1
|
Lu H, Wise SS, Toyoda JH, Speer RM, Croom-Perez TJ, Meaza I, Kouokam JC, Wise JY, Hoyle G, Chen N, Wise JP, Kondo K, Toba H, Takizawa H, Wise JP. Particulate hexavalent chromium exposure induces DNA double-strand breaks and inhibits homologous recombination repair in rat and human lung tissues. CHEMOSPHERE 2025; 370:143982. [PMID: 39701314 PMCID: PMC11750071 DOI: 10.1016/j.chemosphere.2024.143982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
Lung cancer is an important human health concern because of its high mortality rate, with many cases caused by environmental chemicals other than tobacco. Particulate hexavalent chromium [Cr(VI)] is a well-established human lung carcinogen, but how Cr(VI) induces lung cancer is poorly understood. Chromosome instability, a hallmark of lung cancer, is considered a major driving factor in Cr(VI)-induced lung cancer. Our previous studies in cultured human lung cells showed that particulate Cr(VI) induces DNA double-strand breaks during the late S and G2 phases of the cell cycle, which are repaired by homologous recombination, one of the main repair pathways of DNA double-strand breaks. Our previous data showed that prolonged exposure to Cr(VI) inhibits homologous recombination repair by targeting RAD51, a key protein that mediates homologous recombination. Therefore, particulate Cr(VI)-induced DNA damage combined with failure of DNA repair can lead to chromosome instability. In this study we translated these results to rat lung tissue and lung tumor tissue from Cr(VI)-exposed workers. Wistar rats were exposed to zinc chromate in a saline solution or saline alone by oropharyngeal aspiration with a single dose repeated weekly for 90 days. We observed DNA double-strand breaks increased in a concentration-dependent manner, but homologous recombination repair decreased in rat lungs after 90 days of exposure. Notably, these effects were more pronounced in bronchioles than alveoli. We also considered these effects in Cr(VI)-associated human lung tumors and observed increased DNA double-strand breaks and reduced RAD51 levels in lung tumor tissue compared with adjacent normal lung tissue. Thus, Cr(VI)-induced induction of DNA double-strand breaks, and inhibition of homologous recombination repair translates from cultured cells to experimental animals, normal lung tissue adjacent to the tumor, and Cr(VI)-associated human lung tumors.
Collapse
Affiliation(s)
- Haiyan Lu
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Sandra S. Wise
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Jennifer H. Toyoda
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Rachel M. Speer
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Tayler J Croom-Perez
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Idoia Meaza
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - J. Calvin Kouokam
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Jamie Young Wise
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
| | - Gary Hoyle
- Environmental and Occupational Health Sciences, University of Louisville, Louisville, Kentucky 40292
| | - Ning Chen
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky 40292
| | - John Pierce Wise
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
- Pediatric Research Institute, University of Louisville, Louisville, Kentucky 40292
| | - Kazuya Kondo
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University Tokushima, Japan
| | - Hiroaki Toba
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University Tokushima, Japan
| | - Hiromitsu Takizawa
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University Tokushima, Japan
| | - John Pierce Wise
- Wise Laboratory of Environmental and Genetic Toxicology, University of Louisville, Louisville, Kentucky 40292
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292
| |
Collapse
|
2
|
Yu W, Cai X, Wang C, Peng X, Xu L, Gao Y, Tian T, Zhu G, Pan Y, Chu H, Liang S, Chen C, Kim NH, Yuan B, Zhang J, Jiang H. FOXM1 affects oxidative stress, mitochondrial function, and the DNA damage response by regulating p21 in aging oocytes. Theriogenology 2024; 229:66-74. [PMID: 39163804 DOI: 10.1016/j.theriogenology.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Fertilization capacity and embryo survival rate are decreased in postovulatory aging oocytes, which results in a reduced reproductive rate in female animals. However, the key regulatory genes and related regulatory mechanisms involved in the process of postovulatory aging in oocytes remain unclear. In this study, RNA-Seq revealed that 3237 genes were differentially expressed in porcine oocytes between the MII and aging stages (MII + 24 h). The expression level of FOXM1 was increased at the aging stage, and FOXM1 was also observed to be enriched in many key biological processes, such as cell senescence, response to oxidative stress, and transcription, during porcine oocyte aging. Previous studies have shown that FOXM1 is involved in the regulation of various biological processes, such as oxidative stress, DNA damage repair, mitochondrial function, and cellular senescence, which suggests that FOXM1 may play a crucial role in the process of postovulatory aging. Therefore, in this study, we investigated the effects and mechanisms of FOXM1 on oxidative stress, mitochondrial function, DNA damage, and apoptosis during oocyte aging. Our study revealed that aging oocytes exhibited significantly increased ROS levels and significantly decreased GSH, SOD, T-AOC, and CAT levels than did oocytes at the MII stage and that FOXM1 inhibition exacerbated the changes in these levels in aging oocytes. In addition, FOXM1 inhibition increased the levels of DNA damage, apoptosis, and cell senescence in aging oocytes. A p21 inhibitor alleviated the effects of FOXM1 inhibition on oxidative stress, mitochondrial function, and DNA damage and thus alleviated the degree of senescence in aging oocytes. These results indicate that FOXM1 plays a crucial role in porcine oocyte aging. This study contributes to the understanding of the function and mechanism of FOXM1 during porcine oocyte aging and provides a theoretical basis for preventing oocyte aging and optimizing conditions for the in vitro culture of oocytes.
Collapse
Affiliation(s)
- Wenjie Yu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xiaoshi Cai
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Chen Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xinyue Peng
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Lingxia Xu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Yan Gao
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Tian Tian
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China; Center of Reproductive Medicine & Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Guangying Zhu
- Department of Mental Health, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Yuan Pan
- Center of Reproductive Medicine & Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Hongzhong Chu
- General Animal Husbandry Center of Ili Kazakh Autonomous Prefecture, Yining, 835000, China
| | - Shuang Liang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Chengzhen Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529000, China
| | - Bao Yuan
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Jiabao Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Hao Jiang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China.
| |
Collapse
|
3
|
Randolph ME, Afifi M, Gorthi A, Weil R, Wilky BA, Weinreb J, Ciero P, Hoeve NT, van Diest PJ, Raman V, Bishop AJ, Loeb DM. RNA helicase DDX3 regulates RAD51 localization and DNA damage repair in Ewing sarcoma. iScience 2024; 27:108925. [PMID: 38323009 PMCID: PMC10844834 DOI: 10.1016/j.isci.2024.108925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/09/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
We previously demonstrated that RNA helicase DDX3X (DDX3) can be a therapeutic target in Ewing sarcoma (EWS), but its role in EWS biology remains unclear. The present work demonstrates that DDX3 plays a unique role in DNA damage repair (DDR). We show that DDX3 interacts with several proteins involved in homologous recombination, including RAD51, RECQL1, RPA32, and XRCC2. In particular, DDX3 colocalizes with RAD51 and RNA:DNA hybrid structures in the cytoplasm of EWS cells. Inhibition of DDX3 RNA helicase activity increases cytoplasmic RNA:DNA hybrids, sequestering RAD51 in the cytoplasm, which impairs nuclear translocation of RAD51 to sites of double-stranded DNA breaks, thus increasing sensitivity of EWS to radiation treatment, both in vitro and in vivo. This discovery lays the foundation for exploring new therapeutic approaches directed at manipulating DDR protein localization in solid tumors.
Collapse
Affiliation(s)
- Matthew E. Randolph
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marwa Afifi
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Aparna Gorthi
- Greehey Children’s Cancer Research Institute and Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Rachel Weil
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Breelyn A. Wilky
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Joshua Weinreb
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul Ciero
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Natalie ter Hoeve
- Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Paul J. van Diest
- Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Venu Raman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- Department of Pharmacology, Johns Hopkins University, Baltimore, MD, USA
| | - Alexander J.R. Bishop
- Greehey Children’s Cancer Research Institute and Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - David M. Loeb
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
4
|
Wang YY, Cheng KH, Hung AC, Lo S, Chen PY, Wu YC, Hou MF, Yuan SSF. Differential impact of cytoplasmic vs. nuclear RAD51 expression on breast cancer progression and patient prognosis. Int J Oncol 2024; 64:12. [PMID: 38063232 PMCID: PMC10734667 DOI: 10.3892/ijo.2023.5600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
RAD51 recombinase is one of the DNA damage repair proteins associated with breast cancer risk. Apart from its function to maintain genomic integrity within the cell nucleus, RAD51 localized to the cytoplasm has also been implicated in breast malignancy. However, limited information exists on the roles of cytoplasmic vs. nuclear RAD51 in breast cancer progression and patient prognosis. In the present study, the association of cytoplasmic and nuclear RAD51 with clinical outcomes of patients with breast cancer was analyzed, revealing that elevated cytoplasmic RAD51 expression was associated with breast cancer progression, including increased cancer stage, grade, tumor size, lymph node metastasis and chemoresistance, along with reduced patient survival. By contrast, elevated nuclear RAD51 expression largely had the inverse effect. Results from in vitro investigations supported the cancer‑promoting effect of RAD51, showing that overexpression of RAD51 promoted breast cancer cell growth, chemoresistance and metastatic ability, while knockdown of RAD51 repressed these malignant behaviors. The current data suggest that differential expression of subcellular RAD51 had a distinct impact on breast cancer progression and patient survival. Specifically, cytoplasmic RAD51 in contrast to nuclear RAD51 was potentially an adverse marker in breast cancer.
Collapse
Affiliation(s)
- Yen-Yun Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807
- Drug Development and Value Creation Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Kuang-Hung Cheng
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan, R.O.C
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Amos C. Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Steven Lo
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Pang-Yu Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Yi-Chia Wu
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Shyng-Shiou F. Yuan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan, R.O.C
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
5
|
Pádua JDB, Mariano CFA, Fabro AT, Lizarte Neto FS, Zuliani RL, Sares CTG, dos Santos JS, Sankarankutty AK, Tirapelli DPDC, Silveira VDS, de Molfetta GA, Júnior WADS, Brunaldi MO. mRNA Expression and Methylation of the RAD51, ATM, ATR, BRCA1, and BRCA2 Genes in Gastric Adenocarcinoma. Biomark Insights 2024; 19:11772719231225206. [PMID: 38293680 PMCID: PMC10826385 DOI: 10.1177/11772719231225206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
Background Immunohistochemical prognostic significance of the homologous recombination-related proteins RAD51, ATM, BRCA1, and BRCA2 is known in gastric adenocarcinoma, one of the deadliest cancers. Objective and design This retrospective cohort study aimed to evaluate mRNA expression and promoter methylation of some homologous recombination-related genes in this neoplasm. Methods We evaluated mRNA expression and methylation of RAD51, ATM, ATR, BRCA1, and BRCA2 in tumor and non-tumor frozen samples from gastrectomy specimens by RT-qPCR and MS-HRM, correlating our results with previous immunohistochemistry data and prognostic features. Results RAD51, ATR, BRCA1, BRCA2, and ATM mRNA expression was detected in 93.75% (45/48), 93.75% (45/48), 91.67% (44/48), 83.33% (40/48), and 89.58% (43/48) of the tumors; partial or complete methylation, in 94.87% (37/39), 0 (0/42), 97.56% (40/41), 100% (41/41), and 0 (0/40), respectively. Most gene pairs showed significant weak to moderate positive correlations of tumoral mRNA expression with each other: RAD51 with ATR (P = .027), BRCA1 (P < .001), and BRCA2 (P < .001); ATR with BRCA1 (P = .007), and ATM (P = .001); BRCA1 with BRCA2 (P = 0.001). BRCA1 mRNA was reduced in tumors compared with non-neoplastic mucosa (0.345 vs 1.272, P = .015) and, excluding neoadjuvant therapy cases, in T3 to T4 tumors compared with T2 (0.414 vs 0.954, P = .035). Greater tumoral RAD51 mRNA levels correlated with perineural invasion (1.822 vs 0.725, P = .010) and death (1.664 vs 0.929, P = .036), but not with survival time. There was an inverse association between nuclear immunohistochemical positivity for ATR and its mRNA levels (0.487 vs 0.907, P = .032), and no significant correlation for the other markers. Conclusions Our results suggest RAD51, BRCA1, and BRCA2 methylation as a frequent epigenetic mechanism in gastric cancer, support the hypothesis that reduced BRCA1 expression participates in disease progression, and show an association between RAD51 mRNA and perineural invasion and mortality that may be considered unexpected, considering the former immunohistochemical studies. The lack of correlation between immunohistochemistry and mRNA, and even the inverse association, for ATR, can be seen as indicative of action of post-transcriptional or post-translational regulatory mechanisms, to be better investigated.
Collapse
Affiliation(s)
- Joel Del Bel Pádua
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Carolline Fontes Alves Mariano
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Alexandre Todorovic Fabro
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | | | - Rogério Lenotti Zuliani
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, USP, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | | - Mariângela Ottoboni Brunaldi
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
6
|
Meaza I, Williams AR, Lu H, Kouokam JC, Toyoda JH, Croom-Perez TJ, Wise SS, Aboueissa AEM, Wise JP. Prolonged particulate hexavalent chromium exposure induces RAD51 foci inhibition and cytoplasmic accumulation in immortalized and primary human lung bronchial epithelial cells. Toxicol Appl Pharmacol 2023; 479:116711. [PMID: 37805091 PMCID: PMC10841504 DOI: 10.1016/j.taap.2023.116711] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Hexavalent chromium [Cr(VI)] is a human lung carcinogen with widespread exposure risks. Cr(VI) causes DNA double strand breaks that if unrepaired, progress into chromosomal instability (CIN), a key driving outcome in Cr(VI)-induced tumors. The ability of Cr(VI) to cause DNA breaks and inhibit repair is poorly understood in human lung epithelial cells, which are extremely relevant since pathology data show Cr(VI)-induced tumors originate from bronchial epithelial cells. In the present study, we considered immortalized and primary human bronchial epithelial cells. Cells were treated with zinc chromate at concentrations ranging 0.05 to 0.4μg/cm2 for acute (24 h) and prolonged (120 h) exposures. DNA double strand breaks (DSBs) were measured by neutral comet assay and the status of homologous recombination repair, the main pathway to fix Cr(VI)-induced DSBs, was measured by RAD51 foci formation with immunofluorescence, RAD51 localization with confocal microscopy and sister chromatid exchanges. We found acute and prolonged Cr(VI) exposure induced DSBs. Acute exposure induced homologous recombination repair, but prolonged exposure inhibited it resulting in chromosome instability in immortalized and primary human bronchial epithelial cells.
Collapse
Affiliation(s)
- Idoia Meaza
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Aggie R Williams
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Haiyan Lu
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - J Calvin Kouokam
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Jennifer H Toyoda
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Tayler J Croom-Perez
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 6900 Lake Nona Blvd., Orlando, FL 32827, United States of America
| | - Sandra S Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | | | - John Pierce Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America.
| |
Collapse
|
7
|
Hong L, Braden DC, Zhao Y, Skoko JJ, Chang F, Woodcock SR, Uvalle C, Casey A, Wood K, Salvatore SR, Asan A, Harkness T, Fagunloye A, Razzaghi M, Straub A, Spies M, Brown DD, Lee AV, Schopfer F, Freeman BA, Neumann CA. Small molecule nitroalkenes inhibit RAD51-mediated homologous recombination and amplify triple-negative breast cancer cell killing by DNA-directed therapies. Redox Biol 2023; 66:102856. [PMID: 37633047 PMCID: PMC10472314 DOI: 10.1016/j.redox.2023.102856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023] Open
Abstract
Nitro fatty acids (NO2-FAs) are endogenously generated lipid signaling mediators from metabolic and inflammatory reactions between conjugated diene fatty acids and nitric oxide or nitrite-derived reactive species. NO2-FAs undergo reversible Michael addition with hyperreactive protein cysteine thiolates to induce posttranslational protein modifications that can impact protein function. Herein, we report a novel mechanism of action of natural and non-natural nitroalkenes structurally similar to (E) 10-nitro-octadec-9-enoic acid (CP-6), recently de-risked by preclinical Investigational New Drug-enabling studies and Phase 1 and Phase 2 clinical trials and found to induce DNA damage in a TNBC xenograft by inhibiting homologous-recombination (HR)-mediated repair of DNA double-strand breaks (DSB). CP-6 specifically targets Cys319, essential in RAD51-controlled HR-mediated DNA DSB repair in cells. A nitroalkene library screen identified two structurally different nitroalkenes, a non-natural fatty acid [(E) 8-nitro-nonadec-7-enoic acid (CP-8)] and a dicarboxylate ester [dimethyl (E)nitro-oct-4-enedioate (CP-23)] superior to CP-6 in TNBC cells killing, synergism with three different inhibitors of the poly ADP-ribose polymerase (PARP) and γ-IR. CP-8 and CP-23 effectively inhibited γ-IR-induced RAD51 foci formation and HR in a GFP-reported assay but did not affect benign human epithelial cells or cell cycle phases. In vivo, CP-8 and CP-23's efficacies diverged as only CP-8 showed promising anticancer activities alone and combined with the PARP inhibitor talazoparib in an HR-proficient TNBC mouse model. As preliminary preclinical toxicology analysis also suggests CP-8 as safe, our data endorse CP-8 as a novel anticancer molecule for treating cancers sensitive to homologous recombination-mediated DNA repair inhibitors.
Collapse
Affiliation(s)
- Lisa Hong
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Dennis C Braden
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Yaoning Zhao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA; School of Medicine, Tsinghua University, Beijing, China
| | - John J Skoko
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Fei Chang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Crystall Uvalle
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison Casey
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Katherine Wood
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sonia R Salvatore
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alparslan Asan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Trey Harkness
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Adeola Fagunloye
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Mortezaali Razzaghi
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA, USA
| | - Adam Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maria Spies
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA, USA
| | - Daniel D Brown
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adrian V Lee
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francisco Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Hong L, Braden DC, Zhao Y, Skoko JJ, Chang F, Woodcock SR, Uvalle C, Casey A, Wood K, Salvatore SR, Asan A, Harkness T, Fagunloye A, Razzaghi M, Straub A, Spies M, Brown DD, Lee AV, Schopfer F, Freeman BA, Neumann CA. Small molecule nitroalkenes inhibit RAD51-mediated homologous recombination and amplify triple-negative breast cancer cell killing by DNA-directed therapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.11.552990. [PMID: 37645906 PMCID: PMC10462009 DOI: 10.1101/2023.08.11.552990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Nitro fatty acids (NO 2 -FAs) are endogenously generated lipid signaling mediators from metabolic and inflammatory reactions between conjugated diene fatty acids and nitric oxide or nitrite-derived reactive species. NO 2 -FAs undergo reversible Michael addition with hyperreactive protein cysteine thiolates to induce posttranslational protein modifications that can impact protein function. Herein, we report a novel mechanism of action of natural and non-natural nitroalkenes structurally similar to ( E ) 10-nitro-octadec-9-enoic acid (CP-6), recently de-risked by preclinical Investigational New Drug-enabling studies and Phase 1 and Phase 2 clinical trials and found to induce DNA damage in a TNBC xenograft by inhibiting homologous-recombination (HR)-mediated repair of DNA double-strand breaks (DSB). CP-6 specifically targets Cys319, essential in RAD51-controlled HR-mediated DNA DSB repair in cells. A nitroalkene library screen identified two structurally different nitroalkenes, a non-natural fatty acid [( E ) 8-nitro- nonadec-7-enoic acid (CP-8)] and a dicarboxylate ester [dimethyl ( E )nitro-oct-4-enedioate (CP- 23)] superior to CP-6 in TNBC cells killing, synergism with three different inhibitors of the poly ADP-ribose polymerase (PARP) and γ-IR. CP-8 and CP-23 effectively inhibited γ-IR-induced RAD51 foci formation and HR in a GFP-reported assay but did not affect benign human epithelial cells or cell cycle phases. In vivo, CP-8 and CP-23's efficacies diverged as only CP-8 showed promising anticancer activities alone and combined with the PARP inhibitor talazoparib in an HR-proficient TNBC mouse model. As preliminary preclinical toxicology analysis also suggests CP-8 as safe, our data endorse CP-8 as a novel anticancer molecule for treating cancers sensitive to homologous recombination-mediated DNA repair inhibitors.
Collapse
|
9
|
Randolph ME, Afifi M, Gorthi A, Weil R, Wilky BA, Weinreb J, Ciero P, ter Hoeve N, van Diest PJ, Raman V, Bishop AJR, Loeb DM. RNA Helicase DDX3 Regulates RAD51 Localization and DNA Damage Repair in Ewing Sarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.10.544474. [PMID: 37333164 PMCID: PMC10274875 DOI: 10.1101/2023.06.10.544474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
We previously demonstrated that RNA helicase DDX3X (DDX3) can be a therapeutic target in Ewing sarcoma (EWS), but its role in EWS biology remains unclear. The present work demonstrates that DDX3 plays a unique role in DNA damage repair (DDR). We show that DDX3 interacts with several proteins involved in homologous recombination, including RAD51, RECQL1, RPA32, and XRCC2. In particular, DDX3 colocalizes with RAD51 and RNA:DNA hybrid structures in the cytoplasm of EWS cells. Inhibition of DDX3 RNA helicase activity increases cytoplasmic RNA:DNA hybrids, sequestering RAD51 in the cytoplasm, which impairs nuclear translocation of RAD51 to sites of double-stranded DNA breaks thus increasing sensitivity of EWS to radiation treatment, both in vitro and in vivo. This discovery lays the foundation for exploring new therapeutic approaches directed at manipulating DDR protein localization in solid tumors.
Collapse
Affiliation(s)
- Matthew E. Randolph
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Marwa Afifi
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Aparna Gorthi
- Greehey Children’s Cancer Research Institute and Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, TX
| | - Rachel Weil
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Breelyn A. Wilky
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Joshua Weinreb
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Paul Ciero
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Natalie ter Hoeve
- Department of Pathology, University Medical Centre Utrecht, The Netherlands
| | - Paul J. van Diest
- Department of Pathology, University Medical Centre Utrecht, The Netherlands
| | - Venu Raman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
- Department of Radiology, Johns Hopkins University, Baltimore, MD
- Department of Pharmacology, Johns Hopkins University, Baltimore, MD
| | - Alexander J. R. Bishop
- Greehey Children’s Cancer Research Institute and Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, TX
| | - David M. Loeb
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
10
|
Thomas M, Dubacq C, Rabut E, Lopez BS, Guirouilh-Barbat J. Noncanonical Roles of RAD51. Cells 2023; 12:cells12081169. [PMID: 37190078 DOI: 10.3390/cells12081169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Homologous recombination (HR), an evolutionary conserved pathway, plays a paramount role(s) in genome plasticity. The pivotal HR step is the strand invasion/exchange of double-stranded DNA by a homologous single-stranded DNA (ssDNA) covered by RAD51. Thus, RAD51 plays a prime role in HR through this canonical catalytic strand invasion/exchange activity. The mutations in many HR genes cause oncogenesis. Surprisingly, despite its central role in HR, the invalidation of RAD51 is not classified as being cancer prone, constituting the "RAD51 paradox". This suggests that RAD51 exercises other noncanonical roles that are independent of its catalytic strand invasion/exchange function. For example, the binding of RAD51 on ssDNA prevents nonconservative mutagenic DNA repair, which is independent of its strand exchange activity but relies on its ssDNA occupancy. At the arrested replication forks, RAD51 plays several noncanonical roles in the formation, protection, and management of fork reversal, allowing for the resumption of replication. RAD51 also exhibits noncanonical roles in RNA-mediated processes. Finally, RAD51 pathogenic variants have been described in the congenital mirror movement syndrome, revealing an unexpected role in brain development. In this review, we present and discuss the different noncanonical roles of RAD51, whose presence does not automatically result in an HR event, revealing the multiple faces of this prominent actor in genomic plasticity.
Collapse
Affiliation(s)
- Mélissa Thomas
- INSERM U1016, UMR 8104 CNRS, Institut Cochin, Université de Paris Cité, 24 rue du Faubourg St. Jacques, F-75014 Paris, France
| | - Caroline Dubacq
- Institut de Biologie Paris Seine, IBPS, Neuroscience Paris Seine, NPS, INSERM, CNRS, Sorbonne Université, F-75005 Paris, France
| | - Elise Rabut
- INSERM U1016, UMR 8104 CNRS, Institut Cochin, Université de Paris Cité, 24 rue du Faubourg St. Jacques, F-75014 Paris, France
| | - Bernard S Lopez
- INSERM U1016, UMR 8104 CNRS, Institut Cochin, Université de Paris Cité, 24 rue du Faubourg St. Jacques, F-75014 Paris, France
| | - Josée Guirouilh-Barbat
- INSERM U1016, UMR 8104 CNRS, Institut Cochin, Université de Paris Cité, 24 rue du Faubourg St. Jacques, F-75014 Paris, France
| |
Collapse
|
11
|
Soni UK, Wang Y, Pandey RN, Roberts R, Pressey JG, Hegde RS. Molecularly Defined Subsets of Ewing Sarcoma Tumors Differ in Their Responses to IGF1R and WEE1 Inhibition. Clin Cancer Res 2023; 29:458-471. [PMID: 36394520 PMCID: PMC9843438 DOI: 10.1158/1078-0432.ccr-22-2587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022]
Abstract
PURPOSE Targeted cancer therapeutics have not significantly benefited patients with Ewing sarcoma with metastatic or relapsed disease. Understanding the molecular underpinnings of drug resistance can lead to biomarker-driven treatment selection. EXPERIMENTAL DESIGN Receptor tyrosine kinase (RTK) pathway activation was analyzed in tumor cells derived from a panel of Ewing sarcoma tumors, including primary and metastatic tumors from the same patient. Phospho-RTK arrays, Western blots, and IHC were used. Protein localization and the levels of key markers were determined using immunofluorescence. DNA damage tolerance was measured through PCNA ubiquitination levels and the DNA fiber assay. Effects of pharmacologic inhibition were assessed in vitro and key results validated in vivo using patient-derived xenografts. RESULTS Ewing sarcoma tumors fell into two groups. In one, IGF1R was predominantly nuclear (nIGF1R), DNA damage tolerance pathway was upregulated, and cells had low replication stress and RRM2B levels and high levels of WEE1 and RAD21. These tumors were relatively insensitive to IGF1R inhibition. The second group had high replication stress and RRM2B, low levels of WEE1 and RAD21, membrane-associated IGF1R (mIGF1R) signaling, and sensitivity to IGF1R or WEE1-targeted inhibitors. Moreover, the matched primary and metastatic tumors differed in IGF1R localization, levels of replication stress, and inhibitor sensitivity. In all instances, combined IGF1R and WEE1 inhibition led to tumor regression. CONCLUSIONS IGF1R signaling mechanisms and replication stress levels can vary among Ewing sarcoma tumors (including in the same patient), influencing the effects of IGF1R and WEE1 treatment. These findings make the case for using biopsy-derived predictive biomarkers at multiple stages of Ewing sarcoma disease management.
Collapse
Affiliation(s)
- Upendra Kumar Soni
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yuhua Wang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ram Naresh Pandey
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ryan Roberts
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joseph G. Pressey
- Abigail Wexner Research Institute at Nationwide Children's Hospital, Research II, Columbus, Ohio
| | - Rashmi S. Hegde
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
12
|
Quercetin and Isorhamnetin Reduce Benzo[a]pyrene-Induced Genotoxicity by Inducing RAD51 Expression through Downregulation of miR-34a. Int J Mol Sci 2022; 23:ijms232113125. [PMID: 36361910 PMCID: PMC9653982 DOI: 10.3390/ijms232113125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 01/24/2023] Open
Abstract
Benzo[a]pyrene (B[a]P) is metabolized in the liver into highly reactive mutagenic and genotoxic metabolites, which induce carcinogenesis. The mutagenic factors, including B[a]P-7,8-dihydrodiol-9,10-epoxide (BPDE) and reactive oxygen species, generated during B[a]P metabolism can cause DNA damage, such as BPDE-DNA adducts, 8-oxo-dG, and double-strand breaks (DSBs). In this study, we mechanistically investigated the effects of quercetin and its major metabolite isorhamnetin on the repair of B[a]P-induced DNA DSBs. Whole-transcriptome analysis showed that quercetin and isorhamnetin each modulate the expression levels of genes involved in DNA repair, especially those in homologous recombination. RAD51 was identified as a key gene whose expression level was decreased in B[a]P-treated cells and increased by quercetin or isorhamnetin treatment. Furthermore, the number of γH2AX foci induced by B[a]P was significantly decreased by quercetin or isorhamnetin, whereas RAD51 mRNA and protein levels were increased. Additionally, among the five microRNAs (miRs) known to downregulate RAD51, miR-34a level was significantly downregulated by quercetin or isorhamnetin. The protective effect of quercetin or isorhamnetin was lower in cells transfected with a miR-34a mimic than in non-transfected cells, and the B[a]P-induced DNA DSBs remained unrepaired. Our results show that quercetin and isorhamnetin each upregulates RAD51 by downregulating miR-34a and thereby suppresses B[a]P-induced DNA damage.
Collapse
|
13
|
Wasserzug‐Pash P, Rothman R, Reich E, Zecharyahu L, Schonberger O, Weiss Y, Srebnik N, Cohen‐Hadad Y, Weintraub A, Ben‐Ami I, Holzer H, Klutstein M. Loss of heterochromatin and retrotransposon silencing as determinants in oocyte aging. Aging Cell 2022; 21:e13568. [PMID: 35166017 PMCID: PMC8920445 DOI: 10.1111/acel.13568] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/11/2022] [Accepted: 01/27/2022] [Indexed: 12/13/2022] Open
Abstract
Mammalian oocyte quality reduces with age. We show that prior to the occurrence of significant aneuploidy (9M in mouse), heterochromatin histone marks are lost, and oocyte maturation is impaired. This loss occurs in both constitutive and facultative heterochromatin marks but not in euchromatic active marks. We show that heterochromatin loss with age also occurs in human prophase I-arrested oocytes. Moreover, heterochromatin loss is accompanied in mouse oocytes by an increase in RNA processing and associated with an elevation in L1 and IAP retrotransposon expression and in DNA damage and DNA repair proteins nuclear localization. Artificial inhibition of the heterochromatin machinery in young oocytes causes an elevation in retrotransposon expression and oocyte maturation defects. Inhibiting retrotransposon reverse-transcriptase through azidothymidine (AZT) treatment in older oocytes partially rescues their maturation defects and activity of the DNA repair machinery. Moreover, activating the heterochromatin machinery via treatment with the SIRT1 activating molecule SRT-1720, or overexpression of Sirt1 or Ezh2 via plasmid electroporation into older oocytes causes an upregulation in constitutive heterochromatin, downregulation of retrotransposon expression, and elevated maturation rates. Collectively, our work demonstrates a significant process in oocyte aging, characterized by the loss of heterochromatin-associated chromatin marks and activation of specific retrotransposons, which cause DNA damage and impair oocyte maturation.
Collapse
Affiliation(s)
- Peera Wasserzug‐Pash
- Institute of Dental SciencesFaculty of Dental MedicineThe Hebrew University of JerusalemJerusalemIsrael
| | - Rachel Rothman
- Institute of Dental SciencesFaculty of Dental MedicineThe Hebrew University of JerusalemJerusalemIsrael
| | - Eli Reich
- Institute of Dental SciencesFaculty of Dental MedicineThe Hebrew University of JerusalemJerusalemIsrael
| | - Lital Zecharyahu
- Institute of Dental SciencesFaculty of Dental MedicineThe Hebrew University of JerusalemJerusalemIsrael
| | - Oshrat Schonberger
- IVF UnitDepartment of Obstetrics and GynecologyShaare Zedek Medical Center and Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Yifat Weiss
- IVF UnitDepartment of Obstetrics and GynecologyShaare Zedek Medical Center and Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Naama Srebnik
- IVF UnitDepartment of Obstetrics and GynecologyShaare Zedek Medical Center and Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Yaara Cohen‐Hadad
- IVF UnitDepartment of Obstetrics and GynecologyShaare Zedek Medical Center and Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Amir Weintraub
- IVF UnitDepartment of Obstetrics and GynecologyShaare Zedek Medical Center and Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Ido Ben‐Ami
- IVF UnitDepartment of Obstetrics and GynecologyShaare Zedek Medical Center and Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Hananel Holzer
- Department of Obstetrics and GynecologyHadassah‐Hebrew University Medical CenterKiryat HadassahJerusalemIsrael
| | - Michael Klutstein
- Institute of Dental SciencesFaculty of Dental MedicineThe Hebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
14
|
Panek A, Miszczyk J. ATM and RAD51 Repair Pathways in Human Lymphocytes Irradiated with 70 MeV Therapeutic Proton Beam. Radiat Res 2021; 197:396-402. [PMID: 34958667 DOI: 10.1667/rade-21-00109.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 11/17/2021] [Indexed: 11/03/2022]
Abstract
The repair of radiation-induced DNA damage is a key factor differentiating patients in terms of the therapeutic efficacy and toxicity to surrounding normal tissue. Proton energy substantially determines the types of cancers that can be treated. The present work investigated the DNA double-strand break repair systems, represented by phosphorylated ATM and Rad51. The status of proton therapy energy used to treat major types of cancer is summarized. Here, human lymphocytes from eight healthy donors (male and female) were irradiated with a spread-out Bragg peak using a therapeutic 70 MeV proton beam or with reference X rays. For both types of radiation, the kinetics of pATM and Rad51 repair protein activation (0-24 h) were estimated as determinants of homologous and non-homologous double-strand break repair. Additionally, γ-H2AX was used as the gold standard marker of double-strand breaks. Our results showed that at 30 min postirradiation there was significantly greater accumulation of γ-H2AX (0.6-fold), pATM (2.0-fold), and Rad51 (0.6-fold) in the proton-irradiated cells compared with the X-ray-treated cells. At 24 h post irradiation, for both types of radiation and all investigated proteins, the foci number was still significantly higher when compared with control. Furthermore, the mean value of pATM and Rad51 repair effectiveness was higher in cells exposed to protons than in cells exposed to X rays; however, the difference was significant only for pATM. The largest inter-individual differences in the repair capabilities were noted for Rad51. The association between the frequency of repair protein foci and the frequency of lymphocyte viability at 1 h post irradiation showed a positive correlation for protons but a negative correlation for X rays. These findings indicate that the accumulation of radiation-induced repair protein foci after proton versus X-ray irradiation differs between patients, consequently affecting the cellular responses to particle therapy and conventional radiation therapy.
Collapse
Affiliation(s)
- Agnieszka Panek
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31-342 Krakow, Poland
| | - Justyna Miszczyk
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31-342 Krakow, Poland
| |
Collapse
|
15
|
Tobeiha M, Rajabi A, Raisi A, Mohajeri M, Yazdi SM, Davoodvandi A, Aslanbeigi F, Vaziri M, Hamblin MR, Mirzaei H. Potential of natural products in osteosarcoma treatment: Focus on molecular mechanisms. Biomed Pharmacother 2021; 144:112257. [PMID: 34688081 DOI: 10.1016/j.biopha.2021.112257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most frequent type of bone cancer found in children and adolescents, and commonly arises in the metaphyseal region of tubular long bones. Standard therapeutic approaches, such as surgery, chemotherapy, and radiation therapy, are used in the management of osteosarcoma. In recent years, the mortality rate of osteosarcoma has decreased due to advances in treatment methods. Today, the scientific community is investigating the use of different naturally derived active principles against various types of cancer. Natural bioactive compounds can function against cancer cells in two ways. Firstly they can act as classical cytotoxic compounds by non-specifically affecting macromolecules, such as DNA, enzymes, and microtubules, which are also expressed in normal proliferating cells, but to a greater extent by cancer cells. Secondly, they can act against oncogenic signal transduction pathways, many of which are activated in cancer cells. Some bioactive plant-derived agents are gaining increasing attention because of their anti-cancer properties. Moreover, some naturally-derived compounds can significantly promote the effectiveness of standard chemotherapy drugs, and in certain cases are able to ameliorate drug-induced adverse effects caused by chemotherapy. In the present review we summarize the effects of various naturally-occurring bioactive compounds against osteosarcoma.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahshad Mohajeri
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Aslanbeigi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - MohamadSadegh Vaziri
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
16
|
Wang Z, Dai Z, Wang B, Gao Y, Gao X, Wang L, Zhou S, Yang L, Qiu X, Liu Z. Targeting c-MET to Enhance the Efficacy of Olaparib in Prostate Cancer. Onco Targets Ther 2021; 14:4383-4389. [PMID: 34377000 PMCID: PMC8349550 DOI: 10.2147/ott.s291267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Prostate cancer is the second leading cause of cancer death in men worldwide. Olaparib is clinically approved for the treatment prostate cancer, but cytotoxicity and off-target effects including DNA damage limit its clinical applications. In the current study, new strategies to improve the therapeutic efficacy of olaparib for the treatment of prostate cancer were investigated. Methods Two prostate cancer cell lines were exposed to the c-MET inhibitor PHA665752 and/or the PARP inhibitor olaparib. Cell counting kit-8, colony formation assays, and transwell assays were conducted to evaluate the cytotoxicity of olaparib alone or in combination with PHA665752 in prostate cancer cell lines. Western blotting, immunofluorescence staining, and the comet assay were used to assess the effects of PHA665752 on olaparib-induced DNA damage. Results Combined inhibition of c-MET and PARP resulted in effective and synergistic blocking of the growth of prostate cancer cell lines. Invasion and migration were significantly suppressed when the agents were combined. Mechanistically, dual blocking of PARP and c-MET in prostate cancer cell lines was associated with an impaired DNA damage response. Interestingly, immunofluorescence staining analysis of RAD51 protein indicated that the c-MET inhibitor PHA665752 significantly impaired homologous repair via downregulated translocation of RAD51 into the nucleus in prostate cancer cells. Conclusion The combination of the c-MET inhibitor PHA665752 and the PARP inhibitor olaparib may be a promising therapeutic strategy in patients with prostate cancer.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Zhihong Dai
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Bingwei Wang
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Yuren Gao
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Xiang Gao
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Liang Wang
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Sihai Zhou
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Liqin Yang
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Xiaofu Qiu
- Department of Urology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zhiyu Liu
- Department of Urinary Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
17
|
Tye S, Ronson GE, Morris JR. A fork in the road: Where homologous recombination and stalled replication fork protection part ways. Semin Cell Dev Biol 2021; 113:14-26. [PMID: 32653304 PMCID: PMC8082280 DOI: 10.1016/j.semcdb.2020.07.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022]
Abstract
In response to replication hindrances, DNA replication forks frequently stall and are remodelled into a four-way junction. In such a structure the annealed nascent strand is thought to resemble a DNA double-strand break and remodelled forks are vulnerable to nuclease attack by MRE11 and DNA2. Proteins that promote the recruitment, loading and stabilisation of RAD51 onto single-stranded DNA for homology search and strand exchange in homologous recombination (HR) repair and inter-strand cross-link repair also act to set up RAD51-mediated protection of nascent DNA at stalled replication forks. However, despite the similarities of these pathways, several lines of evidence indicate that fork protection is not simply analogous to the RAD51 loading step of HR. Protection of stalled forks not only requires separate functions of a number of recombination proteins, but also utilises nucleases important for the resection steps of HR in alternative ways. Here we discuss how fork protection arises and how its differences with HR give insights into the differing contexts of these two pathways.
Collapse
Affiliation(s)
- Stephanie Tye
- Section of Structural and Synthetic Biology, Department of Infectious Disease, Imperial College London, London, SW7 2AZ, UK
| | - George E Ronson
- University of Birmingham, College of Medical Dental Schools, Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, Vincent Drive, Edgbaston, Birmingham, B15 2TT, UK
| | - Joanna R Morris
- University of Birmingham, College of Medical Dental Schools, Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, Vincent Drive, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
18
|
PUMA facilitates EMI1-promoted cytoplasmic Rad51 ubiquitination and inhibits DNA repair in stem and progenitor cells. Signal Transduct Target Ther 2021; 6:129. [PMID: 33785736 PMCID: PMC8009889 DOI: 10.1038/s41392-021-00510-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/20/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Maintenance of genetic stability via proper DNA repair in stem and progenitor cells is essential for the tissue repair and regeneration, while preventing cell transformation after damage. Loss of PUMA dramatically increases the survival of mice after exposure to a lethal dose of ionizing radiation (IR), while without promoting tumorigenesis in the long-term survivors. This finding suggests that PUMA (p53 upregulated modulator of apoptosis) may have a function other than regulates apoptosis. Here, we identify a novel role of PUMA in regulation of DNA repair in embryonic or induced pluripotent stem cells (PSCs) and immortalized hematopoietic progenitor cells (HPCs) after IR. We found that PUMA-deficient PSCs and HPCs exhibited a significant higher double-strand break (DSB) DNA repair activity via Rad51-mediated homologous recombination (HR). This is because PUMA can be associated with early mitotic inhibitor 1 (EMI1) and Rad51 in the cytoplasm to facilitate EMI1-mediated cytoplasmic Rad51 ubiquitination and degradation, thereby inhibiting Rad51 nuclear translocation and HR DNA repair. Our data demonstrate that PUMA acts as a repressor for DSB DNA repair and thus offers a new rationale for therapeutic targeting of PUMA in regenerative cells in the context of DNA damage.
Collapse
|
19
|
Wang Y, Zuo M, Jin H, Lai M, Luo J, Cheng Z. Inhibition of ELF3 confers synthetic lethality of PARP inhibitor in non-small cell lung cancer. J Recept Signal Transduct Res 2020; 41:304-311. [PMID: 32814472 DOI: 10.1080/10799893.2020.1808676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND E74 Like ETS Transcription Factor 3 (ELF3) functions as a transcriptional factor to regulate non-small cell lung cancer (NSCLC) differentiation and progression. Poly(ADP-ribose) polymerase (PARP) inhibitors demonstrate anti-tumor effect in NSCLC. This study aimed to investigate whether ELF3 confers synthetic lethal with PARP inhibitor in NSCLC. MATERIALS AND METHODS The sensitivity of PARP inhibitor, Olaparib, to different NSCLC cell lines was determined by half maximal inhibitory concentration (IC50). Expression of ELF3 in NSCLC cell lines was evaluated by western blot. The effects of ELF3 on cytotoxicity of Olaparib to NSCLC were investigated by MTT (3-(4,5- di methyl thiazol -2-yl)-2,5-di phenyl tetrazolium bromide) and colony formation assays. The underlying mechanism involved in synthetic lethality with ELF3 and PARP inhibitors in NSCLC were detected by immunofluorescence and Western blot. RESULTS ELF3 was up-regulated in NSCLC cell lines exhibiting resistance to PARP inhibitor, Olaparib. Knock down of ELF3 decreased the sensitivity and enhanced cytotoxicity of Olaparib to NSCLC cells. Moreover, knock down of ELF3 increased S139 phosphorylated histone H2AX (γH2AX), and inhibited homologous recombination activity via down-regulation of DNA repair protein RAD51 homolog 1 (RAD51), thus showing deficiency in DNA damage repair. Over-expression of ELF3 could up-regulate phosphorylation of AKT (Protein kinase B), while knock down of ELF3 regulated homologous recombination-mediated DNA repair via down-regulation of phosphorylation of AKT. CONCLUSION Knock down of ELF3 revealed homologous recombination deficiency via AKT signaling pathway, and synthetic lethality with ELF3 inhibition and PARP inhibitor indicated the clinical significance of PARP inhibitor in ELF3-deficient NSCLC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Min Zuo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Hongtao Jin
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Meina Lai
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jinfeng Luo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Zhiqiang Cheng
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
20
|
Tang HL, Xu L, Chen XQ. [Bortezomib interferes with DNA repair and exerts synergistic anti-multiple myeloma activity with doxorubicin]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:417-421. [PMID: 32536140 PMCID: PMC7342068 DOI: 10.3760/cma.j.issn.0253-2727.2020.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Indexed: 12/30/2022]
Affiliation(s)
- H L Tang
- Department of Hematology, Xijing Hospital, Air Force Medical University, Hematologic Diseases Center of Chinese People's Liberation Army, Xi'an 710032, China
| | - L Xu
- Department of Hematology, Xijing Hospital, Air Force Medical University, Hematologic Diseases Center of Chinese People's Liberation Army, Xi'an 710032, China
| | - X Q Chen
- Department of Hematology, Xijing Hospital, Air Force Medical University, Hematologic Diseases Center of Chinese People's Liberation Army, Xi'an 710032, China
| |
Collapse
|
21
|
Hridy AU, Shabnaz S, Asaduzzaman MD, Shahriar M, Bhuiyan MA, Safiqul Islam M, Moazzem Hossen SM, Emran TB. Genetic Variations of RAD51 and XRCC2 Genes Increase the Risk of Colorectal Cancer in Bangladeshi Population. Asian Pac J Cancer Prev 2020; 21:1445-1451. [PMID: 32458654 PMCID: PMC7541868 DOI: 10.31557/apjcp.2020.21.5.1445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/01/2020] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES In case of Bangladeshi population, no report is observed till now showing the genetic variations of RAD51 (rs1801320) and XRCC2 (rs3218536) genes polymorphism having association with colorectal cancer risk. For this reason the aim of this study is to ascertain their interrelation with colorectal cancer occurrence in Bangladeshi population. MATERIALS AND METHODS A case control study was conducted where 200 colorectal cancer patients and 200 healthy volunteers were figured for this research using Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP). RESULTS Here, in case of RAD51 (rs1801320), G/C heterozygous genotype was found significant (p=0.037; OR=1.64; 95% CI=1.03 to 2.6). On the other hand, G/G genotype was not found statistically significant (p=0.423; OR=1.61; 95% CI=0.49 to 5.22) and significance was observed for GC+GG (p=0.030; OR=1.63; 95% CI=1.05 to 2.55). In case of XRCC2 (rs3218536), C/T heterozygous genotype was remarked statistically significant (p=0.033; OR=1.60; 95% CI=1.04 to 2.46). The T/T genotype was not recorded statistically significant (p=0.237; OR=1.65; 95% CI=0.72 to 3.76) but significance found for CT+TT (p=0.027; OR=1.61; 95% CI=1.05 to 2.45). Moreover, it is found that the risk factor of developing CRC is observed in G/C, C/T heterozygote and GC+GG, CT+TT (heterozygote+ mutant) in RAD51 (rs1801320) and XRCC2 (rs3218536) respectively although no significance is observed in case of G/G and T/T mutant. CONCLUSIONS So, the association of RAD51 (rs1801320) and XRCC2 (rs3218536) genes polymorphism with colorectal cancer risk is observed in Bangladeshi population.
Collapse
Affiliation(s)
- Anika Uddin Hridy
- Department of Pharmacy, University of Asia Pacific, 74/A, Green Road, Dhaka-1205, Bangladesh.
| | - Samia Shabnaz
- Department of Pharmacy, University of Asia Pacific, 74/A, Green Road, Dhaka-1205, Bangladesh.
| | - M d Asaduzzaman
- Department of Pharmacy, University of Asia Pacific, 74/A, Green Road, Dhaka-1205, Bangladesh.
| | - Mohammad Shahriar
- Department of Pharmacy, University of Asia Pacific, 74/A, Green Road, Dhaka-1205, Bangladesh.
| | - Mohiuddin Ahmed Bhuiyan
- Department of Pharmacy, University of Asia Pacific, 74/A, Green Road, Dhaka-1205, Bangladesh.
| | - Mohammad Safiqul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali-3814, Bangladesh.
| | - S M Moazzem Hossen
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong-4331, Bangladesh.
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chandanaish, Chittagong-4381, Bangladesh.
| |
Collapse
|
22
|
[Bortezomib interferes with DNA repair and exerts synergistic anti-multiple myeloma activity with doxorubicin]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41. [PMID: 32536140 PMCID: PMC7342068 DOI: 10.3760/cma.j.issn.0253-2727.2020.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Arctigenin Enhances the Cytotoxic Effect of Doxorubicin in MDA-MB-231 Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21082997. [PMID: 32340377 PMCID: PMC7215735 DOI: 10.3390/ijms21082997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 12/23/2022] Open
Abstract
Several reports have described the anti-cancer activity of arctigenin, a lignan extracted from Arctium lappa L. Here, we investigated the effect of arctigenin (ATG) on doxorubicin (DOX)-induced cell death using MDA-MB-231 human breast cancer cells. The results showed that DOX-induced cell death was enhanced by ATG/DOX co-treatment in a concentration-dependent manner and that this was associated with increased DOX uptake and the suppression of multidrug resistance-associated protein 1 (MRP1) gene expression in MDA-MB-231 cells. ATG enhanced DOX-induced DNA damage and decreased the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the expressions of RAD51 and survivin. Cell death caused by ATG/DOX co-treatment was mediated by the nuclear translocation of apoptosis inducing factor (AIF), reductions in cellular and mitochondrial Bcl-2 and Bcl-xL, and increases in mitochondrial BAX levels. However, caspase-3 and -7 did not participate in DOX/ATG-induced cell death. We also found that DOX/ATG-induced cell death was linked with activation of the p38 signaling pathway and suppressions of the phosphorylations and expressions of Akt and c-Jun N-terminal kinase. Taken together, these results show that ATG enhances the cytotoxic activity of DOX in MDA-MB-231 human breast cancer cells by inducing prolonged p21 expression and p38-mediated AIF-dependent cell death. In conclusion, our findings suggest that ATG might alleviate the side effects and improve the therapeutic efficacy of DOX.
Collapse
|
24
|
Pearson SJ, Elswood J, Barhoumi R, Ming-Whitfield B, Rijnkels M, Porter WW. Loss of SIM2s inhibits RAD51 binding and leads to unresolved replication stress. Breast Cancer Res 2019; 21:125. [PMID: 31775907 PMCID: PMC6882179 DOI: 10.1186/s13058-019-1207-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
Background Mutations in genes associated with homologous recombination (HR) increase an individual’s risk of developing triple-negative breast cancer (TNBC). Although known for their role in repairing dsDNA breaks, HR repair elements also stabilize and restart stalled replication forks. Essential to these functions are RAD51 and its paralogs, each of which has a unique role in preventing replication fork collapse and restart. However, progress toward understanding the regulation of these factors has been slow. With such a pivotal role in the maintenance of genomic integrity, furthering our understanding of this pathway through the discovery of new factors involved in HR is important. Recently, we showed that singleminded-2s (SIM2s) is stabilized in response to dsDNA breaks and is required for effective HR. Methods Initial analysis of the effect loss of SIM2s has on replication stress resolution was conducted using DNA combing assays in established breast cancer cell lines. Further analysis was conducted via immunostaining to determine the effect loss of SIM2s has on factor recruitment. In vivo confirmation was achieved through the use of a mammary epithelial cell conditional knockout mouse model before SIM2s’ role in RAD51 recruitment was determined by immunoblotting. Results Here, we show loss of SIM2s decreases replication fork stability, leading to fork collapse in response to genotoxic stress. Furthermore, loss of SIM2s results in aberrant separation of sister chromatids during mitosis, which has been previously shown to result in chromosomal fragmentation and aneuploidy. Interestingly, loss of SIM2s was shown to result in failure of RAD51 to localize to sites of replication stress in both breast cancer cell lines and primary mammary epithelial cells. Finally, we observed SIM2 is stabilized in response to genotoxic stress and interacts with RAD51, which is necessary for RAD51-DNA binding. Conclusions Together, these results show a role for SIM2s in the resolution of replication stress and further characterize the necessity of SIM2s for effective RAD51 loading in response to DNA damage or stress, ultimately promoting genomic integrity and thus preventing the accumulation of cancer-promoting mutations.
Collapse
Affiliation(s)
- Scott J Pearson
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Jessica Elswood
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Rola Barhoumi
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Brittini Ming-Whitfield
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Monique Rijnkels
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Weston W Porter
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA. .,Present Address: Veterinary Integrative Biosciences, Texas A&M University, College of Veterinary Medicine, College Station, TX, 77843, USA.
| |
Collapse
|
25
|
Garcin EB, Gon S, Sullivan MR, Brunette GJ, Cian AD, Concordet JP, Giovannangeli C, Dirks WG, Eberth S, Bernstein KA, Prakash R, Jasin M, Modesti M. Differential Requirements for the RAD51 Paralogs in Genome Repair and Maintenance in Human Cells. PLoS Genet 2019; 15:e1008355. [PMID: 31584931 PMCID: PMC6795472 DOI: 10.1371/journal.pgen.1008355] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/16/2019] [Accepted: 08/07/2019] [Indexed: 12/16/2022] Open
Abstract
Deficiency in several of the classical human RAD51 paralogs [RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3] is associated with cancer predisposition and Fanconi anemia. To investigate their functions, isogenic disruption mutants for each were generated in non-transformed MCF10A mammary epithelial cells and in transformed U2OS and HEK293 cells. In U2OS and HEK293 cells, viable ablated clones were readily isolated for each RAD51 paralog; in contrast, with the exception of RAD51B, RAD51 paralogs are cell-essential in MCF10A cells. Underlining their importance for genomic stability, mutant cell lines display variable growth defects, impaired sister chromatid recombination, reduced levels of stable RAD51 nuclear foci, and hyper-sensitivity to mitomycin C and olaparib, with the weakest phenotypes observed in RAD51B-deficient cells. Altogether these observations underscore the contributions of RAD51 paralogs in diverse DNA repair processes, and demonstrate essential differences in different cell types. Finally, this study will provide useful reagents to analyze patient-derived mutations and to investigate mechanisms of chemotherapeutic resistance deployed by cancers.
Collapse
Affiliation(s)
- Edwige B. Garcin
- Cancer Research Center of Marseille; CNRS; Inserm; Institut Paoli-Calmettes; Aix-Marseille Université, Marseille, France
| | - Stéphanie Gon
- Cancer Research Center of Marseille; CNRS; Inserm; Institut Paoli-Calmettes; Aix-Marseille Université, Marseille, France
| | - Meghan R. Sullivan
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, United States of America
| | - Gregory J. Brunette
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, United States of America
| | - Anne De Cian
- Museum National d'Histoire Naturelle, Inserm U1154, CNRS UMR 7196, Sorbonne Universités, Paris, France
| | - Jean-Paul Concordet
- Museum National d'Histoire Naturelle, Inserm U1154, CNRS UMR 7196, Sorbonne Universités, Paris, France
| | - Carine Giovannangeli
- Museum National d'Histoire Naturelle, Inserm U1154, CNRS UMR 7196, Sorbonne Universités, Paris, France
| | - Wilhelm G. Dirks
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German, Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Sonja Eberth
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German, Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Kara A. Bernstein
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, United States of America
| | - Rohit Prakash
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Mauro Modesti
- Cancer Research Center of Marseille; CNRS; Inserm; Institut Paoli-Calmettes; Aix-Marseille Université, Marseille, France
| |
Collapse
|
26
|
Yang RM, Nanayakkara D, Kalimutho M, Mitra P, Khanna KK, Dray E, Gonda TJ. MYB regulates the DNA damage response and components of the homology-directed repair pathway in human estrogen receptor-positive breast cancer cells. Oncogene 2019; 38:5239-5249. [PMID: 30971760 DOI: 10.1038/s41388-019-0789-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 02/20/2019] [Accepted: 03/07/2019] [Indexed: 11/09/2022]
Abstract
Over 70% of human breast cancers are estrogen receptor-positive (ER+), most of which express MYB. In these and other cell types, the MYB transcription factor regulates the expression of many genes involved in cell proliferation, differentiation, tumorigenesis, and apoptosis. So far, no clear link has been established between MYB and the DNA damage response in breast cancer. Here, we found that silencing MYB in the ER+ breast cancer cell line MCF-7 led to increased DNA damage accumulation, as marked by increased γ-H2AX foci following induction of double-stranded breaks. We further found that this was likely mediated by decreased homologous recombination-mediated repair (HRR), since silencing MYB impaired the formation of RAD51 foci in response to DNA damage. Moreover, cells depleted for MYB exhibited reduced expression of several key genes involved in HRR including BRCA1, PALB2, and TOPBP1. Taken together, these data imply that MYB and its targets play an important role in the response of ER+ breast cancer cells to DNA damage, and suggest that induction of DNA damage along with inhibition of MYB activity could offer therapeutic benefits for ER+ breast cancer and possibly other cancer types.
Collapse
Affiliation(s)
- Ren-Ming Yang
- School of Pharmacy, University of Queensland, Brisbane, QLD, 4102, Australia.,Keck School of Medicine at the Children's Hospital Los Angeles Campus, University of Southern California, Los Angeles, CA, 90027, USA
| | - Devathri Nanayakkara
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Murugan Kalimutho
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Partha Mitra
- School of Pharmacy, University of Queensland, Brisbane, QLD, 4102, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, TRI, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Kum Kum Khanna
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Eloise Dray
- Institute of Health and Biomedical Innovations, QUT at the Translational Research Institute, Brisbane, QLD, 4102, Australia. .,Mater Research/UQ at the Translational Research Institute, Brisbane, QLD, 4102, Australia. .,University of Texas Health, San Antonio, Department of Biochemistry and Structural Biology, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| | - Thomas J Gonda
- School of Pharmacy, University of Queensland, Brisbane, QLD, 4102, Australia. .,University of South Australia Cancer Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
27
|
Son MY, Hasty P. Homologous recombination defects and how they affect replication fork maintenance. AIMS GENETICS 2019; 5:192-211. [PMID: 31435521 PMCID: PMC6690234 DOI: 10.3934/genet.2018.4.192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/18/2019] [Indexed: 01/07/2023]
Abstract
Homologous recombination (HR) repairs DNA double strand breaks (DSBs) and stabilizes replication forks (RFs). RAD51 is the recombinase for the HR pathway. To preserve genomic integrity, RAD51 forms a filament on the 3' end of a DSB and on a single-stranded DNA (ssDNA) gap. But unregulated HR results in undesirable chromosomal rearrangements. This review describes the multiple mechanisms that regulate HR with a focus on those mechanisms that promote and contain RAD51 filaments to limit chromosomal rearrangements. If any of these pathways break down and HR becomes unregulated then disease, primarily cancer, can result.
Collapse
Affiliation(s)
- Mi Young Son
- Department of Molecular Medicine and Institute of Biotechnology, UT Health San Antonio, 15355 Lambda Drive, San Antonio, USA
| | - Paul Hasty
- Department of Molecular Medicine and Institute of Biotechnology, UT Health San Antonio, 15355 Lambda Drive, San Antonio, USA
- The Mays Cancer Center, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, USA
| |
Collapse
|
28
|
Li Y, Xiao X, Bossé Y, Gorlova O, Gorlov I, Han Y, Byun J, Leighl N, Johansen JS, Barnett M, Chen C, Goodman G, Cox A, Taylor F, Woll P, Wichmann HE, Manz J, Muley T, Risch A, Rosenberger A, Han J, Siminovitch K, Arnold SM, Haura EB, Bolca C, Holcatova I, Janout V, Kontic M, Lissowska J, Mukeria A, Ognjanovic S, Orlowski TM, Scelo G, Swiatkowska B, Zaridze D, Bakke P, Skaug V, Zienolddiny S, Duell EJ, Butler LM, Houlston R, Artigas MS, Grankvist K, Johansson M, Shepherd FA, Marcus MW, Brunnström H, Manjer J, Melander O, Muller DC, Overvad K, Trichopoulou A, Tumino R, Liu G, Bojesen SE, Wu X, Le Marchand L, Albanes D, Bickeböller H, Aldrich MC, Bush WS, Tardon A, Rennert G, Teare MD, Field JK, Kiemeney LA, Lazarus P, Haugen A, Lam S, Schabath MB, Andrew AS, Bertazzi PA, Pesatori AC, Christiani DC, Caporaso N, Johansson M, McKay JD, Brennan P, Hung RJ, Amos CI. Genetic interaction analysis among oncogenesis-related genes revealed novel genes and networks in lung cancer development. Oncotarget 2019; 10:1760-1774. [PMID: 30956756 PMCID: PMC6442994 DOI: 10.18632/oncotarget.26678] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 01/22/2019] [Indexed: 12/31/2022] Open
Abstract
The development of cancer is driven by the accumulation of many oncogenesis-related genetic alterations and tumorigenesis is triggered by complex networks of involved genes rather than independent actions. To explore the epistasis existing among oncogenesis-related genes in lung cancer development, we conducted pairwise genetic interaction analyses among 35,031 SNPs from 2027 oncogenesis-related genes. The genotypes from three independent genome-wide association studies including a total of 24,037 lung cancer patients and 20,401 healthy controls with Caucasian ancestry were analyzed in the study. Using a two-stage study design including discovery and replication studies, and stringent Bonferroni correction for multiple statistical analysis, we identified significant genetic interactions between SNPs in RGL1:RAD51B (OR=0.44, p value=3.27x10-11 in overall lung cancer and OR=0.41, p value=9.71x10-11 in non-small cell lung cancer), SYNE1:RNF43 (OR=0.73, p value=1.01x10-12 in adenocarcinoma) and FHIT:TSPAN8 (OR=1.82, p value=7.62x10-11 in squamous cell carcinoma) in our analysis. None of these genes have been identified from previous main effect association studies in lung cancer. Further eQTL gene expression analysis in lung tissues provided information supporting the functional role of the identified epistasis in lung tumorigenesis. Gene set enrichment analysis revealed potential pathways and gene networks underlying molecular mechanisms in overall lung cancer as well as histology subtypes development. Our results provide evidence that genetic interactions between oncogenesis-related genes play an important role in lung tumorigenesis and epistasis analysis, combined with functional annotation, provides a valuable tool for uncovering functional novel susceptibility genes that contribute to lung cancer development by interacting with other modifier genes.
Collapse
Affiliation(s)
- Yafang Li
- Baylor College of Medicine, Houston, TX, USA
| | | | | | - Olga Gorlova
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, USA
| | - Ivan Gorlov
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, USA
| | | | | | - Natasha Leighl
- University Health Network, The Princess Margaret Cancer Centre, Toronto, CA, USA
| | - Jakob S. Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Matt Barnett
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chu Chen
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Angela Cox
- Department of Oncology, University of Sheffield, Sheffield, UK
| | - Fiona Taylor
- Department of Oncology, University of Sheffield, Sheffield, UK
| | - Penella Woll
- Department of Oncology, University of Sheffield, Sheffield, UK
| | - H. Erich Wichmann
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Judith Manz
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Muley
- Thoraxklinik at University Hospital Heidelberg, Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| | - Angela Risch
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
- German Center for Lung Research (DKFZ), Heidelberg, Germany
- University of Salzburg and Cancer Cluster, Salzburg, Austria
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Jiali Han
- Indiana University, Bloomington, IN, USA
| | | | | | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ciprian Bolca
- Institute of Pneumology “Marius Nasta”, Bucharest, Romania
| | - Ivana Holcatova
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Vladimir Janout
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Milica Kontic
- Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jolanta Lissowska
- M. Sklodowska-Curie Cancer Center, Institute of Oncology, Warsaw, Poland
| | - Anush Mukeria
- Department of Epidemiology and Prevention, N.N. Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Simona Ognjanovic
- International Organization for Cancer Prevention and Research, Belgrade, Serbia
| | - Tadeusz M. Orlowski
- Department of Surgery, National Tuberculosis and Lung Diseases Research Institute, Warsaw, Poland
| | - Ghislaine Scelo
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Beata Swiatkowska
- Nofer Institute of Occupational Medicine, Department of Environmental Epidemiology, Lodz, Poland
| | - David Zaridze
- Department of Epidemiology and Prevention, N.N. Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Vidar Skaug
- National Institute of Occupational Health, Oslo, Norway
| | | | - Eric J. Duell
- Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | | | | | - María Soler Artigas
- Department of Health Sciences, Genetic Epidemiology Group, University of Leicester, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | | | | | - Michael W. Marcus
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Jonas Manjer
- Faculty of Medicine, Lund University, Lund, Sweden
| | | | - David C. Muller
- School of Public Health, St. Mary’s Campus, Imperial College London, London, UK
| | - Kim Overvad
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | | | - Rosario Tumino
- Molecular and Nutritional Epidemiology Unit CSPO (Cancer Research and Prevention Centre), Scientific Institute of Tuscany, Florence, Italy
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Stig E. Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Demetrios Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Melinda C. Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William S. Bush
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Adonina Tardon
- IUOPA, University of Oviedo and CIBERESP, Faculty of Medicine, Campus del Cristo s/n, Oviedo, Spain
| | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - M. Dawn Teare
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - John K. Field
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA, USA
| | - Aage Haugen
- National Institute of Occupational Health, Oslo, Norway
| | - Stephen Lam
- British Columbia Cancer Agency, Vancouver, Canada
| | - Matthew B. Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Pier Alberto Bertazzi
- Department of Preventive Medicine, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Angela C. Pesatori
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - David C. Christiani
- Department of Epidemiology, Program in Molecular and Genetic Epidemiology Harvard School of Public Health, Boston, MA, USA
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mattias Johansson
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - James D. McKay
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Paul Brennan
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Rayjean J. Hung
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | |
Collapse
|
29
|
Maertens O, Kuzmickas R, Manchester HE, Emerson CE, Gavin AG, Guild CJ, Wong TC, De Raedt T, Bowman-Colin C, Hatchi E, Garraway LA, Flaherty KT, Pathania S, Elledge SJ, Cichowski K. MAPK Pathway Suppression Unmasks Latent DNA Repair Defects and Confers a Chemical Synthetic Vulnerability in BRAF-, NRAS-, and NF1-Mutant Melanomas. Cancer Discov 2019; 9:526-545. [PMID: 30709805 PMCID: PMC10151004 DOI: 10.1158/2159-8290.cd-18-0879] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/05/2018] [Accepted: 01/29/2019] [Indexed: 11/16/2022]
Abstract
Although the majority of BRAF-mutant melanomas respond to BRAF/MEK inhibitors, these agents are not typically curative. Moreover, they are largely ineffective in NRAS- and NF1-mutant tumors. Here we report that genetic and chemical suppression of HDAC3 potently cooperates with MAPK pathway inhibitors in all three RAS pathway-driven tumors. Specifically, we show that entinostat dramatically enhances tumor regression when combined with BRAF/MEK inhibitors, in both models that are sensitive or relatively resistant to these agents. Interestingly, MGMT expression predicts responsiveness and marks tumors with latent defects in DNA repair. BRAF/MEK inhibitors enhance these defects by suppressing homologous recombination genes, inducing a BRCA-like state; however, addition of entinostat triggers the concomitant suppression of nonhomologous end-joining genes, resulting in a chemical synthetic lethality caused by excessive DNA damage. Together, these studies identify melanomas with latent DNA repair defects, describe a promising drug combination that capitalizes on these defects, and reveal a tractable therapeutic biomarker. SIGNIFICANCE: BRAF/MEK inhibitors are not typically curative in BRAF-mutant melanomas and are ineffective in NRAS- and NF1-mutant tumors. We show that HDAC inhibitors dramatically enhance the efficacy of BRAF/MEK inhibitors in sensitive and insensitive RAS pathway-driven melanomas by coordinately suppressing two DNA repair pathways, and identify a clinical biomarker that predicts responsiveness.See related commentary by Lombard et al., p. 469.This article is highlighted in the In This Issue feature, p. 453.
Collapse
Affiliation(s)
- Ophélia Maertens
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Ludwig Center at Harvard, Boston, Massachusetts
| | - Ryan Kuzmickas
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Haley E Manchester
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Chloe E Emerson
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Alessandra G Gavin
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Caroline J Guild
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Terence C Wong
- Department of Medical Oncology, Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Thomas De Raedt
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Christian Bowman-Colin
- Harvard Medical School, Boston, Massachusetts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elodie Hatchi
- Harvard Medical School, Boston, Massachusetts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Levi A Garraway
- Harvard Medical School, Boston, Massachusetts
- Ludwig Center at Harvard, Boston, Massachusetts
- Department of Medical Oncology, Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Keith T Flaherty
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Shailja Pathania
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, Massachusetts
| | - Stephen J Elledge
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Ludwig Center at Harvard, Boston, Massachusetts
- Department of Genetics, Howard Hughes Medical Institute, Boston, Massachusetts
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
- Ludwig Center at Harvard, Boston, Massachusetts
| |
Collapse
|
30
|
Chernikova SB, Nguyen RB, Truong JT, Mello SS, Stafford JH, Hay MP, Olson A, Solow-Cordero DE, Wood DJ, Henry S, von Eyben R, Deng L, Gephart MH, Aroumougame A, Wiese C, Game JC, Győrffy B, Brown JM. Dynamin impacts homology-directed repair and breast cancer response to chemotherapy. J Clin Invest 2018; 128:5307-5321. [PMID: 30371505 PMCID: PMC6264728 DOI: 10.1172/jci87191] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 09/13/2018] [Indexed: 12/31/2022] Open
Abstract
After the initial responsiveness of triple-negative breast cancers (TNBCs) to chemotherapy, they often recur as chemotherapy-resistant tumors, and this has been associated with upregulated homology-directed repair (HDR). Thus, inhibitors of HDR could be a useful adjunct to chemotherapy treatment of these cancers. We performed a high-throughput chemical screen for inhibitors of HDR from which we obtained a number of hits that disrupted microtubule dynamics. We postulated that high levels of the target molecules of our screen in tumors would correlate with poor chemotherapy response. We found that inhibition or knockdown of dynamin 2 (DNM2), known for its role in endocytic cell trafficking and microtubule dynamics, impaired HDR and improved response to chemotherapy of cells and of tumors in mice. In a retrospective analysis, levels of DNM2 at the time of treatment strongly predicted chemotherapy outcome for estrogen receptor-negative and especially for TNBC patients. We propose that DNM2-associated DNA repair enzyme trafficking is important for HDR efficiency and is a powerful predictor of sensitivity to breast cancer chemotherapy and an important target for therapy.
Collapse
Affiliation(s)
- Sophia B. Chernikova
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Rochelle B. Nguyen
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Jessica T. Truong
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Stephano S. Mello
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Jason H. Stafford
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Michael P. Hay
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | - Douglas J. Wood
- Data Coordinating Center, Department of Biomedical Data Science, and
| | - Solomon Henry
- Data Coordinating Center, Department of Biomedical Data Science, and
| | - Rie von Eyben
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Lei Deng
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | | | - Asaithamby Aroumougame
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Claudia Wiese
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - John C. Game
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
- Semmelweis University 2nd Department of Pediatrics, Budapest, Hungary
| | - J. Martin Brown
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| |
Collapse
|
31
|
Lee MG, Lee KS, Nam KS. The association of changes in RAD51 and survivin expression levels with the proton beam sensitivity of Capan‑1 and Panc‑1 human pancreatic cancer cells. Int J Oncol 2018; 54:744-752. [PMID: 30483758 DOI: 10.3892/ijo.2018.4642] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/11/2018] [Indexed: 11/05/2022] Open
Abstract
Fewer than 20% of patients diagnosed with pancreatic cancer can be treated with surgical resection. The effects of proton beam irradiation were evaluated on the cell viabilities in Panc‑1 and Capan‑1 pancreatic cancer cells. The cells were irradiated with proton beams at the center of Bragg peaks with a 6‑cm width using a proton accelerator. Cell proliferation was assessed with the MTT assay, gene expression was analyzed with semi‑quantitative or quantitative reverse transcription‑polymerase chain reaction analyses and protein expression was evaluated by western blotting. The results demonstrated that Capan‑1 cells had lower cell viability than Panc‑1 cells at 72 h after proton beam irradiation. Furthermore, the cleaved poly (ADP‑ribose) polymerase protein level was increased by irradiation in Capan‑1 cells, but not in Panc‑1 cells. Additionally, it was determined that histone H2AX phosphorylation in the two cell lines was increased by irradiation. Although a 16 Gy proton beam was only slightly up‑regulated cyclin‑dependent kinase inhibitor 1 (p21) protein expression in Capan‑1 cells, p21 expression levels in Capan‑1 and Panc‑1 cells were significantly increased at 72 h after irradiation. Furthermore, it was observed that the expression of DNA repair protein RAD51 homolog 1 (RAD51), a homogenous repair enzyme, was decreased in what appeared to be a dose‑dependent manner by irradiation in Capan‑1 cells. Contrastingly, the transcription of survivin in Panc‑1 was significantly enhanced. The results suggest that RAD51 and survivin are potent markers that determine the therapeutic efficacy of proton beam therapy in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Min-Gu Lee
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju, Gyeongsanbuk-do 38066, Republic of Korea
| | - Kyu-Shik Lee
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju, Gyeongsanbuk-do 38066, Republic of Korea
| | - Kyung-Soo Nam
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju, Gyeongsanbuk-do 38066, Republic of Korea
| |
Collapse
|
32
|
Li BX, Chen J, Chao B, Zheng Y, Xiao X. A Lamin-Binding Ligand Inhibits Homologous Recombination Repair of DNA Double-Strand Breaks. ACS CENTRAL SCIENCE 2018; 4:1201-1210. [PMID: 30276254 PMCID: PMC6161055 DOI: 10.1021/acscentsci.8b00379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Indexed: 05/02/2023]
Abstract
Nuclear lamins are type V intermediate filament proteins. Lamins, including LA, LB1, LB2, and LC, are the major protein components forming the nuclear lamina to support the mechanical stability of the mammalian cell nucleus. Increasing evidence has shown that LA participates in homologous recombination (HR) repair of DNA double-strand breaks (DSBs) . However, the mechanisms underlying this process are incompletely understood. We recently identified the first lamin-binding ligand 1 (LBL1) that directly binds LA and inhibited cancer cell growth. We provided here further mechanistic investigations of LBL1 and revealed that LA interacts with the HR recombinase Rad51 to protect Rad51 from degradation. LBL1 inhibits LA-Rad51 interaction leading to accelerated proteasome-mediated degradation of Rad51, culminating in inhibition of HR repair of DSBs. These results uncover a novel post-translational regulation of Rad51 by LA and suggest that targeting the LA-Rad51 axis may represent a promising strategy to develop cancer therapeutics.
Collapse
Affiliation(s)
- Bingbing X. Li
- Program
in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
- E-mail:
| | - Jingjin Chen
- Program
in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Bo Chao
- Program
in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Yixian Zheng
- Department
of Embryology, Carnegie Institution for
Science, 3520 San Martin
Drive, Baltimore, Maryland 21218, United States
| | - Xiangshu Xiao
- Program
in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
- Knight
Cancer Institute, Oregon Health & Science
University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
- E-mail:
| |
Collapse
|
33
|
Nonstructural Protein 5A Impairs DNA Damage Repair: Implications for Hepatitis C Virus-Mediated Hepatocarcinogenesis. J Virol 2018; 92:JVI.00178-18. [PMID: 29563287 DOI: 10.1128/jvi.00178-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/11/2018] [Indexed: 01/01/2023] Open
Abstract
RAD51-associated protein 1 (RAD51AP1) is a member of the multiprotein complexes postulated to carry out RAD51-mediated homologous recombination and DNA repair in mammalian cells. In the present study, we showed that hepatitis C virus (HCV) NS5A directly bound RAD51AP1 and increased the protein level of RAD51AP1 through modulation of the ubiquitin-proteasome pathway. We also demonstrated that RAD51AP1 protein levels were increased in the liver tissues of HCV-infected patients and NS5A-transgenic mice. Importantly, NS5A impaired DNA repair by disrupting the RAD51/RAD51AP1/UAF1 complex and rendered HCV-infected cells more sensitive to DNA damage. Silencing of RAD51AP1 expression resulted in a decrease of viral propagation. We further demonstrated that RAD51AP1 was involved in the assembly step of the HCV life cycle by protecting viral RNA. These data suggest that HCV exploits RAD51AP1 to promote viral propagation and thus that host DNA repair is compromised in HCV-infected cells. Overall, our findings provide mechanistic insight into the pathogenesis of HCV infection.IMPORTANCE Chronic infection with HCV is the leading cause of hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying HCV-induced HCC are not fully understood. Here we demonstrate that the HCV NS5A protein physically interacts with RAD51AP1 and increases the RAD51AP1 protein level through modulation of the ubiquitin-proteasome pathway. HCV coopts host RAD51AP1 to protect viral RNA at an assembly step of the HCV life cycle. Note that the RAD51 protein accumulates in the cytoplasm of HCV-infected cells, and thus the RAD51/RAD51AP1/UAF1-mediated DNA damage repair system in the nucleus is compromised in HCV-infected cells. Our data may provide new insight into the molecular mechanisms of HCV-induced pathogenesis.
Collapse
|
34
|
Ginsenosides synergize with mitomycin C in combating human non-small cell lung cancer by repressing Rad51-mediated DNA repair. Acta Pharmacol Sin 2018; 39:449-458. [PMID: 28836581 DOI: 10.1038/aps.2017.53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/05/2017] [Indexed: 02/06/2023]
Abstract
The use of ginseng extract as an adjuvant for cancer treatment has been reported in both animal models and clinical applications, but its molecular mechanisms have not been fully elucidated. Mitomycin C (MMC), an anticancer antibiotic used as a first- or second-line regimen in the treatment for non-small cell lung carcinoma (NSCLC), causes serious adverse reactions when used alone. Here, by using both in vitro and in vivo experiments, we provide evidence for an optimal therapy for NSCLC with total ginsenosides extract (TGS), which significantly enhanced the MMC-induced cytotoxicity against NSCLC A549 and PC-9 cells in vitro when used in combination with relatively low concentrations of MMC. A NSCLC xenograft mouse model was used to confirm the in vivo synergistic effects of the combination of TGS with MMC. Further investigation revealed that TGS could significantly reverse MMC-induced S-phase cell cycle arrest and inhibit Rad51-mediated DNA damage repair, which was evidenced by the inhibitory effects of TGS on the levels of phospho-MEK1/2, phospho-ERK1/2 and Rad51 protein and the translocation of Rad51 from the cytoplasm to the nucleus in response to MMC. In summary, our results demonstrate that TGS could effectively enhance the cytotoxicity of MMC against NSCLC cells in vitro and in vivo, thereby revealing a novel adjuvant anticancer mechanism of TGS. Combined treatment with TGS and MMC can significantly lower the required concentration of MMC and can further reduce the risk of side effects, suggesting a better treatment option for NSCLC patients.
Collapse
|
35
|
Turchick A, Hegan DC, Jensen RB, Glazer PM. A cell-penetrating antibody inhibits human RAD51 via direct binding. Nucleic Acids Res 2017; 45:11782-11799. [PMID: 29036688 PMCID: PMC5714174 DOI: 10.1093/nar/gkx871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/16/2017] [Indexed: 12/11/2022] Open
Abstract
RAD51, a key factor in homology-directed repair (HDR), has long been considered an attractive target for cancer therapy, but few specific inhibitors have been found. A cell-penetrating, anti-DNA, lupus autoantibody, 3E10, was previously shown to inhibit HDR, sensitize tumors to radiation, and mediate synthetic lethal killing of BRCA2-deficient cancer cells, effects that were initially attributed to its affinity for DNA. However, as the molecular basis for its ability to inhibit DNA repair, we report that 3E10 directly binds to the N-terminus of RAD51, sequesters RAD51 in the cytoplasm, and impedes RAD51 binding to DNA. Further, we generate separation-of-function mutations in the complementarity-determining regions of 3E10 revealing that inhibition of HDR tracks with binding to RAD51 but not to DNA, whereas cell penetration is linked to DNA binding. The consequences of these mutations on putative 3E10 interactions with RAD51 and DNA are correlated with in silico molecular modeling. Taken together, the results identify 3E10 as a novel inhibitor of RAD51 by direct binding, accounting for its ability to suppress HDR and providing the molecular basis to guide pre-clinical development of 3E10 as an anti-cancer agent.
Collapse
Affiliation(s)
- Audrey Turchick
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Denise C Hegan
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ryan B Jensen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Peter M Glazer
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.,Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
36
|
Dai J, Voloshin O, Potapova S, Camerini-Otero RD. Meiotic Knockdown and Complementation Reveals Essential Role of RAD51 in Mouse Spermatogenesis. Cell Rep 2017; 18:1383-1394. [PMID: 28178517 DOI: 10.1016/j.celrep.2017.01.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/30/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022] Open
Abstract
Meiotic homologous recombination (HR) is important for proper chromosomal segregation during gametogenesis and facilitates evolutionary adaptation via genomic reshuffling. In most eukaryotes, HR is mediated by two recombinases, the ubiquitous RAD51 and the meiosis-specific DMC1. The role of RAD51 in mammalian meiosis is unclear and study of its function is limited due to embryonic lethality of RAD51 knockouts. Here, we developed an in vivo meiotic knockdown and protein complementation system to study RAD51 during mouse spermatogenesis. We show that RAD51 is crucial during meiotic prophase and its loss leads to depletion of late prophase I spermatocytes through a p53-dependent apoptotic pathway. This phenotype is distinct from that observed in the DMC1 knockdown. Our meiotic knockdown and complementation system establishes an experimental platform for mechanistic studies of meiotic proteins with unknown functions or essential genes for which a testis-specific knockout is not possible.
Collapse
Affiliation(s)
- Jieqiong Dai
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Oleg Voloshin
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Svetlana Potapova
- Division of Intramural Research, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - R Daniel Camerini-Otero
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Venkatachalam S, Mettler E, Fottner C, Miederer M, Kaina B, Weber MM. The impact of the IGF-1 system of cancer cells on radiation response - An in vitro study. Clin Transl Radiat Oncol 2017; 7:1-8. [PMID: 29594222 PMCID: PMC5862664 DOI: 10.1016/j.ctro.2017.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/29/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022] Open
Abstract
Background Overexpression of the insulin-like growth factor-1 receptor (IGF-1R) is associated with increased cell proliferation, differentiation, transformation, and tumorigenicity. Additionally, signaling involved in the resistance of cancer cells to radiotherapy originates from IGF-1R. The purpose of this study was to investigate the role of the IGF-1 system in the radiation response and further evaluate its effect on the expression of DNA repair pathway genes. Methods To inhibit the IGF-1 system, we stably transfected the Caco-2 cell line to express a kinase-deficient IGF-1R mutant. We then studied the effects of this mutation on cell growth, the response to radiation, and clonogenic survival, as well as using a cell viability assay to examine DNA damage and repair. Finally, we performed immunofluorescence for γ-H2AX to examine double-strand DNA breaks and evaluated the expression of 84 key genes involved in DNA repair with a real-time PCR array. Results Mutant IGF-1R cells exhibited significantly blunted cell growth and viability, compared to wild-type cells, as well as reduced clonogenic survival after γ-irradiation. However, mutant IGF-1R cells did not show any significant delays in the repair of radiation-induced DNA double-strand breaks. Furthermore, expression of mutant IGF-1R significantly down-regulated the mRNA levels of BRCA2, a major protein involved in homologous recombination DNA repair. Conclusion These results indicate that blocking the IGF-1R-mediated signaling cascade, through the expression of a kinase-deficient IGF-1R mutant, reduces cell growth and sensitizes cancer cells to ionizing radiation. Therefore, the IGF-1R system could be a potential target to enhance radio-sensitivity and the efficacy of cancer treatments.
Collapse
Key Words
- BAX, BCL-2-associated X
- BCL-2, B-cell lymphoma 2
- BRCA2
- Caco-2-KR4, IGF-1R/KR clone number 4
- Colorectal carcinmoma
- Dominant negative mutant
- HRR, homologous recombination repair
- IGF-1R, insulin-like growth factor 1 receptor
- IGF-1R/KR, kinase-deficient IGF-1R
- IRS-1, insulin receptor substrate 1
- Insulin-like growth factor-1 receptor
- MVP, major vault protein
- NHEJ, non-homologous end joining
- PTEN, phosphatase and tensin homolog
- RAD 51
- Radiosensitivity
- SF, surviving fractions
Collapse
Affiliation(s)
| | - Esther Mettler
- Department of Endocrinology and Metabolic Diseases, University Medical Center, Mainz, Germany
- Corresponding author.
| | - Christian Fottner
- Department of Endocrinology and Metabolic Diseases, University Medical Center, Mainz, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Medical Center, Mainz, Germany
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, Mainz, Germany
| | - Matthias M. Weber
- Department of Endocrinology and Metabolic Diseases, University Medical Center, Mainz, Germany
| |
Collapse
|
38
|
Browning CL, Wise JP. Prolonged exposure to particulate chromate inhibits RAD51 nuclear import mediator proteins. Toxicol Appl Pharmacol 2017; 331:101-107. [PMID: 28554658 PMCID: PMC5568470 DOI: 10.1016/j.taap.2017.05.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Particulate hexavalent chromium (Cr(VI)) is a human lung carcinogen and a human health concern. The induction of structural chromosome instability is considered to be a driving mechanism of Cr(VI)-induced carcinogenesis. Homologous recombination repair protects against Cr(VI)-induced chromosome damage, due to its highly accurate repair of Cr(VI)-induced DNA double strand breaks. However, recent studies demonstrate Cr(VI) inhibits homologous recombination repair through the misregulation of RAD51. RAD51 is an essential protein in HR repair that facilitates the search for a homologous sequence. Recent studies show prolonged Cr(VI) exposure prevents proper RAD51 subcellular localization, causing it to accumulate in the cytoplasm. Since nuclear import of RAD51 is crucial to its function, this study investigated the effect of Cr(VI) on the RAD51 nuclear import mediators, RAD51C and BRCA2. We show acute (24h) Cr(VI) exposure induces the proper localization of RAD51C and BRCA2. In contrast, prolonged (120h) exposure increased the cytoplasmic localization of both proteins, although RAD51C localization was more severely impaired. These results correlate temporally with the previously reported Cr(VI)-induced RAD51 cytoplasmic accumulation. In addition, we found Cr(VI) does not inhibit interaction between RAD51 and its nuclear import mediators. Altogether, our results suggest prolonged Cr(VI) exposure inhibits the nuclear import of RAD51C, and to a lesser extent, BRCA2, which results in the cytoplasmic accumulation of RAD51. Cr(VI)-induced inhibition of nuclear import may play a key role in its carcinogenic mechanism since the nuclear import of many tumor suppressor proteins and DNA repair proteins is crucial to their function.
Collapse
Affiliation(s)
- Cynthia L Browning
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, United States; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, United States.
| | - John Pierce Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, United States; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, United States.
| |
Collapse
|
39
|
van de Ven AL, Tangutoori S, Baldwin P, Qiao J, Gharagouzloo C, Seitzer N, Clohessy JG, Makrigiorgos GM, Cormack R, Pandolfi PP, Sridhar S. Nanoformulation of Olaparib Amplifies PARP Inhibition and Sensitizes PTEN/TP53-Deficient Prostate Cancer to Radiation. Mol Cancer Ther 2017; 16:1279-1289. [PMID: 28500233 DOI: 10.1158/1535-7163.mct-16-0740] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/09/2017] [Accepted: 04/28/2017] [Indexed: 12/17/2022]
Abstract
The use of PARP inhibitors in combination with radiotherapy is a promising strategy to locally enhance DNA damage in tumors. Here we show that radiation-resistant cells and tumors derived from a Pten/Trp53-deficient mouse model of advanced prostate cancer are rendered radiation sensitive following treatment with NanoOlaparib, a lipid-based injectable nanoformulation of olaparib. This enhancement in radiosensitivity is accompanied by radiation dose-dependent changes in γ-H2AX expression and is specific to NanoOlaparib alone. In animals, twice-weekly intravenous administration of NanoOlaparib results in significant tumor growth inhibition, whereas previous studies of oral olaparib as monotherapy have shown no therapeutic efficacy. When NanoOlaparib is administered prior to radiation, a single dose of radiation is sufficient to triple the median mouse survival time compared to radiation only controls. Half of mice treated with NanoOlaparib + radiation achieved a complete response over the 13-week study duration. Using ferumoxytol as a surrogate nanoparticle, MRI studies revealed that NanoOlaparib enhances the intratumoral accumulation of systemically administered nanoparticles. NanoOlaparib-treated tumors showed up to 19-fold higher nanoparticle accumulation compared to untreated and radiation-only controls, suggesting that the in vivo efficacy of NanoOlaparib may be potentiated by its ability to enhance its own accumulation. Together, these data suggest that NanoOlaparib may be a promising new strategy for enhancing the radiosensitivity of radiation-resistant tumors lacking BRCA mutations, such as those with PTEN and TP53 deletions. Mol Cancer Ther; 16(7); 1279-89. ©2017 AACR.
Collapse
Affiliation(s)
- Anne L van de Ven
- Department of Physics, Northeastern University, Boston, Massachusetts.,Nanomedicine Science & Technology Center, Northeastern University, Boston, Massachusetts
| | - Shifalika Tangutoori
- Nanomedicine Science & Technology Center, Northeastern University, Boston, Massachusetts.,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Paige Baldwin
- Nanomedicine Science & Technology Center, Northeastern University, Boston, Massachusetts.,Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Ju Qiao
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts
| | - Codi Gharagouzloo
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Nina Seitzer
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, Massachusetts
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, Massachusetts
| | - G Mike Makrigiorgos
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert Cormack
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, Massachusetts
| | - Srinivas Sridhar
- Department of Physics, Northeastern University, Boston, Massachusetts. .,Nanomedicine Science & Technology Center, Northeastern University, Boston, Massachusetts.,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
40
|
Reindl J, Girst S, Walsh DWM, Greubel C, Schwarz B, Siebenwirth C, Drexler GA, Friedl AA, Dollinger G. Chromatin organization revealed by nanostructure of irradiation induced γH2AX, 53BP1 and Rad51 foci. Sci Rep 2017; 7:40616. [PMID: 28094292 PMCID: PMC5240115 DOI: 10.1038/srep40616] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/07/2016] [Indexed: 11/28/2022] Open
Abstract
The spatial distribution of DSB repair factors γH2AX, 53BP1 and Rad51 in ionizing radiation induced foci (IRIF) in HeLa cells using super resolution STED nanoscopy after low and high linear energy transfer (LET) irradiation was investigated. 53BP1 and γH2AX form IRIF with same mean size of (540 ± 40) nm after high LET irradiation while the size after low LET irradiation is significantly smaller. The IRIF of both repair factors show nanostructures with partial anti-correlation. These structures are related to domains formed within the chromatin territories marked by γH2AX while 53BP1 is mainly situated in the perichromatin region. The nanostructures have a mean size of (129 ± 6) nm and are found to be irrespective of the applied LET and the labelled damage marker. In contrast, Rad51 shows no nanostructure and a mean size of (143 ± 13) nm independent of LET. Although Rad51 is surrounded by 53BP1 it strongly anti-correlates meaning an exclusion of 53BP1 next to DSB when decision for homologous DSB repair happened.
Collapse
Affiliation(s)
- Judith Reindl
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Stefanie Girst
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Dietrich W M Walsh
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany.,Department of Radiation Oncology, Technische Universitaet Muenchen, 81675 Munich, Germany
| | - Christoph Greubel
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Benjamin Schwarz
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Christian Siebenwirth
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany.,Department of Radiation Oncology, Technische Universitaet Muenchen, 81675 Munich, Germany
| | - Guido A Drexler
- Department of Radiation Oncology, Ludwig-Maximilians-Universitaet Muenchen, 80336 Munich, Germany
| | - Anna A Friedl
- Department of Radiation Oncology, Ludwig-Maximilians-Universitaet Muenchen, 80336 Munich, Germany
| | - Günther Dollinger
- Angewandte Physik und Messtechnik, Universitaet der Bundeswehr Muenchen, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| |
Collapse
|
41
|
Alayev A, Salamon RS, Manna S, Schwartz NS, Berman AY, Holz MK. Estrogen induces RAD51C expression and localization to sites of DNA damage. Cell Cycle 2016; 15:3230-3239. [PMID: 27753535 DOI: 10.1080/15384101.2016.1241927] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Homologous recombination (HR) is a conserved process that maintains genome stability and cell survival by repairing DNA double-strand breaks (DSBs). The RAD51-related family of proteins is involved in repair of DSBs; consequently, deregulation of RAD51 causes chromosomal rearrangements and stimulates tumorigenesis. RAD51C has been identified as a potential tumor suppressor and a breast and ovarian cancer susceptibility gene. Recent studies have also implicated estrogen as a DNA-damaging agent that causes DSBs. We found that in ERα-positive breast cancer cells, estrogen transcriptionally regulates RAD51C expression in ERα-dependent mechanism. Moreover, estrogen induces RAD51C assembly into nuclear foci at DSBs, which is a precursor to RAD51 complex recruitment to the nucleus. Additionally, disruption of ERα signaling by either anti-estrogens or siRNA prevented estrogen induced upregulation of RAD51C. We have also found an association of a worse clinical outcome between RAD51C expression and ERα status of tumors. These findings provide insight into the mechanism of genomic instability in ERα-positive breast cancer and suggest that individuals with mutations in RAD51C that are exposed to estrogen would be more susceptible to accumulation of DNA damage, leading to cancer progression.
Collapse
Affiliation(s)
- Anya Alayev
- a Department of Biology , Yeshiva University , New York , NY , USA
| | - Rachel S Salamon
- a Department of Biology , Yeshiva University , New York , NY , USA
| | - Subrata Manna
- a Department of Biology , Yeshiva University , New York , NY , USA
| | - Naomi S Schwartz
- a Department of Biology , Yeshiva University , New York , NY , USA
| | - Adi Y Berman
- a Department of Biology , Yeshiva University , New York , NY , USA
| | - Marina K Holz
- a Department of Biology , Yeshiva University , New York , NY , USA.,b Department of Molecular Pharmacology and the Albert Einstein Cancer Center , Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
42
|
Senavirathne G, Mahto SK, Hanne J, O'Brian D, Fishel R. Dynamic unwrapping of nucleosomes by HsRAD51 that includes sliding and rotational motion of histone octamers. Nucleic Acids Res 2016; 45:685-698. [PMID: 27738136 PMCID: PMC5314800 DOI: 10.1093/nar/gkw920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/01/2016] [Accepted: 10/10/2016] [Indexed: 01/24/2023] Open
Abstract
Wrapping of genomic DNA into nucleosomes poses thermodynamic and kinetic barriers to biological processes such as replication, transcription, repair and recombination. Previous biochemical studies have demonstrated that in the presence of adenosine triphosphate (ATP) the human RAD51 (HsRAD51) recombinase can form a nucleoprotein filament (NPF) on double-stranded DNA (dsDNA) that is capable of unwrapping the nucleosomal DNA from the histone octamer (HO). Here, we have used single molecule Förster Resonance Energy Transfer (smFRET) to examine the real time nucleosome dynamics in the presence of the HsRAD51 NPF. We show that oligomerization of HsRAD51 leads to stepwise, but stochastic unwrapping of the DNA from the HO in the presence of ATP. The highly reversible dynamics observed in single-molecule trajectories suggests an antagonistic mechanism between HsRAD51 binding and rewrapping of the DNA around the HO. These stochastic dynamics were independent of the nucleosomal DNA sequence or the asymmetry created by the presence of a linker DNA. We also observed sliding and rotational oscillations of the HO with respect to the nucleosomal DNA. These studies underline the dynamic nature of even tightly associated protein-DNA complexes such as nucleosomes.
Collapse
Affiliation(s)
- Gayan Senavirathne
- Department of Cancer Biology and Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Santosh K Mahto
- Department of Cancer Biology and Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Jeungphill Hanne
- Department of Cancer Biology and Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Daniel O'Brian
- Department of Cancer Biology and Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Richard Fishel
- Department of Cancer Biology and Genetics, The Ohio State University Medical Center, Columbus, OH 43210, USA .,Physics Department, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
43
|
The role of Rad51 in safeguarding mitochondrial activity during the meiotic cell cycle in mammalian oocytes. Sci Rep 2016; 6:34110. [PMID: 27677401 PMCID: PMC5039699 DOI: 10.1038/srep34110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/07/2016] [Indexed: 12/03/2022] Open
Abstract
Rad51 is a conserved eukaryotic protein that mediates the homologous recombination repair of DNA double-strand breaks that occur during mitosis and meiosis. In addition, Rad51 promotes mitochondrial DNA synthesis when replication stress is increased. Rad51 also regulates cell cycle progression by preserving the G2/M transition in embryonic stem cells. In this study, we report a novel function of Rad51 in regulating mitochondrial activity during in vitro maturation of mouse oocytes. Suppression of Rad51 by injection of Rad51 dsRNA into germinal vesicle-stage oocytes resulted in arrest of meiosis in metaphase I. Rad51-depleted oocytes showed chromosome misalignment and failures in spindle aggregation, affecting the completion of cytokinesis. We found that Rad51 depletion was accompanied by decreased ATP production and mitochondrial membrane potential and increased DNA degradation. We further demonstrated that the mitochondrial defect activated autophagy in Rad51-depleted oocytes. Taken together, we concluded that Rad51 functions to safeguard mitochondrial integrity during the meiotic maturation of oocytes.
Collapse
|
44
|
Foertsch F, Szambowska A, Weise A, Zielinski A, Schlott B, Kraft F, Mrasek K, Borgmann K, Pospiech H, Grosse F, Melle C. S100A11 plays a role in homologous recombination and genome maintenance by influencing the persistence of RAD51 in DNA repair foci. Cell Cycle 2016; 15:2766-79. [PMID: 27590262 DOI: 10.1080/15384101.2016.1220457] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) is an essential process in maintenance of chromosomal stability. A key player of HR is the strand exchange factor RAD51 whose assembly at sites of DNA damage is tightly regulated. We detected an endogenous complex of RAD51 with the calcium-binding protein S100A11, which is localized at sites of DNA repair in HaCaT cells as well as in normal human epidermal keratinocytes (NHEK) synchronized in S phase. In biochemical assays, we revealed that S100A11 enhanced the RAD51 strand exchange activity. When cells expressing a S100A11 mutant lacking the ability to bind Ca(2+), a prolonged persistence of RAD51 in repair sites and nuclear γH2AX foci was observed suggesting an incomplete DNA repair. The same phenotype became apparent when S100A11 was depleted by RNA interference. Furthermore, down-regulation of S100A11 resulted in both reduced sister chromatid exchange confirming the restriction of the recombination capacity of the cells, and in an increase of chromosomal aberrations reflecting the functional requirement of S100A11 for the maintenance of genomic stability. Our data indicate that S100A11 is involved in homologous recombination by regulating the appearance of RAD51 in DSB repair sites. This function requires the calcium-binding activity of S100A11.
Collapse
Affiliation(s)
- Franziska Foertsch
- a Biomolecular Photonics Group , Jena University Hospital , Jena , Germany
| | - Anna Szambowska
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany
| | - Anja Weise
- c Institute of Human Genetics , Jena University Hospital , Jena , Germany
| | - Alexandra Zielinski
- d Radiobiology & Experimental Radiooncology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Bernhard Schlott
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany
| | - Florian Kraft
- c Institute of Human Genetics , Jena University Hospital , Jena , Germany
| | - Kristin Mrasek
- c Institute of Human Genetics , Jena University Hospital , Jena , Germany
| | - Kerstin Borgmann
- d Radiobiology & Experimental Radiooncology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Helmut Pospiech
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany.,e Faculty of Biochemistry and Molecular Medicine , University of Oulu , Finland
| | - Frank Grosse
- b Research Group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute , Jena , Germany
| | - Christian Melle
- a Biomolecular Photonics Group , Jena University Hospital , Jena , Germany
| |
Collapse
|
45
|
Alshareeda AT, Negm OH, Aleskandarany MA, Green AR, Nolan C, TigHhe PJ, Madhusudan S, Ellis IO, Rakha EA. Clinical and biological significance of RAD51 expression in breast cancer: a key DNA damage response protein. Breast Cancer Res Treat 2016; 159:41-53. [DOI: 10.1007/s10549-016-3915-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 07/15/2016] [Indexed: 01/15/2023]
|
46
|
Browning CL, Qin Q, Kelly DF, Prakash R, Vanoli F, Jasin M, Wise JP. Prolonged Particulate Hexavalent Chromium Exposure Suppresses Homologous Recombination Repair in Human Lung Cells. Toxicol Sci 2016; 153:70-8. [PMID: 27449664 DOI: 10.1093/toxsci/kfw103] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Genomic instability is one of the primary models of carcinogenesis and a feature of almost all cancers. Homologous recombination (HR) repair protects against genomic instability by maintaining high genomic fidelity during the repair of DNA double strand breaks. The defining step of HR repair is the formation of the Rad51 nucleofilament, which facilitates the search for a homologous sequence and invasion of the template DNA strand. Particulate hexavalent chromium (Cr(VI)), a human lung carcinogen, induces DNA double strand breaks and chromosome instability. Since the loss of HR repair increases Cr(VI)-induced chromosome instability, we investigated the effect of extended Cr(VI) exposure on HR repair. We show acute (24 h) Cr(VI) exposure induces a normal HR repair response. In contrast, prolonged (120 h) exposure to particulate Cr(VI) inhibited HR repair and Rad51 nucleofilament formation. Prolonged Cr(VI) exposure had a profound effect on Rad51, evidenced by reduced protein levels and Rad51 mislocalization to the cytoplasm. The response of proteins involved in Rad51 nuclear import and nucleofilament formation displayed varying responses to prolonged Cr(VI) exposure. BRCA2 formed nuclear foci after prolonged Cr(VI) exposure, while Rad51C foci formation was suppressed. These results suggest that particulate Cr(VI), a major chemical carcinogen, inhibits HR repair by targeting Rad51, causing DNA double strand breaks to be repaired by a low fidelity, Rad51-independent repair pathway. These results further enhance our understanding of the underlying mechanism of Cr(VI)-induced chromosome instability and thus, carcinogenesis.
Collapse
Affiliation(s)
- Cynthia L Browning
- *Wise Laboratory of Environmental and Genetic Toxicology, Portland, Maine 04104 Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine 04469
| | - Qin Qin
- *Wise Laboratory of Environmental and Genetic Toxicology, Portland, Maine 04104 Virginia Tech Carilion Research Institute, Roanoke, Virginia 24016
| | - Deborah F Kelly
- Virginia Tech Carilion Research Institute, Roanoke, Virginia 24016
| | - Rohit Prakash
- Memorial Sloan Kettering Cancer Center, New York 10065, New York
| | - Fabio Vanoli
- Memorial Sloan Kettering Cancer Center, New York 10065, New York
| | - Maria Jasin
- Memorial Sloan Kettering Cancer Center, New York 10065, New York
| | - John Pierce Wise
- *Wise Laboratory of Environmental and Genetic Toxicology, Portland, Maine 04104 Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine 04469
| |
Collapse
|
47
|
Feng W, Deng Y, Wu J, Zhang H, Liang J, Xian H, Yang S. Overexpression of BRCA1 attenuates the sensitivity of PC9 cells to gefitinib. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:12869-12876. [PMID: 26722478 PMCID: PMC4680423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/25/2015] [Indexed: 06/05/2023]
Abstract
Gefitinib is an orally active antitumor agent which inhibits uncontrolled cell proliferation by interrupting epidermal growth factor receptor (EGFR) signaling pathways. Various in vitro and in vivo studies have revealed that the upregulated expression of breast cancer susceptibility gene 1 (BRCA1) is associated with chemoresistance and reduced survival following chemotherapies. In this study, a gefitinib-highly-sensitive cell line, PC-9, was used to investigate the effect of BRCA1 expression on the sensitivity of PC-9 cells to gefitinib. PC-9 cells were stably transfected with BRCA-1 (HA-tagged). Transfected and untransfected PC-9 cells were treated with gefitinib, phosphorylated γH2AX was examined by western blot to determine the DNA damages. Following the treatment of gefitinib, the inhibition of proliferation of the PC-9 cells, PC-9-pcDNA3.1 cells, and BRCA1-transfected PC-9 cells were determined. Also, a comet assay was performed to determine the DNA damage caused by gefitinib. The treatment of gefitinib for 6 hr, 12 h, and 24 hr significantly increased the cellular expression of phosphorylated γH2AX. With the treatment of gefitinib, the inhibition of proliferation of BRCA-1 overexpressed PC-9 cells was significantly lower than that of the non-transfected PC-9 cells, indicating the overexpression of BRCA1 plays a role in attenuating the sensitivity of PC-9 cells to gefitinib. The comet assay revealed that BRCA1 transfected cells showed a shorter comet tail, indicating the overexpression of BRCA1 attenuated the DNA damages caused by gefitinib. The overexpression of BRCA1 reduced the DNA damages, and enhanced DNA repair mechanisms. Also, gefitinib-mediated inhibition of cell proliferation is attenuated by the expression of BRCA1.
Collapse
Affiliation(s)
- Weineng Feng
- Department of Head and Neck/Thoracic Medical Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| | - Yanming Deng
- Department of Head and Neck/Thoracic Medical Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| | - Jing Wu
- Institute of Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| | - Hua Zhang
- Department of Head and Neck/Thoracic Medical Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| | - Jianmao Liang
- Department of Head and Neck/Thoracic Medical Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| | - Haibing Xian
- Department of Head and Neck/Thoracic Medical Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| | - Shuang Yang
- Department of Head and Neck/Thoracic Medical Oncology, The First People’s Hospital of FoshanFoshan 528000, Guangdong, P. R. China
| |
Collapse
|
48
|
Okimoto S, Sun J, Fukuto A, Horikoshi Y, Matsuda S, Matsuda T, Ikura M, Ikura T, Machida S, Kurumizaka H, Miyamoto Y, Oka M, Yoneda Y, Kiuchi Y, Tashiro S. hCAS/CSE1L regulates RAD51 distribution and focus formation for homologous recombinational repair. Genes Cells 2015; 20:681-94. [PMID: 26123175 DOI: 10.1111/gtc.12262] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/14/2015] [Indexed: 01/05/2023]
Abstract
Homologous recombinational repair (HR) is one of the major repair systems for DNA double-strand breaks. RAD51 is a key molecule in HR, and the RAD51 concentration in the cell nucleus increases after DNA damage induction. However, the mechanism that regulates the intracellular distribution of RAD51 is still unclear. Here, we show that hCAS/CSE1L associates with RAD51 in human cells. We found that hCAS/CSE1L negatively regulates the nuclear protein level of RAD51 under normal conditions. hCAS/CSE1L is also required to repress the DNA damage-induced focus formation of RAD51. Moreover, we show that hCAS/CSE1L plays roles in the regulation of the HR activity and in chromosome stability. These findings suggest that hCAS/CSE1L is responsible for controlling the HR activity by directly interacting with RAD51.
Collapse
Affiliation(s)
- Satoshi Okimoto
- Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8551, Japan.,Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Jiying Sun
- Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Atsuhiko Fukuto
- Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8551, Japan.,Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Yasunori Horikoshi
- Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8551, Japan.,Research Center for the Mathematics on Chromatin Live Dynamics, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Shun Matsuda
- Research Center for Environmental Quality Management, Kyoto University, Otsu, Shiga, 520-0811, Japan
| | - Tomonari Matsuda
- Research Center for Environmental Quality Management, Kyoto University, Otsu, Shiga, 520-0811, Japan
| | - Masae Ikura
- Laboratory of Chromatin Regulatory Network, Department of Mutagenesis, Radiation Biology Center, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tsuyoshi Ikura
- Laboratory of Chromatin Regulatory Network, Department of Mutagenesis, Radiation Biology Center, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shinichi Machida
- Laboratory of Structural Biology, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hitoshi Kurumizaka
- Laboratory of Structural Biology, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Yoichi Miyamoto
- National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Masahiro Oka
- National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yoshihiro Yoneda
- National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Satoshi Tashiro
- Department of Cellular Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8551, Japan.,Research Center for the Mathematics on Chromatin Live Dynamics, Hiroshima University, Hiroshima, 734-8551, Japan
| |
Collapse
|
49
|
Zhong X, Luo G, Zhou X, Luo W, Wu X, Zhong R, Wang Y, Xu F, Wang J. Rad51 in regulating the radiosensitivity of non-small cell lung cancer with different epidermal growth factor receptor mutation status. Thorac Cancer 2015; 7:50-60. [PMID: 26816539 PMCID: PMC4718133 DOI: 10.1111/1759-7714.12274] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 04/12/2015] [Indexed: 02/05/2023] Open
Abstract
Background Non‐small cell lung cancer (NSCLC) harboring kinase‐domain mutations in epidermal growth factor receptors (EGFR) has been observed to be sensitive to ionizing radiation (IR). We explore Rad51‐dependent homologous recombination (HR) DNA repair in regulating radiosensitivity in two NSCLC cell lines with different EGFR mutation status. Methods NSCLC cell lines, wild‐type EGFR A549 and mutant EGFR H820 with an in‐frame deletion in exon 19 of EGFR (ΔE746–E750), were cultured. Radiosensitivity was estimated by colony forming assay. Rad51 expression was evaluated by quantitative real time‐polymerase chain reaction and Western‐blot. Lentiviral small hairpin ribonucleic acid‐Rad51 and ΔE746–E750 deletion mutant EGFR were constructed and transfected into cells. Flowcytometry assay was used to analyze DNA double strand breaks, cell cycle alterations, and apoptosis. Results A549 had a higher survival factor (SF)2 (0.66 vs. 0.44) and lower α/β value (4.07 vs. 9.01). Compared with the A549 cell, the H820 cell exhibited defective arrest in the S‐phase, a higher rate of G2/M accumulation, early apoptosis, and residual γ‐H2AX. Downregulated Rad51 expression decreased SF2 (0.42 vs. 0.31) and increased the α/β ratio (7.51 vs. 10.5), G2/M accumulation, early apoptosis, and γ‐H2AX in two cell lines. H820 had a low IR‐induced Rad51 expression and nuclear translocation. Exogenous expression of the ΔE746–E750 deletion mutant EGFR caused the A549 cell to become more radiosensitive. Conclusions An EGFR mutated NSCLC cell line is sensitive to IR, which is correlated with reduced IR‐induced Rad51 expression and nuclear translocation. The signaling pathway of EGFR maintaining Rad51 protein levels maybe a novel lung cancer therapeutic target to overcome radioresistance.
Collapse
Affiliation(s)
- Xing Zhong
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Guomin Luo
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Xiaojuan Zhou
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Wen Luo
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Xia Wu
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Renming Zhong
- State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Yanping Wang
- Laboratory of Molecular Diagnosis of Cancer West China Hospital Sichuan University Chengdu China
| | - Feng Xu
- State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China; Abdominal oncology West China Hospital Sichuan University Chengdu China
| | - Jin Wang
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| |
Collapse
|
50
|
Aleskandarany M, Caracappa D, Nolan CC, Macmillan RD, Ellis IO, Rakha EA, Green AR. DNA damage response markers are differentially expressed in BRCA-mutated breast cancers. Breast Cancer Res Treat 2015; 150:81-90. [PMID: 25690937 PMCID: PMC4344553 DOI: 10.1007/s10549-015-3306-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/09/2015] [Indexed: 01/07/2023]
Abstract
Cells have stringent DNA repair pathways that are specific for each different set of DNA lesions which is accomplished through the integration of complex array of proteins. However, BRCA-mutated breast cancer (BC) has defective DNA repair mechanisms. This study aims to investigate differential expression of a large panel of DNA repair markers to characterise DNA repair mechanisms in BRCA-associated tumours compared to sporadic tumours in an attempt to characterise these tumours in routine practice. Immunohistochemistry and tissue microarray technology were applied to a cohort of clinically annotated series of sporadic (n = 1849), BRCA1-mutated (n = 48), and BRCA2-mutated (n = 27) BC. The following DNA damage response (DDR) markers are used; BRCA1, BRCA2, RAD51, Ku70/Ku80, BARD, PARP1 (cleaved), PARP1 (non-cleaved), and P53 in addition to basal cytokeratins, ER, PR, and HER2. A significant proportion of BRCA1 tumours were positive for PARP1 (non-cleaved), and negative for BARD1 and RAD51 compared with sporadic BC. BRCA2 tumours were significantly positive for PARP1 (non-cleaved) compared with sporadic tumours. RAD51 was significantly higher in BRCA1 compared with BRCA2 tumours (p = 0.005). When BRCA1/2 BCs were compared to triple-negative (TN) sporadic tumours of the studied DDR proteins, BARD1 (p < 0.001), PARP1 (non-cleaved) (p < 0.001), and P53 (p = 0.002) remained significantly different in BRCA1/2 tumours compared with TN BC. DNA repair markers showed differential expression in BRCA-mutated tumours, with a substantial degree of disruption of DNA repair pathways in sporadic BC especially TN BC. DNA double-strand break (DSB) repair is assisted by PARP1 expression in BRCA-mutated tumours, whereas the loss of DSB repair via RAD51 is predominant in BRCA1 rather than BRCA2 BC.
Collapse
Affiliation(s)
- Mohammed Aleskandarany
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK,
| | | | | | | | | | | | | |
Collapse
|