51
|
Tammaro M, Liao S, Beeharry N, Yan H. DNA double-strand breaks with 5' adducts are efficiently channeled to the DNA2-mediated resection pathway. Nucleic Acids Res 2015; 44:221-31. [PMID: 26420828 PMCID: PMC4705695 DOI: 10.1093/nar/gkv969] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/15/2015] [Indexed: 11/14/2022] Open
Abstract
DNA double-strand breaks (DSBs) with 5′ adducts are frequently formed from many nucleic acid processing enzymes, in particular DNA topoisomerase 2 (TOP2). The key intermediate of TOP2 catalysis is the covalent complex (TOP2cc), consisting of two TOP2 subunits covalently linked to the 5′ ends of the nicked DNA. In cells, TOP2ccs can be trapped by cancer drugs such as etoposide and then converted into DNA double-strand breaks (DSBs) that carry adducts at the 5′ end. The repair of such DSBs is critical to the survival of cells, but the underlying mechanism is still not well understood. We found that etoposide-induced DSBs are efficiently resected into 3′ single-stranded DNA in cells and the major nuclease for resection is the DNA2 protein. DNA substrates carrying model 5′ adducts were efficiently resected in Xenopus egg extracts and immunodepletion of Xenopus DNA2 also strongly inhibited resection. These results suggest that DNA2-mediated resection is a major mechanism for the repair of DSBs with 5′ adducts.
Collapse
Affiliation(s)
- Margaret Tammaro
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Shuren Liao
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Neil Beeharry
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Hong Yan
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| |
Collapse
|
52
|
Masunaga SI, Uzawa A, Hirayama R, Matsumoto Y, Sakurai Y, Tanaka H, Tano K, Sanada Y, Suzuki M, Maruhashi A, Ono K. The Effect of p53 Status of Tumor Cells on Radiosensitivity of Irradiated Tumors With Carbon-Ion Beams Compared With γ-Rays or Reactor Neutron Beams. World J Oncol 2015; 6:398-409. [PMID: 28983338 PMCID: PMC5624688 DOI: 10.14740/wjon941w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2015] [Indexed: 11/13/2022] Open
Abstract
Background The aim of the study was to clarify the effect of p53 status of tumor cells on radiosensitivity of solid tumors following accelerated carbon-ion beam irradiation compared with γ-rays or reactor neutron beams, referring to the response of intratumor quiescent (Q) cells. Methods Human head and neck squamous cell carcinoma cells transfected with mutant TP53 (SAS/mp53) or with neo vector (SAS/neo) were injected subcutaneously into hind legs of nude mice. Tumor-bearing mice received 5-bromo-2’-deoxyuridine (BrdU) continuously to label all intratumor proliferating (P) cells. They received γ-rays or accelerated carbon-ion beams at a high or reduced dose-rate. Other tumor-bearing mice received reactor thermal or epithermal neutrons at a reduced dose-rate. Immediately or 9 hours after the high dose-rate irradiation (HDRI), or immediately after the reduced dose-rate irradiation (RDRI), the tumor cells were isolated and incubated with a cytokinesis blocker, and the micronucleus (MN) frequency in cells without BrdU labeling (Q cells) was determined using immunofluorescence staining for BrdU. Results The difference in radiosensitivity between the total (P + Q) and Q cells after γ-ray irradiation was markedly reduced with reactor neutron beams or carbon-ion beams, especially with a higher linear energy transfer (LET) value. Following γ-ray irradiation, SAS/neo tumor cells, especially intratumor Q cells, showed a marked reduction in sensitivity due to the recovery from radiation-induced damage, compared with the total or Q cells within SAS/mp53 tumors that showed little repair capacity. In both total and Q cells within both SAS/neo and SAS/mp53 tumors, carbon-ion beam irradiation, especially with a higher LET, showed little recovery capacity through leaving an interval between HDRI and the assay or decreasing the dose-rate. The recovery from radiation-induced damage after γ-ray irradiation was a p53-dependent event, but little recovery was found after carbon-ion beam irradiation. With RDRI, the radiosensitivity to reactor thermal and epithermal neutron beams was slightly higher than that to carbon-ion beams. Conclusion For tumor control, including intratumor Q-cell control, accelerated carbon-ion beams, especially with a higher LET, and reactor thermal and epithermal neutron beams were very useful for suppressing the recovery from radiation-induced damage irrespective of p53 status of tumor cells.
Collapse
Affiliation(s)
- Shin-Ichiro Masunaga
- Particle Radiation Biology, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Akiko Uzawa
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, 4-9-1, Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ryoichi Hirayama
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, 4-9-1, Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yoshitaka Matsumoto
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Sakurai
- Radiation Medical Physics, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Hiroki Tanaka
- Radiation Medical Physics, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Keizo Tano
- Particle Radiation Biology, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Yu Sanada
- Particle Radiation Biology, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Minoru Suzuki
- Particle Radiation Oncology, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Akira Maruhashi
- Radiation Medical Physics, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Koji Ono
- Particle Radiation Oncology, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| |
Collapse
|
53
|
Bohrer RC, Coutinho ARS, Duggavathi R, Bordignon V. The Incidence of DNA Double-Strand Breaks Is Higher in Late-Cleaving and Less Developmentally Competent Porcine Embryos. Biol Reprod 2015; 93:59. [PMID: 26134870 DOI: 10.1095/biolreprod.115.130542] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 06/30/2015] [Indexed: 12/21/2022] Open
Abstract
Studies in different species, including human, mice, bovine, and swine, demonstrated that early-cleaving embryos have higher capacity to develop to the blastocyst stage and produce better quality embryos with superior capacity to establish pregnancy than late-cleaving embryos. It has also been shown that experimentally induced DNA damage delays embryo cleavage kinetics and reduces blastocyst formation. To gain additional insights into the effects of genome damage on embryo cleavage kinetics and development, the present study compared the occurrence of DNA double-strand breaks (DSBs) with the expression profile of genes involved in DNA repair and cell cycle control between early- and late-cleaving embryos. Porcine oocytes matured in vitro were activated, and then early-cleaving (before 24 h) and late-cleaving (between 24 and 48 h) embryos were identified and cultured separately. Developing embryos, on Days 3, 5, and 7, were used to evaluate the total cell number and presence of DSBs (by counting the number of immunofluorescent foci for phosphorylated histone H2A.x [H2AX139ph] and RAD51 proteins) and to quantify transcripts of genes involved in DNA repair and cell cycle control by quantitative RT-PCR. Early-cleaving embryos had fewer DSBs, lower transcript levels for genes encoding DNA repair and cell cycle checkpoint proteins, and more cells than late-cleaving embryos. Interestingly, at the blastocyst stage, embryos that developed from early- and late-cleaving groups had similar number of DSBs as well as transcript levels of genes induced by DNA damage. This indicates that only embryos with less DNA damage and/or superior capacity for DNA repair are able to progress to the blastocyst stage. Collectively, findings in this study revealed a negative correlation between the occurrence of DSBs and embryo cleavage kinetics and embryo developmental capacity to the blastocyst stage.
Collapse
Affiliation(s)
| | - Ana Rita S Coutinho
- Department of Animal Science, McGill University, Ste. Anne de Bellevue, Quebec, Canada
| | - Raj Duggavathi
- Department of Animal Science, McGill University, Ste. Anne de Bellevue, Quebec, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Ste. Anne de Bellevue, Quebec, Canada
| |
Collapse
|
54
|
Kast K, Rhiem K. Familial breast cancer - targeted therapy in secondary and tertiary prevention. Breast Care (Basel) 2015; 10:27-31. [PMID: 25960722 DOI: 10.1159/000380756] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The introduction of an increasing number of individualized molecular targeted therapies into clinical routine mirrors their importance in modern cancer prevention and treatment. Well-known examples for targeted agents are the monoclonal antibody trastuzumab and the selective estrogen receptor modulator tamoxifen. The identification of an unaltered gene in tumor tissue in colon cancer (KRAS) is a predictor for the patient's response to targeted therapy with a monoclonal antibody (cetuximab). Targeted therapy for hereditary breast and ovarian cancer has become a reality with the approval of olaparib for platin-sensitive late relapsed BRCA-associated ovarian cancer in December 2014. This manuscript reviews the status quo of poly-ADP-ribose polymerase inhibitors (PARPi) in the therapy of breast and ovarian cancer as well as the struggle for carboplatin as a potential standard of care for triple-negative and, in particular, BRCA-associated breast cancer. Details of the mechanism of action with information on tumor development are provided, and an outlook for further relevant research is given. The efficacy of agents against molecular targets together with the identification of an increasing number of cancer-associated genes will open the floodgates to a new era of treatment decision-making based on molecular tumor profiles. Current clinical trials involving patients with BRCA-associated cancer explore the efficacy of the molecular targeted therapeutics platinum and PARPi.
Collapse
Affiliation(s)
- Karin Kast
- Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Kerstin Rhiem
- Center of Familial Breast and Ovarian Cancer, Department of Gynecology and Obstetrics, University Hospital Cologne, Germany
| |
Collapse
|
55
|
Du Z, Zhang W, Zhou Y, Yu D, Chen X, Chang J, Qiao Y, Zhang M, Huang Y, Wu C, Xiao Z, Tan W, Lin D. Associations of ATM Polymorphisms With Survival in Advanced Esophageal Squamous Cell Carcinoma Patients Receiving Radiation Therapy. Int J Radiat Oncol Biol Phys 2015; 93:181-9. [PMID: 26094126 DOI: 10.1016/j.ijrobp.2015.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/02/2015] [Accepted: 05/01/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE To investigate whether single nucleotide polymorphisms (SNPs) in the ataxia telangiectasia mutated (ATM) gene are associated with survival in patients with esophageal squamous cell carcinoma (ESCC) receiving radiation therapy or chemoradiation therapy or surgery only. METHODS AND MATERIALS Four tagSNPs of ATM were genotyped in 412 individuals with clinical stage III or IV ESCC receiving radiation therapy or chemoradiation therapy, and in 388 individuals with stage I, II, or III ESCC treated with surgery only. Overall survival time of ESCC among different genotypes was estimated by Kaplan-Meier plot, and the significance was examined by log-rank test. The hazard ratios (HRs) and 95% confidence intervals (CIs) for death from ESCC among different genotypes were computed by a Cox proportional regression model. RESULTS We found 2 SNPs, rs664143 and rs664677, associated with survival time of ESCC patients receiving radiation therapy. Individuals with the rs664143A allele had poorer median survival time compared with the rs664143G allele (14.0 vs 20.0 months), with the HR for death being 1.45 (95% CI 1.12-1.89). Individuals with the rs664677C allele also had worse median survival time than those with the rs664677T allele (14.0 vs 23.5 months), with the HR of 1.57 (95% CI 1.18-2.08). Stratified analysis showed that these associations were present in both stage III and IV cancer and different radiation therapy techniques. Significant associations were also found between the SNPs and locosregional progression or progression-free survival. No association between these SNPs and survival time was detected in ESCC patients treated with surgery only. CONCLUSION These results suggest that the ATM polymorphisms might serve as independent biomarkers for predicting prognosis in ESCC patients receiving radiation therapy.
Collapse
Affiliation(s)
- Zhongli Du
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuling Zhou
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dianke Yu
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiabin Chen
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Chang
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Qiao
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Zhang
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Huang
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Wu
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefen Xiao
- Department of Radiation Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wen Tan
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis (Beijing Key Laboratory for Carcinogenesis and Cancer Prevention), Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
56
|
Hsieh YH, Chang WS, Tsai CW, Tsai JP, Hsu CM, Jeng LB, Bau DT. DNA double-strand break repair gene XRCC7 genotypes were associated with hepatocellular carcinoma risk in Taiwanese males and alcohol drinkers. Tumour Biol 2015; 36:4101-6. [PMID: 25944161 DOI: 10.1007/s13277-014-2934-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/03/2014] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide, the prevalence and mortality rates of which are very high in Taiwan. The study aimed at evaluating the contribution of XRCC7 G6721T, together with cigarette smoking and alcohol drinking lifestyles, to the risk of HCC. In this hospital-based case-control study, the association of XRCC7 single nucleotide polymorphism G6721T with HCC risk was examined by polymerase chain reaction (PCR)-restriction fragment length polymorphism (RFLP) among 298 HCC patients and 889 age- and gender-matched healthy controls. The results showed that the percentages of TT, GT, and GG XRCC7 G6721T were 53.0, 41.3, and 5.7 % in the HCC patient group and 48.9, 43.1, and 8.0 % in the non-cancer control group, respectively. We have further stratified the populations by genders, cigarette smoking, and alcohol drinking status to investigate their combinative contributions with XRCC7 G6721T genotype to HCC risk. The results showed that the GG genotype of XRCC7 G6721T conducted a protective effect on HCC susceptibility which was obvious among males and drinkers, but not females, smokers, non-smokers, or non-drinkers (p = 0.0058, 0.0069, 0.1564, 0.2469, 0.9354, and 0.3416, respectively). Our results suggested that the GG and GT genotypes of X-ray repair cross-complementing group 7 (XRCC7) G6721T had no effect on HCC risk to the whole population, but had a protective effect on HCC risk among males and alcohol drinkers.
Collapse
Affiliation(s)
- Yi-Hsien Hsieh
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
57
|
Lee HB, Sebo ZL, Peng Y, Guo Y. An optimized TALEN application for mutagenesis and screening in Drosophila melanogaster. CELLULAR LOGISTICS 2015. [PMID: 26196022 PMCID: PMC4501208 DOI: 10.1080/21592799.2015.1023423] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transcription activator-like effector nucleases (TALENs) emerged as powerful tools for locus-specific genome engineering. Due to the ease of TALEN assembly, the key to streamlining TALEN-induced mutagenesis lies in identifying efficient TALEN pairs and optimizing TALEN mRNA injection concentrations to minimize the effort to screen for mutant offspring. Here we present a simple methodology to quantitatively assess bi-allelic TALEN cutting, as well as approaches that permit accurate measures of somatic and germline mutation rates in Drosophila melanogaster. We report that percent lethality from pilot injection of candidate TALEN mRNAs into Lig4 null embryos can be used to effectively gauge bi-allelic TALEN cutting efficiency and occurs in a dose-dependent manner. This timely Lig4-dependent embryonic survival assay also applies to CRISPR/Cas9-mediated targeting. Moreover, the somatic mutation rate of individual G0 flies can be rapidly quantitated using SURVEYOR nuclease and capillary electrophoresis, and germline transmission rate determined by scoring progeny of G0 outcrosses. Together, these optimized methods provide an effective step-wise guide for routine TALEN-mediated gene editing in the fly.
Collapse
Key Words
- TALEN
- TALENs, Transcription activator-like effector nucleases; TALEs, TAL effectors; ZFNs, Zinc Finger Nucleases; CRISPR, Clustered Regularly Interspersed Short Palindromic Repeats; Cas9, CRISPR-associated; RVDs, repeat-variable diresidues; DSBs, double-stranded breaks; NHEJ, non-homologous end joining; HR, homologous recombination; RFLP, restriction fragment length polymorphism; HRMA, high resolution melt analysis.
- engineered endonuclease
- genome engineering
- mutagenesis
- screening
Collapse
Affiliation(s)
- Han B Lee
- Graduate Program in Neurobiology of Disease; Mayo Graduate School; Mayo Clinic ; Rochester, MN, USA
| | | | - Ying Peng
- Department of Biochemistry and Molecular Biology; Mayo Clinic ; Rochester, MN, USA
| | - Yi Guo
- Department of Biochemistry and Molecular Biology; Mayo Clinic ; Rochester, MN, USA ; Division of Gastroenterology and Hepatology; Mayo Clinic ; Rochester, MN, USA
| |
Collapse
|
58
|
Smith SJ, Gu L, Phipps EA, Dobrolecki LE, Mabrey KS, Gulley P, Dillehay KL, Dong Z, Fields GB, Chen YR, Ann D, Hickey RJ, Malkas LH. A Peptide mimicking a region in proliferating cell nuclear antigen specific to key protein interactions is cytotoxic to breast cancer. Mol Pharmacol 2015; 87:263-76. [PMID: 25480843 PMCID: PMC4293449 DOI: 10.1124/mol.114.093211] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is a highly conserved protein necessary for proper component loading during the DNA replication and repair process. Proteins make a connection within the interdomain connector loop of PCNA, and much of the regulation is a result of the inherent competition for this docking site. If this target region of PCNA is modified, the DNA replication and repair process in cancer cells is potentially altered. Exploitation of this cancer-associated region has implications for targeted breast cancer therapy. In the present communication, we characterize a novel peptide (caPeptide) that has been synthesized to mimic the sequence identified as critical to the cancer-associated isoform of PCNA. This peptide is delivered into cells using a nine-arginine linking mechanism, and the resulting peptide (R9-cc-caPeptide) exhibits cytotoxicity in a triple-negative breast cancer cell line, MDA-MB-436, while having less of an effect on the normal counterparts (MCF10A and primary breast epithelial cells). The novel peptide was then evaluated for cytotoxicity using various in vivo techniques, including ATP activity assays, flow cytometry, and clonogenetic assays. This cytotoxicity has been observed in other breast cancer cell lines (MCF7 and HCC1937) and other forms of cancer (pancreatic and lymphoma). R9-cc-caPeptide has also been shown to block the association of PCNA with chromatin. Alanine scanning of the peptide sequence, combined with preliminary in silico modeling, gives insight to the disruptive ability and the molecular mechanism of action of the therapeutic peptide in vivo.
Collapse
Affiliation(s)
- Shanna J Smith
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Long Gu
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Elizabeth A Phipps
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Lacey E Dobrolecki
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Karla S Mabrey
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Pattie Gulley
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Kelsey L Dillehay
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Zhongyun Dong
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Gregg B Fields
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Yun-Ru Chen
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - David Ann
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Robert J Hickey
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| | - Linda H Malkas
- Department of Molecular and Cellular Biology (S.J.S., L.G., L.H.M.), Department of Molecular Medicine (R.J.H.), and Department of Diabetes and Metabolic Diseases Research (Y.-R.C., D.A.), Beckman Research Institute at City of Hope, Duarte, California; Department of Medical and Molecular Genetics (E.A.P.) and Department of Medicine (K.S.M., P.G.), Indiana University School of Medicine, Indianapolis, Indiana; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas (L.E.D.); Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio (K.L.D., Z.D.); and Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (G.B.F.)
| |
Collapse
|
59
|
McKelvey SM, Horgan KA, Murphy RA. Chemical form of selenium differentially influences DNA repair pathways following exposure to lead nitrate. J Trace Elem Med Biol 2015; 29:151-69. [PMID: 25023848 DOI: 10.1016/j.jtemb.2014.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
Abstract
Lead, an environmental toxin is known to induce a broad range of physiological and biochemical dysfunctions in humans through a number of mechanisms including the deactivation of antioxidants thus leading to generation of reactive oxygen species (ROS) and subsequent DNA damage. Selenium on the other hand has been proven to play an important role in the protection of cells from free radical damage and oxidative stress, though its effects are thought to be form and dose dependent. As the liver is the primary organ required for metabolite detoxification, HepG2 cells were chosen to assess the protective effects of various selenium compounds following exposure to the genotoxic agent lead nitrate. Initially DNA damage was quantified using a comet assay, gene expression patterns associated with DNA damage and signalling were also examined using PCR arrays and the biological pathways which were most significantly affected by selenium were identified. Interestingly, the organic type selenium compounds (selenium yeast and selenomethionine) conferred protection against lead induced DNA damage in HepG2 cells; this is evident by reduction in the quantity of DNA present in the comet tail of cells cultured in their presence with lead. This trend also followed through the gene expression changes noted in DNA damage pathways analysed. These results were in contrast with those of inorganic sodium selenite which promoted lead induced DNA damage evident in both the comet assay results and the gene expression analysis. Over all this study provided valuable insights into the effects which various selenium compounds had on the DNA damage and signalling pathway indicating the potential for using organic forms of selenium such as selenium enriched yeast to protect against DNA damaging agents.
Collapse
Affiliation(s)
- Shauna M McKelvey
- Alltech Biotechnology Centre, Sarney, Summerhill Rd., Dunboyne, County Meath, Ireland.
| | - Karina A Horgan
- Alltech Biotechnology Centre, Sarney, Summerhill Rd., Dunboyne, County Meath, Ireland
| | - Richard A Murphy
- Alltech Biotechnology Centre, Sarney, Summerhill Rd., Dunboyne, County Meath, Ireland
| |
Collapse
|
60
|
Andrs M, Korabecny J, Jun D, Hodny Z, Bartek J, Kuca K. Phosphatidylinositol 3-Kinase (PI3K) and phosphatidylinositol 3-kinase-related kinase (PIKK) inhibitors: importance of the morpholine ring. J Med Chem 2014; 58:41-71. [PMID: 25387153 DOI: 10.1021/jm501026z] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phosphatidylinositol 3-kinases (PI3Ks) and phosphatidylinositol 3-kinase-related protein kinases (PIKKs) are two related families of kinases that play key roles in regulation of cell proliferation, metabolism, migration, survival, and responses to diverse stresses including DNA damage. To design novel efficient strategies for treatment of cancer and other diseases, these kinases have been extensively studied. Despite their different nature, these two kinase families have related origin and share very similar kinase domains. Therefore, chemical inhibitors of these kinases usually carry analogous structural motifs. The most common feature of these inhibitors is a critical hydrogen bond to morpholine oxygen, initially present in the early nonspecific PI3K and PIKK inhibitor 3 (LY294002), which served as a valuable chemical tool for development of many additional PI3K and PIKK inhibitors. While several PI3K pathway inhibitors have recently shown promising clinical responses, inhibitors of the DNA damage-related PIKKs remain thus far largely in preclinical development.
Collapse
Affiliation(s)
- Martin Andrs
- Biomedical Research Center, University Hospital Hradec Kralove , Sokolska 81, 500 05 Hradec Kralove, Czech Republic
| | | | | | | | | | | |
Collapse
|
61
|
Latimer JJ, Majekwana VJ, Pabón-Padín YR, Pimpley MR, Grant SG. Regulation and disregulation of mammalian nucleotide excision repair: a pathway to nongermline breast carcinogenesis. Photochem Photobiol 2014; 91:493-500. [PMID: 25393451 DOI: 10.1111/php.12387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/27/2014] [Indexed: 12/13/2022]
Abstract
Nucleotide excision repair (NER) is an important modulator of disease, especially in constitutive deficiencies such as the cancer predisposition syndrome Xeroderma pigmentosum. We have found profound variation in NER capacity among normal individuals, between cell-types and during carcinogenesis. NER is a repair system for many types of DNA damage, and therefore many types of genotoxic carcinogenic exposures, including ultraviolet light, products of organic combustion, metals and oxidative stress. Because NER is intimately related to cellular metabolism, requiring components of both the DNA replicative and transcription machinery, it has a narrow range of functional viability. Thus, genes in the NER pathway are expressed at the low levels manifested by, for example, nuclear transcription factors. As NER activity and gene expression vary by cell-type, it is inherently epigenetically regulated. Furthermore, this epigenetic modulation is disregulated during sporadic breast carcinogenesis. Loss of NER is one basis of genomic instability, a required element in cellular transformation, and one that potentially influences response to therapy. In this study, we demonstrate differences in NER capacity in eight adult mouse tissues, and place this result into the context of our previous work on mouse extraembryonic tissues, normal human tissues and sporadic early stage human breast cancer.
Collapse
Affiliation(s)
- Jean J Latimer
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL
| | | | | | | | | |
Collapse
|
62
|
Lan J, Gou N, Gao C, He M, Gu AZ. Comparative and mechanistic genotoxicity assessment of nanomaterials via a quantitative toxicogenomics approach across multiple species. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2014; 48:12937-45. [PMID: 25338269 PMCID: PMC4224493 DOI: 10.1021/es503065q] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 05/19/2023]
Abstract
This study reports a comparative and mechanistic genotoxicity assessment of four engineered nanomaterials (ENMs) across three species, including E. coli, yeast, and human cells, with the aim to reveal the distinct potential genotoxicity mechanisms among the different nanomaterials and their association with physiochemical features. Both the conventional phenotypic alkaline comet test and the newly developed quantitative toxicogenomics assay, that detects and quantifies molecular level changes in the regulation of six DNA damage repair pathways, were employed. The proposed molecular endpoints derived from the toxicogenomics assays, namely TELI (Transcriptional Effect Level Index) and PELI (Protein Effect Level Index), correlated well with the phenotypic DNA damage endpoints from comet tests, suggesting that the molecular genotoxicity assay is suitable for genotoxicity detection. Temporal altered gene or protein expression profiles revealed various potential DNA damage types and relevant genotoxic mechanisms induced by the tested ENMs. nTiO2_a induced a wide spectrum of DNA damage consistently across three species. Three carbon-based ENMs, namely carbon black, single wall carbon nanotube (SWCNT) and fullerene, exhibited distinct, species and ENM property-dependent DNA damage mechanisms. All carbon based ENMs induced relatively weak DNA damage repair response in E. coli, but more severe DNA double strand break in eukaryotes. The differences in cellular structure and defense systems among prokaryotic and eukaryotic species lead to distinct susceptibility and mechanisms for ENM uptake and, thus, varying DNA damages and repair responses. The observation suggested that eukaryotes, especially mammalian cells, are likely more susceptible to genotoxicity than prokaryotes in the ecosystem when exposed to these ENMs.
Collapse
Affiliation(s)
- Jiaqi Lan
- Department
of Civil and Environmental Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Na Gou
- Department
of Civil and Environmental Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Ce Gao
- Department
of Civil and Environmental Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Miao He
- Environmental
simulation and pollution control (ESPC) State Key Joint Laboratory,
School of Environment, Tsinghua University, Beijing, 100084, People’s Republic of China
| | - April Z. Gu
- Department
of Civil and Environmental Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
- Tel.: + 1-617−373−3631; e-mail:
| |
Collapse
|
63
|
MARINA RAQUEL, GONZÁLEZ PAQUITA, FERRERAS MCARMEN, COSTILLA SERAFÍN, BARRIO JUANPABLO. Hepatic Nrf2 expression is altered by quercetin supplementation in X-irradiated rats. Mol Med Rep 2014; 11:539-46. [DOI: 10.3892/mmr.2014.2741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 07/04/2014] [Indexed: 11/06/2022] Open
|
64
|
Zhu B, Yan K, Li L, Lin M, Zhang S, He Q, Zheng D, Yang H, Shao G. K63-linked ubiquitination of FANCG is required for its association with the Rap80-BRCA1 complex to modulate homologous recombination repair of DNA interstand crosslinks. Oncogene 2014; 34:2867-78. [PMID: 25132264 DOI: 10.1038/onc.2014.229] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 05/21/2014] [Accepted: 06/23/2014] [Indexed: 11/09/2022]
Abstract
DNA interstrand crosslinks (ICLs) are extremely deleterious lesions that are repaired by homologous recombination (HR) through coordination of Fanconi anemia (FA) proteins and breast cancer susceptibility gene 1 (BRCA1) product, but the exact role these proteins have remains unclear. Here we report that FANCG was modified by the addition of lysine63-linked polyubiquitin chains (K63Ub) in response to DNA damage. We show that FANCG K63Ub was dispensable for monoubiquitination of FANCD2, but was required for FANCG to interact with the Rap80-BRCA1 (receptor-associated protein 80-BRCA1) complex for subsequent modulation of HR repair of ICLs induced by mitomycin C. Mutation of three lysine residues within FANCG to arginine (K182, K258 and K347, 3KR) reduced FANCG K63Ub modification, as well as its interaction with the Rap80-BRCA1 complex, and therefore impeded HR repair. In addition, we demonstrated that K63Ub-modified FANCG was deubiquitinated by BRCC36 complex in vitro and in vivo. Inhibition of BRCC36 resulted in increased K63Ub modification of FANCG. Taken together, our results identify a new role of FANCG in HR repair of ICL through K63Ub-mediated interaction with the Rap80-BRCA1 complex.
Collapse
Affiliation(s)
- B Zhu
- 1] Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China [2] Institute of Systems Biology, Peking University, Beijing, China
| | - K Yan
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - L Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - M Lin
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - S Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Q He
- Center of Medical and Health Analysis, Peking University, Beijing, China
| | - D Zheng
- School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - H Yang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - G Shao
- 1] Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China [2] Institute of Systems Biology, Peking University, Beijing, China
| |
Collapse
|
65
|
Han J, Won EJ, Lee BY, Hwang UK, Kim IC, Yim JH, Leung KMY, Lee YS, Lee JS. Gamma rays induce DNA damage and oxidative stress associated with impaired growth and reproduction in the copepod Tigriopus japonicus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 152:264-272. [PMID: 24800869 DOI: 10.1016/j.aquatox.2014.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/01/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
Nuclear radioisotope accidents are potentially ecologically devastating due to their impact on marine organisms. To examine the effects of exposure of a marine organism to radioisotopes, we irradiated the intertidal copepod Tigriopus japonicus with several doses of gamma radiation and analyzed the effects on mortality, fecundity, and molting by assessing antioxidant enzyme activities and gene expression patterns. No mortality was observed at 96h, even in response to exposure to a high dose (800Gy) of radiation, but mortality rate was significantly increased 120h (5 days) after exposure to 600 or 800Gy gamma ray radiation. We observed a dose-dependent reduction in fecundity of ovigerous females; even the group irradiated with 50Gy showed a significant reduction in fecundity, suggesting that gamma rays are likely to have a population level effect. In addition, we observed growth retardation, particularly at the nauplius stage, in individuals after gamma irradiation. In fact, nauplii irradiated with more than 200Gy, though able to molt to copepodite stage 1, did not develop into adults. Upon gamma radiation, T. japonicus showed a dose-dependent increase in reactive oxygen species (ROS) levels, the activities of several antioxidant enzymes, and expression of double-stranded DNA break damage genes (e.g. DNA-PK, Ku70, Ku80). At a low level (sub-lethal dose) of gamma irradiation, we found dose-dependent upregulation of p53, implying cellular damage in T. japonicus in response to sub-lethal doses of gamma irradiation, suggesting that T. japonicus is not susceptible to sub-lethal doses of gamma irradiation. Additionally, antioxidant genes, phase II enzyme (e.g. GSTs), and cellular chaperone genes (e.g. Hsps) that are involved in cellular defense mechanisms also showed the same expression patterns for sublethal doses of gamma irradiation (50-200Gy). These findings indicate that sublethal doses of gamma radiation can induce oxidative stress-mediated DNA damage and increase the expression of antioxidant enzymes and proteins with chaperone-related functions, thereby significantly affecting life history parameters such as fecundity and molting in the copepod T. japonicus.
Collapse
Affiliation(s)
- Jeonghoon Han
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 440-746, South Korea
| | - Eun-Ji Won
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 440-746, South Korea
| | - Bo-Young Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 440-746, South Korea
| | - Un-Ki Hwang
- Marine Ecological Risk Assessment Center, West Sea Fisheries Research Institute, National Fisheries Research & Development Institute, Incheon 400-420, South Korea
| | - Il-Chan Kim
- Division of Life Sciences, Korea Polar Research Institute, Incheon 406-840, South Korea
| | - Joung Han Yim
- Division of Life Sciences, Korea Polar Research Institute, Incheon 406-840, South Korea
| | - Kenneth Mei Yee Leung
- School of Biological Sciences and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yong Sung Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, South Korea
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 440-746, South Korea.
| |
Collapse
|
66
|
Bhattacharjee V, Lin Y, Waldman BC, Waldman AS. Induction of recombination between diverged sequences in a mammalian genome by a double-strand break. Cell Mol Life Sci 2014; 71:2359-71. [PMID: 24257896 PMCID: PMC11113419 DOI: 10.1007/s00018-013-1520-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/01/2013] [Accepted: 11/08/2013] [Indexed: 10/26/2022]
Abstract
To investigate whether mammalian cells can carry out recombinational double-strand break (DSB) repair between highly diverged sequences, mouse fibroblasts were transfected with DNA substrates that contained a "recipient" thymidine kinase (tk) gene disrupted by the recognition site for endonuclease I-SceI. Substrates also contained a linked "donor" tk gene sequence. Following DSB induction by I-SceI, selection for tk-expressing clones allowed recovery of repair events occurring by nonhomologous end-joining or recombination with the donor sequence. Although recombinational repair was most efficient when donor and recipient shared near-perfect homology, we recovered recombination events between recipient and donor sequences displaying 20 % nucleotide mismatch. Recombination between such imperfectly matched ("homeologous") sequences occurred at a frequency of 1.7 × 10(-7) events per cell and constituted 3 % of the DSB repair events recovered with the pair of homeologous sequences. Additional experiments were done with a substrate containing a donor sequence comprised of a region sharing high homology with the recipient and an adjacent region homeologous to the recipient. Recombinational DSB repair tracts initiating within high homology propagated into homeology in 11 of 112 repair events. These collective results contrasted with our earlier work in which spontaneous recombination (not intentionally induced by a DSB) between homeologous sequences occurred at an undetectable frequency of less than 10(-9) events per cell, and in which events initiating within high homology propagated into adjoining homeology in one of 81 events examined. Our current work suggests that homology requirements for recombination are effectively relaxed in proximity to a DSB in a mammalian genome.
Collapse
Affiliation(s)
- Vikram Bhattacharjee
- Department of Biological Sciences, University of South Carolina, 700 Sumter Street, Columbia, SC, 29208, USA,
| | | | | | | |
Collapse
|
67
|
Bohrer RC, Duggavathi R, Bordignon V. Inhibition of histone deacetylases enhances DNA damage repair in SCNT embryos. Cell Cycle 2014; 13:2138-48. [PMID: 24841373 DOI: 10.4161/cc.29215] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent studies have shown that DNA damage affects embryo development and also somatic cell reprogramming into induced pluripotent stem (iPS) cells. It has been also shown that treatment with histone deacetylase inhibitors (HDACi) improves development of embryos produced by somatic cell nuclear transfer (SCNT) and enhances somatic cell reprogramming. There is evidence that increasing histone acetylation at the sites of DNA double-strand breaks (DSBs) is critical for DNA damage repair. Therefore, we hypothesized that HDACi treatment enhances cell programming and embryo development by facilitating DNA damage repair. To test this hypothesis, we first established a DNA damage model wherein exposure of nuclear donor cells to ultraviolet (UV) light prior to nuclear transfer reduced the development of SCNT embryos proportional to the length of UV exposure. Detection of phosphorylated histone H2A.x (H2AX139ph) foci confirmed that exposure of nuclear donor cells to UV light for 10 s was sufficient to increase DSBs in SCNT embryos. Treatment with HDACi during embryo culture increased development and reduced DSBs in SCNT embryos produced from UV-treated cells. Transcript abundance of genes involved in either the homologous recombination (HR) or nonhomologous end-joining (NHEJ) pathways for DSBs repair was reduced by HDACi treatment in developing embryos at day 5 after SCNT. Interestingly, expression of HR and NHEJ genes was similar between HDACi-treated and control SCNT embryos that developed to the blastocyst stage. This suggested that the increased number of embryos that could achieve the blastocyst stage in response to HDACi treatment have repaired DNA damage. These results demonstrate that DNA damage in nuclear donor cells is an important component affecting development of SCNT embryos, and that HDACi treatment after nuclear transfer enhances DSBs repair and development of SCNT embryos.
Collapse
Affiliation(s)
| | - Raj Duggavathi
- Department of Animal Science; McGill University; Ste. Anne de Bellevue, Quebec, Canada
| | - Vilceu Bordignon
- Department of Animal Science; McGill University; Ste. Anne de Bellevue, Quebec, Canada
| |
Collapse
|
68
|
Vollebergh MA, Lips EH, Nederlof PM, Wessels LFA, Wesseling J, Vd Vijver MJ, de Vries EGE, van Tinteren H, Jonkers J, Hauptmann M, Rodenhuis S, Linn SC. Genomic patterns resembling BRCA1- and BRCA2-mutated breast cancers predict benefit of intensified carboplatin-based chemotherapy. Breast Cancer Res 2014; 16:R47. [PMID: 24887359 PMCID: PMC4076636 DOI: 10.1186/bcr3655] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 05/02/2014] [Indexed: 12/12/2022] Open
Abstract
Introduction BRCA-mutated breast cancer cells lack the DNA-repair mechanism homologous recombination that is required for error-free DNA double-strand break (DSB) repair. Homologous recombination deficiency (HRD) may cause hypersensitivity to DNA DSB-inducing agents, such as bifunctional alkylating agents and platinum salts. HRD can be caused by BRCA mutations, and by other mechanisms. To identify HRD, studies have focused on triple-negative (TN) breast cancers as these resemble BRCA1-mutated breast cancer closely and might also share this hypersensitivity. However, ways to identify HRD in non-BRCA-mutated, estrogen receptor (ER)-positive breast cancers have remained elusive. The current study provides evidence that genomic patterns resembling BRCA1- or BRCA2-mutated breast cancers can identify breast cancer patients with TN as well as ER-positive, HER2-negative tumors that are sensitive to intensified, DSB-inducing chemotherapy. Methods Array comparative genomic hybridization (aCGH) was used to classify breast cancers. Patients with tumors with similar aCGH patterns as BRCA1- and/or BRCA2-mutated breast cancers were defined as having a BRCA-likeCGH status, others as non-BCRA-likeCGH. Stage-III patients (n = 249) had participated in a randomized controlled trial of adjuvant high-dose (HD) cyclophosphamide-thiotepa-carboplatin (CTC) versus 5-fluorouracil-epirubicin-cyclophosphamide (FE90C) chemotherapy. Results Among patients with BRCA-likeCGH tumors (81/249, 32%), a significant benefit of HD-CTC compared to FE90C was observed regarding overall survival (adjusted hazard ratio 0.19, 95% CI: 0.08 to 0.48) that was not seen for patients with non-BRCA-likeCGH tumors (adjusted hazard ratio 0.90, 95% CI: 0.53 to 1.54) (P = 0.004). Half of all BRCA-likeCGH tumors were ER-positive. Conclusions Distinct aCGH patterns differentiated between HER2-negative patients with a markedly improved outcome after adjuvant treatment with an intensified DNA-DSB-inducing regimen (BRCA-likeCGH patients) and those without benefit (non-BRCA-likeCGH patients).
Collapse
|
69
|
Won EJ, Lee JS. Gamma radiation induces growth retardation, impaired egg production, and oxidative stress in the marine copepod Paracyclopina nana. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 150:17-26. [PMID: 24632311 DOI: 10.1016/j.aquatox.2014.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/16/2014] [Accepted: 02/18/2014] [Indexed: 06/03/2023]
Abstract
Accidental nuclear radioisotope release into the ocean from nuclear power plants is of concern due to ecological and health risks. In this study, we used the marine copepod Paracyclopina nana to examine the effects of radioisotopes on marine organisms upon gamma radiation, and to measure the effects on growth and fecundity, which affect population and community structure. Upon gamma radiation, mortality (LD50 - 96 h=172 Gy) in P. nana was significantly increased in a dose-dependent manner in ovigerous P. nana females. For developmental impairment of gamma-irradiated nauplii, we observed growth retardation; in over 30 Gy-irradiated groups, offspring did not grow to adults. Particularly, over 50 Gy-irradiated ovigerous P. nana females did not have normal bilateral egg sacs, and their offspring did not develop normally to adulthood. Additionally, at over 30 Gy, we found dose-dependent increases in oxidative levels with elevated antioxidant enzyme activities and DNA repair activities. These findings indicate that gamma radiation can induce oxidative stress and DNA damage with growth retardation and impaired reproduction.
Collapse
Affiliation(s)
- Eun-Ji Won
- Department of Biological Sciences, College of Natural Sciences, Sungkyunkwan University, Suwon 440-746, South Korea
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Natural Sciences, Sungkyunkwan University, Suwon 440-746, South Korea.
| |
Collapse
|
70
|
Turner RL, Wilkinson JC, Ornelles DA. E1B and E4 oncoproteins of adenovirus antagonize the effect of apoptosis inducing factor. Virology 2014; 456-457:205-19. [PMID: 24889240 DOI: 10.1016/j.virol.2014.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 02/28/2014] [Accepted: 03/10/2014] [Indexed: 01/03/2023]
Abstract
Adenovirus inundates the productively infected cell with linear, double-stranded DNA and an abundance of single-stranded DNA. The cellular response to this stimulus is antagonized by the adenoviral E1B and E4 early genes. A mutant group C adenovirus that fails to express the E1B-55K and E4orf3 genes is unable to suppress the DNA-damage response. Cells infected with this double-mutant virus display significant morphological heterogeneity at late times of infection and frequently contain fragmented nuclei. Nuclear fragmentation was due to the translocation of apoptosis inducing factor (AIF) from the mitochondria into the nucleus. The release of AIF was dependent on active poly(ADP-ribose) polymerase-1 (PARP-1), which appeared to be activated by viral DNA replication. Nuclear fragmentation did not occur in AIF-deficient cells or in cells treated with a PARP-1 inhibitor. The E1B-55K or E4orf3 proteins independently prevented nuclear fragmentation subsequent to PARP-1 activation, possibly by altering the intracellular distribution of PAR-modified proteins.
Collapse
Affiliation(s)
- Roberta L Turner
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - John C Wilkinson
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States.
| | - David A Ornelles
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States.
| |
Collapse
|
71
|
Ramalho EA, Silva-Filho JL, Cartaxo MF, Cavalcanti CB, Rêgo MJ, Oliveira MB, Beltrão EI. Assessment of changes in the BRCA2 and P53 genes in breast invasive ductal carcinoma in northeast Brazil. Biol Res 2014; 47:3. [PMID: 25027116 PMCID: PMC4060371 DOI: 10.1186/0717-6287-47-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 10/24/2013] [Indexed: 02/06/2023] Open
Abstract
Background BRCA protein interacts with at least 13 different proteins that have been implicated with cancer susceptibility and loss of BRCA function is correlated to sensitivity to DNA crosslinking agents in preclinical models. Results BRCA2 methylation frequency was 44%, p53 Pro22 allele frequency was 32% and heterozygous frequency of Arg/Pro72 genotype was 60% which could be associated as risk factor for metastasis (p = 0.046 OR = 4.190). Regarding to polymorphism of codon 249 the frequency of Arg249 allele presented 82% which was considered not statistically significant. Conclusions There was not statistical significance to BRCA2 promoter methylation with any parameters chosen. However, our findings suggest that patients who present heterozygous genotype at codon 72 of p53 gene may have a major susceptibility to any type of metastasis and this could serve as potential auxiliary biomarker for poor prognosis.
Collapse
|
72
|
Abbotts R, Thompson N, Madhusudan S. DNA repair in cancer: emerging targets for personalized therapy. Cancer Manag Res 2014; 6:77-92. [PMID: 24600246 PMCID: PMC3933425 DOI: 10.2147/cmar.s50497] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Genomic deoxyribonucleic acid (DNA) is under constant threat from endogenous and exogenous DNA damaging agents. Mammalian cells have evolved highly conserved DNA repair machinery to process DNA damage and maintain genomic integrity. Impaired DNA repair is a major driver for carcinogenesis and could promote aggressive cancer biology. Interestingly, in established tumors, DNA repair activity is required to counteract oxidative DNA damage that is prevalent in the tumor microenvironment. Emerging clinical data provide compelling evidence that overexpression of DNA repair factors may have prognostic and predictive significance in patients. More recently, DNA repair inhibition has emerged as a promising target for anticancer therapy. Synthetic lethality exploits intergene relationships where the loss of function of either of two related genes is nonlethal, but loss of both causes cell death. Exploiting this approach by targeting DNA repair has emerged as a promising strategy for personalized cancer therapy. In the current review, we focus on recent advances with a particular focus on synthetic lethality targeting in cancer.
Collapse
Affiliation(s)
- Rachel Abbotts
- University of Nottingham, Academic Unit of Oncology, Division of Oncology, School of Medicine, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | - Nicola Thompson
- University of Nottingham, Academic Unit of Oncology, Division of Oncology, School of Medicine, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | - Srinivasan Madhusudan
- University of Nottingham, Academic Unit of Oncology, Division of Oncology, School of Medicine, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| |
Collapse
|
73
|
Katz SS, Gimble FS, Storici F. To nick or not to nick: comparison of I-SceI single- and double-strand break-induced recombination in yeast and human cells. PLoS One 2014; 9:e88840. [PMID: 24558436 PMCID: PMC3928301 DOI: 10.1371/journal.pone.0088840] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 01/14/2014] [Indexed: 11/19/2022] Open
Abstract
Genetic modification of a chromosomal locus to replace an existing dysfunctional allele with a corrected sequence can be accomplished through targeted gene correction using the cell's homologous recombination (HR) machinery. Gene targeting is stimulated by generation of a DNA double-strand break (DSB) at or near the site of correction, but repair of the break via non-homologous end-joining without using the homologous template can lead to deleterious genomic changes such as in/del mutations, or chromosomal rearrangements. By contrast, generation of a DNA single-strand break (SSB), or nick, can stimulate gene correction without the problems of DSB repair because the uncut DNA strand acts as a template to permit healing without alteration of genetic material. Here, we examine the ability of a nicking variant of the I-SceI endonuclease (K223I I-SceI) to stimulate gene targeting in yeast Saccharomyces cerevisiae and in human embryonic kidney (HEK-293) cells. K223I I-SceI is proficient in both yeast and human cells and promotes gene correction up to 12-fold. We show that K223I I-SceI-driven recombination follows a different mechanism than wild-type I-SceI-driven recombination, thus indicating that the initial DNA break that stimulates recombination is not a low-level DSB but a nick. We also demonstrate that K223I I-SceI efficiently elevates gene targeting at loci distant from the break site in yeast cells. These findings establish the capability of the I-SceI nickase to enhance recombination in yeast and human cells, strengthening the notion that nicking enzymes could be effective tools in gene correction strategies for applications in molecular biology, biotechnology, and gene therapy.
Collapse
Affiliation(s)
- Samantha S. Katz
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Frederick S. Gimble
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Francesca Storici
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
74
|
Zeng T, Li Y, Zhang CL, Yu LH, Zhu ZP, Zhao XL, Xie KQ. Garlic oil suppressed the hematological disorders induced by chemotherapy and radiotherapy in tumor-bearing mice. J Food Sci 2014; 78:H936-42. [PMID: 23772706 DOI: 10.1111/1750-3841.12137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 03/24/2013] [Indexed: 12/31/2022]
Abstract
Although the anticancer effects of garlic and its products have been demonstrated by a variety of studies; however, few studies were conducted to investigate the effects of garlic on the adverse effects of chemo/radiotherapy. In order to clarify the above question and make a more comprehensive understanding of the anticancer effects of garlic, tumor xenograft mice model was established by subcutaneous injection of H22 tumor cells, and was used for the investigation of effects of garlic oil (GO) on the chemo/radiotherapy. In the chemotherapy test, tumor-bearing mice were treated with cyclophosphamide (CTX) or CTX plus GO (25 or 50 mg/kg bw) for 14 d, while the mice received a single 5 Gy total body radiation or radiation plus GO (25 or 50 mg/kg bw) in radiotherapy test. The results showed that GO did not increase the tumor inhibitory rate of CTX/radiation, which indicated that GO could not enhance the chemo/radiosensitivity of cancer cells. However, the decrease of the peripheral total white blood cells (WBCs) count induced by CTX/radiation was significantly suppressed by GO cotreatment. Furthermore, GO cotreatment significantly inhibited the decrease of the DNA contents and the micronuclei ratio of the bone marrow. Lastly, the reduction of the endogenous spleen colonies induced by CTX/radiation was significantly suppressed by GO cotreatment. These findings support the idea that GO consumption may benefit for the cancer patients receiving chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- Tao Zeng
- School of Public Health, Shandong Univ., 44 Wenhua West Road, Shandong Province, Jinan City, 250012, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
DNA damage response genes play vital roles in the maintenance of a healthy genome. Defects in cell cycle checkpoint and DNA repair genes, especially mutation or aberrant downregulation, are associated with a wide spectrum of human disease, including a predisposition to the development of neurodegenerative conditions and cancer. On the other hand, upregulation of DNA damage response and repair genes can also cause cancer, as well as increase resistance of cancer cells to DNA damaging therapy. In recent years, it has become evident that many of the genes involved in DNA damage repair have additional roles in tumorigenesis, most prominently by acting as transcriptional (co-)factors. Although defects in these genes are causally connected to tumor initiation, their role in tumor progression is more controversial and it seems to depend on tumor type. In some tumors like melanoma, cell cycle checkpoint/DNA repair gene upregulation is associated with tumor metastasis, whereas in a number of other cancers the opposite has been observed. Several genes that participate in the DNA damage response, such as RAD9, PARP1, BRCA1, ATM and TP53 have been associated with metastasis by a number of in vitro biochemical and cellular assays, by examining human tumor specimens by immunohistochemistry or by DNA genome-wide gene expression profiling. Many of these genes act as transcriptional effectors to regulate other genes implicated in the pathogenesis of cancer. Furthermore, they are aberrantly expressed in numerous human tumors and are causally related to tumorigenesis. However, whether the DNA damage repair function of these genes is required to promote metastasis or another activity is responsible (e.g., transcription control) has not been determined. Importantly, despite some compelling in vitro evidence, investigations are still needed to demonstrate the role of cell cycle checkpoint and DNA repair genes in regulating metastatic phenotypes in vivo.
Collapse
Affiliation(s)
- Constantinos G. Broustas
- Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Howard B. Lieberman
- Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| |
Collapse
|
76
|
Alsbeih G, Brock W, Story M. Misrepair of DNA double-strand breaks in patient with unidentified chromosomal fragility syndrome and family history of radiosensitivity. Int J Radiat Biol 2014; 90:53-9. [PMID: 24164476 DOI: 10.3109/09553002.2014.859764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE To test the hypothesis that differences in DNA double-strand breaks (DSB) repair fidelity underlies differences in radiosensitivity. MATERIALS AND METHODS A primary fibroblast culture (C42) derived from a pediatric cancer patient treated with reduced radiation doses consequent to a family history of radiosensitivity reminiscent of chromosomal fragility syndrome, was compared to a normal control (C29). DNA DSB rejoining and repair fidelity were studied by Southern blotting and hybridization to specific fragments: Alu repetitive sequence representing the overall DSB rejoining capacity in the genome and a 3.2 Mbp NotI restriction fragment on chromosome 21 for DSB repair fidelity. RESULTS Although both assays showed statistically significant difference (p ≤ 0.05) between the two cell strains in residual misrepaired (un-or mis-rejoined) DSB (24 h after 30 or 80 Gy), the residual damage was lower in the Alu enriched genome assay compared to NotI assay (0.01-0.07 and 0.10-0.37, respectively). CONCLUSIONS These results suggest that, in comparison to classic DSB repair experiment, an assay of measuring DNA DSB repair fidelity can provide better resolution and a more accurate estimate of misrepair of radiation-induced DNA damage, which underlies genomic instability and increased radiosensitivity.
Collapse
Affiliation(s)
- Ghazi Alsbeih
- King Faisal Specialist Hospital & Research Centre , Riyadh , Saudi Arabia
| | | | | |
Collapse
|
77
|
Ghoneum M, Elbaghdady HAM, El-Shebly AA, Pan D, Assanah E, Lawson G. Protective effect of hydroferrate fluid, MRN-100, against lethality and hematopoietic tissue damage in γ-radiated Nile tilapia, Oreochromis niloticus. JOURNAL OF RADIATION RESEARCH 2013; 54:852-862. [PMID: 23589025 PMCID: PMC3766301 DOI: 10.1093/jrr/rrt029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 02/05/2013] [Accepted: 03/05/2013] [Indexed: 06/02/2023]
Abstract
Hydroferrate fluid, MRN-100, an iron-based compound derived from bivalent and trivalent ferrates, is a potent antioxidant compound. Therefore, we examined the protective effect of MRN-100 against γ-radiation-induced lethality and damage to hematopoietic tissues in fish. A total of 216 Nile tilapia fish (Oreochromis niloticus) were randomly divided into four groups. Group 1 served as a control that was administered no radiation and no MRN-100 treatment. Group 2 was exposed only to γ-radiation (15 Gy). Groups 3 and 4 were pre-treated with MRN-100 at doses of either 1 ml/l or 3 ml/l in water for 1 week, and subsequently exposed to radiation while continuing to receive MRN-100 for 27 days. The survival rate was measured, and biochemical and histopathological analyses of hematopoietic tissues were performed for the different treatment groups at 1 and 4 weeks post-radiation. Exposure to radiation reduced the survival rate to 27.7%, while treatment with MRN-100 maintained the survival rate at 87.2%. In addition, fish exposed to γ-radiation for 1 week showed a significant decrease in the total number of white blood cells (WBCs) and red blood cells (RBCs) series. However, treatment with MRN-100 protected the total WBC count and the RBCs series when compared with irradiated fish. Furthermore, significant histological lesions were observed in the hepatopancreas, spleen and gills of irradiated fish. However, treatment with MRN-100 protected the histopathology of various organs. We conclude that MRN-100 is a radioprotective agent in fish and may be useful as an adjuvant treatment to counteract the adverse side effects associated with radiation exposure.
Collapse
Affiliation(s)
- Mamdooh Ghoneum
- Departments of Otolaryngology, Charles Drew University of Medicine and Science, 1621 East, 120th Street, Los Angeles, California 90059
| | | | - Abdallah A. El-Shebly
- National Institute of Oceanography and Fisheries, 101 Kaser El Aini Street, Cairo, Egypt
| | - Deyu Pan
- Internal Medicine, Charles Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059
| | - Edward Assanah
- Research, Charles Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059
| | - Greg Lawson
- Pathology and Laboratory Medicine, Division of Laboratory Animal Medicine, David Geffen School of Medicine at UCLA, 724 Westwood Blvd., Seventh Floor, Los Angeles, CA 90095
| |
Collapse
|
78
|
Yang Y, Tian S, Brown B, Chen P, Hu H, Xia L, Zhang J, Cai X, Chen Z, Pan X, Ge J, Yu K, Zhuang J. The Rb1 gene inhibits the viability of retinoblastoma cells by regulating homologous recombination. Int J Mol Med 2013; 32:137-43. [PMID: 23670186 DOI: 10.3892/ijmm.2013.1374] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/09/2013] [Indexed: 11/06/2022] Open
Abstract
Retinoblastoma is a childhood ocular tumor caused by the inactivation of both alleles of the retinoblastoma gene (Rb1). Without Rb1 gene function, chromosomal aberrations are observed in retinoblastoma cells. The instability of the genome is closely associated with the repair of DNA double-strand breaks (DSBs). However, the precise molecular mechanism of action of Rb1 in DNA DSB repair remains unclear. Thus, in this study, we aimed to investigate whether the Rb1 gene affects DNA stability by assaying DNA DSB repair and also whether it regulates the proliferation of retinoblastoma cells. Rb1 immunofluorescence and RT-PCR were performed, demonstrating that the Rb1 gene is silenced in SO-Rb50 retinoblastoma cells, and the karyotype analysis of SO-Rb50 cells indicated that the loss of Rb1 function led to genomic instability; both numerical and structural chromosomal aberrations were observed in our study. In addition, the DNA DSB repair efficiency of the SO-Rb50 cells was measured by γ-H2AX immunofluorescence, a commonly used in situ marker of DNA DSBs, following exposure to ionizing radiation (IR) (2.5 and 5.0 Gy). We found that the DNA repair efficiency was significantly increased following IR-induced damage (P<0.01). However, there was no significant difference in DNA repair efficiency between the cells expressing exogenous Rb1 and the control (P>0.05). The assay for the screening of the effect of Rb1 on the sub-pathway of DNA DSB repair, non-homologous end joining (NHEJ) and homologous recombination (HR), indicated that Rb1 did not affect NHEJ activity, although it significantly promoted the HR pathway (HR levels increased by 2.46-fold) compared with the control (P<0.01). Furthermore, we found that the cell viability of the SO-Rb50 cells transfected with exogenous Rb1 was significantly inhibited (P<0.01) and cell cycle assay indicated that exogenous Rb1 induced S phase arrest (P<0.001) which also inhibited the proliferation of retinoblastoma cells (SO-Rb50) in vitro. Therefore, this study provides new insight into the mechanisms of action of the Rb1 gene in regulating the proliferation of retinoblastoma cells.
Collapse
Affiliation(s)
- Ying Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Ghoneum M, Badr El-Din NK, Abdel Fattah SM, Tolentino L. Arabinoxylan rice bran (MGN-3/Biobran) provides protection against whole-body γ-irradiation in mice via restoration of hematopoietic tissues. JOURNAL OF RADIATION RESEARCH 2013; 54:419-29. [PMID: 23287771 PMCID: PMC3650744 DOI: 10.1093/jrr/rrs119] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The aim of the current study is to examine the protective effect of MGN-3 on overall maintenance of hematopoietic tissue after γ-irradiation. MGN-3 is an arabinoxylan from rice bran that has been shown to be a powerful antioxidant and immune modulator. Swiss albino mice were treated with MGN-3 prior to irradiation and continued to receive MGN-3 for 1 or 4 weeks. Results were compared with mice that received radiation (5 Gy γ rays) only, MGN-3 (40 mg/kg) only and control mice (receiving neither radiation nor MGN-3). At 1 and 4 weeks post-irradiation, different hematological, histopathological and biochemical parameters were examined. Mice exposed to irradiation alone showed significant depression in their complete blood count (CBC) except for neutrophilia. Additionally, histopathological studies showed hypocellularity of their bone marrow, as well as a remarkable decrease in splenic weight/relative size and in number of megakaryocytes. In contrast, pre-treatment with MGN-3 resulted in protection against irradiation-induced damage to the CBC parameters associated with complete bone marrow cellularity, as well as protection of the aforementioned splenic changes. Furthermore, MGN-3 exerted antioxidative activity in whole-body irradiated mice, and provided protection from irradiation-induced loss of body and organ weight. In conclusion, MGN-3 has the potential to protect progenitor cells in the bone marrow, which suggests the possible use of MGN-3/Biobran as an adjuvant treatment to counteract the severe adverse side effects associated with radiation therapy.
Collapse
Affiliation(s)
- Mamdooh Ghoneum
- Department of Otolaryngology, Charles Drew University of Medicine and Science, Los Angeles, California 90059, USA.
| | | | | | | |
Collapse
|
80
|
Ji G, Yan L, Liu W, Huang C, Gu A, Wang X. Polymorphisms in double-strand breaks repair genes are associated with impaired fertility in Chinese population. Reproduction 2013; 145:463-70. [PMID: 23630330 DOI: 10.1530/rep-12-0370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The DNA double-strand breaks (DSBs) repair pathway plays a critical role in repairing double-strand breaks, and genetic variants in DSBs repair pathway genes are potential risk factors for various diseases. To test the hypothesis that polymorphisms in DSBs genes are associated with susceptibility to male infertility, we examined 11 single nucleotide polymorphisms in eight key DSBs genes (XRCC3, XRCC2, BRCA2, RAG1, XRCC5, LIG4, XRCC4 and ATM) in 580 infertility cases and 580 controls from a Chinese population-based case-control study (NJMU Infertility Study). Genotypes were determined using the OpenArray platform, and sperm DNA fragmentation was detected using the TUNEL assay. The adjusted odds ratio (OR) and 95% CI were estimated using logistic regression. The results indicate that LIG4 rs1805388 (Ex2+54C>T, Thr9Ile) T allele could increase the susceptibility to male infertility (adjusted OR=2.78; 95% CI, 1.77-4.36 for TT genotype; and adjusted OR=1.58; 95% CI, 1.77-4.36 for TC genotype respectively). In addition, the homozygous variant genotype GG of RAG1 rs2227973 (A>G, K820R) was associated with a significantly increased risk of male infertility (adjusted OR, 1.44; 95% CI, 1.01-2.04). Moreover, linear regression analysis revealed that carriers of LIG4 rs1805388 or RAG1 rs2227973 variants had a significantly higher level of sperm DNA fragmentation and that T allele carriers of LIG4 rs1805388 also had a lower level of sperm concentration when compared with common homozygous genotype carriers. This study demonstrates, for the first time, to our knowledge, that functional variants of RAG1 rs2227973 and LIG4 rs1805388 are associated with susceptibility to male infertility.
Collapse
Affiliation(s)
- Guixiang Ji
- Nanjing Institute of Environmental Sciences/Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Environmental Protection, Nanjing 210042, People's Republic of China
| | | | | | | | | | | |
Collapse
|
81
|
Masunaga SI, Sakurai Y, Tanaka H, Suzuki M, Kondo N, Narabayashi M, Maruhashi A, Ono K. Wortmannin efficiently suppresses the recovery from radiation-induced damage in pimonidazole-unlabeled quiescent tumor cell population. JOURNAL OF RADIATION RESEARCH 2013; 54:221-9. [PMID: 23097299 PMCID: PMC3589932 DOI: 10.1093/jrr/rrs094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Labeling of proliferating (P) cells in mice bearing EL4 tumors was achieved by continuous administration of 5-bromo-2'-deoxyuridine (BrdU). Tumors were irradiated with γ-rays at 1 h after pimonidazole administration followed by caffeine or wortmannin treatment. Twenty-four hours later, assessment of the responses of quiescent (Q) and total (= P + Q) cell populations were based on the frequencies of micronucleation and apoptosis using immunofluorescence staining for BrdU. The response of the pimonidazole-unlabeled tumor cell fractions was assessed by means of apoptosis frequency using immunofluorescence staining for pimonidazole. The pimonidazole-unlabeled cell fraction showed significantly enhanced radio-sensitivity compared with the whole cell fraction more remarkably in Q cells than total cells. However, a significantly greater decrease in radio-sensitivity in the pimonidazole-unlabeled than the whole cell fraction, evaluated using an assay performed 24 hours after irradiation, was more clearly observed in Q cells than total cells. In both the pimonidazole-unlabeled and the whole cell fractions, wortmannin efficiently suppressed the reduction in sensitivity due to delayed assay. Wortmannin combined with γ-ray irradiation is useful for suppressing the recovery from radiation-induced damage especially in the pimonidazole-unlabeled cell fraction within the total and Q tumor cell populations.
Collapse
Affiliation(s)
- Shin-Ichiro Masunaga
- Particle Radiation Oncology Research Center, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Dolan D, Nelson G, Zupanic A, Smith G, Shanley D. Systems modelling of NHEJ reveals the importance of redox regulation of Ku70/80 in the dynamics of dna damage foci. PLoS One 2013; 8:e55190. [PMID: 23457464 PMCID: PMC3566652 DOI: 10.1371/journal.pone.0055190] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 12/19/2012] [Indexed: 12/21/2022] Open
Abstract
The presence of DNA double-stranded breaks in a mammalian cell typically activates the Non-Homologous End Joining (NHEJ) pathway to repair the damage and signal to downstream systems that govern cellular decisions such as apoptosis or senescence. The signalling system also stimulates effects such as the generation of reactive oxygen species (ROS) which in turn feed back into the damage response. Although the overall process of NHEJ is well documented, we know little of the dynamics and how the system operates as a whole. We have developed a computational model which includes DNA Protein Kinase (DNA-PK) dependent NHEJ (D-NHEJ) and back-up NHEJ mechanisms (B-NHEJ) and use it to explain the dynamic response to damage induced by different levels of gamma irradiation in human fibroblasts. Our work suggests that the observed shift from fast to slow repair of DNA damage foci at higher levels of damage cannot be explained solely by inherent stochasticity in the NHEJ system. Instead, our model highlights the importance of Ku oxidation which leads to increased Ku dissociation rates from DNA damage foci and shifts repair in favour of the less efficient B-NHEJ system.
Collapse
Affiliation(s)
- David Dolan
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | | | | | | | | |
Collapse
|
83
|
Masunaga SI, Sakurai Y, Tanaka H, Suzuki M, Kondo N, Narabayashi M, Tano K, Maruhashi A, Ono K. Usefulness of Daily Fractionated Administration of Wortmannin Combined With γ-Ray Irradiation in Terms of Local Tumor Response and Lung Metastasis. World J Oncol 2013; 4:26-36. [PMID: 29147327 PMCID: PMC5649916 DOI: 10.4021/wjon640w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2013] [Indexed: 11/30/2022] Open
Abstract
Background To evaluate the usefulness of fractionated administration of wortmannin combined with γ-ray irradiation in terms of local tumor response and lung metastatic potential, referring to the response of intratumor quiescent (Q) cells. Methods B16-BL6 melanoma tumor-bearing C57BL/6 mice were continuously given 5-bromo-2’-deoxyuridine (BrdU) to label all proliferating (P) cells. The tumor-bearing mice then received γ-ray irradiation after wortmannin treatment through a single or 4 consecutive daily intraperitoneal administrations up to a total dose of 4 mg/kg in combination with an acute hypoxia-releasing agent (nicotinamide) or mild temperature hyperthermia (MTH). Immediately after the irradiation, cells from some tumors were isolated and incubated with a cytokinesis blocker. The responses of the Q and total (= P + Q) cell populations were assessed based on the frequency of micronuclei using immunofluorescence staining for BrdU. In other tumor-bearing mice, 17 days after irradiation, macroscopic lung metastases were enumerated. Results Wortmannin raised the sensitivity of Q cells more remarkably than the total cell population in both single and daily administrations. Daily administration of wortmannin elevated the sensitivity of both the total and Q cell populations, but especially the total cell population, compared with single administration. Daily administration, especially combined with MTH, decreased the number of lung metastases. Conclusion Daily fractionated administration of wortmannin in combination with γ-ray irradiation was thought to be more promising than single administration because of its potential to enhance local tumor response and repress lung metastatic potential.
Collapse
Affiliation(s)
- Shin-Ichiro Masunaga
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Yoshinori Sakurai
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Hiroki Tanaka
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Minoru Suzuki
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Natsuko Kondo
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Masaru Narabayashi
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Keizo Tano
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Akira Maruhashi
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| | - Koji Ono
- Radiation Life and Medical Science Research Division, Research Reactor Institute, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka, 590-0494, Japan
| |
Collapse
|
84
|
Current evidence on the relationship between three polymorphisms in the XRCC7 gene and cancer risk. Mol Biol Rep 2012; 40:81-6. [PMID: 23108991 DOI: 10.1007/s11033-012-2018-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 10/01/2012] [Indexed: 01/20/2023]
Abstract
Inconsistency of the association of polymorphisms of XRCC7 with cancer is noted. Three commonly studied XRCC7 polymorphisms including rs7003908 (T>G), rs7830743 (A>G), and rs10109984 (T>C) were selected to explore their association with risk of development of cancer by meta-analysis of published case-control studies. The results showed that no significant associations with cancer risk were found in any model in terms of rs7003908, rs7830743 and rs10109984 when all studies were pooled into the meta-analysis. But when stratified by cancer type, statistically significantly elevated cancer risk was only found in prostate cancer for rs7003908 (GG vs. TT: OR = 1.845, 95 % CI = 1.178-2.888; dominant model: OR = 1.423, 95 % CI = 1.050-1.929; recessive model: OR = 1.677, 95 % CI = 1.133-2.482). In the subgroup analysis by ethnicity or study design, no significantly increased risks were found for all three polymorphisms. This meta-analysis suggests that XRCC7 rs7003908 polymorphism may contribute to cancer susceptibility for prostate cancer, which is recommended to be included in future large-sample studies and functional assays.
Collapse
|
85
|
Small-molecule inhibitors of DNA damage-repair pathways: an approach to overcome tumor resistance to alkylating anticancer drugs. Future Med Chem 2012; 4:1093-111. [PMID: 22709253 DOI: 10.4155/fmc.12.58] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A major challenge in the future development of cancer therapeutics is the identification of biological targets and pathways, and the subsequent design of molecules to combat the drug-resistant cells hiding in virtually all cancers. This therapeutic approach is justified based upon the limited advances in cancer cures over the past 30 years, despite the development of many novel chemotherapies and earlier detection, which often fail due to drug resistance. Among the various targets to overcome tumor resistance are the DNA repair systems that can reverse the cytotoxicity of many clinically used DNA-damaging agents. Some progress has already been made but much remains to be done. We explore some components of the DNA-repair process, which are involved in repair of alkylation damage of DNA, as targets for the development of novel and effective molecules designed to improve the efficacy of existing anticancer drugs.
Collapse
|
86
|
Baxter S, Lambert AR, Kuhar R, Jarjour J, Kulshina N, Parmeggiani F, Danaher P, Gano J, Baker D, Stoddard BL, Scharenberg AM. Engineering domain fusion chimeras from I-OnuI family LAGLIDADG homing endonucleases. Nucleic Acids Res 2012; 40:7985-8000. [PMID: 22684507 PMCID: PMC3439895 DOI: 10.1093/nar/gks502] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although engineered LAGLIDADG homing endonucleases (LHEs) are finding increasing applications in biotechnology, their generation remains a challenging, industrial-scale process. As new single-chain LAGLIDADG nuclease scaffolds are identified, however, an alternative paradigm is emerging: identification of an LHE scaffold whose native cleavage site is a close match to a desired target sequence, followed by small-scale engineering to modestly refine recognition specificity. The application of this paradigm could be accelerated if methods were available for fusing N- and C-terminal domains from newly identified LHEs into chimeric enzymes with hybrid cleavage sites. Here we have analyzed the structural requirements for fusion of domains extracted from six single-chain I-OnuI family LHEs, spanning 40–70% amino acid identity. Our analyses demonstrate that both the LAGLIDADG helical interface residues and the linker peptide composition have important effects on the stability and activity of chimeric enzymes. Using a simple domain fusion method in which linker peptide residues predicted to contact their respective domains are retained, and in which limited variation is introduced into the LAGLIDADG helix and nearby interface residues, catalytically active enzymes were recoverable for ∼70% of domain chimeras. This method will be useful for creating large numbers of chimeric LHEs for genome engineering applications.
Collapse
Affiliation(s)
- Sarah Baxter
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
McAlpine JN, Porter H, Köbel M, Nelson BH, Prentice LM, Kalloger SE, Senz J, Milne K, Ding J, Shah SP, Huntsman DG, Gilks CB. BRCA1 and BRCA2 mutations correlate with TP53 abnormalities and presence of immune cell infiltrates in ovarian high-grade serous carcinoma. Mod Pathol 2012; 25:740-50. [PMID: 22282309 DOI: 10.1038/modpathol.2011.211] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We characterized BRCA1 and BRCA2 status (mutation/methylation) in a consecutive series of cases of ovarian carcinoma in order to identify differences in clinicopathological features, molecular characteristics, and outcome between the pelvic high-grade serous cancers with (i) germline or somatic mutations in BRCA1 or BRCA2, (ii) methylation of BRCA1, and (iii) normal BRCA1 or BRCA2. In all, 131 women were identified prospectively, who were undergoing surgical staging and agreed to germline testing for BRCA1 and BRCA2 mutations. Histopathology, germline and somatic BRCA1 or BRCA2 mutations, BRCA1 methylation, and BRCA1 and BRCA2 mRNA expression levels distinguished four subgroups. In all, 103 cases were high-grade serous carcinoma and of these 31 (30%) had germline or somatic BRCA1 or BRCA2 mutations (20% BRCA1 and 10% BRCA2) (group 1), 21 (20%) had methylation of BRCA1 (group 2), and in 51 (50%) there was no BRCA loss (group 3). Group 4 consisted of 28 cases of non-high-grade serous, none of which had BRCA loss. BRCA1 and BRCA2 mRNA expression levels correlated with designated group (P=0.0008). Among high-grade serous carcinomas, there were no differences between groups 1-3 with respect to stage, ascites, CA125 level, platinum sensitivity, cytoreduction rate, neoadjuvant chemotherapy, or survival. Tumors with BRCA1 or BRCA2 mutations had increased immune infiltrates (CD20 and TIA-1) compared with high-grade serous without mutations (P=0.034, 0.027). TP53 expression differed between groups (P<0.0001), with abnormal TP53 expression in 49/50 tumors from groups 1 and 2. Wild-type TP53 expression was associated with worse outcome in high-grade serous (P<0.001). BRCA loss (mutation/methylation) is a common event in the pelvic high-grade serous (50%). TP53 abnormalities and increased immune cell infiltrates are significantly more common in high-grade serous with germline and somatic mutations in BRCA1 or BRCA2, compared with tumors lacking BRCA abnormalities.
Collapse
Affiliation(s)
- Jessica N McAlpine
- Department of Gynecology and Obstetrics, Division of Gynecologic Oncology, University of British Columbia, Vancouver, BC, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Barnes DR, Antoniou AC. Unravelling modifiers of breast and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers: update on genetic modifiers. J Intern Med 2012; 271:331-43. [PMID: 22443199 DOI: 10.1111/j.1365-2796.2011.02502.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pathogenic mutations in the tumour suppressor genes BRCA1 and BRCA2 confer increased risks for breast and ovarian cancer and account for approximately 15% of the excess familial risk of breast cancer amongst first-degree relatives of patients with breast cancer. There is considerable evidence indicating that these risks vary by other genetic and environmental factors clustering in families. In the past few years, based on the availability of genome-wide association data and samples from large collaborative studies, several common alleles have been found to modify breast or ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. These common alleles explain a small proportion of the genetic variability in breast or ovarian cancer risk for mutation carriers, suggesting more modifiers remain to be identified. We review the so far identified genetic modifiers of breast and ovarian cancer risk and consider the implications for risk prediction. BRCA1 and BRCA2 mutation carriers could be some of the first to benefit from clinical applications of common variants identified through genome-wide association studies. However, to be able to provide more individualized risk estimates, it will be important to understand how the associations vary with different tumour characteristics and their interactions with other genetic and environmental modifiers.
Collapse
Affiliation(s)
- D R Barnes
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
89
|
Wang W, Gao Y, Yan F, Wang M, Hu F, Wang D, Cao Q, Qin C, Yin C, Zhang Z, Pan X. Association of Ku70 A-31G polymorphism and risk of renal cell carcinoma in a Chinese population. DNA Cell Biol 2012; 31:1314-20. [PMID: 22455395 DOI: 10.1089/dna.2011.1540] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The DNA repair gene Ku70 plays a key role in the DNA double-strand breaks (DSBs) repair system. Defects in DSBs repair capacity can lead to genomic instability. We hypothesized that the Ku70 A-31G polymorphism (rs132770) was associated with the risk of renal cell carcinoma (RCC). In a hospital-based case-control study of 620 RCC patients and 623 cancer-free controls frequency matched by age and sex, we genotyped the functional polymorphism Ku70 A-31G (rs132770). Thirty-eight normal renal tissue samples with different genotypes were tested to estimate the Ku70 mRNA expression by real-time quantitative reverse transcription. Compared with the GG genotype, the GA and GA/AA genotypes had a significantly decreased risk of RCC [adjusted odds ratio (OR) = 0.62, 95% confidence interval (CI) = 0.44-0.87 for GA, and OR = 0.62, 95% CI = 0.45-0.86 for GA/AA]. The in vivo experiments with normal renal tissues revealed that a statistically significantly higher Ku70 mRNA expression was identified in samples with GA/AA genotypes compared with those with GG genotypes (p = 0.001). These results suggested that the Ku70 A-31G polymorphism is involved in the etiology of RCC and, thus, may be a marker for genetic susceptibility to RCC in the Chinese populations.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Chiruvella KK, Sebastian R, Sharma S, Karande AA, Choudhary B, Raghavan SC. Time-Dependent Predominance of Nonhomologous DNA End-Joining Pathways during Embryonic Development in Mice. J Mol Biol 2012; 417:197-211. [DOI: 10.1016/j.jmb.2012.01.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 01/18/2012] [Accepted: 01/20/2012] [Indexed: 12/26/2022]
|
91
|
Wei S, Xiong M, Zhan DQ, Liang BY, Wang YY, Gutmann DH, Huang ZY, Chen XP. Ku80 functions as a tumor suppressor in hepatocellular carcinoma by inducing S-phase arrest through a p53-dependent pathway. Carcinogenesis 2012; 33:538-47. [PMID: 22226916 DOI: 10.1093/carcin/bgr319] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Ku80 is a component of the protein complex called DNA-dependent protein kinase, which is involved in DNA double-strand break repair and multiple other functions. Previous studies revealed that Ku80 haplo-insufficient and poly (adenosine diphosphate-ribose) polymerase-null transgenic mice developed hepatocellular carcinoma (HCC) at a high frequency. The role of Ku80 has never been investigated in human HCC. Ku80 expressions in HCC and adjacent liver tissue were investigated by using immunohistochemical staining and western blot. Ku80 was transfected into a Ku80-deficient HCC cell line SMMC7721 cells, and the growth features of the Ku80-expressing cells and vector-transfected cells were studied both in vitro and in vivo. Cell cycle analysis and RNA interference were employed to investigate the mechanisms underlying the growth regulation associated with Ku80 expression. Ku80 was found frequently downregulated in HCC compared with adjacent liver tissue. Ku80 downregulation was significantly correlated with elevated hepatitis B virus-DNA load and severity of liver cirrhosis. Overexpression of Ku80 in SMMC7721 cells significantly suppressed cell proliferation in vitro and in vivo. Ku80 overexpression caused S-phase cell cycle arrest and was associated with upregulation of p53 and p21(CIP1/WAF1), and the inhibition of p53 or p21(CIP1/WAF1) expression by RNA interference overcame the growth suppression and S-phase arrest in the Ku80-expressing cells. A novel mechanism was revealed that Ku80 functions as a tumor suppressor in HCC by inducing S-phase arrest through a p53-dependent pathway.
Collapse
Affiliation(s)
- Shuang Wei
- Research Laboratory and Hepatic Surgical Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Da Dao, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Morgenroth A, Vogg AT, Mottaghy FM, Schmaljohann J. Targeted endoradiotherapy using nucleotides. Methods 2011; 55:203-14. [PMID: 21782950 DOI: 10.1016/j.ymeth.2011.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/24/2011] [Accepted: 06/28/2011] [Indexed: 11/15/2022] Open
Abstract
Increased cellular proliferation is an integral part of the cancer phenotype. Hence, the sustained and continued demand on supply of DNA building blocks during the DNA replication presents a potential target for therapeutic intervention. For this propose, the α and Auger electron emitting nucleotides analogs are attractive for targeted endoradiotherapy, given that DNA of malignant cells is selectively addressed. This review summarizes development and preclinical and clinical studies of endoradiotherapeutic acting nucleoside analogs with a special focus on thymidine analogs.
Collapse
Affiliation(s)
- Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH, Pauwelsstraße 30, D-52074 Aachen, Germany.
| | | | | | | |
Collapse
|
93
|
Tsai YS, Lin CS, Chiang SL, Lee CH, Lee KW, Ko YC. Areca nut induces miR-23a and inhibits repair of DNA double-strand breaks by targeting FANCG. Toxicol Sci 2011; 123:480-90. [PMID: 21750350 DOI: 10.1093/toxsci/kfr182] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Previous investigations have shown that areca nut extracts (ANE) or arecoline (ARE) causes DNA damage, which in turn contributes to oral cell carcinogenesis. To understand the role of microRNA (miRNA) in ANE-associated carcinogenesis, miRNA expression profile was examined in ANE-treated normal human oral fibroblasts. Among the miRNAs changed by ANE exposure, we found that ANE-induced miR-23a overexpression was correlated with an increase of γ-H2AX, a DNA damage marker. In addition, DNA double-strand breaks (DSB) repair that was determined by an in vivo plasmid-based assay was reduced in ANE-treated or miR-23a-overexpressed cells, suggesting the role of miR-23a in DSB repair. FANCG is one of Fanconi anemia susceptibility genes that participate in DSB repair pathway to prevent chromosomal aberrations. FANCG was predicted as a candidate target of miR-23a by TargetScan algorithm. This was confirmed by ectopic overexpression or knockdown of miR-23a. The correlation between miR-23a overexpression and areca nut-chewing habit could also be found in oral cancer patients. Finally, we showed that ANE-induced/ARE-induced miRNAs were significantly associated with the functional categories of "genetic disorders" and "cancer" using network-based analyses. In conclusion, our data showed for the first time that ANE-induced miR-23a was correlated with a reduced FANCG expression and DSB repair, which might contribute to ANE-associated human malignancies.
Collapse
Affiliation(s)
- Yi-Shan Tsai
- Center of Excellence for Environmental Medicine, Kaohsiung Medical University, No. 100, Shi-Chuan 1stRoad, Kaohsiung 807, Taiwan
| | | | | | | | | | | |
Collapse
|
94
|
Magae J, Furukawa C, Ogata H. Dose-Rate Effect on Proliferation Suppression in Human Cell Lines Continuously Exposed to γ Rays. Radiat Res 2011; 176:447-58. [DOI: 10.1667/rr2408.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Junji Magae
- Institute of Research and Innovation, 1201 Takada, Kashiwa 277-0861, Japan
| | - Chiharu Furukawa
- Institute of Research and Innovation, 1201 Takada, Kashiwa 277-0861, Japan
| | - Hiromitsu Ogata
- National Institute of Public Health, 2-3-6, Minami, Wako, Saitama 351-0197 Japan
| |
Collapse
|
95
|
Mungenast AE, Tsai LH. Addressing the complex etiology of Alzheimer’s disease: the role of p25/Cdk5. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder characterized by the progressive loss of forebrain neurons and the deterioration of learning and memory. Therapies for AD have primarily focused upon either the inhibition of amyloid synthesis or its deposition in the brain, but clinical testing to date has not yet found an effective amelioration of cognitive symptoms. Synaptic loss closely correlates with the degree of dementia in AD patients. However, mouse AD models that target the amyloid-β pathway generally do not exhibit a profound loss of synapses, despite extensive synaptic dysfunction. The increased generation of p25, an activator of the cyclin-dependent kinase 5 (Cdk5) has been found in both human patients and mouse models of neurodegeneration. The current work reviews our knowledge, to date, on the role of p25/Cdk5 in Alzheimer’s disease, with a focus upon the interaction of amyloid-β and p25/Cdk5 in synaptic dysfunction and neuronal loss.
Collapse
Affiliation(s)
- Alison E Mungenast
- Picower Institute for Learning & Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li-Huei Tsai
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
96
|
Vollebergh MA, Lips EH, Nederlof PM, Wessels LFA, Schmidt MK, van Beers EH, Cornelissen S, Holtkamp M, Froklage FE, de Vries EGE, Schrama JG, Wesseling J, van de Vijver MJ, van Tinteren H, de Bruin M, Hauptmann M, Rodenhuis S, Linn SC. An aCGH classifier derived from BRCA1-mutated breast cancer and benefit of high-dose platinum-based chemotherapy in HER2-negative breast cancer patients. Ann Oncol 2011; 22:1561-1570. [PMID: 21135055 PMCID: PMC3121967 DOI: 10.1093/annonc/mdq624] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/07/2010] [Accepted: 09/14/2010] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Breast cancer cells deficient for BRCA1 are hypersensitive to agents inducing DNA double-strand breaks (DSB), such as bifunctional alkylators and platinum agents. Earlier, we had developed a comparative genomic hybridisation (CGH) classifier based on BRCA1-mutated breast cancers. We hypothesised that this BRCA1-like(CGH) classifier could also detect loss of function of BRCA1 due to other causes besides mutations and, consequently, might predict sensitivity to DSB-inducing agents. PATIENTS AND METHODS We evaluated this classifier in stage III breast cancer patients, who had been randomly assigned between adjuvant high-dose platinum-based (HD-PB) chemotherapy, a DSB-inducing regimen, and conventional anthracycline-based chemotherapy. Additionally, we assessed BRCA1 loss through mutation or promoter methylation and immunohistochemical basal-like status in the triple-negative subgroup (TN subgroup). RESULTS We observed greater benefit from HD-PB chemotherapy versus conventional chemotherapy among patients with BRCA1-like(CGH) tumours [41/230 = 18%, multivariate hazard ratio (HR) = 0.12, 95% confidence interval (CI) 0.04-0.43] compared with patients with non-BRCA1-like(CGH) tumours (189/230 = 82%, HR = 0.78, 95% CI 0.50-1.20), with a significant difference (test for interaction P = 0.006). Similar results were obtained for overall survival (P interaction = 0.04) and when analyses were restricted to the TN subgroup. Sixty-three percent (20/32) of assessable BRCA1-like(CGH) tumours harboured either a BRCA1 mutation (n = 8) or BRCA1 methylation (n = 12). CONCLUSION BRCA1 loss as assessed by CGH analysis can identify patients with substantially improved outcome after adjuvant DSB-inducing chemotherapy when compared with standard anthracycline-based chemotherapy in our series.
Collapse
Affiliation(s)
- M A Vollebergh
- Division of Molecular Biology; Division of Medical Oncology
| | - E H Lips
- Division of Experimental Therapy
| | - P M Nederlof
- Division of Experimental Therapy; Division of Molecular Pathology
| | - L F A Wessels
- Department of Bioinformatics and Statistics, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam; Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft
| | - M K Schmidt
- Division of Experimental Therapy; Department of Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| | | | | | | | | | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen
| | | | | | - M J van de Vijver
- Department of Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam; Department of Pathology, Academic Medical Center
| | - H van Tinteren
- Department of Biometrics, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - M Hauptmann
- Department of Bioinformatics and Statistics, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| | | | - S C Linn
- Division of Molecular Biology; Division of Medical Oncology.
| |
Collapse
|
97
|
Qi L, Liu CY, Wu WQ, Gu ZL, Guo CY. Protective effect of flavonoids from Astragalus complanatus on radiation induced damages in mice. Fitoterapia 2011; 82:383-92. [DOI: 10.1016/j.fitote.2010.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/09/2010] [Accepted: 11/09/2010] [Indexed: 02/06/2023]
|
98
|
Yan H, Toczylowski T, McCane J, Chen C, Liao S. Replication protein A promotes 5'-->3' end processing during homology-dependent DNA double-strand break repair. ACTA ACUST UNITED AC 2011; 192:251-61. [PMID: 21263027 PMCID: PMC3172182 DOI: 10.1083/jcb.201005110] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The single-strand DNA–binding protein RPA promotes 5′-strand resection to generate 3′ single strands for homology-dependent DNA double-strand repair. Replication protein A (RPA), the eukaryotic single-strand deoxyribonucleic acid (DNA [ss-DNA])–binding protein, is involved in DNA replication, nucleotide damage repair, mismatch repair, and DNA damage checkpoint response, but its function in DNA double-strand break (DSB) repair is poorly understood. We investigated the function of RPA in homology-dependent DSB repair using Xenopus laevis nucleoplasmic extracts as a model system. We found that RPA is required for single-strand annealing, one of the homology-dependent DSB repair pathways. Furthermore, RPA promotes the generation of 3′ single-strand tails (ss-tails) by stimulating both the Xenopus Werner syndrome protein (xWRN)–mediated unwinding of DNA ends and the subsequent Xenopus DNA2 (xDNA2)–mediated degradation of the 5′ ss-tail. Purified xWRN, xDNA2, and RPA are sufficient to carry out the 5′-strand resection of DNA that carries a 3′ ss-tail. These results provide strong biochemical evidence to link RPA to a specific DSB repair pathway and reveal a novel function of RPA in the generation of 3′ ss-DNA for homology-dependent DSB repair.
Collapse
Affiliation(s)
- Hong Yan
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | | | |
Collapse
|
99
|
Evaluating the radioprotective effect of hesperidin in the liver of Swiss albino mice. Eur J Pharmacol 2011; 658:206-12. [PMID: 21371459 DOI: 10.1016/j.ejphar.2011.02.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 02/04/2023]
Abstract
The present study was aimed to evaluate the radioprotective efficacy of hesperidin, a flavonone glycoside against X-ray radiation-induced cellular damage in the liver of Swiss albino mice. The first phase of the study was carried out to fix the effective concentration of hesperidin by performing a 30 days of survival studies using different graded doses [12.5, 25, 50 and 100mg/kg body weight] of hesperidin administered orally to mice via intragastric intubations for seven consecutive days prior to exposure of whole body radiation (10 Gy). Based on the results of survival studies, the effective dose of hesperidin was fixed which was then administered to animals orally via intragastric intubations for seven consecutive days prior to exposure of whole body radiation (4 Gy) to evaluate its radioprotective efficacy by performing various biochemical estimations, comet assay, DNA fragmentation assay and histopathological studies in the liver of Swiss albino mice. The results indicated that radiation-induced decrease in the levels of endogenous antioxidant enzymes and increase in lipid peroxidative index, DNA damage and comet parameters were altered by pre-administration with the effective dose of hesperidin [25mg/kg body weight] which restored the antioxidant status to near normal and decreased the levels of lipid peroxidative index, DNA damage and comet parameters. These results were further confirmed by histopathological examinations which indicated that pre-administration with the effective dose of hesperidin reduced the hepatic damage induced by radiation. Thus the current study shows hesperidin to be an effective radioprotector against radiation induced damage in the liver of mice.
Collapse
|
100
|
Coleman KA, Greenberg RA. The BRCA1-RAP80 complex regulates DNA repair mechanism utilization by restricting end resection. J Biol Chem 2011; 286:13669-80. [PMID: 21335604 DOI: 10.1074/jbc.m110.213728] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The tumor suppressor protein BRCA1 is a constituent of several different protein complexes and is required for homology-directed repair (HDR) of DNA double strand breaks (DSBs). The most recently discovered BRCA1-RAP80 complex is recruited to ubiquitin structures on chromatin surrounding the break. Deficiency of any member of this complex confers hypersensitivity to DNA-damaging agents by undefined mechanisms. In striking contrast to other BRCA1-containing complexes that are known to promote HDR, we demonstrate that the BRCA1-RAP80 complex restricts end resection in S/G(2) phase of the cell cycle, thereby limiting HDR. RAP80 or BRCC36 deficiency resulted in elevated Mre11-CtIP-dependent 5' end resection with a concomitant increase in HDR mechanisms that rely on 3' single-stranded overhangs. We propose a model in which the BRCA1-RAP80 complex limits nuclease accessibility to DSBs, thus preventing excessive end resection and potentially deleterious homology-directed DSB repair mechanisms that can impair genome integrity.
Collapse
Affiliation(s)
- Kara A Coleman
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6160, USA
| | | |
Collapse
|