1
|
Zech HB, von Bargen C, Oetting A, Möckelmann N, Möller-Koop C, Witt M, Struve N, Petersen C, Betz C, Rothkamm K, Münscher A, Clauditz TS, Rieckmann T. Tissue microarray analyses of the essential DNA repair factors ATM, DNA-PKcs and Ku80 in head and neck squamous cell carcinoma. Radiat Oncol 2024; 19:150. [PMID: 39478631 PMCID: PMC11523811 DOI: 10.1186/s13014-024-02541-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) negative for Human Papillomavirus (HPV) has remained a difficult to treat entity, whereas tumors positive for HPV are characterized by radiosensitivity and favorable patient outcome. On the cellular level, radiosensitivity is largely governed by the tumor cells` ability to repair radiation-induced DNA double-strand breaks (DSBs), but no biomarker is established that could guide clinical decision making. Therefore, we tested the impact of the expression levels of ATM, the central kinase of the DNA damage response as well as DNA-PKcs and Ku80, two major factors in the main DSB repair pathway non-homologous end joining (NHEJ). METHODS A tissue microarray of a single center HNSCC cohort was stained for ATM, DNA-PKcs and Ku80 and the expression scored based on staining intensity and the percentages of tumor cells stained. Scores were correlated with clinicopathological parameters and survival. RESULTS Samples from 427 HNSCC patients yielded interpretable stainings and were scored following an established algorithm. The majority of tumors showed strong expression of both NHEJ factors, whereas the expression of ATM varied more. The expression scores of ATM and DNA-PKcs were not associated with patient survival. For HPV-negative HNSCC, the minority of tumors without strong Ku80 expression trended towards superior survival when treatment included radiotherapy. Focusing stronger on staining intensity to define the subgroup with lowest and therefore potentially insufficient expression levels in the HPV-negative subgroup, we observed significantly better overall survival for patients treated with radiotherapy but not with surgery alone. CONCLUSIONS Our data suggest that HPV-negative HNSCC with particularly low Ku80 expression represent a highly radiosensitive subpopulation. Confirmation in independent cohorts is required.
Collapse
Affiliation(s)
- Henrike Barbara Zech
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clara von Bargen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Agnes Oetting
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nikolaus Möckelmann
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Otorhinolaryngology, Marienkrankenhaus, Hamburg, Germany
| | - Christina Möller-Koop
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Witt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Struve
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Betz
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adrian Münscher
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Otorhinolaryngology, Marienkrankenhaus, Hamburg, Germany
| | | | - Thorsten Rieckmann
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2
|
Chen X, Chen C, Luo C, Liu J, Lin Z. Discovery of UMI-77 as a novel Ku70/80 inhibitor sensitizing cancer cells to DNA damaging agents in vitro and in vivo. Eur J Pharmacol 2024; 975:176647. [PMID: 38754534 DOI: 10.1016/j.ejphar.2024.176647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/18/2024]
Abstract
The emergence of chemoresistance poses a significant challenge to the efficacy of DNA-damaging agents in cancer treatment, in part due to the inherent DNA repair capabilities of cancer cells. The Ku70/80 protein complex (Ku) plays a central role in double-strand breaks (DSBs) repair through the classical non-homologous end joining (c-NHEJ) pathway, and has proven to be one of the most promising drug target for cancer treatment when combined with radiotherapy or chemotherapy. In this study, we conducted a high-throughput screening of small-molecule inhibitors targeting the Ku complex by using a fluorescence polarization-based DNA binding assay. From a library of 11,745 small molecules, UMI-77 was identified as a potent Ku inhibitor, with an IC50 value of 2.3 μM. Surface plasmon resonance and molecular docking analyses revealed that UMI-77 directly bound the inner side of Ku ring, thereby disrupting Ku binding with DNA. In addition, UMI-77 also displayed potent inhibition against MUS81-EME1, a key player in homologous recombination (HR), demonstrating its potential for blocking both NHEJ- and HR-mediated DSB repair pathways. Further cell-based studies showed that UMI-77 could impair bleomycin-induced DNA damage repair, and significantly sensitized multiple cancer cell lines to the DNA-damaging agents. Finally, in a mouse xenograft tumor model, UMI-77 significantly enhanced the chemotherapeutic efficacy of etoposide with little adverse physiological effects. Our work offers a new avenue to combat chemoresistance in cancer treatment, and suggests that UMI-77 could be further developed as a promising candidate in cancer treatment.
Collapse
Affiliation(s)
- Xuening Chen
- College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Changkun Chen
- College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Chengmiao Luo
- College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jianyong Liu
- College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zhonghui Lin
- College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
3
|
Fabbrizi MR, Doggett TJ, Hughes JR, Melia E, Dufficy ER, Hill RM, Goula A, Phoenix B, Parsons JL. Inhibition of key DNA double strand break repair protein kinases enhances radiosensitivity of head and neck cancer cells to X-ray and proton irradiation. Cell Death Discov 2024; 10:282. [PMID: 38866739 PMCID: PMC11169544 DOI: 10.1038/s41420-024-02059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
Ionising radiation (IR) is widely used in cancer treatment, including for head and neck squamous cell carcinoma (HNSCC), where it induces significant DNA damage leading ultimately to tumour cell death. Among these lesions, DNA double strand breaks (DSBs) are the most threatening lesion to cell survival. The two main repair mechanisms that detect and repair DSBs are non-homologous end joining (NHEJ) and homologous recombination (HR). Among these pathways, the protein kinases ataxia telangiectasia mutated (ATM), ataxia telangiectasia and Rad3-related (ATR) and the DNA dependent protein kinase catalytic subunit (DNA-Pkcs) play key roles in the sensing of the DSB and subsequent coordination of the downstream repair events. Consequently, targeting these kinases with potent and specific inhibitors is considered an approach to enhance the radiosensitivity of tumour cells. Here, we have investigated the impact of inhibition of ATM, ATR and DNA-Pkcs on the survival and growth of six radioresistant HPV-negative HNSCC cell lines in combination with either X-ray irradiation or proton beam therapy, and confirmed the mechanistic pathway leading to cell radiosensitisation. Using inhibitors targeting ATM (AZD1390), ATR (AZD6738) and DNA-Pkcs (AZD7648), we observed that this led to significantly decreased clonogenic survival of HNSCC cell lines following both X-ray and proton irradiation. Radiosensitisation of HNSCC cells grown as 3D spheroids was also observed, particularly following ATM and DNA-Pkcs inhibition. We confirmed that the inhibitors in combination with X-rays and protons led to DSB persistence, and increased micronuclei formation. Cumulatively, our data suggest that targeting DSB repair, particularly via ATM and DNA-Pkcs inhibition, can exacerbate the impact of ionising radiation in sensitising HNSCC cell models.
Collapse
Affiliation(s)
- Maria Rita Fabbrizi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK
| | - Thomas J Doggett
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Jonathan R Hughes
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK
| | - Emma Melia
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK
| | - Elizabeth R Dufficy
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK
| | - Rhianna M Hill
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Amalia Goula
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK
| | - Ben Phoenix
- School of Physics and Astronomy, University of Birmingham, Edgbaston, UK
| | - Jason L Parsons
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK.
- School of Physics and Astronomy, University of Birmingham, Edgbaston, UK.
| |
Collapse
|
4
|
Pavlova G, Belyashova A, Savchenko E, Panteleev D, Shamadykova D, Nikolaeva A, Pavlova S, Revishchin A, Golbin D, Potapov A, Antipina N, Golanov A. Reparative properties of human glioblastoma cells after single exposure to a wide range of X-ray doses. Front Oncol 2022; 12:912741. [PMID: 35992802 PMCID: PMC9386365 DOI: 10.3389/fonc.2022.912741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Radiation therapy induces double-stranded DNA breaks in tumor cells, which leads to their death. A fraction of glioblastoma cells repair such breaks and reinitiate tumor growth. It was necessary to identify the relationship between high radiation doses and the proliferative activity of glioblastoma cells, and to evaluate the contribution of DNA repair pathways, homologous recombination (HR), and nonhomologous end joining (NHEJ) to tumor-cell recovery. We demonstrated that the GO1 culture derived from glioblastoma cells from Patient G, who had previously been irradiated, proved to be less sensitive to radiation than the Sus\fP2 glioblastoma culture was from Patient S, who had not been exposed to radiation before. GO1 cell proliferation decreased with radiation dose, and MTT decreased to 35% after a single exposure to 125 Gγ. The proliferative potential of glioblastoma culture Sus\fP2 decreased to 35% after exposure to 5 Gγ. At low radiation doses, cell proliferation and the expression of RAD51 were decreased; at high doses, cell proliferation was correlated with Ku70 protein expression. Therefore, HR and NHEJ are involved in DNA break repair after exposure to different radiation doses. Low doses induce HR, while higher doses induce the faster but less accurate NHEJ pathway of double-stranded DNA break repair.
Collapse
Affiliation(s)
- Galina Pavlova
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
- Department of Medical Genetics, Sechenov First Moscow State Medical University, Moscow, Russia
- *Correspondence: Galina Pavlova,
| | - Alexandra Belyashova
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Ekaterina Savchenko
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Dmitri Panteleev
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Dzhirgala Shamadykova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Anna Nikolaeva
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Svetlana Pavlova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Alexander Revishchin
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Denis Golbin
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Alexander Potapov
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Natalia Antipina
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Andrey Golanov
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| |
Collapse
|
5
|
Yang X, Yang F, Lan L, Wen N, Li H, Sun X. Potential value of PRKDC as a therapeutic target and prognostic biomarker in pan-cancer. Medicine (Baltimore) 2022; 101:e29628. [PMID: 35801800 PMCID: PMC9259106 DOI: 10.1097/md.0000000000029628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND While protein kinase, DNA-activated, catalytic subunit (PRKDC) plays an important role in double-strand break repair to retain genomic stability, there is still no pan-cancer analysis based on large clinical information on the relationship between PRKDC and different tumors. For the first time, this research used numerous databases to perform a pan-cancer review for PRKDC to explore the possible mechanism of PRKDC in the etiology and outcomes in various tumors. METHODS PRKDC's expression profile and prognostic significance in pan-cancer were investigated based on various databases and online platforms, including TIMER2, GEPIA2, cBioPortal, CPTAC, and SangerBox. We applied the TIMER to identified the interlink of PRKDC and the immune infiltration in assorted tumors, and the SangerBox online platform was adopted to find out the relevance between PRKDC and immune checkpoint genes, tumor mutation burden, and microsatellite instability in tumors. GeneMANIA tool was employed to create a protein-protein interaction analysis, gene set enrichment analysis was conducted to performed gene enrichment analysis. RESULTS Overall, tumor tissue presented a higher degree of PRKDC expression than adjacent normal tissue. Meanwhile, patients with high PRKDC expression have a worse prognosis. PRKDC mutations were present in almost all The Cancer Genome Atlas tumors and might lead to a better survival prognosis. The PRKDC expression level was shown a positive correlation with tumor-infiltrating immune cells. PRKDC high expression cohorts were enriched in "cell cycle" "oocyte meiosis" and "RNA-degradation" signaling pathways. CONCLUSIONS This study revealed the potential value of PRKDC in tumor immunology and as a therapeutic target and prognostic biomarker in pan-cancer.
Collapse
Affiliation(s)
- Xiawei Yang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Feng Yang
- Department of Gynocology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Liugen Lan
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Ning Wen
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Haibin Li
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xuyong Sun
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory for Transplantation Medicine, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Transplantation Medicine Research Center of Engineering Technology, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
6
|
Prognostic significance of metabolic tumour volume and maximum standard uptake value of fluor-18-fluorodeoxyglucose positron emission tomography with computed tomography in nasopharyngeal carcinoma. Contemp Oncol (Pozn) 2021; 25:153-159. [PMID: 34729034 PMCID: PMC8547178 DOI: 10.5114/wo.2021.109620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/19/2021] [Indexed: 11/17/2022] Open
Abstract
Aim of the study To evaluate the prognostic role of markers of fluor-18-fluorodeoxyglucose positron emission tomography with computed tomography (18F-FDG-PET-CT), such as maximum standard uptake value (SUVmax) and metabolic tumour volume (MTV) measured at primary and nodal disease, and their clinical significance in terms of predicting treatment outcomes and survival. Material and methods Between January 2017 and January 2020, 20 case records of nasopharyngeal carcinoma patients who underwent 18F-FDG-PET-CT as part of staging workup before radiotherapy and as a part of response evaluation after radiotherapy were retrospectively reviewed. Results At a median follow-up of 34.7 months, the 2-year progression-free survival (PFS) was 70% and 2-year overall survival (OS) was 79%. Patients with a lower nodal SUVmax (SUVmax-N) had a better 2-year PFS (91% vs. 46%; p = 0.035) and 2-year OS (95% vs. 58%; p = 0.015). A high SUVmax-N of > 10.58 was a negative predictor of OS (95% confidence interval [CI]: 0.93-1; p = 0.003) as well as PFS (95% CI: 0.64-1; p = 0.017). Also, a high MTV > 25.8 cm3 was a negative predictor of PFS (95% CI: 0.58-0.98; p = 0.048). MTV was an independent predictor of PFS and OS on univariate analysis, whereas it was not significant in the Cox regression multivariate analysis. Conclusions High values of MTV and SUVmax-N can be considered as independent prognostic factors of OS and PFS in nasopharyngeal cancer patients treated with concurrent chemoradiation, highlighting the need for more intensified treatment.
Collapse
|
7
|
Hu S, Hui Z, Lirussi F, Garrido C, Ye XY, Xie T. Small molecule DNA-PK inhibitors as potential cancer therapy: a patent review (2010-present). Expert Opin Ther Pat 2021; 31:435-452. [PMID: 33347360 DOI: 10.1080/13543776.2021.1866540] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: DNA-dependent protein kinase (DNA-PK) plays a crucial role in the repair of DSBs via non-homologous end joining (NHEJ). Several DNA-PK inhibitors are being investigated for potential anticancer treatment in clinical trials.Area covered: This review aims to give an overview of patents published since 2010 by analyzing the patent space and structure features of scaffolds used in those patents. It also discusses the recent clinical developments and provides perspectives on future challenges and directions in this field.Expert opinion: As a key component of the DNA damage response (DDR) pathway, DNA-PK appears to be a viable drug target for anticancer therapy. The clinical investigation of a DNA-PK inhibitor employs both a monotherapy and a combination strategy. In the combination strategy, a DNA-PK inhibitor is typically combined with a DSB inducer, radiation, a chemotherapy agent, or a PARP inhibitor, etc. Patent analyses suggest that diverse structures comprising different scaffolds from mono-heteroaryl to bicyclic heteroaryl to tricyclic heteroaryl are capable to achieve good DNA-PK inhibitory activity and good DNA-PK selectivity over other closely related enzymes. Several DNA-PK inhibitors are currently being evaluated in clinics, with the hope to get approval in the near future.
Collapse
Affiliation(s)
- Suwen Hu
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, Zhejiang, People's Republic of China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Zhejiang Province, People's Republic of China.,;cCollaborative Innovation Center of Chinese Medicines from Zhejiang Province, Zhejiang Province, People's Republic of China.,;dKey Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, People's Republic of China.,;eHangzhou Huadong Medicine Group, Pharmaceutical Research Institute Co. Ltd, Hanzhou City, Zhejiang Province, People's Republic of China
| | - Zi Hui
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, Zhejiang, People's Republic of China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Zhejiang Province, People's Republic of China.,;cCollaborative Innovation Center of Chinese Medicines from Zhejiang Province, Zhejiang Province, People's Republic of China.,;Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, People's Republic of China
| | - Frédéric Lirussi
- ;fINSERM, U1231, Label LipSTIC, and Ligue Nationale Contre Le Cancer, Dijon, France.,;gUniversité De Bourgogne-Franche Comté, I-SITE, France.,;hDepartment of Pharmacology-Toxicology & Metabolomics, University hospital of Besançon (CHU), 2 Boulevard Fleming, 25030 BESANCON, France
| | - Carmen Garrido
- ;INSERM, U1231, Label LipSTIC, and Ligue Nationale Contre Le Cancer, Dijon, France.,;Université De Bourgogne-Franche Comté, I-SITE, France.,;iAnti-cancer Center George-François Leclerc, CGFL, Dijon, France
| | - Xiang-Yang Ye
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, Zhejiang, People's Republic of China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Zhejiang Province, People's Republic of China.,;cCollaborative Innovation Center of Chinese Medicines from Zhejiang Province, Zhejiang Province, People's Republic of China.,;Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, People's Republic of China
| | - Tian Xie
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang Province, Zhejiang, People's Republic of China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Zhejiang Province, People's Republic of China.,;cCollaborative Innovation Center of Chinese Medicines from Zhejiang Province, Zhejiang Province, People's Republic of China.,;Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
8
|
LncRNA linc00312 suppresses radiotherapy resistance by targeting DNA-PKcs and impairing DNA damage repair in nasopharyngeal carcinoma. Cell Death Dis 2021; 12:69. [PMID: 33431817 PMCID: PMC7801696 DOI: 10.1038/s41419-020-03302-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 02/05/2023]
Abstract
Radioresistance is the main obstacle in the clinical management of nasopharyngeal carcinoma (NPC). linc00312 is deregulated in a number of human cancers, including NPC. However, the detailed functions and underlying mechanisms of linc00312 in regulating radiosensitivity of NPC remains unknown. In this study, cox regression analysis was used to assess the association between linc00312 and NPC patients’ survival after radiotherapy. Our results reveal that linc00312 is significantly down-regulated in NPC tissues and patients with higher expression of linc00312 are significantly associated with longer overall survival and better short-term radiotherapy efficacy. Overexpression of linc00312 could increase the sensitivity of NPC cells to ionizing radiation, as indicated by clonogenic survival assay, comet assay, and flow cytometry. Mechanistically, RNA pull down and RNA immunoprecipitation were performed to investigate the binding proteins of linc00312. linc00312 directly binds to DNA-PKcs, hinders the recruitment of DNA-PKcs to Ku80, and inhibits phosphorylation of AKT–DNA–PKcs axis, therefore inhibiting the DNA damage signal sensation and transduction in the NHEJ repair pathway. In addition, linc00312 impairs DNA repair and cell cycle control by suppressing MRN–ATM–CHK2 signal and ATR–CHK1 signal. In summary, we identified DNA-PKcs as the binding protein of linc00312 and revealed a novel mechanism of linc00312 in the DNA damage response, providing evidence for a potential therapeutic strategy in NPC.
Collapse
|
9
|
Mohiuddin IS, Kang MH. DNA-PK as an Emerging Therapeutic Target in Cancer. Front Oncol 2019; 9:635. [PMID: 31380275 PMCID: PMC6650781 DOI: 10.3389/fonc.2019.00635] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022] Open
Abstract
The DNA-dependent protein kinase (DNA-PK) plays an instrumental role in the overall survival and proliferation of cells. As a member of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, DNA-PK is best known as a mediator of the cellular response to DNA damage. In this context, DNA-PK has emerged as an intriguing therapeutic target in the treatment of a variety of cancers, especially when used in conjunction with genotoxic chemotherapy or ionizing radiation. Beyond the DNA damage response, DNA-PK activity is necessary for multiple cellular functions, including the regulation of transcription, progression of the cell cycle, and in the maintenance of telomeres. Here, we review what is currently known about DNA-PK regarding its structure and established roles in DNA repair. We also discuss its lesser-known functions, the pharmacotherapies inhibiting its function in DNA repair, and its potential as a therapeutic target in a broader context.
Collapse
Affiliation(s)
- Ismail S Mohiuddin
- Cancer Center, Department of Pediatrics, Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Min H Kang
- Cancer Center, Department of Pediatrics, Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
10
|
Geng W, Tian D, Wang Q, Shan S, Zhou J, Xu W, Shan H. DNA‑PKcs inhibitor increases the sensitivity of gastric cancer cells to radiotherapy. Oncol Rep 2019; 42:561-570. [PMID: 31173270 PMCID: PMC6610038 DOI: 10.3892/or.2019.7187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 05/23/2019] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer (GC) is a severe public health problem worldwide, particularly in China. Radiotherapy is the main locoregional treatment for various types of unresectable tumor, including GC. However, many patients fail to respond to radiotherapy due to the intrinsic radioresistance of cancer cells. This study was designed to investigate the effects and potential mechanism of radiosensitization associated with DNA-dependent protein kinase catalytic subunit (DNA-PKcs) inhibitor in human GC cell lines in vitro. Among the six GC cell lines (SGC7901, HGC-27, MKN45, MKN74, BGC823 and MGC803) that were exposed to increasing doses of IR (0, 2, 4, 6 and 8 Gy), the mean lethal dose and quasi-threshold dose measurements indicated that BGC823 and MGC803 were relatively insensitive to ionizing radiation (IR). IR induced significant elevation of γ H2A histone family member X (γH2AX) in MKN45 cells compared with BGC823 cells. DNA-PKcs and phospho-DNA-PKcs protein levels were increased in BGC823 and MGC803 cells compared with other GC cell lines (SGC7901, HGC-27, MKN45 and MKN74). DNA-PKcs inhibition led to increased sensitivity of BGC823 and MGC803 cells to IR. NU7441 increased γH2AX expression in the nuclei of BGC823 cells following IR. Combination of DNA-PKcs and CK2 inhibition further increased the sensitivity of GC cells to IR. The combination of NU7441 and CX4945 increased γH2AX expression in the nucleus of BGC823 cells following IR compared with treatment with NU7441 alone. Taken together, the findings suggest that DNA-PKcs inhibitor increased the sensitivity of radioresistant BGC823 and MGC803 cells to radiotherapy through the cleaved-caspase3/γH2AX signaling pathway, thus presenting a potential treatment method for GC.
Collapse
Affiliation(s)
- Wei Geng
- Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224005, P.R. China
| | - Dalong Tian
- Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224005, P.R. China
| | - Qiang Wang
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Shunlin Shan
- Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224005, P.R. China
| | - Jianwei Zhou
- Cancer Center of The 82nd Hospital of PLA, Huaian, Jiangsu 223001, P.R. China
| | - Wenxia Xu
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Husheng Shan
- Cancer Center of The 82nd Hospital of PLA, Huaian, Jiangsu 223001, P.R. China
| |
Collapse
|
11
|
Ihara M, Ashizawa K, Shichijo K, Kudo T. Expression of the DNA-dependent protein kinase catalytic subunit is associated with the radiosensitivity of human thyroid cancer cell lines. JOURNAL OF RADIATION RESEARCH 2019; 60:171-177. [PMID: 30476230 PMCID: PMC6430255 DOI: 10.1093/jrr/rry097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/13/2018] [Indexed: 05/02/2023]
Abstract
The prognosis and treatment of thyroid cancer depends on the type and stage of the disease. Radiosensitivity differs among cancer cells owing to their varying capacity for repair after irradiation. Radioactive iodine can be used to destroy thyroid cancer cells. However, patient prognosis and improvement after irradiation varies. Therefore, predictive measures are important for avoiding unnecessary exposure to radiation. We describe a new method for predicting the effects of radiation in individual cases of thyroid cancer based on the DNA-dependent protein kinase (DNA-PK) activity level in cancer cells. The radiation sensitivity, DNA-PK activity, and cellular levels of DNA-PK complex subunits in five human thyroid cancer cell lines were analyzed in vitro. A positive correlation was observed between the D10 value (radiation dose that led to 10% survival) of cells and DNA-PK activity. This correlation was not observed after treatment with NU7441, a DNA-PK-specific inhibitor. A significant correlation was also observed between DNA-PK activity and expression levels of the DNA-PK catalytic subunit (DNA-PKcs). Cells expressing low DNA-PKcs levels were radiation-sensitive, and cells expressing high DNA-PKcs levels were radiation-resistant. Our results indicate that radiosensitivity depends on the expression level of DNA-PKcs in thyroid cancer cell lines. Thus, the DNA-PKcs expression level is a potential predictive marker of the success of radiation therapy for thyroid tumors.
Collapse
Affiliation(s)
- Makoto Ihara
- Department of Radioisotope Medicine, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki, Japan
- Corresponding author. Department of Radioisotope Medicine, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki 852-8523, Japan. Tel: +81-95-819-71013; Fax: +81-95-849-7104;
| | - Kiyoto Ashizawa
- Department of Radioisotope Medicine, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki, Japan
| | - Kazuko Shichijo
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki, Japan
| | - Takashi Kudo
- Department of Radioisotope Medicine, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki, Japan
| |
Collapse
|
12
|
George VC, Ansari SA, Chelakkot VS, Chelakkot AL, Chelakkot C, Menon V, Ramadan W, Ethiraj KR, El-Awady R, Mantso T, Mitsiogianni M, Panagiotidis MI, Dellaire G, Vasantha Rupasinghe HP. DNA-dependent protein kinase: Epigenetic alterations and the role in genomic stability of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 780:92-105. [PMID: 31395353 DOI: 10.1016/j.mrrev.2018.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/13/2018] [Indexed: 12/28/2022]
Abstract
DNA-dependent protein kinase (DNA-PK), a member of phosphatidylinositol-kinase family, is a key protein in mammalian DNA double-strand break (DSB) repair that helps to maintain genomic integrity. DNA-PK also plays a central role in immune cell development and protects telomerase during cellular aging. Epigenetic deregulation due to endogenous and exogenous factors may affect the normal function of DNA-PK, which in turn could impair DNA repair and contribute to genomic instability. Recent studies implicate a role for epigenetics in the regulation of DNA-PK expression in normal and cancer cells, which may impact cancer progression and metastasis as well as provide opportunities for treatment and use of DNA-PK as a novel cancer biomarker. In addition, several small molecules and biological agents have been recently identified that can inhibit DNA-PK function or expression, and thus hold promise for cancer treatments. This review discusses the impact of epigenetic alterations and the expression of DNA-PK in relation to the DNA repair mechanisms with a focus on its differential levels in normal and cancer cells.
Collapse
Affiliation(s)
- Vazhappilly Cijo George
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, Canada; Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Shabbir Ahmed Ansari
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Vipin Shankar Chelakkot
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | | | - Chaithanya Chelakkot
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Varsha Menon
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Wafaa Ramadan
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Raafat El-Awady
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates; Cancer Biology Department, National Cancer Institute and College of Medicine, Cairo University, Cairo, Egypt
| | - Theodora Mantso
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, Canada; Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Melina Mitsiogianni
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, Canada; Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Mihalis I Panagiotidis
- Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Graham Dellaire
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - H P Vasantha Rupasinghe
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, Canada; Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
13
|
Li J, Li H, Zhan D, Xiang M, Yang J, Zuo Y, Yu Y, Zhou H, Jiang D, Luo H, Chen Z, Yu Z, Xu Z. Niclosamide sensitizes nasopharyngeal carcinoma to radiation by downregulating Ku70/80 expression. J Cancer 2018; 9:736-744. [PMID: 29556331 PMCID: PMC5858495 DOI: 10.7150/jca.20963] [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: 05/10/2017] [Accepted: 12/13/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate whether niclosamide could sensitize the nasopharyngeal carcinoma cells to radiation and further explore the underlying mechanisms. CCK-8 assay was used to determine the effect of niclosamide on the proliferation of NPC cells. Colony formation assay was used to evaluate the radiosensitizing effect of niclosamide on NPC cells. Flow cytometry analysis was used to determine the apoptosis of NPC cells induced by niclosamide. Immunofluorescent staining was used to detect the formation of γ-H2AX foci and the localization of Ku70/80 proteins in NPC cells. Real-time PCR quantification analysis was used to examine the level of Ku70/80 mRNA. DNA damage repair-related proteins were detected by western blot analysis. Our results showed that niclosamide markedly suppressed the proliferation of NPC cells. Niclosamide pretreatment followed by irradiation reduced the colony forming ability of NPC cells. Niclosamide in combination with irradiation significantly increased the apoptotic rate of NPC cells. Niclosamide significantly reduced the transcriptional level of K70/80 but not the translocation of Ku70/80 protein induced by irradiation. In conclusion, our study demonstrated that niclosamide could inhibit the growth of NPC cells and sensitize the NPC cells to radiation via suppressing the transcription of Ku70/80.
Collapse
Affiliation(s)
- Jingjing Li
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Haiwen Li
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Dechao Zhan
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Mei Xiang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Jun Yang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Yufang Zuo
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Yin Yu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Hechao Zhou
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Danxian Jiang
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Haiqing Luo
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zihong Chen
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zhonghua Yu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zumin Xu
- Cancer Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| |
Collapse
|
14
|
Dong J, Ren Y, Zhang T, Wang Z, Ling CC, Li GC, He F, Wang C, Wen B. Inactivation of DNA-PK by knockdown DNA-PKcs or NU7441 impairs non-homologous end-joining of radiation-induced double strand break repair. Oncol Rep 2018; 39:912-920. [PMID: 29344644 PMCID: PMC5802037 DOI: 10.3892/or.2018.6217] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022] Open
Abstract
The DNA-dependent protein kinase (DNA-PK) complex plays a pivotal role in non-homologous end-joining (NHEJ) repair. We investigated the mechanism of NU7441, a highly selective DNA-PK inhibitor, in NHEJ-competent mouse embryonic fibroblast (MEF) cells and NHEJ-deficient cells and explored the feasibility of its application in radiosensitizing nasopharyngeal carcinoma (NPC) cells. We generated wild-type and DNA-PKcs−/− MEF cells. Clonogenic survival assays, flow cytometry, and immunoblotting were performed to study the effect of NU7441 on survival, cell cycle, and DNA repair. NU7441 profoundly radiosensitized wild-type MEF cells and SUNE-1 cells, but not DNA-PKcs−/− MEF cells. NU7441 significantly suppressed radiation-induced DSB repair post-irradiation through unrepaired and lethal DNA damage, the cell cycle arrest. The effect was associated with the activation of cell cycle checkpoints. The present study revealed a mechanism by which inhibition of DNA-PK sensitizes cells to irradiation suggesting that radiotherapy in combination with DNA-PK inhibitor is a promising paradigm for the management of NPC which merits further investigation.
Collapse
Affiliation(s)
- Jun Dong
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yufeng Ren
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Tian Zhang
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhenyu Wang
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Clifton C Ling
- Department of Medical Physics and Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Gloria C Li
- Department of Medical Physics and Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Fuqiu He
- Department of Medical Physics and Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Chengtao Wang
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bixiu Wen
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
15
|
Jin PY, Lu HJ, Tang Y, Fan SH, Zhang ZF, Wang Y, Li XN, Wu DM, Lu J, Zheng YL. Retracted: The effect of DNA-PKcs gene silencing on proliferation, migration, invasion and apoptosis, and in vivo tumorigenicity of human osteosarcoma MG-63 cells. Biomed Pharmacother 2017; 96:1324-1334. [PMID: 29203385 DOI: 10.1016/j.biopha.2017.11.079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/14/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to explore the role by which the DNA-dependent protein kinase complex catalytic subunit (DNA-PKcs) influences osteosarcoma MG-63 cell apoptosis, proliferation, migration and invasion. Osteosarcoma tissues and adjacent normal tissues were obtained from 57 osteosarcoma patients. Human osteosarcoma MG-63 cells were assigned into designated groups including the blank, siRNA-negative control (NC) and siRNA-DNA-PKcs groups. RT-qPCR and Western blotting methods were employed to evaluate the mRNA and protein expressions of DNA-PKcs. A cell counting kit-8 (CCK-8) assay was performed to assess cell viability. The evaluation of cell migration and invasion were conducted by means of Scratch test and Transwell assay. Flow cytometry with PI and annexin V/PI double staining was applied for the analysis of the cell cycle and apoptosis. Twenty-Four Balb/c nude mice were recruited and randomly divided into the blank, siRNA-NC and siRNA-DNA-PKcs groups. Tumorigenicity of the Balb/c nude mice was conducted to evaluate the rate of tumor formation, as well as for the assessment of tumor size and weight, and confirm the number of lung metastatic nodules in the mice post transfection. Osteosarcoma tissues were found to possess greater expression of DNA-PKcs than that of the adjacent normal tissues. DNA-PKcs expression in osteosarcoma tissues were correlated with the clinical stage and metastasis. Compared with the blank and siRNA-NC groups, proliferation, miration, as well as the invasion abilities of the MG-63 cells increased. Furthermore, an increase in apoptosis and cells at the G1 stage in the MG-63 cells was observed, while there were reductions in the cells detected at the S stage. The mRNA and protein expressions of CyclinD1, PCNA, Bcl-2 decreased while those of Bax increased in the siRNA-DNA-PKcs group. The tumor formation rate, tumor diameter, weight and lung metastatic nodules among the nude mice in the siRNA-DNA-PKcs group were all lower than those in the blank and siRNA-NC groups. The observations and findings of the study suggested that the silencing of DNA-PKcs inhibits the proliferation, migration and invasion, while acting to promote cell apoptosis in MG-63 cells and osteosarcoma growth in nude mice.
Collapse
Affiliation(s)
- Pei-Ying Jin
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Hong-Jie Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Yao Tang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China
| | - Yan Wang
- Department of Oncology, Beijing Hospital, Beijing 100730, PR China
| | - Xu-Ning Li
- Department of Oncology, Beijing Hospital, Beijing 100730, PR China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China.
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China.
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, PR China.
| |
Collapse
|
16
|
Abstract
Cellular chromosomal DNA is the principal target through which ionising radiation exerts it diverse biological effects. This chapter summarises the relevant DNA damage signalling and repair pathways used by normal and tumour cells in response to irradiation. Strategies for tumour radiosensitisation are reviewed which exploit tumour-specific DNA repair deficiencies or signalling pathway addictions, with a special focus on growth factor signalling, PARP, cancer stem cells, cell cycle checkpoints and DNA replication. This chapter concludes with a discussion of DNA repair-related candidate biomarkers of tumour response which are of crucial importance for implementing precision medicine in radiation oncology.
Collapse
|
17
|
Mesenchymal subtype of glioblastomas with high DNA-PKcs expression is associated with better response to radiotherapy and temozolomide. J Neurooncol 2017; 132:287-294. [DOI: 10.1007/s11060-016-2367-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/25/2016] [Indexed: 01/29/2023]
|
18
|
Estrada-Bernal A, Chatterjee M, Haque SJ, Yang L, Morgan MA, Kotian S, Morrell D, Chakravarti A, Williams TM. MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer cells involves inhibition of DNA double-strand break repair pathways. Cell Cycle 2016; 14:3713-24. [PMID: 26505547 DOI: 10.1080/15384101.2015.1104437] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Over 90% of pancreatic adenocarcinoma PC express oncogenic mutant KRAS that constitutively activates the Raf-MEK-MAPK pathway conferring resistance to both radiation and chemotherapy. MEK inhibitors have shown promising anti-tumor responses in recent preclinical and clinical studies, and are currently being tested in combination with radiation in clinical trials. Here, we have evaluated the radiosensitizing potential of a novel MEK1/2 inhibitor GSK1120212 (GSK212,or trametinib) and evaluated whether MEK1/2 inhibition alters DNA repair mechanisms in multiple PC cell lines. METHODS Radiosensitization and DNA double-strand break (DSB) repair were evaluated by clonogenic assays, comet assay, nuclear foci formation (γH2AX, DNA-PK, 53BP1, BRCA1, and RAD51), and by functional GFP-reporter assays for homologous recombination (HR) and non-homologous end-joining (NHEJ). Expression and activation of DNA repair proteins were measured by immunoblotting. RESULTS GSK212 blocked ERK1/2 activity and radiosensitized multiple KRAS mutant PC cell lines. Prolonged pre-treatment with GSK212 for 24-48 hours was required to observe significant radiosensitization. GSK212 treatment resulted in delayed resolution of DNA damage by comet assays and persistent γH2AX nuclear foci. GSK212 treatment also resulted in altered BRCA1, RAD51, DNA-PK, and 53BP1 nuclear foci appearance and resolution after radiation. Using functional reporters, GSK212 caused repression of both HR and NHEJ repair activity. Moreover, GSK212 suppressed the expression and activation of a number of DSB repair pathway intermediates including BRCA1, DNA-PK, RAD51, RRM2, and Chk-1. CONCLUSION GSK212 confers radiosensitization to KRAS-driven PC cells by suppressing major DNA-DSB repair pathways. These data provide support for the combination of MEK1/2 inhibition and radiation in the treatment of PC.
Collapse
Affiliation(s)
- Adriana Estrada-Bernal
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - Moumita Chatterjee
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - S Jaharul Haque
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - Linlin Yang
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - Meredith A Morgan
- b University of Michigan Comprehensive Cancer Center ; Ann Arbor , MI , USA
| | - Shweta Kotian
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - David Morrell
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - Arnab Chakravarti
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| | - Terence M Williams
- a The Ohio State University Medical Center; Arthur G James Comprehensive Cancer Center and Richard J Solove Research Institute ; Columbus , OH USA
| |
Collapse
|
19
|
Lu J, Tang M, Li H, Xu Z, Weng X, Li J, Yu X, Zhao L, Liu H, Hu Y, Tan Z, Yang L, Zhong M, Zhou J, Fan J, Bode AM, Yi W, Gao J, Sun L, Cao Y. EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma. Cancer Lett 2016; 380:191-200. [PMID: 27255972 DOI: 10.1016/j.canlet.2016.05.032] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 02/05/2023]
Abstract
We conducted this research to explore the role of latent membrane protein 1 (LMP1) encoded by the Epstein-Barr virus (EBV) in modulating the DNA damage response (DDR) and its regulatory mechanisms in radioresistance. Our results revealed that LMP1 repressed the repair of DNA double strand breaks (DSBs) by inhibiting DNA-dependent protein kinase (DNA-PK) phosphorylation and activity. Moreover, LMP1 reduced the phosphorylation of AMP-activated protein kinase (AMPK) and changed its subcellular location after irradiation, which appeared to occur through a disruption of the physical interaction between AMPK and DNA-PK. The decrease in AMPK activity was associated with LMP1-mediated glycolysis and resistance to apoptosis induced by irradiation. The reactivation of AMPK significantly promoted radiosensitivity both in vivo and in vitro. The AMPKα (Thr172) reduction was associated with a poorer clinical outcome of radiation therapy in NPC patients. Our data revealed a new mechanism of LMP1-mediated radioresistance and provided a mechanistic rationale in support of the use of AMPK activators for facilitating NPC radiotherapy.
Collapse
Affiliation(s)
- Jingchen Lu
- Department of Medical Oncology, Xiangya Hospital, Central South University, Changsha, China; Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Min Tang
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongde Li
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhijie Xu
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinxian Weng
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiangjiang Li
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinfang Yu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yongbin Hu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Zheqiong Tan
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lifang Yang
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China; Molecular Imaging Center, Central South University, Changsha, China
| | - Meizuo Zhong
- Department of Medical Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Zhou
- Key Laboratory of Chinese Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Key Laboratory of Chinese Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Wei Yi
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jinghe Gao
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lunquan Sun
- Molecular Imaging Center, Central South University, Changsha, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China; Molecular Imaging Center, Central South University, Changsha, China.
| |
Collapse
|
20
|
Molina S, Guerif S, Garcia A, Debiais C, Irani J, Fromont G. DNA-PKcs Expression Is a Predictor of Biochemical Recurrence After Permanent Iodine 125 Interstitial Brachytherapy for Prostate Cancer. Int J Radiat Oncol Biol Phys 2016; 95:965-972. [DOI: 10.1016/j.ijrobp.2016.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 01/27/2023]
|
21
|
Kim DH, Oh SY, Kim SY, Lee S, Koh MS, Lee JH, Lee S, Kim SH, Park HS, Hur WJ, Jeong JS, Ju MH, Seol YM, Choi YJ, Chung JS, Kim HJ. DNA ligase4 as a prognostic marker in nasopharyngeal cancer patients treated with radiotherapy. Asian Pac J Cancer Prev 2015; 15:10985-9. [PMID: 25605214 DOI: 10.7314/apjcp.2014.15.24.10985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The capability for DNA double-strand breaks (DSBs) repair is crucial for inherent radiosensitivity of tumor and normal cells. We have investigated the clinicopathologic significance of DNA repair gene expression in nasopharyngeal (NP) carcinoma. MATERIALS AND METHODS A total of 65 NP cancer patients who received radiotherapy were included. The immunopositivity to Ku 70, DNA-PKcs, MRN, RAD50, XRCC4, and LIG4 were examined in all tumor tissues. RESULTS The patients comprised 42 males and 23 females, with a median age of 56 years (range, 18-84). The expression levels of RAD50 (0,+1,+2,+3) were 27.7%, 32.3%, 21.5%, and 18.5%. LIG4 (±) were 43.1% and 56.9% respectively. The 5-year OS rate of patients with LIG4 (±) were 90% and 67.9%, respectively (p=0.035). The 5-year TTP rate of patients with LIG4 (±) were 75.9%, 55.5%, respectively (P=0.039). CONCLUSIONS Our results suggest the possibility of predicting the radiosensitivity of NP cancer by performing immunohistochemical analysis of LIG4.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea E-mail :
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Huang CY, Tsai CW, Hsu CM, Shih LC, Chang WS, Shui HA, Bau DT. The role of XRCC6/Ku70 in nasopharyngeal carcinoma. Int J Oral Maxillofac Surg 2015; 44:1480-5. [PMID: 26149939 DOI: 10.1016/j.ijom.2015.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 12/09/2022]
Abstract
The association between XRCC6/Ku70, an upstream player in the DNA double-strand break repair system, and the risk of nasopharyngeal carcinoma (NPC) was examined. In this case-control study, 176 NPC patients and 352 cancer-free controls were genotyped, and the associations of XRCC6 promoter T-991C (rs5751129), promoter G-57C (rs2267437), promoter G-31A (rs132770), and intron 3 (rs132774) polymorphisms with NPC risk were evaluated. NPC tissue samples were also assessed for their XRCC6 mRNA and protein expression by real-time quantitative reverse transcription PCR and Western blotting, respectively. With regard to the XRCC6 promoter T-991C, the TC and CC genotypes were associated with a significantly increased risk of NPC compared with wild-type TT genotype (adjusted odds ratio 2.02 and 3.42, 95% confidence interval 1.21-3.32 and 1.28-8.94, P=0.0072 and 0.0165, respectively). The mRNA and protein expression levels for NPC tissues revealed significantly lower XRCC6 mRNA and protein expression in the NPC samples with TC/CC genotypes compared to those with the TT genotype (P=0.0210 and 0.0164, respectively). These findings suggest that XRCC6 may play an important role in the carcinogenesis of NPC and could serve as a chemotherapeutic target for personalized medicine and therapy.
Collapse
Affiliation(s)
- C-Y Huang
- Graduate Institute of Medical Sciences, National Defence Medical Centre, Taipei, Taiwan, ROC; Taichung Armed Forces General Hospital, Taichung, Taiwan, ROC
| | - C-W Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, ROC; Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan, ROC
| | - C-M Hsu
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan, ROC
| | - L-C Shih
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan, ROC
| | - W-S Chang
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan, ROC; Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, ROC
| | - H-A Shui
- Graduate Institute of Medical Sciences, National Defence Medical Centre, Taipei, Taiwan, ROC
| | - D-T Bau
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, ROC; Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan, ROC; Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
23
|
Li X, Tian J, Bo Q, Li K, Wang H, Liu T, Li J. Targeting DNA-PKcs increased anticancer drug sensitivity by suppressing DNA damage repair in osteosarcoma cell line MG63. Tumour Biol 2015; 36:9365-72. [DOI: 10.1007/s13277-015-3642-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/03/2015] [Indexed: 11/24/2022] Open
|
24
|
Su M, Zhao L, Wei H, Lin R, Zhang X, Zou C. 18F-fluorodeoxyglucose positron emission tomography for predicting tumor response to radiochemotherapy in nasopharyngeal carcinoma. Strahlenther Onkol 2015; 191:642-8. [PMID: 25981635 DOI: 10.1007/s00066-015-0842-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/09/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of this study was to evaluate the value of (18)F-fluorodeoxyglucose (FDG) positron emission tomography (PET) in predicting tumor response to radiochemotherapy in nasopharyngeal carcinoma (NPC). MATERIALS AND METHODS From July 2012 to March 2014, 46 NPC patients who had undergone PET scanning before receiving definitive intensity-modulated radiotherapy (IMRT) treatment in our hospital were enrolled. Factors potentially affecting tumor response to treatment were studied by multiple logistic regression analysis. RESULTS After radiochemotherapy, 32 patients had a clinical complete response (CR), making the CR rate 69.6%. Multiple logistic regression analysis demonstrated that the maximal standard uptake value (SUV max) of the primary tumor was the only factor related to tumor response (p = 0.001), and that the logistic model had a high positive predictive value (90.6%). The area under the receiver operating characteristic (ROC) curve was 0.809, with a best cutoff threshold at 10.05. Patients with SUV max ≤ 10 had a higher CR rate than those with SUV max > 10 (p < 0.001). CONCLUSION The SUV max of the primary tumor before treatment is an independent predictor of tumor response in NPC.
Collapse
Affiliation(s)
- Meng Su
- Department of Radiation Oncology and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Street, 325000, Wenzhou, Zhejiang province, P.R. China
| | | | | | | | | | | |
Collapse
|
25
|
Gustafsson AS, Abramenkovs A, Stenerlöw B. Suppression of DNA-dependent protein kinase sensitize cells to radiation without affecting DSB repair. Mutat Res 2014; 769:1-10. [PMID: 25771720 DOI: 10.1016/j.mrfmmm.2014.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/04/2014] [Accepted: 06/16/2014] [Indexed: 06/04/2023]
Abstract
Efficient and correct repair of DNA double-strand break (DSB) is critical for cell survival. Defects in the DNA repair may lead to cell death, genomic instability and development of cancer. The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) is an essential component of the non-homologous end joining (NHEJ) which is the major DSB repair pathway in mammalian cells. In the present study, by using siRNA against DNA-PKcs in four human cell lines, we examined how low levels of DNA-PKcs affected cellular response to ionizing radiation. Decrease of DNA-PKcs levels by 80-95%, induced by siRNA treatment, lead to extreme radiosensitivity, similar to that seen in cells completely lacking DNA-PKcs and low levels of DNA-PKcs promoted cell accumulation in G2/M phase after irradiation and blocked progression of mitosis. Surprisingly, low levels of DNA-PKcs did not affect the repair capacity and the removal of 53BP1 or γ-H2AX foci and rejoining of DSB appeared normal. This was in strong contrast to cells completely lacking DNA-PKcs and cells treated with the DNA-PKcs inhibitor NU7441, in which DSB repair were severely compromised. This suggests that there are different mechanisms by which loss of DNA-PKcs functions can sensitize cells to ionizing radiation. Further, foci of phosphorylated DNA-PKcs (T2609 and S2056) co-localized with DSB and this was independent of the amount of DNA-PKcs but foci of DNA-PKcs was only seen in siRNA-treated cells. Our study emphasizes on the critical role of DNA-PKcs for maintaining survival after radiation exposure which is uncoupled from its essential function in DSB repair. This could have implications for the development of therapeutic strategies aiming to radiosensitize tumors by affecting the DNA-PKcs function.
Collapse
Affiliation(s)
- Ann-Sofie Gustafsson
- Section of Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Science, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85 Uppsala, Sweden.
| | - Andris Abramenkovs
- Section of Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Science, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85 Uppsala, Sweden
| | - Bo Stenerlöw
- Section of Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Science, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85 Uppsala, Sweden
| |
Collapse
|
26
|
Qian D, Zhang B, Zeng XL, Le Blanc JM, Guo YH, Xue C, Jiang C, Wang HH, Zhao TS, Meng MB, Zhao LJ, Hao JH, Wang P, Xie D, Lu B, Yuan ZY. Inhibition of human positive cofactor 4 radiosensitizes human esophageal squmaous cell carcinoma cells by suppressing XLF-mediated nonhomologous end joining. Cell Death Dis 2014; 5:e1461. [PMID: 25321468 PMCID: PMC4649520 DOI: 10.1038/cddis.2014.416] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 12/24/2022]
Abstract
Radiotherapy has the widest application to esophageal squamous cell carcinoma (ESCC) patients. Factors associated with DNA damage repair have been shown to function in cell radiosensitivity. Human positive cofactor 4 (PC4) has a role in nonhomologous end joining (NHEJ) and is involved in DNA damage repair. However, the clinical significance and biological role of PC4 in cancer progression and cancer cellular responses to chemoradiotherapy (CRT) remain largely unknown. The aim of the present study was to investigate the potential roles of PC4 in the radiosensitivity of ESCC. In this study, we showed that knockdown of PC4 substantially increased ESCC cell sensitivity to ionizing radiation (IR) both in vitro and in vivo and enhanced radiation-induced apoptosis and mitotic catastrophe (MC). Importantly, we demonstrated that silencing of PC4 suppressed NHEJ by downregulating the expression of XLF in ESCC cells, whereas reconstituting the expression of XLF protein in the PC4-knockdown ESCC cells restored NHEJ activity and radioresistance. Moreover, high expression of PC4 positively correlated with ESCC resistance to CRT and was an independent predictor for short disease-specific survival of ESCC patients in both of our cohorts. These findings suggest that PC4 protects ESCC cells from IR-induced death by enhancing the NHEJ-promoting activity of XLF and could be used as a novel radiosensitivity predictor and a promising therapeutic target for ESCCs.
Collapse
Affiliation(s)
- D Qian
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - B Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - X-L Zeng
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - J M Le Blanc
- Department of Radiation Oncology, Bodine Cancer Center, Thomas Jefferson University School of Medicine, Philadelphia, PA, USA
| | - Y-H Guo
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - C Xue
- Department of Radiation Oncology, Bodine Cancer Center, Thomas Jefferson University School of Medicine, Philadelphia, PA, USA
| | - C Jiang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - H-H Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - T-S Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - M-B Meng
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - L-J Zhao
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - J-H Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - P Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - D Xie
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - B Lu
- 1] Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China [2] Department of Radiation Oncology, Bodine Cancer Center, Thomas Jefferson University School of Medicine, Philadelphia, PA, USA
| | - Z-Y Yuan
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
27
|
Yang J, Fuller PJ, Morgan J, Shibata H, McDonnell DP, Clyne CD, Young MJ. Use of phage display to identify novel mineralocorticoid receptor-interacting proteins. Mol Endocrinol 2014; 28:1571-84. [PMID: 25000480 DOI: 10.1210/me.2014-1101] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The mineralocorticoid receptor (MR) plays a central role in salt and water homeostasis via the kidney; however, inappropriate activation of the MR in the heart can lead to heart failure. A selective MR modulator that antagonizes MR signaling in the heart but not the kidney would provide the cardiovascular protection of current MR antagonists but allow for normal electrolyte balance. The development of such a pharmaceutical requires an understanding of coregulators and their tissue-selective interactions with the MR, which is currently limited by the small repertoire of MR coregulators described in the literature. To identify potential novel MR coregulators, we used T7 phage display to screen tissue-selective cDNA libraries for MR-interacting proteins. Thirty MR binding peptides were identified, from which three were chosen for further characterization based on their nuclear localization and their interaction with other MR-interacting proteins or, in the case of x-ray repair cross-complementing protein 6, its known status as an androgen receptor coregulator. Eukaryotic elongation factor 1A1, structure-specific recognition protein 1, and x-ray repair cross-complementing protein 6 modulated MR-mediated transcription in a ligand-, cell- and/or promoter-specific manner and colocalized with the MR upon agonist treatment when imaged using immunofluorescence microscopy. These results highlight the utility of phage display for rapid and sensitive screening of MR binding proteins and suggest that eukaryotic elongation factor 1A1, structure-specific recognition protein 1, and x-ray repair cross-complementing protein 6 may be potential MR coactivators whose activity is dependent on the ligand, cellular context, and target gene promoter.
Collapse
Affiliation(s)
- Jun Yang
- MIMR-PHI Medical Research Institute (J.Y., P.J.F., J.M., C.D.C., M.J.Y.), Department of Medicine (J.Y., P.J.F., M.J.Y.), Monash University, Clayton, Victoria 3168, Australia; Department of Endocrinology, Metabolism, Rheumatology, and Nephrology (H.S.), Oita University, Yufu 879-5593, Japan; and Department of Pharmacology and Cancer Biology (D.P.M.), Duke University Medical Center, Durham, North Carolina 27710
| | | | | | | | | | | | | |
Collapse
|
28
|
The potential role of Ku80 in primary central nervous system lymphoma as a prognostic factor. Contemp Oncol (Pozn) 2013; 17:58-63. [PMID: 23788963 PMCID: PMC3685349 DOI: 10.5114/wo.2013.33775] [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: 02/06/2012] [Revised: 08/24/2012] [Accepted: 11/14/2012] [Indexed: 01/15/2023] Open
Abstract
The aim of our study was to detect the expression of Ku80 in primary central nervous system lymphoma and to evaluate the relationship between Ku80 expression level and clinical outcomes. Thirty-eight patients with primary central nervous system lymphoma (PCNSL) were included in this retrospective study. The expression of Ku80 in tumor samples was determined by immunohistochemistry. One thousand neoplastic cells per specimen were counted. The expression levels were compared with the clinical data and statistically analyzed. The results of this study show that the expression of Ku80 can be found in the majority of PCNSLs. The mean expression level of Ku80 in 38 PCNSL is 64.1 ±24.5. A significant difference in Ku80 expression could be found between the age < 65 years group and age ≥ 65 years group (P = 0.006). Kaplan-Meier analysis revealed that patients who showed a high Ku80 expression had a significantly shorter median survival time (MST) than patients who had low Ku80 expression (P = 0.036). Patients’ age, tumor location, and treatment protocol were significantly related to prognosis in PCNSL (P < 0.05). The expression of Ku80 was observed in the majority of PCNSLs. Ku80 was a predictive factor for survival in this study. In addition to Ku80, other clinical variables including age, tumor location and therapeutic protocol are correlated significantly with overall survival.
Collapse
|
29
|
Hsu FM, Zhang S, Chen BPC. Role of DNA-dependent protein kinase catalytic subunit in cancer development and treatment. Transl Cancer Res 2012; 1:22-34. [PMID: 22943041 DOI: 10.3978/j.issn.2218-676x.2012.04.01] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a key component of the non-homologous end-joining (NHEJ) pathway, is involved in DNA double-strand break repair, immunocompetence, genomic integrity, and epidermal growth factor receptor signaling. Clinical studies indicate that expression and activity of DNA-PKcs is correlated with cancer progression and response to treatment. Various anti-DNA-PKcs strategies have been developed and tested in preclinical studies to exploit the benefit of DNA-PKcs inhibition in sensitization of radiotherapy and in combined modality therapy with other antitumor agents. In this article, we review the association between DNA-PKcs and cancer development and discuss current approaches and mechanisms for inhibition of DNA-PKcs. The future challenges are to understand how DNA-PKcs activity is correlated with cancer susceptibility and to identify those patients who would most benefit from DNA-PKcs inhibition.
Collapse
Affiliation(s)
- Feng-Ming Hsu
- Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | |
Collapse
|
30
|
Wang W, Pan X, Huo X, Yan F, Wang M, Wang D, Gao Y, Cao Q, Luo D, Qin C, Yin C, Zhang Z. A functional polymorphism C-1310G in the promoter region of Ku70/XRCC6 is associated with risk of renal cell carcinoma. Mol Carcinog 2012; 51 Suppl 1:E183-90. [PMID: 22593040 DOI: 10.1002/mc.21914] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 01/13/2012] [Accepted: 03/19/2012] [Indexed: 11/11/2022]
Abstract
The DNA repair gene Ku70 plays a key role in the DNA double strand break (DSB) repair system. Defects in DSB repair capacity can lead to genomic instability. We hypothesized that the Ku70 C-1310G polymorphism (rs2267437) was associated with risk of renal cell carcinoma (RCC). We genotyped the Ku70 C-1310G polymorphism in a case-control study of 620 patients and 623 controls in a Chinese population and assessed the effects of C-1310G polymorphism on RCC susceptibility and survival. We then examined the functionality of this polymorphism. Compared with the Ku70-1310CC genotype, the CG and CG/GG genotypes had a significantly increased risk of RCC [adjusted odds ratio (OR) = 1.47, 95% confidence interval (CI) = 1.16-1.87 for CG and OR = 1.47, 95% CI = 1.16-1.86 for CG/GG]. However, the C-1310G polymorphism did not influence the survival of RCC. The in vivo experiments with normal renal tissues revealed statistically significantly lower Ku70 mRNA expression in samples with CG/GG genotypes relative to those with the CC genotype (P < 0.05). In vitro luciferase assays in various cell lines showed lower luciferase activity for the -1310G allele than for the -1310C allele. These results suggest that the Ku70 C-1310G polymorphism is involved in the etiology of RCC and thus may be a marker for genetic susceptibility to RCC in Chinese populations. Larger studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Nanjing Medical University, Yizheng, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bouchaert P, Guerif S, Debiais C, Irani J, Fromont G. DNA-PKcs expression predicts response to radiotherapy in prostate cancer. Int J Radiat Oncol Biol Phys 2012; 84:1179-85. [PMID: 22494583 DOI: 10.1016/j.ijrobp.2012.02.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 12/24/2022]
Abstract
PURPOSE Double-strand breaks, the most lethal DNA lesions induced by ionizing radiation, are mainly repaired by the nonhomologous end-joining system. The expression of the nonhomologous end-joining pathway has never been studied in prostate cancer, and its prognostic value for patients undergoing radiotherapy remains unknown. METHODS Pretreatment biopsies from 238 patients treated with exclusive external beam radiotherapy for localized prostate cancer with ≥ 2 years of follow-up were reviewed to reassess the Gleason score. Of these 238 cases, 179 were suitable for in situ analysis and were included in the tissue microarrays. Expression of the nonhomologous end-joining proteins Ku70, Ku80, DNA-dependent protein kinase, catalytic subunits (DNA-PKcs), and X-ray repair cross complementing 4-like factor was studied by immunohistochemistry, together with the proliferation marker Ki67. RESULTS The predictive value of the Gleason score for biochemical relapse (using the Phoenix criteria) was markedly improved after review (P<.0001) compared with the initial score (P=.003). The clinical stage, pretreatment prostate-specific antigen level, and perineural invasion status were also associated with progression-free survival (P=.005, P<.0001, and P=.03, respectively). High proliferation (>4%) tends to be associated with biochemical recurrence; however, the difference did not reach statistical significance (P=.06). Although the expression of Ku70, Ku80, and X-ray repair cross complementing 4-like factor was not predictive of relapse, positive DNA-PKcs nuclear staining was closely associated with biochemical recurrence (P=.0002). On multivariate analysis, only the Gleason score, prostate-specific antigen level, and DNA-PKcs status remained predictive of recurrence (P=.003, P=.002, and P=.01, respectively). CONCLUSIONS The results of the present study highly suggest that DNA-PKcs could be a predictive marker of recurrence after radiotherapy, independently of the classic prognostic markers, including the Gleason score modified after review.
Collapse
Affiliation(s)
- Patrick Bouchaert
- Department of Pathology, CHU-Universite de Poitiers, Poitiers, France
| | | | | | | | | |
Collapse
|
32
|
Azad A, Jackson S, Cullinane C, Natoli A, Neilsen PM, Callen DF, Maira SM, Hackl W, McArthur GA, Solomon B. Inhibition of DNA-dependent protein kinase induces accelerated senescence in irradiated human cancer cells. Mol Cancer Res 2011; 9:1696-707. [PMID: 22009179 DOI: 10.1158/1541-7786.mcr-11-0312] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
DNA-dependent protein kinase (DNA-PK) plays a pivotal role in the repair of DNA double-strand breaks (DSB) and is centrally involved in regulating cellular radiosensitivity. Here, we identify DNA-PK as a key therapeutic target for augmenting accelerated senescence in irradiated human cancer cells. We find that BEZ235, a novel inhibitor of DNA-PK and phosphoinositide 3-kinase (PI3K)/mTOR, abrogates radiation-induced DSB repair resulting in cellular radiosensitization and growth delay of irradiated tumor xenografts. Importantly, radiation enhancement by BEZ235 coincides with a prominent p53-dependent accelerated senescence phenotype characterized by positive β-galactosidase staining, G(2)-M cell-cycle arrest, enlarged and flattened cellular morphology, and increased p21 expression and senescence-associated cytokine secretion. Because this senescence response to BEZ235 is accompanied by unrepaired DNA DSBs, we examined whether selective targeting of DNA-PK also induces accelerated senescence in irradiated cells. Significantly, we show that specific pharmacologic inhibition of DNA-PK, but not PI3K or mTORC1, delays DSB repair leading to accelerated senescence after radiation. We additionally show that PRKDC knockdown using siRNA promotes a striking accelerated senescence phenotype in irradiated cells comparable with that of BEZ235. Thus, in the context of radiation treatment, our data indicate that inhibition of DNA-PK is sufficient for the induction of accelerated senescence. These results validate DNA-PK as an important therapeutic target in irradiated cancer cells and establish accelerated senescence as a novel mechanism of radiosensitization induced by DNA-PK blockade.
Collapse
Affiliation(s)
- Arun Azad
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Xie P, Li M, Zhao H, Sun X, Fu Z, Yu J. 18F-FDG PET or PET-CT to evaluate prognosis for head and neck cancer: a meta-analysis. J Cancer Res Clin Oncol 2011; 137:1085-93. [PMID: 21229262 DOI: 10.1007/s00432-010-0972-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 12/22/2010] [Indexed: 12/22/2022]
Abstract
PURPOSE The purpose of this meta-analysis was to evaluate the prognostic value of standard uptake value (SUV) from serial Fluorine-18-fluorodeoxyglucose positron emission tomography/computed tomography ((18)F-FDG PET/CT) in patients with head and neck cancer. METHODS We searched for articles limited to head and neck cancer, dealt with the impact of SUV on survival and published in English. The endpoints were disease-free survival (DFS), overall survival (OS), and local control (LC). Two reviewers extracted data independently. RESULTS Thirty-five studies were identified; of which, 26 studies involving 1,415 patients met the inclusion criteria. Pooled survival data suggested better DFS, OS, and LC in patients with low SUV of pre-treatment, and the odds ratio (OR) was 0.23, 0.24, and 0.27, respectively. Patients having tumors with low SUV of post-treatment also had significantly better DFS (OR = 0.17) and OS (OR = 0.28) than those with high SUV. CONCLUSIONS The present meta-analysis showed that (18)F-FDG uptake, as measured by the SUV before treatment and metabolic response after treatment, are valuable for predicting long-term survival in head and neck cancer. High (18)F-FDG uptake may be useful for identifying patients requiring more aggressive treatment.
Collapse
Affiliation(s)
- Peng Xie
- Department of Radiation Oncology, Shandong Tumor Hospital and Institute, Key Laboratory of Radiation Oncology of Shandong Province, Jiyan Road 440, 250117, Jinan, Shandong Province, China
| | | | | | | | | | | |
Collapse
|
34
|
Moeller BJ, Yordy JS, Williams MD, Giri U, Raju U, Molkentine DP, Byers LA, Heymach JV, Story MD, Lee JJ, Sturgis EM, Weber RS, Garden AS, Ang KK, Schwartz DL. DNA repair biomarker profiling of head and neck cancer: Ku80 expression predicts locoregional failure and death following radiotherapy. Clin Cancer Res 2011; 17:2035-43. [PMID: 21349997 DOI: 10.1158/1078-0432.ccr-10-2641] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE Radiotherapy plays an integral role in the treatment of head and neck squamous cell carcinoma (HNSCC). Although proteins involved in DNA repair may predict HNSCC response to radiotherapy, none has been validated in this context. We examined whether differential expression of double-strand DNA break (DSB) repair proteins in HNSCC, the chief mediators of DNA repair following irradiation, predict for treatment outcomes. EXPERIMENTAL DESIGN Archival HNSCC tumor specimens (n = 89) were assembled onto a tissue microarray and stained with antibodies raised against 38 biomarkers. The biomarker set was enriched for proteins involved in DSB repair, in addition to established mechanistic markers of radioresistance. Staining was correlated with treatment response and survival alongside established clinical and pathologic covariates. Results were validated in an independent intramural cohort (n = 34). RESULTS Ku80, a key mediator of DSB repair, correlated most closely with clinical outcomes. Ku80 was overexpressed in half of all tumors, and its expression was independent of all other covariates examined. Ku80 overexpression was an independent predictor for both locoregional failure and mortality following radiotherapy (P < 0.01). The predictive power of Ku80 overexpression was confined largely to HPV-negative HNSCC, where it conferred a nine-fold greater risk of death at two years. CONCLUSIONS Ku80 overexpression is a common feature of HNSCC, and is a candidate DNA repair-related biomarker for radiation treatment failure and death, particularly in patients with high-risk HPV-negative disease. It is a promising, mechanistically rational biomarker to select individual HPV-negative HNSCC patients for strategies to intensify treatment.
Collapse
Affiliation(s)
- Benjamin J Moeller
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Canazza A, De Grazia U, Fumagalli L, Brait L, Ghielmetti F, Fariselli L, Croci D, Salmaggi A, Ciusani E. In vitro effects of Cyberknife-driven intermittent irradiation on glioblastoma cell lines. Neurol Sci 2011; 32:579-88. [PMID: 21301910 DOI: 10.1007/s10072-011-0485-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 01/22/2011] [Indexed: 01/23/2023]
Abstract
Radiosurgery is used increasingly upon recurrence of high-grade gliomas to deliver a high dose of focused radiation to a defined target. The purpose of our study was to compare intermittent irradiation (IIR) by using a CyberKnife (CK) with continuous irradiation (CIR) by using a conventional linear accelerator (LINAC). A significant decrease in surviving fraction was observed after IIR irradiation compared with after CIR at a dose of 8 Gy. Three hours after irradiation, most of the DNA damage was repaired in U87. Slightly higher basal levels of Ku70/80 mRNA were found in U87 compared with A172, while radiation treatment induced only minor regulation of Ku70/80 and Rad51 transcription in either cell lines. IIR treatment using CK significantly decreased the survival in U87 and A172 compared with CIR. Although the two cell lines differed in DNA repair capability, the role of Ku70/80 and Rad51 in the cell line radiosensitivity seemed marginal.
Collapse
Affiliation(s)
- Alessandra Canazza
- Laboratory of Clinical Investigation, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Xie P, Yue JB, Zhao HX, Sun XD, Kong L, Fu Z, Yu JM. Prognostic value of 18F-FDG PET-CT metabolic index for nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2010; 136:883-9. [PMID: 19936788 DOI: 10.1007/s00432-009-0729-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 11/05/2009] [Indexed: 01/18/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the prognostic value of metabolic tumor volume (MTV) and metabolic index (MI) from fluorine-18-fluorodeoxyglucose positron emission tomography/computed tomography ((18)F-FDG PET/CT) in patients with nasopharyngeal carcinoma (NPC). METHODS From October 2002 to July 2004, 41 patients with NPC who underwent (18)F-FDG PET-CT scan before and after radiotherapy were reviewed retrospectively. All patients received intensity-modulated radiotherapy using 6MV X-rays. We examined the association of MTV and the results of long-term follow-up of the patients. RESULTS Patients having tumors with an MTV below 30 cm(3) had significantly better 5-year overall survival (OS) (84.6:46.7%, P = 0.006) and disease-free survival (DFS) (73.1:40.0%, P = 0.014) than patients with an MTV of 30 cm(3) or greater. And the patients with MI below 130 had significantly higher 5-year OS (88.0:43.8%, P = 0.002) and DFS (76.0:37.5%, P = 0.005) than other patients. In the Cox multivariate analysis, MI and metabolic response (MR) were predictive of DFS, and we did not find a significant relationship between standard uptake value (SUV) and OS or DFS. CONCLUSIONS The present study shows that tumor volume parameters, especially the combination of MTV and SUV in the "metabolic index", are valuable for predicting long-term survival. High MI may be useful for identifying patients requiring more aggressive treatment.
Collapse
Affiliation(s)
- Peng Xie
- Department of Radiation Oncology, Shandong Tumor Hospital and Institute, Jiyan Road 440, 250117 Jinan, Shandong, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Xie P, Yue JB, Fu Z, Feng R, Yu JM. Prognostic value of 18F-FDG PET/CT before and after radiotherapy for locally advanced nasopharyngeal carcinoma. Ann Oncol 2010; 21:1078-82. [DOI: 10.1093/annonc/mdp430] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
38
|
Willems P, De Ruyck K, Van den Broecke R, Makar A, Perletti G, Thierens H, Vral A. A polymorphism in the promoter region of Ku70/XRCC6, associated with breast cancer risk and oestrogen exposure. J Cancer Res Clin Oncol 2009; 135:1159-68. [DOI: 10.1007/s00432-009-0556-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 01/26/2009] [Indexed: 01/09/2023]
|
39
|
Pavón MA, Parreño M, León X, Sancho FJ, Céspedes MV, Casanova I, Lopez-Pousa A, Mangues MA, Quer M, Barnadas A, Mangues R. Ku70 predicts response and primary tumor recurrence after therapy in locally advanced head and neck cancer. Int J Cancer 2008; 123:1068-79. [PMID: 18546291 DOI: 10.1002/ijc.23635] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
5-Fluorouracil and cisplatin-based induction chemotherapy (IC) is commonly used to treat locally advanced head and neck squamous cell carcinoma (HNSCC). The role of nonhomologous end joining (NHEJ) genes (Ku70, Ku80 and DNA-PKcs) in double-strand break (DSB) repair, genomic instability and apoptosis suggest a possible impact on tumor response to radiotherapy, 5-fluorouracil or cisplatin, as these agents are direct or indirect inductors of DSBs. We evaluated the relationship between Ku80, Ku70 or DNA PKcs mRNA expression in pretreatment tumor biopsies, and tumor response to IC or local recurrence, in 50 patients with HNSCC. Additionally, in an independent cohort of 75 patients with HNSCC, we evaluated the relationship between tumor Ku70 protein expression and the same clinical outcomes or patient survival. Tumors in the responder group had significantly higher mRNA levels for Ku70, Ku80 and DNA-PKcs than those in the nonresponder group. Ku70 mRNA was the marker most significantly associated with response to IC. Moreover, high tumor Ku70 mRNA expression was associated with significantly longer local recurrence-free survival (LRFS). Ku70 protein expression was also significantly related to response, and patients with higher percentage of tumor cells expressing Ku70 had longer LRFS. In addition, the percentage of Ku70 positive cells, tumor localization and node involvement were significantly associated with overall survival of patient. Therefore, Ku70 expression is a candidate predictive marker that could distinguish patients who are likely to benefit from chemoradiotherapy or radiotherapy after the induction chemotherapy treatment, suggesting a contribution of the NHEJ system in HNSCC clinical outcome.
Collapse
Affiliation(s)
- Miguel Angel Pavón
- Grup d'Oncogènesi i Antitumorals, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER) and Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Xing J, Wu X, Vaporciyan AA, Spitz MR, Gu J. Prognostic significance of ataxia-telangiectasia mutated, DNA-dependent protein kinase catalytic subunit, and Ku heterodimeric regulatory complex 86-kD subunit expression in patients with nonsmall cell lung cancer. Cancer 2008; 112:2756-64. [PMID: 18457328 DOI: 10.1002/cncr.23533] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The double-strand break (DSB) repair capacity has been implicated in the survival of patients in several cancer types. However, little is known about the prognostic importance of the key DSB repair genes-ataxia-telangiectasia mutated (ATM), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), and the Ku heterodimeric regulatory complex 86-kD subunit (Ku80)-in nonsmall cell lung cancer (NSCLC). To address this issue, the authors determined the messenger RNA (mRNA) expression of these genes in patients NSCLC and assessed their prognostic relevance. METHODS mRNA expression levels of ATM, DNA-PKcs, and Ku80 were measured in tumor and adjacent normal tissues from 140 patients with NSCLC by using quantitative real-time polymerase chain reaction analysis. Then, a Cox proportional hazards regression model and Kaplan-Meier plots were used to evaluate the association between the tumor:normal (T/N) expression ratios of the 3 genes and the overall survival rate and duration in patients with NSCLC. RESULTS mRNA expression of ATM and DNA-PKcs, but not of Ku80, was significantly higher in tumor tissues than in adjacent normal tissues (P=.003 and P<.001, respectively). The high T/N expression ratios of ATM and DNA-PKcs were associated significantly with a 1.82-fold increased risk of death (95% confidence interval, 1.05-2.70) and a 2.13-fold increased risk of death (95% confidence interval, 1.21-3.76), respectively. However, no significant association with risk was observed for Ku80. Kaplan-Meier analyses revealed that patients with high T/N expression ratios of ATM or DNA-PKcs had notably shorter median survival than patients with low ratios. CONCLUSIONS The current findings suggested that the T/N expression ratios of ATM and DNA-PKcs may be useful for identifying NSCLC patients with a poor prognosis who may benefit from more aggressive therapy.
Collapse
Affiliation(s)
- Jinliang Xing
- Department of Epidemiology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Lee SW, Nam SY, Im KC, Kim JS, Choi EK, Ahn SD, Park SH, Kim SY, Lee BJ, Kim JH. Prediction of prognosis using standardized uptake value of 2-[18F] fluoro-2-deoxy-d-glucose positron emission tomography for nasopharyngeal carcinomas. Radiother Oncol 2008; 87:211-6. [DOI: 10.1016/j.radonc.2008.01.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 12/07/2007] [Accepted: 01/12/2008] [Indexed: 10/22/2022]
|
42
|
Kasten-Pisula U, Vronskaja S, Overgaard J, Dikomey E. In normal human fibroblasts variation in DSB repair capacity cannot be ascribed to radiation-induced changes in the localisation, expression or activity of major NHEJ proteins. Radiother Oncol 2008; 86:321-8. [PMID: 18158193 DOI: 10.1016/j.radonc.2007.11.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Accepted: 11/30/2007] [Indexed: 10/22/2022]
|
43
|
Chang HW, Kim SY, Yi SL, Son SH, Song DY, Moon SY, Kim JH, Choi EK, Ahn SD, Shin SS, Lee KK, Lee SW. Expression of Ku80 correlates with sensitivities to radiation in cancer cell lines of the head and neck. Oral Oncol 2006; 42:979-86. [PMID: 16472552 DOI: 10.1016/j.oraloncology.2005.12.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Accepted: 12/08/2005] [Indexed: 01/03/2023]
Abstract
The Ku protein is essential for the repair of a majority of DNA double-strand breaks in mammalian cells. The purpose of this study was to investigate the relationship between the expression of Ku70/80 and sensitivity to radiation in cancer cell lines of the head and neck. The sensitivity to radiation in various head and neck cancer cell lines (AMC-HN-1 to -9) was analyzed by colony forming assay. Of the nine cell lines examined, the most radiosensitive cell line (AMC-HN-3) and the most radioresistant cell line (AMC-HN-9) were selected for this experiments. The expression of Ku70/80 was examined after irradiation using real time PCR, Western blotting and immunofluorescence in two different cell lines. Cell cycle distribution after irradiation were analysed. A differential radioresponse was demonstrated by expression of Ku70/80 in AMC-HN-3 and AMC-HN-9 cells. While the expression of Ku70 was slightly increased in the radioresistant AMC-HN-9 cell line, the expression of Ku80 was remarkably increased, suggesting a correlation between Ku80 expression and radiation resistance. Overexpression of Ku80 plays an important role in the repair of DNA damage induced by radiation. Ku80 expression may provide an effective predictive assay of radiosensitivity in head and neck cancers.
Collapse
Affiliation(s)
- Hyo Won Chang
- Department of Otolaryngology, University of Ulsan, College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Davis JE, Smith MC, Coman WB, Moss DJ. Epstein–Barr virus: the future for screening, treatment and monitoring of nasopharyngeal carcinoma. Future Virol 2006. [DOI: 10.2217/17460794.1.2.211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is often not diagnosed until an advanced stage of the disease, and has a poor 5-year survival with current therapies. Thus, screening programs to identify high-risk patients at early disease stages are essential to improve patient outcomes, most likely through using Epstein–Barr virus (EBV) DNA monitoring in conjunction with tumor-specific markers. EBV-specific cytotoxic T lymphocytes (CTLs) have been utilized successfully for long-term regression of EBV-associated B-cell lymphomas, such as post-transplant lymphoproliferative disease. This strategy has recently been adapted to raise latent membrane proteins 1 and 2, and EBV nuclear antigen 1-specific CD8+ and CD4+ T cells to target EBV proteins expressed in NPC tumors. Future challenges will be focused on developing multiple-target therapies, including improving CTL persistence and tumor specificity. Understanding the role of EBV infection and protein expression in NPC will be pivotal in the development of screening protocols and novel treatments, including vaccines.
Collapse
Affiliation(s)
| | | | | | - Denis J Moss
- The Queensland Institute of Medical Research, The EBV Biology Laboratory, PO Box Royal Brisbane Hospital, Brisbane 4006, Queensland, Australia
- The Princess Alexandra Hospital, The Head and Neck Clinic, Woolloongabba, Brisbane, Queensland, Australia and, The Queensland Institute of Medical Research, The EBV Biology Laboratory, PO Box Royal Brisbane Hospital, Brisbane 4006, Queensland, Australia
- The Princess Alexandra Hospital, The Head and Neck Clinic, Woolloongabba, Brisbane, Queensland, Australia
- The Queensland Institute of Medical Research, The EBV Biology Laboratory, PO Box Royal Brisbane Hospital, Brisbane 4006, Queensland, Australia
| |
Collapse
|