101
|
Abstract
Rad51 is a key component of homologous recombination (HR) to repair DNA double-strand breaks and it forms Rad51 recombinase filaments of broken single-stranded DNA to promote HR. In addition to its role in DNA repair and cell cycle progression, Rad51 contributes to the reprogramming process during the generation of induced pluripotent stem cells. In light of this, we performed reprogramming experiments to examine the effect of co-expression of Rad51 and four reprogramming factors, Oct4, Sox2, Klf4, and c-Myc, on the reprogramming efficiency. Co-expression of Rad51 significantly increased the numbers of alkaline phosphatase-positive colonies and embryonic stem cell-like colonies during the process of reprogramming. Co-expression ofRad51 significantly increased the expression of epithelial markers at an early stage of reprogramming compared with control cells. Phosphorylated histone H2AX (γH2AX), which initiates the DNA double-strand break repair system, was highly accumulated in reprogramming intermediates upon co-expression of Rad51. This study identified a novel role of Rad51 in enhancing the reprogramming efficiency, possibly by facilitating mesenchymal-to-epithelial transition and by regulating a DNA damage repair pathway during the early phase of the reprogramming process.
Collapse
Affiliation(s)
- Jae-Young Lee
- Dept. of Biomedical Science, College of Life Science, CHA University, Seoul 06135, Korea
| | - Dae-Kwan Kim
- Dept. of Biomedical Science, College of Life Science, CHA University, Seoul 06135, Korea
| | - Jeong-Jae Ko
- Dept. of Biomedical Science, College of Life Science, CHA University, Seoul 06135, Korea
| | - Keun Pil Kim
- Dept. of Life Science, Chung-Ang University, Seoul 06975, Korea
| | - Kyung-Soon Park
- Dept. of Biomedical Science, College of Life Science, CHA University, Seoul 06135, Korea
| |
Collapse
|
102
|
The Tumor-Associated Variant RAD51 G151D Induces a Hyper-Recombination Phenotype. PLoS Genet 2016; 12:e1006208. [PMID: 27513445 PMCID: PMC4981402 DOI: 10.1371/journal.pgen.1006208] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/29/2016] [Indexed: 12/20/2022] Open
Abstract
The RAD51 protein plays a key role in the homology-directed repair of DNA double-strand breaks and is important for maintaining genome stability. Here we report on a novel human RAD51 variant found in an aggressive and therapy-refractive breast carcinoma. Expression of the RAD51 G151D variant in human breast epithelial cells increases the levels of homology-directed repair. Expression of RAD51 G151D in cells also promotes high levels of chromosomal aberrations and sister chromatid exchanges. In vitro, the purified RAD51 G151D protein directly and significantly enhances DNA strand exchange activity in the presence of RPA. In concordance with this result, co-incubation of G151D with BRCA2 resulted in a much higher level of strand-exchange activity compared to WT RAD51. Strikingly, the RAD51 G151D variant confers resistance to multiple DNA damaging agents, including ionizing radiation, mitomycin C, and doxorubicin. Our findings demonstrate that the RAD51 G151D somatic variant has a novel hyper-recombination phenotype and suggest that this property of the protein is important for the repair of DNA damage, leading to drug resistance. Therapeutic resistance is a major hurdle for the treatment and eradication of cancer. Furthermore, the development of therapeutic resistance significantly decreases patient survival and negatively impacts the quality of life of patients battling cancer. Cancer cells utilize a number of previously described mechanisms in order to overcome sensitivity to cancer therapeutics, including overexpression of RAD51. However, in this study we report a novel gain-of-function heterozygous somatic variant, RAD51 G151D, identified in a highly refractory and aggressive breast adenocarcinoma. RAD51 G151D induces a hyper-recombination phenotype in human cells resulting in increased resistance to therapeutics via enhanced HDR of DSBs. We further demonstrate enhanced DNA strand exchange activity in the presence of RPA, providing a possible mechanism for the hyper-recombination phenotype observed in cells. Our study presents a novel hyper-recombinant RAD51 tumor-associated variant (RAD51 G151D), providing the first evidence that links altered RAD51 function with therapeutic resistance as well as a novel genetic marker to identify patients at high risk for aggressive and refractory disease.
Collapse
|
103
|
Tran M, Tsarouhas V, Kegel A. Early development of Drosophila embryos requires Smc5/6 function during oogenesis. Biol Open 2016; 5:928-41. [PMID: 27288507 PMCID: PMC4958276 DOI: 10.1242/bio.019000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Mutations in structural maintenance of chromosomes (Smc) proteins are frequently associated with chromosomal abnormalities commonly observed in developmental disorders. However, the role of Smc proteins in development still remains elusive. To investigate Smc5/6 function during early embryogenesis we examined smc5 and smc6 mutants of the fruit fly Drosophila melanogaster using a combination of reverse genetics and microscopy approaches. Smc5/6 exhibited a maternally contributed function in maintaining chromosome stability during early embryo development, which manifested as female subfertility in its absence. Loss of Smc5/6 caused an arrest and a considerable delay in embryo development accompanied by fragmented nuclei and increased anaphase-bridge formation, respectively. Surprisingly, early embryonic arrest was attributable to the absence of Smc5/6 during oogenesis, which resulted in insufficient repair of pre-meiotic and meiotic DNA double-strand breaks. Thus, our findings contribute to the understanding of Smc proteins in higher eukaryotic development by highlighting a maternal function in chromosome maintenance and a link between oogenesis and early embryogenesis. Summary: Early emerging problems during oogenesis, such as DNA double-strand breaks, can affect chromosome duplication and segregation in embryogenesis in Drosophila. Moreover, environmental cues including temperature are important for proper oogenesis.
Collapse
Affiliation(s)
- Martin Tran
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm S-17177, Sweden
| | - Vasilios Tsarouhas
- Department of Molecular Bioscience, The Wenner-Gren Institute, Stockholm University, Stockholm S-10691, Sweden
| | - Andreas Kegel
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm S-17177, Sweden
| |
Collapse
|
104
|
Wang L, Zheng JN, Pei DS. The emerging roles of Jab1/CSN5 in cancer. Med Oncol 2016; 33:90. [DOI: 10.1007/s12032-016-0805-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/04/2016] [Indexed: 12/13/2022]
|
105
|
Stanley EC, Azzinaro PA, Vierra DA, Howlett NG, Irvine SQ. The Simple Chordate Ciona intestinalis Has a Reduced Complement of Genes Associated with Fanconi Anemia. Evol Bioinform Online 2016; 12:133-48. [PMID: 27279728 PMCID: PMC4898443 DOI: 10.4137/ebo.s37920] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 12/26/2022] Open
Abstract
Fanconi anemia (FA) is a human genetic disease characterized by congenital defects, bone marrow failure, and increased cancer risk. FA is associated with mutation in one of 24 genes. The protein products of these genes function cooperatively in the FA pathway to orchestrate the repair of DNA interstrand cross-links. Few model organisms exist for the study of FA. Seeking a model organism with a simpler version of the FA pathway, we searched the genome of the simple chordate Ciona intestinalis for homologs of the human FA-associated proteins. BLAST searches, sequence alignments, hydropathy comparisons, maximum likelihood phylogenetic analysis, and structural modeling were used to infer the likelihood of homology between C. intestinalis and human FA proteins. Our analysis indicates that C. intestinalis indeed has a simpler and potentially functional FA pathway. The C. intestinalis genome was searched for candidates for homology to 24 human FA and FA-associated proteins. Support was found for the existence of homologs for 13 of these 24 human genes in C. intestinalis. Members of each of the three commonly recognized FA gene functional groups were found. In group I, we identified homologs of FANCE, FANCL, FANCM, and UBE2T/FANCT. Both members of group II, FANCD2 and FANCI, have homologs in C. intestinalis. In group III, we found evidence for homologs of FANCJ, FANCO, FANCQ/ERCC4, FANCR/RAD51, and FANCS/BRCA1, as well as the FA-associated proteins ERCC1 and FAN1. Evidence was very weak for the existence of homologs in C. intestinalis for any other recognized FA genes. This work supports the notion that C. intestinalis, as a close relative of vertebrates, but having a much reduced complement of FA genes, offers a means of studying the function of certain FA proteins in a simpler pathway than that of vertebrate cells.
Collapse
Affiliation(s)
- Edward C Stanley
- Integrative and Evolutionary Biology Graduate Specialization, University of Rhode Island, Kingston, RI, USA
| | - Paul A Azzinaro
- Cell and Molecular Biology Graduate Specialization, University of Rhode Island, Kingston, RI, USA
| | - David A Vierra
- Cell and Molecular Biology Graduate Specialization, University of Rhode Island, Kingston, RI, USA
| | - Niall G Howlett
- Cell and Molecular Biology Graduate Specialization, University of Rhode Island, Kingston, RI, USA.; Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI, USA
| | - Steven Q Irvine
- Integrative and Evolutionary Biology Graduate Specialization, University of Rhode Island, Kingston, RI, USA.; Department of Biological Sciences, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
106
|
Franz A, Ackermann L, Hoppe T. Ring of Change: CDC48/p97 Drives Protein Dynamics at Chromatin. Front Genet 2016; 7:73. [PMID: 27200082 PMCID: PMC4853748 DOI: 10.3389/fgene.2016.00073] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/16/2016] [Indexed: 12/31/2022] Open
Abstract
The dynamic composition of proteins associated with nuclear DNA is a fundamental property of chromosome biology. In the chromatin compartment dedicated protein complexes govern the accurate synthesis and repair of the genomic information and define the state of DNA compaction in vital cellular processes such as chromosome segregation or transcription. Unscheduled or faulty association of protein complexes with DNA has detrimental consequences on genome integrity. Consequently, the association of protein complexes with DNA is remarkably dynamic and can respond rapidly to cellular signaling events, which requires tight spatiotemporal control. In this context, the ring-like AAA+ ATPase CDC48/p97 emerges as a key regulator of protein complexes that are marked with ubiquitin or SUMO. Mechanistically, CDC48/p97 functions as a segregase facilitating the extraction of substrate proteins from the chromatin. As such, CDC48/p97 drives molecular reactions either by directed disassembly or rearrangement of chromatin-bound protein complexes. The importance of this mechanism is reflected by human pathologies linked to p97 mutations, including neurodegenerative disorders, oncogenesis, and premature aging. This review focuses on the recent insights into molecular mechanisms that determine CDC48/p97 function in the chromatin environment, which is particularly relevant for cancer and aging research.
Collapse
Affiliation(s)
- André Franz
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Institute for Genetics, University of Cologne Cologne, Germany
| | - Leena Ackermann
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Institute for Genetics, University of Cologne Cologne, Germany
| | - Thorsten Hoppe
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Institute for Genetics, University of Cologne Cologne, Germany
| |
Collapse
|
107
|
Tsai RYL. Balancing self-renewal against genome preservation in stem cells: How do they manage to have the cake and eat it too? Cell Mol Life Sci 2016; 73:1803-23. [PMID: 26886024 PMCID: PMC5040593 DOI: 10.1007/s00018-016-2152-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/18/2016] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
Stem cells are endowed with the awesome power of self-renewal and multi-lineage differentiation that allows them to be major contributors to tissue homeostasis. Owing to their longevity and self-renewal capacity, they are also faced with a higher risk of genomic damage compared to differentiated cells. Damage on the genome, if not prevented or repaired properly, will threaten the survival of stem cells and culminate in organ failure, premature aging, or cancer formation. It is therefore of paramount importance that stem cells remain genomically stable throughout life. Given their unique biological and functional requirement, stem cells are thought to manage genotoxic stress somewhat differently from non-stem cells. The focus of this article is to review the current knowledge on how stem cells escape the barrage of oxidative and replicative DNA damage to stay in self-renewal. A clear statement on this subject should help us better understand tissue regeneration, aging, and cancer.
Collapse
Affiliation(s)
- Robert Y L Tsai
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, 2121 W. Holcombe Blvd, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA.
| |
Collapse
|
108
|
Peiris TH, Ramirez D, Barghouth PG, Ofoha U, Davidian D, Weckerle F, Oviedo NJ. Regional signals in the planarian body guide stem cell fate in the presence of genomic instability. Development 2016; 143:1697-709. [PMID: 27013241 DOI: 10.1242/dev.131318] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 03/10/2016] [Indexed: 12/28/2022]
Abstract
Cellular fate decisions are influenced by their topographical location in the adult body. For instance, tissue repair and neoplastic growth are greater in anterior than in posterior regions of adult animals. However, the molecular underpinnings of these regional differences are unknown. We identified a regional switch in the adult planarian body upon systemic disruption of homologous recombination with RNA-interference of Rad51 Rad51 knockdown increases DNA double-strand breaks (DSBs) throughout the body, but stem cells react differently depending on their location along the anteroposterior axis. In the presence of extensive DSBs, cells in the anterior part of the body resist death, whereas cells in the posterior region undergo apoptosis. Furthermore, we found that proliferation of cells with DNA damage is induced in the presence of brain tissue and that the retinoblastoma pathway enables overproliferation of cells with DSBs while attending to the demands of tissue growth and repair. Our results implicate both autonomous and non-autonomous mechanisms as key mediators of regional cell behavior and cellular transformation in the adult body.
Collapse
Affiliation(s)
- T Harshani Peiris
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Daniel Ramirez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Paul G Barghouth
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Udokanma Ofoha
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Devon Davidian
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Frank Weckerle
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA
| | - Néstor J Oviedo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA 95343, USA Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| |
Collapse
|
109
|
Schay G, Borka B, Kernya L, Bulyáki É, Kardos J, Fekete M, Fidy J. Without Binding ATP, Human Rad51 Does Not Form Helical Filaments on ssDNA. J Phys Chem B 2016; 120:2165-78. [PMID: 26890079 DOI: 10.1021/acs.jpcb.5b12220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Construction of the presynaptic filament (PSF) of proper helical structure by Rad51 recombinases is a prerequisite of the progress of homologous recombination repair. We studied the contribution of ATP-binding to this structure of wt human Rad51 (hRad51). We exploited the protein-dissociation effect of high hydrostatic pressure to determine the free energy of dissociation of the protomer interfaces in hRad51 oligomer states and used electron microscopy to obtain topological parameters. Without cofactors ATP and Ca(2+) and template DNA, hRad51 did not exist in monomer form, but it formed rodlike long filaments without helical order. ΔG(diss) indicated a strong inherent tendency of aggregation. Binding solely ssDNA left the filament unstructured with slightly increased ΔG(diss). Adding only ATP and Ca(2+) to the buffer disintegrated the self-associated rods into rings and short helices of further increased ΔG(diss). Rad51 binding to ssDNA only with ATP and Ca bound could lead to ordered helical filament formation of proper pitch size with interface contacts of K(d) ∼ 2 × 10(-11) M, indicating a structure of outstanding stability. ATP/Ca binding increased the ΔG(diss) of protomer contacts in the filament by 16 kJ/mol. The results emphasize that ATP-binding in the PSF of hRad51 has an essential, yet purely structural, role.
Collapse
Affiliation(s)
- Gusztáv Schay
- Department of Biophysics and Radiation Biology, Semmelweis University , Tűzoltó utca 37-47, Budapest H-1094, Hungary
| | - Bálint Borka
- Department of Biophysics and Radiation Biology, Semmelweis University , Tűzoltó utca 37-47, Budapest H-1094, Hungary
| | - Linda Kernya
- MTA-ELTE NAP B Neuroimmunology Research Group, Department of Biochemistry, Eötvös Loránd University , Pázmány P. sétány 1/C, Budapest H-1117, Hungary
| | - Éva Bulyáki
- MTA-ELTE NAP B Neuroimmunology Research Group, Department of Biochemistry, Eötvös Loránd University , Pázmány P. sétány 1/C, Budapest H-1117, Hungary
| | - József Kardos
- MTA-ELTE NAP B Neuroimmunology Research Group, Department of Biochemistry, Eötvös Loránd University , Pázmány P. sétány 1/C, Budapest H-1117, Hungary
| | - Melinda Fekete
- Department of Biophysics and Radiation Biology, Semmelweis University , Tűzoltó utca 37-47, Budapest H-1094, Hungary
| | - Judit Fidy
- Department of Biophysics and Radiation Biology, Semmelweis University , Tűzoltó utca 37-47, Budapest H-1094, Hungary
| |
Collapse
|
110
|
Ahuja AK, Jodkowska K, Teloni F, Bizard AH, Zellweger R, Herrador R, Ortega S, Hickson ID, Altmeyer M, Mendez J, Lopes M. A short G1 phase imposes constitutive replication stress and fork remodelling in mouse embryonic stem cells. Nat Commun 2016; 7:10660. [PMID: 26876348 PMCID: PMC4756311 DOI: 10.1038/ncomms10660] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 01/08/2016] [Indexed: 12/15/2022] Open
Abstract
Embryonic stem cells (ESCs) represent a transient biological state, where pluripotency is coupled with fast proliferation. ESCs display a constitutively active DNA damage response (DDR), but its molecular determinants have remained elusive. Here we show in cultured ESCs and mouse embryos that H2AX phosphorylation is dependent on Ataxia telangiectasia and Rad3 related (ATR) and is associated with chromatin loading of the ssDNA-binding proteins RPA and RAD51. Single-molecule analysis of replication intermediates reveals massive ssDNA gap accumulation, reduced fork speed and frequent fork reversal. All these marks of replication stress do not impair the mitotic process and are rapidly lost at differentiation onset. Delaying the G1/S transition in ESCs allows formation of 53BP1 nuclear bodies and suppresses ssDNA accumulation, fork slowing and reversal in the following S-phase. Genetic inactivation of fork slowing and reversal leads to chromosomal breakage in unperturbed ESCs. We propose that rapid cell cycle progression makes ESCs dependent on effective replication-coupled mechanisms to protect genome integrity. In fast proliferating embryonic stem cells (ESC) the DNA damage response is activated by mechanisms that are as yet elusive. Here, Ahuja et al. link the DNA damage response to replication stress in mouse ESCs, caused by a short G1 phase, and propose fork remodelling as maintaining genome stability in embryos.
Collapse
Affiliation(s)
- Akshay K Ahuja
- Institute of Molecular Cancer Research, University of Zurich, Zurich CH-8057, Switzerland
| | - Karolina Jodkowska
- DNA Replication Group, Molecular Oncology Programme, CNIO, Madrid E-28029, Spain
| | - Federico Teloni
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich CH-8057, Switzerland
| | - Anna H Bizard
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, University of Copenhagen, Panum Institute, Copenhagen N DK-2200, Denmark
| | - Ralph Zellweger
- Institute of Molecular Cancer Research, University of Zurich, Zurich CH-8057, Switzerland
| | - Raquel Herrador
- Institute of Molecular Cancer Research, University of Zurich, Zurich CH-8057, Switzerland
| | - Sagrario Ortega
- Transgenic Mice Core Unit, Biotechnology Programme, CNIO, Madrid E-28029, Spain
| | - Ian D Hickson
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, University of Copenhagen, Panum Institute, Copenhagen N DK-2200, Denmark
| | - Matthias Altmeyer
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich CH-8057, Switzerland
| | - Juan Mendez
- DNA Replication Group, Molecular Oncology Programme, CNIO, Madrid E-28029, Spain
| | - Massimo Lopes
- Institute of Molecular Cancer Research, University of Zurich, Zurich CH-8057, Switzerland
| |
Collapse
|
111
|
Shao J, Xu Z, Peng X, Chen M, Zhu Y, Xu L, Zhu H, Yang B, Luo P, He Q. Gefitinib Synergizes with Irinotecan to Suppress Hepatocellular Carcinoma via Antagonizing Rad51-Mediated DNA-Repair. PLoS One 2016; 11:e0146968. [PMID: 26752698 PMCID: PMC4709237 DOI: 10.1371/journal.pone.0146968] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/23/2015] [Indexed: 11/24/2022] Open
Abstract
Chemotherapy is the only choice for most of the advanced hepatocellular carcinoma (HCC) patients, while few agents were available, making it an urgent need to develop new chemotherapy strategies. A phase II clinical trial suggested that the efficacy of irinotecan in HCC was limited due to dose-dependent toxicities. Here, we found that gefitinib exhibited synergistic activity in combination with SN-38, an active metabolite of irinotecan, in HCC cell lines. And the enhanced apoptosis induced by gefitinib plus SN-38 was a result from caspase pathway activation. Mechanistically, gefitinib dramatically promoted the ubiquitin–proteasome-dependent degradation of Rad51 protein, suppressed the DNA repair, gave rise to more DNA damages, and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of gefitinib combined with irinotecan was further validated in a HepG2 xenograft mice model. Taken together, our data demonstrated for the first time that the combination of irinotecan and gefitinib showed potential benefit in HCC, which suggests that Rad51 is a promising target and provides a rationale for clinical trials investigating the efficacy of the combination of topoisomerase I inhibitors and gefitinib in HCC.
Collapse
Affiliation(s)
- Jinjin Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhifei Xu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xueming Peng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Min Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanrun Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Xu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Luo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- * E-mail: (PL); (QH)
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- * E-mail: (PL); (QH)
| |
Collapse
|
112
|
Ameziane N, May P, Haitjema A, van de Vrugt HJ, van Rossum-Fikkert SE, Ristic D, Williams GJ, Balk J, Rockx D, Li H, Rooimans MA, Oostra AB, Velleuer E, Dietrich R, Bleijerveld OB, Maarten Altelaar AF, Meijers-Heijboer H, Joenje H, Glusman G, Roach J, Hood L, Galas D, Wyman C, Balling R, den Dunnen J, de Winter JP, Kanaar R, Gelinas R, Dorsman JC. A novel Fanconi anaemia subtype associated with a dominant-negative mutation in RAD51. Nat Commun 2015; 6:8829. [PMID: 26681308 PMCID: PMC4703882 DOI: 10.1038/ncomms9829] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/07/2015] [Indexed: 12/17/2022] Open
Abstract
Fanconi anaemia (FA) is a hereditary disease featuring hypersensitivity to DNA cross-linker-induced chromosomal instability in association with developmental abnormalities, bone marrow failure and a strong predisposition to cancer. A total of 17 FA disease genes have been reported, all of which act in a recessive mode of inheritance. Here we report on a de novo g.41022153G>A; p.Ala293Thr (NM_002875) missense mutation in one allele of the homologous recombination DNA repair gene RAD51 in an FA-like patient. This heterozygous mutation causes a novel FA subtype, 'FA-R', which appears to be the first subtype of FA caused by a dominant-negative mutation. The patient, who features microcephaly and mental retardation, has reached adulthood without the typical bone marrow failure and paediatric cancers. Together with the recent reports on RAD51-associated congenital mirror movement disorders, our results point to an important role for RAD51-mediated homologous recombination in neurodevelopment, in addition to DNA repair and cancer susceptibility.
Collapse
Affiliation(s)
- Najim Ameziane
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, House of Biomedicine, 7 Avenue des Hauts-Fourneaux, Esch/Alzette L-4362, Luxembourg
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109-5234, USA
| | - Anneke Haitjema
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Henri J. van de Vrugt
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
- Division of Biological Stress Response, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Sari E. van Rossum-Fikkert
- Department of Genetics, Cancer Genomics Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- Department of Radiation Oncology, Erasmus Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Dejan Ristic
- Department of Genetics, Cancer Genomics Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- Department of Radiation Oncology, Erasmus Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Gareth J. Williams
- Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California 94720, USA
| | - Jesper Balk
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Davy Rockx
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Hong Li
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109-5234, USA
| | - Martin A. Rooimans
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Anneke B. Oostra
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Eunike Velleuer
- Department of Paediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Ralf Dietrich
- Deutsche Fanconi-Anämie-Hilfe e.V., Böckenweg 4, 59427 Unna, Germany
| | - Onno B. Bleijerveld
- Mass Spectrometry and Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - A. F. Maarten Altelaar
- Mass Spectrometry and Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Hanne Meijers-Heijboer
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Hans Joenje
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Gustavo Glusman
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109-5234, USA
| | - Jared Roach
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109-5234, USA
| | - Leroy Hood
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109-5234, USA
| | - David Galas
- Luxembourg Centre for Systems Biomedicine, House of Biomedicine, 7 Avenue des Hauts-Fourneaux, Esch/Alzette L-4362, Luxembourg
- Pacific Northwest Diabetes Research Institute, 720 Broadway, Seattle, Washington 98122, USA
| | - Claire Wyman
- Department of Genetics, Cancer Genomics Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- Department of Radiation Oncology, Erasmus Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, House of Biomedicine, 7 Avenue des Hauts-Fourneaux, Esch/Alzette L-4362, Luxembourg
| | - Johan den Dunnen
- Department of Human and Clinical Genetics, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands
| | - Johan P. de Winter
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| | - Roland Kanaar
- Department of Genetics, Cancer Genomics Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- Department of Radiation Oncology, Erasmus Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Richard Gelinas
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109-5234, USA
| | - Josephine C. Dorsman
- Department of Clinical Genetics, VU University Medical Center, Van der Boechorststraat 7, Amsterdam 1081 BT, The Netherlands
| |
Collapse
|
113
|
Cdk12 is essential for embryonic development and the maintenance of genomic stability. Cell Death Differ 2015; 23:1038-48. [PMID: 26658019 PMCID: PMC4987723 DOI: 10.1038/cdd.2015.157] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/28/2015] [Accepted: 10/30/2015] [Indexed: 01/06/2023] Open
Abstract
The maintenance of genomic integrity during early embryonic development is important in order to ensure the proper development of the embryo. Studies from cultured cells have demonstrated that cyclin-dependent kinase 12 (Cdk12) is a multifunctional protein that maintains genomic stability and the pluripotency of embryonic stem cells. Perturbation of its functions is also known to be associated with pathogenesis and drug resistance in human cancers. However, the biological significance of Cdk12 in vivo is unclear. Here we bred mice that are deficient in Cdk12 and demonstrated that Cdk12 depletion leads to embryonic lethality shortly after implantation. We also used an in vitro culture system of blastocysts to examine the molecular mechanisms associated with the embryonic lethality of Cdk12-deficient embryos. Cdk12−/− blastocysts fail to undergo outgrowth of the inner cell mass because of an increase in the apoptosis of these cells. Spontaneous DNA damage was revealed by an increase in 53BP1 foci among cells cultured from Cdk12−/− embryos. Furthermore, the expression levels of various DNA damage response genes, namely Atr, Brca1, Fanci and Fancd2, are reduced in Cdk12−/− embryos. These findings indicate that Cdk12 is important for the correct expression of some DNA damage response genes and indirectly has an influence on the efficiency of DNA repair. Our report also highlights that DNA breaks occurring during DNA replication are frequent in mouse embryonic cells and repair of such damage is critical to the successful development of mouse embryos.
Collapse
|
114
|
Lu LY, Yu X. Double-strand break repair on sex chromosomes: challenges during male meiotic prophase. Cell Cycle 2015; 14:516-25. [PMID: 25565522 DOI: 10.1080/15384101.2014.998070] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
During meiotic prophase, DNA double-strand break (DSB) repair-mediated homologous recombination (HR) occurs for exchange of genetic information between homologous chromosomes. Unlike autosomes or female sex chromosomes, human male sex chromosomes X and Y share little homology. Although DSBs are generated throughout male sex chromosomes, homologous recombination does not occur for most regions and DSB repair process is significantly prolonged. As a result, male sex chromosomes are coated with many DNA damage response proteins and form a unique chromatin structure known as the XY body. Interestingly, associated with the prolonged DSB repair, transcription is repressed in the XY body but not in autosomes, a phenomenon known as meiotic sex chromosome inactivation (MSCI), which is critical for male meiosis. Here using mice as model organisms, we briefly summarize recent progress on DSB repair in meiotic prophase and focus on the mechanism and function of DNA damage response in the XY body.
Collapse
Affiliation(s)
- Lin-Yu Lu
- a Women's Hospital ; School of Medicine ; Zhejiang University ; Hangzhou , Zhejiang , China
| | | |
Collapse
|
115
|
DNA Damage Signalling and Repair Inhibitors: The Long-Sought-After Achilles' Heel of Cancer. Biomolecules 2015; 5:3204-59. [PMID: 26610585 PMCID: PMC4693276 DOI: 10.3390/biom5043204] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
For decades, radiotherapy and chemotherapy were the two only approaches exploiting DNA repair processes to fight against cancer. Nowadays, cancer therapeutics can be a major challenge when it comes to seeking personalized targeted medicine that is both effective and selective to the malignancy. Over the last decade, the discovery of new targeted therapies against DNA damage signalling and repair has offered the possibility of therapeutic improvements in oncology. In this review, we summarize the current knowledge of DNA damage signalling and repair inhibitors, their molecular and cellular effects, and future therapeutic use.
Collapse
|
116
|
Mahajan K, Mahajan NP. Cross talk of tyrosine kinases with the DNA damage signaling pathways. Nucleic Acids Res 2015; 43:10588-601. [PMID: 26546517 PMCID: PMC4678820 DOI: 10.1093/nar/gkv1166] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/21/2015] [Indexed: 01/19/2023] Open
Abstract
Tyrosine kinases respond to extracellular and intracellular cues by activating specific cellular signaling cascades to regulate cell cycle, growth, proliferation, differentiation and survival. Likewise, DNA damage response proteins (DDR) activated by DNA lesions or chromatin alterations recruit the DNA repair and cell cycle checkpoint machinery to restore genome integrity and cellular homeostasis. Several new examples have been uncovered in recent studies which reveal novel epigenetic and non-epigenetic mechanisms by which tyrosine kinases interact with DDR proteins to dictate cell fate, i.e. survival or apoptosis, following DNA damage. These studies reveal the ability of tyrosine kinases to directly regulate the activity of DNA repair and cell cycle check point proteins by tyrosine phosphorylation. In addition, tyrosine kinases epigenetically regulate DNA damage signaling pathways by modifying the core histones as well as chromatin modifiers at critical tyrosine residues. Thus, deregulated tyrosine kinase driven epigenomic alterations have profound implications in cancer, aging and genetic disorders. Consequently, targeting oncogenic tyrosine kinase induced epigenetic alterations has gained significant traction in overcoming cancer cell resistance to various therapies. This review discusses mechanisms by which tyrosine kinases interact with DDR pathways to regulate processes critical for maintaining genome integrity as well as clinical strategies for targeted cancer therapies.
Collapse
Affiliation(s)
- Kiran Mahajan
- Tumor Biology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA Department of Oncological Sciences, University of South Florida, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Nupam P Mahajan
- Drug Discovery Department, Moffitt Cancer Center, University of South Florida, 12902 Magnolia Drive, Tampa, FL 33612, USA Department of Oncological Sciences, University of South Florida, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
117
|
Wang AT, Kim T, Wagner JE, Conti BA, Lach FP, Huang AL, Molina H, Sanborn EM, Zierhut H, Cornes BK, Abhyankar A, Sougnez C, Gabriel SB, Auerbach AD, Kowalczykowski SC, Smogorzewska A. A Dominant Mutation in Human RAD51 Reveals Its Function in DNA Interstrand Crosslink Repair Independent of Homologous Recombination. Mol Cell 2015; 59:478-90. [PMID: 26253028 DOI: 10.1016/j.molcel.2015.07.009] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 05/26/2015] [Accepted: 07/07/2015] [Indexed: 12/17/2022]
Abstract
Repair of DNA interstrand crosslinks requires action of multiple DNA repair pathways, including homologous recombination. Here, we report a de novo heterozygous T131P mutation in RAD51/FANCR, the key recombinase essential for homologous recombination, in a patient with Fanconi anemia-like phenotype. In vitro, RAD51-T131P displays DNA-independent ATPase activity, no DNA pairing capacity, and a co-dominant-negative effect on RAD51 recombinase function. However, the patient cells are homologous recombination proficient due to the low ratio of mutant to wild-type RAD51 in cells. Instead, patient cells are sensitive to crosslinking agents and display hyperphosphorylation of Replication Protein A due to increased activity of DNA2 and WRN at the DNA interstrand crosslinks. Thus, proper RAD51 function is important during DNA interstrand crosslink repair outside of homologous recombination. Our study provides a molecular basis for how RAD51 and its associated factors may operate in a homologous recombination-independent manner to maintain genomic integrity.
Collapse
Affiliation(s)
- Anderson T Wang
- Laboratory of Genome Maintenance, The Rockefeller University, New York, NY 10065, USA
| | - Taeho Kim
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA 95616, USA
| | - John E Wagner
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brooke A Conti
- Laboratory of Genome Maintenance, The Rockefeller University, New York, NY 10065, USA
| | - Francis P Lach
- Laboratory of Genome Maintenance, The Rockefeller University, New York, NY 10065, USA
| | - Athena L Huang
- Laboratory of Genome Maintenance, The Rockefeller University, New York, NY 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Erica M Sanborn
- Laboratory of Genome Maintenance, The Rockefeller University, New York, NY 10065, USA
| | - Heather Zierhut
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | - Carrie Sougnez
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Arleen D Auerbach
- Human Genetics and Hematology, The Rockefeller University, New York, NY 10065, USA
| | - Stephen C Kowalczykowski
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA 95616, USA
| | - Agata Smogorzewska
- Laboratory of Genome Maintenance, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
118
|
Langsfeld ES, Bodily JM, Laimins LA. The Deacetylase Sirtuin 1 Regulates Human Papillomavirus Replication by Modulating Histone Acetylation and Recruitment of DNA Damage Factors NBS1 and Rad51 to Viral Genomes. PLoS Pathog 2015; 11:e1005181. [PMID: 26405826 PMCID: PMC4583417 DOI: 10.1371/journal.ppat.1005181] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/01/2015] [Indexed: 11/18/2022] Open
Abstract
Human papillomaviruses (HPV) regulate their differentiation-dependent life cycles by activating a number of cellular pathways, such as the DNA damage response, through control of post-translational protein modification. Sirtuin 1 (SIRT1) is a protein deacetylase that modulates the acetylation of a number of cellular substrates, resulting in activation of pathways controlling gene expression and DNA damage repair. Our studies indicate that SIRT1 levels are increased in cells containing episomes of high-risk HPV types through the combined action of the E6 and E7 oncoproteins. Knockdown of SIRT1 in these cells with shRNAs impairs viral activities including genome maintenance, amplification and late gene transcription, with minimal effects on cellular proliferation ability. Abrogation of amplification was also seen following treatment with the SIRT1 deacetylase inhibitor, EX-527. Importantly, SIRT1 binds multiple regions of the HPV genome in undifferentiated cells, but this association is lost upon of differentiation. SIRT1 regulates the acetylation of Histone H1 (Lys26) and H4 (Lys16) bound to HPV genomes and this may contribute to regulation of viral replication and gene expression. The differentiation-dependent replication of high-risk HPVs requires activation of factors in the Ataxia Telangiectasia Mutated (ATM) pathway and SIRT1 regulates the recruitment of both NBS1 and Rad51 to the viral genomes. These observations demonstrate that SIRT1 is a critical regulator of multiple aspects of the high-risk HPV life cycle. Human papillomaviruses regulate their differentiation-dependent life cycles by activating a number of cellular pathways, such as the DNA damage response, through control of post-translational protein modification. Sirtuin 1 (SIRT1) is a protein deacetylase that regulates the acetylation of a number of cellular substrates, resulting in activation of pathways involved in gene expression and DNA damage repair. We report here that SIRT1 protein levels are elevated in cells stably maintaining genomes of oncogenic HPVs and that SIRT1 knockdown impairs genome maintenance, productive replication and late gene transcription. The DNA damage sensing and repair pathways are critical for the HPV viral life cycle and members of this pathway, such as NBS1 and Rad51, are targets of SIRT1. Our studies demonstrate that SIRT1 binds the HPV genome and regulates both viral chromatin remodeling as well as binding of members of the homologous repair pathway to viral DNA. These findings demonstrate that binding of SIRT1 to the HPV genome is necessary for histone deacetylation and recruitment of DNA damage repair factors and is a critical step in the HPV life cycle.
Collapse
Affiliation(s)
- Erika S. Langsfeld
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Jason M. Bodily
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Laimonis A. Laimins
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
119
|
Mucha B, Kabzinski J, Dziki A, Przybylowska-Sygut K, Sygut A, Majsterek I, Dziki L. Polymorphism within the distal RAD51 gene promoter is associated with colorectal cancer in a Polish population. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:11601-11607. [PMID: 26617897 PMCID: PMC4637713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/21/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers in developed countries. Annually, over one million of new cases in the world are recorded. Majority of CRCs occur sporadically with dominant phenotype of chromosomal instability (CIN). Permanent exposure to DNA damaging agents such as ionizing radiation result in DNA double-stranded breaks, which create favorable conditions for chromosomal aberration to arise. Homologous recombination repair (HRR) is the leading process engaged in maintaining of the genome integrity. RAD51 protein was recognized as crucial in HRR. Single nucleotide polymorphisms are the primary source of genetic variation which presence in the RAD51 promoter region can affect on its expression and consequently modulate HR efficiency. OBJECTIVES The aim of this study was to analyze the distribution of genotypes and allele frequencies of -4791A/T and -4601A/G RAD51 gene polymorphisms, followed by an assessment of their relationship with the risk of CRC. MATERIAL AND METHODS The study included 115 patients with confirmed CRC. Control group was consisted of 118 cancer-free individuals with a negative family history. The genotypes were identified by PCR-RFLP method. CONCLUSION This study revealed statistically significant association between appearance of G/A genotype in position -4601 of RAD51 gene and CRC risk.
Collapse
Affiliation(s)
- Bartosz Mucha
- Department of Clinical Chemistry and Biochemistry, Medical University of LodzPoland
| | - Jacek Kabzinski
- Department of Clinical Chemistry and Biochemistry, Medical University of LodzPoland
| | - Adam Dziki
- Department of General and Colorectal Surgery Medical University in LodzPoland
| | | | - Andrzej Sygut
- Department of General and Vascular Surgery, Medical Center of PabianicePoland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of LodzPoland
| | - Lukasz Dziki
- Department of General and Colorectal Surgery Medical University in LodzPoland
| |
Collapse
|
120
|
Rothkamm K, Barnard S, Moquet J, Ellender M, Rana Z, Burdak-Rothkamm S. DNA damage foci: Meaning and significance. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:491-504. [PMID: 25773265 DOI: 10.1002/em.21944] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 02/13/2015] [Indexed: 06/04/2023]
Abstract
The discovery of DNA damage response proteins such as γH2AX, ATM, 53BP1, RAD51, and the MRE11/RAD50/NBS1 complex, that accumulate and/or are modified in the vicinity of a chromosomal DNA double-strand break to form microscopically visible, subnuclear foci, has revolutionized the detection of these lesions and has enabled studies of the cellular machinery that contributes to their repair. Double-strand breaks are induced directly by a number of physical and chemical agents, including ionizing radiation and radiomimetic drugs, but can also arise as secondary lesions during replication and DNA repair following exposure to a wide range of genotoxins. Here we aim to review the biological meaning and significance of DNA damage foci, looking specifically at a range of different settings in which such markers of DNA damage and repair are being studied and interpreted.
Collapse
Affiliation(s)
- Kai Rothkamm
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
- Department of Radiotherapy, Laboratory of Radiation Biology and Experimental Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephen Barnard
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| | - Jayne Moquet
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| | - Michele Ellender
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| | - Zohaib Rana
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, United Kingdom
| | - Susanne Burdak-Rothkamm
- Department of Cellular Pathology, Oxford University Hospitals, Headley Way, Headington, Oxford, United Kingdom
| |
Collapse
|
121
|
Kostyrko K, Bosshard S, Urban Z, Mermod N. A role for homologous recombination proteins in cell cycle regulation. Cell Cycle 2015; 14:2853-61. [PMID: 26125600 PMCID: PMC4614994 DOI: 10.1080/15384101.2015.1049784] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022] Open
Abstract
Eukaryotic cells respond to DNA breaks, especially double-stranded breaks (DSBs), by activating the DNA damage response (DDR), which encompasses DNA repair and cell cycle checkpoint signaling. The DNA damage signal is transmitted to the checkpoint machinery by a network of specialized DNA damage-recognizing and signal-transducing molecules. However, recent evidence suggests that DNA repair proteins themselves may also directly contribute to the checkpoint control. Here, we investigated the role of homologous recombination (HR) proteins in normal cell cycle regulation in the absence of exogenous DNA damage. For this purpose, we used Chinese Hamster Ovary (CHO) cells expressing the Fluorescent ubiquitination-based cell cycle indicators (Fucci). Systematic siRNA-mediated knockdown of HR genes in these cells demonstrated that the lack of several of these factors alters cell cycle distribution, albeit differentially. The knock-down of MDC1, Rad51 and Brca1 caused the cells to arrest in the G2 phase, suggesting that they may be required for the G2/M transition. In contrast, inhibition of the other HR factors, including several Rad51 paralogs and Rad50, led to the arrest in the G1/G0 phase. Moreover, reduced expression of Rad51B, Rad51C, CtIP and Rad50 induced entry into a quiescent G0-like phase. In conclusion, the lack of many HR factors may lead to cell cycle checkpoint activation, even in the absence of exogenous DNA damage, indicating that these proteins may play an essential role both in DNA repair and checkpoint signaling.
Collapse
Affiliation(s)
- Kaja Kostyrko
- Institute of Biotechnology; University of Lausanne; and Center for Biotechnology UNIL-EPFL; Lausanne, Switzerland
| | - Sandra Bosshard
- Institute of Biotechnology; University of Lausanne; and Center for Biotechnology UNIL-EPFL; Lausanne, Switzerland
| | - Zuzanna Urban
- Institute of Biotechnology; University of Lausanne; and Center for Biotechnology UNIL-EPFL; Lausanne, Switzerland
| | - Nicolas Mermod
- Institute of Biotechnology; University of Lausanne; and Center for Biotechnology UNIL-EPFL; Lausanne, Switzerland
| |
Collapse
|
122
|
Chang HY, Liao CY, Su GC, Lin SW, Wang HW, Chi P. Functional Relationship of ATP Hydrolysis, Presynaptic Filament Stability, and Homologous DNA Pairing Activity of the Human Meiotic Recombinase DMC1. J Biol Chem 2015; 290:19863-73. [PMID: 26088134 DOI: 10.1074/jbc.m115.666289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Indexed: 11/06/2022] Open
Abstract
DMC1 and RAD51 are conserved recombinases that catalyze homologous recombination. DMC1 and RAD51 share similar properties in DNA binding, DNA-stimulated ATP hydrolysis, and catalysis of homologous DNA strand exchange. A large body of evidence indicates that attenuation of ATP hydrolysis leads to stabilization of the RAD51-ssDNA presynaptic filament and enhancement of DNA strand exchange. However, the functional relationship of ATPase activity, presynaptic filament stability, and DMC1-mediated homologous DNA strand exchange has remained largely unexplored. To address this important question, we have constructed several mutant variants of human DMC1 and characterized them biochemically to gain mechanistic insights. Two mutations, K132R and D223N, that change key residues in the Walker A and B nucleotide-binding motifs ablate ATP binding and render DMC1 inactive. On the other hand, the nucleotide-binding cap D317K mutant binds ATP normally but shows significantly attenuated ATPase activity and, accordingly, forms a highly stable presynaptic filament. Surprisingly, unlike RAD51, presynaptic filament stabilization achieved via ATP hydrolysis attenuation does not lead to any enhancement of DMC1-catalyzed homologous DNA pairing and strand exchange. This conclusion is further supported by examining wild-type DMC1 with non-hydrolyzable ATP analogues. Thus, our results reveal an important mechanistic difference between RAD51 and DMC1.
Collapse
Affiliation(s)
- Hao-Yen Chang
- From the Institute of Biochemical Sciences, National Taiwan University, Number 1, Section 4, Roosevelt Road, Taipei 10617 Taiwan
| | - Chia-Yu Liao
- From the Institute of Biochemical Sciences, National Taiwan University, Number 1, Section 4, Roosevelt Road, Taipei 10617 Taiwan
| | - Guan-Chin Su
- From the Institute of Biochemical Sciences, National Taiwan University, Number 1, Section 4, Roosevelt Road, Taipei 10617 Taiwan
| | - Sheng-Wei Lin
- the Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan, and
| | - Hong-Wei Wang
- the Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Peter Chi
- From the Institute of Biochemical Sciences, National Taiwan University, Number 1, Section 4, Roosevelt Road, Taipei 10617 Taiwan, the Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan, and
| |
Collapse
|
123
|
Liu NA, Sun J, Kono K, Horikoshi Y, Ikura T, Tong X, Haraguchi T, Tashiro S. Regulation of homologous recombinational repair by lamin B1 in radiation-induced DNA damage. FASEB J 2015; 29:2514-25. [PMID: 25733566 DOI: 10.1096/fj.14-265546] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/13/2015] [Indexed: 01/05/2023]
Abstract
DNA double-strand breaks (DSBs) are the major lethal lesion induced by ionizing radiation (IR). RAD51-dependent homologous recombination (HR) is one of the most important pathways in DSB repair and genome integrity maintenance. However, the mechanism of HR regulation by RAD51 remains unclear. To understand the mechanism of RAD51-dependent HR, we searched for interacting partners of RAD51 by a proteomics analysis and identified lamin B1 in human cells. Lamins are nuclear lamina proteins that play important roles in the structural organization of the nucleus and the regulation of chromosome functions. Immunoblotting analyses revealed that siRNA-mediated lamin B1 depletion repressed the DNA damage-dependent increase of RAD51 after IR. The repression was abolished by the proteasome inhibitor MG132, suggesting that lamin B1 stabilizes RAD51 by preventing proteasome-mediated degradation in cells with IR-induced DNA damage. We also showed that lamin B1 depletion repressed RAD51 focus formation and decreased the survival rates after IR. On the basis of these results, we propose that lamin B1 promotes DSB repair and cell survival by maintaining the RAD51 protein levels for HR upon DSB induction after IR.
Collapse
Affiliation(s)
- Ning-Ang Liu
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Jiying Sun
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Kazuteru Kono
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Yasunori Horikoshi
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Tsuyoshi Ikura
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Xing Tong
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Tokuko Haraguchi
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Satoshi Tashiro
- *Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, and Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, Hiroshima, Japan; Department of Mutagenesis, Laboratory of Chromatin Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan; Laboratory Center, Medical College of Soochow University, Suzhou, China; and Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| |
Collapse
|
124
|
Prakash R, Zhang Y, Feng W, Jasin M. Homologous recombination and human health: the roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harb Perspect Biol 2015; 7:a016600. [PMID: 25833843 DOI: 10.1101/cshperspect.a016600] [Citation(s) in RCA: 608] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Homologous recombination (HR) is a major pathway for the repair of DNA double-strand breaks in mammalian cells, the defining step of which is homologous strand exchange directed by the RAD51 protein. The physiological importance of HR is underscored by the observation of genomic instability in HR-deficient cells and, importantly, the association of cancer predisposition and developmental defects with mutations in HR genes. The tumor suppressors BRCA1 and BRCA2, key players at different stages of HR, are frequently mutated in familial breast and ovarian cancers. Other HR proteins, including PALB2 and RAD51 paralogs, have also been identified as tumor suppressors. This review summarizes recent findings on BRCA1, BRCA2, and associated proteins involved in human disease with an emphasis on their molecular roles and interactions.
Collapse
Affiliation(s)
- Rohit Prakash
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Yu Zhang
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Weiran Feng
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065 Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065 Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
125
|
Mason JM, Dusad K, Wright WD, Grubb J, Budke B, Heyer WD, Connell PP, Weichselbaum RR, Bishop DK. RAD54 family translocases counter genotoxic effects of RAD51 in human tumor cells. Nucleic Acids Res 2015; 43:3180-96. [PMID: 25765654 PMCID: PMC4381078 DOI: 10.1093/nar/gkv175] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/20/2015] [Indexed: 12/14/2022] Open
Abstract
The RAD54 family DNA translocases have several biochemical activities. One activity, demonstrated previously for the budding yeast translocases, is ATPase-dependent disruption of RAD51-dsDNA binding. This activity is thought to promote dissociation of RAD51 from heteroduplex DNA following strand exchange during homologous recombination. In addition, previous experiments in budding yeast have shown that the same activity of Rad54 removes Rad51 from undamaged sites on chromosomes; mutants lacking Rad54 accumulate nonrepair-associated complexes that can block growth and lead to chromosome loss. Here, we show that human RAD54 also promotes the dissociation of RAD51 from dsDNA and not ssDNA. We also show that translocase depletion in tumor cell lines leads to the accumulation of RAD51 on chromosomes, forming complexes that are not associated with markers of DNA damage. We further show that combined depletion of RAD54L and RAD54B and/or artificial induction of RAD51 overexpression blocks replication and promotes chromosome segregation defects. These results support a model in which RAD54L and RAD54B counteract genome-destabilizing effects of direct binding of RAD51 to dsDNA in human tumor cells. Thus, in addition to having genome-stabilizing DNA repair activity, human RAD51 has genome-destabilizing activity when expressed at high levels, as is the case in many human tumors.
Collapse
Affiliation(s)
- Jennifer M Mason
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA
| | - Kritika Dusad
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA
| | - William Douglass Wright
- Department of Molecular and Cellular Biology, University of California, Davis, Davis CA 95616, USA
| | - Jennifer Grubb
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA
| | - Brian Budke
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA
| | - Wolf-Dietrich Heyer
- Department of Molecular and Cellular Biology, University of California, Davis, Davis CA 95616, USA Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Philip P Connell
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA
| | - Douglas K Bishop
- Department of Radiation and Cellular Oncology, University of Chicago, Cummings Life Science Center, Box 13, 920 East 58th St., Chicago, IL 60637, USA Department of Microbiology and Molecular Genetics, University of California, Davis, Davis CA 95616, USA
| |
Collapse
|
126
|
Genois MM, Plourde M, Éthier C, Roy G, Poirier GG, Ouellette M, Masson JY. Roles of Rad51 paralogs for promoting homologous recombination in Leishmania infantum. Nucleic Acids Res 2015; 43:2701-15. [PMID: 25712090 PMCID: PMC4357719 DOI: 10.1093/nar/gkv118] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 12/28/2022] Open
Abstract
To achieve drug resistance Leishmania parasite alters gene copy number by using its repeated sequences widely distributed through the genome. Even though homologous recombination (HR) is ascribed to maintain genome stability, this eukaryote exploits this potent mechanism driven by the Rad51 recombinase to form beneficial extrachromosomal circular amplicons. Here, we provide insights on the formation of these circular amplicons by analyzing the functions of the Rad51 paralogs. We purified three Leishmania infantum Rad51 paralogs homologs (LiRad51-3, LiRad51-4 and LiRad51-6) all of which directly interact with LiRad51. LiRad51-3, LiRad51-4 and LiRad51-6 show differences in DNA binding and annealing capacities. Moreover, it is also noteworthy that LiRad51-3 and LiRad51-4 are able to stimulate Rad51-mediated D-loop formation. In addition, we succeed to inactivate the LiRad51-4 gene and report a decrease of circular amplicons in this mutant. The LiRad51-3 gene was found to be essential for cell viability. Thus, we propose that the LiRad51 paralogs play crucial functions in extrachromosomal circular DNA amplification to circumvent drug actions and preserve survival.
Collapse
Affiliation(s)
- Marie-Michelle Genois
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Québec City, QC G1V 0A6, Canada Centre de Recherche en Infectiologie, CHUL, 2705 boul. Laurier, Quebec, Quebec G1V 4G2, Canada
| | - Marie Plourde
- Centre de Recherche en Infectiologie, CHUL, 2705 boul. Laurier, Quebec, Quebec G1V 4G2, Canada
| | - Chantal Éthier
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Québec City, QC G1V 0A6, Canada CHU de Québec Research Center, CHUL Pavilion, Oncology Axis, 2705 boul. Laurier, Quebec city, Quebec, G1V 4G2, Canada
| | - Gaétan Roy
- Centre de Recherche en Infectiologie, CHUL, 2705 boul. Laurier, Quebec, Quebec G1V 4G2, Canada
| | - Guy G Poirier
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Québec City, QC G1V 0A6, Canada CHU de Québec Research Center, CHUL Pavilion, Oncology Axis, 2705 boul. Laurier, Quebec city, Quebec, G1V 4G2, Canada
| | - Marc Ouellette
- Centre de Recherche en Infectiologie, CHUL, 2705 boul. Laurier, Quebec, Quebec G1V 4G2, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
127
|
Changes in the expression of DNA double strand break repair genes in primordial follicles from immature and aged rats. Reprod Biomed Online 2015; 30:303-10. [DOI: 10.1016/j.rbmo.2014.11.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 11/19/2022]
|
128
|
Krivokuca AM, Malisic EJ, Dobricic JD, Brotto KV, Cavic MR, Jankovic RN, Tomasevic ZI, Brankovic-Magic MV. RAD51 135G>C and TP53 Arg72Pro polymorphisms and susceptibility to breast cancer in Serbian women. Fam Cancer 2015; 13:173-80. [PMID: 24114315 DOI: 10.1007/s10689-013-9690-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is a complex disease with both genetic and environmental factors involved in its etiology. An important role of polymorphisms in genes involved in DNA repair has been reported related to breast cancer risk. We conducted a case-control study in order to investigate the association of RAD51 135G>C and TP53 Arg72Pro polymorphisms with breast cancer in Serbian women.48 BRCA negative women with breast cancer and family history of breast/ovarian cancer (hereditary group), 107 women with breast cancer but without family history of the disease (sporadic group) and 114 healthy women without a history of the disease (control group) were included. Restriction fragment length polymorphism was used for genotyping. Genotype and allelic frequencies, the odds ratio (OR) and the 95 % confidence interval (CI) were calculated as an estimate of relative risk. The Hardy-Weinberg equilibrium was tested using χ(2) test. Significance was considered for p < 0.05. RAD51 135G>C showed statistically significant association of CC genotype and increased breast cancer risk (OR 10.28, 95 % CI 1.12-94.5) in hereditary group of patients compared to the control group. Regarding the TP53 Arg72Pro, we showed statistical significance for ProPro + ProArg comparing to ArgArg (OR 2.34, 95 %, CI 1.17-4.70) in hereditary compared to sporadic group. RAD51 135G>C contributes to hereditary breast cancer in Serbian population, with CC genotype as a risk factor. We also found that carriers of Pro allele of TP53 codon 72 is related to hereditary cancer comparing to sporadic one, which indicates it as a potential risk factor for hereditary form of disease.
Collapse
Affiliation(s)
- Ana M Krivokuca
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia,
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Stasiak A. Presence control of DNA repair controllers. Cell Cycle 2015; 14:2199. [PMID: 26039553 PMCID: PMC5242304 DOI: 10.1080/15384101.2015.1056618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Andrzej Stasiak
- Center for Integrative Genomics; University of Lausanne & SIB Swiss Institute of Bioinformatics; Lausanne, Switzerland
| |
Collapse
|
130
|
Lodhia KA, Gao S, Aleksic T, Esashi F, Macaulay VM. Suppression of homologous recombination sensitizes human tumor cells to IGF-1R inhibition. Int J Cancer 2014; 136:2961-6. [PMID: 25388513 DOI: 10.1002/ijc.29327] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/27/2014] [Indexed: 01/10/2023]
Abstract
Inhibition of type 1 IGF receptor (IGF-1R) sensitizes to DNA-damaging cancer treatments, and delays repair of DNA double strand breaks (DSBs) by non-homologous end-joining and homologous recombination (HR). In a recent screen for mediators of resistance to IGF-1R inhibitor AZ12253801, we identified RAD51, required for the strand invasion step of HR. These findings prompted us to test the hypothesis that IGF-1R-inhibited cells accumulate DSBs formed at endogenous DNA lesions, and depend on residual HR for their repair. Indeed, initial experiments showed time-dependent accumulation of γH2AX foci in IGF-1R -inhibited or -depleted prostate cancer cells. We then tested effects of suppressing HR, and found that RAD51 depletion enhanced AZ12253801 sensitivity in PTEN wild-type prostate cancer cells but not in cells lacking functional PTEN. Similar sensitization was induced in prostate cancer cells by depletion of BRCA2, required for RAD51 loading onto DNA, and in BRCA2(-/-) colorectal cancer cells, compared with isogenic BRCA2(+/-) cells. We also assessed chemical HR inhibitors, finding that RAD51 inhibitor BO2 blocked RAD51 focus formation and sensitized to AZ12253801. Finally, we tested CDK1 inhibitor RO-3306, which impairs HR by inhibiting CDK1-mediated BRCA1 phosphorylation. R0-3306 suppressed RAD51 focus formation consistent with HR attenuation, and sensitized prostate cancer cells to IGF-1R inhibition, with 2.4-fold reduction in AZ12253801 GI50 and 13-fold reduction in GI80. These data suggest that responses to IGF-1R inhibition are enhanced by genetic and chemical approaches to suppress HR, defining a population of cancers (PTEN wild-type, BRCA mutant) that may be intrinsically sensitive to IGF-1R inhibitory drugs.
Collapse
Affiliation(s)
- Kunal A Lodhia
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, United Kingdom
| | | | | | | | | |
Collapse
|
131
|
Uringa EJ, Baldeyron C, Odijk H, Wassenaar E, van Cappellen WA, Maas A, Hoeijmakers JHJ, Baarends WM, Kanaar R, Essers J. A mRad51-GFP antimorphic allele affects homologous recombination and DNA damage sensitivity. DNA Repair (Amst) 2014; 25:27-40. [PMID: 25463395 DOI: 10.1016/j.dnarep.2014.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/05/2014] [Accepted: 11/07/2014] [Indexed: 10/24/2022]
Abstract
Accurate DNA double-strand break repair through homologous recombination is essential for preserving genome integrity. Disruption of the gene encoding RAD51, the protein that catalyzes DNA strand exchange during homologous recombination, results in lethality of mammalian cells. Proteins required for homologous recombination, also play an important role during DNA replication. To explore the role of RAD51 in DNA replication and DSB repair, we used a knock-in strategy to express a carboxy-terminal fusion of green fluorescent protein to mouse RAD51 (mRAD51-GFP) in mouse embryonic stem cells. Compared to wild-type cells, heterozygous mRad51(+/wt-GFP) embryonic stem cells showed increased sensitivity to DNA damage induced by ionizing radiation and mitomycin C. Moreover, gene targeting was found to be severely impaired in mRad51(+/wt-GFP) embryonic stem cells. Furthermore, we found that mRAD51-GFP foci were not stably associated with chromatin. From these experiments we conclude that this mRad51-GFP allele is an antimorphic allele. When this allele is present in a heterozygous condition over wild-type mRad51, embryonic stem cells are proficient in DNA replication but display defects in homologous recombination and DNA damage repair.
Collapse
Affiliation(s)
- Evert-Jan Uringa
- Department of Reproduction and Development, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Céline Baldeyron
- Department of Genetics, Cancer Genomics Center, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Hanny Odijk
- Department of Genetics, Cancer Genomics Center, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Evelyne Wassenaar
- Department of Reproduction and Development, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Wiggert A van Cappellen
- Erasmus Optical Imaging Centre, Department of Pathology, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Alex Maas
- Department of Genetics, Cancer Genomics Center, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jan H J Hoeijmakers
- Department of Genetics, Cancer Genomics Center, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Willy M Baarends
- Department of Reproduction and Development, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Roland Kanaar
- Department of Genetics, Cancer Genomics Center, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Department of Radiation Oncology, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jeroen Essers
- Department of Genetics, Cancer Genomics Center, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Department of Radiation Oncology, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Department of Surgical Oncology, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
132
|
Charlot F, Chelysheva L, Kamisugi Y, Vrielynck N, Guyon A, Epert A, Le Guin S, Schaefer DG, Cuming AC, Grelon M, Nogué F. RAD51B plays an essential role during somatic and meiotic recombination in Physcomitrella. Nucleic Acids Res 2014; 42:11965-78. [PMID: 25260587 PMCID: PMC4231755 DOI: 10.1093/nar/gku890] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The eukaryotic RecA homologue Rad51 is a key factor in homologous recombination and recombinational repair. Rad51-like proteins have been identified in yeast (Rad55, Rad57 and Dmc1), plants and vertebrates (RAD51B, RAD51C, RAD51D, XRCC2, XRCC3 and DMC1). RAD51 and DMC1 are the strand-exchange proteins forming a nucleofilament for strand invasion, however, the function of the paralogues in the process of homologous recombination is less clear. In yeast the two Rad51 paralogues, Rad55 and Rad57, have been shown to be involved in somatic and meiotic HR and they are essential to the formation of the Rad51/DNA nucleofilament counterbalancing the anti-recombinase activity of the SRS2 helicase. Here, we examined the role of RAD51B in the model bryophyte Physcomitrella patens. Mutant analysis shows that RAD51B is essential for the maintenance of genome integrity, for resistance to DNA damaging agents and for gene targeting. Furthermore, we set up methods to investigate meiosis in Physcomitrella and we demonstrate that the RAD51B protein is essential for meiotic homologous recombination. Finally, we show that all these functions are independent of the SRS2 anti-recombinase protein, which is in striking contrast to what is found in budding yeast where the RAD51 paralogues are fully dependent on the SRS2 anti-recombinase function.
Collapse
Affiliation(s)
- Florence Charlot
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Liudmila Chelysheva
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Yasuko Kamisugi
- Centre for Plant Sciences, Faculty of Biological Sciences, Leeds University, Leeds LS2 9JT, UK
| | - Nathalie Vrielynck
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Anouchka Guyon
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Aline Epert
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Sylvia Le Guin
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Didier G Schaefer
- Laboratoire de Biologie Moleculaire et Cellulaire, Institut de Biologie, Universite de Neuchatel, rue Emile-Argand 11, CH-2007 Neuchatel, Switzerland
| | - Andrew C Cuming
- Centre for Plant Sciences, Faculty of Biological Sciences, Leeds University, Leeds LS2 9JT, UK
| | - Mathilde Grelon
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| | - Fabien Nogué
- INRA, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France AgroParisTech, Institut Jean-Pierre Bourgin UMR1318, Saclay Plant Sciences, Versailles, France
| |
Collapse
|
133
|
Le Cigne A, Menil-Philippot V, Fleury F, Takahashi M, Thiriet C. Transient expression of RAD51 in the late G2-phase is required for cell cycle progression in synchronous Physarum cells. Genes Cells 2014; 19:755-65. [PMID: 25200281 DOI: 10.1111/gtc.12174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 07/30/2014] [Indexed: 11/27/2022]
Abstract
The homologous recombination factor RAD51 is highly conserved. This criterion enabled us to identify a RAD51 ortholog in Physarum polycephalum. We found that the Physarum protein presents a high homology to the human protein and cross-reacted with antibodies directed against the human RAD51. Taking advantage of the natural synchrony of millions of nuclei within a single cell of Physarum, we investigated the fluctuation of the amount of the PpRAD51 throughout the cell cycle. Our results showed that in the late G2-phase, RAD51 was transiently expressed in a large quantity. Furthermore, knocking-down RAD51 in the G2-phase abolished this transient expression before mitosis and affected cell cycle progression. These results support the idea that RAD51 plays a role in the progression of the cell cycle in the late G2-phase.
Collapse
Affiliation(s)
- Anthony Le Cigne
- Faculté des Sciences et des Techniques, UFIP UMR CNRS 6286 & Université de Nantes, 44322, Nantes Cedex 3, France; Division of Mechanism and Regulation of DNA Repair, Faculté des Sciences et des Techniques, UFIP UMR CNRS 6286 & Université de Nantes, 44322, Nantes Cedex 3, France; Division of Epigenetics: Proliferation and Differentiation, Faculté des Sciences et des Techniques, UFIP UMR CNRS 6286 & Université de Nantes, 44322, Nantes Cedex 3, France
| | | | | | | | | |
Collapse
|
134
|
Roy N, Bhattacharyya S, Chakrabarty S, Laskar S, Babu SM, Bhattacharyya MK. Dominant negative mutant of Plasmodium Rad51 causes reduced parasite burden in host by abrogating DNA double-strand break repair. Mol Microbiol 2014; 94:353-66. [PMID: 25145341 DOI: 10.1111/mmi.12762] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2014] [Indexed: 01/05/2023]
Abstract
Malaria parasites survive through repairing a plethora of DNA double-stranded breaks (DSBs) experienced during their asexual growth. In Plasmodium Rad51 mediated homologous recombination (HR) mechanism and homology-independent alternative end-joining mechanism have been identified. Here we address whether loss of HR activity can be compensated by other DSB repair mechanisms. Creating a transgenic Plasmodium line defective in HR function, we demonstrate that HR is the most important DSB repair pathway in malarial parasite. Using mouse malaria model we have characterized the dominant negative effect of PfRad51(K143R) mutant on Plasmodium DSB repair and host-parasite interaction. Our work illustrates that Plasmodium berghei harbouring the mutant protein (PfRad51(K143R)) failed to repair DSBs as evidenced by hypersensitivity to DNA-damaging agent. Mice infected with mutant parasites lived significantly longer with markedly reduced parasite burden. To better understand the effect of mutant PfRad51(K143R) on HR, we used yeast as a surrogate model and established that the presence of PfRad51(K143R) completely inhibited DNA repair, gene conversion and gene targeting. Biochemical experiment confirmed that very low level of mutant protein was sufficient for complete disruption of wild-type PfRad51 activity. Hence our work provides evidence that HR pathway of Plasmodium could be efficiently targeted to curb malaria.
Collapse
Affiliation(s)
- Nabamita Roy
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, Andhra Pradesh, India
| | | | | | | | | | | |
Collapse
|
135
|
Yoon SW, Kim DK, Kim KP, Park KS. Rad51 regulates cell cycle progression by preserving G2/M transition in mouse embryonic stem cells. Stem Cells Dev 2014; 23:2700-11. [PMID: 24991985 DOI: 10.1089/scd.2014.0129] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Homologous recombination (HR) maintains genomic integrity against DNA replication stress and deleterious lesions, such as double-strand breaks (DSBs). Rad51 recombinase is critical for HR events that mediate the exchange of genetic information between parental chromosomes in eukaryotes. Additionally, Rad51 and HR accessory factors may facilitate replication fork progression by preventing replication fork collapse and repair DSBs that spontaneously arise during the normal cell cycle. In this study, we demonstrated a novel role for Rad51 during the cell cycle in mouse embryonic stem cells (mESCs). In mESCs, Rad51 was constitutively expressed throughout the cell cycle, and the formation of Rad51 foci increased as the cells entered S phase. Suppression of Rad51 expression caused cells to accumulate at G2/M phase and activated the DNA damage checkpoint, but it did not affect the self-renewal or differentiation capacity of mESCs. Even though Rad51 suppression significantly inhibited the proliferation rate of mESCs, Rad51 suppression did not affect the replication fork progression and speed, indicating that Rad51 repaired DNA damage and promoted DNA replication in S phase through an independent mechanism. In conclusion, Rad51 may contribute to G2/M transition in mESCs, while preserving genomic integrity in global organization of DNA replication fork.
Collapse
Affiliation(s)
- Sang-Wook Yoon
- 1 Department of Life Science, Chung-Ang University , Seoul, Korea
| | | | | | | |
Collapse
|
136
|
Lim YC, Roberts TL, Day BW, Stringer BW, Kozlov S, Fazry S, Bruce ZC, Ensbey KS, Walker DG, Boyd AW, Lavin MF. Increased sensitivity to ionizing radiation by targeting the homologous recombination pathway in glioma initiating cells. Mol Oncol 2014; 8:1603-15. [PMID: 25017126 DOI: 10.1016/j.molonc.2014.06.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/20/2014] [Accepted: 06/20/2014] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma is deemed the most malignant form of brain tumour, particularly due to its resistance to conventional treatments. A small surviving group of aberrant stem cells termed glioma initiation cells (GICs) that escape surgical debulking are suggested to be the cause of this resistance. Relatively quiescent in nature, GICs are capable of driving tumour recurrence and undergo lineage differentiation. Most importantly, these GICs are resistant to radiotherapy, suggesting that radioresistance contribute to their survival. In a previous study, we demonstrated that GICs had a restricted double strand break (DSB) repair pathway involving predominantly homologous recombination (HR) associated with a lack of functional G1/S checkpoint arrest. This unusual behaviour led to less efficient non-homologous end joining (NHEJ) repair and overall slower DNA DSB repair kinetics. To determine whether specific targeting of the HR pathway with small molecule inhibitors could increase GIC radiosensitivity, we used the Ataxia-telangiectasia mutated inhibitor (ATMi) to ablate HR and the DNA-dependent protein kinase inhibitor (DNA-PKi) to inhibit NHEJ. Pre-treatment with ATMi prior to ionizing radiation (IR) exposure prevented HR-mediated DNA DSB repair as measured by Rad51 foci accumulation. Increased cell death in vitro and improved in vivo animal survival could be observed with combined ATMi and IR treatment. Conversely, DNA-PKi treatment had minimal impact on GICs ability to resolve DNA DSB after IR with only partial reduction in cell survival, confirming the major role of HR. These results provide a mechanistic insight into the predominant form of DNA DSB repair in GICs, which when targeted may be a potential translational approach to increase patient survival.
Collapse
Affiliation(s)
- Yi Chieh Lim
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia; The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Herston, Queensland 4029, Australia
| | - Tara L Roberts
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia; The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Herston, Queensland 4029, Australia
| | - Bryan W Day
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - Brett W Stringer
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - Sergei Kozlov
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - Shazrul Fazry
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - Zara C Bruce
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - Kathleen S Ensbey
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - David G Walker
- BrizBrain and Spine, The Wesley Hospital, Evan Thomson Building, Level 10, Auchenflower, Queensland 4066, Australia
| | - Andrew W Boyd
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia
| | - Martin F Lavin
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4029, Australia; The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Herston, Queensland 4029, Australia.
| |
Collapse
|
137
|
Liang H, Li Y, Luo RY, Shen FJ. An increased risk of ovarian cancer associated with polymorphism in BRCC5 gene in Caucasian populations. Tumour Biol 2014; 35:9179-84. [DOI: 10.1007/s13277-014-2135-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/21/2014] [Indexed: 11/29/2022] Open
|
138
|
Nucleases in homologous recombination as targets for cancer therapy. FEBS Lett 2014; 588:2446-56. [PMID: 24928444 DOI: 10.1016/j.febslet.2014.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/02/2014] [Accepted: 06/02/2014] [Indexed: 11/21/2022]
|
139
|
Shah MM, Dobbin ZC, Nowsheen S, Wielgos M, Katre AA, Alvarez RD, Konstantinopoulos PA, Yang ES, Landen CN. An ex vivo assay of XRT-induced Rad51 foci formation predicts response to PARP-inhibition in ovarian cancer. Gynecol Oncol 2014; 134:331-7. [PMID: 24844596 DOI: 10.1016/j.ygyno.2014.05.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/09/2014] [Accepted: 05/11/2014] [Indexed: 12/16/2022]
Abstract
OBJECTIVE BRCA-positive ovarian cancer patients derive benefit PARP inhibitors. Approximately 50% of ovarian cancer tumors have homologous recombination (HR) deficiencies and are therefore "BRCA-like," possibly rendering them sensitive to PARP inhibition. However, no predictive assay exists to identify these patients. We sought to determine if irradiation-induced Rad51 foci formation, a known marker of HR, correlated to PARP inhibitor response in an ovarian cancer model. METHODS Ovarian cancer cell lines were exposed to PARP-inhibitor ABT-888 to determine effect on growth. Rad51 protein expression prior to irradiation was determined via Western blot. Cultured cells and patient-derived xenograft tumors (PDX) were irradiated and probed for Rad51 foci. In vivo PDX tumors were treated with ABT-888 and carboplatin; these results were correlated with the ex vivo ionizing radiation assay. RESULTS Three of seven cell lines were sensitive to ABT-888. Sensitive lines had the lowest Rad51 foci formation rate after irradiation, indicating functional HR deficiency. Approximately 50% of the PDX samples had decreased Rad51 foci formation. Total Rad51 protein levels were consistently low, suggesting that DNA damage induction is required to characterize HR status. The ex vivo IR assay accurately predicted which PDX models were sensitive to PARP inhibition in vitro and in vivo. ABT-888 alone reduced orthotopic tumor growth by 51% in A2780ip2 cell line, predicted to respond by the ex vivo assay. Three PDX models' response also correlated with the assay. CONCLUSIONS The ex vivo IR assay correlates with response to PARP inhibition. Analysis of total Rad51 protein is not a reliable substitute.
Collapse
Affiliation(s)
- Monjri M Shah
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zachary C Dobbin
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Somaira Nowsheen
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Monica Wielgos
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashwini A Katre
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ronald D Alvarez
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles N Landen
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
140
|
Chan YL, Brown MS, Qin D, Handa N, Bishop DK. The third exon of the budding yeast meiotic recombination gene HOP2 is required for calcium-dependent and recombinase Dmc1-specific stimulation of homologous strand assimilation. J Biol Chem 2014; 289:18076-86. [PMID: 24798326 DOI: 10.1074/jbc.m114.558601] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During meiosis in Saccharomyces cerevisiae, the HOP2 and MND1 genes are essential for recombination. A previous biochemical study has shown that budding yeast Hop2-Mnd1 stimulates the activity of the meiosis-specific strand exchange protein ScDmc1 only 3-fold, whereas analogous studies using mammalian homologs show >30-fold stimulation. The HOP2 gene was recently discovered to contain a second intron that lies near the 3'-end. We show that both HOP2 introns are efficiently spliced during meiosis, forming a predominant transcript that codes for a protein with a C-terminal sequence different from that of the previously studied version of the protein. Using the newly identified HOP2 open reading frame to direct synthesis of wild type Hop2 protein, we show that the Hop2-Mnd1 heterodimer stimulated Dmc1 D-loop activity up to 30-fold, similar to the activity of mammalian Hop2-Mnd1. ScHop2-Mnd1 stimulated ScDmc1 activity in the presence of physiological (micromolar) concentrations of Ca(2+) ions, as long as Mg(2+) was also present at physiological concentrations, leading us to hypothesize that ScDmc1 protomers bind both cations in the active Dmc1 filament. Co-factor requirements and order-of-addition experiments suggested that Hop2-Mnd1-mediated stimulation of Dmc1 involves a process that follows the formation of functional Dmc1-ssDNA filaments. In dramatic contrast to mammalian orthologs, the stimulatory activity of budding yeast Hop2-Mnd1 appeared to be specific to Dmc1; we observed no Hop2-Mnd1-mediated stimulation of the other budding yeast strand exchange protein Rad51. Together, these results support previous genetic experiments indicating that Hop2-Mnd1 specifically stimulates Dmc1 during meiotic recombination in budding yeast.
Collapse
Affiliation(s)
- Yuen-Ling Chan
- From the Departments of Radiation and Cellular Oncology and
| | - M Scott Brown
- From the Departments of Radiation and Cellular Oncology and
| | - Daoming Qin
- Molecular Genetics and Cell Biology, Cummings Life Science Center, University of Chicago, Chicago, Illinois 60637 and
| | - Naofumi Handa
- the Department of Microbiology and Molecular Genetics, University of California, Davis, California 95616
| | - Douglas K Bishop
- From the Departments of Radiation and Cellular Oncology and Molecular Genetics and Cell Biology, Cummings Life Science Center, University of Chicago, Chicago, Illinois 60637 and
| |
Collapse
|
141
|
Chinone A, Matsumoto M. DrRad51 is required for chiasmata formation in meiosis in planarian Dugesia ryukyuensis. Mol Reprod Dev 2014; 81:409-21. [PMID: 24488935 DOI: 10.1002/mrd.22308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/28/2014] [Indexed: 01/01/2023]
Abstract
Rad51, a conserved eukaryotic protein, mediates the homologous-recombination repair of DNA double-strand breaks that occur during both mitosis and meiosis. During prophase I of meiosis, homologous recombination enhances the linkage between homologous chromosomes to increase the accuracy of segregation at anaphase I. In polyploidy situations, however, difficulties with homologous chromosome segregation often disrupt meiosis. Yet, triploid individuals of the planarian Dugesia ryukyuensis are able to produce functional gametes through a specialized form of meiosis. To shed light on the molecular mechanisms that promote successful meiosis in triploid D. ryukyuensis, we investigated rad51 gene function. We isolated three genes of the Rad51 family, the Rad51 homolog Dr-rad51 and the Rad51 paralogs Dr-rad51B and Dr-rad51C. Dr-rad51 was expressed in germ-line and presumably in somatic stem cells, but was not necessary for the regeneration of somatic tissue. RNA-interference (RNAi) depletion of Dr-rad51 during sexualization did not affect chromosome behavior in zygotene oocytes, but did result in the loss of chiasmata at the diplotene stage. Thus, homologous recombination does not appear to be necessary for synapsis, but is needed for crossover and proper segregation in D. ryukyuensis.
Collapse
Affiliation(s)
- Ayako Chinone
- Department of Biosciences and Informatics, Keio University, Yokohama, Japan
| | | |
Collapse
|
142
|
Jekimovs C, Bolderson E, Suraweera A, Adams M, O’Byrne KJ, Richard DJ. Chemotherapeutic compounds targeting the DNA double-strand break repair pathways: the good, the bad, and the promising. Front Oncol 2014; 4:86. [PMID: 24795863 PMCID: PMC4001069 DOI: 10.3389/fonc.2014.00086] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/08/2014] [Indexed: 01/09/2023] Open
Abstract
The repair of DNA double-strand breaks (DSBs) is a critical cellular mechanism that exists to ensure genomic stability. DNA DSBs are the most deleterious type of insult to a cell's genetic material and can lead to genomic instability, apoptosis, or senescence. Incorrectly repaired DNA DSBs have the potential to produce chromosomal translocations and genomic instability, potentially leading to cancer. The prevalence of DNA DSBs in cancer due to unregulated growth and errors in repair opens up a potential therapeutic window in the treatment of cancers. The cellular response to DNA DSBs is comprised of two pathways to ensure DNA breaks are repaired: homologous recombination and non-homologous end joining. Identifying chemotherapeutic compounds targeting proteins involved in these DNA repair pathways has shown promise as a cancer therapy for patients, either as a monotherapy or in combination with genotoxic drugs. From the beginning, there have been a number of chemotherapeutic compounds that have yielded successful responses in the clinic, a number that have failed (CGK-733 and iniparib), and a number of promising targets for future studies identified. This review looks in detail at how the cell responds to these DNA DSBs and investigates the chemotherapeutic avenues that have been and are currently being explored to target this repair process.
Collapse
Affiliation(s)
- Christian Jekimovs
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Emma Bolderson
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Amila Suraweera
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Mark Adams
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kenneth J. O’Byrne
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Derek J. Richard
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
143
|
Gasparini P, Lovat F, Fassan M, Casadei L, Cascione L, Jacob NK, Carasi S, Palmieri D, Costinean S, Shapiro CL, Huebner K, Croce CM. Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradiation. Proc Natl Acad Sci U S A 2014; 111:4536-4541. [PMID: 24616504 PMCID: PMC3970505 DOI: 10.1073/pnas.1402604111] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cell survival after DNA damage relies on DNA repair, the abrogation of which causes genomic instability and development of cancer. However, defective DNA repair in cancer cells can be exploited for cancer therapy using DNA-damaging agents. DNA double-strand breaks are the major lethal lesions induced by ionizing radiation (IR) and can be efficiently repaired by DNA homologous recombination, a system that requires numerous factors including the recombinase RAD51 (RAD51). Therapies combined with adjuvant radiotherapy have been demonstrated to improve the survival of triple-negative breast cancer patients; however, such therapy is challenged by the emergence of resistance in tumor cells. It is, therefore, essential to develop novel therapeutic strategies to overcome radioresistance and improve radiosensitivity. In this study we show that overexpression of microRNA 155 (miR-155) in human breast cancer cells reduces the levels of RAD51 and affects the cellular response to IR. miR-155 directly targets the 3'-untranslated region of RAD51. Overexpression of miR-155 decreased the efficiency of homologous recombination repair and enhanced sensitivity to IR in vitro and in vivo. High miR-155 levels were associated with lower RAD51 expression and with better overall survival of patients in a large series of triple-negative breast cancers. Taken together, our findings indicate that miR-155 regulates DNA repair activity and sensitivity to IR by repressing RAD51 in breast cancer. Testing for expression levels of miR-155 may be useful in the identification of breast cancer patients who will benefit from an IR-based therapeutic approach.
Collapse
Affiliation(s)
- Pierluigi Gasparini
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Francesca Lovat
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Matteo Fassan
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
- Applied Research on Cancer Network (ARC-NET) Research Centre, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Lucia Casadei
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Luciano Cascione
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
- Lymphoma and Genomics Research Program, Institute of Oncology Research, Bellinzona 6500, Switzerland
| | - Naduparambil K. Jacob
- Department of Radiation Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Stefania Carasi
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Dario Palmieri
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Stefan Costinean
- Department of Pathology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; and
| | - Charles L. Shapiro
- Division of Medical Oncology and the Breast Program, James Cancer Hospital and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Wexner Medical Center and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
144
|
Koike-Yusa H, Li Y, Tan EP, Velasco-Herrera MDC, Yusa K. Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nat Biotechnol 2014; 32:267-73. [PMID: 24535568 DOI: 10.1038/nbt.2800] [Citation(s) in RCA: 796] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/17/2013] [Indexed: 12/23/2022]
Abstract
Identification of genes influencing a phenotype of interest is frequently achieved through genetic screening by RNA interference (RNAi) or knockouts. However, RNAi may only achieve partial depletion of gene activity, and knockout-based screens are difficult in diploid mammalian cells. Here we took advantage of the efficiency and high throughput of genome editing based on type II, clustered, regularly interspaced, short palindromic repeats (CRISPR)-CRISPR-associated (Cas) systems to introduce genome-wide targeted mutations in mouse embryonic stem cells (ESCs). We designed 87,897 guide RNAs (gRNAs) targeting 19,150 mouse protein-coding genes and used a lentiviral vector to express these gRNAs in ESCs that constitutively express Cas9. Screening the resulting ESC mutant libraries for resistance to either Clostridium septicum alpha-toxin or 6-thioguanine identified 27 known and 4 previously unknown genes implicated in these phenotypes. Our results demonstrate the potential for efficient loss-of-function screening using the CRISPR-Cas9 system.
Collapse
Affiliation(s)
| | - Yilong Li
- 1] Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK. [2]
| | - E-Pien Tan
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | | | - Kosuke Yusa
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| |
Collapse
|
145
|
Zhang SX, Yang S, Xu CQ, Hou RP, Zhang CZ, Xu CP. Equivocal association of RAD51 polymorphisms with risk of esophageal squamous cell carcinoma in a Chinese population. Asian Pac J Cancer Prev 2014; 15:763-7. [PMID: 24568492 DOI: 10.7314/apjcp.2014.15.2.763] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AIM To study the contribution of genetic variation in RAD51 to risk of esophageal squamous cell carcinoma (ESCC). METHODS Three single nucleotide polymorphisms (SNPs) in RAD51 (rs1801320, rs4144242 and rs4417527) were genotyped in 316 ESCC patients and 316 healthy controls in Anyang area of China using PCR- RFLP (polymerase chain reaction-restriction fragment length polymorphism). Demographic variables between cases and controls were statistically compared by T test and Chi-square test. Hardy-Weinberg equilibrium was evaluated by the Chi-square test. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to measure any association with ESCC. Haplotype frequencies were estimated by Phase 2.1. RESULT The genotype frequencies of rs1801320, rs4144242 and rs4417527 in patients with ESCC demonstrated no significant differences from those in control group (P>0.05). When the haplotypes of these three SNPs were constructed and their relationships with ESCC risk investigated, however, CGG was observed to increase the risk (P=0.020, OR=2. 289). CONCLUSIONS There was no association between the three SNPs of RAD51 and ESCC susceptibility in our Chinese population. However, the CGG haplotype might be a risk factor.
Collapse
Affiliation(s)
- Shu-Xiang Zhang
- Nursing Department, Shandong Provincial Qianfoshan Hospital, Jinan, China E-mail :
| | | | | | | | | | | |
Collapse
|
146
|
Carvalho JFS, Kanaar R. Targeting homologous recombination-mediated DNA repair in cancer. Expert Opin Ther Targets 2014; 18:427-58. [PMID: 24491188 DOI: 10.1517/14728222.2014.882900] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION DNA is the target of many traditional non-specific chemotherapeutic drugs. New drugs or therapeutic approaches with a more rational and targeted component are mandatory to improve the success of cancer therapy. The homologous recombination (HR) pathway is an attractive target for the development of inhibitors because cancer cells rely heavily on HR for repair of DNA double-strand breaks resulting from chemotherapeutic treatments. Additionally, the discovery that poly(ADP)ribose polymerase-1 inhibitors selectively kill cells with genetic defects in HR has spurned an even greater interest in inhibitors of HR. AREAS COVERED HR drives the repair of broken DNA via numerous protein-mediated sequential DNA manipulations. Due to extensive number of steps and proteins involved, the HR pathway provides a rich pool of potential drug targets. This review discusses the latest developments concerning the strategies being explored to inhibit HR. Particular attention is given to the identification of small molecule inhibitors of key HR proteins, including the BRCA proteins and RAD51. EXPERT OPINION Current HR inhibitors are providing the basis for pharmaceutical development of more potent and specific inhibitors to be applied in mono- or combinatorial therapy regimes, while novel targets will be uncovered by experiments aimed to gain a deeper mechanistic understanding of HR and its subpathways.
Collapse
Affiliation(s)
- João F S Carvalho
- Erasmus MC Cancer Institute, Department of Genetics, Department of Radiation Oncology, Cancer Genomics Netherlands , PO Box 2040, 3000 CA Rotterdam , The Netherlands
| | | |
Collapse
|
147
|
Wang Y, Xiao R, Wang H, Cheng Z, Li W, Zhu G, Wang Y, Ma H. The Arabidopsis RAD51 paralogs RAD51B, RAD51D and XRCC2 play partially redundant roles in somatic DNA repair and gene regulation. THE NEW PHYTOLOGIST 2014; 201:292-304. [PMID: 24102485 DOI: 10.1111/nph.12498] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/14/2013] [Indexed: 05/12/2023]
Abstract
The eukaryotic RAD51 gene family has seven ancient paralogs conserved between plants and animals. Among these, RAD51, DMC1, RAD51C and XRCC3 are important for homologous recombination and/or DNA repair, whereas single mutants in RAD51B, RAD51D or XRCC2 show normal meiosis, and the lineages they represent diverged from each other evolutionarily later than the other four paralogs, suggesting possible functional redundancy. The function of Arabidopsis RAD51B, RAD51D and XRCC2 genes in mitotic DNA repair and meiosis was analyzed using molecular genetic, cytological and transcriptomic approaches. The relevant double and triple mutants displayed normal vegetative and reproductive growth. However, the triple mutant showed greater sensitivity than single or double mutants to DNA damage by bleomycin. RNA-Seq transcriptome analysis supported the idea that the triple mutant showed DNA damage similar to that caused by bleomycin. On bleomycin treatment, many genes were altered in the wild-type but not in the triple mutant, suggesting that the RAD51 paralogs have roles in the regulation of gene transcription, providing an explanation for the hypersensitive phenotype of the triple mutant to bleomycin. Our results provide strong evidence that Arabidopsis XRCC2, RAD51B and RAD51D have complex functions in somatic DNA repair and gene regulation, arguing for further studies of these ancient genes that have been maintained in both plants and animals during their long evolutionary history.
Collapse
Affiliation(s)
- Yingxiang Wang
- State Key Laboratory of Genetic Engineering and Institute of Genetics, Institute of Plant Biology, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Rong Xiao
- Department of Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Haifeng Wang
- State Key Laboratory of Genetic Engineering and Institute of Genetics, Institute of Plant Biology, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200433, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Zhihao Cheng
- State Key Laboratory of Genetic Engineering and Institute of Genetics, Institute of Plant Biology, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Wuxing Li
- Department of Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Genfeng Zhu
- State Key Laboratory of Genetic Engineering and Institute of Genetics, Institute of Plant Biology, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Ying Wang
- State Key Laboratory of Genetic Engineering and Institute of Genetics, Institute of Plant Biology, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Hong Ma
- State Key Laboratory of Genetic Engineering and Institute of Genetics, Institute of Plant Biology, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200433, China
- Department of Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| |
Collapse
|
148
|
Fornander LH, Renodon-Cornière A, Kuwabara N, Ito K, Tsutsui Y, Shimizu T, Iwasaki H, Nordén B, Takahashi M. Swi5-Sfr1 protein stimulates Rad51-mediated DNA strand exchange reaction through organization of DNA bases in the presynaptic filament. Nucleic Acids Res 2013; 42:2358-65. [PMID: 24304898 PMCID: PMC3936755 DOI: 10.1093/nar/gkt1257] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Swi5-Sfr1 heterodimer protein stimulates the Rad51-promoted DNA strand exchange reaction, a crucial step in homologous recombination. To clarify how this accessory protein acts on the strand exchange reaction, we have analyzed how the structure of the primary reaction intermediate, the Rad51/single-stranded DNA (ssDNA) complex filament formed in the presence of ATP, is affected by Swi5-Sfr1. Using flow linear dichroism spectroscopy, we observe that the nucleobases of the ssDNA are more perpendicularly aligned to the filament axis in the presence of Swi5-Sfr1, whereas the bases are more randomly oriented in the absence of Swi5-Sfr1. When using a modified version of the natural protein where the N-terminal part of Sfr1 is deleted, which has no affinity for DNA but maintained ability to stimulate the strand exchange reaction, we still observe the improved perpendicular DNA base orientation. This indicates that Swi5-Sfr1 exerts its activating effect through interaction with the Rad51 filament mainly and not with the DNA. We propose that the role of a coplanar alignment of nucleobases induced by Swi5-Sfr1 in the presynaptic Rad51/ssDNA complex is to facilitate the critical matching with an invading double-stranded DNA, hence stimulating the strand exchange reaction.
Collapse
Affiliation(s)
- Louise H Fornander
- Department of Chemical and Biological Engineering, Chalmers University of Technology, S-41296 Gothenburg, Sweden, Research Unit FRE3478, Centre National de la Recherche Scientifique & University of Nantes, F-44322 Nantes cedex 3, France, Graduate School of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan, Structural Biology Research Center, Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization, KEK, Tsukuba, 305-0801, Japan and Department of Life Science, Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Lin T, Ibrahim W, Peng CY, Finegold MJ, Tsai RY. A novel role of nucleostemin in maintaining the genome integrity of dividing hepatocytes during mouse liver development and regeneration. Hepatology 2013; 58:2176-87. [PMID: 23813570 PMCID: PMC3844114 DOI: 10.1002/hep.26600] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 06/17/2013] [Indexed: 01/04/2023]
Abstract
UNLABELLED During liver development and regeneration, hepatocytes undergo rapid cell division and face an increased risk of DNA damage associated with active DNA replication. The mechanism that protects proliferating hepatocytes from replication-induced DNA damage remains unclear. Nucleostemin (NS) is known to be up-regulated during liver regeneration, and loss of NS is associated with increased DNA damage in cancer cells. To determine whether NS is involved in protecting the genome integrity of proliferating hepatocytes, we created an albumin promoter-driven NS conditional-null (albNS(cko) ) mouse model. Livers of albNS(cko) mice begin to show loss of NS in developing hepatocytes from the first postnatal week and increased DNA damage and hepatocellular injury at 1-2 weeks of age. At 3-4 weeks, albNS(cko) livers develop bile duct hyperplasia and show increased apoptotic cells, necrosis, regenerative nodules, and evidence suggestive of hepatic stem/progenitor cell activation. CCl4 treatment enhances degeneration and DNA damage in NS-deleted hepatocytes and increases biliary hyperplasia and A6(+) cells in albNS(cko) livers. After 70% partial hepatectomy, albNS(cko) livers show increased DNA damage in parallel with a blunted and prolonged regenerative response. The DNA damage in NS-depleted hepatocytes is explained by the impaired recruitment of a core DNA repair enzyme, RAD51, to replication-induced DNA damage foci. CONCLUSION This work reveals a novel genome-protective role of NS in developing and regenerating hepatocytes.
Collapse
Affiliation(s)
- Tao Lin
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030 USA
| | - Wessam Ibrahim
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030 USA
| | - Cheng-Yuan Peng
- School of Medicine, China Medical University, Taichung, 40402, Taiwan, Division of Hepatogastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Milton J Finegold
- Gastrointestinal & Hepatobiliary Pathology, Texas Children's Hospital Houston, Texas 77030 USA, Department of Pathology & Immunology, Baylor College of Medicine, Texas 77030 USA
| | - Robert Y.L. Tsai
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030 USA
| |
Collapse
|
150
|
Magwood AC, Malysewich MJ, Cealic I, Mundia MM, Knapp J, Baker MD. Endogenous levels of Rad51 and Brca2 are required for homologous recombination and regulated by homeostatic re-balancing. DNA Repair (Amst) 2013; 12:1122-33. [PMID: 24210700 DOI: 10.1016/j.dnarep.2013.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 12/17/2022]
Abstract
Stable expression of Rad51 siRNA was used to generate mouse hybridoma cell lines in which endogenous Rad51 levels were depleted by as much as 60%. Stable Rad51 knockdowns feature reduced homologous recombination responses. The relative ease with which stable Rad51 knockdowns were recovered was surprising, given the embryonic lethality of Rad51 ablation. Interestingly, Rad51-depleted hybridoma cell lines are characterized by reduced levels of p53 protein. Completely unexpected, was the finding that Rad51-depleted hybridoma cell lines are also reduced for the breast cancer susceptibility 2 (Brca2) protein. Additionally, hybridoma cell lines that are siRNA depleted for mouse Brca2 show a corresponding reduction in Rad51 and p53 proteins. Furthermore, cellular levels of Rad51, Brca2 and p53 can be elevated in these cell lines by ectopic expression of wild-type human Rad51 and wild-type human BRCA2. In marked contrast, hybridoma cell lines that are siRNA depleted for mouse p53 feature relatively normal Rad51 and Brca2 levels. These results suggest that cellular levels of Brca2 and Rad51 are mutually dependent on each other, and that low levels of these proteins provide selective pressure for reduction of p53, which permits cell growth.
Collapse
Affiliation(s)
- Alissa C Magwood
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | | | | | | | | | | |
Collapse
|