1
|
Zhang Y, Shen G, Zhang D, Meng T, Lv Z, Chen L, Li J, Li K. N 6-Methyladenosine modification mediated by METTL3 promotes DNA-PKcs expression to induce anlotinib resistance in osteosarcoma. Clin Transl Med 2025; 15:e70228. [PMID: 39924638 PMCID: PMC11807765 DOI: 10.1002/ctm2.70228] [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: 09/18/2024] [Revised: 01/21/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Acquired anlotinib resistance is still a key challenge in osteosarcoma treatment. Unravelling the mechanisms underlying anlotinib resistance is the key to optimising its efficacy for treating osteosarcoma. Previous studies have explored the pivotal function of the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) with regard to osteosarcoma chemoresistance. METHODS We used bioinformatics analysis to predict DNA-PKcs and Beclin-1 interactions, confirmed through immunofluorescence (IF) and co-immunoprecipitation (co-IP). Dual-luciferase analyses and Methylated RNA immunoprecipitation (MeRIP) were implemented to detect the detected m6A modifications. RNA fluorescence in situ hybridisation (FISH)-IF co-localisation and RNA immunoprecipitation (RIP) were conducted to explore the interplay between PRKDC mRNA and the indicated proteins. RESULTS Anlotinib-treated osteosarcoma cells exhibited increased DNA-PKcs levels, and silencing DNA-PKcs augmented osteosarcoma sensitivity to anlotinib. DNA-PKcs affects anlotinib-induced autophagy by interacting with Beclin-1 and regulating its ubiquitination. Notably, PRKDC mRNA, encoding DNA-PKcs, underwent N6-Methyladenosine (m6A) modification. Methyltransferase-like 3 (METTL3) positively regulated DNA-PKcs expression. Functionally, METTL3 enhances anlotinib resistance in osteosarcoma, which is reversed by PRKDC knockdown. Mechanistically, METTL3 binds to PRKDC mRNA and facilitates m6A methylation. Additionally, m6A methylated PRKDC mRNA is identified via YTH N6-methyladenosine RNA-binding protein 1 (YTHDF1), augmenting its expression. CONCLUSION These findings revealed that DNA-PKcs promotes anlotinib resistance by regulating protective autophagy, while METTL3 induces PRKDC m6A modification, enhancing its expression. Thus, targeting METTL3/PRKDC may be a novel strategy for improving therapeutic efficacy in human osteosarcoma. KEY POINTS DNA-PKcs knockdown heightens osteosarcoma sensitivity to anlotinib. DNA-PKcs modulates anlotinib-induced protective autophagy through interacts with Beclin-1 and regulates its ubiquitination. m6A modification of OLE_LINK82PRKDC mRNA induced by METTL3 contributes to anlotinib resistance in osteosarcoma. m6A methylation of PRKDC mRNA recognised by YTHDF1 amplifies the expression of DNA-PKcs.
Collapse
Affiliation(s)
- Yining Zhang
- The First Clinical College of Cheeloo College of MedicineShandong UniversityJinanShandongChina
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandongChina
| | - Guohong Shen
- Department of PediatricsCentral Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Dan Zhang
- Department of Cardiovascular MedicineJinan Central HospitalJinanShandongChina
| | - Tingting Meng
- Department of Cardiovascular MedicineJinan Central HospitalJinanShandongChina
- Research Center of Translational MedicineCentral Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Zhaorui Lv
- Department of OrthopedicsAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Lei Chen
- The First Clinical College of Cheeloo College of MedicineShandong UniversityJinanShandongChina
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandongChina
| | - Jianmin Li
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandongChina
| | - Ka Li
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
2
|
Zhang Y, Shen G, Meng T, Lv Z, Li X, Li J, Li K. Eicosapentaenoic acid enhances the sensitivity of osteosarcoma to cisplatin by inducing ferroptosis through the DNA-PKcs/AKT/NRF2 pathway and reducing PD-L1 expression to attenuate immune evasion. Int Immunopharmacol 2023; 125:111181. [PMID: 37951196 DOI: 10.1016/j.intimp.2023.111181] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/13/2023]
Abstract
Acquired drug resistance poses a significant challenge in osteosarcoma therapy. Therefore, it is necessary for us to discover and develop an alternative anti-cancer strategy. Previous studies have shown that eicosapentaenoic acid (EPA) significantly increases chemosensitivity in cancer cells. In this study, we discovered that EPA enhances the sensitivity of osteosarcoma to cisplatin (DDP). Interestingly, in addition to inhibiting growth and inducing apoptosis, EPA also enhances DDP-induced ferroptosis. Western blot analysis confirmed that EPA treatment significantly decreases the expression of DNA-dependent protein kinase catalytic subunit (DNA-PKcs), p-AKT, nuclear factor erythroid 2-related factor 2 (NRF2), and glutathione peroxidase 4 (GPX4) in cells. Knockdown of DNA-PKcs by siRNA further enhances the level of ferroptosis induced by EPA. Importantly, EPA can reverse the high expression level of programmed death ligand 1 (PD-L1) induced by DDP. ELISA and western blotting analysis revealed that EPA treatment decreases the levels of IL-6 and p-STAT3, which are increased by DDP treatment. Furthermore, a co-immunoprecipitation (co-IP) assay confirmed the interaction between DNA-PKcs and PD-L1, and knockdown of DNA-PKcs further reduces the expression of PD-L1. This data provides the first evidence that EPA suppresses the DNA-PKcs/AKT/NRF2/GPX4 pathway to enhance ferroptosis, and inhibits IL-6/STAT3 and DNA-PKcs to decrease PD-L1 expression, thereby sensitizing osteosarcoma to DDP. The combination of EPA and DDP presents an encouraging and promising anti-tumor strategy.
Collapse
Affiliation(s)
- Yining Zhang
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Guohong Shen
- Department of Pediatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Tingting Meng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Zhaorui Lv
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Xin Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Jianmin Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Ka Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China.
| |
Collapse
|
3
|
Li G, Yan X. Long non-coding RNA GAS5 promotes cisplatin-chemosensitivity of osteosarcoma cells via microRNA-26b-5p/TP53INP1 axis. J Orthop Surg Res 2023; 18:890. [PMID: 37993867 PMCID: PMC10666340 DOI: 10.1186/s13018-023-04387-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
Osteosarcoma is a common malignant bone tumor. Cisplatin (DDP) achieves a high response rate in osteosarcoma. Here we aim to study the dysregulation of long non-coding RNA the growth arrest-specific transcript 5 (GAS5), and its roles in DDP-resistance of osteosarcoma. The expression of mRNA and microRNA in osteosarcoma tissues and osteosarcoma cell lines were detected by quantitative reverse-transcription polymerase chain reaction, and protein expression levels were measured by western blotting assay. Cell Counting Kit-8 and 5-Ethynyl-2'-deoxyuridine were used to measure cell proliferation. Flow cytometer assay was used to evaluate cell apoptosis. The interactions between miR-26b-5p and GAS5 or tumor protein p53-induced nuclear protein 1 (TP53INP1) were verified by dual luciferase reporter along with biotin RNA pull-down assays. GAS5 was identified to be significantly lowly expressed in osteosarcoma samples especially in cisplatin-resistant (DDP-resistant) tissues. GAS5 was also downregulated in DDP-resistant cells. Over-expressed GAS5 prominently increased the sensitivity of osteosarcoma cells to DDP in vitro. Furthermore, over-expression of GAS5 suppressed cell proliferation and facilitated apoptosis of DDP-resistant cells. Mechanistically, GAS5 sponged miR-26b-5p, over-expression of which reversed the effects of GAS5 on cell proliferation and apoptosis of DDP-resistant cells. In addition, miR-26b-5p targeted TP53INP1. TP53INP1 abrogated the functions of miR-26b-5p on cell proliferation and apoptosis in DDP-resistant cells. Taken together, GAS5 enhanced the sensitivity of osteosarcoma cells to DDP via GAS5/miR-26b-5p/TP53INP1 axis. Therefore, GAS5 may be a potential indicator for the management of osteosarcoma.
Collapse
Affiliation(s)
- Guowei Li
- Department of Spine Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Xue Yan
- Respiration Medicine, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, 121000, Liaoning, China.
| |
Collapse
|
4
|
Origin and Therapies of Osteosarcoma. Cancers (Basel) 2022; 14:cancers14143503. [PMID: 35884563 PMCID: PMC9322921 DOI: 10.3390/cancers14143503] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Osteosarcoma is the most common malignant bone tumor in children, with a 5-year survival rate ranging from 70% to 20% depending on the aggressiveness of the disease. The current treatments have not evolved over the past four decades due in part to the genetic complexity of the disease and its heterogeneity. This review will summarize the current knowledge of OS origin, diagnosis and therapies. Abstract Osteosarcoma (OS) is the most frequent primary bone tumor, mainly affecting children and young adults. Despite therapeutic advances, the 5-year survival rate is 70% but drastically decreases to 20–30% for poor responders to therapies or for patients with metastasis. No real evolution of the survival rates has been observed for four decades, explained by poor knowledge of the origin, difficulties related to diagnosis and the lack of targeted therapies for this pediatric tumor. This review will describe a non-exhaustive overview of osteosarcoma disease from a clinical and biological point of view, describing the origin, diagnosis and therapies.
Collapse
|
5
|
Alemi F, Malakoti F, Vaghari-Tabari M, Soleimanpour J, Shabestani N, Sadigh AR, Khelghati N, Asemi Z, Ahmadi Y, Yousefi B. DNA damage response signaling pathways as important targets for combination therapy and chemotherapy sensitization in osteosarcoma. J Cell Physiol 2022; 237:2374-2386. [PMID: 35383920 DOI: 10.1002/jcp.30721] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/13/2022] [Accepted: 02/25/2022] [Indexed: 11/08/2022]
Abstract
Osteosarcoma (OS) is the most common bone malignancy that occurs most often in young adults, and adolescents with a survival rate of 20% in its advanced stages. Nowadays, increasing the effectiveness of common treatments used in OS has become one of the main problems for clinicians due to cancer cells becoming resistant to chemotherapy. One of the most important mechanisms of resistance to chemotherapy is through increasing the ability of DNA repair because most chemotherapy drugs damage the DNA of cancer cells. DNA damage response (DDR) is a signal transduction pathway involved in preserving the genome stability upon exposure to endogenous and exogenous DNA-damaging factors such as chemotherapy agents. There is evidence that the suppression of DDR may reduce chemoresistance and increase the effectiveness of chemotherapy in OS. In this review, we aim to summarize these studies to better understand the role of DDR in OS chemoresistance in pursuit of overcoming the obstacles to the success of chemotherapy.
Collapse
Affiliation(s)
- Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Vaghari-Tabari
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimanpour
- Department of Orthopedics Surgery, Shohada Teaching Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Shabestani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aydin R Sadigh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Khelghati
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Yasin Ahmadi
- Department of Medical Laboratory Sciences, Faculty of Science, Komar University of Science and Technology, Soleimania, Kurdistan Region, Iraq
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Xu L, Sun Z, Wei X, Tan H, Kong P, Li Z, Yang Q, Dai E, Li J. The inhibition of MARK2 suppresses cisplatin resistance of osteosarcoma stem cells by regulating DNA damage and repair. J Bone Oncol 2020; 23:100290. [PMID: 32368441 PMCID: PMC7184251 DOI: 10.1016/j.jbo.2020.100290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022] Open
Abstract
MARK2 plays an important role in the chemoresistance mechanism of osteosarcoma stem cells. Down-regulation of MARK2 in CD133+ MG-63 and MNNG/HOS cells inhibits the expression of DNA-PKcs by inhibiting the activity of the PI3K/Akt/mTOR pathway. New clues for the osteosarcoma chemotherapy strategy.
Objective This study aims to explore the role of MARK2 in chemotherapeutic resistance and potential mechanism within cisplatin resistance models of CD133+ MG-63 and MNNG/HOS cells. Methods CD133− and CD133+ MG-63 and MNNG/HOS cells were differentiated and obtained by MACS(Magnetic bead sorting). Cell activity was determined by CCK-8 assay. siRNA was employed to down regulate the Microtubule Affinity Regulated Kinase 2 (MARK2) expression. Immunofluorescence detection and RT-qPCR were used to measure the expressions of MARK2 and DNA-PKcs at both protein and mRNA levels. Western blot was applied to test the levels of MARK2, γH2AX (S139), DNA-PKcs, Phospho-PI3 Kinase p85 (Tyr458), Akt, phospho-Akt (T308) antibodies, mTOR, phospho-mTOR (Ser2448). Results Compared with CD133− MG-63 cells, CD133+ MG-63 cells showed significantly strong cisplatin resistance, with high levels of MARK2, DNA-PKcs and potent DNA damage repair ability (p<0.05). Down regulation of MARK2 reduced the cisplatin resistance of CD133+ MG-63 cells, with deceasing expression of DNA-PKcs (p<0.05). PI3K/Akt/mTOR pathway was potentially activated in CD133+ MG-63 cells, and involved in the cisplatin resistance of MG-63 cells. The similar results were observed in CD133+ MNNG/HOS cells. The reduction of MARK2 retarded the activity of PI3K/Akt/mTOR pathway and further impeded the cisplatin resistance in CD133+ MG-63 and MNNG/HOS cell. Conclusion Our data suggested that MARK2 was related to cisplatin resistance in CD133+ MG-63 and MNNG/HOS cells. The decrease of MARK2 restricted the cisplatin resistance of CD133+ MG-63 and MNNG/HOS cells by down regulating the expression of DNA dependent protein kinase catalytic subunit (DNA-PKcs) and inhibiting activity of PI3K/Akt/mTOR signaling pathway, which provides new clues for the osteosarcoma chemotherapy strategy.
Collapse
Affiliation(s)
- Liang Xu
- Department of Orthopaedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Rd, Lixia District, Jinan 250012, Shandong, China.,Department of Orthopaedics, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan 250012, Shandong, China.,Department of Orthopaedics, Shandong Provincial Chest Hospital, Jinan 250013, Shandong, China
| | - Zhengkao Sun
- Department of Orthopaedics, Qilu Hospital of Shandong University (Qingdao), 758 Hefei Rd, North District, Qingdao 266035, Shandong, China
| | - Xianfu Wei
- Department of Orthopaedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Rd, Lixia District, Jinan 250012, Shandong, China
| | - Hongdong Tan
- Department of Orthopaedics, Shandong Provincial Chest Hospital, Jinan 250013, Shandong, China
| | - Peng Kong
- Department of Orthopaedics, Shandong Traditional Chinese Medicine Hospital, Jinan 250014, Shandong, China
| | - Zhenfeng Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Rd, Lixia District, Jinan 250012, Shandong, China
| | - Qiang Yang
- Department of Orthopaedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Rd, Lixia District, Jinan 250012, Shandong, China
| | - E'nuo Dai
- Department of Orthopaedics, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan 250012, Shandong, China
| | - Jianmin Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Rd, Lixia District, Jinan 250012, Shandong, China
| |
Collapse
|
7
|
Hou J, Liu G, Zhang P, Wang B, Yan Q, Wu P, Wang C, Yao W. Experimental Study of Somatic Variants of Osteosarcoma by Whole-Exome Sequencing. Med Sci Monit 2020; 26:e920826. [PMID: 32193367 PMCID: PMC7106971 DOI: 10.12659/msm.920826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background This study aimed to investigate the role of gene mutation site distribution, biological function, pathway enrichment, and gene association analysis in the occurrence, development, and migration of osteosarcoma. Material/Methods Somatic mutation screening was performed using the whole-exome sequencing of osteosarcoma samples, and the distribution of mutations was demonstrated by Circos diagrams. Metascape was used to analyze the GO and KEGG signal pathway enrichment of the genes harboring protein coding alterations, and GeneMANIA was used to analyze the interaction of mutated genes. Results The results showed that the protein coding alterations were found throughout the whole genome in 3 osteosarcoma samples. A large number of identical or related biological processes and pathways were found in osteosarcoma samples. The GeneMANIA analysis of the 10 mutations shared by 3 samples showed that the target gene minichromosome maintenance complex component 4 (MCM4) and 3 lateral genes were most functional, and were all related to DNA replication. The analysis of GO and KEGG signal pathway enrichment showed that the mutated genes were involved mainly in tumor-related metabolic pathways. Three mutated genes were involved in the cell process, and 2 mutated genes were involved in the metabolic process. Known driver gene mutations were also observed in the samples. Conclusions The gene analysis confirmed that patients with osteosarcoma had a wide range of common gene mutations related to each other, which are involved in tumor-related metabolic pathways. These findings provide a basis for further gene-targeted therapy and pathway research.
Collapse
Affiliation(s)
- Jingyu Hou
- Department of Bone and Soft Tissue Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Guoqing Liu
- Department of Bone and Soft Tissue Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Peng Zhang
- Department of Bone and Soft Tissue Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Bangmin Wang
- Department of Bone and Soft Tissue Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Qiang Yan
- Department of Bone and Soft Tissue Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Pin Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Chuchu Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Weitao Yao
- Department of Bone and Soft Tissue Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
8
|
Dogrusöz M, Ruschel Trasel A, Cao J, Ҫolak S, van Pelt SI, Kroes WGM, Teunisse AFAS, Alsafadi S, van Duinen SG, Luyten GPM, van der Velden PA, Amaro A, Pfeffer U, Jochemsen AG, Jager MJ. Differential Expression of DNA Repair Genes in Prognostically-Favorable versus Unfavorable Uveal Melanoma. Cancers (Basel) 2019; 11:cancers11081104. [PMID: 31382494 PMCID: PMC6721581 DOI: 10.3390/cancers11081104] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/26/2019] [Accepted: 07/30/2019] [Indexed: 01/20/2023] Open
Abstract
Expression of DNA repair genes was studied in uveal melanoma (UM) in order to identify genes that may play a role in metastases formation. We searched for genes that are differentially expressed between tumors with a favorable and unfavorable prognosis. Gene-expression profiling was performed on 64 primary UM from the Leiden University Medical Center (LUMC), Leiden, The Netherlands. The expression of 121 genes encoding proteins involved in DNA repair pathways was analyzed: a total of 44 genes differed between disomy 3 and monosomy 3 tumors. Results were validated in a cohort from Genoa and Paris and the The Cancer Genome Atlas (TCGA) cohort. Expression of the PRKDC, WDR48, XPC, and BAP1 genes was significantly associated with clinical outcome after validation. PRKDC was highly expressed in metastasizing UM (p < 0.001), whereas WDR48, XPC, and BAP1 were lowly expressed (p < 0.001, p = 0.006, p = 0.003, respectively). Low expression of WDR48 and XPC was related to a large tumor diameter (p = 0.01 and p = 0.004, respectively), and a mixed/epithelioid cell type (p = 0.007 and p = 0.03, respectively). We conclude that the expression of WDR48, XPC, and BAP1 is significantly lower in UM with an unfavorable prognosis, while these tumors have a significantly higher expression of PRKDC. Pharmacological inhibition of DNA-PKcs resulted in decreased survival of UM cells. PRKDC may be involved in proliferation, invasion and metastasis of UM cells. Unraveling the role of DNA repair genes may enhance our understanding of UM biology and result in the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Mehmet Dogrusöz
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
- Department of Ophthalmology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Andrea Ruschel Trasel
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
- Universidade Federal do Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Jinfeng Cao
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130012, China
| | - Selҫuk Ҫolak
- Department of Molecular Cell Biology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
- Center for Reproductive Medicine, Elisabeth-TweeSteden Hospital, 5022 GC Tilburg, The Netherlands
| | - Sake I van Pelt
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Wilma G M Kroes
- Department of Clinical Genetics, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Amina F A S Teunisse
- Department of Clinical Genetics, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Samar Alsafadi
- Department of Translational Research, PSL Research University, Institute Curie, 75248 Paris, France
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Gregorius P M Luyten
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Pieter A van der Velden
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Adriana Amaro
- Laboratory of Tumor Epigenetics, Department of Integrated Oncology Therapies, IRCCS Ospedale Policlinico San Martino, 16133 Genoa, Italy
| | - Ulrich Pfeffer
- Laboratory of Tumor Epigenetics, Department of Integrated Oncology Therapies, IRCCS Ospedale Policlinico San Martino, 16133 Genoa, Italy
| | - Aart G Jochemsen
- Department of Molecular Cell Biology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, 2333 AZ Leiden, The Netherlands.
| |
Collapse
|
9
|
Yang Y, Li H, He Z, Xie D, Ni J, Lin X. MicroRNA‐488‐3p inhibits proliferation and induces apoptosis by targeting ZBTB2 in esophageal squamous cell carcinoma. J Cell Biochem 2019; 120:18702-18713. [PMID: 31243806 DOI: 10.1002/jcb.29178] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Yi Yang
- Department of Clinical Skills Center, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - He Li
- Department of Otolaryngoloy-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhifeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Deyao Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiangwei Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoming Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
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
|
11
|
Wang Q, Xiong J, Qiu D, Zhao X, Yan D, Xu W, Wang Z, Chen Q, Panday S, Li A, Wang S, Zhou J. Inhibition of PARP1 activity enhances chemotherapeutic efficiency in cisplatin-resistant gastric cancer cells. Int J Biochem Cell Biol 2017; 92:164-172. [PMID: 28827033 DOI: 10.1016/j.biocel.2017.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/19/2017] [Accepted: 08/02/2017] [Indexed: 01/28/2023]
Abstract
Cisplatin (DDP) is the first line chemotherapeutic drug for several cancers, including gastric cancer (GC). Unfortunately, the rapid development of drug resistance remains a significant challenge for the clinical application of cisplatin. There is an urgent need to develop new strategies to overcome DDP resistance for cancer treatment. In this study, four types of human GC cells have been divided into naturally sensitive or naturally resistant categories according to their responses to cisplatin. PARP1 activity (poly (ADP-ribose), PAR) was found to be greatly increased in cisplatin-resistant GC cells. PARP1 inhibitors significantly enhanced cisplatin-induced DNA damage and apoptosis in the resistant GC cells via the inhibition of PAR. Mechanistically, PARP1 inhibitors suppress DNA-PKcs stability and reduce the capability of DNA double-strand break (DSB) repair via the NHEJ pathway. This was also verified in BGC823/DDP GC cells with acquired cisplatin resistance. In conclusion, we identified that PARP1 is a useful interceptive target in cisplatin-resistant GC cells. Our data provide a promising therapeutic strategy against cisplatin resistance in GC cells that has potential translational significance.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jianping Xiong
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Danping Qiu
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xue Zhao
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Donglin Yan
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenxia Xu
- Laboratory of Cancer Biology, Biomedical Research Center, Sir Runrun Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Zhangding Wang
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qi Chen
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sapna Panday
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Aiping Li
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shouyu Wang
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, Key Laboratory of Modern Toxicology of the Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
He SS, Chen Y, Shen XM, Wang HZ, Sun P, Dong J, Guo GF, Chen JG, Xia LP, Hu PL, Qiu HJ, Liu SS, Zhou YX, Wang W, Hu WH, Cai XY. DNA-dependent protein kinase catalytic subunit functions in metastasis and influences survival in advanced-stage laryngeal squamous cell carcinoma. J Cancer 2017; 8:2410-2416. [PMID: 28819445 PMCID: PMC5560160 DOI: 10.7150/jca.20069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/18/2017] [Indexed: 01/15/2023] Open
Abstract
Background: DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is known to function in several types of cancer. In this study, we investigated the expression and clinicopathologic significance of DNA-PKcs in laryngeal squamous cell carcinoma (LSCC). Methods: We conducted a retrospective study of 208 patients with advanced-stage LSCC treated at Sun Yat-sen University Cancer Center, Guangzhou, China. We assessed DNA-PKcs and p16INK4a (p16) status using immunohistochemistry. We examined the association between DNA-PKcs expression and clinicopathologic features and survival outcomes. To evaluate the independent prognostic relevance of DNA-PKcs, we used univariate and multivariate Cox regression models. We estimated overall survival (OS) and distant metastasis-free survival (DMFS) using the Kaplan-Meier method. Results: Immunohistochemical analyses revealed that 163/208 (78.4%) of the LSCC tissue samples exhibited high DNA-PKcs expression. High DNA-PKcs expression was significantly associated with survival outcomes (P = 0.016) and distant metastasis (P = 0.02; chi-squared test). High DNA-PKcs expression was associated with a significantly shorter OS and DMFS than low DNA-PKcs expression (P = 0.029 and 0.033, respectively; log-rank test), and was associated with poor OS in the p16-positive subgroup (P = 0.047). Multivariate analysis identified DNA-PKcs as an independent prognostic indicator of OS and DMFS in all patients (P = 0.039 and 0.037, respectively). Conclusions: Our results suggest that patients with LSCC in whom DNA-PKcs expression is elevated have a higher incidence of distant metastasis and a poorer prognosis. DNA-PKcs may represent a marker of tumor progression in patients with p16-positive LSCC.
Collapse
Affiliation(s)
- Sha-Sha He
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiation, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Yong Chen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiation, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Ming Shen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiology, The First People's Hospital of Foshan (The affiliated Foshan Hospital of Sun Yat-Sen University), Foshan, Guangdong, China
| | - Hong-Zhi Wang
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100021, China
| | - Peng Sun
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Pathology, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Jun Dong
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Gui-Fang Guo
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Ju-Gao Chen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou
| | - Liang-Ping Xia
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Pei-Li Hu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Hui-Juan Qiu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Shou-Sheng Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Yi-Xin Zhou
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Gastric Surgery, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Wei-Han Hu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of Radiation, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiu-Yu Cai
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou.,Department of VIP Region, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
13
|
Inhibiting DNA-PK CS radiosensitizes human osteosarcoma cells. Biochem Biophys Res Commun 2017; 486:307-313. [PMID: 28300555 DOI: 10.1016/j.bbrc.2017.03.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/11/2017] [Indexed: 01/14/2023]
Abstract
Osteosarcoma survival rate has not improved over the past three decades, and the debilitating side effects of the surgical treatment suggest the need for alternative local control approaches. Radiotherapy is largely ineffective in osteosarcoma, indicating a potential role for radiosensitizers. Blocking DNA repair, particularly by inhibiting the catalytic subunit of DNA-dependent protein kinase (DNA-PKCS), is an attractive option for the radiosensitization of osteosarcoma. In this study, the expression of DNA-PKCS in osteosarcoma tissue specimens and cell lines was examined. Moreover, the small molecule DNA-PKCS inhibitor, KU60648, was investigated as a radiosensitizing strategy for osteosarcoma cells in vitro. DNA-PKCS was consistently expressed in the osteosarcoma tissue specimens and cell lines studied. Additionally, KU60648 effectively sensitized two of those osteosarcoma cell lines (143B cells by 1.5-fold and U2OS cells by 2.5-fold). KU60648 co-treatment also altered cell cycle distribution and enhanced DNA damage. Cell accumulation at the G2/M transition point increased by 55% and 45%, while the percentage of cells with >20 γH2AX foci were enhanced by 59% and 107% for 143B and U2OS cells, respectively. These results indicate that the DNA-PKCS inhibitor, KU60648, is a promising radiosensitizing agent for osteosarcoma.
Collapse
|
14
|
Sharma SK, Bagshawe KD. Translating antibody directed enzyme prodrug therapy (ADEPT) and prospects for combination. Expert Opin Biol Ther 2016; 17:1-13. [DOI: 10.1080/14712598.2017.1247802] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Surinder K. Sharma
- Research Department of Oncology, UCL Cancer Institute, University College London, London, UK
| | | |
Collapse
|
15
|
Heymann MF, Brown HK, Heymann D. Drugs in early clinical development for the treatment of osteosarcoma. Expert Opin Investig Drugs 2016; 25:1265-1280. [DOI: 10.1080/13543784.2016.1237503] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Marie-Françoise Heymann
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, Faculty of Medicine, University of Nantes, Nantes, France
- Nantes University Hospital, Nantes, France
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| | - Hannah K. Brown
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| | - Dominique Heymann
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, Faculty of Medicine, University of Nantes, Nantes, France
- Nantes University Hospital, Nantes, France
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| |
Collapse
|
16
|
Shih YL, Chou J, Yeh MY, Chou HM, Chou HC, Lu HF, Shang HS, Chueh FS, Chu YL, Hsueh SC, Chung JG. Casticin induces DNA damage and inhibits DNA repair-associated protein expression in B16F10 mouse melanoma cancer cells. Oncol Rep 2016; 36:2094-100. [DOI: 10.3892/or.2016.5027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/09/2016] [Indexed: 11/06/2022] Open
|
17
|
Li K, Li X, Tian J, Wang H, Pan J, Li J. Downregulation of DNA-PKcs suppresses P-gp expression via inhibition of the Akt/NF-κB pathway in CD133-positive osteosarcoma MG-63 cells. Oncol Rep 2016; 36:1973-80. [DOI: 10.3892/or.2016.4991] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 04/22/2016] [Indexed: 11/06/2022] Open
|
18
|
TANG NOUYING, CHUEH FUSHIN, YU CHIENCHIH, LIAO CHINGLUNG, LIN JENJYH, HSIA TECHUN, WU KINGCHUEN, LIU HSINCHUNG, LU KUNGWEN, CHUNG JINGGUNG. Benzyl isothiocyanate alters the gene expression with cell cycle regulation and cell death in human brain glioblastoma GBM 8401 cells. Oncol Rep 2016; 35:2089-96. [DOI: 10.3892/or.2016.4577] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/12/2015] [Indexed: 11/05/2022] Open
|
19
|
Dungl DA, Maginn EN, Stronach EA. Preventing Damage Limitation: Targeting DNA-PKcs and DNA Double-Strand Break Repair Pathways for Ovarian Cancer Therapy. Front Oncol 2015. [PMID: 26579492 DOI: 10.3389/fonc.2015.00240] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Platinum-based chemotherapy is the cornerstone of ovarian cancer treatment, and its efficacy is dependent on the generation of DNA damage, with subsequent induction of apoptosis. Inappropriate or aberrant activation of the DNA damage response network is associated with resistance to platinum, and defects in DNA repair pathways play critical roles in determining patient response to chemotherapy. In ovarian cancer, tumor cell defects in homologous recombination - a repair pathway activated in response to double-strand DNA breaks (DSB) - are most commonly associated with platinum-sensitive disease. However, despite initial sensitivity, the emergence of resistance is frequent. Here, we review strategies for directly interfering with DNA repair pathways, with particular focus on direct inhibition of non-homologous end joining (NHEJ), another DSB repair pathway. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a core component of NHEJ and it has shown considerable promise as a chemosensitization target in numerous cancer types, including ovarian cancer where it functions to promote platinum-induced survival signaling, via AKT activation. The development of pharmacological inhibitors of DNA-PKcs is on-going, and clinic-ready agents offer real hope to patients with chemoresistant disease.
Collapse
Affiliation(s)
- Daniela A Dungl
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Elaina N Maginn
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Euan A Stronach
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| |
Collapse
|
20
|
Dungl DA, Maginn EN, Stronach EA. Preventing Damage Limitation: Targeting DNA-PKcs and DNA Double-Strand Break Repair Pathways for Ovarian Cancer Therapy. Front Oncol 2015. [PMID: 26579492 DOI: 10.3389/fonc.2015.00240]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Platinum-based chemotherapy is the cornerstone of ovarian cancer treatment, and its efficacy is dependent on the generation of DNA damage, with subsequent induction of apoptosis. Inappropriate or aberrant activation of the DNA damage response network is associated with resistance to platinum, and defects in DNA repair pathways play critical roles in determining patient response to chemotherapy. In ovarian cancer, tumor cell defects in homologous recombination - a repair pathway activated in response to double-strand DNA breaks (DSB) - are most commonly associated with platinum-sensitive disease. However, despite initial sensitivity, the emergence of resistance is frequent. Here, we review strategies for directly interfering with DNA repair pathways, with particular focus on direct inhibition of non-homologous end joining (NHEJ), another DSB repair pathway. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a core component of NHEJ and it has shown considerable promise as a chemosensitization target in numerous cancer types, including ovarian cancer where it functions to promote platinum-induced survival signaling, via AKT activation. The development of pharmacological inhibitors of DNA-PKcs is on-going, and clinic-ready agents offer real hope to patients with chemoresistant disease.
Collapse
Affiliation(s)
- Daniela A Dungl
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Elaina N Maginn
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Euan A Stronach
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| |
Collapse
|
21
|
Dungl DA, Maginn EN, Stronach EA. Preventing Damage Limitation: Targeting DNA-PKcs and DNA Double-Strand Break Repair Pathways for Ovarian Cancer Therapy. Front Oncol 2015; 5:240. [PMID: 26579492 PMCID: PMC4620694 DOI: 10.3389/fonc.2015.00240] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/10/2015] [Indexed: 12/22/2022] Open
Abstract
Platinum-based chemotherapy is the cornerstone of ovarian cancer treatment, and its efficacy is dependent on the generation of DNA damage, with subsequent induction of apoptosis. Inappropriate or aberrant activation of the DNA damage response network is associated with resistance to platinum, and defects in DNA repair pathways play critical roles in determining patient response to chemotherapy. In ovarian cancer, tumor cell defects in homologous recombination – a repair pathway activated in response to double-strand DNA breaks (DSB) – are most commonly associated with platinum-sensitive disease. However, despite initial sensitivity, the emergence of resistance is frequent. Here, we review strategies for directly interfering with DNA repair pathways, with particular focus on direct inhibition of non-homologous end joining (NHEJ), another DSB repair pathway. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a core component of NHEJ and it has shown considerable promise as a chemosensitization target in numerous cancer types, including ovarian cancer where it functions to promote platinum-induced survival signaling, via AKT activation. The development of pharmacological inhibitors of DNA-PKcs is on-going, and clinic-ready agents offer real hope to patients with chemoresistant disease.
Collapse
Affiliation(s)
- Daniela A Dungl
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Elaina N Maginn
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Euan A Stronach
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| |
Collapse
|