA1, which were increasingly O-GlcNAcyated upon irradiation. This is consistent with our findings that the function of HR is affected by OGT inhibition. Besides, we found that OGT and OGA activity modulate chromatin compaction states, providing a potential additional level of DNA-repair regulation.
- Published
- 2021
14. X-ray Irradiation activates immune response in human T-lymphocytes by eliciting a Ca2+signaling cascade
- Author
-
Claudia Fournier, Burkhard Jakob, F. Roedel, T. Smit, Stephanie Hehlgans, Anna Moroni, T. Sponagel, Bastian Roth, Sebastian Fuck, Dominique Tandl, and Gerhard Thiel
- Subjects
education.field_of_study ,Cytosol ,Immune system ,Chemistry ,Cancer cell ,Population ,NFAT ,education ,Transcription factor ,Jurkat cells ,Peripheral blood mononuclear cell ,Cell biology - Abstract
Radiation therapy is efficiently employed for eliminating cancer cells and reducing tumor growth. To further improving its therapeutic application it is mandatory to unravel the molecular effects of ionizing irradiation and to understand whether they support or counteract tumor therapy. Here we examine the impact of X-ray irradiation on immune activation of human T cells with single doses typically employed in tumor therapy. We discover that exposing cells to radiation triggers in a population of leukemic Jurkat T cells and in peripheral blood mononuclear cells (PBMCs) a canonical Ca2+signaling cascade, which elicits immune activation of these cells. An early step in the signaling cascade is the initiation of sustained oscillations of the cytosolic Ca2+concentration, an event mediated by store operated Ca2+entry (SOCE) via an X-ray induced clustering of the Calcium Release-Activated Calcium Modulator 1 with the stromal interaction molecule 1 (Oari1/STIM1). A functional consequence of the Ca2+signaling cascade is the translocation of the transcription factor nuclear factor of activated T cells (NFAT) from the cytosol into the nucleus where it elicits the expression of genes required for immune activation. These data imply that a direct activation of blood immune cells by ionizing irradiation has an impact on toxicity and therapeutic effects of radiation therapy.
- Published
- 2020
15. Differential Repair Protein Recruitment at Sites of Clustered and Isolated DNA Double-Strand Breaks Produced by High-Energy Heavy Ions
- Author
-
Gisela Taucher-Scholz, Burkhard Jakob, Alina Schmidt, Monika Dubiak-Szepietowska, Ellen Janiel, Marco Durante, Jakob, B., Dubiak-Szepietowska, M., Janiel, E., Schmidt, A., Durante, M., and Taucher-Scholz, G.
- Subjects
High energy ,Cell biology ,DNA Repair ,genetic processes ,Biophysics ,lcsh:Medicine ,Cell Cycle Proteins ,Plasma protein binding ,Article ,chemistry.chemical_compound ,Live cell imaging ,Cell Line, Tumor ,Neoplasms ,Humans ,DNA Breaks, Double-Stranded ,Heavy Ions ,lcsh:Science ,Cancer ,Double strand ,Multidisciplinary ,Binding Sites ,Chemistry ,lcsh:R ,fungi ,Nuclear Proteins ,Base excision repair ,DNA ,enzymes and coenzymes (carbohydrates) ,Cell culture ,Protein recruitment ,health occupations ,lcsh:Q ,biological phenomena, cell phenomena, and immunity ,Tumor Suppressor p53-Binding Protein 1 ,ddc:600 ,Synchrotrons ,Protein Binding - Abstract
Scientific reports 10(1), 1443 (2020). doi:10.1038/s41598-020-58084-6, Published by Macmillan Publishers Limited, part of Springer Nature, [London]
- Published
- 2020
16. Correlative Light and Electron Microscopy (CLEM) Analysis of Nuclear Reorganization Induced by Clustered DNA Damage Upon Charged Particle Irradiation
- Author
-
Mikhail Eltsov, Burkhard Jakob, and Susanne Tonnemacher
- Subjects
Electron Microscope Tomography ,Euchromatin ,Uranyl acetate ,law.invention ,lcsh:Chemistry ,chemistry.chemical_compound ,Mice ,law ,ChromEMT ,DNA Breaks, Double-Stranded ,Heavy Ions ,DNA-specific staining ,lcsh:QH301-705.5 ,Spectroscopy ,chromatin structure ,Chemistry ,General Medicine ,CLEM ,Chromatin ,Computer Science Applications ,Phenotype ,ddc:540 ,Osmium ammine B ,DNA repair ,DNA damage ,Catalysis ,Article ,DNA and RNA cytochemistry ,Inorganic Chemistry ,Microscopy, Electron, Transmission ,Cell Line, Tumor ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,electron microscopy ,radiation-induced damage ,Organic Chemistry ,Microscopy, Electron ,carbon ions ,lcsh:Biology (General) ,lcsh:QD1-999 ,Ultrastructure ,Biophysics ,NIH 3T3 Cells ,Electron microscope ,DNA ,DNA Damage - Abstract
International journal of molecular sciences 21(6), 1911 (2020). doi:10.3390/ijms21061911, Published by Molecular Diversity Preservation International, Basel
- Published
- 2020
- Full Text
- View/download PDF
17. Intersectin 1 is a component of the Reelin pathway to regulate neuronal migration and synaptic plasticity in the hippocampus
- Author
-
Gaga Kochlamazashvili, Burkhard Jakob, Volker Haucke, Aziz Gauhar, Tanja Maritzen, Hans H. Bock, and Maria Jäpel
- Subjects
0301 basic medicine ,Low-density lipoprotein receptor-related protein 8 ,Cell Adhesion Molecules, Neuronal ,Very Low-Density Lipoprotein Receptor ,Hippocampus ,Lissencephaly ,Nerve Tissue Proteins ,Receptors, N-Methyl-D-Aspartate ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,medicine ,Animals ,Reelin ,LDL-Receptor Related Proteins ,Mice, Knockout ,Neurons ,Extracellular Matrix Proteins ,Neuronal Plasticity ,Multidisciplinary ,biology ,Serine Endopeptidases ,multidomain scaffold ,endocytosis ,synaptic plasticity ,Reelin signaling ,hippocampus ,Long-term potentiation ,Biological Sciences ,DAB1 ,medicine.disease ,Adaptor Proteins, Vesicular Transport ,Reelin Protein ,030104 developmental biology ,Receptors, LDL ,nervous system ,Synaptic plasticity ,biology.protein ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Brain development and function depend on the directed and coordinated migration of neurons from proliferative zones to their final position. The secreted glycoprotein Reelin is an important factor directing neuronal migration. Loss of Reelin function results in the severe developmental disorder lissencephaly and is associated with neurological diseases in humans. Reelin signals via the lipoprotein receptors very low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2), but the exact mechanism by which these receptors control cellular function is poorly understood. We report that loss of the signaling scaffold intersectin 1 (ITSN1) in mice leads to defective neuronal migration and ablates Reelin stimulation of hippocampal long-term potentiation (LTP). Knockout (KO) mice lacking ITSN1 suffer from dispersion of pyramidal neurons and malformation of the radial glial scaffold, akin to the hippocampal lamination defects observed in VLDLR or ApoER2 mutants. ITSN1 genetically interacts with Reelin receptors, as evidenced by the prominent neuronal migration and radial glial defects in hippocampus and cortex seen in double-KO mice lacking ITSN1 and ApoER2. These defects were similar to, albeit less severe than, those observed in Reelin-deficient or VLDLR/ ApoER2 double-KO mice. Molecularly, ITSN1 associates with the VLDLR and its downstream signaling adaptor Dab1 to facilitate Reelin signaling. Collectively, these data identify ITSN1 as a component of Reelin signaling that acts predominantly by facilitating the VLDLR-Dab1 axis to direct neuronal migration in the cortex and hippocampus and to augment synaptic plasticity.
- Published
- 2017
18. Clustered double-strand breaks in heterochromatin perturb DNA repair after high linear energy transfer irradiation
- Author
-
Sara Timm, Yvonne Lorat, Burkhard Jakob, Gisela Taucher-Scholz, and Claudia E. Rübe
- Subjects
0301 basic medicine ,DNA Repair ,DNA repair ,Heterochromatin ,Linear energy transfer ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Radioresistance ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Linear Energy Transfer ,Radiology, Nuclear Medicine and imaging ,Irradiation ,Cells, Cultured ,Radiotherapy ,Chemistry ,DNA ,Hematology ,Fibroblasts ,Chromatin ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Biophysics ,Nucleus - Abstract
Background and purpose High linear energy transfer (LET) radiotherapy offers superior dose conformity and biological effectiveness compared with low-LET radiotherapy, representing a promising alternative for radioresistant tumours. A prevailing hypothesis is that energy deposition along the high-LET particle trajectories induces DNA lesions that are more complex and clustered and therefore more challenging to repair. The precise molecular mechanisms underlying the differences in radiobiological effects between high-LET and low-LET radiotherapies remain unclear. Material and Methods Human fibroblasts were irradiated with high-LET carbon ions or low-LET photons. At 0.5 h and 5 h post exposure, the DNA-damage pattern in the chromatin ultrastructure was visualised using gold-labelled DNA-repair factors. The induction and repair of single-strand breaks, double-strand breaks (DSBs), and clustered lesions were analysed in combination with terminal dUTP nick-end labelling of DNA breaks. Results High-LET irradiation induced clustered lesions with multiple DSBs along ion trajectories predominantly in heterochromatic regions. The cluster size increased over time, suggesting inefficient DSB repair. Low-LET irradiation induced many isolated DSBs throughout the nucleus, most of which were efficiently rejoined. Conclusions The clustering of DSBs in heterochromatin following high-LET irradiation perturbs efficient DNA repair, leading to greater biological effectiveness of high-LET irradiation versus that of low-LET irradiation.
- Published
- 2016
19. Upgrading the GSI beamline microscope with a confocal fluorescence lifetime scanner to monitor charged particle induced chromatin decondensation in living cells
- Author
-
Burkhard Jakob, Marco Durante, Gisela Taucher-Scholz, Elham Abdollahi, Abdollahi, Elham, Taucher Scholz, Gisela, Durante, Marco, and Jakob, Burkhard
- Subjects
Chromatin decondensation ,Ionizing radiation ,Nuclear and High Energy Physics ,Materials science ,Microscope ,Confocal ,DNA repair ,DNA condensation ,Fluorescence ,Linear particle accelerator ,Photon counting ,Chromatin ,law.invention ,Nuclear magnetic resonance ,Beamline ,law ,Heterochromatin ,FLIM microscopy ,Instrumentation - Abstract
We report the upgrade of the GSI beamline microscope coupled to the linear accelerator UNILAC by a confocal FLIM scanner utilizing time correlated single photon counting technique (TCSPC). The system can now be used to address the radiation induced chromatin decondensation in more detail and with higher sensitivity compared to intensity based methods. This decondensation of heterochromatic areas is one of the early DNA damage responses observed after charged particle irradiation and might facilitate the further processing of the induced lesions. We describe here the establishment of different DNA dyes as chromatin compaction probes usable for quantification of the DNA condensation status in living cells utilizing lifetime imaging. In addition, we find an evidence of heterochromatic chromatin decondensation in ion irradiated murine chromocenters detected after subsequent fixation using FLIM measurements.
- Published
- 2015
20. Phosphorylation of Ku dictates DNA double-strand break (DSB) repair pathway choice in S phase
- Author
-
Shu-Chi Wang, Kyung Jong Lee, Gisela Taucher-Scholz, Janapriya Saha, Anthony J. Davis, Shih Ya Wang, Kazi R. Fattah, Jingxin Sun, Burkhard Jakob, Linfeng Chi, and David J. Chen
- Subjects
0301 basic medicine ,Ku80 ,DNA End-Joining Repair ,DNA Repair ,genetic processes ,Biology ,Genome Integrity, Repair and Replication ,Cell cycle phase ,S Phase ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Genetics ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Homologous Recombination ,Ku Autoantigen ,S phase ,Ku70 ,fungi ,Antigens, Nuclear ,Fibroblasts ,HCT116 Cells ,Molecular biology ,Cell biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,chemistry ,Phosphorylation ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,DNA ,DNA Damage ,Signal Transduction - 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.
- Published
- 2015
21. ATM-dependent phosphorylation of MRE11 controls extent of resection during homology directed repair by signalling through Exonuclease 1
- Author
-
Frank Tobias, Nuri Gueven, Ernst J. Wolvetang, Magtouf Gatei, Amanda W. Kijas, Burkhard Jakob, Patrick Concannon, Gisela Taucher-Scholz, Emma Bolderson, Karen Cerosaletti, Greg Oakley, Kum Kum Khanna, Martin F. Lavin, Lisa Wiesmüller, and Yi Chieh Lim
- Subjects
DNA repair ,DNA damage ,DNA polymerase II ,Recombinational DNA Repair ,Ataxia Telangiectasia Mutated Proteins ,Biology ,DNA repair protein XRCC4 ,Genome Integrity, Repair and Replication ,Molecular biology ,Cell Line ,Homology directed repair ,DNA-Binding Proteins ,Exonuclease 1 ,enzymes and coenzymes (carbohydrates) ,Exodeoxyribonucleases ,MRN complex ,Cell Line, Tumor ,Radiation, Ionizing ,Genetics ,biology.protein ,Humans ,DNA mismatch repair ,Phosphorylation ,DNA Damage ,Signal Transduction - Abstract
The MRE11/RAD50/NBS1 (MRN) complex plays a central role as a sensor of DNA double strand breaks (DSB) and is responsible for the efficient activation of ataxia-telangiectasia mutated (ATM) kinase. Once activated ATM in turn phosphorylates RAD50 and NBS1, important for cell cycle control, DNA repair and cell survival. We report here that MRE11 is also phosphorylated by ATM at S676 and S678 in response to agents that induce DNA DSB, is dependent on the presence of NBS1, and does not affect the association of members of the complex or ATM activation. A phosphosite mutant (MRE11S676AS678A) cell line showed decreased cell survival and increased chromosomal aberrations after radiation exposure indicating a defect in DNA repair. Use of GFP-based DNA repair reporter substrates in MRE11S676AS678A cells revealed a defect in homology directed repair (HDR) but single strand annealing was not affected. More detailed investigation revealed that MRE11S676AS678A cells resected DNA ends to a greater extent at sites undergoing HDR. Furthermore, while ATM-dependent phosphorylation of Kap1 and SMC1 was normal in MRE11S676AS678A cells, there was no phosphorylation of Exonuclease 1 consistent with the defect in HDR. These results describe a novel role for ATM-dependent phosphorylation of MRE11 in limiting the extent of resection mediated through Exonuclease 1.
- Published
- 2015
22. Nanoscale analysis of clustered DNA damage after high-LET irradiation by quantitative electron microscopy – The heavy burden to repair
- Author
-
Gisela Taucher-Scholz, Christina U. Brunner, Yvonne Lorat, Burkhard Jakob, Stefanie Schanz, and Claudia E. Rübe
- Subjects
Genetics ,DNA Repair ,Euchromatin ,DNA repair ,DNA damage ,Heterochromatin ,Cell Biology ,Biology ,Biochemistry ,Chromatin ,Ionizing radiation ,Kinetics ,Microscopy, Electron ,chemistry.chemical_compound ,chemistry ,Radiation, Ionizing ,Biophysics ,Humans ,DNA Breaks, Double-Stranded ,Linear Energy Transfer ,Irradiation ,Molecular Biology ,DNA - Abstract
Low- and high-linear energy transfer (LET) ionising radiation are effective cancer therapies, but produce structurally different forms of DNA damage. Isolated DNA damage is repaired efficiently; however, clustered lesions may be more difficult to repair, and are considered as significant biological endpoints. We investigated the formation and repair of DNA double-strand breaks (DSBs) and clustered lesions in human fibroblasts after exposure to sparsely (low-LET; delivered by photons) and densely (high-LET; delivered by carbon ions) ionising radiation. DNA repair factors (pKu70, 53BP1, γH2AX, and pXRCC1) were detected using immunogold-labelling and electron microscopy, and spatiotemporal DNA damage patterns were analysed within the nuclear ultrastructure at the nanoscale level. By labelling activated Ku-heterodimers (pKu70) the number of DSBs was determined in electron-lucent euchromatin and electron-dense heterochromatin. Directly after low-LET exposure (5 min post-irradiation), single pKu70 dimers, which reflect isolated DSBs, were randomly distributed throughout the entire nucleus with a linear dose correlation up to 30 Gy. Most euchromatic DSBs were sensed and repaired within 40 min, whereas heterochromatic DSBs were processed with slower kinetics. Essentially all DNA lesions induced by low-LET irradiation were efficiently rejoined within 24h post-irradiation. High-LET irradiation caused localised energy deposition within the particle tracks, and generated highly clustered DNA lesions with multiple DSBs in close proximity. The dimensions of these clustered lesions along the particle trajectories depended on the chromatin packing density, with huge DSB clusters predominantly localised in condensed heterochromatin. High-LET irradiation-induced clearly higher DSB yields than low-LET irradiation, with up to ∼ 500 DSBs per μm(3) track volume, and large fractions of these heterochromatic DSBs remained unrepaired. Hence, the spacing and quantity of DSBs in clustered lesions influence DNA repair efficiency, and may determine the radiobiological outcome.
- Published
- 2015
23. Live Cell Imaging to Study Real-Time ATM-Mediated Recruitment of DNA Repair Complexes to Sites of Ionizing Radiation-Induced DNA Damage
- Author
-
Burkhard, Jakob and Gisela, Taucher-Scholz
- Subjects
DNA Repair ,Radiation, Ionizing ,Humans ,DNA Breaks, Double-Stranded ,Ataxia Telangiectasia Mutated Proteins ,DNA Damage - Abstract
Measurements of protein recruitment and the formation of repair complexes at DNA double-strand breaks in real time provide valuable insight into the regulation of the early DNA damage response. Here, we describe the use of live cell microscopy in combination with ionizing radiation as a tool to evaluate the influence of ATM and its site-specific phosphorylation of target proteins on these processes. Recommendations are made for the preparation of the cells and the design of specialized cell chambers for the localized (and/or targeted) irradiation with charged particles at accelerator beamlines as well as the microscopic equipment and protocol to obtain high-resolution, sensitive fluorescence measurements.
- Published
- 2017
24. Live Cell Imaging to Study Real-Time ATM-Mediated Recruitment of DNA Repair Complexes to Sites of Ionizing Radiation-Induced DNA Damage
- Author
-
Gisela Taucher-Scholz and Burkhard Jakob
- Subjects
0301 basic medicine ,Chemistry ,DNA repair ,DNA damage ,Cell ,Molecular biology ,Ionizing radiation ,Green fluorescent protein ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Live cell imaging ,030220 oncology & carcinogenesis ,Biophysics ,medicine ,Irradiation ,DNA - Abstract
Measurements of protein recruitment and the formation of repair complexes at DNA double-strand breaks in real time provide valuable insight into the regulation of the early DNA damage response. Here, we describe the use of live cell microscopy in combination with ionizing radiation as a tool to evaluate the influence of ATM and its site-specific phosphorylation of target proteins on these processes. Recommendations are made for the preparation of the cells and the design of specialized cell chambers for the localized (and/or targeted) irradiation with charged particles at accelerator beamlines as well as the microscopic equipment and protocol to obtain high-resolution, sensitive fluorescence measurements.
- Published
- 2017
25. Die Finanzen unserer Regionalgruppe : ein Sorgenkind
- Author
-
Burkhard, Jakob and Riva, Enrico
- Published
- 2016
- Full Text
- View/download PDF
26. Autophosphorylation and ATM Activation
- Author
-
Martin F. Lavin, Phillip J. Robinson, Sairai So, Marcel Tanuji, Mark E. Graham, David J. Chen, Sergei Kozlov, Philip Chen, Gisela Taucher-Scholz, Keiji Suzuki, Burkhard Jakob, Frank Tobias, and Amanda W. Kijas
- Subjects
DNA repair ,Autophosphorylation ,Cell Biology ,Serine threonine protein kinase ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,enzymes and coenzymes (carbohydrates) ,MRN complex ,Ataxia-telangiectasia ,medicine ,Phosphorylation ,Protein phosphorylation ,Protein kinase A ,Molecular Biology - Abstract
The recognition and signaling of DNA double strand breaks involves the participation of multiple proteins, including the protein kinase ATM (mutated in ataxia-telangiectasia). ATM kinase is activated in the vicinity of the break and is recruited to the break site by the Mre11-Rad50-Nbs1 complex, where it is fully activated. In human cells, the activation process involves autophosphorylation on three sites (Ser(367), Ser(1893), and Ser(1981)) and acetylation on Lys(3016). We now describe the identification of a new ATM phosphorylation site, Thr(P)(1885) and an additional autophosphorylation site, Ser(P)(2996), that is highly DNA damage-inducible. We also confirm that human and murine ATM share five identical phosphorylation sites. We targeted the ATM phosphorylation sites, Ser(367) and Ser(2996), for further study by generating phosphospecific antibodies against these sites and demonstrated that phosphorylation of both was rapidly induced by radiation. These phosphorylations were abolished by a specific inhibitor of ATM and were dependent on ATM and the Mre11-Rad50-Nbs1 complex. As found for Ser(P)(1981), ATM phosphorylated at Ser(367) and Ser(2996) localized to sites of DNA damage induced by radiation, but ATM recruitment was not dependent on phosphorylation at these sites. Phosphorylation at Ser(367) and Ser(2996) was functionally important because mutant forms of ATM were defective in correcting the S phase checkpoint defect and restoring radioresistance in ataxia-telangiectasia cells. These data provide further support for the importance of autophosphorylation in the activation and function of ATM in vivo.
- Published
- 2011
27. Application of fluorescence lifetime imaging microscopy of DNA binding dyes to assess radiation-induced chromatin compaction changes
- Author
-
Burkhard Jakob, Elham Abdollahi, and Gisela Taucher-Scholz
- Subjects
0301 basic medicine ,Fluorescence-lifetime imaging microscopy ,Article ,Hoechst 34580 ,Catalysis ,3T3 cells ,lcsh:Chemistry ,Inorganic Chemistry ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,histone deacetylation inhibitor (HDACi) ,FLIM microcopy ,ddc:570 ,medicine ,Animals ,Irradiation ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,Fluorescent Dyes ,chromatin compaction ,irradiation ,X-Rays ,Organic Chemistry ,DNA ,General Medicine ,Chromatin Assembly and Disassembly ,Fluorescence ,Photon counting ,Computer Science Applications ,Chromatin ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Microscopy, Fluorescence ,chemistry ,pile-up ,NIH 3T3 Cells ,Biophysics ,Benzimidazoles ,Histone deacetylase ,Syto 13 - Abstract
International journal of molecular sciences 19(8), 2399 - (2018). doi:10.3390/ijms19082399, Published by Molecular Diversity Preservation International, Basel
- Published
- 2018
28. Live cell microscopy analysis of radiation-induced DNA double-strand break motion
- Author
-
J. Splinter, Gisela Taucher-Scholz, Burkhard Jakob, Marco Durante, B., Jakob, J., Splinter, Durante, Marco, and G., Taucher Scholz
- Subjects
Microscopy, Confocal ,Multidisciplinary ,DNA Repair ,Cell Survival ,DNA repair ,DNA damage ,Confocal ,DNA ,Biological Sciences ,Biology ,Molecular biology ,Chromatin ,chemistry.chemical_compound ,chemistry ,Live cell imaging ,Cell Line, Tumor ,Cancer cell ,DNA Repair Protein ,Biophysics ,Humans ,DNA Breaks, Double-Stranded - Abstract
We studied the spatiotemporal organization of DNA damage processing by live cell microscopy analysis in human cells. In unirradiated U2OS osteosarcoma and HeLa cancer cells, a fast confined and Brownian-like motion of DNA repair protein foci was observed, which was not altered by radiation. By analyzing the motional activity of GFP-53BP1 foci in live cells up to 12-h after irradiation, we detected an additional slower mobility of damaged chromatin sites showing a mean square displacement of ≈0.6 μm 2 /h after exposure to densely- or sparsely-ionizing radiation, most likely driven by normal diffusion of chromatin. Only occasionally, larger translational motion connected to morphological changes of the whole nucleus could be observed. In addition, there was no general tendency to form repair clusters in the irradiated cells. We conclude that long-range displacements of damaged chromatin domains do not generally occur during DNA double-strand break repair after introduction of multiple damaged sites by charged particles. The occasional and in part transient appearance of cluster formation of radiation-induced foci may represent a higher mobility of chromatin along the ion trajectory. These observations support the hypothesis that spatial proximity of DNA breaks is required for the formation of radiation-induced chromosomal exchanges.
- Published
- 2009
29. Autophosphorylation of DNA-PKCS regulates its dynamics at DNA double-strand breaks
- Author
-
Benjamin P C Chen, Gisela Taucher-Scholz, Pierre Olivier Mari, Eric Weterings, Naoya Uematsu, Dik C. van Gent, David J. Chen, Burkhard Jakob, Keiko Morotomi-Yano, Ken Ichi Yano, and Molecular Genetics
- Subjects
DNA Repair ,DNA repair ,Protein subunit ,CHO Cells ,DNA-Activated Protein Kinase ,Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Cricetulus ,0302 clinical medicine ,Catalytic Domain ,Cricetinae ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Kinase activity ,Protein kinase A ,Ku Autoantigen ,Research Articles ,DNA-PKcs ,030304 developmental biology ,0303 health sciences ,Photobleaching ,Lasers ,fungi ,Autophosphorylation ,Antigens, Nuclear ,DNA ,Cell Biology ,Molecular biology ,Cell biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,chemistry ,030220 oncology & carcinogenesis ,biological phenomena, cell phenomena, and immunity - Abstract
The DNA-dependent protein kinase catalytic subunit (DNA-PKCS) plays an important role during the repair of DNA double-strand breaks (DSBs). It is recruited to DNA ends in the early stages of the nonhomologous end-joining (NHEJ) process, which mediates DSB repair. To study DNA-PKCS recruitment in vivo, we used a laser system to introduce DSBs in a specified region of the cell nucleus. We show that DNA-PKCS accumulates at DSB sites in a Ku80-dependent manner, and that neither the kinase activity nor the phosphorylation status of DNA-PKCS influences its initial accumulation. However, impairment of both of these functions results in deficient DSB repair and the maintained presence of DNA-PKCS at unrepaired DSBs. The use of photobleaching techniques allowed us to determine that the kinase activity and phosphorylation status of DNA-PKCS influence the stability of its binding to DNA ends. We suggest a model in which DNA-PKCS phosphorylation/autophosphorylation facilitates NHEJ by destabilizing the interaction of DNA-PKCS with the DNA ends.
- Published
- 2007
30. X-ray irradiation activates K+ channels via H2O2 signaling
- Author
-
Indra Schroeder, Claudia Fournier, Burkhard Jakob, Gerhard Thiel, Patrick Becker, Bastian Roth, Christine S. Gibhardt, Anna Moroni, and Sebastian Fuck
- Subjects
REDOX REGULATION ,Potassium Channels ,Cell ,Biology ,Article ,Ionizing radiation ,Cell Line ,Cytosol ,medicine ,ION CHANNELS ,Humans ,Cell Nucleus ,Multidisciplinary ,ULTRAVIOLET-LIGHT ,Cell growth ,X-Rays ,HEK 293 cells ,BREAST-CANCER CELLS ,Hydrogen Peroxide ,Glutathione ,Potassium channel ,Cell biology ,medicine.anatomical_structure ,Biochemistry ,INTRACELLULAR HYDROGEN-PEROXIDE ,Calcium ,Signal transduction ,Nucleus ,Oxidation-Reduction ,Signal Transduction - Abstract
Ionizing radiation is a universal tool in tumor therapy but may also cause secondary cancers or cell invasiveness. These negative side effects could be causally related to the human-intermediate-conductance Ca2+-activated-K+-channel (hIK), which is activated by X-ray irradiation and affects cell proliferation and migration. To analyze the signaling cascade downstream of ionizing radiation we use genetically encoded reporters for H2O2 (HyPer) and for the dominant redox-buffer glutathione (Grx1-roGFP2) to monitor with high spatial and temporal resolution, radiation-triggered excursions of H2O2 in A549 and HEK293 cells. The data show that challenging cells with ≥1 Gy X-rays or with UV-A laser micro-irradiation causes a rapid rise of H2O2 in the nucleus and in the cytosol. This rise, which is determined by the rate of H2O2 production and glutathione-buffering, is sufficient for triggering a signaling cascade that involves an elevation of cytosolic Ca2+ and eventually an activation of hIK channels.
- Published
- 2015
31. Induction and processing of the radiation-induced gamma-H2AX signal and its link to the underlying pattern of DSB: A combined experimental and modelling study
- Author
-
Michael Scholz, F. Tommasino, Barbara Meyer, Burkhard Jakob, Thomas Friedrich, Marco Durante, Tommasino, Francesco, Friedrich, Thoma, Jakob, Burkhard, Meyer, Barbara, Durante, Marco, and Scholz, Michael
- Subjects
Models, Molecular ,Genetics and Molecular Biology (all) ,lcsh:Medicine ,Radiation induced ,Context (language use) ,CHO Cells ,Radiation ,Biology ,Signal ,Biochemistry ,Fluorescence ,Histones ,Cricetulus ,High doses ,Animals ,DNA Breaks, Double-Stranded ,Irradiation ,Agricultural and Biological Sciences (all) ,Biochemistry, Genetics and Molecular Biology (all) ,Medicine (all) ,Phosphorylation ,lcsh:Science ,Cell Nucleu ,Cells, Cultured ,Genetics ,Cell Nucleus ,Kinetic ,Multidisciplinary ,Animal ,X-Rays ,lcsh:R ,Dose-Response Relationship, Radiation ,Fibroblasts ,Kinetics ,Histone ,Gamma h2ax ,CHO Cell ,Yield (chemistry) ,X-Ray ,Fibroblast ,lcsh:Q ,Cricetulu ,Biological system ,Research Article ,Signal Transduction - Abstract
We present here an analysis of DSB induction and processing after irradiation with X-rays in an extended dose range based on the use of the γH2AX assay. The study was performed by quantitative flow cytometry measurements, since the use of foci counting would result in reasonable accuracy only in a limited dose range of a few Gy. The experimental data are complemented by a theoretical analysis based on the GLOBLE model. In fact, original aim of the study was to test GLOBLE predictions against new experimental data, in order to contribute to the validation of the model. Specifically, the γH2AX signal kinetics has been investigated up to 24 h after exposure to increasing photon doses between 2 and 500 Gy. The prolonged persistence of the signal at high doses strongly suggests dose dependence in DSB processing after low LET irradiation. Importantly, in the framework of our modelling analysis, this is related to a gradually increased fraction of DSB clustering at the micrometre scale. The parallel study of γH2AX dose response curves shows the onset of a pronounced saturation in two cell lines at a dose of about 20 Gy. This dose is much lower than expected according to model predictions based on the values usually adopted for the DSB induction yield (≈ 30 DSB/Gy) and for the γH2AX foci extension of approximately 2 Mbp around the DSB. We show and discuss how theoretical predictions and experimental findings can be in principle reconciled by combining an increased DSB induction yield with the assumption of a larger genomic extension for the single phosphorylated regions. As an alternative approach, we also considered in our model the possibility of a 3D spreading-mechanism of the H2AX phosphorylation around the induced DSB, and applied it to the analysis of both the aspects considered. Our results are found to be supportive for the basic assumptions on which GLOBLE is built. Apart from giving new insights into the H2AX phosphorylation process, experiments performed at high doses are of relevance in the context of radiation therapy, where hypo-fractionated schemes become increasingly popular.
- Published
- 2015
32. Nucleolar localization of aprataxin is dependent on interaction with nucleolin and on active ribosomal DNA transcription
- Author
-
Gisela Taucher-Scholz, Amila Suraweera, Nuri Gueven, Valeérie Schreiber, Martin F. Lavin, Olivier J. Becherel, Burkhard Jakob, and Geoff W. Birrell
- Subjects
Cerebellar Ataxia ,Transcription, Genetic ,Apraxias ,Nucleolus ,Recombinant Fusion Proteins ,Molecular Sequence Data ,aptX ,In Vitro Techniques ,Biology ,DNA, Ribosomal ,Cell Line ,Transcription (biology) ,Genetics ,RNA polymerase I ,Humans ,Amino Acid Sequence ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,Genetics (clinical) ,Aprataxin ,Nucleophosmin ,Binding Sites ,Base Sequence ,Nuclear Proteins ,RNA-Binding Proteins ,General Medicine ,Ribosomal RNA ,Phosphoproteins ,Molecular biology ,DNA-Binding Proteins ,RNA, Ribosomal ,RNA Interference ,Pol1 Transcription Initiation Complex Proteins ,Nucleolin ,Cell Nucleolus ,HeLa Cells ,Protein Binding - Abstract
The APTX gene, mutated in patients with the neurological disorder ataxia with oculomotor apraxia type 1 (AOA1), encodes a novel protein aprataxin. We describe here, the interaction and interdependence between aprataxin and several nucleolar proteins, including nucleolin, nucleophosmin and upstream binding factor-1 (UBF-1), involved in ribosomal RNA (rRNA) synthesis and cellular stress signalling. Interaction between aprataxin and nucleolin occurred through their respective N-terminal regions. In AOA1 cells lacking aprataxin, the stability of nucleolin was significantly reduced. On the other hand, down-regulation of nucleolin by RNA interference did not affect aprataxin protein levels but abolished its nucleolar localization suggesting that the interaction with nucleolin is involved in its nucleolar targeting. GFP-aprataxin fusion protein co-localized with nucleolin, nucleophosmin and UBF-1 in nucleoli and inhibition of ribosomal DNA transcription altered the distribution of aprataxin in the nucleolus, suggesting that the nature of the nucleolar localization of aprataxin is also dependent on ongoing rRNA synthesis. In vivo rRNA synthesis analysis showed only a minor decrease in AOA1 cells when compared with controls cells. These results demonstrate a cross-dependence between aprataxin and nucleolin in the nucleolus and while aprataxin does not appear to be directly involved in rRNA synthesis its nucleolar localization is dependent on this synthesis.
- Published
- 2006
33. Live Cell Imaging of Heavy-Ion-Induced Radiation Responses by Beamline Microscopy
- Author
-
G. Taucher-Scholz, Burkhard Jakob, Nuri Gueven, Martin F. Lavin, and J. H. Rudolph
- Subjects
DNA damage ,DNA repair ,Cell Culture Techniques ,Biophysics ,Biology ,Radiation Dosage ,chemistry.chemical_compound ,Live cell imaging ,Image Interpretation, Computer-Assisted ,Microscopy ,Humans ,Heavy Ions ,Linear Energy Transfer ,Radiology, Nuclear Medicine and imaging ,Irradiation ,Microscopy, Video ,Radiation ,Nuclear Proteins ,Dose-Response Relationship, Radiation ,DNA ,Equipment Design ,Robotics ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Equipment Failure Analysis ,Protein Transport ,chemistry ,DNA Damage ,HeLa Cells ,Nucleotide excision repair - Abstract
To study the dynamics of protein recruitment to DNA lesions, ion beams can be used to generate extremely localized DNA damage within restricted regions of the nuclei. This inhomogeneous spatial distribution of lesions can be visualized indirectly and rapidly in the form of radiation-induced foci using immunocytochemical detection or GFP-tagged DNA repair proteins. To analyze faster protein translocations and a possible contribution of radiation-induced chromatin movement in DNA damage recognition in live cells, we developed a remote-controlled system to obtain high-resolution fluorescence images of living cells during ion irradiation with a frame rate of the order of seconds. Using scratch replication labeling, only minor chromatin movement at sites of ion traversal was observed within the first few minutes of impact. Furthermore, time-lapse images of the GFP-coupled DNA repair protein aprataxin revealed accumulations within seconds at sites of ion hits, indicating a very fast recruitment to damaged sites. Repositioning of the irradiated cells after fixation allowed the comparison of live cell observation with immunocytochemical staining and retrospective etching of ion tracks. These results demonstrate that heavy-ion radiation-induced changes in subnuclear structures can be used to determine the kinetics of early protein recruitment in living cells and that the changes are not dependent on large-scale chromatin movement at short times postirradiation.
- Published
- 2005
34. Microscopic visualization of a biological response to charged particle traversal
- Author
-
G. Becker, M. Scholz, Gisela Taucher-Scholz, and Burkhard Jakob
- Subjects
Nuclear and High Energy Physics ,Range (particle radiation) ,Radiobiology ,DNA damage ,Nanotechnology ,Charged particle ,Ionizing radiation ,chemistry.chemical_compound ,chemistry ,Biophysics ,Radiation damage ,Irradiation ,Instrumentation ,DNA - Abstract
Understanding the molecular mechanisms underlying biological effects of charged particle radiation has become increasingly important in view of the use of ion beams in tumor therapy. Elucidating how the enhanced efficiency of densely ionizing radiation in cell killing is related to the initial causative lesions, namely DNA double-strand breaks, constitutes a major task in radiobiology. The inhomogeneous spatial distribution of energy deposition leading to the induction of more complex and less reparable DNA lesions is the basis for high-LET effects. But the cellular response to radiation damage also involves the interplay between repair and signal transduction proteins with the aim of coordinating the processing of DNA damage and cell cycle progression to allow time for repair. Charged particles are used as a probe for the production of localized subcellular damage to study these aspects of the biological response to ionizing radiation. Immunocytochemical techniques applied in combination with confocal laser microscopy allow to monitor the relocalization of DNA damage response proteins within individual nuclei following irradiation. In particular, the rapid accumulation of the signalling protein p21 at sites of heavy ion-induced DNA damage reflects the microscopic distribution of dose deposited within nuclei of irradiated human fibroblasts. The biological response pattern for p21 is presented for high and low energy ion beams, involving different particle species and representing a wide range of radiation qualities.
- Published
- 2003
35. DNA end resection is needed for the repair of complex lesions in G1-phase human cells
- Author
-
Oliver Ringel, Gisela Taucher-Scholz, Marco Durante, Maren Herrlitz, Burkhard Jakob, and Nicole B. Averbeck
- Subjects
G2 Phase ,DNA End-Joining Repair ,DNA repair ,Biology ,Cell Line ,S Phase ,Histones ,Cell Cycle News & Views ,Exonuclease 1 ,MRE11 Homologue Protein ,Report ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Molecular Biology ,Replication protein A ,X-Rays ,G1 Phase ,Cell Biology ,Cell cycle ,Molecular biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,DNA Repair Enzymes ,Exodeoxyribonucleases ,Cell killing ,Cancer research ,Homologous recombination ,Developmental Biology - Abstract
Repair of DNA double strand breaks (DSBs) is influenced by the chemical complexity of the lesion. Clustered lesions (complex DSBs) are generally considered more difficult to repair and responsible for early and late cellular effects after exposure to genotoxic agents. Resection is commonly used by the cells as part of the homologous recombination (HR) pathway in S- and G2-phase. In contrast, DNA resection in G1-phase may lead to an error-prone microhomology-mediated end joining. We induced DNA lesions with a wide range of complexity by irradiation of mammalian cells with X-rays or accelerated ions of different velocity and mass. We found replication protein A (RPA) foci indicating DSB resection both in S/G2- and G1-cells, and the fraction of resection-positive cells correlates with the severity of lesion complexity throughout the cell cycle. Besides RPA, Ataxia telangiectasia and Rad3-related (ATR) was recruited to complex DSBs both in S/G2- and G1-cells. Resection of complex DSBs is driven by meiotic recombination 11 homolog A (MRE11), CTBP-interacting protein (CtIP), and exonuclease 1 (EXO1) but seems not controlled by the Ku heterodimer or by phosphorylation of H2AX. Reduced resection capacity by CtIP depletion increased cell killing and the fraction of unrepaired DSBs after exposure to densely ionizing heavy ions, but not to X-rays. We conclude that in mammalian cells resection is essential for repair of complex DSBs in all phases of the cell-cycle and targeting this process sensitizes mammalian cells to cytotoxic agents inducing clustered breaks, such as in heavy-ion cancer therapy.
- Published
- 2014
36. ATM alters the otherwise robust chromatin mobility at sites of DNA Double-Strand Breaks (DSBs) in human cells
- Author
-
Burkhard Jakob, Marco Durante, Gisela Taucher-Scholz, Annabelle Becker, Becker, A., Durante, M., Taucher-Scholz, G., and Jakob, B.
- Subjects
DNA Repair ,Chromosomal Proteins, Non-Histone ,DNA damage ,DNA repair ,Radiation Biophysics ,Poly ADP ribose polymerase ,Biophysics ,lcsh:Medicine ,Gene Expression ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Biochemistry ,Cell Line ,chemistry.chemical_compound ,Genes, Reporter ,Nucleic Acids ,Molecular Cell Biology ,Genetics ,Medicine and Health Sciences ,Humans ,DNA Breaks, Double-Stranded ,lcsh:Science ,Transcription factor ,Multidisciplinary ,Biology and life sciences ,Cohesin ,Chromosome Biology ,Chemistry ,Radiology and Imaging ,lcsh:R ,Radiobiology ,DNA ,Cell Biology ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,Cell biology ,Radiation Effects ,Gene Knockdown Techniques ,Multiprotein Complexes ,lcsh:Q ,Poly(ADP-ribose) Polymerases ,Research Article ,Transcription Factors - Abstract
Ionizing radiation induces DNA double strand breaks (DSBs) which can lead to the formation of chromosome rearrangements through error prone repair. In mammalian cells the positional stability of chromatin contributes to the maintenance of genome integrity. DSBs exhibit only a small, submicron scale diffusive mobility, but a slight increase in the mobility of chromatin domains by the induction of DSBs might influence repair fidelity and the formation of translocations. The radiation-induced local DNA decondensation in the vicinity of DSBs is one factor potentially enhancing the mobility of DSB-containing chromatin domains. Therefore in this study we focus on the influence of different chromatin modifying proteins, known to be activated by the DNA damage response, on the mobility of DSBs. IRIF (ionizing radiation induced foci) in U2OS cells stably expressing 53BP1-GFP were used as a surrogate marker of DSBs. Low angle charged particle irradiation, known to trigger a pronounced DNA decondensation, was used for the defined induction of linear tracks of IRIF. Our results show that movement of IRIF is independent of the investigated chromatin modifying proteins like ACF1 or PARP1 and PARG. Also depletion of proteins that tether DNA strands like MRE11 and cohesin did not alter IRIF dynamics significantly. Inhibition of ATM, a key component of DNA damage response signaling, resulted in a pronounced confinement of DSB mobility, which might be attributed to a diminished radiation induced decondensation. This confinement following ATM inhibition was confirmed using X-rays, proving that this effect is not restricted to densely ionizing radiation. In conclusion, repair sites of DSBs exhibit a limited mobility on a small spatial scale that is mainly unaffected by depletion of single remodeling or DNA tethering proteins. However, it relies on functional ATM kinase which is considered to influence the chromatin structure after irradiation. © 2014 Becker et al.
- Published
- 2014
37. DNA end resection is needed for the repair of complex lesions in G1-phase human cells
- Author
-
Nicole B Averbeck, Oliver Ringel, Maren Herrlitz, Burkhard Jakob, Marco Durante, Gisela Taucher-Scholz, Nicole B Averbeck, Oliver Ringel, Maren Herrlitz, Burkhard Jakob, Marco Durante, and Gisela Taucher-Scholz
- Published
- 2015
- Full Text
- View/download PDF
38. Photooxidative Damage in Young Leaves of Declining Grapevine: Does It Result from a New and Possibly Viroid-Related Disease?
- Author
-
Yuri Gamalei, Hans J. Gross, Ulrich Heber, Rainer Wolf, and Burkhard Jakob
- Subjects
Physiology ,Botany ,Cell Biology ,Plant Science ,General Medicine ,Ancient history ,Biology ,Vitis vinifera - Abstract
Burkhard Jakob', Yuri Gamalei, Rainer Wolf, Ulrich Heber' and Hans J. Gross 1 Julius-von-Sachs-Institute, University of Wiirzburg, Wiirzburg D-97082, Germany 2 Komarov-Institute of Botany, Russian Academy of Sciences, St. Petersburg, Russia 3 Theodor-Boveri-Institute, University of Wiirzburg, Wiirzburg D-97074, Germany 4 Institute of Biochemistry, University of Wiirzburg, Wiirzburg D-97074, Germany
- Published
- 1997
39. Clustered DNA damage induces pan-nuclear H2AX phosphorylation mediated by ATM and DNA-PK
- Author
-
Burkhard Jakob, Barbara Meyer, Kay-Obbe Voss, Frank Tobias, Gisela Taucher-Scholz, Marco Durante, Meyer, Barbara, Voss, Kay Obbe, Tobias, Frank, Jakob, Burkhard, Durante, Marco, and Taucher Scholz, Gisela
- Subjects
Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,DNA-Activated Protein Kinase ,Genome Integrity, Repair and Replication ,Protein-Serine-Threonine Kinase ,Histones ,Ataxia Telangiectasia Mutated Protein ,chemistry.chemical_compound ,Mice ,Cricetinae ,Radiation, Ionizing ,Cell Cycle Protein ,DNA Breaks, Double-Stranded ,Phosphorylation ,Cell Nucleu ,Cells, Cultured ,Nuclear Protein ,Kinase ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Histone ,Trans-Activator ,biological phenomena, cell phenomena, and immunity ,Tumor Suppressor p53-Binding Protein 1 ,Human ,DNA damage ,DNA repair ,DNA-Binding Protein ,Protein Serine-Threonine Kinases ,Biology ,Cell Line ,Genetics ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Cell Nucleus ,Tumor Suppressor Protein ,Animal ,Tumor Suppressor Proteins ,Fluorescence recovery after photobleaching ,Molecular biology ,MDC1 ,enzymes and coenzymes (carbohydrates) ,chemistry ,Intracellular Signaling Peptides and Protein ,Trans-Activators ,DNA - Abstract
DNA double-strand breaks (DSB) are considered as the most deleterious DNA lesions, and their repair is further complicated by increasing damage complexity. However, the molecular effects of clustered lesions are yet not fully understood. As the locally restricted phosphorylation of H2AX to form γH2AX is a key step in facilitating efficient DSB repair, we investigated this process after localized induction of clustered damage by ionizing radiation. We show that in addition to foci at damaged sites, H2AX is also phosphorylated in undamaged chromatin over the whole-cell nucleus in human and rodent cells, but this is not related to apoptosis. This pan-nuclear γH2AX is mediated by the kinases ataxia telangiectasia mutated and DNA-dependent protein kinase (DNA–PK) that also phosphorylate H2AX at DSBs. The pan-nuclear response is dependent on the amount of DNA damage and is transient even under conditions of impaired DSB repair. Using fluorescence recovery after photobleaching (FRAP), we found that MDC1, but not 53BP1, binds to the nuclear-wide γH2AX. Consequently, the accumulation of MDC1 at DSBs is reduced. Altogether, we show that a transient dose-dependent activation of the kinases occurring on complex DNA lesions leads to their nuclear-wide distribution and H2AX phosphorylation, yet without eliciting a full pan-nuclear DNA damage response.
- Published
- 2013
40. Species conserved DNA damage response at the inactive human X chromosome
- Author
-
Gisela Taucher-Scholz, Iris Müller, Nicole B. Averbeck, Kay-Obbe Voss, Marco Durante, B. Merk, Burkhard Jakob, Muller, I., Merk, B., Voss, K. -O., Averbeck, N., Jakob, B., Durante, M., and Taucher-Scholz, G.
- Subjects
Chromocenter ,DNA Repair ,Heterochromatin ,DNA damage ,Health, Toxicology and Mutagenesis ,Cell ,Fluorescent Antibody Technique ,Biology ,DNA damage response ,Genome ,Particle irradiation ,Histones ,chemistry.chemical_compound ,Mice ,Inactive X chromosome ,Genetics ,medicine ,Constitutive heterochromatin ,Animals ,Humans ,X chromosome ,Chromosomes, Human, X ,fungi ,Fibroblasts ,Molecular biology ,Chromatin ,Cell biology ,medicine.anatomical_structure ,chemistry ,NIH 3T3 Cells ,Female ,DNA ,DNA Damage - Abstract
Chromatin modifications are long known as an essential part of the orchestrated response resulting in the repair of radiation-induced DNA double-strand breaks (DSBs). Only recently, however, the influence of the chromatin architecture itself on the DNA damage response has been recognised. Thus for heterochromatic DSBs the sensing and early recruitment of repair factors to the lesion occurs within the heterochromatic compartments, but the damage sites are subsequently relocated from the inside to the outside of the heterochromatin. While previous studies were accomplished at the constitutive heterochromatin of centromeric regions in mouse and flies, here we examine the DSB repair at the facultative heterochromatin of the inactive X chromosome (Xi) in humans. Using heavy ion irradiation we show that at later times after irradiation the DSB damage streaks bend around the Xi verifying that the relocation process is conserved between species and not specialised to repetitive sequences only. In addition, to measure chromatin relaxation at rare positions within the genome, we established live cell microscopy at the GSI microbeam thus allowing the aimed irradiation of small nuclear structures like the Xi. Chromatin decondensation at DSBs within the Xi is clearly visible within minutes as a continuous decrease of the DNA staining over time, comparable to the DNA relaxation revealed at DSBs in mouse chromocenters. Furthermore, despite being conserved between species, slight differences in the underlying regulation of these processes in heterochromatic DSBs are apparent. © 2013 Elsevier B.V.
- Published
- 2013
41. Photobleaching setup for the biological end-station of the darmstadt heavy-ion microprobe
- Author
-
Bernd E. Fischer, K.-O. Voss, Christina Trautmann, B. Merk, I. Müller, Burkhard Jakob, Marco Durante, Gisela Taucher-Scholz, Merk, B., Voss, K. -O., Muller, I., Fischer, B. E., Jakob, B., Taucher-Scholz, G., Trautmann, C., and Durante, M.
- Subjects
Nuclear and High Energy Physics ,Microprobe ,Microscope ,business.industry ,Chemistry ,Fluorescence recovery after photobleaching ,Laser ,Photobleaching ,law.invention ,Numerical aperture ,Optics ,law ,Fluorescence microscope ,FRAP ,Focal Spot Size ,business ,Live cell imaging ,Instrumentation ,Ku80 - Abstract
We report the upgrade of the epifluorescence microscope of the GSI heavy-ion microprobe with a galvo-scanned, 488 nm laser diode. The laser is focussed into the object plane by the water-immersion objective resulting in a focal spot size of about 1 μm. To increase temporal and spatial resolution a water-immersion objective with a high numerical aperture is integrated into the custom-build microscope. The upgraded system can now be used to bleach GFP-tagged proteins recruited to DNA damage induced by targeted single-ion irradiation. The system is demonstrated on NIH 3T3 cells with Ku80-GFP ion-targeted in heterochromatic and euchromatic DNA. Fluorescence recovery after photobleaching (FRAP) is shown to be significantly slower in heterochromatin. © 2013 Elsevier B.V. All rights reserved.
- Published
- 2013
42. Photoproduction and Detoxification of Hydroxyl radicals in Chloroplasts and Leaves and Relation to Photoinactivation of Photosystems I and II
- Author
-
Ulrich Heber and Burkhard Jakob
- Subjects
Spinacia ,biology ,Physiology ,Radical ,Mehler reaction ,food and beverages ,DCMU ,Cell Biology ,Plant Science ,General Medicine ,Photochemistry ,biology.organism_classification ,Chloroplast membrane ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Thylakoid ,Ferredoxin ,Photosystem - Abstract
The light-dependent production of hydroxyl radicals (HO*) by thylakoids, chloroplasts and leaves of Spinacia oleracea was investigated using dimethylsulfoxide as HO' trapping agent. Maximum rates of HO' production by thylakoids as indicated by the formation of methane sulfinic acid were observed under aerobic conditions in the absence of added electron acceptors. They were higher than 2 /anol (mgChlh)"1. Saturation of HO* production occurred at the low photon flux density of 100 /rniol m~2 s"1. Trapping of HO* by dimethylsulfoxide suppressed, but did not eliminate light-dependent inactivation of PSI and II suggesting that HO' formation contributed to the photosensitivity of isolated thylakoids. DCMU inhibited HO' formation. Importantly, methylviologen decreased HO* formation in the absence, but stimulated it in the presence of Fe 3+. In intact chloroplasts, HO' formation became appreciable only after KCN had been added to inhibit effective H2O2 scavenging by ascorbate peroxidase. It was stimulated by ferrisulfate, but not by ferricyanide which does not penetrate the chloroplast envelope. Infiltrated spinach leaves behaved similar in principle to intact chloroplasts in regard to HO* formation but HO' production was very slow if detectable at all by the formation of methylsulfini c acid indicating effective radical detoxification. HO' formation is interpreted to be the result of a Fenton-type reaction which produces HO' in chloroplasts from H2O2 and reduced ferredoxin, when O2 is electron acceptor in the Mehler reaction and radical detoxification reactions are inhibited.
- Published
- 1996
43. Reduced contribution of thermally labile sugar lesions to DNA double strand break formation after exposure to heavy ions
- Author
-
Satyendra K, Singh, Alena, Bencsik-Theilen, Emil, Mladenov, Burkhard, Jakob, Gisela, Taucher-Scholz, and George, Iliakis
- Subjects
Research ,High LET ,Carbohydrates ,DNA double strand breaks (DSB) ,Heavy Ion Radiotherapy ,DNA ,Ionizing radiation (IR) ,Heavy ions ,Electrophoresis, Gel, Pulsed-Field ,Mice ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Linear Energy Transfer ,Radiation chemistry ,Labile lesions ,Relative Biological Effectiveness - Abstract
In cells exposed to low linear energy transfer (LET) ionizing-radiation (IR), double-strand-breaks (DSBs) form within clustered-damage-sites (CDSs) from lesions disrupting the DNA sugar-phosphate backbone. It is commonly assumed that all DSBs form promptly and are immediately detected by the cellular DNA-damage-response (DDR) apparatus. However, there is evidence that the pool of DSBs detected by physical methods, such as pulsed-field gel electrophoresis (PFGE), comprises not only promptly forming DSBs (prDSBs) but also DSBs developing during lysis at high temperatures from thermally-labile sugar-lesions (TLSLs). We recently demonstrated that conversion of TLSLs to DNA breaks and ultimately to DSBs also occurs in cells during the first hour of post-irradiation incubation at physiological temperatures. Thus, TLSL-dependent DSBs (tlDSBs) are not an avoidable technique-related artifact, but a reality the cell always faces. The biological consequences of tlDSBs and the dependence of their formation on LET require in-depth investigation. Heavy-ions (HI) are a promising high-LET radiation modality used in cancer treatment. HI are also encountered in space and generate serious radiation protection problems to prolonged space missions. Here, we study, therefore, the effect of HI on the yields of tlDSBs and prDSBs. We report a reduction in the yield of tlDBSs stronger than that earlier reported for neutrons, and with pronounced cell line dependence. We conclude that with increasing LET the complexity of CDSs increases resulting in a commensurate increase in the yield prDSBs and a decrease in tlDSBs. The consequences of these effects to the relative biological effectiveness are discussed.
- Published
- 2012
44. PCNA-dependent accumulation of CDKN1A into nuclear foci after ionizing irradiation
- Author
-
Daniela Fink, Gisela Taucher-Scholz, Frank Tobias, J. H. Rudolph, Christine Blattner, Burkhard Jakob, and Claudia Wiese
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,DNA Repair ,DNA damage ,DNA repair ,Biology ,Biochemistry ,Proliferating Cell Nuclear Antigen ,Radiation, Ionizing ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Molecular Biology ,Cell Nucleus ,X-Rays ,DNA replication ,Cell Biology ,Base excision repair ,Hydrogen Peroxide ,Fibroblasts ,Oxidants ,Molecular biology ,Chromatin ,Proliferating cell nuclear antigen ,Cell nucleus ,Protein Transport ,medicine.anatomical_structure ,Gamma Rays ,biology.protein ,Nucleotide excision repair ,Protein Binding - Abstract
The cyclin-dependent kinase inhibitor CDKN1A/p21 confers cell-cycle arrest in response to DNA damage and inhibits DNA replication through its direct interaction with the proliferating cell nuclear antigen (PCNA) and cyclin/cyclin-dependent kinase complexes. Previously, we reported that in response to densely ionizing radiation CDKN1A rapidly is recruited to the sites of particle traversal, and that CDKN1A foci formation in response to heavy ions is independent of its transactivation by TP53. Here, we show that exposure of normal human fibroblasts to X-rays or to H2O2 also induces nuclear accumulations of CDKN1A. We find that CDKN1A foci formation in response to radiation damage is dependent on its dephosphorylation and on its direct physical interaction with PCNA. Live cell imaging analyses of ectopically expressed EGFP-CDKN1A and dsRed-PCNA show rapid recruitment of both proteins into foci after radiation damage. Detailed dynamic measurements reveal a slightly delayed recruitment of CDKN1A compared to PCNA, which is best described by bi-exponential curve fitting, taking the preceding binding of PCNA to DNA into account. We propose a regulatory role for CDKN1A in mediating PCNA function after radiation damage, and provide evidence that this role is distinct from its involvement in nucleotide excision repair and unrelated to double-strand break repair.
- Published
- 2011
45. ATM protein-dependent phosphorylation of Rad50 protein regulates DNA repair and cell cycle control
- Author
-
Ji-Hoon Lee, Magtouf Gatei, Geoff W. Birrell, Detlev Schindler, Burkhard Jakob, Thilo Dörk, Shazrul Fazry, Tanya T. Paull, Amanda W. Kijas, Reinhard Kalb, Martin F. Lavin, Olivier J. Becherel, Gisela Taucher-Scholz, Regina Waltes, Yaniv Lerenthal, Philip Chen, and Nuri Gueven
- Subjects
Genome instability ,DNA Repair ,DNA repair ,Chromosomal Proteins, Non-Histone ,Eukaryotic DNA replication ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,Radiation Tolerance ,Genomic Instability ,S Phase ,Ataxia Telangiectasia ,Humans ,CHEK1 ,Phosphorylation ,Molecular Biology ,Replication protein A ,Adaptor Proteins, Signal Transducing ,Tumor Suppressor Proteins ,Cell Cycle ,Cell Biology ,G2-M DNA damage checkpoint ,DNA repair protein XRCC4 ,Cell biology ,Acid Anhydride Hydrolases ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,DNA Repair Enzymes ,MRN complex ,Mutant Proteins ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
The Mre11/Rad50/NBN complex plays a central role in coordinating the cellular response to DNA double-strand breaks. The importance of Rad50 in that response is evident from the recent description of a patient with Rad50 deficiency characterized by chromosomal instability and defective ATM-dependent signaling. We report here that ATM (defective in ataxia-telangiectasia) phosphorylates Rad50 at a single site (Ser-635) that plays an important adaptor role in signaling for cell cycle control and DNA repair. Although a Rad50 phosphosite-specific mutant (S635G) supported normal activation of ATM in Rad50-deficient cells, it was defective in correcting DNA damage-induced signaling through the ATM-dependent substrate SMC1. This mutant also failed to correct radiosensitivity, DNA double-strand break repair, and an S-phase checkpoint defect in Rad50-deficient cells. This was not due to disruption of the Mre11/Rad50/NBN complex revealing for the first time that phosphorylation of Rad50 plays a key regulatory role as an adaptor for specific ATM-dependent downstream signaling through SMC1 for DNA repair and cell cycle checkpoint control in the maintenance of genome integrity.
- Published
- 2011
46. Autophosphorylation and ATM activation: additional sites add to the complexity
- Author
-
Sergei V, Kozlov, Mark E, Graham, Burkhard, Jakob, Frank, Tobias, Amanda W, Kijas, Marcel, Tanuji, Philip, Chen, Phillip J, Robinson, Gisela, Taucher-Scholz, Keiji, Suzuki, Sairai, So, David, Chen, and Martin F, Lavin
- Subjects
MRE11 Homologue Protein ,Tumor Suppressor Proteins ,Nuclear Proteins ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Acid Anhydride Hydrolases ,S Phase ,DNA-Binding Proteins ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,Ataxia Telangiectasia ,Mice ,DNA Repair Enzymes ,Multiprotein Complexes ,Radiation, Ionizing ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Protein Kinase Inhibitors ,Cell Line, Transformed ,Signal Transduction - Abstract
The recognition and signaling of DNA double strand breaks involves the participation of multiple proteins, including the protein kinase ATM (mutated in ataxia-telangiectasia). ATM kinase is activated in the vicinity of the break and is recruited to the break site by the Mre11-Rad50-Nbs1 complex, where it is fully activated. In human cells, the activation process involves autophosphorylation on three sites (Ser(367), Ser(1893), and Ser(1981)) and acetylation on Lys(3016). We now describe the identification of a new ATM phosphorylation site, Thr(P)(1885) and an additional autophosphorylation site, Ser(P)(2996), that is highly DNA damage-inducible. We also confirm that human and murine ATM share five identical phosphorylation sites. We targeted the ATM phosphorylation sites, Ser(367) and Ser(2996), for further study by generating phosphospecific antibodies against these sites and demonstrated that phosphorylation of both was rapidly induced by radiation. These phosphorylations were abolished by a specific inhibitor of ATM and were dependent on ATM and the Mre11-Rad50-Nbs1 complex. As found for Ser(P)(1981), ATM phosphorylated at Ser(367) and Ser(2996) localized to sites of DNA damage induced by radiation, but ATM recruitment was not dependent on phosphorylation at these sites. Phosphorylation at Ser(367) and Ser(2996) was functionally important because mutant forms of ATM were defective in correcting the S phase checkpoint defect and restoring radioresistance in ataxia-telangiectasia cells. These data provide further support for the importance of autophosphorylation in the activation and function of ATM in vivo.
- Published
- 2010
47. Biological dose estimation of UVA laser microirradiation utilizing charged particle-induced protein foci
- Author
-
G. Taucher-Scholz, J. Splinter, Ken Ichi Yano, Burkhard Jakob, Marco Durante, Marion C. Lang, Stefan W. Hell, David J. Chen, Johann Engelhardt, J., Splinter, B., Jakob, M., Lang, K., Yano, J., Engelhardt, S. W., Hell, D. J., Chen, Durante, Marco, and G., Taucher Scholz
- Subjects
DNA damage ,Ultraviolet Rays ,Health, Toxicology and Mutagenesis ,Biology ,Toxicology ,Ionizing radiation ,Cell Line ,Replication Protein A ,Genetics ,Dosimetry ,Humans ,Telomeric Repeat Binding Protein 2 ,Irradiation ,Telomeric Repeat Binding Protein 1 ,Replication protein A ,Genetics (clinical) ,Cell Nucleus ,Equivalent dose ,Lasers ,Dose-Response Relationship, Radiation ,Alpha particle ,Original Articles ,Alpha Particles ,Molecular biology ,Charged particle ,Biophysics ,DNA Damage ,Protein Binding - Abstract
The induction of localized DNA damage within a discrete nuclear volume is an important tool in DNA repair studies. Both charged particle irradiation and laser microirradiation (LMI) systems allow for such a localized damage induction, but the results obtained are difficult to compare, as the delivered laser dose cannot be measured directly. Therefore, we revisited the idea of a biological dosimetry based on the microscopic evaluation of irradiation-induced Replication Protein A (RPA) foci numbers. Considering that local dose deposition is characteristic for both LMI and charged particles, we took advantage of the defined dosimetry of particle irradiation to estimate the locally applied laser dose equivalent. Within the irradiated nuclear sub-volumes, the doses were in the range of several hundreds of Gray. However, local dose estimation is limited by the saturation of the RPA foci numbers with increasing particle doses. Even high-resolution 4Pi microscopy did not abrogate saturation as it was not able to resolve single lesions within individual RPA foci. Nevertheless, 4Pi microscopy revealed multiple and distinct 53BP1- and gamma H2AX-stained substructures within the lesion flanking chromatin domains. Monitoring the local recruitment of the telomere repeat-binding factors TRF1 and TRF2 showed that both proteins accumulated at damage sites after UVA-LMI but not after densely ionizing charged particle irradiation. Hence, our results indicate that the local dose delivered by UVA-LMI is extremely high and cannot be accurately translated into an equivalent ionizing radiation dose, despite the sophisticated techniques used in this study.
- Published
- 2010
48. CK2 phosphorylation-dependent interaction between aprataxin and MDC1 in the DNA damage response
- Author
-
Cheng Peng, Peter J. McKinnon, Junjie Chen, G. Taucher-Scholz, Martin F. Lavin, Markus Fusser, Sachin Katyal, Olivier J. Becherel, Bernd Epe, Burkhard Jakob, Nuri Gueven, Amanda W. Kijas, Stephen J. Smerdon, and A.L. Cherry
- Subjects
Models, Molecular ,DNA Repair ,DNA repair ,DNA damage ,Molecular Sequence Data ,Cell Cycle Proteins ,Biology ,Genome Integrity, Repair and Replication ,medicine.disease_cause ,Cell Line ,03 medical and health sciences ,QH301 ,Mice ,Genetics ,medicine ,Animals ,Humans ,Linear Energy Transfer ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Binding site ,Phosphorylation ,Casein Kinase II ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Aprataxin ,chemistry.chemical_classification ,0303 health sciences ,Mutation ,DNA ligase ,Binding Sites ,030302 biochemistry & molecular biology ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Molecular biology ,Cell biology ,MDC1 ,DNA-Binding Proteins ,chemistry ,Trans-Activators ,Casein kinase 2 ,DNA Damage - Abstract
Aprataxin, defective in the neurodegenerative disorder ataxia oculomotor apraxia type 1, resolves abortive DNA ligation intermediates during DNA repair. Here, we demonstrate that aprataxin localizes at sites of DNA damage induced by high LET radiation and binds to mediator of DNA-damage checkpoint protein 1 (MDC1/NFBD1) through a phosphorylation-dependent interaction. This interaction is mediated via the aprataxin FHA domain and multiple casein kinase 2 di-phosphorylated S-D-T-D motifs in MDC1. X-ray structural and mutagenic analysis of aprataxin FHA domain, combined with modelling of the pSDpTD peptide interaction suggest an unusual FHA binding mechanism mediated by a cluster of basic residues at and around the canonical pT-docking site. Mutation of aprataxin FHA Arg29 prevented its interaction with MDC1 and recruitment to sites of DNA damage. These results indicate that aprataxin is involved not only in single strand break repair but also in the processing of a subset of double strand breaks presumably through its interaction with MDC1.
- Published
- 2009
49. Positional stability of damaged chromatin domains along radiation tracks in mammalian cells
- Author
-
G. Taucher-Scholz, J. Splinter, and Burkhard Jakob
- Subjects
Time Factors ,DNA damage ,Cell ,Biophysics ,Radiation ,Biology ,Models, Biological ,Chromosomes ,Cell Line ,Lesion ,Replication Protein A ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Irradiation ,Cell Nucleus ,Range (particle radiation) ,Intracellular Signaling Peptides and Proteins ,Fibroblasts ,Charged particle ,Chromatin ,Protein Structure, Tertiary ,Crystallography ,medicine.anatomical_structure ,Microscopy, Fluorescence ,medicine.symptom ,Tumor Suppressor p53-Binding Protein 1 ,DNA Damage ,HeLa Cells - Abstract
Irradiation of cell nuclei with charged particles leads to the spatially defined production of DNA damage along the particle trajectories, thus facilitating studies on the dynamics of radiation-induced protein foci associated with lesion processing. Here we used visual inspection and computational analysis of the track morphology after immunodetection to describe the patterns of formation of gamma-H2AX foci and the repair-related proteins 53BP1 and RPA. We addressed the influence of lesion density on gamma-H2AX formation and the mobility of damaged chromatin sites by using low-angle irradiation of cell monolayers with low-energy carbon or uranium ions. We show the discrete formation of gamma-H2AX foci and the recruitment of repair-related proteins along ion trajectories over an LET range from 200 to 14300 keV/microm in human fibroblasts and in HeLa cells. The marked DSBs exhibited a limited mobility that was independent of the LET. The moderate extent of mobility in human fibroblasts pointed to a relatively stable positioning of the damaged chromatin domains during repair, in contrast to HeLa cells, which showed significant changes in the streak patterns in a fraction of cells, suggesting greater mobility in the local processing of DSBs. Our data indicate that the presence of single or multiple DSBs is not associated with an altered potential for movement of damaged chromatin. We infer that the repair of high-LET radiation-induced DSBs in mammalian cells is not coupled to an increased motional activity of lesions enhancing the probability of translocations.
- Published
- 2009
50. Targeted irradiation of Mammalian cells using a heavy-ion microprobe
- Author
-
Burkhard Jakob, Claudia Fournier, Gudrun Becker, Bernd E. Fischer, Markus Heiß, and Gisela Taucher-Scholz
- Subjects
Microprobe ,Biophysics ,Analytical chemistry ,Cell Culture Techniques ,chemistry.chemical_element ,Electron ,Ion ,Optics ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Heavy Ions ,Linear Energy Transfer ,Irradiation ,Helium ,Cells, Cultured ,Range (particle radiation) ,Radiation ,business.industry ,Scattering ,Chemistry ,Microbeam ,Equipment Design ,Fibroblasts ,Equipment Failure Analysis ,Particle Accelerators ,business - Abstract
The existing focusing heavy-ion microprobe at the Gesellschaft für Schwerionenforschung in Darmstadt (Germany) has been modified to enable the targeted irradiation of single, selected cells with a defined number of ions. With this setup, ions in the range from helium to uranium with linear energy transfers (LETs) up to approximately 15,000 keV/microm can be positioned with a precision of a few micrometers in the nuclei of single cells that are growing in culture on a thin polypropylene film. To achieve this accuracy, the microbeam traverses a thin vacuum window with minimal scattering. Electron emission from that window is used for particle detection. The cells are kept in a specially designed dish that is mounted directly behind the vacuum window in a setup allowing the precise movement and the imaging of the sample with microscopic methods. The cells are located by an integrated software program that also controls the rapid deflection and switching of the beam. In this paper, the setup is described in detail together with the first experiments showing its performance. We describe the ability of the microprobe to reliably hit randomly positioned etched nuclear tracks in CR-39 with single ions as well as the ability to visualize the ion hits using immunofluorescence staining for 53BP1 as a marker of DNA damage in the targeted cell nuclei.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.