401
|
Maidarti M, Clarkson YL, McLaughlin M, Anderson RA, Telfer EE. Inhibition of PTEN activates bovine non-growing follicles in vitro but increases DNA damage and reduces DNA repair response. Hum Reprod 2019; 34:297-307. [PMID: 30521029 PMCID: PMC6343469 DOI: 10.1093/humrep/dey354] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Does ovarian follicle activation by phosphatase homologue of chromosome-10 (PTEN) inhibition affect DNA damage and repair in bovine oocytes and granulosa cells? SUMMARY ANSWER PTEN inhibition promotes bovine non-growing follicle activation but results in increased DNA damage and impaired DNA repair capacity in ovarian follicles in vitro. WHAT IS KNOWN ALREADY Inhibition of PTEN is known to activate primordial follicles but may compromise further developmental potential. In breast cancer cells, PTEN inhibition represses nuclear translocation of breast cancer susceptibility 1 (BRCA1) and Rad51; this impairs DNA repair resulting in an accumulation of damaged DNA, which contributes to cell senescence. STUDY DESIGN, SIZE, DURATION Bovine ovarian tissue fragments were exposed to control medium alone or containing either 1 or 10 μM bpv(HOpic), a pharmacological inhibitor of PTEN, in vitro for 24 h. A sub-group of tissue fragments were collected for Western blot analysis after bpv(HOpic) exposure. The remainder were incubated in control medium for a further 5 days and then analysed histologically and by immunohistochemistry to detect DNA damage and repair pathways. PARTICIPANTS/MATERIALS, SETTING, METHODS Bovine ovaries were obtained from abattoir-slaughtered heifers. Tissue fragments were exposed to either control medium alone or medium containing either 1 μM or 10 μM bpv(HOpic) for 24 h. Tissue fragments collected after 24 h were subjected to Akt quantification by Western blotting (six to nine fragments per group per experiment). Follicle stage and morphology were classified in remaining fragments. Immunohistochemical analysis included nuclear exclusion of FOXO3 as a marker of follicle activation, γH2AX as a marker of DNA damage, meiotic recombination 11 (MRE11), ataxia telangiectasia mutated (ATM), Rad51, breast cancer susceptibility 1 (BRCA1) and breast cancer susceptibility 2 (BRCA2) as DNA repair factors. A total of 29 550 follicles from three independent experiments were analysed. MAIN RESULTS AND THE ROLE OF CHANCE Tissue fragments exposed to bpv(HOpic) had increased Akt phosphorylation at serine 473 (pAkt/Akt ratio, 2.25- and 6.23-fold higher in 1 and 10 μM bpv(HOpic) respectively compared to control, P < 0.05). These tissue fragments contained a significantly higher proportion of growing follicles compared to control (78.6% in 1 μM and 88.7% in 10 μM versus 70.5% in control; P < 0.001). The proportion of morphologically healthy follicles did not differ significantly between 1 μM bpv(HOpic) and control (P < 0.001) but follicle health was lower in 10 μM compared to 1 μM and control in all follicle types (P < 0.05). DNA damage in oocytes, indicated by expression of γH2AX, increased following exposure to 1 μM bpv(HOpic) (non-growing, 83%; primary follicles, 76%) and 10 μM (non-growing, 77%; primary, 84%) compared to control (non-growing, 30% and primary, 59%) (P < 0.05 for all groups). A significant reduction in expression of DNA repair proteins MRE11, ATM and Rad51 was observed in oocytes of non-growing and primary follicles of treatment groups (primary follicles in controls versus 10 μM bpv(HOpic): MRE, 68% versus 47%; ATM, 47% versus 18%; Rad51, 48% versus 24%), P < 0.05 for all groups. Higher dose bpv(HOpic) also resulted in lower expression of BRCA1 compared to control and 1 μM bpv(HOpic) (P < 0.001) in non-growing and primary follicles. BRCA2 expression was increased in oocytes of primary follicles in 1 μM bpv(HOpic) (36%) compared to control (20%, P = 0.010) with a marked decrease in 10 μM (1%, P ≤ 0.001). Granulosa cells of primary and secondary follicles in bpv(HOpic) groups showed more DNA damage compared to control (P < 0.05). However, bpv(HOpic) did not impact granulosa cell DNA repair capacity in secondary follicles, but BRCA1 declined significantly in higher dose bpv(HOpic). LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This study focuses on non-growing follicle activation after 6 days culture and may not reflect DNA damage and repair capacity in later stages of oocyte and follicle growth. WIDER IMPLICATIONS OF THE FINDINGS In vitro activation of follicle growth may compromise the bidirectional signalling between oocyte and granulosa cells necessary for optimal oocyte and follicle health. This large animal model may be useful in optimising follicle activation protocols with a view to transfer for clinical application. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by Indonesia endowment fund for education. No competing interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Mila Maidarti
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Institute of Cell Biology and Genes and Development Group, CDBS Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| | - Yvonne L Clarkson
- Institute of Cell Biology and Genes and Development Group, CDBS Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| | - Marie McLaughlin
- Institute of Cell Biology and Genes and Development Group, CDBS Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Institute of Cell Biology and Genes and Development Group, CDBS Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
402
|
Lee K, Zhang Y, Kleinstiver BP, Guo JA, Aryee MJ, Miller J, Malzahn A, Zarecor S, Lawrence‐Dill CJ, Joung JK, Qi Y, Wang K. Activities and specificities of CRISPR/Cas9 and Cas12a nucleases for targeted mutagenesis in maize. PLANT BIOTECHNOLOGY JOURNAL 2019; 17:362-372. [PMID: 29972722 PMCID: PMC6320322 DOI: 10.1111/pbi.12982] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/14/2018] [Accepted: 06/27/2018] [Indexed: 05/11/2023]
Abstract
CRISPR/Cas9 and Cas12a (Cpf1) nucleases are two of the most powerful genome editing tools in plants. In this work, we compared their activities by targeting maize glossy2 gene coding region that has overlapping sequences recognized by both nucleases. We introduced constructs carrying SpCas9-guide RNA (gRNA) and LbCas12a-CRISPR RNA (crRNA) into maize inbred B104 embryos using Agrobacterium-mediated transformation. On-target mutation analysis showed that 90%-100% of the Cas9-edited T0 plants carried indel mutations and 63%-77% of them were homozygous or biallelic mutants. In contrast, 0%-60% of Cas12a-edited T0 plants had on-target mutations. We then conducted CIRCLE-seq analysis to identify genome-wide potential off-target sites for Cas9. A total of 18 and 67 potential off-targets were identified for the two gRNAs, respectively, with an average of five mismatches compared to the target sites. Sequencing analysis of a selected subset of the off-target sites revealed no detectable level of mutations in the T1 plants, which constitutively express Cas9 nuclease and gRNAs. In conclusion, our results suggest that the CRISPR/Cas9 system used in this study is highly efficient and specific for genome editing in maize, while CRISPR/Cas12a needs further optimization for improved editing efficiency.
Collapse
Affiliation(s)
- Keunsub Lee
- Crop Bioengineering CenterIowa State UniversityAmesIAUSA
- Department of AgronomyIowa State UniversityAmesIAUSA
| | - Yingxiao Zhang
- Department of Plant Science and Landscape ArchitectureUniversity of MarylandCollege ParkMDUSA
| | - Benjamin P. Kleinstiver
- Molecular Pathology UnitCenter for Cancer Research, and Center for Computational and Integrative BiologyMassachusetts General HospitalCharlestownMAUSA
- Department of PathologyHarvard Medical SchoolBostonMAUSA
| | - Jimmy A. Guo
- Molecular Pathology UnitCenter for Cancer Research, and Center for Computational and Integrative BiologyMassachusetts General HospitalCharlestownMAUSA
| | - Martin J. Aryee
- Department of PathologyHarvard Medical SchoolBostonMAUSA
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - Jonah Miller
- Crop Bioengineering CenterIowa State UniversityAmesIAUSA
| | - Aimee Malzahn
- Department of Plant Science and Landscape ArchitectureUniversity of MarylandCollege ParkMDUSA
| | - Scott Zarecor
- Crop Bioengineering CenterIowa State UniversityAmesIAUSA
- Department of Genetics, Development and Cell BiologyIowa State UniversityAmesIAUSA
| | - Carolyn J. Lawrence‐Dill
- Crop Bioengineering CenterIowa State UniversityAmesIAUSA
- Department of AgronomyIowa State UniversityAmesIAUSA
- Department of Genetics, Development and Cell BiologyIowa State UniversityAmesIAUSA
- Bioinformatics and Computational Biology ProgramIowa State UniversityAmesIAUSA
| | - J. Keith Joung
- Molecular Pathology UnitCenter for Cancer Research, and Center for Computational and Integrative BiologyMassachusetts General HospitalCharlestownMAUSA
- Department of PathologyHarvard Medical SchoolBostonMAUSA
| | - Yiping Qi
- Department of Plant Science and Landscape ArchitectureUniversity of MarylandCollege ParkMDUSA
- Institute for Bioscience and Biotechnology ResearchUniversity of MarylandRockvilleMDUSA
| | - Kan Wang
- Crop Bioengineering CenterIowa State UniversityAmesIAUSA
- Department of AgronomyIowa State UniversityAmesIAUSA
| |
Collapse
|
403
|
Novel quinazolin-4-one derivatives as potentiating agents of doxorubicin cytotoxicity. Bioorg Chem 2019; 82:204-210. [DOI: 10.1016/j.bioorg.2018.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
|
404
|
A germline HLTF mutation in familial MDS induces DNA damage accumulation through impaired PCNA polyubiquitination. Leukemia 2019; 33:1773-1782. [PMID: 30696947 DOI: 10.1038/s41375-019-0385-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/27/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023]
Abstract
Although several causal genes of familial myelodysplastic syndromes (MDS) have been identified, the genetic landscape and the molecular pathogenesis are not totally understood. To explore novel driver genes and their pathogenetic significance, we performed whole-exome sequence analysis of four individuals from a familial MDS pedigree and 10 candidate single-nucleotide variants (C9orf43, CYP7B1, EFHB, ENTPD7, FAM160B2, HELZ2, HLTF, INPP5J, ITPKB, and RYK) were identified. Knockdown screening revealed that Hltf downregulation enhanced colony-forming capacity of primary murine bone marrow (BM) stem/progenitor cells. γH2AX immunofluorescent staining assay revealed increased DNA damage in a human acute myeloid leukemia (AML) cell line ectopically expressing HLTF E259K, which was not observed in cells expressing wild-type HLTF. Silencing of HLTF in human AML cells also led to DNA damage, indicating that HLTF E259K is a loss-of-function mutation. Molecularly, we found that an E259K mutation reduced the binding capacity of HLTF with ubiquitin-conjugating enzymes, methanesulfonate sensitive 2 and ubiquitin-conjugating enzyme E2N, resulting in impaired polyubiquitination of proliferating cell nuclear antigen (PCNA) in HLTF E259K-transduced cells. In summary, our results indicate that a familial MDS-associated HLTF E259K germline mutation induces accumulation of DNA double-strand breaks, possibly through impaired PCNA polyubiquitination.
Collapse
|
405
|
Rager JE, Suh M, Chappell GA, Thompson CM, Proctor DM. Review of transcriptomic responses to hexavalent chromium exposure in lung cells supports a role of epigenetic mediators in carcinogenesis. Toxicol Lett 2019; 305:40-50. [PMID: 30690063 DOI: 10.1016/j.toxlet.2019.01.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 01/17/2019] [Accepted: 01/25/2019] [Indexed: 12/14/2022]
Abstract
Inhalation exposure to hexavalent chromium [Cr(VI)] is associated with increased risk of lung cancer with a mode of action (MOA) postulated to involve non-mutagenic key events, yet molecular-level events remain uncertain. Previously-published transcriptomic studies in the lung and lung cells were reviewed to evaluate molecular events in the MOA. This study aimed to (i) identify biological pathways that are consistently modulated by Cr(VI) in the lung through the compilation of transcriptomic-based databases, (ii) predict interactions between epigenetic regulators and transcriptional responses, and (iii) relate findings to previous literature to postulate a mechanism of action underlying Cr(VI)-induced lung cancer involving changes in genomic/epigenomic signatures. This cross-study comparison identified 372 genes with Cr(VI)-induced expression alterations in multiple studies. Pathway enrichment analyses of the commonly modulated genes demonstrated that pathways involved in cytotoxicity / cell proliferation were highly enriched, as well as the general suppression of genes involved in DNA damage repair. These signaling alterations were predicted to be regulated by DNA methylation, histone modifications, and microRNAs; and published evidence substantiates the role of these epigenetic regulators in Cr(VI)-induced carcinogenicity. Findings support the influence of epigenetic alterations on cell signaling related to Cr(VI)-induced cytotoxicity/cell proliferation, and decreases in DNA repair signaling leading to tumorigenesis.
Collapse
Affiliation(s)
- Julia E Rager
- ToxStrategies, Inc., Austin, TX, 78759, United States
| | - Mina Suh
- ToxStrategies, Inc., Mission Viejo, CA, 92692, United States
| | | | | | | |
Collapse
|
406
|
Cai Y, Zhou L, Gao Y, Liu W, Shao Y, Zheng Y. Contribution of Base Damages to the Molecular Radiosensitization Mechanism of Platinum Chemotherapeutic Drugs. ChemistrySelect 2019. [DOI: 10.1002/slct.201803400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Yanming Cai
- Research Institute of Photocatalysis, S; tate Key Laboratory of Photocatalysis on Energy and Environment; Fuzhou University; Fuzhou 350116 P.R. China
| | - Limei Zhou
- Research Institute of Photocatalysis, S; tate Key Laboratory of Photocatalysis on Energy and Environment; Fuzhou University; Fuzhou 350116 P.R. China
| | - Yingxia Gao
- Research Institute of Photocatalysis, S; tate Key Laboratory of Photocatalysis on Energy and Environment; Fuzhou University; Fuzhou 350116 P.R. China
| | - Wenhui Liu
- Research Institute of Photocatalysis, S; tate Key Laboratory of Photocatalysis on Energy and Environment; Fuzhou University; Fuzhou 350116 P.R. China
| | - Yu Shao
- Research Institute of Photocatalysis, S; tate Key Laboratory of Photocatalysis on Energy and Environment; Fuzhou University; Fuzhou 350116 P.R. China
| | - Yi Zheng
- Research Institute of Photocatalysis, S; tate Key Laboratory of Photocatalysis on Energy and Environment; Fuzhou University; Fuzhou 350116 P.R. China
| |
Collapse
|
407
|
Alblihy A, Mesquita KA, Sadiq MT, Madhusudan S. Development and implementation of precision therapies targeting base-excision DNA repair in BRCA1-associated tumors. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1567266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Adel Alblihy
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Katia A. Mesquita
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Maaz T. Sadiq
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | - Srinivasan Madhusudan
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| |
Collapse
|
408
|
Ebrahimi S, Hashemy SI. MicroRNA-mediated redox regulation modulates therapy resistance in cancer cells: clinical perspectives. Cell Oncol (Dordr) 2019; 42:131-141. [PMID: 30645730 DOI: 10.1007/s13402-018-00421-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Chemotherapy and radiation therapy are the most common types of cancer therapy. The development of chemo/radio-resistance remains, however, a major obstacle. Altered redox balances are among of the main factors mediating therapy resistance. Therefore, redox regulatory strategies are urgently needed to overcome this problem. Recently, microRNAs have been found to act as major redox regulatory factors affecting chemo/radio-resistance. MicroRNAs play critical roles in regulating therapeutic resistance through the regulation of antioxidant enzymes, redox-sensitive signaling pathways, cancer stem cells, DNA repair mechanisms and autophagy. CONCLUSIONS Here, we summarize current knowledge on microRNA-mediated redox regulatory mechanisms underlying chemo/radio-resistance. This knowledge may form a basis for a better clinical management of cancer patients.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
409
|
Ma J, Kumar A, Muroya Y, Yamashita S, Sakurai T, Denisov SA, Sevilla MD, Adhikary A, Seki S, Mostafavi M. Observation of dissociative quasi-free electron attachment to nucleoside via excited anion radical in solution. Nat Commun 2019; 10:102. [PMID: 30626877 PMCID: PMC6327028 DOI: 10.1038/s41467-018-08005-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 12/11/2018] [Indexed: 01/28/2023] Open
Abstract
Damage to DNA via dissociative electron attachment has been well-studied in both the gas and condensed phases; however, understanding this process in bulk solution at a fundamental level is still a challenge. Here, we use a picosecond pulse of a high energy electron beam to generate electrons in liquid diethylene glycol and observe the electron attachment dynamics to ribothymidine at different stages of electron relaxation. Our transient spectroscopic results reveal that the quasi-free electron with energy near the conduction band effectively attaches to ribothymidine leading to a new absorbing species that is characterized in the UV-visible region. This species exhibits a nearly concentration-independent decay with a time constant of ~350 ps. From time-resolved studies under different conditions, combined with data analysis and theoretical calculations, we assign this intermediate to an excited anion radical that undergoes N1-C1′ glycosidic bond dissociation rather than relaxation to its ground state. Radiation-induced low-energy electrons in solution are implicated in DNA damage, but their relaxation dynamics are not well understood. Here the authors observe how quasi-free electrons dissociate glycosidic bonds via an excited nucleoside anion radical, whereas solvated electrons reside on the nucleoside as a relatively stable anion radical.
Collapse
Affiliation(s)
- Jun Ma
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan.
| | - Anil Kumar
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, MI, 48309, USA
| | - Yusa Muroya
- Department of Beam Materials Science, Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Shinichi Yamashita
- Nuclear Professional School, School of Engineering, The University of Tokyo, 2-22 Shirakata Shirane, Tokai-mura, Naka-gun, Ibaraki, 319-1188, Japan
| | - Tsuneaki Sakurai
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Sergey A Denisov
- Laboratoire de Chimie Physique, UMR 8000 CNRS/Université Paris-Sud, Bât. 349, 91405, Orsay, Cedex, France
| | - Michael D Sevilla
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, MI, 48309, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, MI, 48309, USA
| | - Shu Seki
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan.
| | - Mehran Mostafavi
- Laboratoire de Chimie Physique, UMR 8000 CNRS/Université Paris-Sud, Bât. 349, 91405, Orsay, Cedex, France.
| |
Collapse
|
410
|
Zhou Z, Lu H, Zhu S, Gomaa A, Chen Z, Yan J, Washington K, El-Rifai W, Dang C, Peng D. Activation of EGFR-DNA-PKcs pathway by IGFBP2 protects esophageal adenocarcinoma cells from acidic bile salts-induced DNA damage. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:13. [PMID: 30626422 PMCID: PMC6327430 DOI: 10.1186/s13046-018-1021-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/26/2018] [Indexed: 12/26/2022]
Abstract
Background The incidence of esophageal adenocarcinoma (EAC) is rising rapidly in the US and Western countries. The development of Barrett’s esophagus (BE) and its progression to EAC have been linked to chronic gastroesophageal reflux disease (GERD). Exposure of BE and EAC cells to acidic bile salts (ABS) in GERD conditions induces high levels of oxidative stress and DNA damage. In this study, we investigated the role of insulin-like growth factor binding protein 2 (IGFBP2) in regulating ABS-induced DNA double-strand breaks. Methods Real-time RT-PCR, western blot, immunohistochemistry, immunofluorescence, co-immunoprecipitation, flow cytometry, and cycloheximide (CHX) chase assays were used in this study. To mimic GERD conditions, a cocktail of acidic bile salts (pH 4) was used in 2D and 3D organotypic culture models. Overexpression and knockdown of IGFBP2 in EAC cells were established to examine the functional and mechanistic roles of IGFBP2 in ABS-induced DNA damage. Results Our results demonstrated high levels of IGFBP2 mRNA and protein in EAC cell lines as compared to precancerous Barrett’s cell lines, and IGFBP2 is frequently overexpressed in EACs (31/57). Treatment of EAC cells with ABS, to mimic GERD conditions, induced high levels of IGFBP2 expression. Knocking down endogenous IGFBP2 in FLO1 cells (with constitutive high levels of IGFBP2) led to a significant increase in DNA double-strand breaks and apoptosis, following transient exposure to ABS. On the other hand, overexpression of exogenous IGFBP2 in OE33 cells (with low endogenous levels of IGFBP2) had a protective effect against ABS-induced double-strand breaks and apoptosis. We found that IGFBP2 is required for ABS-induced nuclear accumulation and phosphorylation of EGFR and DNA-PKcs, which are necessary for DNA damage repair activity. Using co-immunoprecipitation assay, we detected co-localization of IGFBP2 with EGFR and DNA-PKcs, following acidic bile salts treatment. We further demonstrated, using cycloheximide chase assay, that IGFBP2 promotes EGFR protein stability in response to ABS exposure. Conclusions IGFBP2 protects EAC cells against ABS-induced DNA damage and apoptosis through stabilization and activation of EGFR - DNA-PKcs signaling axis. Electronic supplementary material The online version of this article (10.1186/s13046-018-1021-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhangjian Zhou
- Department of Surgical Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta W. Road, Xi'an, 710061, Shaanxi, China.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA
| | - Heng Lu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA
| | - Shoumin Zhu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA
| | - Ahmed Gomaa
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA
| | - Zheng Chen
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA
| | - Jin Yan
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA.,Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wael El-Rifai
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA.,Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Chengxue Dang
- Department of Surgical Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta W. Road, Xi'an, 710061, Shaanxi, China.
| | - Dunfa Peng
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA. .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136-1015, USA.
| |
Collapse
|
411
|
Toulany M. Targeting DNA Double-Strand Break Repair Pathways to Improve Radiotherapy Response. Genes (Basel) 2019; 10:genes10010025. [PMID: 30621219 PMCID: PMC6356315 DOI: 10.3390/genes10010025] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/07/2018] [Accepted: 12/27/2018] [Indexed: 12/13/2022] Open
Abstract
More than half of cancer patients receive radiotherapy as a part of their cancer treatment. DNA double-strand breaks (DSBs) are considered as the most lethal form of DNA damage and a primary cause of cell death and are induced by ionizing radiation (IR) during radiotherapy. Many malignant cells carry multiple genetic and epigenetic aberrations that may interfere with essential DSB repair pathways. Additionally, exposure to IR induces the activation of a multicomponent signal transduction network known as DNA damage response (DDR). DDR initiates cell cycle checkpoints and induces DSB repair in the nucleus by non-homologous end joining (NHEJ) or homologous recombination (HR). The canonical DSB repair pathways function in both normal and tumor cells. Thus, normal-tissue toxicity may limit the targeting of the components of these two pathways as a therapeutic approach in combination with radiotherapy. The DSB repair pathways are also stimulated through cytoplasmic signaling pathways. These signaling cascades are often upregulated in tumor cells harboring mutations or the overexpression of certain cellular oncogenes, e.g., receptor tyrosine kinases, PIK3CA and RAS. Targeting such cytoplasmic signaling pathways seems to be a more specific approach to blocking DSB repair in tumor cells. In this review, a brief overview of cytoplasmic signaling pathways that have been reported to stimulate DSB repair is provided. The state of the art of targeting these pathways will be discussed. A greater understanding of the underlying signaling pathways involved in DSB repair may provide valuable insights that will help to design new strategies to improve treatment outcomes in combination with radiotherapy.
Collapse
Affiliation(s)
- Mahmoud Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Roentgenweg 11, 72076 Tuebingen, Germany.
| |
Collapse
|
412
|
Smith CJ, Perfetti TA, King JA. Rodent 2-year cancer bioassays and in vitro and in vivo genotoxicity tests insufficiently predict risk or model development of human carcinomas. TOXICOLOGY RESEARCH AND APPLICATION 2019. [DOI: 10.1177/2397847319849648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Carr J Smith
- Albemarle Corporation, Mobile, AL, USA
- Department of Nurse Anesthesia, Florida State University, Panama City, FL, USA
| | | | - Judy A King
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, USA
| |
Collapse
|
413
|
Zhu LS, Wang DQ, Cui K, Liu D, Zhu LQ. Emerging Perspectives on DNA Double-strand Breaks in Neurodegenerative Diseases. Curr Neuropharmacol 2019; 17:1146-1157. [PMID: 31362659 PMCID: PMC7057204 DOI: 10.2174/1570159x17666190726115623] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/03/2019] [Accepted: 07/01/2019] [Indexed: 11/22/2022] Open
Abstract
DNA double-strand breaks (DSBs) are common events that were recognized as one of the most toxic lesions in eukaryotic cells. DSBs are widely involved in many physiological processes such as V(D)J recombination, meiotic recombination, DNA replication and transcription. Deregulation of DSBs has been reported in multiple diseases in human beings, such as the neurodegenerative diseases, with which the underlying mechanisms are needed to be illustrated. Here, we reviewed the recent insights into the dysfunction of DSB formation and repair, contributing to the pathogenesis of neurodegenerative disorders including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) and ataxia telangiectasia (A-T).
Collapse
Affiliation(s)
| | | | | | | | - Ling-Qiang Zhu
- Address correspondence to this author at the Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China; Tel: 862783692625; Fax: 862783692608; E-mail:
| |
Collapse
|
414
|
Wan R, Mo Y, Tong R, Gao M, Zhang Q. Determination of Phosphorylated Histone H2AX in Nanoparticle-Induced Genotoxic Studies. Methods Mol Biol 2019; 1894:145-159. [PMID: 30547460 DOI: 10.1007/978-1-4939-8916-4_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
DNA double-strand breaks (DSBs), one of the most severe lesions of DNA damage triggered by various genotoxic insults, can lead to chromosome change, genomic instability, and even tumorigenesis if not repaired efficiently. In response to DNA damage, histone H2AX molecules are rapidly phosphorylated at serine 139 near the site of DNA DSBs and form γ-H2AX foci. As an early important cellular event linked to DNA damage and repair, γ-H2AX is a highly sensitive biomarker for "monitoring" DNA damage and consequently is a useful tool in genetic toxicology screen. We and other researchers have used γ-H2AX as a marker to assess the potential genotoxic effects of some nanoparticles in vitro and in vivo. In this chapter, we describe several useful methods for γ-H2AX detection, which can be used to evaluate the potential genotoxic effects of nanoparticles.
Collapse
Affiliation(s)
- Rong Wan
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Ruirui Tong
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Meiqin Gao
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
415
|
Jimeno S, Mejías-Navarro F, Prados-Carvajal R, Huertas P. Controlling the balance between chromosome break repair pathways. DNA Repair (Amst) 2019; 115:95-134. [DOI: 10.1016/bs.apcsb.2018.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
416
|
Decitabine exerted synergistic effects with oxaliplatin in colorectal cancer cells with intrinsic resistance to decitabine. Biochem Biophys Res Commun 2019; 509:249-254. [DOI: 10.1016/j.bbrc.2018.12.115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 12/15/2018] [Indexed: 11/18/2022]
|
417
|
Liu S, Shao Y, Wang Q, Zhai Y, Li X. Genotoxic stress causes the accumulation of DNA-dependent protein kinase catalytic subunit phosphorylated at serine 2056 at nuclear speckles and alters pre-mRNA alternative splicing. FEBS Open Bio 2018; 9:304-314. [PMID: 30761255 PMCID: PMC6356181 DOI: 10.1002/2211-5463.12569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 11/25/2018] [Accepted: 12/05/2018] [Indexed: 01/28/2023] Open
Abstract
RNA splicing has emerged as a critical player in the DNA damage response (DDR). However, the underlying mechanism(s) by which pre‐mRNA splicing is coordinately regulated by genotoxic stress has remained largely unclear. Here, we show that a DDR factor, DNA‐dependent protein kinase (DNA‐PK), participates in the modulation of pre‐mRNA splicing in the presence of DNA double‐strand break (DSB)‐induced genotoxic stress. Through indirect immunostaining, we made the surprising discovery that DNA‐PK catalytic subunits (DNA‐PKcs) autophosphorylated at serine 2056 (S2056) accumulate at nuclear speckles (dynamic nuclear structures that are enriched with splicing factors), following their dissociation from DSB lesions. Inactivation of DNA‐PKcs, either using a small molecule inhibitor or by RNA interference, alters alternative splicing of a set of pre‐mRNAs in A549 cells treated with the topoisomerase II inhibitor mitoxantrone, indicative of an involvement of DNA‐PKcs in modulating pre‐mRNA splicing following genotoxic stress. These findings indicate a novel physical and functional connection between the DNA damage response and pre‐mRNA splicing, and enhance our understanding of how mRNA splicing is involved in the cellular response to DSB lesions.
Collapse
Affiliation(s)
- Shuang Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China
| | - Yuan Shao
- Beijing Key Laboratory of DNA Damage Response College of Life Sciences Capital Normal University China
| | - Qi Wang
- Beijing Key Laboratory of DNA Damage Response College of Life Sciences Capital Normal University China
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China
| | - Xialu Li
- Beijing Key Laboratory of DNA Damage Response College of Life Sciences Capital Normal University China
| |
Collapse
|
418
|
Liu Y, Zhang J, Feng S, Zhao T, Li Z, Wang L, Wang P, Du H, Yuan S, Sun L. A Novel Camptothecin Derivative 3j Inhibits Nsclc Proliferation Via Induction of Cell Cycle Arrest By Topo I-Mediated DNA Damage. Anticancer Agents Med Chem 2018; 19:365-374. [PMID: 30523769 DOI: 10.2174/1871520619666181207102037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim of this study is to investigate the inhibitory effect of camptothecin derivative 3j on Non-Small Cell Lung Cancer (NSCLCs) cells and the potential anti-tumor mechanisms. BACKGROUND Camptothecin compounds are considered as the third largest natural drugs which are widely investigated in the world and they suffered restriction because of serious toxicity, such as hemorrhagic cystitis and bone marrow suppression. METHODS Using cell proliferation assay and S180 tumor mice model, a series of 20(S)-O-substituted benzoyl 7- ethylcamptothecin compounds were screened and evaluated the antitumor activities in vitro and in vivo. Camptothecin derivative 3j was selected for further study using flow cytometry in NSCLCs cells. Cell cycle related protein cyclin A2, CDK2, cyclin D and cyclin E were detected by Western Blot. Then, computer molecular docking was used to confirm the interaction between 3j and Topo I. Also, DNA relaxation assay and alkaline comet assay were used to investigate the mechanism of 3j on DNA damage. RESULTS Our results demonstrated that camptothecin derivative 3j showed a greater antitumor effect in eleven 20(S)-O-substituted benzoyl 7-ethylcamptothecin compounds in vitro and in vivo. The IC50 of 3j was 1.54± 0.41 µM lower than irinotecan with an IC50 of 13.86±0.80 µM in NCI-H460 cell, which was reduced by 8 fold. In NCI-H1975 cell, the IC50 of 3j was 1.87±0.23 µM lower than irinotecan (IC50±SD, 5.35±0.38 µM), dropped by 1.8 fold. Flow cytometry analysis revealed that 3j induced significant accumulation in a dose-dependent manner. After 24h of 3j (10 µM) treatment, the percentage of NCI-H460 cell in S-phase significantly increased (to 93.54 ± 4.4%) compared with control cells (31.67 ± 3.4%). Similarly, the percentage of NCI-H1975 cell in Sphase significantly increased (to 83.99 ± 2.4%) compared with control cells (34.45 ± 3.9%) after treatment with 10µM of 3j. Moreover, increased levels of cyclin A2, CDK2, and decreased levels of cyclin D, cyclin E further confirmed that cell cycle arrest was induced by 3j. Furthermore, molecular docking studies suggested that 3j interacted with Topo I-DNA and DNA-relaxation assay simultaneously confirmed that 3j suppressed the activity of Topo I. Research on the mechanism showed that 3j exhibited anti-tumour activity via activating the DNA damage response pathway and suppressing the repair pathway in NSCLC cells. CONCLUSION Novel camptothecin derivative 3j has been demonstrated as a promising antitumor agent and remains to be assessed in further studies.
Collapse
Affiliation(s)
- Yang Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jingyin Zhang
- Department of Gynaecology and Obstetrics, Southeast University Affiliated Zhongda Hospital, Nanjing, Jiangsu, China
| | - Shuyun Feng
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tingli Zhao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhengzheng Li
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lai Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Puhai Wang
- Jiangsu Provincial Institue of Materia Medica, Nanjing Technology University, Nanjing, Jiangsu, China
| | - Hongzhi Du
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shengtao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
419
|
Targeting the DNA-PK complex: Its rationale use in cancer and HIV-1 infection. Biochem Pharmacol 2018; 160:80-91. [PMID: 30529192 DOI: 10.1016/j.bcp.2018.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023]
Abstract
The DNA-PK complex is the major component of the predominant mechanism of DSB repair in humans. In addition, this complex is involved in many other processes such as DNA recombination, genome maintenance, apoptosis and transcription regulation. Several studies have linked the decrease of the DNA-PK activity with cancer initiation, due to defects in the repair. On another hand, higher DNA-PK expression and activity have been observed in various other tumor cells and have been linked with a decrease of the efficiency of anti-tumor drugs. It has also been shown that DNA-PK is critical for the integration of the HIV-1 DNA into the cell host genome and promotes replication and transcription of the virus. Targeting this complex makes therefore sense to treat these two pathologies. However, according to the status of HIV-1 replication (active versus latent replication) or to the tumor grade cells (initiation versus metastasis), the way to target this DNA-PK complex might be rather different. In this review, we discuss the importance of DNA-PK complex in two major pathologies i.e. HIV-1 infection and cancer, and the rationale use of therapies aiming to target this complex.
Collapse
|
420
|
Faraoni I, Graziani G. Role of BRCA Mutations in Cancer Treatment with Poly(ADP-ribose) Polymerase (PARP) Inhibitors. Cancers (Basel) 2018; 10:E487. [PMID: 30518089 PMCID: PMC6316750 DOI: 10.3390/cancers10120487] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/29/2022] Open
Abstract
Inhibition of poly(ADP-ribose) polymerase (PARP) activity induces synthetic lethality in mutated BRCA1/2 cancers by selectively targeting tumor cells that fail to repair DNA double strand breaks (DSBs). Clinical studies have confirmed the validity of the synthetic lethality approach and four different PARP inhibitors (PARPi; olaparib, rucaparib, niraparib and talazoparib) have been approved as monotherapies for BRCA-mutated or platinum-sensitive recurrent ovarian cancer and/or for BRCA-mutated HER2-negative metastatic breast cancer. PARPi therapeutic efficacy is higher against tumors harboring deleterious germline or somatic BRCA mutations than in BRCA wild-type tumors. BRCA mutations or intrinsic tumor sensitivity to platinum compounds are both regarded as indicators of deficiency in DSB repair by homologous recombination as well as of favorable response to PARPi. However, not all BRCA-mutated or platinum-responsive patients obtain clinical benefit from these agents. Conversely, a certain percentage of patients with wild-type BRCA or platinum-resistant tumors can still get benefit from PARPi. Thus, additional reliable markers need to be validated in clinical trials to select patients potentially eligible for PARPi-based therapies, in the absence of deleterious BRCA mutations or platinum sensitivity. In this review, we summarize the mechanisms of action of PARPi and the clinical evidence supporting their use as anticancer drugs as well as the additional synthetic lethal partners that might confer sensitivity to PARPi in patients with wild-type BRCA tumors.
Collapse
Affiliation(s)
- Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, 00173 Rom, Italy.
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, 00173 Rom, Italy.
| |
Collapse
|
421
|
Otsuka K, Tomita M. Concurrent live imaging of DNA double-strand break repair and cell-cycle progression by CRISPR/Cas9-mediated knock-in of a tricistronic vector. Sci Rep 2018; 8:17309. [PMID: 30470841 PMCID: PMC6251881 DOI: 10.1038/s41598-018-35642-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 11/08/2018] [Indexed: 12/31/2022] Open
Abstract
Cell-cycle progression can be arrested by ionizing radiation-induced DNA double-strand breaks (DSBs). Although DSBs are patched by DSB repair systems, which comprise proteins such as p53-binding protein 1 (53BP1), the relationship between DSB repair progression and cell-cycle status in living cells is unclear. The probe FUCCI (fluorescent ubiquitination-based cell-cycle indicator) was previously developed for visualizing cell-cycle status. Here, we established novel live-imaging probes based on custom-designed plasmids designated “Focicles” harboring a tricistronic compartment encoding distinct fluorescent proteins ligated to the murine 53BP1 foci-forming region (FFR) and two cell-cycle indicators that are known components of FUCCI (hCdt1 and hGmnn). We used CRISPR/Cas9-mediated genome editing to obtain Focicle knock-in cell lines in NIH3T3 cells, which were subject to X-ray irradiation that induced comparable numbers of Focicle and endogenous-53BP1 foci. In addition, the Focicle probes enabled the kinetic analysis of both DSB repair and cell-cycle arrest/progression after irradiation, demonstrating that the Focicle knock-in cells progressed to cell division after DNA damage elimination. These newly developed probes can help to gain a better understanding of the dynamics of DSB repair and cell-cycle control to in turn guide cancer treatment development and cancer-risk assessments.
Collapse
Affiliation(s)
- Kensuke Otsuka
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, 201-8511, Japan.
| | - Masanori Tomita
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, 201-8511, Japan
| |
Collapse
|
422
|
de Camargo AC, Schwember AR, Parada R, Garcia S, Maróstica MR, Franchin M, Regitano-d'Arce MAB, Shahidi F. Opinion on the Hurdles and Potential Health Benefits in Value-Added Use of Plant Food Processing By-Products as Sources of Phenolic Compounds. Int J Mol Sci 2018; 19:E3498. [PMID: 30404239 PMCID: PMC6275048 DOI: 10.3390/ijms19113498] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/23/2022] Open
Abstract
Plant foods, their products and processing by-products are well recognized as important sources of phenolic compounds. Recent studies in this field have demonstrated that food processing by-products are often richer sources of bioactive compounds as compared with their original feedstock. However, their final application as a source of nutraceuticals and bioactives requires addressing certain hurdles and challenges. This review discusses recent knowledge advances in the use of plant food processing by-products as sources of phenolic compounds with special attention to the role of genetics on the distribution and biosynthesis of plant phenolics, as well as their profiling and screening, potential health benefits, and safety issues. The potentialities in health improvement from food phenolics in animal models and in humans is well substantiated, however, considering the emerging market of plant food by-products as potential sources of phenolic bioactives, more research in humans is deemed necessary.
Collapse
Affiliation(s)
- Adriano Costa de Camargo
- Departamento de Ciencias Vegetales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Casilla 306-22, Santiago, Chile.
- Department of Food Science and Technology, Londrina State University, Londrina 86051-990, Parana State, Brazil.
- Department of Agri-Food Industry, Food & Nutrition, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba 13418-900, São Paulo State, Brazil.
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| | - Andrés R Schwember
- Departamento de Ciencias Vegetales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Casilla 306-22, Santiago, Chile.
| | - Roberto Parada
- Departamento de Ciencias Vegetales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Casilla 306-22, Santiago, Chile.
| | - Sandra Garcia
- Department of Food Science and Technology, Londrina State University, Londrina 86051-990, Parana State, Brazil.
| | - Mário Roberto Maróstica
- Department of Food and Nutrition, University of Campinas-UNICAMP, Campinas 13083-862, São Paulo State, Brazil.
| | - Marcelo Franchin
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba 13414-903, São Paulo State, Brazil.
| | - Marisa Aparecida Bismara Regitano-d'Arce
- Department of Agri-Food Industry, Food & Nutrition, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba 13418-900, São Paulo State, Brazil.
| | - Fereidoon Shahidi
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
423
|
Gopalakrishnan V, Dahal S, Radha G, Sharma S, Raghavan SC, Choudhary B. Characterization of DNA double-strand break repair pathways in diffuse large B cell lymphoma. Mol Carcinog 2018; 58:219-233. [DOI: 10.1002/mc.22921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/18/2018] [Accepted: 10/07/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Vidya Gopalakrishnan
- Institute of Bioinformatics and Applied Biotechnology; Electronics City; Bangalore India
- Manipal Academy of Higher Education; Manipal Karnataka India
| | - Sumedha Dahal
- Department of Biochemistry; Indian Institute of Science; Bangalore India
| | - Gudapureddy Radha
- Department of Biochemistry; Indian Institute of Science; Bangalore India
| | - Shivangi Sharma
- Department of Biochemistry; Indian Institute of Science; Bangalore India
| | | | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology; Electronics City; Bangalore India
| |
Collapse
|
424
|
Sun Y, Wang H, Zhang K, Liu J, Wang P, Wang X, Liu Q. Sonodynamic therapy induces oxidative stress, DNA damage and apoptosis in glioma cells. RSC Adv 2018; 8:36245-36256. [PMID: 35558463 PMCID: PMC9088833 DOI: 10.1039/c8ra07099g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 06/01/2021] [Accepted: 10/15/2018] [Indexed: 02/05/2023] Open
Abstract
Malignant glioma remains one of the most challenging diseases to treat because of the invasive growth of glioma cells and the existence of the blood-brain barrier (BBB), which blocks drug delivery to the brain. New strategies are urgently needed to overcome these shortcomings and improve the outcomes. Ultrasound represents a promising noninvasive and reversible BBB opening approach and the related sonodynamic therapy (SDT) is rapidly emerging. This study aims to explore the ultrasound parameters for BBB opening and the cell killing effect of SDT in human glioma U373 cells by using a recently reported sonosensitizer, sinoporphyrin sodium (DVDMS). The in vitro BBB model indicated that SDT caused a time-dependent permeability increase, which peaked at 2 h post treatment and then recovered gradually. The results of toxicology tests showed significant U373 cell viability loss and apoptosis increase after DVDMS-SDT, accompanied by enhanced cleaved-caspase-3 level and DNA fragmentation, in which reactive oxygen species (ROS) were a major triggering intermediate during DVDMS-SDT. Furthermore, DVDMS-SDT produced DNA damage and the underlying mechanisms were evaluated, in order to provide a fundamental basis for DVDMS-SDT application in glioma therapy. The findings indicated that the DNA molecules could be temporarily regulated by SDT and DNA double-strand breaks (DSBs), which increased the difficulty of cellular self-repair, thus aggravating cell apoptosis and inhibiting glioma cell invasive growth. Therefore, this study supports the use of SDT as an alternative approach for glioma therapy.
Collapse
Affiliation(s)
- Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| | - Haiping Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| | - Kun Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| | - Jingfei Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| | - Quanhong Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University Xi'an Shaanxi 710119 People's Republic of China +86-029-85310275
| |
Collapse
|
425
|
Çağlayan M, Wilson SH. Pol μ dGTP mismatch insertion opposite T coupled with ligation reveals promutagenic DNA repair intermediate. Nat Commun 2018; 9:4213. [PMID: 30310068 PMCID: PMC6181931 DOI: 10.1038/s41467-018-06700-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/08/2018] [Indexed: 01/01/2023] Open
Abstract
Incorporation of mismatched nucleotides during DNA replication or repair leads to transition or transversion mutations and is considered as a predominant source of base substitution mutagenesis in cancer cells. Watson-Crick like dG:dT base pairing is considered to be an important source of genome instability. Here we show that DNA polymerase (pol) μ insertion of 7,8-dihydro-8′-oxo-dGTP (8-oxodGTP) or deoxyguanosine triphosphate (dGTP) into a model double-strand break DNA repair substrate with template base T results in efficient ligation by DNA ligase. These results indicate that pol μ-mediated dGTP mismatch insertion opposite template base T coupled with ligation could be a feature of mutation prone nonhomologous end joining during double-strand break repair. Incorporation of mismatched nucleotides during DNA replication or repair can lead to mutagenesis. Here the authors reveal that DNA ligase can ligate NHEJ intermediates following incorporation of 8-oxodGTP or dGTP opposite T by DNA Polymerase mu (Pol mu) in vitro, which suggests that Pol mu could cause promutagenic mismatches during DSB repair.
Collapse
Affiliation(s)
- Melike Çağlayan
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA. .,Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, 32610, USA.
| | - Samuel H Wilson
- Genome Integrity and Structural Biology Laboratory, National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
426
|
Liu Y, Korn JA, Dang A, Tureček F. Hydrogen-Rich Cation Radicals of DNA Dinucleotides: Generation and Structure Elucidation by UV-Vis Action Spectroscopy. J Phys Chem B 2018; 122:9665-9680. [PMID: 30269486 DOI: 10.1021/acs.jpcb.8b07925] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hydrogen-rich DNA dinucleotide cation radicals (dGG + 2H)+•, (dCG + 2H)+•, and (dGC + 2H)+• represent transient species comprising protonated and hydrogen atom adducted nucleobase rings that serve as models for proton and radical migrations in ionized DNA. These DNA cation radicals were generated in the gas phase by electron-transfer dissociation of dinucleotide dication-crown-ether complexes and characterized by UV-vis photodissociation action spectra, ab initio calculations of structures and relative energies, and time-dependent density functional theory calculations of UV-vis absorption spectra. Theoretical calculations indicate that (dGG + 2H)+• cation radicals formed by electron transfer underwent an exothermic conformational collapse that was accompanied by guanine ring stacking and facile internucleobase hydrogen atom transfer, forming 3'-guanine C-8-H radicals. In contrast, exothermic hydrogen transfer from the 5'-cytosine radical onto the guanine ring in (dCG + 2H)+• was kinetically hampered, resulting in the formation of a mixture of 5'-cytosine and 3'-guanine radicals. Conformational folding and nucleobase stacking were energetically unfavorable in (dGC + 2H)+• that retained its structure of a 3'-cytosine radical, as formed by one-electron reduction of the dication. Hydrogen-rich guanine (G + H)• and cytosine (C + H)• radicals were calculated to have vastly different basicities in water, as illustrated by the respective p Ka values of 20.0 and 4.6, which is pertinent to their different abilities to undergo proton-transfer reactions in solution.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, Bagley Hall , University of Washington , P.O. Box 351700, Seattle , Washington 98195-1700 , United States
| | - Joseph A Korn
- Department of Chemistry, Bagley Hall , University of Washington , P.O. Box 351700, Seattle , Washington 98195-1700 , United States
| | - Andy Dang
- Department of Chemistry, Bagley Hall , University of Washington , P.O. Box 351700, Seattle , Washington 98195-1700 , United States
| | - František Tureček
- Department of Chemistry, Bagley Hall , University of Washington , P.O. Box 351700, Seattle , Washington 98195-1700 , United States
| |
Collapse
|
427
|
Gasinska A, Biesaga B, Widla AJ, Darasz Z. Positive effect of single nucleotide RAD51 135G>C polymorphism and low Ku70 protein expression on female rectal cancer patients survival after preoperative radiotherapy. TURKISH JOURNAL OF GASTROENTEROLOGY 2018; 30:3-14. [PMID: 30289394 DOI: 10.5152/tjg.2018.17486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIMS This is a retrospective analysis of 103 patients having locally advanced rectal cancer who received short-course radiotherapy (SCRT). The objective of the study was to check whether a polymorphism in the RAD51 gene (135 G>C), Ku70 protein expression, and tumor microenvironment: proliferation rate measured by BrdUrdLI and Ki-67LI, hypoxia (glucose transporter-1 expression), P53 protein expression, and DNA ploidy can influence DNA repair capacity, the factors contributing to patient overall survival (OS) and the incidence of recurrences and metastases. MATERIALS AND METHODS RAD51 (135 G>C) polymorphism was evaluated using restriction fragment length polymorphism polymerase chain reaction, and proteins were identified using immunohistochemistry. RESULTS There were 3 (2.9%) tumors with RAD51 CC, 75 (72.8%) with GG, and 25 (24.3%) with GC genotypes. The median follow-up time was 63.1 months (range 2-120). Patients with CC genotype survived significantly longer than those with GG and GC genotypes and did not develop any recurrences or distant metastases. Female patients with Ku70 expression (<75.1) or RAD51CC genotype (impaired DNA damage repair and radiosensitive) had significantly longer OS (p=0.013) than those with Ku70>75.1 % or RAD51GG,GC (radioresistant phenotype) and male patients in the log-rank test. In multivariate analysis, positive prognostic factors for OS in the male patients were grade=1 and <17 days break in the treatment, whereas in the female subgroup, only radiosensitive phenotype (Ku70 <75.1% or RAD51CC genotype). CONCLUSION To the best of our knowledge, this is the first study to provide evidence for the positive effect of CC genotype of RAD51 or low Ku70 expression on OS in females with rectal cancer after SCRT.
Collapse
Affiliation(s)
- Anna Gasinska
- Department of Applied Radiobiology, Maria Sklodowska - Curie Institute, Oncology Center, Cracow Branch, Poland
| | - Beata Biesaga
- Department of Applied Radiobiology, Maria Sklodowska - Curie Institute, Oncology Center, Cracow Branch, Poland
| | - Anna Janecka Widla
- Department of Applied Radiobiology, Maria Sklodowska - Curie Institute, Oncology Center, Cracow Branch, Poland
| | - Zbigniew Darasz
- Department of Surgery, Maria Sklodowska - Curie Institute, Oncology Center, Cracow Branch, Poland
| |
Collapse
|
428
|
Panou V, Gadiraju M, Wolin A, Weipert CM, Skarda E, Husain AN, Patel JD, Rose B, Zhang SR, Weatherly M, Nelakuditi V, Knight Johnson A, Helgeson M, Fischer D, Desai A, Sulai N, Ritterhouse L, Røe OD, Turaga KK, Huo D, Segal J, Kadri S, Li Z, Kindler HL, Churpek JE. Frequency of Germline Mutations in Cancer Susceptibility Genes in Malignant Mesothelioma. J Clin Oncol 2018; 36:2863-2871. [PMID: 30113886 PMCID: PMC6804864 DOI: 10.1200/jco.2018.78.5204] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The aim of the current study was to determine the prevalence and clinical predictors of germline cancer susceptibility mutations in patients with malignant mesothelioma (MM). METHODS We performed targeted capture and next-generation sequencing of 85 cancer susceptibility genes on germline DNA from 198 patients with pleural, peritoneal, and tunica vaginalis MM. RESULTS Twenty-four germline mutations were identified in 13 genes in 23 (12%) of 198 patients. BAP1 mutations were the most common (n = 6; 25%). The remaining were in genes involved in DNA damage sensing and repair (n = 14), oxygen sensing (n = 2), endosome trafficking (n = 1), and cell growth (n = 1). Pleural site (odds ratio [OR], 0.23; 95% CI, 0.10 to 0.58; P < .01), asbestos exposure (OR, 0.28; 95% CI, 0.11 to 0.72; P < .01), and older age (OR, 0.95; 95% CI, 0.92 to 0.99; P = .01) were associated with decreased odds of carrying a germline mutation, whereas having a second cancer diagnosis (OR, 3.33; 95% CI, 1.22 to 9.07; P = .02) significantly increased the odds. The odds of carrying a mutation in BAP1 (OR, 1,658; 95% CI, 199 to 76,224; P < .001), BRCA2 (OR, 5; 95% CI, 1.0 to 14.7; P = .03), CDKN2A (OR, 53; 95% CI, 6 to 249; P < .001), TMEM127 (OR, 88; 95% CI, 1.7 to 1,105; P = .01), VHL (OR, 51; 95% CI, 1.1 to 453; P = .02), and WT1 (OR, 20; 95% CI, 0.5 to 135; P = .049) were significantly higher in MM cases than in a noncancer control population. Tumor sequencing identified mutations in a homologous recombination pathway gene in 52% (n = 29 of 54). CONCLUSION A significant proportion of patients with MM carry germline mutations in cancer susceptibility genes, especially those with peritoneal MM, minimal asbestos exposure, young age, and a second cancer diagnosis. These data support clinical germline genetic testing for patients with MM and provide a rationale for additional investigation of the homologous recombination pathway in MM.
Collapse
Affiliation(s)
- Vasiliki Panou
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Meghana Gadiraju
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arthur Wolin
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Caroline M. Weipert
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Emily Skarda
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Aliya N. Husain
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jyoti D. Patel
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Buerkley Rose
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Shannon R. Zhang
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Madison Weatherly
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Viswateja Nelakuditi
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Amy Knight Johnson
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Maria Helgeson
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - David Fischer
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arpita Desai
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nanna Sulai
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lauren Ritterhouse
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Oluf D. Røe
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kiran K. Turaga
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dezheng Huo
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jeremy Segal
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sabah Kadri
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Zejuan Li
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hedy L. Kindler
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jane E. Churpek
- Vasiliki Panou and Oluf D. Røe, Aalborg University Hospital, Aalborg, Denmark; Meghana Gadiraju, Arthur Wolin, Caroline M. Weipert, Emily Skarda, Aliya N. Husain, Jyoti D. Patel, Buerkley Rose, Shannon R. Zhang, Madison Weatherly, Viswateja Nelakuditi, Amy Knight Johnson, Maria Helgeson, David Fischer, Arpita Desai, Nanna Sulai, Lauren Ritterhouse, Kiran K. Turaga, Dezheng Huo, Jeremy Segal, Sabah Kadri, Zejuan Li, Hedy L. Kindler, and Jane E. Churpek, The University of Chicago, Chicago, IL; Oluf D. Røe, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger; and Oluf D. Røe, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
429
|
Zhang J, Dang F, Ren J, Wei W. Biochemical Aspects of PD-L1 Regulation in Cancer Immunotherapy. Trends Biochem Sci 2018; 43:1014-1032. [PMID: 30287140 DOI: 10.1016/j.tibs.2018.09.004] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
Abstract
PD-L1, frequently expressed in human cancers, engages with PD-1 on immune cells and contributes to cancer immune evasion. As such, antibodies blocking the PD-1/PD-L1 interaction reactivate cytotoxic T cells to eradicate cancer cells. However, a majority of cancer patients fail to respond to PD-1/PD-L1 blockade with unclear underlying mechanism(s). Recent studies revealed that PD-L1 expression levels on tumor cells might affect the clinical response to anti-PD-1/PD-L1 therapies. Hence, understanding molecular mechanisms for controlling PD-L1 expression will be important to improve the clinical response rate and efficacy of PD-1/PD-L1 blockade. In this review, we primarily focus on summarizing PD-L1 regulation and its potential roles in regulating antitumor immune response, with purpose to optimize anti-PD-1/PD-L1 therapies, benefiting a wider cancer patient population.
Collapse
Affiliation(s)
- Jinfang Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; These authors contributed equally to this work
| | - Fabin Dang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; These authors contributed equally to this work
| | - Junming Ren
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
430
|
Toh MR, Chiang JB, Chong ST, Chan SH, Ishak NDB, Courtney E, Lee WH, Syed Abdillah Al SMFB, Carson Allen J, Lim KH, Davila S, Tan P, Lim WK, Tan IBH, Ngeow J. Germline Pathogenic Variants in Homologous Recombination and DNA Repair Genes in an Asian Cohort of Young-Onset Colorectal Cancer. JNCI Cancer Spectr 2018; 2:pky054. [PMID: 31360874 PMCID: PMC6649855 DOI: 10.1093/jncics/pky054] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Growing evidence suggests a role for cancer susceptibility genes such as BRCA2 and PALB2 in young-onset colorectal cancers. Using a cohort of young colorectal cancer patients, we sought to identify and provide functional evidence for germline pathogenic variants of DNA repair genes not typically associated with colorectal cancer. METHODS We recruited 88 patients with young-onset colorectal cancers seen at a general oncology center. Whole-exome sequencing was performed to identify variants in DNA repair and colorectal cancer predisposition genes. Pathogenic BRCA2 and PALB2 variants were analyzed using immunoblot and immunofluorescence on patient-derived lymphoblastoid cells. RESULTS In general, our cohort displayed characteristic features of young-onset colorectal cancers. Most patients had left-sided tumors and were diagnosed at late stages. Four patients had familial adenomatous polyposis, as well as pathogenic APC variants. We identified 12 pathogenic variants evenly distributed between DNA repair and colorectal cancer predisposition genes. Six patients had pathogenic variants in colorectal cancer genes: APC (n = 4) and MUTYH monoallelic (n = 2). Another six had pathogenic variants in DNA repair genes: ATM (n = 1), BRCA2 (n = 1), PALB2 (n = 1), NTHL1 (n = 1), and WRN (n = 2). Pathogenic variants BRCA2 c.9154C>T and PALB2 c.1059delA showed deficient homologous recombination repair, evident from the impaired RAD51 nuclear localization and foci formation. CONCLUSION A substantial portion of pathogenic variants in young-onset colorectal cancer was found in DNA repair genes not previously associated with colorectal cancer. This may have implications for the management of patients. Further studies are needed to ascertain the enrichment of pathogenic DNA repair gene variants in colorectal cancers.
Collapse
Affiliation(s)
- Ming Ren Toh
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jian Bang Chiang
- Duke-NUS Medical School, Singapore 169857, Singapore
- Division of Medical Oncology, National Cancer Center, Singapore 169610, Singapore
| | - Siao Ting Chong
- Division of Medical Oncology, National Cancer Center, Singapore 169610, Singapore
| | - Sock Hoai Chan
- Division of Medical Oncology, National Cancer Center, Singapore 169610, Singapore
| | | | - Eliza Courtney
- Division of Medical Oncology, National Cancer Center, Singapore 169610, Singapore
| | - Wei Hao Lee
- Division of Medical Oncology, National Cancer Center, Singapore 169610, Singapore
| | | | | | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Sonia Davila
- Singhealth Duke-NUS Institute of Precision Medicine (PRISM), Singapore 169856, Singapore
| | - Patrick Tan
- Singhealth Duke-NUS Institute of Precision Medicine (PRISM), Singapore 169856, Singapore
- Cancer & Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Cancer Science Institute of Singapore, National University Singapore, Singapore 117599, Singapore
| | - Weng Khong Lim
- Singhealth Duke-NUS Institute of Precision Medicine (PRISM), Singapore 169856, Singapore
- Cancer & Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Iain Bee Huat Tan
- Division of Molecular and Cellular Research, National Cancer Center, Singapore 169610, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Joanne Ngeow
- Duke-NUS Medical School, Singapore 169857, Singapore
- Division of Medical Oncology, National Cancer Center, Singapore 169610, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798, Singapore
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
| |
Collapse
|
431
|
Abstract
Double-stranded DNA breaks activate a DNA damage checkpoint in G2 phase to trigger a cell cycle arrest, which can be reversed to allow for recovery. However, damaged G2 cells can also permanently exit the cell cycle, going into senescence or apoptosis, raising the question how an individual cell decides whether to recover or withdraw from the cell cycle. Here we find that the decision to withdraw from the cell cycle in G2 is critically dependent on the progression of DNA repair. We show that delayed processing of double strand breaks through HR-mediated repair results in high levels of resected DNA and enhanced ATR-dependent signalling, allowing p21 to rise to levels at which it drives cell cycle exit. These data imply that cells have the capacity to discriminate breaks that can be repaired from breaks that are difficult to repair at a time when repair is still ongoing. Cells with damaged DNA can permanently exit the cell cycle during the G2 phase or recover spontaneously entering mitosis. Here the authors reveal that the decision to exit from the cell cycle in G2 is dependent on the presence of repair intermediates associated with homologous recombination.
Collapse
|
432
|
Gustafsson NMS, Färnegårdh K, Bonagas N, Ninou AH, Groth P, Wiita E, Jönsson M, Hallberg K, Lehto J, Pennisi R, Martinsson J, Norström C, Hollers J, Schultz J, Andersson M, Markova N, Marttila P, Kim B, Norin M, Olin T, Helleday T. Targeting PFKFB3 radiosensitizes cancer cells and suppresses homologous recombination. Nat Commun 2018; 9:3872. [PMID: 30250201 PMCID: PMC6155239 DOI: 10.1038/s41467-018-06287-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
The glycolytic PFKFB3 enzyme is widely overexpressed in cancer cells and an emerging anti-cancer target. Here, we identify PFKFB3 as a critical factor in homologous recombination (HR) repair of DNA double-strand breaks. PFKFB3 rapidly relocates into ionizing radiation (IR)-induced nuclear foci in an MRN-ATM-γH2AX-MDC1-dependent manner and co-localizes with DNA damage and HR repair proteins. PFKFB3 relocalization is critical for recruitment of HR proteins, HR activity, and cell survival upon IR. We develop KAN0438757, a small molecule inhibitor that potently targets PFKFB3. Pharmacological PFKFB3 inhibition impairs recruitment of ribonucleotide reductase M2 and deoxynucleotide incorporation upon DNA repair, and reduces dNTP levels. Importantly, KAN0438757 induces radiosensitization in transformed cells while leaving non-transformed cells unaffected. In summary, we identify a key role for PFKFB3 enzymatic activity in HR repair and present KAN0438757, a selective PFKFB3 inhibitor that could potentially be used as a strategy for the treatment of cancer.
Collapse
Affiliation(s)
- Nina M S Gustafsson
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden.
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden.
| | - Katarina Färnegårdh
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
- Drug Discovery and Development Platform, Science for Life Laboratory, Department of Organic Chemistry, Stockholm University, Box 1030, S-171 21, Solna, Sweden
| | - Nadilly Bonagas
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | - Anna Huguet Ninou
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Petra Groth
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | - Elisee Wiita
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | | | - Kenth Hallberg
- SARomics Biostructures AB, Medicon Village, SE-223 81, Lund, Sweden
- Sprint Bioscience, 141 57, Huddinge, Sweden
| | - Jemina Lehto
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Rosa Pennisi
- Department of Sciences, Roma Tre University, 446 00146 Rome, Italy
| | | | | | - Jessica Hollers
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Johan Schultz
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | | | | | - Petra Marttila
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | - Baek Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Pharmacy, Kyung-Hee University, 02447, Seoul, South Korea
| | - Martin Norin
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Thomas Olin
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden.
- Sheffield Cancer Centre, Department of Oncology and Metabolism, University of Sheffield, S10 2RX, Sheffield, UK.
| |
Collapse
|
433
|
Kidane D. Molecular Mechanisms of H. pylori-Induced DNA Double-Strand Breaks. Int J Mol Sci 2018; 19:ijms19102891. [PMID: 30249046 PMCID: PMC6213211 DOI: 10.3390/ijms19102891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/11/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022] Open
Abstract
Infections contribute to carcinogenesis through inflammation-related mechanisms. H. pylori infection is a significant risk factor for gastric carcinogenesis. However, the molecular mechanism by which H. pylori infection contributes to carcinogenesis has not been fully elucidated. H. pylori-associated chronic inflammation is linked to genomic instability via reactive oxygen and nitrogen species (RONS). In this article, we summarize the current knowledge of H. pylori-induced double strand breaks (DSBs). Furthermore, we provide mechanistic insight into how processing of oxidative DNA damage via base excision repair (BER) leads to DSBs. We review recent studies on how H. pylori infection triggers NF-κB/inducible NO synthase (iNOS) versus NF-κB/nucleotide excision repair (NER) axis-mediated DSBs to drive genomic instability. This review discusses current research findings that are related to mechanisms of DSBs and repair during H. pylori infection.
Collapse
Affiliation(s)
- Dawit Kidane
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA.
| |
Collapse
|
434
|
Merigliano C, Mascolo E, Burla R, Saggio I, Vernì F. The Relationship Between Vitamin B6, Diabetes and Cancer. Front Genet 2018; 9:388. [PMID: 30271425 PMCID: PMC6146109 DOI: 10.3389/fgene.2018.00388] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/29/2018] [Indexed: 11/15/2022] Open
Abstract
Pyridoxal 5′-phosphate (PLP), the active form of vitamin B6, works as cofactor in numerous enzymatic reactions and it behaves as antioxidant molecule. PLP deficiency has been associated to many human pathologies including cancer and diabetes and the mechanism behind this connection is now becoming clearer. Inadequate intake of this vitamin increases the risk of many cancers; furthermore, PLP deprivation impairs insulin secretion in rats, whereas PLP supplementation prevents diabetic complications and improves gestational diabetes. Growing evidence shows that diabetes and cancer are correlated not only because they share same risk factors but also because diabetic patients have a higher risk of developing tumors, although the underlying mechanisms remain elusive. In this review, we will explore data obtained in Drosophila revealing the existence of a connection between vitamin B6, DNA damage and diabetes, as flies in the past decade turned out to be a promising model also for metabolic diseases including diabetes. We will focus on recent studies that revealed a specific role for PLP in maintaining chromosome integrity and glucose homeostasis, and we will show that these aspects are correlated. In addition, we will discuss recent data identifying PLP as a putative linking factor between diabetes and cancer.
Collapse
Affiliation(s)
- Chiara Merigliano
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma, Rome, Italy
| | - Elisa Mascolo
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma, Rome, Italy
| | - Romina Burla
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma, Rome, Italy
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma, Rome, Italy.,Institute of Structural Biology, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Fiammetta Vernì
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
435
|
Song Z, Wang Y, Xiao Q, Yu Z, Zhao L, Wu H, Sun M, Chai Z, Hou P, Geng X, Liu W, Wei M. Poly(ADP-ribose) polymerase-3 overexpression is associated with poor prognosis in patients with breast cancer following chemotherapy. Oncol Lett 2018; 16:5621-5630. [PMID: 30344717 PMCID: PMC6176245 DOI: 10.3892/ol.2018.9398] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 05/18/2017] [Indexed: 12/31/2022] Open
Abstract
Double strand breaks induced by genotoxic agents, if inappropriately repaired, will cause cell death or induce cancer. Poly(ADP-ribose) polymerase-3 (PARP-3) serves a role in double strand break repair, and may be involved in tumorigenesis. To the best of our knowledge, the role of PARP-3 in breast cancer has not yet been examined. In the present study, the expression of PARP-3 was investigated in 493 breast cancer samples and 54 tumor-adjacent control samples using tissue-microarray-based immunohistochemistry. PARP-3 expression was higher in breast cancer samples compared with control samples. PARP-3 overexpression was significantly associated with histological grade II–III (P=0.012). In addition, PARP-3 overexpression was significantly associated with shorter disease-free survival (DFS; P=0.027) time and exhibited a tendency toward shorter overall survival (OS; P=0.183) time in patients with breast cancer compared with patients with lower PARP-3 expression, particularly in BRCA1-positive patients (P=0.004 for disease-free survival and P=0.095 for OS). Multivariate Cox regression analysis indicated that PARP-3 was an independent prognostic factor in patients with breast cancer. Furthermore, it was revealed that PARP-3 overexpression was associated with shorter survival time in patients with cyclophosphamide/doxorubicin or epirubicin/5-fluorouracil (CAF/CEF) chemotherapy compared with low PARP-3 expression, but not in patients with CAF/CEF + docetaxel chemotherapy. The present study suggested that PARP-3 may be used as a biomarker for predicting the clinical outcome of patients receiving chemotherapy, and targeting PARP-3 may be a potential therapeutic strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Zhiguo Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Yong Wang
- Department of General Practice, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Qinghuan Xiao
- Deparment of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Mingli Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Zhangguo Chai
- Outpatient Department, Shenyang Artillery Academy, Shenyang, Liaoning 110867, P.R. China
| | - Ping Hou
- Liaoning Blood Center, Shenyang, Liaoning 110044, P.R. China
| | - Xiaoqiang Geng
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Wensi Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
436
|
Usman M, Volpi EV. DNA damage in obesity: Initiator, promoter and predictor of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 778:23-37. [PMID: 30454680 DOI: 10.1016/j.mrrev.2018.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/29/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
Epidemiological evidence linking obesity with increased risk of cancer is steadily growing, although the causative aspects underpinning this association are only partially understood. Obesity leads to a physiological imbalance in the regulation of adipose tissue and its normal functioning, resulting in hyperglycaemia, dyslipidaemia and inflammation. These states promote the generation of oxidative stress, which is exacerbated in obesity by a decline in anti-oxidant defence systems. Oxidative stress can have a marked impact on DNA, producing mutagenic lesions that could prove carcinogenic. Here we review the current evidence for genomic instability, sustained DNA damage and accelerated genome ageing in obesity. We explore the notion of genotoxicity, ensuing from systemic oxidative stress, as a key oncogenic factor in obesity. Finally, we advocate for early, pre-malignant assessment of genome integrity and stability to inform surveillance strategies and interventions.
Collapse
Affiliation(s)
- Moonisah Usman
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
| | - Emanuela V Volpi
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK.
| |
Collapse
|
437
|
Barichievy S, Naidoo J, Boullé M, Scholefield J, Parihar SP, Coussens AK, Brombacher F, Sigal A, Mhlanga MM. Viral Apoptosis Evasion via the MAPK Pathway by Use of a Host Long Noncoding RNA. Front Cell Infect Microbiol 2018; 8:263. [PMID: 30123777 PMCID: PMC6086015 DOI: 10.3389/fcimb.2018.00263] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/13/2018] [Indexed: 11/30/2022] Open
Abstract
An emerging realization of infectious disease is that pathogens can cause a high incidence of genetic instability within the host as a result of infection-induced DNA lesions. These often lead to classical hallmarks of cancer, one of which is the ability to evade apoptosis despite the presence of numerous genetic mutations that should be otherwise lethal. The Human Immunodeficiency Virus type 1 (HIV-1) is one such pathogen as it induces apoptosis in CD4+ T cells but is largely non-cytopathic in macrophages. As a consequence there is long-term dissemination of the pathogen specifically by these infected yet surviving host cells. Apoptosis is triggered by double-strand breaks (DSBs), such as those induced by integrating retroviruses like HIV-1, and is coordinated by the p53-regulated long noncoding RNA lincRNA-p21. As is typical for a long noncoding RNA, lincRNA-p21 mediates its activities in a complex with one of its two protein binding partners, namely HuR and hnRNP-K. In this work, we monitor the cellular response to infection to determine how HIV-1 induces DSBs in macrophages yet evades apoptosis in these cells. We show that the virus does so by securing the pro-survival MAP2K1/ERK2 cascade early upon entry, in a gp120-dependent manner, to orchestrate a complex dysregulation of lincRNA-p21. By sequestering the lincRNA-p21 partner HuR in the nucleus, HIV-1 enables lincRNA-p21 degradation. Simultaneously, the virus permits transcription of pro-survival genes by sequestering lincRNA-p21's other protein partner hnRNP-K in the cytoplasm via the MAP2K1/ERK2 pathway. Of particular note, this MAP2K1/ERK2 pro-survival cascade is switched off during T cell maturation and is thus unavailable for similar viral manipulation in mature CD4+ T cells. We show that the introduction of MAP2K1, ERK2, or HDM2 inhibitors in HIV-infected macrophages results in apoptosis, providing strong evidence that the viral-mediated apoptotic block can be released, specifically by restoring the nuclear interaction of lincRNA-p21 and its apoptosis protein partner hnRNP-K. Together, these results reveal a unique example of pathogenic control over mammalian apoptosis and DNA damage via a host long noncoding RNA, and present MAP2K1/ERK2 inhibitors as a novel therapeutic intervention strategy for HIV-1 infection in macrophages.
Collapse
Affiliation(s)
- Samantha Barichievy
- Gene Expression and Biophysics Group, Synthetic Biology ERA, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa.,Discovery Sciences, IMED Biotech Unit, AstraZeneca AB R&D, Gothenburg, Sweden
| | - Jerolen Naidoo
- Gene Expression and Biophysics Group, Synthetic Biology ERA, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa.,Division of Chemical Systems and Synthetic Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Mikaël Boullé
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa.,Max Planck Institute for Infection Biology Berlin, Germany
| | - Janine Scholefield
- Gene Expression and Biophysics Group, Synthetic Biology ERA, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa
| | - Suraj P Parihar
- Division of Immunology and South African Medical Research Council Immunology of Infectious Diseases, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicinem University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Anna K Coussens
- Division of Medical Microbiology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology and South African Medical Research Council Immunology of Infectious Diseases, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicinem University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Alex Sigal
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa.,Max Planck Institute for Infection Biology Berlin, Germany
| | - Musa M Mhlanga
- Gene Expression and Biophysics Group, Synthetic Biology ERA, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa.,Division of Chemical Systems and Synthetic Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Gene Expression and Biophysics Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
438
|
Stringer JM, Winship A, Liew SH, Hutt K. The capacity of oocytes for DNA repair. Cell Mol Life Sci 2018; 75:2777-2792. [PMID: 29748894 PMCID: PMC11105623 DOI: 10.1007/s00018-018-2833-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/27/2018] [Accepted: 05/02/2018] [Indexed: 12/18/2022]
Abstract
Female fertility and offspring health are critically dependent on the maintenance of an adequate supply of high-quality oocytes. Like somatic cells, oocytes are subject to a variety of different types of DNA damage arising from endogenous cellular processes and exposure to exogenous genotoxic stressors. While the repair of intentionally induced DNA double strand breaks in gametes during meiotic recombination is well characterised, less is known about the ability of oocytes to repair pathological DNA damage and the relative contribution of DNA repair to oocyte quality is not well defined. This review will discuss emerging data suggesting that oocytes are in fact capable of efficient DNA repair and that DNA repair may be an important mechanism for ensuring female fertility, as well as the transmission of high-quality genetic material to subsequent generations.
Collapse
Affiliation(s)
- Jessica M Stringer
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Amy Winship
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Seng H Liew
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Karla Hutt
- Ovarian Biology Laboratory, Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| |
Collapse
|
439
|
Estiar MA, Mehdipour P. ATM in breast and brain tumors: a comprehensive review. Cancer Biol Med 2018; 15:210-227. [PMID: 30197789 PMCID: PMC6121044 DOI: 10.20892/j.issn.2095-3941.2018.0022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/16/2018] [Indexed: 02/05/2023] Open
Abstract
The ATM gene is mutated in the syndrome, ataxia-telangiectasia (AT), which is characterized by predisposition to cancer. Patients with AT have an elevated risk of breast and brain tumors Carrying mutations in ATM, patients with AT have an elevated risk of breast and brain tumors. An increased frequency of ATM mutations has also been reported in patients with breast and brain tumors; however, the magnitude of this risk remains uncertain. With the exception of a few common mutations, the spectrum of ATM alterations is heterogeneous in diverse populations, and appears to be remarkably dependent on the ethnicity of patients. This review aims to provide an easily accessible summary of common variants in different populations which could be useful in ATM screening programs. In addition, we have summarized previous research on ATM, including its molecular functions. We attempt to demonstrate the significance of ATM in exploration of breast and brain tumors and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mehrdad Asghari Estiar
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran 14155-6447, Iran
| | - Parvin Mehdipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran 14155-6447, Iran
| |
Collapse
|
440
|
Zhang M, Wang L, An K, Cai J, Li G, Yang C, Liu H, Du F, Han X, Zhang Z, Zhao Z, Pei D, Long Y, Xie X, Zhou Q, Sun Y. Lower genomic stability of induced pluripotent stem cells reflects increased non-homologous end joining. Cancer Commun (Lond) 2018; 38:49. [PMID: 30045759 PMCID: PMC6060453 DOI: 10.1186/s40880-018-0313-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 06/11/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) share many common features, including similar morphology, gene expression and in vitro differentiation profiles. However, genomic stability is much lower in iPSCs than in ESCs. In the current study, we examined whether changes in DNA damage repair in iPSCs are responsible for their greater tendency towards mutagenesis. METHODS Mouse iPSCs, ESCs and embryonic fibroblasts were exposed to ionizing radiation (4 Gy) to introduce double-strand DNA breaks. At 4 h later, fidelity of DNA damage repair was assessed using whole-genome re-sequencing. We also analyzed genomic stability in mice derived from iPSCs versus ESCs. RESULTS In comparison to ESCs and embryonic fibroblasts, iPSCs had lower DNA damage repair capacity, more somatic mutations and short indels after irradiation. iPSCs showed greater non-homologous end joining DNA repair and less homologous recombination DNA repair. Mice derived from iPSCs had lower DNA damage repair capacity than ESC-derived mice as well as C57 control mice. CONCLUSIONS The relatively low genomic stability of iPSCs and their high rate of tumorigenesis in vivo appear to be due, at least in part, to low fidelity of DNA damage repair.
Collapse
Affiliation(s)
- Minjie Zhang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Ke An
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jun Cai
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Guochao Li
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Caiyun Yang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Huixian Liu
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Fengxia Du
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Xiao Han
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zilong Zhang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zitong Zhao
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Duanqing Pei
- The Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, P. R. China
| | - Yuan Long
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Yingli Sun
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China.
| |
Collapse
|
441
|
The Human T-Cell Leukemia Virus Type 1 Basic Leucine Zipper Factor Attenuates Repair of Double-Stranded DNA Breaks via Nonhomologous End Joining. J Virol 2018; 92:JVI.00672-18. [PMID: 29769340 DOI: 10.1128/jvi.00672-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/10/2018] [Indexed: 12/27/2022] Open
Abstract
Adult T-cell leukemia (ATL) is a fatal malignancy of CD4+ T cells infected with human T-cell leukemia virus type 1 (HTLV-1). ATL cells often exhibit random gross chromosomal rearrangements that are associated with the induction and improper repair of double-stranded DNA breaks (DSBs). The viral oncoprotein Tax has been reported to impair DSB repair but has not been shown to be consistently expressed throughout all phases of infection. The viral oncoprotein HTLV-1 basic leucine zipper (bZIP) factor (HBZ) is consistently expressed prior to and throughout disease progression, but it is unclear whether it also influences DSB repair. We report that HBZ attenuates DSB repair by nonhomologous end joining (NHEJ), in a manner dependent upon the bZIP domain. HBZ was found to interact with two vital members of the NHEJ core machinery, Ku70 and Ku80, and to be recruited to DSBs in a bZIP-dependent manner in vitro We observed that HBZ expression also resulted in a bZIP-dependent delay in DNA protein kinase (DNA-PK) activation following treatment with etoposide. Although Tax is reported to interact with Ku70, we did not find Tax expression to interfere with HBZ:Ku complex formation. However, as Tax was reported to saturate NHEJ, we found that this effect masked the attenuation of NHEJ by HBZ. Overall, these data suggest that DSB repair mechanisms are impaired not only by Tax but also by HBZ and show that HBZ expression may significantly contribute to the accumulation of chromosomal abnormalities during HTLV-1-mediated oncogenesis.IMPORTANCE Human T-cell leukemia virus type 1 (HTLV-1) infects 15 million to 20 million people worldwide. Approximately 90% of infected individuals are asymptomatic and may remain undiagnosed, increasing the risk that they will unknowingly transmit the virus. About 5% of the HTLV-1-positive population develop adult T-cell leukemia (ATL), a fatal disease that is not highly responsive to treatment. Although ATL development remains poorly understood, two viral proteins, Tax and HBZ, have been implicated in driving disease progression by manipulating host cell signaling and transcriptional pathways. Unlike Tax, HBZ expression is consistently observed in all infected individuals, making it important to elucidate the specific role of HBZ in disease progression. Here, we present evidence that HBZ could promote the accumulation of double-stranded DNA breaks (DSBs) through the attenuation of the nonhomologous end joining (NHEJ) repair pathway. This effect may lead to genome instability, ultimately contributing to the development of ATL.
Collapse
|
442
|
Shin JH, Jung S, Ramakrishna S, Kim HH, Lee J. In vivo gene correction with targeted sequence substitution through microhomology-mediated end joining. Biochem Biophys Res Commun 2018; 502:116-122. [PMID: 29787760 DOI: 10.1016/j.bbrc.2018.05.130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 11/21/2022]
Abstract
Genome editing technology using programmable nucleases has rapidly evolved in recent years. The primary mechanism to achieve precise integration of a transgene is mainly based on homology-directed repair (HDR). However, an HDR-based genome-editing approach is less efficient than non-homologous end-joining (NHEJ). Recently, a microhomology-mediated end-joining (MMEJ)-based transgene integration approach was developed, showing feasibility both in vitro and in vivo. We expanded this method to achieve targeted sequence substitution (TSS) of mutated sequences with normal sequences using double-guide RNAs (gRNAs), and a donor template flanking the microhomologies and target sequence of the gRNAs in vitro and in vivo. Our method could realize more efficient sequence substitution than the HDR-based method in vitro using a reporter cell line, and led to the survival of a hereditary tyrosinemia mouse model in vivo. The proposed MMEJ-based TSS approach could provide a novel therapeutic strategy, in addition to HDR, to achieve gene correction from a mutated sequence to a normal sequence.
Collapse
Affiliation(s)
- Jeong Hong Shin
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, South Korea; Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Soobin Jung
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, South Korea; Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, South Korea; Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, South Korea; Yonsei-IBS Institute, Yonsei University, Seoul, South Korea.
| | - Junwon Lee
- Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea; Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
443
|
Zhu M, Liu W, Shi L, Xiao X, Wu W, Wu L, Zhou Z. Expression of DNA doublestrand repair proteins in oral leukoplakia and the risk of malignant transformation. Oncol Lett 2018; 15:9827-9835. [PMID: 29928356 PMCID: PMC6004653 DOI: 10.3892/ol.2018.8574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 01/05/2018] [Indexed: 12/16/2022] Open
Abstract
The present study assessed the expression of the DNA doublestrand repair (DDR) proteins ATM serine/threonine kinase (ATM), checkpoint kinase 2 (CHEK2) and γH2A histone family member X (γH2AFX) in oral leukoplakia (OL) and evaluated their clinical significance and usefulness as biomarkers for predicting OL transformation. Retrospectively, ATM, CHEK2 and γH2AFX protein levels were evaluated using immunohistochemical analysis in 61 OL, 33 oral squamous cell carcinoma (OSCC) and 15 normal oral mucosa tissues. OL tissues were classified into two groups according to the epithelial dysplasia pathology: The low risk dysplasia group (n=41) and the high-risk dysplasia group (n=20). The results of the present study revealed that the expression of ATM and γH2AFX in OSCC was significantly increased compared with that in OL with low-risk dysplasia and normal oral mucosa tissues. There was no statistically significant difference in CHEK2 expression among the groups. ATM expression was correlated with that of γH2AFX in OSCC tissue. The prognostic values of the DDR proteins and their correlation with clinical and pathological parameters were evaluated further in 99 OL patients with low risk dysplasia. Multivariate analysis revealed that increased expression of ATM and γH2AFX was significantly associated with an increased risk of malignant transformation. Immunohistochemical analysis of ATM and γH2AFX protein expression provided useful prognostic information on the carcinogenesis of OL. Increased ATM and γH2AFX expression may indicate a poor prognosis.
Collapse
Affiliation(s)
- Minwen Zhu
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wei Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Linjun Shi
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xuan Xiao
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wenyan Wu
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Lan Wu
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Zengtong Zhou
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
444
|
DNA damage repair in ovarian cancer: unlocking the heterogeneity. J Ovarian Res 2018; 11:50. [PMID: 29925418 PMCID: PMC6011341 DOI: 10.1186/s13048-018-0424-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/08/2018] [Indexed: 01/13/2023] Open
Abstract
Treatment for advanced ovarian cancer is rarely curative; three quarters of patients with advanced disease relapse and ultimately die with resistant disease. Improving patient outcomes will require the introduction of new treatments and better patient selection. Abrogations in the DNA damage response (DDR) may allow such stratifications. A defective DNA-damage response (DDR) is a defining hallmark of high grade serous ovarian cancer (HGSOC). Indeed, current evidence indicates that all HGSOCs harbour a defect in at least one major DDR pathway. However, defective DDR is not mediated through a single mechanism but rather results from a variety of (epi)genetic lesions affecting one or more of the five major DNA repair pathways. Understanding the relationship between these pathways and how these are abrogated will be necessary in order to facilitate appropriate selection of both existing and novel agents. Here we review the current understanding of the DDR with regard to ovarian, and particularly high grade serous, cancer, with reference to existing and emerging treatments as appropriate.
Collapse
|
445
|
Gee ME, Faraahi Z, McCormick A, Edmondson RJ. DNA damage repair in ovarian cancer: unlocking the heterogeneity. J Ovarian Res 2018. [PMID: 29925418 DOI: 10.1186/s13048-018-0424-x] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Treatment for advanced ovarian cancer is rarely curative; three quarters of patients with advanced disease relapse and ultimately die with resistant disease. Improving patient outcomes will require the introduction of new treatments and better patient selection. Abrogations in the DNA damage response (DDR) may allow such stratifications.A defective DNA-damage response (DDR) is a defining hallmark of high grade serous ovarian cancer (HGSOC). Indeed, current evidence indicates that all HGSOCs harbour a defect in at least one major DDR pathway. However, defective DDR is not mediated through a single mechanism but rather results from a variety of (epi)genetic lesions affecting one or more of the five major DNA repair pathways. Understanding the relationship between these pathways and how these are abrogated will be necessary in order to facilitate appropriate selection of both existing and novel agents.Here we review the current understanding of the DDR with regard to ovarian, and particularly high grade serous, cancer, with reference to existing and emerging treatments as appropriate.
Collapse
Affiliation(s)
- Mary Ellen Gee
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK.,Department of Obstetrics and Gynaecology, Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Level 5, Research, Oxford Road, Manchester, UK
| | - Zahra Faraahi
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK
| | - Aiste McCormick
- Northern Institute for Cancer Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4AD, UK
| | - Richard J Edmondson
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK. .,Department of Obstetrics and Gynaecology, Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Level 5, Research, Oxford Road, Manchester, UK.
| |
Collapse
|
446
|
Gee ME, Faraahi Z, McCormick A, Edmondson RJ. DNA damage repair in ovarian cancer: unlocking the heterogeneity. J Ovarian Res 2018. [PMID: 29925418 DOI: 10.1186/s13048-018-0424-x]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Treatment for advanced ovarian cancer is rarely curative; three quarters of patients with advanced disease relapse and ultimately die with resistant disease. Improving patient outcomes will require the introduction of new treatments and better patient selection. Abrogations in the DNA damage response (DDR) may allow such stratifications.A defective DNA-damage response (DDR) is a defining hallmark of high grade serous ovarian cancer (HGSOC). Indeed, current evidence indicates that all HGSOCs harbour a defect in at least one major DDR pathway. However, defective DDR is not mediated through a single mechanism but rather results from a variety of (epi)genetic lesions affecting one or more of the five major DNA repair pathways. Understanding the relationship between these pathways and how these are abrogated will be necessary in order to facilitate appropriate selection of both existing and novel agents.Here we review the current understanding of the DDR with regard to ovarian, and particularly high grade serous, cancer, with reference to existing and emerging treatments as appropriate.
Collapse
Affiliation(s)
- Mary Ellen Gee
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK.,Department of Obstetrics and Gynaecology, Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Level 5, Research, Oxford Road, Manchester, UK
| | - Zahra Faraahi
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK
| | - Aiste McCormick
- Northern Institute for Cancer Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4AD, UK
| | - Richard J Edmondson
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK. .,Department of Obstetrics and Gynaecology, Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Level 5, Research, Oxford Road, Manchester, UK.
| |
Collapse
|
447
|
Higelin J, Catanese A, Semelink-Sedlacek LL, Oeztuerk S, Lutz AK, Bausinger J, Barbi G, Speit G, Andersen PM, Ludolph AC, Demestre M, Boeckers TM. NEK1 loss-of-function mutation induces DNA damage accumulation in ALS patient-derived motoneurons. Stem Cell Res 2018; 30:150-162. [PMID: 29929116 DOI: 10.1016/j.scr.2018.06.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/22/2018] [Accepted: 06/07/2018] [Indexed: 10/14/2022] Open
Abstract
Mutations in genes coding for proteins involved in DNA damage response (DDR) and repair, such as C9orf72 and FUS (Fused in Sarcoma), are associated with neurodegenerative diseases and lead to amyotrophic lateral sclerosis (ALS). Heterozygous loss-of-function mutations in NEK1 (NIMA-related kinase 1) have also been recently found to cause ALS. NEK1 codes for a multifunctional protein, crucially involved in mitotic checkpoint control and DDR. To resolve pathological alterations associated with NEK1 mutation, we compared hiPSC-derived motoneurons carrying a NEK1 mutation with mutant C9orf72 and wild type neurons at basal level and after DNA damage induction. Motoneurons carrying a C9orf72 mutation exhibited cell specific signs of increased DNA damage. This phenotype was even more severe in NEK1c.2434A>T neurons that showed significantly increased DNA damage at basal level and impaired DDR after induction of DNA damage in an maturation-dependent manner. Our results provide first mechanistic insight in pathophysiological alterations induced by NEK1 mutations and point to a converging pathomechanism of different gene mutations causative for ALS. Therefore, our study contributes to the development of novel therapeutic strategies to reduce DNA damage accumulation in neurodegenerative diseases and ALS.
Collapse
Affiliation(s)
- Julia Higelin
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany; International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Alberto Catanese
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany; International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | | | - Sertap Oeztuerk
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany; International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | | | - Gotthold Barbi
- Institute for Human Genetics, University Ulm, Ulm, Germany
| | - Günter Speit
- Institute for Human Genetics, University Ulm, Ulm, Germany
| | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umea University, Umea, Sweden
| | | | - Maria Demestre
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.
| |
Collapse
|
448
|
Zhou ZR, Yang ZZ, Yu XL, Guo XM. Highlights on molecular targets for radiosensitization of breast cancer cells: Current research status and prospects. Cancer Med 2018; 7:3110-3117. [PMID: 29856131 PMCID: PMC6051209 DOI: 10.1002/cam4.1588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/16/2022] Open
Abstract
In the past, searching for effective radiotherapy sensitization molecular targets and improving the radiation sensitivity of malignant tumors was the hot topic for the oncologists, but with little achievements. We will summarize the research results about breast cancer irradiation sensitization molecular targets over the past two decades; we mainly focus on the following aspects: DNA damage repair and radiation sensitization, cell cycle regulation and radiation sensitization, cell autophagy regulation and radiation sensitization, and radiation sensitivity prediction and breast cancer radiotherapy scheme making. And based on this summary, we will put forward some of our viewpoints.
Collapse
Affiliation(s)
- Zhi-Rui Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhao-Zhi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Li Yu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Mao Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
449
|
Ji J, Chen W, Lian W, Chen R, Yang J, Zhang Q, Weng Q, Khan Z, Hu J, Chen X, Zou P, Chen X, Liang G. (S)-crizotinib reduces gastric cancer growth through oxidative DNA damage and triggers pro-survival akt signal. Cell Death Dis 2018; 9:660. [PMID: 29855474 PMCID: PMC5981313 DOI: 10.1038/s41419-018-0667-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/30/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC), a common gastrointestinal malignancy worldwide, has poor prognosis and frequent recurrence. There is a great need to identify effective therapy for GC. Crizotinib is a multi-targeted, clinically available oral tyrosine kinase inhibitor approved for lung cancer, but its use for the highly heterogeneous disease of GC is unknown. The goal of this study was to investigate the anti-cancer mechanisms of the (S)-crizotinib in inhibiting GC growth. Human GC cell lines (SGC-7901 and BGC-823) and the (S)-crizotinib-resistant BGC-823/R were cultured for determining the effects of (S)-crizotinib on cell viability, apoptosis, oxidant generation, and cell cycle progression. Involvement of ROS, Akt signaling, MTH1, and DNA damage was tested with respective pharmacological blockade. The in vivo anti-tumor effects of (S)-crizotinib were determined using xenograft tumor mice. Results indicated that (S)-crizotinib decreased GC cell viability, induced growth arrest and apoptosis, and increased levels of γH2AX and Ser1981-phosphorylated ATM, which were inhibited by NAC. The anti-cancer mechanism of (S)-crizotinib was independent of MTH1. Moreover, ATM-activated Akt, a pro-survival signal, whose inhibition further enhanced (S)-crizotinib-induced inhibition of GC cell growth and tumor growth in xenograft mice, and re-sensitized resistant GC cells to (S)-crizotinib. (S)-crizotinib reduced GC cell and tumor growth through oxidative DNA damage mechanism and triggered pro-survival Akt signaling. We conclude that inclusion of Akt inhibition (to block the survival signaling) with (S)-crizotinib may provide an effective and novel combination therapy for GC in the clinical setting.
Collapse
Affiliation(s)
- Jiansong Ji
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Weiqian Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Weishuai Lian
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jinqing Yang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qianqian Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Qiaoyou Weng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Zia Khan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Hu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Peng Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaoming Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
450
|
Thongsroy J, Patchsung M, Pongpanich M, Settayanon S, Mutirangura A. Reduction in replication-independent endogenous DNA double-strand breaks promotes genomic instability during chronological aging in yeast. FASEB J 2018; 32:fj201800218RR. [PMID: 29812972 DOI: 10.1096/fj.201800218rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The mechanism that causes genomic instability in nondividing aging cells is unknown. Our previous study of mutant yeast suggested that 2 types of replication-independent endogenous DNA double-strand breaks (RIND-EDSBs) exist and that they play opposing roles. The first type, known as physiologic RIND-EDSBs, were ubiquitous in the G0 phase of both yeast and human cells in certain genomic locations and may act as epigenetic markers. Low RIND-EDSB levels were found in mutants that lacked chromatin-condensing proteins, such as the high-mobility group box (HMGB) proteins and Sir2. The second type is referred to as pathologic RIND-EDSBs. High pathological RIND-EDSB levels were found in DSB repair mutants. Under normal physiologic conditions, these excess RIND-EDSBs are repaired in much the same way as DNA lesions. Here, chronological aging in yeast reduced physiological RIND-EDSBs and cell viability. A strong correlation was observed between the reduction in RIND-EDSBs and viability in aging yeast cells ( r = 0.94, P < 0.0001). We used galactose-inducible HO endonuclease (HO) and nhp6a∆, an HMGB protein mutant, to evaluate the consequences of reduced physiological RIND-EDSB levels. The HO-induced cells exhibited a sustained reduction in RIND-EDSBs at various levels for several days. Interestingly, we found that lower physiologic RIND-EDSB levels resulted in decreased cell viability ( r = 0.69, P < 0.0001). Treatment with caffeine, a DSB repair inhibitor, increased pathological RIND-EDSBs, which were distinguished from physiologic RIND-EDSBs by their lack of sequences prior to DSB in untreated cells [odds ratio (OR) ≤1]. Caffeine treatment in both the HO-induced and nhp6a∆ cells markedly increased OR ≤1 breaks. Therefore, physiological RIND-EDSBs play an epigenetic role in preventing pathological RIND-EDSBs, a type of DNA damage. In summary, the reduction of physiological RIND-EDSB level is a genomic instability mechanism in chronologically aging cells.-Thongsroy, J., Patchsung, M., Pongpanich, M., Settayanon, S., Mutirangura, A. Reduction in replication-independent endogenous DNA double-strand breaks promotes genomic instability during chronological aging in yeast.
Collapse
Affiliation(s)
- Jirapan Thongsroy
- School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | - Maturada Patchsung
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Monnat Pongpanich
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Omics Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Sirapat Settayanon
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Apiwat Mutirangura
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|