151
|
Runge R, Arlt J, Oehme L, Freudenberg R, Kotzerke J. Comparison of clonogenic cell survival and DNA damage induced by 188Re and X-rays in rat thyroid cells. Nuklearmedizin 2017; 56:47-54. [PMID: 27781237 DOI: 10.3413/nukmed-0842-16-08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/14/2016] [Indexed: 11/20/2022]
Abstract
AIM Ionizing radiation produces DNA lesions among which DNA double strand breaks (DSB) are the most critical events. Radiation of various energy types might differ in their biological effectiveness. Here, we compared cell survival and DNA damage induced by 188Re and X-rays using γH2AX foci as a measure of DSB. The correlation between survival and residual foci was also analyzed. METHODS PCCl3 cells were irradiated with 200 kV X-rays (1.2 Gy/min) or 0.5-25 MBq/ml 188Re (1 h irradiation) achieving doses up to 10 Gy. By blocking of sodium iodide symporter (NIS) essentially extracellular activity could be guaranteed. Survival fractions (SF) were detected by colony forming assay. Initial and residual γH2AX foci (15 min and 24 h after irradiation) were assessed by immunostaining. The relationship between SF and residual radiation induced γH2AX foci (RIF) was evaluated by Spearman and Pearson correlation tests. RESULTS We did not find significant differences between the survival curves in terms of the radiation quality. The D37 values were 4.6 Gy and 4.2 Gy for 188Re or X-ray, respectively. The initial foci numbers were in the same range for 188Re and X-ray, but higher levels of residual foci persisted after X-rays in comparison to 188Re (1 GyX-ray 6.5 ± 0.2; 1 GyRe-188 4.8 ± 0.2 RIF). Accordingly, for 188Re a higher extent of DSB repair was found. The Spearman test revealed a significant (p < 0.01) correlation between SF and residual RIF for both radiation modalities. CONCLUSION No differences in terms of radiation were found for SF and initial foci. However, residual foci were lower for 188Re than for X-rays. A prediction of SF by residual foci should consider the properties of the radiation qualities that influence foci removal and DSB repair.
Collapse
Affiliation(s)
- Roswitha Runge
- Dr. rer.medic. Roswitha Runge, Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus, Fetscherstr. 74, 01307 Dresden, Tel. +49 (0) 351 458 5481, Fax +49 (0) 351 458 5347, E-Mail:
| | | | | | | | | |
Collapse
|
152
|
Jun DW, Hwang M, Kim YH, Kim KT, Kim S, Lee CH. DDRI-9: a novel DNA damage response inhibitor that blocks mitotic progression. Oncotarget 2017; 7:17699-710. [PMID: 26848527 PMCID: PMC4951243 DOI: 10.18632/oncotarget.7135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 01/19/2016] [Indexed: 12/17/2022] Open
Abstract
The DNA damage response (DDR) is an emerging target for cancer therapy. By modulating the DDR, including DNA repair and cell cycle arrest, the efficacy of anticancer drugs can be enhanced and side effects reduced. We previously screened a chemical library and identified novel DDR inhibitors including DNA damage response inhibitor-9 (DDRI-9; 1H-Purine-2,6-dione,7-[(4-fluorophenyl)methyl]-3,7-dihydro-3-methyl-8-nitro). In this study, we characterized DDRI-9 activity and found that it inhibited phosphorylated histone variant H2AX foci formation upon DNA damage, delayed DNA repair, and enhanced the cytotoxicity of etoposide and ionizing radiation. It also reduced the foci formation of DNA repair-related proteins, including the protein kinase ataxia-telangiectasia mutated, DNA-dependent protein kinase, breast cancer type 1 susceptibility protein, and p53-binding protein 1, but excluding mediator of DNA damage checkpoint protein 1. Cell cycle analysis revealed that DDRI-9 blocked mitotic progression. Like other mitotic inhibitors, DDRI-9 treatment resulted in the accumulation of mitotic protein and induced cell death. Thus, DDRI-9 may affect both DDR signal amplification and mitotic progression. This study suggests that DDRI-9 is a good lead molecule for the development of anticancer drugs.
Collapse
Affiliation(s)
- Dong Wha Jun
- New Experimental Therapeutics Branch, Division of Convergence Technology, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Mihwa Hwang
- New Experimental Therapeutics Branch, Division of Convergence Technology, National Cancer Center, Goyang, Gyeonggi, Korea.,System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Yun-Hee Kim
- Molecular Imaging and Therapy Branch, Division of Convergence Technology, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Kyung-Tae Kim
- Molecular Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Sunshin Kim
- New Experimental Therapeutics Branch, Division of Convergence Technology, National Cancer Center, Goyang, Gyeonggi, Korea
| | - Chang-Hun Lee
- Cancer Cell and Molecular Biology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Korea
| |
Collapse
|
153
|
Feldman S, Wuerffel R, Achour I, Wang L, Carpenter PB, Kenter AL. 53BP1 Contributes to Igh Locus Chromatin Topology during Class Switch Recombination. THE JOURNAL OF IMMUNOLOGY 2017; 198:2434-2444. [PMID: 28159901 DOI: 10.4049/jimmunol.1601947] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/05/2017] [Indexed: 01/01/2023]
Abstract
In B lymphocytes, Ig class switch recombination (CSR) is induced by activation-induced cytidine deaminase, which initiates a cascade of events leading to DNA double-strand break formation in switch (S) regions. Resolution of DNA double-strand breaks proceeds through formation of S-S synaptic complexes. S-S synapsis is mediated by a chromatin loop that spans the C region domain of the Igh locus. S-S junctions are joined via a nonhomologous end joining DNA repair process. CSR occurs via an intrachromosomal looping out and deletion mechanism that is 53BP1 dependent. However, the mechanism by which 53BP1 facilitates deletional CSR and inhibits inversional switching events remains unknown. We report a novel architectural role for 53BP1 in Igh chromatin looping in mouse B cells. Long-range interactions between the Eμ and 3'Eα enhancers are significantly diminished in the absence of 53BP1. In contrast, germline transcript promoter:3'Eα looping interactions are unaffected by 53BP1 deficiency. Furthermore, 53BP1 chromatin occupancy at sites in the Igh locus is B cell specific, is correlated with histone H4 lysine 20 marks, and is subject to chromatin spreading. Thus, 53BP1 is required for three-dimensional organization of the Igh locus and provides a plausible explanation for the link with 53BP1 enforcement of deletional CSR.
Collapse
Affiliation(s)
- Scott Feldman
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612-7344; and
| | - Robert Wuerffel
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612-7344; and
| | - Ikbel Achour
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612-7344; and
| | - Lili Wang
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612-7344; and
| | - Phillip B Carpenter
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX 77030
| | - Amy L Kenter
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612-7344; and
| |
Collapse
|
154
|
Leventakos K, Tsiodras S, Kelesidis T, Kefala M, Kottaridi C, Spathis A, Gouloumi AR, Pouliakis A, Pappas A, Sioulas V, Chrelias C, Karakitsos P, Panayiotides I. γH2Ax Expression as a Potential Biomarker Differentiating between Low and High Grade Cervical Squamous Intraepithelial Lesions (SIL) and High Risk HPV Related SIL. PLoS One 2017; 12:e0170626. [PMID: 28118377 PMCID: PMC5261776 DOI: 10.1371/journal.pone.0170626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/07/2017] [Indexed: 01/10/2023] Open
Abstract
Background γH2AX is a protein biomarker for double-stranded DNA breakage; its expression was studied in cervical squamous intraepithelial lesions and carcinomas. Methods Immunostaining for phospho-γH2AX was performed in sections from histologically confirmed cervical SIL and carcinomas, as well as from normal cervices used as controls. In total, 275 cases were included in the study: 112 low grade SIL (LGSIL), 99 high grade SIL (HGSIL), 24 squamous cell carcinoma (SCC), 12 adenocarcinoma and 28 cervical specimens with no essential lesions. Correlation of histological grading, high risk vs. low risk HPV virus presence, activated vs. non-activated status (by high risk HPV mRNA expression) and γH2AX expression in both basal and surface segments of the squamous epithelium was performed. Results Gradual increase of both basal and surface γH2AX expression was noted up from normal cervices to LGSIL harboring a low risk HPV type, to LGSIL harboring a high risk virus at a non-activated state (p<0.05). Thereafter, both basal and surface γH2AX expression dropped in LGSIL harboring a high risk virus at an activated state and in HGSIL. Conclusions γH2AX could serve as a potential biomarker discriminating between LGSIL and HGSIL, as well as between LGSIL harboring high risk HPV at an activated state.
Collapse
Affiliation(s)
- Konstantinos Leventakos
- 2 Department of Pathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- 4 Department of Internal Medicine, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios Tsiodras
- 4 Department of Internal Medicine, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- * E-mail:
| | - Theodore Kelesidis
- 4 Department of Internal Medicine, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Kefala
- 2 Department of Pathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christine Kottaridi
- Department of Cytopathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris Spathis
- Department of Cytopathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Alina-Roxani Gouloumi
- 2 Department of Pathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Abraham Pouliakis
- Department of Cytopathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Asimakis Pappas
- 3 Department of Obstetrics and Gynecology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasileios Sioulas
- 3 Department of Obstetrics and Gynecology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Chrelias
- 3 Department of Obstetrics and Gynecology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Karakitsos
- Department of Cytopathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Panayiotides
- 2 Department of Pathology, University Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
155
|
Hodjat M, Baeeri M, Rezvanfar MA, Rahimifard M, Gholami M, Abdollahi M. On the mechanism of genotoxicity of ethephon on embryonic fibroblast cells. Toxicol Mech Methods 2017; 27:173-180. [DOI: 10.1080/15376516.2016.1273425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Mahshid Hodjat
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Rezvanfar
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahban Rahimifard
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
156
|
Crefcoeur RP, Zgheib O, Halazonetis TD. A Model to Investigate Single-Strand DNA Responses in G1 Human Cells via a Telomere-Targeted, Nuclease-Deficient CRISPR-Cas9 System. PLoS One 2017; 12:e0169126. [PMID: 28046023 PMCID: PMC5207518 DOI: 10.1371/journal.pone.0169126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/11/2016] [Indexed: 12/22/2022] Open
Abstract
DNA replication stress has the potential to compromise genomic stability and, therefore, cells have developed elaborate mechanisms to detect and resolve problems that may arise during DNA replication. The presence of single-stranded DNA (ssDNA) is often associated with DNA replication stress and serves as a signal for both checkpoint and repair responses. In this study, we exploited a CRISPR-Cas9 system to induce regions of ssDNA in the genome. Specifically, single-guide RNAs bearing sequence complementarity to human telomeric repeats, were used to target nuclease-deficient Cas9 (dCas9) to telomeres. Such targeting was associated with the formation of DNA-RNA hybrids, leaving one telomeric DNA strand single-stranded. This ssDNA then recruited DNA repair and checkpoint proteins, such as RPA, ATRIP, BLM and Rad51, at the telomeres. Interestingly, targeting of all these proteins to telomeric ssDNA was observed even in cells that were in the G1 phase of the cell cycle. Therefore, this system has the potential to serve as a platform for further investigation of DNA replication stress responses at specific loci in the human genome and in all phases of the cell cycle.
Collapse
Affiliation(s)
- Remco P. Crefcoeur
- Department of Molecular Biology, University of Geneva, Geneva, Switzerland
| | - Omar Zgheib
- Department of Molecular Biology, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
157
|
Vaclová T, Woods NT, Megías D, Gomez-Lopez S, Setién F, García Bueno JM, Macías JA, Barroso A, Urioste M, Esteller M, Monteiro ANA, Benítez J, Osorio A. Germline missense pathogenic variants in the BRCA1 BRCT domain, p.Gly1706Glu and p.Ala1708Glu, increase cellular sensitivity to PARP inhibitor olaparib by a dominant negative effect. Hum Mol Genet 2016; 25:5287-5299. [PMID: 27742776 DOI: 10.1093/hmg/ddw343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/03/2016] [Indexed: 12/29/2022] Open
Abstract
BRCA1-deficient cells show defects in DNA repair and rely on other members of the DNA repair machinery, which makes them sensitive to PARP inhibitors (PARPi). Although carrying a germline pathogenic variant in BRCA1/2 is the best determinant of response to PARPi, a significant percentage of the patients do not show sensitivity and/or display increased toxicity to the agent. Considering previously suggested mutation-specific BRCA1 haploinsufficiency, we aimed to investigate whether there are any differences in cellular response to PARPi olaparib depending on the BRCA1 mutation type. Lymphoblastoid cell lines derived from carriers of missense pathogenic variants in the BRCA1 BRCT domain (c.5117G > A, p.Gly1706Glu and c.5123C > A, p.Ala1708Glu) showed higher sensitivity to olaparib than cells with truncating variants or wild types (WT). Response to olaparib depended on a basal PARP enzymatic activity, but did not correlate with PARP1 expression. Interestingly, cellular sensitivity to the agent was associated with the level of BRCA1 recruitment into γH2AX foci, being the lowest in cells with missense variants. Since these variants lead to partially stable protein mutants, we propose a model in which the mutant protein acts in a dominant negative manner on the WT BRCA1, impairing the recruitment of BRCA1 into DNA damage sites and, consequently, increasing cellular sensitivity to PARPi. Taken together, our results indicate that carriers of different BRCA1 mutations could benefit from olaparib in a distinct way and show different toxicities to the agent, which could be especially relevant for a potential future use of PARPi as prophylactic agents in BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Tereza Vaclová
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Nicholas T Woods
- Eppley Institute for Research in Cancer and Allied Diseases, Molecular and Biochemical Etiology Program, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Diego Megías
- Confocal Microscopy Core Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sergio Gomez-Lopez
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fernando Setién
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | | | - José Antonio Macías
- Hereditary Cancer Unit, Medical Oncology Service, Hospital Morales Meseguer, Murcia, Spain
| | - Alicia Barroso
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Miguel Urioste
- Familial Cancer Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Alvaro N A Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, FL, USA
| | - Javier Benítez
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Spanish Network on Rare Diseases (CIBERER), Madrid, Spain.,Genotyping Unit (CEGEN), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Osorio
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Spanish Network on Rare Diseases (CIBERER), Madrid, Spain
| |
Collapse
|
158
|
Kong EY, Cheng SH, Yu KN. Zebrafish as an In Vivo Model to Assess Epigenetic Effects of Ionizing Radiation. Int J Mol Sci 2016; 17:ijms17122108. [PMID: 27983682 PMCID: PMC5187908 DOI: 10.3390/ijms17122108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/01/2016] [Accepted: 12/09/2016] [Indexed: 12/14/2022] Open
Abstract
Exposure to ionizing radiations (IRs) is ubiquitous in our environment and can be categorized into “targeted” effects and “non-targeted” effects. In addition to inducing deoxyribonucleic acid (DNA) damage, IR exposure leads to epigenetic alterations that do not alter DNA sequence. Using an appropriate model to study the biological effects of radiation is crucial to better understand IR responses as well as to develop new strategies to alleviate exposure to IR. Zebrafish, Danio rerio, is a scientific model organism that has yielded scientific advances in several fields and recent studies show the usefulness of this vertebrate model in radiation biology. This review briefly describes both “targeted” and “non-targeted” effects, describes the findings in radiation biology using zebrafish as a model and highlights the potential of zebrafish to assess the epigenetic effects of IR, including DNA methylation, histone modifications and miRNA expression. Other in vivo models are included to compare observations made with zebrafish, or to illustrate the feasibility of in vivo models when the use of zebrafish was unavailable. Finally, tools to study epigenetic modifications in zebrafish, including changes in genome-wide DNA methylation, histone modifications and miRNA expression, are also described in this review.
Collapse
Affiliation(s)
- Eva Yi Kong
- Department of Physics and Materials Science, City University of Hong Kong, Hong Kong, China.
| | - Shuk Han Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Hong Kong, China.
| | - Kwan Ngok Yu
- Department of Physics and Materials Science, City University of Hong Kong, Hong Kong, China.
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
159
|
Khonsari H, Schneider M, Al-Mahdawi S, Chianea YG, Themis M, Parris C, Pook MA, Themis M. Lentivirus-meditated frataxin gene delivery reverses genome instability in Friedreich ataxia patient and mouse model fibroblasts. Gene Ther 2016; 23:846-856. [PMID: 27518705 PMCID: PMC5143368 DOI: 10.1038/gt.2016.61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/05/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023]
Abstract
Friedreich ataxia (FRDA) is a progressive neurodegenerative disease caused by deficiency of frataxin protein, with the primary sites of pathology being the large sensory neurons of the dorsal root ganglia and the cerebellum. FRDA is also often accompanied by severe cardiomyopathy and diabetes mellitus. Frataxin is important in mitochondrial iron-sulfur cluster (ISC) biogenesis and low-frataxin expression is due to a GAA repeat expansion in intron 1 of the FXN gene. FRDA cells are genomically unstable, with increased levels of reactive oxygen species and sensitivity to oxidative stress. Here we report the identification of elevated levels of DNA double strand breaks (DSBs) in FRDA patient and YG8sR FRDA mouse model fibroblasts compared to normal fibroblasts. Using lentivirus FXN gene delivery to FRDA patient and YG8sR cells, we obtained long-term overexpression of FXN mRNA and frataxin protein levels with reduced DSB levels towards normal. Furthermore, γ-irradiation of FRDA patient and YG8sR cells revealed impaired DSB repair that was recovered on FXN gene transfer. This suggests that frataxin may be involved in DSB repair, either directly by an unknown mechanism, or indirectly via ISC biogenesis for DNA repair enzymes, which may be essential for the prevention of neurodegeneration.
Collapse
Affiliation(s)
- H Khonsari
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
- Synthetic Biology Theme, Institute of Environment, Health & Societies, Brunel University London, Uxbridge, Middlesex, UK
| | - M Schneider
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
- Synthetic Biology Theme, Institute of Environment, Health & Societies, Brunel University London, Uxbridge, Middlesex, UK
| | - S Al-Mahdawi
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
- Synthetic Biology Theme, Institute of Environment, Health & Societies, Brunel University London, Uxbridge, Middlesex, UK
| | - Y G Chianea
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
- Synthetic Biology Theme, Institute of Environment, Health & Societies, Brunel University London, Uxbridge, Middlesex, UK
| | - M Themis
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
| | - C Parris
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
| | - M A Pook
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
- Synthetic Biology Theme, Institute of Environment, Health & Societies, Brunel University London, Uxbridge, Middlesex, UK
| | - M Themis
- Division of Biosciences, Department of Life Sciences, College of Health & Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
- Synthetic Biology Theme, Institute of Environment, Health & Societies, Brunel University London, Uxbridge, Middlesex, UK
- Division of Ecology and Evolution, Department of Life Sciences, Imperial College London, London, UK
| |
Collapse
|
160
|
Liu W. Epigenetics in Schistosomes: What We Know and What We Need Know. Front Cell Infect Microbiol 2016; 6:149. [PMID: 27891322 PMCID: PMC5104962 DOI: 10.3389/fcimb.2016.00149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/28/2016] [Indexed: 01/26/2023] Open
Abstract
Schistosomes are metazoan parasites and can cause schistosomiasis. Epigenetic modifications include DNA methylation, histone modifications and non-coding RNAs. Some enzymes involved in epigenetic modification and microRNA processes have been developed as drugs to treat the disease. Compared with humans and vertebrates, an in-depth understanding of epigenetic modifications in schistosomes is starting to be realized. DNA methylation, histone modifications and non-coding RNAs play important roles in the development and reproduction of schistosomes and in interactions between the host and schistosomes. Therefore, exploring and investigating the epigenetic modifications in schistosomes will facilitate drug development and therapy for schistosomiasis. Here, we review the role of epigenetic modifications in the development, growth and reproduction of schistosomes, and the interactions between the host and schistosome. We further discuss potential epigenetic targets for drug discovery for the treatment of schistosomiasis.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science Shanghai, China
| |
Collapse
|
161
|
Gemenetzidis E, Gammon L, Biddle A, Emich H, Mackenzie IC. Invasive oral cancer stem cells display resistance to ionising radiation. Oncotarget 2016; 6:43964-77. [PMID: 26540568 PMCID: PMC4791279 DOI: 10.18632/oncotarget.6268] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/06/2015] [Indexed: 11/28/2022] Open
Abstract
There is a significant amount of evidence to suggest that human tumors are driven and maintained by a sub-population of cells, known as cancer stem cells (CSC). In the case of head and neck cancer, such cells have been characterised by high expression levels of CD44 cell surface glycoprotein, while we have previously shown the presence of two diverse oral CSC populations in vitro, with different capacities for cell migration and proliferation. Here, we examined the response of oral CSC populations to ionising radiation (IR), a front-line measure for the treatment of head and neck tumors. We show that oral CSC initially display resistance to IR-induced growth arrest as well as relative apoptotic resistance. We propose that this is a result of preferential activation of the DNA damagerepair pathway in oral CSC with increased activation of ATM and BRCA1, elevated levels of DNA repair proteins RAD52, XLF, and a significantly faster rate of DNA double-strand-breaks clearance 24 hours following IR. By visually identifying CSC sub-populations undergoing EMT, we show that EMT-CSC represent the majority of invasive cells, and are more radio-resistant than any other population in re-constructed 3D tissues. We provide evidence that IR is not sufficient to eliminate CSC in vitro, and that sensitization of CD44hi/ESAlow cells to IR, followed by secondary EMT blockade, could be critical in order to reduce primary tumor recurrence, but more importantly to be able to eradicate cells capable of invasion and distant metastasis.
Collapse
Affiliation(s)
- Emilios Gemenetzidis
- Blizard Institute Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Luke Gammon
- Blizard Institute Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Adrian Biddle
- Blizard Institute Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Helena Emich
- Blizard Institute Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ian C Mackenzie
- Blizard Institute Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
162
|
The Fra-1-miR-134-SDS22 feedback loop amplifies ERK/JNK signaling and reduces chemosensitivity in ovarian cancer cells. Cell Death Dis 2016; 7:e2384. [PMID: 27685628 PMCID: PMC5059884 DOI: 10.1038/cddis.2016.289] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/04/2016] [Accepted: 08/10/2016] [Indexed: 12/12/2022]
Abstract
The Fra-1 transcription factor is frequently upregulated in multiple types of tumors. Here we found that Fra-1 promotes miR-134 expression. miR-134 activates JNK and ERK by targeting SDS22, which in turn induces Fra-1 expression and leads to miR-134 upregulation. In addition, miR-134 augmented H2AX S139 phosphorylation by activating JNK and promoted non-homologous end joining (NHEJ)-mediated DNA repair. Therefore, ectopic miR-134 expression reduced chemosensitivity in ovarian cancer cells. Furthermore, miR-134 promotes cell proliferation, migration and invasion of ovarian cancer cells, and enhances tumor growth in vivo. Of particular significance, both Fra-1 and miR-134 are upregulated in ovarian cancer tissues, and Fra-1 and miR-134 expression is positively correlated. High levels of miR-134 expression were associated with a reduced median survival of ovarian cancer patients. Our study revealed that a Fra-1-miR-134 axis drives a positive feedback loop that amplifies ERK/JNK signaling and reduces chemosensitivity in ovarian cancer cells.
Collapse
|
163
|
Compromized DNA repair as a basis for identification of cancer radiotherapy patients with extreme radiosensitivity. Cancer Lett 2016; 383:212-219. [PMID: 27693457 DOI: 10.1016/j.canlet.2016.09.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/06/2016] [Accepted: 09/08/2016] [Indexed: 01/09/2023]
Abstract
A small percentage of cancer radiotherapy patients develop abnormally severe side effects as a consequence of intrinsic radiosensitivity. We analysed the γ-H2AX response to ex-vivo irradiation of peripheral blood lymphocytes (PBL) and plucked eyebrow hair follicles from 16 patients who developed severe late radiation toxicity following radiotherapy, and 12 matched control patients. Longer retention of the γ-H2AX signal and lower colocalization efficiency of repair factors in over-responding patients confirmed that DNA repair in these individuals was compromised. Five of the radiosensitive patients harboured LoF mutations in DNA repair genes. An extensive range of quantitative parameters of the γ-H2AX response were studied with the objective to establish a predictor for radiosensitivity status. The most powerful predictor was the combination of the fraction of the unrepairable component of γ-H2AX foci and repair rate in PBL, both derived from non-linear regression analysis of foci repair kinetics. We introduce a visual representation of radiosensitivity status that allocates a position for each patient on a two-dimensional "radiosensitivity map". This analytical approach provides the basis for larger prospective studies to further refine the algorithm, ultimately to triage capability.
Collapse
|
164
|
Biphasic reduction of histone H3 phosphorylation in response to N-nitroso compounds induced DNA damage. Biochim Biophys Acta Gen Subj 2016; 1860:1836-44. [DOI: 10.1016/j.bbagen.2016.05.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/08/2016] [Accepted: 05/20/2016] [Indexed: 02/08/2023]
|
165
|
Vogel KU, Bell LS, Galloway A, Ahlfors H, Turner M. The RNA-Binding Proteins Zfp36l1 and Zfp36l2 Enforce the Thymic β-Selection Checkpoint by Limiting DNA Damage Response Signaling and Cell Cycle Progression. THE JOURNAL OF IMMUNOLOGY 2016; 197:2673-2685. [PMID: 27566829 DOI: 10.4049/jimmunol.1600854] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/26/2016] [Indexed: 11/19/2022]
Abstract
The RNA-binding proteins Zfp36l1 and Zfp36l2 act redundantly to enforce the β-selection checkpoint during thymopoiesis, yet their molecular targets remain largely unknown. In this study, we identify these targets on a genome-wide scale in primary mouse thymocytes and show that Zfp36l1/l2 regulate DNA damage response and cell cycle transcripts to ensure proper β-selection. Double-negative 3 thymocytes lacking Zfp36l1/l2 share a gene expression profile with postselected double-negative 3b cells despite the absence of intracellular TCRβ and reduced IL-7 signaling. Our findings show that in addition to controlling the timing of proliferation at β-selection, posttranscriptional control by Zfp36l1/l2 limits DNA damage responses, which are known to promote thymocyte differentiation. Zfp36l1/l2 therefore act as posttranscriptional safeguards against chromosomal instability and replication stress by integrating pre-TCR and IL-7 signaling with DNA damage and cell cycle control.
Collapse
Affiliation(s)
| | - Lewis S Bell
- Dept. of Medicine, University of Cambridge, MRC-Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Alison Galloway
- Centre for Gene Regulation and Expression, School of Life Science, University of Dundee, Dundee DD1 5EH, UK
| | - Helena Ahlfors
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Martin Turner
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| |
Collapse
|
166
|
Affiliation(s)
- Midori Shimada
- a Department of Cell Biology , Graduate School of Medical Sciences, Nagoya City University , Nagoya , Japan
| | - Makoto Nakanishi
- b Division of Cancer Cell Biology, Department of Cancer Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| |
Collapse
|
167
|
Heiser K, Nicholas C, Garcea RL. Activation of DNA damage repair pathways by murine polyomavirus. Virology 2016; 497:346-356. [PMID: 27529739 DOI: 10.1016/j.virol.2016.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/08/2016] [Accepted: 07/26/2016] [Indexed: 11/27/2022]
Abstract
Nuclear replication of DNA viruses activates DNA damage repair (DDR) pathways, which are thought to detect and inhibit viral replication. However, many DNA viruses also depend on these pathways in order to optimally replicate their genomes. We investigated the relationship between murine polyomavirus (MuPyV) and components of DDR signaling pathways including CHK1, CHK2, H2AX, ATR, and DNAPK. We found that recruitment and retention of DDR proteins at viral replication centers was independent of H2AX, as well as the viral small and middle T-antigens. Additionally, infectious virus production required ATR kinase activity, but was independent of CHK1, CHK2, or DNAPK signaling. ATR inhibition did not reduce the total amount of viral DNA accumulated, but affected the amount of virus produced, indicating a defect in virus assembly. These results suggest that MuPyV may utilize a subset of DDR proteins or non-canonical DDR signaling pathways in order to efficiently replicate and assemble.
Collapse
Affiliation(s)
- Katie Heiser
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado at Boulder, Jennie Smoly Caruthers Biotechnology Building, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Catherine Nicholas
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado at Boulder, Jennie Smoly Caruthers Biotechnology Building, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Robert L Garcea
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado at Boulder, Jennie Smoly Caruthers Biotechnology Building, 3415 Colorado Avenue, Boulder, CO 80303, USA.
| |
Collapse
|
168
|
Novak M, Žegura B, Baebler Š, Štern A, Rotter A, Stare K, Filipič M. Influence of selected anti-cancer drugs on the induction of DNA double-strand breaks and changes in gene expression in human hepatoma HepG2 cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:14751-14761. [PMID: 26392091 DOI: 10.1007/s11356-015-5420-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/14/2015] [Indexed: 06/05/2023]
Abstract
In chemotherapy, various anti-cancer drugs with different mechanisms of action are used and may represent different risk of undesirable delayed side effects in treated patients as well as in occupationally exposed populations. The aim of the present study was to evaluate genotoxic potential of four widely used anti-cancer drugs with different mechanisms of action: 5-fluorouracil (5-FU), cisplatin (CDDP) and etoposide (ET) that cause cell death by targeting DNA function and imatinib mesylate (IM) that inhibits targeted protein kinases in cancer cells in an experimental model with human hepatoma HepG2 cells. After 24 h of exposure all four anti-cancer drugs at non-cytotoxic concentrations induced significant increase in formation of DNA double strand breaks (DSBs), with IM being the least effective. The analysis of the changes in the expression of genes involved in the response to DNA damage (CDKN1A, GADD45A, MDM2), apoptosis (BAX, BCL2) and oncogenesis (MYC, JUN) showed that 5-FU, CDDP and ET upregulated the genes involved in DNA damage response, while the anti-apoptotic gene BCL2 and oncogene MYC were downregulated. On the contrary, IM did not change the mRNA level of the studied genes, showing different mechanism of action that probably does not involve direct interaction with DNA processing. Genotoxic effects of the tested anti-cancer drugs were observed at their therapeutic concentrations that may consequently lead to increased risk for development of delayed adverse effects in patients. In addition, considering the genotoxic mechanism of action of 5-FU, CDDP and ET an increased risk can also not be excluded in occupationally exposed populations. The results also indicate that exposure to 5-FU, CDDP and ET represent a higher risk for delayed effects such as cancer, reproductive effects and heritable disease than exposure to IM.
Collapse
Affiliation(s)
- Matjaž Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia
- Ecological Engineering Institute, Maribor, Slovenia
- Jozef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia
| | - Špela Baebler
- Department of Biotechnology and System Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Alja Štern
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia
| | - Ana Rotter
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia
| | - Katja Stare
- Department of Biotechnology and System Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia.
| |
Collapse
|
169
|
Shimada M, Goshima T, Matsuo H, Johmura Y, Haruta M, Murata K, Tanaka H, Ikawa M, Nakanishi K, Nakanishi M. Essential role of autoactivation circuitry on Aurora B-mediated H2AX-pS121 in mitosis. Nat Commun 2016; 7:12059. [PMID: 27389782 PMCID: PMC4941122 DOI: 10.1038/ncomms12059] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/20/2016] [Indexed: 02/06/2023] Open
Abstract
Proper deposition and activation of Aurora B at the centromere is critical for faithful chromosome segregation in mammals. However, the mechanistic basis for abrupt Aurora B kinase activation at the centromere has not yet been fully understood. We demonstrate here that Aurora B-mediated phosphorylation of histone H2AX at serine 121 (H2AX-pS121) promotes Aurora B autophosphorylation and is essential for proper chromosome segregation. Aurora B-mediated H2AX-pS121 is specifically detected at the centromere during mitosis. H2AX depletion results in a severe defect in activation and deposition of Aurora B at this locus. A phosphomimic mutant of H2AX at S121 interacts with activated Aurora B more efficiently than wild-type in vitro. Taken together, these results propose a model in which Aurora B-mediated H2AX-pS121 probably provide a platform for Aurora B autoactivation circuitry at centromeres and thus play a pivotal role in proper chromosome segregation. Aurora B activation at the centromere is critical for faithful chromosome segregation in mammals. Here the authors show that Aurora B-mediated phosphorylation of histone H2AX at serine 121 is essential for Aurora B auto-activation circuitry at centromeres, ensuring proper chromosome segregation.
Collapse
Affiliation(s)
- Midori Shimada
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Takahiro Goshima
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiromi Matsuo
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yoshikazu Johmura
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Mayumi Haruta
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Kazuhiro Murata
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiromitsu Tanaka
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki 859-3298, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Keiko Nakanishi
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 489-0392, Japan
| | - Makoto Nakanishi
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.,Division of Cancer Cell Biology, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
170
|
Feng YL, Xiang JF, Kong N, Cai XJ, Xie AY. Buried territories: heterochromatic response to DNA double-strand breaks. Acta Biochim Biophys Sin (Shanghai) 2016; 48:594-602. [PMID: 27151295 DOI: 10.1093/abbs/gmw033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/28/2016] [Indexed: 12/22/2022] Open
Abstract
Cellular response to DNA double-strand breaks (DSBs), the most deleterious type of DNA damage, is highly influenced by higher-order chromatin structure in eukaryotic cells. Compared with euchromatin, the compacted structure of heterochromatin not only protects heterochromatic DNA from damage, but also adds an extra layer of control over the response to DSBs occurring in heterochromatin. One key step in this response is the decondensation of heterochromatin structure. This decondensation process facilitates the DNA damage signaling and promotes proper heterochromatic DSB repair, thus helping to prevent instability of heterochromatic regions of genomes. This review will focus on the functions of the ataxia telangiectasia mutated (ATM) signaling cascade involving ATM, heterochromatin protein 1 (HP1), Krüppel-associated box (KRAB)-associated protein-1 (KAP-1), tat-interacting protein 60 (Tip60), and many other protein factors in DSB-induced decondensation of heterochromatin and subsequent repair of heterochromatic DSBs. As some subsets of DSBs may be repaired in heterochromatin independently of the ATM signaling, a possible repair model is also proposed for ATM-independent repair of these heterochromatic DSBs.
Collapse
Affiliation(s)
- Yi-Li Feng
- Key Laboratory of Surgery of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Ji-Feng Xiang
- Key Laboratory of Surgery of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Na Kong
- Key Laboratory of Surgery of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Xiu-Jun Cai
- Key Laboratory of Surgery of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - An-Yong Xie
- Key Laboratory of Surgery of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
171
|
LoRusso PM, Li J, Burger A, Heilbrun LK, Sausville EA, Boerner SA, Smith D, Pilat MJ, Zhang J, Tolaney SM, Cleary JM, Chen AP, Rubinstein L, Boerner JL, Bowditch A, Cai D, Bell T, Wolanski A, Marrero AM, Zhang Y, Ji J, Ferry-Galow K, Kinders RJ, Parchment RE, Shapiro GI. Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of the Poly(ADP-ribose) Polymerase (PARP) Inhibitor Veliparib (ABT-888) in Combination with Irinotecan in Patients with Advanced Solid Tumors. Clin Cancer Res 2016; 22:3227-37. [PMID: 26842236 PMCID: PMC4930710 DOI: 10.1158/1078-0432.ccr-15-0652] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 01/26/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE PARP is essential for recognition and repair of DNA damage. In preclinical models, PARP inhibitors modulate topoisomerase I inhibitor-mediated DNA damage. This phase I study determined the MTD, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib, an orally bioavailable PARP1/2 inhibitor, in combination with irinotecan. EXPERIMENTAL DESIGN Patients with advanced solid tumors were treated with 100 mg/m(2) irinotecan on days 1 and 8 of a 21-day cycle. Twice-daily oral dosing of veliparib (10-50 mg) occurred on days 3 to 14 (cycle 1) and days -1 to 14 (subsequent cycles) followed by a 6-day rest. PK studies were conducted with both agents alone and in combination. Paired tumor biopsies were obtained after irinotecan alone and veliparib/irinotecan to evaluate PARP1/2 inhibition and explore DNA damage signals (nuclear γ-H2AX and pNBS1). RESULTS Thirty-five patients were treated. DLTs included fatigue, diarrhea, febrile neutropenia, and neutropenia. The MTD was 100 mg/m(2) irinotecan (days 1 and 8) combined with veliparib 40 mg twice daily (days -1-14) on a 21-day cycle. Of 31 response-evaluable patients, there were six (19%) partial responses. Veliparib exhibited linear PK, and there were no apparent PK interactions between veliparib and irinotecan. At all dose levels, veliparib reduced tumor poly(ADP-ribose) (PAR) content in the presence of irinotecan. Several samples showed increases in γ-H2AX and pNBS1 after veliparib/irinotecan compared with irinotecan alone. CONCLUSIONS Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Preliminary antitumor activity justifies further evaluation of the combination. Clin Cancer Res; 22(13); 3227-37. ©2016 AACR.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Jing Li
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Angelika Burger
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Lance K Heilbrun
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Scott A Boerner
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Daryn Smith
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Mary Jo Pilat
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan. Wayne State University, Detroit, Michigan
| | - Jie Zhang
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Sara M Tolaney
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - James M Cleary
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Lawrence Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Julie L Boerner
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Adam Bowditch
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Dongpo Cai
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Tracy Bell
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Andrew Wolanski
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Allison M Marrero
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Yiping Zhang
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jiuping Ji
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Katherine Ferry-Galow
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert J Kinders
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ralph E Parchment
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Geoffrey I Shapiro
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
172
|
German P, Saenz D, Szaniszlo P, Aguilera-Aguirre L, Pan L, Hegde ML, Bacsi A, Hajas G, Radak Z, Ba X, Mitra S, Papaconstantinou J, Boldogh I. 8-Oxoguanine DNA glycosylase1-driven DNA repair-A paradoxical role in lung aging. Mech Ageing Dev 2016; 161:51-65. [PMID: 27343030 DOI: 10.1016/j.mad.2016.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/16/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Age-associated changes in lung structure and function are some of the most important predictors of overall health, cognitive activities and longevity. Common to all aging cells is an increase in oxidatively modified DNA bases, primarily 8-oxo-7,8-dihydroguanine (8-oxoG). It is repaired via DNA base excision repair pathway driven by 8-oxoguanine DNA glycosylase-1 (OGG1-BER), whose role in aging has been the focus of many studies. This study hypothesizes that signaling and consequent gene expression during cellular response to OGG1-BER "wires" senescence/aging processes. To test OGG1-BER was mimicked by repeatedly exposing diploid lung fibroblasts cells and airways of mice to 8-oxoG base. Results showed that repeated exposures led to G1 cell cycle arrest and pre-matured senescence of cultured cells in which over 1000 genes were differentially expressed -86% of them been identical to those in naturally senesced cells. Gene ontology analysis of gene expression displayed biological processes driven by small GTPases, phosphoinositide 3-kinase and mitogen activated kinase cascades both in cultured cells and lungs. These results together, points to a new paradigm about the role of DNA damage and repair by OGG1 in aging and age-associated disease processes.
Collapse
Affiliation(s)
- Peter German
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - David Saenz
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Peter Szaniszlo
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Leopoldo Aguilera-Aguirre
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Lang Pan
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Muralidhar L Hegde
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Attila Bacsi
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Gyorgy Hajas
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Zsolt Radak
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Xueqing Ba
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Sankar Mitra
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - John Papaconstantinou
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| |
Collapse
|
173
|
Coordinated Regulation of TIP60 and Poly(ADP-Ribose) Polymerase 1 in Damaged-Chromatin Dynamics. Mol Cell Biol 2016; 36:1595-607. [PMID: 26976643 DOI: 10.1128/mcb.01085-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/09/2016] [Indexed: 11/20/2022] Open
Abstract
The dynamic exchange of histones alleviates the nucleosome barrier and simultaneously facilitates various aspects of cellular DNA metabolism, such as DNA repair and transcription. In response to DNA damage, the acetylation of Lys5 in the histone variant H2AX, catalyzed by TIP60, plays a key role in promoting histone exchange; however, the detailed molecular mechanism still is unclear. Here, we show that the TIP60 complex includes poly(ADP-ribose) polymerase 1 (PARP-1). PARP-1 is required for the rapid exchange of H2AX on chromatin at DNA damage sites. It is known that PARP-1 binds dynamically to damaged chromatin and is crucial for the subsequent recruitment of other repair factors, and its auto-poly(ADP-ribosyl)ation is required for the dynamics. We also show that the acetylation of histone H2AX at Lys5 by TIP60, but not the phosphorylation of H2AX, is required for the ADP-ribosylation activity of PARP-1 and its dynamic binding to damaged chromatin. Our results indicate the reciprocal regulation of K5 acetylation of H2AX and PARP-1, which could modulate the chromatin structure to facilitate DNA metabolism at damage sites. This could explain the rather undefined roles of PARP-1 in various DNA damage responses.
Collapse
|
174
|
Namas R, Renauer P, Ognenovski M, Tsou PS, Sawalha AH. Histone H2AX phosphorylation as a measure of DNA double-strand breaks and a marker of environmental stress and disease activity in lupus. Lupus Sci Med 2016; 3:e000148. [PMID: 27158526 PMCID: PMC4854117 DOI: 10.1136/lupus-2016-000148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/09/2016] [Accepted: 04/12/2016] [Indexed: 11/23/2022]
Abstract
Objective Defective or inefficient DNA double-strand break (DSB) repair results in failure to preserve genomic integrity leading to apoptotic cell death, a hallmark of systemic lupus erythematosus (SLE). Compelling evidence linked environmental factors that increase oxidative stress with SLE risk and the formation of DSBs. In this study, we sought to further explore genotoxic stress sensitivity in SLE by investigating DSB accumulation as a marker linking the effect of environmental stressors and the chromatin microenvironment. Methods DSBs were quantified in peripheral blood mononuclear cell subsets from patients with SLE, healthy controls, and patients with rheumatoid arthritis (RA) by measuring phosphorylated H2AX (phospho-H2AX) levels with flow cytometry. Phospho-H2AX levels were assessed in G0/G1, S and G2 cell-cycle phases using propidium iodide staining, and after oxidative stress using 0.5 µM hydrogen peroxide exposure for 0, 2, 5, 10, 30 and 60 min. Results DSB levels were significantly increased in CD4+ T cells, CD8+ T cells and monocytes in SLE compared with healthy controls (p=2.16×10−4, 1.68×10−3 and 4.74×10−3, respectively) and RA (p=1.05×10−3, 1.78×10−3 and 2.43×10−2, respectively). This increase in DSBs in SLE was independent of the cell-cycle phase, and correlated with disease activity. In CD4+ T cells, CD8+ T cells and monocytes, oxidative stress exposure induced significantly higher DSB accumulation in SLE compared with healthy controls (60 min; p=1.64×10−6, 8.11×10−7 and 2.04×10−3, respectively). Conclusions Our data indicate that SLE T cells and monocytes have increased baseline DSB levels and an increased sensitivity to acquiring DSBs in response to oxidative stress. Although the mechanism underlying DSB sensitivity in SLE requires further investigation, accumulation of DSB may serve a biomarker for disease activity in SLE and help explain increased apoptotic cell accumulation in this disease.
Collapse
Affiliation(s)
- Rajaie Namas
- Division of Rheumatology, Department of Internal Medicine , University of Michigan , Ann Arbor, Michigan , USA
| | - Paul Renauer
- Division of Rheumatology, Department of Internal Medicine , University of Michigan , Ann Arbor, Michigan , USA
| | - Mikhail Ognenovski
- Division of Rheumatology, Department of Internal Medicine , University of Michigan , Ann Arbor, Michigan , USA
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine , University of Michigan , Ann Arbor, Michigan , USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
175
|
Maucksch U, Runge R, Wunderlich G, Freudenberg R, Naumann A, Kotzerke J. Comparison of the radiotoxicity of the 99mTc-labeled compounds 99mTc-pertechnetate, 99mTc-HMPAO and 99mTc-MIBI. Int J Radiat Biol 2016; 92:698-706. [DOI: 10.3109/09553002.2016.1168533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Ute Maucksch
- University Hospital/Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Roswitha Runge
- University Hospital/Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Gerd Wunderlich
- University Hospital/Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Robert Freudenberg
- University Hospital/Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Anne Naumann
- University Hospital/Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Jörg Kotzerke
- University Hospital/Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| |
Collapse
|
176
|
Mazouzi A, Stukalov A, Müller AC, Chen D, Wiedner M, Prochazkova J, Chiang SC, Schuster M, Breitwieser FP, Pichlmair A, El-Khamisy SF, Bock C, Kralovics R, Colinge J, Bennett KL, Loizou JI. A Comprehensive Analysis of the Dynamic Response to Aphidicolin-Mediated Replication Stress Uncovers Targets for ATM and ATMIN. Cell Rep 2016; 15:893-908. [PMID: 27149854 DOI: 10.1016/j.celrep.2016.03.077] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 01/21/2016] [Accepted: 03/21/2016] [Indexed: 01/01/2023] Open
Abstract
The cellular response to replication stress requires the DNA-damage-responsive kinase ATM and its cofactor ATMIN; however, the roles of this signaling pathway following replication stress are unclear. To identify the functions of ATM and ATMIN in response to replication stress, we utilized both transcriptomics and quantitative mass-spectrometry-based phosphoproteomics. We found that replication stress induced by aphidicolin triggered widespread changes in both gene expression and protein phosphorylation patterns. These changes gave rise to distinct early and late replication stress responses. Furthermore, our analysis revealed previously unknown targets of ATM and ATMIN downstream of replication stress. We demonstrate ATMIN-dependent phosphorylation of H2AX and of CRMP2, a protein previously implicated in Alzheimer's disease but not in the DNA damage response. Overall, our dataset provides a comprehensive resource for discovering the cellular responses to replication stress and, potentially, associated pathologies.
Collapse
Affiliation(s)
- Abdelghani Mazouzi
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Alexey Stukalov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria; Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - André C Müller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Doris Chen
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Marc Wiedner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Jana Prochazkova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Shih-Chieh Chiang
- Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Florian P Breitwieser
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Andreas Pichlmair
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Sherif F El-Khamisy
- Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Robert Kralovics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Jacques Colinge
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Vienna, Austria.
| |
Collapse
|
177
|
Zhang C, Luo T, Cui S, Gu Y, Bian C, Chen Y, Yu X, Wang Z. Poly(ADP-ribose) protects vascular smooth muscle cells from oxidative DNA damage. BMB Rep 2016; 48:354-9. [PMID: 25748172 PMCID: PMC4578623 DOI: 10.5483/bmbrep.2015.48.6.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Indexed: 11/20/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) undergo death during atherosclerosis, a
widespread cardiovascular disease. Recent studies suggest that oxidative damage
occurs in VSMCs and induces atherosclerosis. Here, we analyzed oxidative damage
repair in VSMCs and found that VSMCs are hypersensitive to oxidative damage.
Further analysis showed that oxidative damage repair in VSMCs is suppressed by a
low level of poly (ADP-ribosyl)ation (PARylation), a key post-translational
modification in oxidative damage repair. The low level of PARylation is not
caused by the lack of PARP-1, the major poly(ADP-ribose) polymerase activated by
oxidative damage. Instead, the expression of poly(ADP-ribose) glycohydrolase,
PARG, the enzyme hydrolyzing poly(ADP-ribose), is significantly higher in VSMCs
than that in the control cells. Using PARG inhibitor to suppress PARG activity
facilitates oxidative damage-induced PARylation as well as DNA damage repair.
Thus, our study demonstrates a novel molecular mechanism for oxidative
damage-induced VSMCs death. This study also identifies the use of PARG
inhibitors as a potential treatment for atherosclerosis. [BMB Reports 2015;
48(6): 354-359]
Collapse
Affiliation(s)
- Chao Zhang
- Vascular Surgery Department of Xuanwu Hospital, Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China
| | - Tao Luo
- Vascular Surgery Department of Xuanwu Hospital, Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China
| | - Shijun Cui
- Vascular Surgery Department of Xuanwu Hospital, Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China
| | - Yongquan Gu
- Vascular Surgery Department of Xuanwu Hospital, Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China
| | - Chunjing Bian
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yibin Chen
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xiaochun Yu
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Zhonggao Wang
- Vascular Surgery Department of Xuanwu Hospital, Institute of Vascular Surgery, Capital Medical University, Beijing 100053, China
| |
Collapse
|
178
|
Phosphorylation-Dependent Regulation of the DNA Damage Response of Adaptor Protein KIBRA in Cancer Cells. Mol Cell Biol 2016; 36:1354-65. [PMID: 26929199 DOI: 10.1128/mcb.01004-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/12/2016] [Indexed: 11/20/2022] Open
Abstract
Multifunctional adaptor proteins encompassing various protein-protein interaction domains play a central role in the DNA damage response pathway. In this report, we show that KIBRA is a physiologically interacting reversible substrate of ataxia telangiectasia mutated (ATM) kinase. We identified the site of phosphorylation in KIBRA as threonine 1006, which is embedded within the serine/threonine (S/T) Q consensus motif, by site-directed mutagenesis, and we further confirmed the same with a phospho-(S/T) Q motif-specific antibody. Results from DNA repair functional assays such as the γ-H2AX assay, pulsed-field gel electrophoresis (PFGE), Comet assay, terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, and clonogenic cell survival assay using stable overexpression clones of wild-type (wt.) KIBRA and active (T1006E) and inactive (T1006A) KIBRA phosphorylation mutants showed that T1006 phosphorylation on KIBRA is essential for optimal DNA double-strand break repair in cancer cells. Further, results from stable retroviral short hairpin RNA-mediated knockdown (KD) clones of KIBRA and KIBRA knockout (KO) model cells generated by a clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 system showed that depleting KIBRA levels compromised the DNA repair functions in cancer cells upon inducing DNA damage. All these phenotypic events were reversed upon reconstitution of KIBRA into cells lacking KIBRA knock-in (KI) model cells. All these results point to the fact that phosphorylated KIBRA might be functioning as a scaffolding protein/adaptor protein facilitating the platform for further recruitment of other DNA damage response factors. In summary, these data demonstrate the imperative functional role of KIBRAper se(KIBRA phosphorylation at T1006 site as a molecular switch that regulates the DNA damage response, possibly via the nonhomologous end joining [NHEJ] pathway), suggesting that KIBRA could be a potential therapeutic target for modulating chemoresistance in cancer cells.
Collapse
|
179
|
He D, Xiang J, Li B, Liu H. The dynamic behavior of Ect2 in response to DNA damage. Sci Rep 2016; 6:24504. [PMID: 27074761 PMCID: PMC4830932 DOI: 10.1038/srep24504] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/30/2016] [Indexed: 12/16/2022] Open
Abstract
Ect2 is a BRCT-containing guanidine exchange factor for Rho GTPases. It is essential for cytokinesis and is also involved in tumorigenesis. Since most BRCT-containing proteins are involved in DNA damage response and/or DNA repair, we tested whether Ect2 plays similar roles. We report that in primary mouse embryonic fibroblasts (MEFs), DNA damage quickly led to Ect2 relocalization to the chromatin and DNA damage foci-like structures. Ect2 knockdown did not affect foci localization of γH2AX, TopBP1, or Brca1, or activation of Atm, yet it impeded p53 Ser15 phosphorylation and activation, and resulted in defects in apoptosis and activation of S and G2/M checkpoints in response to DNA damage. These results suggest that Ect2 plays a role in DNA damage response. Interestingly, Ect2 is down-regulated at late stages of DNA damage response. Although p53 and E2F1 have been shown to regulate Ect2 transcription, DNA damage-induced Ect2 down-regulation occurred in p53-/- or Atm-/- MEFs and E2F1 knockdown cells. Instead, DNA damage-induced Ect2 down-regulation is mainly attributable to decreased protein stability. Like Ect2 knockdown, Ect2 destabilization may help the cell to recover from DNA damage response. These results suggest that Ect2 plays roles in multiple aspects of DNA damage response.
Collapse
Affiliation(s)
- Dan He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinnan Xiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
180
|
Wen L, Liang C, Chen E, Chen W, Liang F, Zhi X, Wei T, Xue F, Li G, Yang Q, Gong W, Feng X, Bai X, Liang T. Regulation of Multi-drug Resistance in hepatocellular carcinoma cells is TRPC6/Calcium Dependent. Sci Rep 2016; 6:23269. [PMID: 27011063 PMCID: PMC4806320 DOI: 10.1038/srep23269] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 03/03/2016] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is notoriously refractory to chemotherapy because of its tendency to develop multi-drug resistance (MDR), whose various underlying mechanisms make it difficult to target. The calcium signalling pathway is associated with many cellular biological activities, and is also a critical player in cancer. However, its role in modulating tumour MDR remains unclear. In this study, stimulation by doxorubicin, hypoxia and ionizing radiation was used to induce MDR in HCC cells. A sustained aggregation of intracellular calcium was observed upon these stimuli, while inhibition of calcium signalling enhanced the cells' sensitivity to various drugs by attenuating epithelial-mesenchymal transition (EMT), Hif1-α signalling and DNA damage repair. The effect of calcium signalling is mediated via transient receptor potential canonical 6 (TRPC6), a subtype of calcium-permeable channel. An in vivo xenograft model of HCC further confirmed that inhibiting TRPC6 enhanced the efficacy of doxorubicin. In addition, we deduced that STAT3 activation is a downstream signalling pathway in MDR. Collectively, this study demonstrated that the various mechanisms regulating MDR in HCC cells are calcium dependent through the TRPC6/calcium/STAT3 pathway. We propose that targeting TRPC6 in HCC may be a novel antineoplastic strategy, especially combined with chemotherapy.
Collapse
Affiliation(s)
- Liang Wen
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Chao Liang
- Department of General Surgery, Ningbo Medical Treatment Center Lihuili Hospital, Ningbo, PR China
| | - Enjiang Chen
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Wei Chen
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Feng Liang
- Department of Neurosurgery Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xiao Zhi
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Tao Wei
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Fei Xue
- Department of Hepatobiliary and Pancreatic Surgery, Henan Province People’s Hospital, Henan, PR China
| | - Guogang Li
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Qi Yang
- Center for Immunology and Microbial Disease,Albany Medical College, ME-205 47 New Scotland Ave., MC-151, Albany, New York, USA
| | - Weihua Gong
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xinhua Feng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, PR China
| | - Xueli Bai
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Tingbo Liang
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
- Collaborative Innovation Center for Cancer Medicine, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
181
|
Wang Y, Gao D, Chu B, Gao C, Cao D, Liu H, Jiang Y. Exposure of CCRF-CEM cells to acridone derivative 8a triggers tumor death via multiple mechanisms. Proteomics 2016; 16:1177-90. [DOI: 10.1002/pmic.201500317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Yini Wang
- Department of Chemistry; Tsinghua University; Beijing P. R. China
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
| | - Dan Gao
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Bizhu Chu
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Chunmei Gao
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Deliang Cao
- Department of Medical Microbiology; Immunology and Cell Biology; Simmons Cancer Institute; Southern Illinois University School of Medicine, Springfield; IL USA
| | - Hongxia Liu
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Yuyang Jiang
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
- School of Medicine; Tsinghua University; Beijing P. R. China
| |
Collapse
|
182
|
Jacobs KM, Misri S, Meyer B, Raj S, Zobel CL, Sleckman BP, Hallahan DE, Sharma GG. Unique epigenetic influence of H2AX phosphorylation and H3K56 acetylation on normal stem cell radioresponses. Mol Biol Cell 2016; 27:1332-45. [PMID: 26941327 PMCID: PMC4831886 DOI: 10.1091/mbc.e16-01-0017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/22/2016] [Indexed: 01/08/2023] Open
Abstract
Normal stem cells from tissues often exhibiting radiation injury are highly radiosensitive and exhibit a muted DNA damage response, in contrast to differentiated progeny. These radioresponses can be attributed to unique epigenetic regulation in stem cells, identifying potential therapeutic targets for radioprotection. Normal tissue injury resulting from cancer radiotherapy is often associated with diminished regenerative capacity. We examined the relative radiosensitivity of normal stem cell populations compared with non–stem cells within several radiosensitive tissue niches and culture models. We found that these stem cells are highly radiosensitive, in contrast to their isogenic differentiated progeny. Of interest, they also exhibited a uniquely attenuated DNA damage response (DDR) and muted DNA repair. Whereas stem cells exhibit reduced ATM activation and ionizing radiation–induced foci, they display apoptotic pannuclear H2AX-S139 phosphorylation (γH2AX), indicating unique radioresponses. We also observed persistent phosphorylation of H2AX-Y142 along the DNA breaks in stem cells, which promotes apoptosis while inhibiting DDR signaling. In addition, down-regulation of constitutively elevated histone-3 lysine-56 acetylation (H3K56ac) in stem cells significantly decreased their radiosensitivity, restored DDR function, and increased survival, signifying its role as a key contributor to stem cell radiosensitivity. These results establish that unique epigenetic landscapes affect cellular heterogeneity in radiosensitivity and demonstrate the nonubiquitous nature of radiation responses. We thus elucidate novel epigenetic rheostats that promote ionizing radiation hypersensitivity in various normal stem cell populations, identifying potential molecular targets for pharmacological radioprotection of stem cells and hopefully improving the efficacy of future cancer treatment.
Collapse
Affiliation(s)
- Keith M Jacobs
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108
| | - Sandeep Misri
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108
| | - Barbara Meyer
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108
| | - Suyash Raj
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108
| | - Cheri L Zobel
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108
| | - Barry P Sleckman
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63108 Department of Pathology, Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63108
| | - Dennis E Hallahan
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108 Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63108
| | - Girdhar G Sharma
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, St. Louis, MO 63108 Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63108
| |
Collapse
|
183
|
Nyeste A, Bencsura P, Vida I, Hegyi Z, Homolya L, Fodor E, Welker E. Expression of the Prion Protein Family Member Shadoo Causes Drug Hypersensitivity That Is Diminished by the Coexpression of the Wild Type Prion Protein. J Biol Chem 2016; 291:4473-86. [PMID: 26721882 DOI: 10.1074/jbc.m115.679035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Indexed: 11/06/2022] Open
Abstract
The prion protein (PrP) seems to exert both neuroprotective and neurotoxic activities. The toxic activities are associated with the C-terminal globular parts in the absence of the flexible N terminus, specifically the hydrophobic domain (HD) or the central region (CR). The wild type prion protein (PrP-WT), having an intact flexible part, exhibits neuroprotective qualities by virtue of diminishing many of the cytotoxic effects of these mutant prion proteins (PrPΔHD and PrPΔCR) when coexpressed. The prion protein family member Doppel, which possesses a three-dimensional fold similar to the C-terminal part of PrP, is also harmful to neuronal and other cells in various models, a phenotype that can also be eliminated by the coexpression of PrP-WT. In contrast, another prion protein family member, Shadoo (Sho), a natively disordered protein possessing structural features similar to the flexible N-terminal tail of PrP, exhibits PrP-WT-like protective properties. Here, we report that, contrary to expectations, Sho expression in SH-SY5Y or HEK293 cells induces the same toxic phenotype of drug hypersensitivity as PrPΔCR. This effect is exhibited in a dose-dependent manner and is also counteracted by the coexpression of PrP-WT. The opposing effects of Shadoo in different model systems revealed here may be explored to help discern the relationship of the various toxic activities of mutant PrPs with each other and the neurotoxic effects seen in neurodegenerative diseases, such as transmissible spongiform encephalopathy and Alzheimer disease.
Collapse
Affiliation(s)
- Antal Nyeste
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Petra Bencsura
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - István Vida
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and the Institute of Chemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Zoltán Hegyi
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - László Homolya
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - Elfrieda Fodor
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Ervin Welker
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary, the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| |
Collapse
|
184
|
Zhu Q, Sharma N, He J, Wani G, Wani AA. USP7 deubiquitinase promotes ubiquitin-dependent DNA damage signaling by stabilizing RNF168. Cell Cycle 2016; 14:1413-25. [PMID: 25894431 DOI: 10.1080/15384101.2015.1007785] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
During DNA damage response (DDR), histone ubiquitination by RNF168 is a critical event, which orchestrates the recruitment of downstream DDR factors, e.g. BRCA1 and 53BP1. Here, we report USP7 deubiquitinase regulates the stability of RNF168. We showed that USP7 disruption impairs H2A and ultraviolet radiation (UVR)-induced γH2AX monoubiquitination, and decreases the levels of pBmi1, Bmi1, RNF168 and BRCA1. The effect of USP7 disruption was recapitulated by siRNA-mediated USP7 depletion. The USP7 disruption also compromises the formation of UVR-induced foci (UVRIF) and ionizing radiation-induced foci (IRIF) of monoubiquitinated H2A (uH2A) and polyubiquitinated H2AX/A, and subsequently affects UVRIF and IRIF of BRCA1 as well as the IRIF of 53BP1. USP7 was shown to physically bind RNF168 in vitro and in vivo. Overexpression of wild-type USP7, but not its interaction-defective mutant, prevents UVR-induced RNF168 degradation. The USP7 mutant is unable to cleave Ub-conjugates of RNF168 in vivo. Importantly, ectopic expression of RNF168, or both RNF8 and RNF168 together in USP7-disrupted cells, significantly rescue the formation of UVRIF and IRIF of polyubiquitinated H2A and BRCA1. Taken together, these findings reveal an important role of USP7 in regulating ubiquitin-dependent signaling via stabilization of RNF168.
Collapse
Affiliation(s)
- Qianzheng Zhu
- a Department of Radiology ; The Ohio State University ; Columbus , OH USA
| | | | | | | | | |
Collapse
|
185
|
Francia S, Cabrini M, Matti V, Oldani A, d'Adda di Fagagna F. DICER, DROSHA and DNA damage response RNAs are necessary for the secondary recruitment of DNA damage response factors. J Cell Sci 2016; 129:1468-76. [PMID: 26906421 PMCID: PMC4852722 DOI: 10.1242/jcs.182188] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/09/2016] [Indexed: 12/18/2022] Open
Abstract
The DNA damage response (DDR) plays a central role in preserving genome integrity. Recently, we reported that the endoribonucleases DICER and DROSHA contribute to DDR activation by generating small non-coding RNAs, termed DNA damage response RNA (DDRNA), carrying the sequence of the damaged locus. It is presently unclear whether DDRNAs act by promoting the primary recognition of DNA lesions or the secondary recruitment of DDR factors into cytologically detectable foci and consequent signal amplification. Here, we demonstrate that DICER and DROSHA are dispensable for primary recruitment of the DDR sensor NBS1 to DNA damage sites. Instead, the accumulation of the DDR mediators MDC1 and 53BP1 (also known as TP53BP1), markers of secondary recruitment, is reduced in DICER- or DROSHA-inactivated cells. In addition, NBS1 (also known as NBN) primary recruitment is resistant to RNA degradation, consistent with the notion that RNA is dispensable for primary recognition of DNA lesions. We propose that DICER, DROSHA and DDRNAs act in the response to DNA damage after primary recognition of DNA lesions and, together with γH2AX, are essential for enabling the secondary recruitment of DDR factors and fuel the amplification of DDR signaling. Summary: We show that DICER, DROSHA and DNA damage response RNAs are necessary for the secondary recruitment of DNA damage response factors but not essential for primary recognition of DNA lesions.
Collapse
Affiliation(s)
- Sofia Francia
- IFOM Foundation - The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, Milan 20139, Italy Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), Via Abbiategrasso 207, Pavia 27100, Italy
| | - Matteo Cabrini
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), Via Abbiategrasso 207, Pavia 27100, Italy
| | - Valentina Matti
- IFOM Foundation - The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, Milan 20139, Italy
| | - Amanda Oldani
- IFOM Foundation - The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, Milan 20139, Italy
| | - Fabrizio d'Adda di Fagagna
- IFOM Foundation - The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, Milan 20139, Italy Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), Via Abbiategrasso 207, Pavia 27100, Italy
| |
Collapse
|
186
|
Weyemi U, Redon CE, Choudhuri R, Aziz T, Maeda D, Boufraqech M, Parekh PR, Sethi TK, Kasoji M, Abrams N, Merchant A, Rajapakse VN, Bonner WM. The histone variant H2A.X is a regulator of the epithelial-mesenchymal transition. Nat Commun 2016; 7:10711. [PMID: 26876487 PMCID: PMC4756313 DOI: 10.1038/ncomms10711] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 01/12/2016] [Indexed: 02/08/2023] Open
Abstract
The epithelial–mesenchymal transition (EMT), considered essential for metastatic cancer, has been a focus of much research, but important questions remain. Here, we show that silencing or removing H2A.X, a histone H2A variant involved in cellular DNA repair and robust growth, induces mesenchymal-like characteristics including activation of EMT transcription factors, Slug and ZEB1, in HCT116 human colon cancer cells. Ectopic H2A.X re-expression partially reverses these changes, as does silencing Slug and ZEB1. In an experimental metastasis model, the HCT116 parental and H2A.X-null cells exhibit a similar metastatic behaviour, but the cells with re-expressed H2A.X are substantially more metastatic. We surmise that H2A.X re-expression leads to partial EMT reversal and increases robustness in the HCT116 cells, permitting them to both form tumours and to metastasize. In a human adenocarcinoma panel, H2A.X levels correlate inversely with Slug and ZEB1 levels. Together, these results point to H2A.X as a regulator of EMT. The histone H2A variants are involved in DNA repair, gene regulation and cancer development. In this study, the authors unravel an additional role for H2A.X in the regulation of mesenchymal-like traits and activation of the EMT transcription factors, Slug and ZEB1, in colon cancer cells.
Collapse
Affiliation(s)
- Urbain Weyemi
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Christophe E Redon
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Rohini Choudhuri
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Towqir Aziz
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Daisuke Maeda
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Myriem Boufraqech
- Endocrine Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, USA
| | - Palak R Parekh
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Taresh K Sethi
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Manjula Kasoji
- Center for Cancer Research Collaborative Bioinformatics Resource, National Cancer Institute, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Natalie Abrams
- Center for Cancer Research Collaborative Bioinformatics Resource, National Cancer Institute, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Anand Merchant
- Center for Cancer Research Collaborative Bioinformatics Resource, National Cancer Institute, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - Vinodh N Rajapakse
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| | - William M Bonner
- Developmental Therapeutics Branch, Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892, USA
| |
Collapse
|
187
|
Histone modifications in DNA damage response. SCIENCE CHINA-LIFE SCIENCES 2016; 59:257-70. [PMID: 26825946 DOI: 10.1007/s11427-016-5011-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
Abstract
DNA damage is a relatively common event in eukaryotic cell and may lead to genetic mutation and even cancer. DNA damage induces cellular responses that enable the cell either to repair the damaged DNA or cope with the damage in an appropriate way. Histone proteins are also the fundamental building blocks of eukaryotic chromatin besides DNA, and many types of post-translational modifications often occur on tails of histones. Although the function of these modifications has remained elusive, there is ever-growing studies suggest that histone modifications play vital roles in several chromatin-based processes, such as DNA damage response. In this review, we will discuss the main histone modifications, and their functions in DNA damage response.
Collapse
|
188
|
Jiang L, Wu X, He F, Liu Y, Hu X, Takeda S, Qing Y. Genetic Evidence for Genotoxic Effect of Entecavir, an Anti-Hepatitis B Virus Nucleotide Analog. PLoS One 2016; 11:e0147440. [PMID: 26800464 PMCID: PMC4723259 DOI: 10.1371/journal.pone.0147440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/04/2016] [Indexed: 02/05/2023] Open
Abstract
Nucleoside analogues (NAs) have been the most frequently used treatment option for chronic hepatitis B patients. However, they may have genotoxic potentials due to their interference with nucleic acid metabolism. Entecavir, a deoxyguanosine analog, is one of the most widely used oral antiviral NAs against hepatitis B virus. It has reported that entecavir gave positive responses in both genotoxicity and carcinogenicity assays. However the genotoxic mechanism of entecavir remains elusive. To evaluate the genotoxic mechanisms, we analyzed the effect of entecavir on a panel of chicken DT40 B-lymphocyte isogenic mutant cell line deficient in DNA repair and damage tolerance pathways. Our results showed that Parp1-/- mutant cells defective in single-strand break (SSB) repair were the most sensitive to entecavir. Brca1-/-, Ubc13-/- and translesion-DNA-synthesis deficient cells including Rad18-/- and Rev3-/- were hypersensitive to entecavir. XPA-/- mutant deficient in nucleotide excision repair was also slightly sensitive to entecavir. γ-H2AX foci forming assay confirmed the existence of DNA damage by entecavir in Parp1-/-, Rad18-/- and Brca1-/- mutants. Karyotype assay further showed entecavir-induced chromosomal aberrations, especially the chromosome gaps in Parp1-/-, Brca1-/-, Rad18-/- and Rev3-/- cells when compared with wild-type cells. These genetic comprehensive studies clearly identified the genotoxic potentials of entecavir and suggested that SSB and postreplication repair pathways may suppress entecavir-induced genotoxicity.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaohua Wu
- Regenerative Medicine Research Center, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fang He
- Center of Infectious Diseases, Division of Molecular Biology of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying Liu
- Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoqing Hu
- Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| | - Yong Qing
- Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
189
|
Hayes SA, Pandiri AR, Ton TVT, Hong HHL, Clayton NP, Shockley KR, Peddada SD, Gerrish K, Wyde M, Sills RC, Hoenerhoff MJ. Renal Cell Carcinomas in Vinylidene Chloride-exposed Male B6C3F1 Mice Are Characterized by Oxidative Stress and TP53 Pathway Dysregulation. Toxicol Pathol 2016; 44:71-87. [PMID: 26682919 PMCID: PMC4752433 DOI: 10.1177/0192623315610820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vinylidene chloride (VDC) has been widely used in the production of plastics and flame retardants. Exposure of B6C3F1 mice to VDC in the 2-year National Toxicology Program carcinogenicity bioassay resulted in a dose-dependent increases in renal cell hyperplasia, renal cell adenoma, and renal cell carcinomas (RCCs). Among those differentially expressed genes from controls and RCC of VDC-exposed mice, there was an overrepresentation of genes from pathways associated with chronic xenobiotic and oxidative stress as well as c-Myc overexpression and dysregulation of TP53 cell cycle checkpoint and DNA damage repair pathways in RCC. Trend analysis comparing RCC, VDC-exposed kidney, and chamber control kidney showed a conservation of pathway dysregulation in terms of overrepresentation of xenobiotic and oxidative stress, and DNA damage and cell cycle checkpoint pathways in both VDC-exposed kidney and RCC, suggesting that these mechanisms play a role in the pathogenesis of RCC in VDC-exposed mice.
Collapse
Affiliation(s)
- Schantel A Hayes
- Charles River Laboratories, Pathology Associates, Research Triangle Park, North Carolina, USA
| | - Arun R Pandiri
- Experimental Pathology Laboratories Inc., Research Triangle Park, North Carolina, USA Cellular and Molecular Pathology Branch, Division of the National Toxicologic Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Thai-vu T Ton
- Cellular and Molecular Pathology Branch, Division of the National Toxicologic Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Hue-Hua L Hong
- Cellular and Molecular Pathology Branch, Division of the National Toxicologic Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Natasha P Clayton
- Cellular and Molecular Pathology Branch, Division of the National Toxicologic Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Keith R Shockley
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Shyamal D Peddada
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kevin Gerrish
- Microarray Core, Toxicology and Pharmacology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Michael Wyde
- Toxicology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Robert C Sills
- Cellular and Molecular Pathology Branch, Division of the National Toxicologic Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Mark J Hoenerhoff
- Cellular and Molecular Pathology Branch, Division of the National Toxicologic Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
190
|
Januchta W, Serocki M, Dzierzbicka K, Cholewinski G, Gensicka M, Skladanowski A. Synthesis and biological evaluation of novel analogues of batracylin with synthetic amino acids and adenosine: an unexpected effect on centromere segregation in tumor cells through a dual inhibition of topoisomerase IIα and Aurora B. RSC Adv 2016. [DOI: 10.1039/c6ra04957e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In the search for new anticancer agents we designed and synthesized batracylin derivatives with linking synthetic amino acid side chains of different lengths and adenosine.
Collapse
Affiliation(s)
- Wioleta Januchta
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Marcin Serocki
- Department of Pharmaceutical Technology and Biochemistry
- Faculty of Chemistry
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Grzegorz Cholewinski
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Monika Gensicka
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Andrzej Skladanowski
- Department of Pharmaceutical Technology and Biochemistry
- Faculty of Chemistry
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| |
Collapse
|
191
|
Solarczyk KJ, Kordon M, Berniak K, Dobrucki JW. Two stages of XRCC1 recruitment and two classes of XRCC1 foci formed in response to low level DNA damage induced by visible light, or stress triggered by heat shock. DNA Repair (Amst) 2016; 37:12-21. [PMID: 26630398 DOI: 10.1016/j.dnarep.2015.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/13/2015] [Accepted: 10/13/2015] [Indexed: 12/18/2022]
Abstract
Induction of local photosensitised DNA damage has been used to study recruitment of repair factors, spatial organisation and subsequent stages of the repair processes. However, the damage induced by a focused laser beam interacting with a photosensitiser may not fully reflect the types of damage and repair encountered in cells of an animal under typical conditions in vivo. We report on two characteristic stages of recruitment of XRCC1 (a protein engaged in BER and SSB repair pathways), in response to low level DNA damage induced by visible light. We demonstrate that, when just a few DNA breaks are induced in a small region of the nucleus, the recruited XRCC1 is initially distributed uniformly throughout this region, and rearranges into several small stationary foci within minutes. In contrast, when heavy damage of various types (including oxidative damage) is induced in cells pre-sensitized with a DNA-binding drug ethidium bromide, XRCC1 is also recruited but fails to rearrange from the stage of the uniform distribution to the stage of several small foci, indicating that this heavy damage interferes with the progress and completion of the repair processes. We hypothesize that that first stage may reflect recruitment of XRCC1 to poly(ADP-ribose) moieties in the region surrounding the single-strand break, while the second-binding directly to the DNA lesions. We also show that moderate damage or stress induces formation of two types of XRCC1-containing foci differing in their mobility. A large subset of DNA damage-induced XRCC1 foci is associated with a major component of PML nuclear bodies--the Sp100 protein.
Collapse
Affiliation(s)
- Kamil J Solarczyk
- Division of Cell Biophysics Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Magdalena Kordon
- Division of Cell Biophysics Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Krzysztof Berniak
- Division of Cell Biophysics Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Jurek W Dobrucki
- Division of Cell Biophysics Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University ul. Gronostajowa 7, 30-387 Kraków, Poland.
| |
Collapse
|
192
|
Suzuki M, Bandoski C, Bartlett JD. Fluoride induces oxidative damage and SIRT1/autophagy through ROS-mediated JNK signaling. Free Radic Biol Med 2015; 89:369-78. [PMID: 26431905 PMCID: PMC4684823 DOI: 10.1016/j.freeradbiomed.2015.08.015] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/20/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023]
Abstract
Fluoride is an effective caries prophylactic, but at high doses can also be an environmental health hazard. Acute or chronic exposure to high fluoride doses can result in dental enamel and skeletal and soft tissue fluorosis. Dental fluorosis is manifested as mottled, discolored, porous enamel that is susceptible to dental caries. Fluoride induces cell stress, including endoplasmic reticulum stress and oxidative stress, which leads to impairment of ameloblasts responsible for dental enamel formation. Recently we reported that fluoride activates SIRT1 and autophagy as an adaptive response to protect cells from stress. However, it still remains unclear how SIRT1/autophagy is regulated in dental fluorosis. In this study, we demonstrate that fluoride exposure generates reactive oxygen species (ROS) and the resulting oxidative damage is counteracted by SIRT1/autophagy induction through c-Jun N-terminal kinase (JNK) signaling in ameloblasts. In the mouse-ameloblast-derived cell line LS8, fluoride induced ROS, mitochondrial damage including cytochrome-c release, up-regulation of UCP2, attenuation of ATP synthesis, and H2AX phosphorylation (γH2AX), which is a marker of DNA damage. We evaluated the effects of the ROS inhibitor N-acetylcysteine (NAC) and the JNK inhibitor SP600125 on fluoride-induced SIRT1/autophagy activation. NAC decreased fluoride-induced ROS generation and attenuated JNK and c-Jun phosphorylation. NAC decreased SIRT1 phosphorylation and formation of the autophagy marker LC3II, which resulted in an increase in the apoptosis mediators γH2AX and cleaved/activated caspase-3. SP600125 attenuated fluoride-induced SIRT1 phosphorylation, indicating that fluoride activates SIRT1/autophagy via the ROS-mediated JNK pathway. In enamel organs from rats or mice treated with 50, 100, or 125 ppm fluoride for 6 weeks, cytochrome-c release and the DNA damage markers 8-oxoguanine, p-ATM, and γH2AX were increased compared to those in controls (0 ppm fluoride). These results suggest that fluoride-induced ROS generation causes mitochondrial damage and DNA damage, which may lead to impairment of ameloblast function. To counteract this impairment, SIRT1/autophagy is induced via JNK signaling to protect cells/ameloblasts from fluoride-induced oxidative damage that may cause dental fluorosis.
Collapse
Affiliation(s)
- Maiko Suzuki
- Department of Mineralized Tissue Biology, The Forsyth Institute & Harvard School of Dental Medicine, 245 First Street, Cambridge, MA
| | - Cheryl Bandoski
- Department of Mineralized Tissue Biology, The Forsyth Institute & Harvard School of Dental Medicine, 245 First Street, Cambridge, MA
| | - John D Bartlett
- Department of Mineralized Tissue Biology, The Forsyth Institute & Harvard School of Dental Medicine, 245 First Street, Cambridge, MA.
| |
Collapse
|
193
|
Yu V, Rahimy M, Korrapati A, Xuan Y, Zou AE, Krishnan AR, Tsui T, Aguilera JA, Advani S, Crotty Alexander LE, Brumund KT, Wang-Rodriguez J, Ongkeko WM. Electronic cigarettes induce DNA strand breaks and cell death independently of nicotine in cell lines. Oral Oncol 2015; 52:58-65. [PMID: 26547127 DOI: 10.1016/j.oraloncology.2015.10.018] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 09/29/2015] [Accepted: 10/20/2015] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Evaluate the cytotoxicity and genotoxicity of short- and long-term e-cigarette vapor exposure on a panel of normal epithelial and head and neck squamous cell carcinoma (HNSCC) cell lines. MATERIALS AND METHODS HaCaT, UMSCC10B, and HN30 were treated with nicotine-containing and nicotine-free vapor extract from two popular e-cigarette brands for periods ranging from 48 h to 8 weeks. Cytotoxicity was assessed using Annexin V flow cytometric analysis, trypan blue exclusion, and clonogenic assays. Genotoxicity in the form of DNA strand breaks was quantified using the neutral comet assay and γ-H2AX immunostaining. RESULTS E-cigarette-exposed cells showed significantly reduced cell viability and clonogenic survival, along with increased rates of apoptosis and necrosis, regardless of e-cigarette vapor nicotine content. They also exhibited significantly increased comet tail length and accumulation of γ-H2AX foci, demonstrating increased DNA strand breaks. CONCLUSION E-cigarette vapor, both with and without nicotine, is cytotoxic to epithelial cell lines and is a DNA strand break-inducing agent. Further assessment of the potential carcinogenic effects of e-cigarette vapor is urgently needed.
Collapse
Affiliation(s)
- Vicky Yu
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Mehran Rahimy
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Avinaash Korrapati
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Yinan Xuan
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Angela E Zou
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Aswini R Krishnan
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Tzuhan Tsui
- Division of Pulmonary and Critical Care, University of California, San Diego, La Jolla, California, United States
| | - Joseph A Aguilera
- Department of Radiation Medicine and Applied Sciences, Moores Cancer Center, University of California, San Diego, La Jolla, California, United States
| | - Sunil Advani
- Department of Radiation Medicine and Applied Sciences, Moores Cancer Center, University of California, San Diego, La Jolla, California, United States
| | - Laura E Crotty Alexander
- Division of Pulmonary and Critical Care, University of California, San Diego, La Jolla, California, United States; Pulmonary and Critical Care Section, VA San Diego Healthcare System, La Jolla, California, United States
| | - Kevin T Brumund
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| | - Jessica Wang-Rodriguez
- Department of Pathology, VA San Diego Healthcare System, La Jolla, California, United States
| | - Weg M Ongkeko
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
194
|
Lin JJ, Wu CC, Hsu SC, Weng SW, Ma YS, Huang YP, Lin JG, Chung JG. Alpha-phellandrene-induced DNA damage and affect DNA repair protein expression in WEHI-3 murine leukemia cells in vitro. ENVIRONMENTAL TOXICOLOGY 2015; 30:1322-30. [PMID: 24861204 DOI: 10.1002/tox.22003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/28/2014] [Accepted: 05/09/2014] [Indexed: 05/24/2023]
Abstract
Although there are few reports regarding α-phellandrene (α-PA), a natural compound from Schinus molle L. essential oil, there is no report to show that α-PA induced DNA damage and affected DNA repair associated protein expression. Herein, we investigated the effects of α-PA on DNA damage and repair associated protein expression in murine leukemia cells. Flow cytometric assay was used to measure the effects of α-PA on total cell viability and the results indicated that α-PA induced cell death. Comet assay and 4,6-diamidino-2-phenylindole dihydrochloride staining were used for measuring DNA damage and condensation, respectively, and the results indicated that α-PA induced DNA damage and condensation in a concentration-dependent manner. DNA gel electrophoresis was used to examine the DNA damage and the results showed that α-PA induced DNA damage in WEHI-3 cells. Western blotting assay was used to measure the changes of DNA damage and repair associated protein expression and the results indicated that α-PA increased p-p53, p-H2A.X, 14-3-3-σ, and MDC1 protein expression but inhibited the protein of p53, MGMT, DNA-PK, and BRCA-1.
Collapse
Affiliation(s)
- Jen-Jyh Lin
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
- Division of Cardiology, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chih-Chung Wu
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, 711, Taiwan
| | - Shu-Chun Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Shu-Wen Weng
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
- Department of Chinese Medicine, Taichung Hospital, Department of Health, Executive Yuan, Taichung, 403, Taiwan
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, 84001, Taiwan
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung, 82445, Taiwan
| | - Yi-Ping Huang
- Department of Physiology, China Medical University, Taichung, 404, Taiwan
| | - Jaung-Geng Lin
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 413, Taiwan, R.O.C
| |
Collapse
|
195
|
Cloutier JM, Mahadevaiah SK, ElInati E, Nussenzweig A, Tóth A, Turner JMA. Histone H2AFX Links Meiotic Chromosome Asynapsis to Prophase I Oocyte Loss in Mammals. PLoS Genet 2015; 11:e1005462. [PMID: 26509888 PMCID: PMC4624946 DOI: 10.1371/journal.pgen.1005462] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/23/2015] [Indexed: 11/19/2022] Open
Abstract
Chromosome abnormalities are common in the human population, causing germ cell loss at meiotic prophase I and infertility. The mechanisms driving this loss are unknown, but persistent meiotic DNA damage and asynapsis may be triggers. Here we investigate the contribution of these lesions to oocyte elimination in mice with chromosome abnormalities, e.g. Turner syndrome (XO) and translocations. We show that asynapsed chromosomes trigger oocyte elimination at diplonema, which is linked to the presence of phosphorylated H2AFX (γH2AFX). We find that DNA double-strand break (DSB) foci disappear on asynapsed chromosomes during pachynema, excluding persistent DNA damage as a likely cause, and demonstrating the existence in mammalian oocytes of a repair pathway for asynapsis-associated DNA DSBs. Importantly, deletion or point mutation of H2afx restores oocyte numbers in XO females to wild type (XX) levels. Unexpectedly, we find that asynapsed supernumerary chromosomes do not elicit prophase I loss, despite being enriched for γH2AFX and other checkpoint proteins. These results suggest that oocyte loss cannot be explained simply by asynapsis checkpoint models, but is related to the gene content of asynapsed chromosomes. A similar mechanistic basis for oocyte loss may operate in humans with chromosome abnormalities. Chromosome abnormalities, such as aneuploidies and structural variants (i.e. translocations, inversions), are strikingly common in the human population, causing disorders such as Down syndrome and Turner syndrome. One important consequence of chromosome abnormalities in mammals is errors during meiosis, the specialized cell division that generates sperm and eggs for reproduction. As a result of these meiotic errors, patients with chromosome abnormalities oftentimes suffer from infertility due to loss of developing germ cells. The precise molecular mechanism for germ cell losses and infertility due to chromosome abnormalities is not well understood, but is hypothesized to result from a surveillance mechanism, which has evolved to prevent aneuploidies from developing from abnormal germ cells. In mammals, meiotic surveillance mechanisms have been hypothesized to monitor for unrepaired DNA double-strand breaks (DSB) and/or chromosome pairing/synapsis errors. Here we test these hypotheses using a variety of chromosomally variant mouse models. We find that germ cell loss in female mice with chromosome abnormalities is dependent on phosphorylation of the histone variant H2AFX, an epigenetic mark involved in the transcriptional silencing of asynapsed chromosomes during meiosis. These data inform a silencing-based mechanism of germ cell loss in patients with chromosome abnormalities and for the prophase I surveillance system which safeguards against aneuploidy.
Collapse
Affiliation(s)
| | | | - Elias ElInati
- The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
| | - André Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Attila Tóth
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - James M. A. Turner
- The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
- * E-mail:
| |
Collapse
|
196
|
Targeting Suppressor of Variegation 3-9 Homologue 2 (SUV39H2) in Acute Lymphoblastic Leukemia (ALL). Transl Oncol 2015; 8:368-375. [PMID: 26500027 PMCID: PMC4631083 DOI: 10.1016/j.tranon.2015.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 01/26/2023] Open
Abstract
Although recent progress in understanding the biology and optimizing the treatment of acute lymphoblastic leukemia (ALL) has improved cure rates of childhood ALL to nearly 90%, the cure rate in adult ALL remains less than 50%. The poor prognosis in adult ALL has in part been attributed to larger proportion of high-risk leukemia showing drug resistance. Thus, identifying novel therapeutic targets in ALL is needed for further improvements in treatment outcomes of adult ALL. Genetic aberration of chromatin-modifying molecules has been recently reported in subtypes of ALL, and targeting components of chromatin complexes has shown promising efficacy in preclinical studies. Suppressor of variegation 3-9 homologue 2 (SUV39H2), also known as KMT1B, is a SET-domain–containing histone methyltransferase that is upregulated in solid cancers, but its expression is hardly detectable in normal tissues. Here, we show that SUV39H2 is highly expressed in ALL cells but not in blood cells from healthy donors and also that SUV39H2 mRNA is expressed at significantly higher levels in bone marrow or blood cells from patients with ALL obtained at diagnosis compared with those obtained at remission (P = .007). In four ALL cell lines (Jurkat and CEM derived from T-ALL and RS4;11 and REH derived from B-ALL), SUV39H2 knockdown resulted in a significant decrease in cell viability (~ 77%, P < .001), likely through induction of apoptosis. On the other hand, SUV39H2 overexpression made cells more resistant to chemotherapy. We conclude that SUV39H2 is a promising therapeutic target and further investigation of this therapeutic approach in ALL is warranted.
Collapse
|
197
|
Lavin MF, Kozlov S, Gatei M, Kijas AW. ATM-Dependent Phosphorylation of All Three Members of the MRN Complex: From Sensor to Adaptor. Biomolecules 2015; 5:2877-902. [PMID: 26512707 PMCID: PMC4693261 DOI: 10.3390/biom5042877] [Citation(s) in RCA: 110] [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: 08/27/2015] [Revised: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 11/16/2022] Open
Abstract
The recognition, signalling and repair of DNA double strand breaks (DSB) involves the participation of a multitude of proteins and post-translational events that ensure maintenance of genome integrity. Amongst the proteins involved are several which when mutated give rise to genetic disorders characterised by chromosomal abnormalities, cancer predisposition, neurodegeneration and other pathologies. ATM (mutated in ataxia-telangiectasia (A-T) and members of the Mre11/Rad50/Nbs1 (MRN complex) play key roles in this process. The MRN complex rapidly recognises and locates to DNA DSB where it acts to recruit and assist in ATM activation. ATM, in the company of several other DNA damage response proteins, in turn phosphorylates all three members of the MRN complex to initiate downstream signalling. While ATM has hundreds of substrates, members of the MRN complex play a pivotal role in mediating the downstream signalling events that give rise to cell cycle control, DNA repair and ultimately cell survival or apoptosis. Here we focus on the interplay between ATM and the MRN complex in initiating signaling of breaks and more specifically on the adaptor role of the MRN complex in mediating ATM signalling to downstream substrates to control different cellular processes.
Collapse
Affiliation(s)
- Martin F Lavin
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia.
| | - Sergei Kozlov
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia.
| | - Magtouf Gatei
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia.
| | - Amanda W Kijas
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia.
| |
Collapse
|
198
|
Borràs M, Armengol G, De Cabo M, Barquinero JF, Barrios L. Comparison of methods to quantify histone H2AX phosphorylation and its usefulness for prediction of radiosensitivity. Int J Radiat Biol 2015; 91:915-24. [DOI: 10.3109/09553002.2015.1101501] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
199
|
Abstract
DNA damage is correlated with and may drive the ageing process. Neurons in the brain are postmitotic and are excluded from many forms of DNA repair; therefore, neurons are vulnerable to various neurodegenerative diseases. The challenges facing the field are to understand how and when neuronal DNA damage accumulates, how this loss of genomic integrity might serve as a 'time keeper' of nerve cell ageing and why this process manifests itself as different diseases in different individuals.
Collapse
Affiliation(s)
- Hei-man Chow
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Karl Herrup
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
200
|
Matsui Y, Sunatani Y, Hayashi N, Okino K, Okushi Y, Mukai K, Adachi H, Yamaya H, Iwabuchi K, Yokoyama H. DNA double-strand breaks induced intractable glomerular fibrosis in renal allografts. Clin Exp Nephrol 2015; 20:479-88. [PMID: 26440363 DOI: 10.1007/s10157-015-1174-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/24/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUNDS The relationship between DNA damage and glomerular fibrosis in renal allografts remains unclear. METHODS We examined renal allograft specimens from 35 patients in which DNA double-strand breaks (DSBs) and glomerular fibrosis were detected by phospho-histone H2A.X (γ-H2AX) expression and collagen (COL) types III, IV, and VI accumulation. We also examined the in vitro relationship between DNA damage and COL accumulation by mitomycin C (MMc)-induced DNA damage in human glomerular endothelial cells (HRGEc). RESULTS The γ-H2AX and COL type VI, which mainly accumulated in the subendothelial and mesangial regions, were positively correlated with the duration of the post-renal transplant (RT) period. In multiple regression analysis, the duration of the post-RT period and cg in the Banff '07 classification were identified as a significant predictor of COL type VI accumulation and γ-H2AX expression in the glomerular capillaries. In addition, the γ-H2AX-positive area was also identified as a predictor of glomerular accumulation of COL type VI. COL type VI was detected in the cytoplasm of the HRGEc, which was secreted into the supernatant after MMc stimulation with γ-H2AX expression. The number of γ-H2AX (-)/COL type VI (+) cells was inversely associated with the number of γ-H2AX (+)/COL type VI (-) cells during 24-h MMc treatment. CONCLUSIONS Our findings suggest that the long-term RT induces DSBs and HRGEc-secreted COL type VI accumulation in the glomerular capillaries, which might progress to intractable glomerular fibrosis.
Collapse
Affiliation(s)
- Yuki Matsui
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Yumi Sunatani
- Department of Biochemistry I, School of Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Norifumi Hayashi
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Kazuaki Okino
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Yuki Okushi
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Kiyotaka Mukai
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiroki Adachi
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Hideki Yamaya
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Kuniyoshi Iwabuchi
- Department of Biochemistry I, School of Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Hitoshi Yokoyama
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan.
| |
Collapse
|