1
|
Ho JJ, Cheng E, Wong CJ, St-Germain JR, Dunham WH, Raught B, Gingras AC, Brown GW. The BLM-TOP3A-RMI1-RMI2 proximity map reveals that RAD54L2 suppresses sister chromatid exchanges. EMBO Rep 2025; 26:1290-1314. [PMID: 39870965 PMCID: PMC11894219 DOI: 10.1038/s44319-025-00374-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/29/2025] Open
Abstract
Homologous recombination is a largely error-free DNA repair mechanism conserved across all domains of life and is essential for the maintenance of genome integrity. Not only are the mutations in homologous recombination repair genes probable cancer drivers, some also cause genetic disorders. In particular, mutations in the Bloom (BLM) helicase cause Bloom Syndrome, a rare autosomal recessive disorder characterized by increased sister chromatid exchanges and predisposition to a variety of cancers. The pathology of Bloom Syndrome stems from the impaired activity of the BLM-TOP3A-RMI1-RMI2 (BTRR) complex which suppresses crossover recombination to prevent potentially deleterious genome rearrangements. We provide a comprehensive BTRR proximal proteome, revealing proteins that suppress crossover recombination. We find that RAD54L2, a SNF2-family protein, physically interacts with BLM and suppresses sister chromatid exchanges. RAD54L2 is important for recruitment of BLM to chromatin and requires an intact ATPase domain to promote non-crossover recombination. Thus, the BTRR proximity map identifies a regulator of recombination.
Collapse
Affiliation(s)
- Jung Jennifer Ho
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Edith Cheng
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Cassandra J Wong
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON, Canada
| | - Jonathan R St-Germain
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Wade H Dunham
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Grant W Brown
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada.
| |
Collapse
|
2
|
Palma-Rojo E, Barquinero JF, Pérez-Alija J, González JR, Armengol G. Differential biological effect of low doses of ionizing radiation depending on the radiosensitivity in a cell line model. Int J Radiat Biol 2024; 100:1527-1540. [PMID: 39288264 DOI: 10.1080/09553002.2024.2400514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/18/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE Exposure to low doses (LD) of ionizing radiation (IR), such as the ones employed in computed tomography (CT) examination, can be associated with cancer risk. However, cancer development could depend on individual radiosensitivity. In the present study, we evaluated the differences in the response to a CT-scan radiation dose of 20 mGy in two lymphoblastoid cell lines with different radiosensitivity. MATERIALS AND METHODS Several parameters were studied: gene expression, DNA damage, and its repair, as well as cell viability, proliferation, and death. Results were compared with those after a medium dose of 500 mGy. RESULTS After 20 mGy of IR, the radiosensitive (RS) cell line showed an increase in DNA damage, and higher cell proliferation and apoptosis, whereas the radioresistant (RR) cell line was insensitive to this LD. Interestingly, the RR cell line showed a higher expression of an antioxidant gene, which could be used by the cells as a protective mechanism. After a dose of 500 mGy, both cell lines were affected by IR but with significant differences. The RS cells presented an increase in DNA damage and apoptosis, but a decrease in cell proliferation and cell viability, as well as less antioxidant response. CONCLUSIONS A differential biological effect was observed between two cell lines with different radiosensitivity, and these differences are especially interesting after a CT scan dose. If this is confirmed by further studies, one could think that individuals with radiosensitivity-related genetic variants may be more vulnerable to long-term effects of IR, potentially increasing cancer risk after LD exposure.
Collapse
Affiliation(s)
- Elia Palma-Rojo
- Unitat d'Antropologia Biològica, Departament de Biologia Animal, Biologia Vegetal i Ecologia, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Joan-Francesc Barquinero
- Unitat d'Antropologia Biològica, Departament de Biologia Animal, Biologia Vegetal i Ecologia, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Jaime Pérez-Alija
- Servei de Radiofísica i Radioprotecció, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Juan R González
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
| | - Gemma Armengol
- Unitat d'Antropologia Biològica, Departament de Biologia Animal, Biologia Vegetal i Ecologia, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| |
Collapse
|
3
|
Audrey A, Kok YP, Yu S, de Haan L, van de Kooij B, van den Tempel N, Chen M, de Boer HR, van der Vegt B, van Vugt MATM. RAD52-dependent mitotic DNA synthesis is required for genome stability in Cyclin E1-overexpressing cells. Cell Rep 2024; 43:114116. [PMID: 38625790 DOI: 10.1016/j.celrep.2024.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/28/2024] [Accepted: 03/29/2024] [Indexed: 04/18/2024] Open
Abstract
Overexpression of Cyclin E1 perturbs DNA replication, resulting in DNA lesions and genomic instability. Consequently, Cyclin E1-overexpressing cancer cells increasingly rely on DNA repair, including RAD52-mediated break-induced replication during interphase. We show that not all DNA lesions induced by Cyclin E1 overexpression are resolved during interphase. While DNA lesions upon Cyclin E1 overexpression are induced in S phase, a significant fraction of these lesions is transmitted into mitosis. Cyclin E1 overexpression triggers mitotic DNA synthesis (MiDAS) in a RAD52-dependent fashion. Chemical or genetic inactivation of MiDAS enhances mitotic aberrations and persistent DNA damage. Mitosis-specific degradation of RAD52 prevents Cyclin E1-induced MiDAS and reduces the viability of Cyclin E1-overexpressing cells, underscoring the relevance of RAD52 during mitosis to maintain genomic integrity. Finally, analysis of breast cancer samples reveals a positive correlation between Cyclin E1 amplification and RAD52 expression. These findings demonstrate the importance of suppressing mitotic defects in Cyclin E1-overexpressing cells through RAD52.
Collapse
Affiliation(s)
- Anastasia Audrey
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Yannick P Kok
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Shibo Yu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Lauren de Haan
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Bert van de Kooij
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Nathalie van den Tempel
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Mengting Chen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - H Rudolf de Boer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands.
| |
Collapse
|
4
|
Kuchi Bhotla H, Balasubramanian B, Rengasamy KRR, Arumugam VA, Alagamuthu KK, Chithravel V, Chaudhary A, Alanazi AM, Pappuswamy M, Meyyazhagan A. Genotoxic repercussion of high-intensity radiation (x-rays) on hospital radiographers. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2023; 64:123-131. [PMID: 36541415 DOI: 10.1002/em.22523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 05/24/2023]
Abstract
Recent technological advances in the medical field have increased the plausibility of exposing humans to high-intensity wavelength radiations like x-rays and gamma rays while diagnosing or treating specific medical maladies. These radiations induce nucleotide changes and chromosomal alterations in the exposed population, intentionally or accidentally. A radiological investigation is regularly used in identifying the disease, especially by the technicians working in intensive care units. The current study observes the genetic damages like chromosomal abnormalities (CA) in clinicians who are occupationally exposed to high-intensity radiations (x-rays) at their workplaces using universal cytogenetic tools like micronucleus assay (MN), sister chromatid exchange and comet assay. The study was conducted between 100 exposed practitioners from the abdominal scanning, chest scanning, cranial and orthopedic or bone scanning department and age-matched healthy controls. We observed a slightly higher rate of MN and CA (p < .05) in orthopedic and chest department practitioners than in other departments concerning increasing age and duration of exposure at work. Our results emphasize taking extra precautionary measures in clinical and hospital radiation laboratories to protect the practitioners.
Collapse
Affiliation(s)
| | | | - Kannan R R Rengasamy
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Vijaya Anand Arumugam
- Medical Genetics and Epigenetics Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Karthick Kumar Alagamuthu
- Department of Biotechnology, Selvamm Arts and Science College (Autonomous), Namakkal, Tamil Nadu, India
| | | | - Aditi Chaudhary
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, India
| | - Amer M Alanazi
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Arun Meyyazhagan
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, India
| |
Collapse
|
5
|
Rampias T, Klinakis A. Using Sister Chromatid Exchange Assay to Detect Homologous Recombination Deficiency in Epigenetically Deregulated Urothelial Carcinoma Cells. Methods Mol Biol 2023; 2684:133-144. [PMID: 37410231 DOI: 10.1007/978-1-0716-3291-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Sister chromatid exchange (SCE) is the process of exchanging regions between two sister chromatids during DNA replication. Exchanges between replicated chromatids and their sisters can be visualized in cells when DNA synthesis in one chromatid is labelled by 5-bromo-2'-deoxyuridine (BrdU). Homologous recombination (HR) is considered as the principal mechanism responsible for the sister chromatid exchange (SCE) upon replication fork collapse, and therefore SCE frequency upon genotoxic conditions reflects the capacity of HR repair to respond to replication stress. During tumorigenesis, inactivating mutations or altered transcriptome can affect a plethora of epigenetic factors that participate in DNA repair processes, and there are an increasing number of reports which demonstrate a link between epigenetic deregulation in cancer and homologous recombination deficiency (HRD). Therefore, the SCE assay can provide valuable information regarding the HR functionality in tumors with epigenetic deficiencies. In this chapter, we provide a method to visualize SCEs. The technique outlined below is characterized by high sensitivity and specificity and has been successfully applied to human bladder cancer cell lines. In this context, this technique could be used to characterize the dynamics of HR repair in tumors with deregulated epigenome.
Collapse
Affiliation(s)
- Theodoros Rampias
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Apostolos Klinakis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
6
|
Sister chromatid exchanges induced by perturbed replication can form independently of BRCA1, BRCA2 and RAD51. Nat Commun 2022; 13:6722. [PMID: 36344511 PMCID: PMC9640580 DOI: 10.1038/s41467-022-34519-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Sister chromatid exchanges (SCEs) are products of joint DNA molecule resolution, and are considered to form through homologous recombination (HR). Indeed, SCE induction upon irradiation requires the canonical HR factors BRCA1, BRCA2 and RAD51. In contrast, replication-blocking agents, including PARP inhibitors, induce SCEs independently of BRCA1, BRCA2 and RAD51. PARP inhibitor-induced SCEs are enriched at difficult-to-replicate genomic regions, including common fragile sites (CFSs). PARP inhibitor-induced replication lesions are transmitted into mitosis, suggesting that SCEs can originate from mitotic processing of under-replicated DNA. Proteomics analysis reveals mitotic recruitment of DNA polymerase theta (POLQ) to synthetic DNA ends. POLQ inactivation results in reduced SCE numbers and severe chromosome fragmentation upon PARP inhibition in HR-deficient cells. Accordingly, analysis of CFSs in cancer genomes reveals frequent allelic deletions, flanked by signatures of POLQ-mediated repair. Combined, we show PARP inhibition generates under-replicated DNA, which is processed into SCEs during mitosis, independently of canonical HR factors.
Collapse
|
7
|
Hamed KA, El-Fiky SA, Gawish AM, Khalil WKB, Mohamed HRH. Alleviation of nicotine-induced reproductive disorder, clastogenicity, and histopathological alterations by fenugreek saponin bulk and nanoparticles in male rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:47488-47501. [PMID: 35182342 PMCID: PMC9232449 DOI: 10.1007/s11356-022-19123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/04/2022] [Indexed: 06/14/2023]
Abstract
Nicotine is the most abundant ingredient in cigarette smoking and has serious side effects on the lung, heart, reproductive system, and many other human organs. Saponins extracted from many plants exhibit multiple biological actions such as anti-cancer effects. Therefore, the possible protective effect of fenugreek saponin (FS) and nanofenugreek saponin (NFS) against nicotine-induced toxicity in male rats was investigated in this study. Animals were divided into a control group and the nicotine (1.5 mg/kg/day), FS (25, 50, and 100 mg/kg/day), or/and NFS (20, 40, and 80 mg/kg/day) administered groups. Micronucleus assay, histopathological, and sperm abnormality examinations as well as measurement of the acetylcholinesterase (AChE) gene expression were conducted. Our findings revealed that nicotine treatment induced significant increases in the incidence of micronucleus, sperm abnormalities, and expression levels of AChE in addition to inducing histopathological changes in rat testis. On the other hand, administration of FS or NFS with nicotine significantly decreased the incidence of micronuclei and the percentage of sperm abnormalities as well as the expression levels of AChE gene. Moreover, nicotine-induced histological alterations were reduced by given FS or NFS with nicotine. In conclusion, nicotine-induced sperm abnormalities, chromosomal damage, and histological injuries were mitigated by administration of FS or NFS with nicotine, and thus, FS and NFS could be used as ameliorating agents against nicotine toxicity.
Collapse
Affiliation(s)
- Karima A Hamed
- Department of Cell Biology, National Research Centre, 33 El-Bohous StDokki, P.O. 12622, Giza, 12622, Egypt
| | - Samia A El-Fiky
- Department of Cell Biology, National Research Centre, 33 El-Bohous StDokki, P.O. 12622, Giza, 12622, Egypt
| | - Azza M Gawish
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Wagdy K B Khalil
- Department of Cell Biology, National Research Centre, 33 El-Bohous StDokki, P.O. 12622, Giza, 12622, Egypt
| | - Hanan R H Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
8
|
Abstract
Homologous recombination (HR) repairs DNA double-strand breaks by using a homologous template to retrieve sequence information lost at the break site. The broken DNA molecule first engages with the homologous donor molecule and is then separated from it to complete the process. Depending on the HR subpathways used, the separation step can lead to crossovers (COs) between the participating molecules. Such events can cause genomic alterations and eventually cancer if a donor molecule other than the identical sister chromatid is used. Here, we characterize two subpathways of HR with different propensities to form COs. We show the unexpected dominance of the CO-forming subpathway and characterize the processes involved in CO formation and subpathway choice in cancer and normal, untransformed cells. Homologous recombination (HR) is an important DNA double-strand break (DSB) repair pathway that copies sequence information lost at the break site from an undamaged homologous template. This involves the formation of a recombination structure that is processed to restore the original sequence but also harbors the potential for crossover (CO) formation between the participating molecules. Synthesis-dependent strand annealing (SDSA) is an HR subpathway that prevents CO formation and is thought to predominate in mammalian cells. The chromatin remodeler ATRX promotes an alternative HR subpathway that has the potential to form COs. Here, we show that ATRX-dependent HR outcompetes RECQ5-dependent SDSA for the repair of most two-ended DSBs in human cells and leads to the frequent formation of COs, assessed by measuring sister chromatid exchanges (SCEs). We provide evidence that subpathway choice is dependent on interaction of both ATRX and RECQ5 with proliferating cell nuclear antigen. We also show that the subpathway usage varies among different cancer cell lines and compare it to untransformed cells. We further observe HR intermediates arising as ionizing radiation (IR)-induced ultra-fine bridges only in cells expressing ATRX and lacking MUS81 and GEN1. Consistently, damage-induced MUS81 recruitment is only observed in ATRX-expressing cells. Cells lacking BLM show similar MUS81 recruitment and IR-induced SCE formation as control cells. Collectively, these results suggest that the ATRX pathway involves the formation of HR intermediates whose processing is entirely dependent on MUS81 and GEN1 and independent of BLM. We propose that the predominant ATRX-dependent HR subpathway forms joint molecules distinct from classical Holliday junctions.
Collapse
|
9
|
Sakamoto Y, Kokuta T, Teshigahara A, Iijima K, Kitao H, Takata M, Tauchi H. Mitotic cells can repair DNA double-strand breaks via a homology-directed pathway. JOURNAL OF RADIATION RESEARCH 2021; 62:25-33. [PMID: 33009557 PMCID: PMC7779344 DOI: 10.1093/jrr/rraa095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/21/2020] [Indexed: 05/04/2023]
Abstract
The choice of repair pathways of DNA double-strand breaks (DSBs) is dependent upon the cell cycle phases. While homologous recombination repair (HRR) is active between the S and G2 phases, its involvement in mitotic DSB repair has not been examined in detail. In the present study, we developed a new reporter assay system to detect homology-directed repair (HDR), a major pathway used for HRR, in combination with an inducible DSB-generation system. As expected, the maximal HDR activity was observed in the late S phase, along with minimal activity in the G1 phase and at the G1/S boundary. Surprisingly, significant HDR activity was observed in M phase, and the repair efficiency was similar to that observed in late S phase. HDR was also confirmed in metaphase cells collected with continuous colcemid exposure. ChIP assays revealed the recruitment of RAD51 to the vicinity of DSBs in M phase. In addition, the ChIP assay for gamma-H2AX and phosphorylated DNA-PKcs indicated that a part of M-phase cells with DSBs could proceed into the next G1 phase. These results provide evidence showing that a portion of mitotic cell DSBs are undoubtedly repaired through action of the HDR repair pathway.
Collapse
Affiliation(s)
- Yuki Sakamoto
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Tetsuya Kokuta
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Ai Teshigahara
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Kenta Iijima
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
- Department of Cancer Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroyuki Kitao
- Department of Molecular Cancer Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Minoru Takata
- Radiation Biology Center, Kyoto University, Yoshida-Konoe Cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Tauchi
- Corresponding author. Department of Biological Sciences, Faculty of Science, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan. Tel: +81-29-228-8383; Fax: +81-29-228-8403;
| |
Collapse
|
10
|
Erdogan M, Fabritius A, Basquin J, Griesbeck O. Targeted In Situ Protein Diversification and Intra-organelle Validation in Mammalian Cells. Cell Chem Biol 2020; 27:610-621.e5. [PMID: 32142629 DOI: 10.1016/j.chembiol.2020.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/22/2019] [Accepted: 02/14/2020] [Indexed: 02/08/2023]
Abstract
Engineered proteins must be phenotypically selected for function in the appropriate physiological context. Here, we present a versatile approach that allows generating panels of mammalian cells that express diversified heterologous protein libraries in the cytosol or subcellular compartments under stable conditions and in a single-variant-per-cell manner. To this end we adapt CRISPR/Cas9 editing technology to diversify targeted stretches of a protein of interest in situ. We demonstrate the utility of the approach by in situ engineering and intra-lysosome specific selection of an extremely pH-resistant long Stokes shift red fluorescent protein variant. Tailoring properties to specific conditions of cellular sub-compartments or organelles of mammalian cells can be an important asset to optimize various proteins, protein-based tools, and biosensors for distinct functions.
Collapse
Affiliation(s)
- Mutlu Erdogan
- Tools for Bio-Imaging, Max-Planck-Institut für Neurobiologie, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Arne Fabritius
- Tools for Bio-Imaging, Max-Planck-Institut für Neurobiologie, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Jérome Basquin
- Structural Cell Biology, Max-Planck-Institut für Biochemie, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Oliver Griesbeck
- Tools for Bio-Imaging, Max-Planck-Institut für Neurobiologie, Am Klopferspitz 18, Martinsried 82152, Germany.
| |
Collapse
|
11
|
Abstract
Sister chromatid exchange (SCE) is the phenomenon of partial DNA exchange during DNA replication. SCE detection has been developed through eliciting DNA's semiconservative replicative nature. Thymidine analogues such as 5'-bromodeoxyuridine (BrdU) and ethynyldeoxyuridine (EdU) are incorporated into the newly synthesized DNA for two cell cycles. The addition of Colcemid to the culture blocks and synchronizes cells at mitosis, and conventional cytogenetic preparations are made. Differential staining methods with Hoechst dye and Giemsa (Fluorescence Plus Giemsa staining), antibody detection against BrdU, or highly specific Click reaction to EdU, allow the newly synthesized DNA within a chromatid to be recognized. SCEs represent a point of DNA template exchange during DNA synthesis, visualized by differential chromatid staining or harlequin chromosomes. We will introduce three basic protocols in this chapter including non-fluorescence and fluorescence methods for SCE microscopic analysis. SCE is a very sensitive marker of genotoxic stress during replication.
Collapse
|
12
|
Wójcik E, Szostek M. Assessment of genome stability in various breeds of cattle. PLoS One 2019; 14:e0217799. [PMID: 31163060 PMCID: PMC6548367 DOI: 10.1371/journal.pone.0217799] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/18/2019] [Indexed: 02/06/2023] Open
Abstract
Chromosomal instability is a type of genome instability involving changes in genetic information at the chromosomal level. The basic tests used to identify this form of instability are sister chromatid exchange (SCE) tests and identification of fragile sites (FS). SCE is the process by which sister chromatids become fragmented as a result of DNA strand breakage and reassembly, followed by exchange of these fragments. FS can be observed in the form of breaks, gaps or constrictions on chromosomes, which often result from multiple nucleotide repeats in DNA that are difficult to replicate. The research material was the peripheral blood of ten breeds of cattle raised in Poland, including four native breeds covered by a genetic resources conservation programme, i.e. Polish Red, Polish Red-and-White, White-Backed, and Polish Black-and-White, as well as Polish Holstein-Friesian, Simmental, Montbéliarde, Jersey, Limousine and Danish Red. Two tests were performed on chromosomes obtained from in vitro cultures: SCE and FS. The average frequency of SCE was 5.08 ± 1.31, while the incidence of FS was 3.45 ± 0.94. Differences in the incidence of SCE and FS were observed between breeds. The least damage was observed in the Polish Red and White-Backed breeds, and the most in Polish Holstein-Friesians. The most damage was observed in the interstitial part of the chromosomes. Age was shown to significantly affect the incidence of SCE and FS. Younger cows showed less damage than older ones (SCE: 4.84 ± 1.25; 5.34 ± 1.24; FS: 3.10 ± 0.88, 3.80 ± 0.92).
Collapse
Affiliation(s)
- Ewa Wójcik
- Institute of Bioengineering and Animal Breeding, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Małgorzata Szostek
- Institute of Bioengineering and Animal Breeding, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| |
Collapse
|
13
|
Soni A, Murmann-Konda T, Magin S, Iliakis G. A method for the cell-cycle-specific analysis of radiation-induced chromosome aberrations and breaks. Mutat Res 2019; 815:10-19. [PMID: 30999232 DOI: 10.1016/j.mrfmmm.2019.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 06/09/2023]
Abstract
The classical G2-assay is widely used to assess cell-radiosensitivity and cancer phenotype: Cells are exposed to low doses of ionizing-radiation (IR) and collected for cytogenetic- analysis ˜1.5 h later. In this way, chromosome-damage is measured in cells irradiated in G2-phase, without retrieving information regarding kinetics of chromosome-break-repair. Modification of the assay to include analysis at multiple time-points after IR, has enabled kinetic-analysis of chromatid-break-repair and assessment of damage in a larger proportion of G2-phase cells. This modification, however, increases the probability that at later time points not only cells irradiated in G2-phase, but also cells irradiated in S-phase will reach metaphase. However, the response of cells irradiated in G2-phase can be mechanistically different from that of cells irradiated in S-phase. Therefore, indiscriminate analysis may confound the interpretation of experiments designed to elucidate mechanisms of chromosome-break-repair and the contributions of the different DSB-repair-pathways in this response. Here we report an EdU based modification of the assay that enables S- and G2-phase specific analysis of chromatid break repair. Our results show that the majority of metaphases captured during the first 2 h after IR originate from cells irradiated in G2-phase (EdU- metaphases) in both rodent and human cells. Metaphases originating from cells irradiated in S-phase (EdU+ metaphases) start appearing at 2 h and 4 h after IR in rodent and human cells, respectively. The kinetics of chromatid-break-repair are similar in cells irradiated in G2- and S-phase of the cell-cycle, both in rodent and human cells. The protocol is applicable to classical-cytogenetic experiments and allows the cell-cycle specific analysis of chromosomal-aberrations. Finally, the protocol can be applied to the kinetic analysis of chromosome-breaks in prematurely-condensed-chromosomes of G2-phase cells. In summary, the developed protocol provides means to enhance the analysis of IR-induced-cytogenetic-damage by providing information on the cell-cycle phase where DNA damage is inflicted.
Collapse
Affiliation(s)
- Aashish Soni
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Tamara Murmann-Konda
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Simon Magin
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - George Iliakis
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany.
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Recent lymphoma genome sequencing projects have shed light on the genomic landscape of indolent and aggressive lymphomas, as well as some of the molecular mechanisms underlying recurrent mutations and translocations in these entities. Here, we review these recent genomic discoveries, focusing on acquired DNA repair defects in lymphoma. In addition, we highlight recently identified actionable molecular vulnerabilities associated with recurrent mutations in chronic lymphocytic leukemia (CLL), which serves as a model entity. RECENT FINDINGS The results of several large lymphoma genome sequencing projects have recently been reported, including CLL, T-PLL and DLBCL. We align these discoveries with proposed mechanisms of mutation acquisition in B-cell lymphomas. Moreover, novel autochthonous mouse models of CLL have recently been generated and we discuss how these models serve as preclinical tools to drive the development of novel targeted therapeutic interventions. Lastly, we highlight the results of early clinical data on novel compounds targeting defects in the DNA damage response of CLL with a particular focus on deleterious ATM mutations. SUMMARY Defects in DNA repair pathways are selected events in cancer, including lymphomas. Specifically, ATM deficiency is associated with PARP1- and DNA-PKcs inhibitor sensitivity in vitro and in vivo.
Collapse
|
15
|
Juhász S, Elbakry A, Mathes A, Löbrich M. ATRX Promotes DNA Repair Synthesis and Sister Chromatid Exchange during Homologous Recombination. Mol Cell 2018; 71:11-24.e7. [PMID: 29937341 DOI: 10.1016/j.molcel.2018.05.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/20/2018] [Accepted: 05/10/2018] [Indexed: 01/15/2023]
Abstract
ATRX is a chromatin remodeler that, together with its chaperone DAXX, deposits the histone variant H3.3 in pericentromeric and telomeric regions. Notably, ATRX is frequently mutated in tumors that maintain telomere length by a specific form of homologous recombination (HR). Surprisingly, in this context, we demonstrate that ATRX-deficient cells exhibit a defect in repairing exogenously induced DNA double-strand breaks (DSBs) by HR. ATRX operates downstream of the Rad51 removal step and interacts with PCNA and RFC-1, which are collectively required for DNA repair synthesis during HR. ATRX depletion abolishes DNA repair synthesis and prevents the formation of sister chromatid exchanges at exogenously induced DSBs. DAXX- and H3.3-depleted cells exhibit identical HR defects as ATRX-depleted cells, and both ATRX and DAXX function to deposit H3.3 during DNA repair synthesis. This suggests that ATRX facilitates the chromatin reconstitution required for extended DNA repair synthesis and sister chromatid exchange during HR.
Collapse
Affiliation(s)
- Szilvia Juhász
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Amira Elbakry
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Arthur Mathes
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Markus Löbrich
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany.
| |
Collapse
|
16
|
Anzalone DA, Sampino S, Czernik M, Iuso D, Ptak GE. Polychlorinated biphenyls (PCBs) alter DNA methylation and genomic integrity of sheep fetal cells in a simplified in vitro model of pregnancy exposure. Toxicol In Vitro 2018; 46:39-46. [DOI: 10.1016/j.tiv.2017.09.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 09/11/2017] [Accepted: 09/18/2017] [Indexed: 01/08/2023]
|
17
|
Knittel G, Rehkämper T, Korovkina D, Liedgens P, Fritz C, Torgovnick A, Al-Baldawi Y, Al-Maarri M, Cun Y, Fedorchenko O, Riabinska A, Beleggia F, Nguyen PH, Wunderlich FT, Ortmann M, Montesinos-Rongen M, Tausch E, Stilgenbauer S, P Frenzel L, Herling M, Herling C, Bahlo J, Hallek M, Peifer M, Buettner R, Persigehl T, Reinhardt HC. Two mouse models reveal an actionable PARP1 dependence in aggressive chronic lymphocytic leukemia. Nat Commun 2017; 8:153. [PMID: 28751718 PMCID: PMC5532225 DOI: 10.1038/s41467-017-00210-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) remains an incurable disease. Two recurrent cytogenetic aberrations, namely del(17p), affecting TP53, and del(11q), affecting ATM, are associated with resistance against genotoxic chemotherapy (del17p) and poor outcome (del11q and del17p). Both del(17p) and del(11q) are also associated with inferior outcome to the novel targeted agents, such as the BTK inhibitor ibrutinib. Thus, even in the era of targeted therapies, CLL with alterations in the ATM/p53 pathway remains a clinical challenge. Here we generated two mouse models of Atm- and Trp53-deficient CLL. These animals display a significantly earlier disease onset and reduced overall survival, compared to controls. We employed these models in conjunction with transcriptome analyses following cyclophosphamide treatment to reveal that Atm deficiency is associated with an exquisite and genotype-specific sensitivity against PARP inhibition. Thus, we generate two aggressive CLL models and provide a preclinical rational for the use of PARP inhibitors in ATM-affected human CLL. ATM and TP53 mutations are associated with poor prognosis in chronic lymphocytic leukaemia (CLL). Here the authors generate mouse models of Tp53- and Atm-defective CLL mimicking the high-risk form of human disease and show that Atm-deficient CLL is sensitive to PARP1 inhibition.
Collapse
Affiliation(s)
- Gero Knittel
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany. .,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany. .,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany.
| | - Tim Rehkämper
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Darya Korovkina
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Paul Liedgens
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Christian Fritz
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Alessandro Torgovnick
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Yussor Al-Baldawi
- Department of Radiology, Medical Faculty, University Hospital of Cologne, Cologne, 50931, Germany
| | - Mona Al-Maarri
- Max-Planck-Institute for Metabolism Research, Cologne, 50931, Germany
| | - Yupeng Cun
- Department of Translational Genomics, University of Cologne, Cologne, 50931, Germany
| | - Oleg Fedorchenko
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Arina Riabinska
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Filippo Beleggia
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Phuong-Hien Nguyen
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | | | - Monika Ortmann
- Institute of Pathology, University Hospital of Cologne, Cologne, 50931, Germany
| | | | - Eugen Tausch
- Department of Internal Medicine III, Ulm University, Ulm, 89070, Germany
| | | | - Lukas P Frenzel
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Marco Herling
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, 50931, Germany
| | - Carmen Herling
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Jasmin Bahlo
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany
| | - Michael Hallek
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Martin Peifer
- Department of Translational Genomics, University of Cologne, Cologne, 50931, Germany
| | - Reinhard Buettner
- Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany.,Institute of Pathology, University Hospital of Cologne, Cologne, 50931, Germany
| | - Thorsten Persigehl
- Department of Radiology, Medical Faculty, University Hospital of Cologne, Cologne, 50931, Germany
| | - H Christian Reinhardt
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany. .,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany. .,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany. .,Center of Molecular Medicine, University of Cologne, Cologne, 50931, Germany.
| |
Collapse
|
18
|
Patel A, Anderson J, Kraft D, Finnon R, Finnon P, Scudamore CL, Manning G, Bulman R, Brown N, Bouffler S, O'Neill P, Badie C. The Influence of the CTIP Polymorphism, Q418P, on Homologous Recombination and Predisposition to Radiation-Induced Tumorigenesis (mainly rAML) in Mice. Radiat Res 2016; 186:638-649. [DOI: 10.1667/rr14495.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Agata Patel
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Jennifer Anderson
- DNA Damage Group, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Daniela Kraft
- GSI Helmholtzzentrum für Schwerionenforschung GmbH Planckstraße 1, 64291 Darmstadt, 11-05-52, Germany and
| | - Rosemary Finnon
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Paul Finnon
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Cheryl L. Scudamore
- Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom
| | - Grainne Manning
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Robert Bulman
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Natalie Brown
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Simon Bouffler
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| | - Peter O'Neill
- DNA Damage Group, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Christophe Badie
- Cancer Genetics and Cytogenetics Group, Radiation Effects Department, Centre for Radiation Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, United Kingdom
| |
Collapse
|
19
|
Foertsch F, Szambowska A, Weise A, Zielinski A, Schlott B, Kraft F, Mrasek K, Borgmann K, Pospiech H, Grosse F, Melle C. S100A11 plays a role in homologous recombination and genome maintenance by influencing the persistence of RAD51 in DNA repair foci. Cell Cycle 2016; 15:2766-79. [PMID: 27590262 DOI: 10.1080/15384101.2016.1220457] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) is an essential process in maintenance of chromosomal stability. A key player of HR is the strand exchange factor RAD51 whose assembly at sites of DNA damage is tightly regulated. We detected an endogenous complex of RAD51 with the calcium-binding protein S100A11, which is localized at sites of DNA repair in HaCaT cells as well as in normal human epidermal keratinocytes (NHEK) synchronized in S phase. In biochemical assays, we revealed that S100A11 enhanced the RAD51 strand exchange activity. When cells expressing a S100A11 mutant lacking the ability to bind Ca(2+), a prolonged persistence of RAD51 in repair sites and nuclear γH2AX foci was observed suggesting an incomplete DNA repair. The same phenotype became apparent when S100A11 was depleted by RNA interference. Furthermore, down-regulation of S100A11 resulted in both reduced sister chromatid exchange confirming the restriction of the recombination capacity of the cells, and in an increase of chromosomal aberrations reflecting the functional requirement of S100A11 for the maintenance of genomic stability. Our data indicate that S100A11 is involved in homologous recombination by regulating the appearance of RAD51 in DSB repair sites. This function requires the calcium-binding activity of S100A11.
Collapse
Affiliation(s)
- Franziska Foertsch
- a Biomolecular Photonics Group , Jena University Hospital , Jena , Germany
| | - Anna Szambowska
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany
| | - Anja Weise
- c Institute of Human Genetics , Jena University Hospital , Jena , Germany
| | - Alexandra Zielinski
- d Radiobiology & Experimental Radiooncology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Bernhard Schlott
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany
| | - Florian Kraft
- c Institute of Human Genetics , Jena University Hospital , Jena , Germany
| | - Kristin Mrasek
- c Institute of Human Genetics , Jena University Hospital , Jena , Germany
| | - Kerstin Borgmann
- d Radiobiology & Experimental Radiooncology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Helmut Pospiech
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany.,e Faculty of Biochemistry and Molecular Medicine , University of Oulu , Finland
| | - Frank Grosse
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany
| | - Christian Melle
- a Biomolecular Photonics Group , Jena University Hospital , Jena , Germany
| |
Collapse
|
20
|
Spies J, Waizenegger A, Barton O, Sürder M, Wright WD, Heyer WD, Löbrich M. Nek1 Regulates Rad54 to Orchestrate Homologous Recombination and Replication Fork Stability. Mol Cell 2016; 62:903-917. [PMID: 27264870 DOI: 10.1016/j.molcel.2016.04.032] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/23/2016] [Accepted: 04/26/2016] [Indexed: 11/17/2022]
Abstract
Never-in-mitosis A-related kinase 1 (Nek1) has established roles in apoptosis and cell cycle regulation. We show that human Nek1 regulates homologous recombination (HR) by phosphorylating Rad54 at Ser572 in late G2 phase. Nek1 deficiency as well as expression of unphosphorylatable Rad54 (Rad54-S572A) cause unresolved Rad51 foci and confer a defect in HR. Phospho-mimic Rad54 (Rad54-S572E), in contrast, promotes HR and rescues the HR defect associated with Nek1 loss. Although expression of phospho-mimic Rad54 is beneficial for HR, it causes Rad51 removal from chromatin and degradation of stalled replication forks in S phase. Thus, G2-specific phosphorylation of Rad54 by Nek1 promotes Rad51 chromatin removal during HR in G2 phase, and its absence in S phase is required for replication fork stability. In summary, Nek1 regulates Rad51 removal to orchestrate HR and replication fork stability.
Collapse
Affiliation(s)
- Julian Spies
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Anja Waizenegger
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Olivia Barton
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Michael Sürder
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - William D Wright
- Section of Microbiology, University of California, Davis, Davis, CA 95616-8665, USA
| | - Wolf-Dietrich Heyer
- Section of Microbiology, University of California, Davis, Davis, CA 95616-8665, USA
| | - Markus Löbrich
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany.
| |
Collapse
|
21
|
Knittel G, Liedgens P, Reinhardt HC. Targeting ATM-deficient CLL through interference with DNA repair pathways. Front Genet 2015; 6:207. [PMID: 26113859 PMCID: PMC4461826 DOI: 10.3389/fgene.2015.00207] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/28/2015] [Indexed: 11/13/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in the Western world and accounts for approximately 30% of adult leukemias and 25% of non-Hodgkin lymphomas. The median age at diagnosis is 72 years. During recent years numerous genetic aberrations have been identified that are associated with an aggressive course of the disease and resistance against genotoxic chemotherapies. The DNA damage-responsive proapoptotic ATM-CHK2-p53 signaling pathway is frequently mutationally inactivated in CLL either through large deletions on chromosome 11q (ATM) or 17p (TP53), or through protein-damaging mutations. Here, we focus on the role of ATM signaling for the immediate DNA damage response, DNA repair and leukemogenesis. We further discuss novel therapeutic concepts for the targeted treatment of ATM-defective CLLs. We specifically highlight the potential use of PARP1 and DNA-PKcs inhibitors for the treatment of ATM-mutant CLL clones. Lastly, we briefly discuss the current state of genetically engineered mouse models of the disease and emphasize the use of these preclinical tools as a common platform for the development and validation of novel therapeutic agents.
Collapse
Affiliation(s)
- Gero Knittel
- Department of Internal Medicine, University Hospital of CologneCologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of CologneCologne, Germany
| | - Paul Liedgens
- Department of Internal Medicine, University Hospital of CologneCologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of CologneCologne, Germany
| | - Hans C. Reinhardt
- Department of Internal Medicine, University Hospital of CologneCologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of CologneCologne, Germany
| |
Collapse
|
22
|
Rodríguez-Mercado JJ, Hernández-de la Cruz H, Felipe-Reyes M, Jaramillo-Cruz E, Altamirano-Lozano MA. Evaluation of cytogenetic and DNA damage caused by thallium(I) acetate in human blood cells. ENVIRONMENTAL TOXICOLOGY 2015; 30:572-580. [PMID: 24318865 DOI: 10.1002/tox.21934] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 11/25/2013] [Indexed: 06/02/2023]
Abstract
Although thallium is detrimental to all living organisms, information regarding the mutagenic and genotoxic effects of this element and its compounds remains scarce. Therefore, we tested the genotoxic and cytotoxic effects of thallium(I) acetate on human peripheral blood cells in vitro using structural chromosomal aberrations (SCAs), sister chromatid exchanges (SCEs), and single-cell gel electrophoresis (at pH >13 or 12.1) analysis. Whole blood samples were incubated with 0.5, 1, 5, 10, 50, or 100 µg/mL thallium salt. Exposure to this metal compound resulted in a clear dose-dependent reduction in the mitotic and replicative indices. An increase in SCAs was evident in the treated group compared with the control group, and significant differences were observed in the percentage of cells with SCAs when metaphase cells were treated with 0.5-10 µg/mL of thallium(I). The SCE test did not reveal any significant differences. We observed that a 1-h treatment with thallium(I) at pH > 13 significantly increased the comet length for all the concentrations tested; however, at pH 12.1, only the two highest concentrations affected the comet length. These results suggested that thallium(I) acetate induces cytotoxic, cytostatic, and clastogenic effects, as well as DNA damage.
Collapse
Affiliation(s)
- Juan J Rodríguez-Mercado
- Unidad de Investigación en Genética y Toxicología Ambiental (UIGTA), Laboratorio L5 PA, Unidad Multidisciplinaria de Investigación Experimental (UMIE-Z), Facultad de Estudios Superiores-Zaragoza, Campus II, UNAM, AP 9-020, CP 15000, Ciudad de México, México
| | | | | | | | | |
Collapse
|
23
|
Bakr A, Oing C, Köcher S, Borgmann K, Dornreiter I, Petersen C, Dikomey E, Mansour WY. Involvement of ATM in homologous recombination after end resection and RAD51 nucleofilament formation. Nucleic Acids Res 2015; 43:3154-66. [PMID: 25753674 PMCID: PMC4381069 DOI: 10.1093/nar/gkv160] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 02/18/2015] [Indexed: 12/03/2022] Open
Abstract
Ataxia-telangiectasia mutated (ATM) is needed for the initiation of the double-strand break (DSB) repair by homologous recombination (HR). ATM triggers DSB end resection by stimulating the nucleolytic activity of CtIP and MRE11 to generate 3′-ssDNA overhangs, followed by RPA loading and RAD51 nucleofilament formation. Here we show for the first time that ATM is also needed for later steps in HR after RAD51 nucleofilament formation. Inhibition of ATM after completion of end resection did not affect RAD51 nucleofilament formation, but resulted in HR deficiency as evidenced by (i) an increase in the number of residual RAD51/γH2AX foci in both S and G2 cells, (ii) the decrease in HR efficiency as detected by HR repair substrate (pGC), (iii) a reduced SCE rate and (iv) the radiosensitization of cells by PARP inhibition. This newly described role for ATM was found to be dispensable in heterochromatin-associated DSB repair, as KAP1-depletion did not alleviate the HR-deficiency when ATM was inhibited after end resection. Moreover, we demonstrated that ATR can partly compensate for the deficiency in early, but not in later, steps of HR upon ATM inhibition. Taken together, we describe here for the first time that ATM is needed not only for the initiation but also for the completion of HR.
Collapse
Affiliation(s)
- A Bakr
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - C Oing
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - S Köcher
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - K Borgmann
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - I Dornreiter
- Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, Hamburg 20251, Germany
| | - C Petersen
- Department of Radiotherapy & Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - E Dikomey
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - W Y Mansour
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany Tumor Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| |
Collapse
|
24
|
De Pascalis I, Pilato B, Mazzotta A, Dell'Endice TS, Rubini V, Simone G, Paradiso A, Aiello V, Mangia A. Sister chromatid exchange: A possible approach to characterize familial breast cancer patients. Oncol Rep 2014; 33:930-4. [PMID: 25434423 DOI: 10.3892/or.2014.3628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/17/2014] [Indexed: 11/05/2022] Open
Abstract
Sister chromatid exchange (SCE) frequency is widely used as an indicator of spontaneous chromosome instability. We investigated SCE frequency in the peripheral blood lymphocytes of familial and sporadic breast cancer (BC) patients from the Apulian Caucasian Population. Eighty-one patients were enrolled: 22 with familial history and 59 sporadic patients. Eleven familial patients had an 'increased risk' of BRCA gene mutation (BRCAPro ≥ 10%) and were candidates for BRCA1 and BRCA2 mutation analysis. For these reasons, we stratified the 22 familial BC patients in two group: 'low-risk' (n=11) and 'high-risk' (n=11) patients for BRCA gene mutations. Two of these 11 'high-risk' patients (18%) had pathogenic mutations in the BRCA2 gene. The subjects were not cigarette smokers or alcohol or drug users, and had no genetic disorders or chronic diseases affecting the family. Our results showed a significant increase in SCE frequency in the familial (5.305 ± 1.088/metaphase) (P<0.0001) and the sporadic patients (3.943 ± 0.552) (P<0.0001) compared to the controls (3.197 ± 0.649). We found that the SCE frequency was always significantly higher in familial than in sporadic patients, regardless of their clinicopathological characteristics. Moreover, we observed that the frequency of SCE in BRCA2 mutation carrier patients was higher compared to patients without mutations in BRCA1/2 genes. These findings highlight an intrinsic genomic instability in familial patients, and we suggest that SCE frequency may be used as a biomarker to better characterize familial BC.
Collapse
Affiliation(s)
- Ivana De Pascalis
- Functional Biomorphology Laboratory, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Brunella Pilato
- Molecular Genetics Laboratory, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Annalisa Mazzotta
- Functional Biomorphology Laboratory, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | | | - Vincenza Rubini
- Pathology Department, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Giovanni Simone
- Pathology Department, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Angelo Paradiso
- Experimental Medical Oncology, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | | | - Anita Mangia
- Functional Biomorphology Laboratory, NCRC, IRCSS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| |
Collapse
|
25
|
Lee K, Gollahon LS. Zscan4 interacts directly with human Rap1 in cancer cells regardless of telomerase status. Cancer Biol Ther 2014; 15:1094-105. [PMID: 24840609 DOI: 10.4161/cbt.29220] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Telomeres are repetitive sequences at the ends of chromosomes protected by DNA binding proteins of the shelterin complex that form capping structures. Through the interaction of shelterin complex-associated proteins, telomere length maintenance is regulated. Recently, the newly identified embryonic stem cell marker, Zinc finger and SCAN domain-containing 4 gene (Zscan4), was shown to be a telomere-associated protein, co-localizing to the shelterin complex. Furthermore, it was shown to play an essential role in genomic stability by regulating telomere elongation. Although it is known that Zscan4 regulates TRF2, POT1b, and Rap1 expression in embryonic stem cells, the relationship and the exact mechanism of action for ZSscan4-mediated telomere maintenance in cancer cells is unknown. In this study, we investigated Zscan4 expression and interactions with Rap1 in telomerase positive (HeLa, MCF7) and ALT pathway (SaOS2, U2OS) cancer cells. Through western, pulldown, siRNA, and overexpression assays we demonstrate, for the first time, that Zscan4 directly associates with Rap1 (physical association protein). Furthermore, by generating truncated versions of Zscan4, we identified its zinc finger domain as the Rap1 binding site. Using bimolecular fluorescence complementation, we further validate this functional interaction in human cancer cells. Our results indicate that Zscan4 functions as a mediator of telomere length through its direct interaction with Rap1, possibly regulating shelterin complex-controlled telomere elongation in both telomerase positive and alternative lengthening of telomere pathways. This direct interaction between Zscan4 and Rap1 may explain how Zscan4 rapidly increases telomere length, yielding important information about the role of these proteins in telomere biology.
Collapse
Affiliation(s)
- Kyungwoo Lee
- Department of Biological Sciences; Texas Tech University; Lubbock, TX USA
| | - Lauren S Gollahon
- Department of Biological Sciences; Texas Tech University; Lubbock, TX USA; Texas Tech University Imaging Center; Lubbock, TX USA
| |
Collapse
|
26
|
Mutagenicity and genotoxicity of dicapthon insecticide. Cytotechnology 2014; 66:741-51. [PMID: 24477548 DOI: 10.1007/s10616-013-9623-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/19/2013] [Indexed: 10/25/2022] Open
Abstract
Mutagenic and genotoxic effects of dicapthon were investigated by using the bacterial reverse mutation assay in Salmonella typhimurium TA97, TA98, TA100 and TA102 strains with or without metabolic activation system (S9 mix), and chromosome aberrations (CAs), sister chromatid exchanges (SCEs), and micronucleus (MN) tests in human peripheral blood lymphocytes in vitro. Dicapthon was dissolved in dimethyl sulfoxide for all test systems. 0.1, 1, 10 and 100 μg/plate doses of dicapthon were found to be weakly mutagenic on S. typhimurium TA 98 without S9 mix. The human peripheral lymphocytes were treated with four experimental concentrations of dicapthon (25, 50, 100, and 200 μg/mL) for 24 and 48 h. Dicapthon increased the frequency of SCE only at the 100 μg/mL concentration for the 24 and 48 h applications. Dicapthon also induced abnormal cell frequency, CA/cell ratio and frequency of MN dose dependently for 24 and 48 h. Dicapthon showed a statistically significant cytotoxic effect by decreasing the mitotic index in all concentrations and a cytostatic effect by decreasing nuclear division index in 100 and 200 μg/mL concentrations for both treatment periods when compared with both untreated and solvent controls. These values decreased also in a dose dependent manner.
Collapse
|
27
|
Kakarougkas A, Ismail A, Klement K, Goodarzi AA, Conrad S, Freire R, Shibata A, Lobrich M, Jeggo PA. Opposing roles for 53BP1 during homologous recombination. Nucleic Acids Res 2013; 41:9719-31. [PMID: 23969417 PMCID: PMC3834810 DOI: 10.1093/nar/gkt729] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although DNA non-homologous end-joining repairs most DNA double-strand breaks (DSBs) in G2 phase, late repairing DSBs undergo resection and repair by homologous recombination (HR). Based on parallels to the situation in G1 cells, previous work has suggested that DSBs that undergo repair by HR predominantly localize to regions of heterochromatin (HC). By using H3K9me3 and H4K20me3 to identify HC regions, we substantiate and extend previous evidence, suggesting that HC-DSBs undergo repair by HR. Next, we examine roles for 53BP1 and BRCA1 in this process. Previous studies have shown that 53BP1 is pro-non-homologous end-joining and anti-HR. Surprisingly, we demonstrate that in G2 phase, 53BP1 is required for HR at HC-DSBs with its role being to promote phosphorylated KAP-1 foci formation. BRCA1, in contrast, is dispensable for pKAP-1 foci formation but relieves the barrier caused by 53BP1. As 53BP1 is retained at irradiation-induced foci during HR, we propose that BRCA1 promotes displacement but retention of 53BP1 to allow resection and any necessary HC modifications to complete HR. In contrast to this role for 53BP1 in HR in G2 phase, we show that it is dispensable for HR in S phase, where HC regions are likely relaxed during replication.
Collapse
Affiliation(s)
- Andreas Kakarougkas
- DNA Double Strand Break Repair Laboratory, University of Sussex, Brighton BN1 9RQ, UK, Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, Alberta T2N 4N1, Canada, Radiation Biology and DNA Repair Laboratory, Darmstadt University of Technology, 64287 Darmstadt, Germany and Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Ofra s/n, 38320 La Laguna, Tenerife, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Geuting V, Reul C, Löbrich M. ATM release at resected double-strand breaks provides heterochromatin reconstitution to facilitate homologous recombination. PLoS Genet 2013; 9:e1003667. [PMID: 23935532 PMCID: PMC3731223 DOI: 10.1371/journal.pgen.1003667] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/10/2013] [Indexed: 01/20/2023] Open
Abstract
Non-homologous end-joining (NHEJ) and homologous recombination (HR) represent the two main pathways for repairing DNA double-strand breaks (DSBs). During the G2 phase of the mammalian cell cycle, both processes can operate and chromatin structure is one important factor which determines DSB repair pathway choice. ATM facilitates the repair of heterochromatic DSBs by phosphorylating and inactivating the heterochromatin building factor KAP-1, leading to local chromatin relaxation. Here, we show that ATM accumulation and activity is strongly diminished at DSBs undergoing end-resection during HR. Such DSBs remain unrepaired in cells devoid of the HR factors BRCA2, XRCC3 or RAD51. Strikingly, depletion of KAP-1 or expression of phospho-mimic KAP-1 allows repair of resected DSBs in the absence of BRCA2, XRCC3 or RAD51 by an erroneous PARP-dependent alt-NHEJ process. We suggest that DSBs in heterochromatin elicit initial local heterochromatin relaxation which is reversed during HR due to the release of ATM from resection break ends. The restored heterochromatic structure facilitates HR and prevents usage of error-prone alternative processes. Double-strand breaks (DSBs) are critical DNA lesions because they can lead to cell death or, which is even more devastating, the formation of genomic rearrangements. Cells are equipped with two main pathways to repair such lesions, homologous recombination (HR) and non-homologous end-joining (NHEJ). HR is an error-free process and completely restores the genetic information, whereas NHEJ has the potential to form genomic rearrangements. We have previously shown that the structure of the chromatin is one important factor which determines the choice between these two pathways, such that DSBs localizing to highly condensed heterochromatic regions are mainly repaired by HR and breaks in more open euchromatic DNA undergo repair by NHEJ. Here, we investigate this aspect of DSB repair pathway choice. We show that DSB end-resection, which channels DSB repair into the process of HR, counteracts the profound local relaxation which initially takes place at the break site and reconstitutes the heterochromatic structure. Cells which are genetically modified, such that they cannot reconstitute the heterochromatic structure at resected DSBs, fail to employ HR and instead repair heterochromatic DSBs by alternative NHEJ mechanisms. Thus, chromatin modifications which occur during the process of end-resection prevent error-prone repair pathways from generating genomic rearrangements.
Collapse
Affiliation(s)
- Verena Geuting
- Darmstadt University of Technology, Radiation Biology and DNA Repair, Darmstadt, Germany
| | | | | |
Collapse
|
29
|
Tug E, Kayhan G, Kan D, Guntekin S, Ergun MA. The evaluation of long-term effects of ionizing radiation through measurement of current sister chromatid exchange (SCE) rates in radiology technologists, compared with previous SCE values. Mutat Res 2013; 757:28-30. [PMID: 23867852 DOI: 10.1016/j.mrgentox.2013.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 02/12/2013] [Accepted: 04/05/2013] [Indexed: 11/29/2022]
Abstract
Ionizing radiation is a strong physical mutagen, causing breakage of phosphodiester bonds in DNA at any stage of the mitotic cycle. Analysis of sister chromatid exchange (SCE) has come into use as a sensitive DNA-damage indicator. We investigated the SCE rates in radiology technologists who are occupationally and chronically exposed to ionizing radiation. The study included 39 radiology technologists and 35 sex- and age-matched healthy controls. There was a statistically significant difference in the SCE frequency between radiology technologists and controls (p<0.0001). Additionally, previous SCE data of 10 radiology technologists were compared with current results regarding radiation exposure time. There was statistically significant difference between previous and current SCE values (p=0.005). The significant increase in the frequency of SCE in radiology technologists emphasizes the importance of radiation-protection procedures in order to minimize radiation exposure and avoid possible genotoxic effects. Comparison of two studies that measured SCE values of radiology technologists after 8 years also suggests that the genotoxic effect is reversible. In conclusion, radiation is still an important mutagenic agent despite improvements in daily working hours and conditions.
Collapse
Affiliation(s)
- E Tug
- Gazi University, Faculty of Medicine, Department of Medical Genetics, Ankara, Turkey.
| | | | | | | | | |
Collapse
|
30
|
Riabinska A, Daheim M, Herter-Sprie GS, Winkler J, Fritz C, Hallek M, Thomas RK, Kreuzer KA, Frenzel LP, Monfared P, Martins-Boucas J, Chen S, Reinhardt HC. Therapeutic Targeting of a Robust Non-Oncogene Addiction to PRKDC in ATM-Defective Tumors. Sci Transl Med 2013; 5:189ra78. [DOI: 10.1126/scitranslmed.3005814] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
31
|
Abstract
Proper repair of DNA double strand breaks (DSBs) is vital for the preservation of genomic integrity. There are two main pathways that repair DSBs, Homologous recombination (HR) and Non-homologous end-joining (NHEJ). HR is restricted to the S and G2 phases of the cell cycle due to the requirement for the sister chromatid as a template, while NHEJ is active throughout the cell cycle and does not rely on a template. The balance between both pathways is essential for genome stability and numerous assays have been developed to measure the efficiency of the two pathways. Several proteins are known to affect the balance between HR and NHEJ and the complexity of the break also plays a role. In this review we describe several repair assays to determine the efficiencies of both pathways. We discuss how disturbance of the balance between HR and NHEJ can lead to disease, but also how it can be exploited for cancer treatment.
Collapse
Affiliation(s)
- Inger Brandsma
- Department of Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | | |
Collapse
|
32
|
Thompson LH. Losing and finding myself in DNA repair. DNA Repair (Amst) 2012; 11:637-48. [PMID: 23012750 DOI: 10.1016/j.dnarep.2011.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Larry H Thompson
- Biology & Biotechnology Division, L452, Lawrence Livermore National Laboratory, Livermore, CA 94551-0808, USA.
| |
Collapse
|
33
|
Thompson LH. Recognition, signaling, and repair of DNA double-strand breaks produced by ionizing radiation in mammalian cells: the molecular choreography. Mutat Res 2012; 751:158-246. [PMID: 22743550 DOI: 10.1016/j.mrrev.2012.06.002] [Citation(s) in RCA: 261] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 06/09/2012] [Accepted: 06/16/2012] [Indexed: 12/15/2022]
Abstract
The faithful maintenance of chromosome continuity in human cells during DNA replication and repair is critical for preventing the conversion of normal diploid cells to an oncogenic state. The evolution of higher eukaryotic cells endowed them with a large genetic investment in the molecular machinery that ensures chromosome stability. In mammalian and other vertebrate cells, the elimination of double-strand breaks with minimal nucleotide sequence change involves the spatiotemporal orchestration of a seemingly endless number of proteins ranging in their action from the nucleotide level to nucleosome organization and chromosome architecture. DNA DSBs trigger a myriad of post-translational modifications that alter catalytic activities and the specificity of protein interactions: phosphorylation, acetylation, methylation, ubiquitylation, and SUMOylation, followed by the reversal of these changes as repair is completed. "Superfluous" protein recruitment to damage sites, functional redundancy, and alternative pathways ensure that DSB repair is extremely efficient, both quantitatively and qualitatively. This review strives to integrate the information about the molecular mechanisms of DSB repair that has emerged over the last two decades with a focus on DSBs produced by the prototype agent ionizing radiation (IR). The exponential growth of molecular studies, heavily driven by RNA knockdown technology, now reveals an outline of how many key protein players in genome stability and cancer biology perform their interwoven tasks, e.g. ATM, ATR, DNA-PK, Chk1, Chk2, PARP1/2/3, 53BP1, BRCA1, BRCA2, BLM, RAD51, and the MRE11-RAD50-NBS1 complex. Thus, the nature of the intricate coordination of repair processes with cell cycle progression is becoming apparent. This review also links molecular abnormalities to cellular pathology as much a possible and provides a framework of temporal relationships.
Collapse
Affiliation(s)
- Larry H Thompson
- Biology & Biotechnology Division, L452, Lawrence Livermore National Laboratory, P.O. Box 808, Livermore, CA 94551-0808, United States.
| |
Collapse
|
34
|
Tekcan A, Elbistan M, Ulusoy AN. Sister chromatid exchanges in breast cancer patients who underwent chemotherapy. J Toxicol Sci 2012; 37:235-43. [DOI: 10.2131/jts.37.235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Akin Tekcan
- Department of Medical Biology and Genetics, Faculty of Medicine, Ondokuz Mayis University, Turkey
| | - Mehmet Elbistan
- Department of Medical Biology and Genetics, Faculty of Medicine, Ondokuz Mayis University, Turkey
| | - Ali Naki Ulusoy
- Department of General Surgery, Faculty of Medicine, Ondokuz Mayis University, Turkey
| |
Collapse
|
35
|
The role of homologous recombination in radiation-induced double-strand break repair. Radiother Oncol 2011; 101:7-12. [PMID: 21737170 DOI: 10.1016/j.radonc.2011.06.019] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/08/2011] [Accepted: 06/08/2011] [Indexed: 11/23/2022]
Abstract
DNA double-strand breaks (DSBs) represent the most biologically significant lesions induced by ionizing radiation (IR). HR is the predominant pathway for repairing one-ended DSBs arising in S-phase when the replication fork encounters single-stranded breaks or base damages. Here, we discuss recent findings that two-ended DSBs directly induced by X- or γ-rays in late S- or G2-phase are repaired predominantly by NHEJ, with HR only repairing a sub-fraction of such DSBs. This sub-fraction represents DSBs which localize to heterochromatic DNA regions and, which in control cells, are repaired with slow kinetics over many hours post irradiation. The observation that defined DSB populations are repaired by either NHEJ or HR suggests an assignment of specific tasks for each of the two processes. Furthermore, heavy ion induced complex DSBs, which are in general more slowly repaired than X- or γ-ray induced breaks, are nearly always repaired by HR independent of chromatin localization suggesting that the speed of repair is an important factor determining the DSB repair pathway usage. Finally, NHEJ and HR can, under certain conditions, also compensate for each other such that DSBs normally repaired by one pathway can undergo repair by the other if genetic failures necessitate the pathway switch.
Collapse
|