1
|
Xiang H, Luo R, Wang Y, Yang B, Xu S, Huang W, Tang S, Fang R, Chen L, Zhu N, Yu Z, Akesu S, Wei C, Xu C, Zhou Y, Gu J, Zhao J, Hou Y, Ding C. Proteogenomic insights into the biology and treatment of pan-melanoma. Cell Discov 2024; 10:78. [PMID: 39039072 PMCID: PMC11263678 DOI: 10.1038/s41421-024-00688-7] [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: 08/04/2023] [Accepted: 05/03/2024] [Indexed: 07/24/2024] Open
Abstract
Melanoma is one of the most prevalent skin cancers, with high metastatic rates and poor prognosis. Understanding its molecular pathogenesis is crucial for improving its diagnosis and treatment. Integrated analysis of multi-omics data from 207 treatment-naïve melanomas (primary-cutaneous-melanomas (CM, n = 28), primary-acral-melanomas (AM, n = 81), primary-mucosal-melanomas (MM, n = 28), metastatic-melanomas (n = 27), and nevi (n = 43)) provides insights into melanoma biology. Multivariate analysis reveals that PRKDC amplification is a prognostic molecule for melanomas. Further proteogenomic analysis combined with functional experiments reveals that the cis-effect of PRKDC amplification may lead to tumor proliferation through the activation of DNA repair and folate metabolism pathways. Proteome-based stratification of primary melanomas defines three prognosis-related subtypes, namely, the ECM subtype, angiogenesis subtype (with a high metastasis rate), and cell proliferation subtype, which provides an essential framework for the utilization of specific targeted therapies for particular melanoma subtypes. The immune classification identifies three immune subtypes. Further analysis combined with an independent anti-PD-1 treatment cohort reveals that upregulation of the MAPK7-NFKB signaling pathway may facilitate T-cell recruitment and increase the sensitivity of patients to immunotherapy. In contrast, PRKDC may reduce the sensitivity of melanoma patients to immunotherapy by promoting DNA repair in melanoma cells. These results emphasize the clinical value of multi-omics data and have the potential to improve the understanding of melanoma treatment.
Collapse
Affiliation(s)
- Hang Xiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bing Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shaoshuai Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rundong Fang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lingli Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Na Zhu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sujie Akesu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanyuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianying Gu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Shanghai, China.
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Liu Y, Jiang C, Liu Q, Huang R, Wang M, Guo X. CircRNAs: emerging factors for regulating glucose metabolism in colorectal cancer. Clin Transl Oncol 2023:10.1007/s12094-023-03131-7. [PMID: 36944731 DOI: 10.1007/s12094-023-03131-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/19/2023] [Indexed: 03/23/2023]
Abstract
Colorectal cancer is a malignant disease with a high incidence and low survival rate, and the effectiveness of traditional treatments, such as surgery and radiotherapy, is very limited. CircRNAs, a kind of stable endogenous circular RNA, generally function by sponging miRNAs and binding or translating proteins. CircRNAs have been identified to play an important role in regulating the proliferation and metabolism of CRC. In recent years, many reports have indicated that by regulating the expression of glycolysis-related proteins, such as GLUT1 and HK2, or directly translating proteins, circRNAs can promote the Warburg effect in cancer cells, thereby driving CRC metabolism. Moreover, the Warburg effect increases lactate production in cancer cells and promotes acidification of the TME, which further drives cancer progression. In this review, we summarized the remarkable role of circRNAs in regulating glucose metabolism in CRC in recent years, which might be useful for finding new targets for the clinical treatment of CRC.
Collapse
Affiliation(s)
- Yulin Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Chenjun Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Qianqian Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Runchun Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Mancai Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
- General Surgery Department, The Second Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Xiaohu Guo
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China.
- General Surgery Department, The Second Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
3
|
Tirilomi A, Elakad O, Yao S, Li Y, Hinterthaner M, Danner BC, Ströbel P, Tirilomis T, Bohnenberger H, von Hammerstein-Equord A. Expression and prognostic impact of DNA-PK in human lung cancer. Medicine (Baltimore) 2023; 102:e33143. [PMID: 36862864 PMCID: PMC9981429 DOI: 10.1097/md.0000000000033143] [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: 03/04/2023] Open
Abstract
Among all cancer patient's lung cancer is the leading cause of death. Prognostic biomarkers continue to be investigated for the detection and stratification of lung cancer for clinical use. The DNA-dependent protein kinase is involved in mechanisms of DNA damage repair. Deregulation and overexpression of DNA-dependent protein kinase is associated with poor prognosis in various tumor entities. In this study, we investigated the expression of DNA-dependent protein kinase in relation to clinicopathological features and overall survival in patients with lung cancer. By immunohistochemistry, expression of DNA-dependent protein kinase was analyzed in 205 cases of lung cancer; 95 cases of adenocarcinoma, 83 cases of squamous cell lung carcinoma and 27 cases of small cell lung cancer and correlated with clinicopathological characteristics as well as patient's overall survival. In patients with adenocarcinoma, a significant correlation between strong expression of DNA-dependent protein kinase and worse overall survival was found. No significant association was observed in patients with squamous cell lung carcinoma and small cell lung cancer. Strong detection of DNA-dependent protein kinase expression was most evident in small cell lung cancer (81.48 %), followed by squamous cell lung carcinoma (62.65 %) and adenocarcinoma (61.05 %). In our study, expression of DNA-dependent protein kinase was associated with poor overall survival in patients with adenocarcinoma. DNA-dependent protein kinase could serve as a new prognostic biomarker.
Collapse
Affiliation(s)
- Anna Tirilomi
- Department of Cardio-Thoracic and Vascular Surgery, University Medical Center, Göttingen, Germany
| | - Omar Elakad
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Sha Yao
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Yuchan Li
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Marc Hinterthaner
- Department of Cardio-Thoracic and Vascular Surgery, University Medical Center, Göttingen, Germany
| | - Bernhard C. Danner
- Department of Cardio-Thoracic and Vascular Surgery, University Medical Center, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Theodor Tirilomis
- Department of Cardio-Thoracic and Vascular Surgery, University Medical Center, Göttingen, Germany
| | | | - Alexander von Hammerstein-Equord
- Department of Cardio-Thoracic and Vascular Surgery, University Medical Center, Göttingen, Germany
- * Correspondence: Alexander von Hammerstein-Equord, Department of Cardio-Thoracic and Vascular Surgery, University Medical Center, Robert-Koch-Str. 40, Göttingen 37075, Germany (e-mail: )
| |
Collapse
|
4
|
Liu J, Huang J, Cheng X, Liao Z, Gao X. miR-556-3p/Disabled Homolog 2-Interacting Protein (dab2ip) Promotes Cancer Progression by Down-Regulating Bcl-2-Like Protein 11 (BIM) Expression in Colorectal Cancer. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is a major threat affecting human health. Studies have shown that miR-556-3p can regulate dab2ip and promote tumor deterioration, and up-regulation of BIM inhibits CRC cell progression. However, the interaction between miR-556-3p/dab2ip and BIM in CRC is unknown.
We examined miR-556-3p expression in CRC tissues and cells by RT-qPCR. The impact of miR-556-3p/dab2ip and BIM on CRC cell behaviors were assessed by western blot, transwell and MTT assay. miR-556-3p was highly expressed in CRC and its overexpression increased CRC cell proliferation and migration
as well as up-regulated dab2ip and Ki-67 expression. Besides, miR-556-3p could target the BIM and overexpressed miR-556-3p decreased BIM expression. However, silencing of BIM abrogated the impact of overexpressed miR-556-3p on CRC cell proliferation and migration. In conclusion, miR-556-3p/dab2ip
promotes cell growth by down-regulating the expression of BIM, thereby promoting the progression of CRC.
Collapse
Affiliation(s)
- Jiaqi Liu
- Department of General Surgery, Beihai People’s Hospital, Beihai City, Guangxi Zhuang Autonomous Region, 536000, China
| | - Jingping Huang
- Department of Nutrition, Beihai People’s Hospital, Beihai City, Guangxi Zhuang Autonomous Region, 536000, China
| | - Xueyuan Cheng
- Department of General Surgery, Beihai People’s Hospital, Beihai City, Guangxi Zhuang Autonomous Region, 536000, China
| | - Zuowei Liao
- Department of General Surgery, Beihai People’s Hospital, Beihai City, Guangxi Zhuang Autonomous Region, 536000, China
| | - Xueyuan Gao
- Department of General Surgery, Beihai People’s Hospital, Beihai City, Guangxi Zhuang Autonomous Region, 536000, China
| |
Collapse
|
5
|
Valikhani M, Rahimian E, Ahmadi SE, Chegeni R, Safa M. Involvement of classic and alternative non-homologous end joining pathways in hematologic malignancies: targeting strategies for treatment. Exp Hematol Oncol 2021; 10:51. [PMID: 34732266 PMCID: PMC8564991 DOI: 10.1186/s40164-021-00242-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/13/2021] [Indexed: 12/31/2022] Open
Abstract
Chromosomal translocations are the main etiological factor of hematologic malignancies. These translocations are generally the consequence of aberrant DNA double-strand break (DSB) repair. DSBs arise either exogenously or endogenously in cells and are repaired by major pathways, including non-homologous end-joining (NHEJ), homologous recombination (HR), and other minor pathways such as alternative end-joining (A-EJ). Therefore, defective NHEJ, HR, or A-EJ pathways force hematopoietic cells toward tumorigenesis. As some components of these repair pathways are overactivated in various tumor entities, targeting these pathways in cancer cells can sensitize them, especially resistant clones, to radiation or chemotherapy agents. However, targeted therapy-based studies are currently underway in this area, and furtherly there are some biological pitfalls, clinical issues, and limitations related to these targeted therapies, which need to be considered. This review aimed to investigate the alteration of DNA repair elements of C-NHEJ and A-EJ in hematologic malignancies and evaluate the potential targeted therapies against these pathways.
Collapse
Affiliation(s)
- Mohsen Valikhani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences, Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Rahimian E, Amini A, Alikarami F, Pezeshki SMS, Saki N, Safa M. DNA repair pathways as guardians of the genome: Therapeutic potential and possible prognostic role in hematologic neoplasms. DNA Repair (Amst) 2020; 96:102951. [PMID: 32971475 DOI: 10.1016/j.dnarep.2020.102951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022]
Abstract
DNA repair pathways, which are also identified as guardians of the genome, protect cells from frequent damage that can lead to DNA breaks. The most deleterious types of damage are double-strand breaks (DSBs), which are repaired by homologous recombination (HR) and non-homologous end joining (NHEJ). Single strand breaks (SSBs) can be corrected through base excision repair (BER), nucleotide excision repair (NER), and mismatch repair (MMR). Failure to restore DNA lesions or inappropriately repaired DNA damage culminates in genomic instability and changes in the regulation of cellular functions. Intriguingly, particular mutations and translocations are accompanied by special types of leukemia. Besides, expression patterns of certain repair genes are altered in different hematologic malignancies. Moreover, analysis of mutations in key mediators of DNA damage repair (DDR) pathways, as well as investigation of their expression and function, may provide us with emerging biomarkers of response/resistance to treatment. Therefore, defective DDR pathways can offer a rational starting point for developing DNA repair-targeted drugs. In this review, we address genetic alterations and gene/protein expression changes, as well as provide an overview of DNA repair pathways.
Collapse
Affiliation(s)
- Elahe Rahimian
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Amini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Alikarami
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA 19104, USA
| | - Seyed Mohammad Sadegh Pezeshki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Li Y, Zang H, Zhang X, Huang G. circ_0136666 Facilitates the Progression of Colorectal Cancer via miR-383/CREB1 Axis. Cancer Manag Res 2020; 12:6795-6806. [PMID: 32821160 PMCID: PMC7424319 DOI: 10.2147/cmar.s251952] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/09/2020] [Indexed: 12/23/2022] Open
Abstract
Background The changes in dietary patterns cause an increased incidence of colorectal cancer (CRC) globally. We aimed to explore the mechanism behind circular RNA circ_0136666 in the progression of CRC. Materials and Methods The expression of circ_0136666, miR-383 and cAMP response element binding protein 1 (CREB1) was detected by real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, apoptosis and glycolysis were measured by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry and glucose or lactate detection kit, respectively. The combination between miR-383 and circ_0136666 or CREB1 in 293T cells was predicted by Circular RNA Interactome or Starbase software and confirmed by dual-luciferase reporter assay. Western blot assay was performed to detect the abundance of CREB1, hexokinase 2 (HK2) and lactate dehydrogenase A (LDHA) in CRC cells. Murine xenograft model was established to verify the function of circ_0136666 in vivo. Results circ_0136666 was aberrantly up-regulated in CRC tissues and cells, and it promoted the proliferation and glycolysis and inhibited the apoptosis of CRC cells. circ_0136666 accelerated the progression of CRC through directly targeting and down-regulating miR-383. CREB1 could bind to miR-383 in 293T cells. The overexpression of CREB1 reversed the inhibitory effects of miR-383 accumulation on the proliferation and glycolysis and the promoting impact on the apoptosis of CRC cells. The enrichment of CREB1 was modulated by circ_0136666/miR-383 signaling in CRC cells. The glycolysis-related proteins (HK2 and LDHA) were modulated by circ_0136666/miR-383/CREB1 axis in CRC cells. circ_0136666 accelerated the growth of CRC tumors via circ_0136666/miR-383/CREB1 axis in vivo. Conclusion circ_0136666 deteriorated CRC through miR-383/CREB1 axis. circ_0136666/miR-383/CREB1 axis might be an underlying therapeutic target for CRC therapy.
Collapse
Affiliation(s)
- Yuhui Li
- Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Hongliang Zang
- Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xue Zhang
- Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Guomin Huang
- Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
8
|
DNA-PK in human malignant disorders: Mechanisms and implications for pharmacological interventions. Pharmacol Ther 2020; 215:107617. [PMID: 32610116 DOI: 10.1016/j.pharmthera.2020.107617] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
The DNA-PK holoenzyme is a fundamental element of the DNA damage response machinery (DDR), which is responsible for cellular genomic stability. Consequently, and predictably, over the last decades since its identification and characterization, numerous pre-clinical and clinical studies reported observations correlating aberrant DNA-PK status and activity with cancer onset, progression and responses to therapeutic modalities. Notably, various studies have established in recent years the role of DNA-PK outside the DDR network, corroborating its role as a pleiotropic complex involved in transcriptional programs that operate biologic processes as epithelial to mesenchymal transition (EMT), hypoxia, metabolism, nuclear receptors signaling and inflammatory responses. In particular tumor entities as prostate cancer, immense research efforts assisted mapping and describing the overall signaling networks regulated by DNA-PK that control metastasis and tumor progression. Correspondingly, DNA-PK emerges as an obvious therapeutic target in cancer and data pertaining to various pharmacological approaches have been published, largely in context of combination with DNA-damaging agents (DDAs) that act by inflicting DNA double strand breaks (DSBs). Currently, new generation inhibitors are tested in clinical trials. Several excellent reviews have been published in recent years covering the biology of DNA-PK and its role in cancer. In the current article we are aiming to systematically describe the main findings on DNA-PK signaling in major cancer types, focusing on both preclinical and clinical reports and present a detailed current status of the DNA-PK inhibitors repertoire.
Collapse
|
9
|
Zhang Y, Yang WK, Wen GM, Tang H, Wu CA, Wu YX, Jing ZL, Tang MS, Liu GL, Li DZ, Li YH, Deng YJ. High expression of PRKDC promotes breast cancer cell growth via p38 MAPK signaling and is associated with poor survival. Mol Genet Genomic Med 2019; 7:e908. [PMID: 31513357 PMCID: PMC6825841 DOI: 10.1002/mgg3.908] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/14/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND DNA-Dependent Protein Kinase Catalytic Subunit (PRKDC), a key component of the DNA damage repair pathway, is associated with chemotherapy resistance and tumor progression. METHODS Here we analyzed transcriptome data of ~2,000 breast cancer patients and performed functional studies in vitro to investigate the function of PRKDC in breast cancer. RESULTS Our results revealed overexpression of PRKDC in multiple breast cancer subtypes. Consistent with patients' data, overexpression of PRKDC was also observed in breast cancer cell lines compared to normal breast epithelial cells. Knockdown of PRKDC in MCF-7 and T47D breast cancer cell lines resulted in proliferation inhibition, reduced colony formation and G2/M cell cycle arrest. Furthermore, we showed that PRKDC knockdown induced proliferation inhibition through activation of p38 MAPK, but not ERK MAPK, signaling pathway in breast cancer cells. Blockage of p38 MAPK signaling could largely rescue proliferation inhibition and cell cycle arrest induced by PRKDC knockdown. Moreover, we analyzed gene expression and clinical data from six independent breast cancer cohorts containing ~1,000 patients. In all cohorts, our results consistently showed that high expression of PRKDC was significantly associated with poor survival in both treated and untreated breast cancer patients. CONCLUSION Together, our results suggest that high expression of PRKDC facilitates breast cancer cell growth via regulation of p38 MAPK signaling, and is a prognostic marker for poor survival in breast cancer patients.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, P.R. China.,Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Wei-Kang Yang
- Department of Prevention and Health Care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, P.R. China
| | - Guo-Ming Wen
- Department of Outpatient, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, P.R. China
| | - Hongping Tang
- Department of Pathology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, P.R. China
| | - Chuan-An Wu
- Department of Prevention and Health Care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, P.R. China
| | - Yan-Xia Wu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Zhi-Liang Jing
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Min-Shan Tang
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Guang-Long Liu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Da-Zhou Li
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Yan-Hua Li
- Department of Pathology, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, P.R. China
| | - Yong-Jian Deng
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
10
|
Zhang Y, Wen GM, Wu CA, Jing ZL, Li DZ, Liu GL, Wei XX, Tang MS, Li YH, Zhong Y, Deng YJ, Yang WK. PRKDC is a prognostic marker for poor survival in gastric cancer patients and regulates DNA damage response. Pathol Res Pract 2019; 215:152509. [PMID: 31255330 DOI: 10.1016/j.prp.2019.152509] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/27/2019] [Accepted: 06/20/2019] [Indexed: 01/01/2023]
Abstract
A hallmark of gastric cancer is the high rate of genomic instability associated with deregulation of DNA damage repair pathways. DNA-Dependent Protein Kinase Catalytic Subunit (PRKDC) is a key component of the non-homologous end-joining (NHEJ) pathway. By reanalyzing transcriptome data of 80 pairs of gastric cancer tumors and the adjacent normal tissues from non-treated patients, we identified PRKDC as the top upregulated DNA damage repair genes in gastric cancer. High expression of PRKDC is associated with poor survival of gastric cancer patients, and genomic amplification of the gene is frequently observed across most gastric cancer subtypes. Knockdown of PRKDC in gastric cell lines resulted in reduced proliferation and cell cycle arrest. Furthermore, we showed that loss of PRKDC induced DNA damage and enhanced gastric cancer cell chemosensitivity to DNA-damaging reagents. Together, our results suggest that PRKDC is a prognostic marker of poor survival and is a putative target to overcome chemoresistance in gastric cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, PR China.
| | - Guo-Ming Wen
- Department of Outpatient, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, PR China
| | - Chuan-An Wu
- Department of Prevention and health care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, PR China
| | - Zhi-Liang Jing
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, PR China
| | - Da-Zhou Li
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, PR China
| | - Guang-Long Liu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, PR China
| | - Xu-Xuan Wei
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, PR China
| | - Min-Shan Tang
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, PR China
| | - Yan-Hua Li
- Department of Pathology, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, PR China
| | - Yan Zhong
- Department of Pathology, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, PR China
| | - Yong-Jian Deng
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, PR China.
| | - Wei-Kang Yang
- Department of Prevention and health care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, PR China.
| |
Collapse
|
11
|
Sun JS, Yang XH. Expression of DNA-dependent protein kinase catalytic subunit in laryngeal squamous cell carcinoma and its importance. Exp Ther Med 2018; 15:3295-3301. [PMID: 29545847 PMCID: PMC5840916 DOI: 10.3892/etm.2018.5826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 06/08/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the expression and distribution of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) in tumor tissues and adjacent normal mucosa tissues of patients with laryngeal squamous cell carcinoma (LSCC), and further analyze the association between the expression and the clinicopathological parameters of patients with LSCC. Clinical data of tumor tissues and corresponding adjacent normal mucosa tissues of pathologically diagnosed LSCC in 96 cases were collected in the present study. Of these specimens, the mRNA and protein expression levels of DNA-PKcs in LSCC tissues and the adjacent normal mucosa tissues were analyzed via reverse transcription-quantitative polymerase chain reaction and western blot analysis. Immunohistochemistry was used to detect expression and distribution of DNA-PKcs protein in LSCC tissues and corresponding adjacent normal mucosa tissues. The association between DNA-PKcs expression and the specific clinicopathologic features was evaluated by the χ2 test. Kaplan-Meier and Cox proportional hazards regression models were used to analyze the data. It was revealed that the expression of DNA-PKcs mRNA and protein was significantly higher in LSCC tissues than the adjacent normal mucosa tissues (P<0.05). DNA-PKcs was expressed predominantly in the nucleus. DNA-PKcs expression showed significant correlation with the differentiation degree of LSCC (P<0.05), and changes of DNA-PKcs expression gradually increased with the decrease of the differentiation degree. However, DNA-PKcs expression was not significantly associated with sex, age, lymph node metastasis or TMN stage (P>0.05). Patients with LSCC exhibited higher DNA-PKcs expression had markedly shorter survival than those with lower DNA-PKcs expression. In conclusion, the present results suggested that the expression levels of DNA-PKcs were significantly increased in LSCC tumor tissues than in adjacent normal mucosa. DNA-PKcs expression was correlated with differentiation of LSCC, and may become a novel prognostic marker for patients with LSCC.
Collapse
Affiliation(s)
- Jian-Song Sun
- Department of Otolaryngology-Head and Neck Surgery, The People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Xiu-Hai Yang
- Department of Otolaryngology-Head and Neck Surgery, The People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
12
|
Sun G, Yang L, Dong C, Ma B, Shan M, Ma B. PRKDC regulates chemosensitivity and is a potential prognostic and predictive marker of response to adjuvant chemotherapy in breast cancer patients. Oncol Rep 2017; 37:3536-3542. [DOI: 10.3892/or.2017.5634] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 04/05/2017] [Indexed: 11/06/2022] Open
|
13
|
Down-regulation of protein kinase, DNA-activated, catalytic polypeptide attenuates tumor progression and is an independent prognostic predictor of survival in prostate cancer. Urol Oncol 2016; 35:111.e15-111.e23. [PMID: 27856181 DOI: 10.1016/j.urolonc.2016.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/27/2016] [Accepted: 10/13/2016] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Protein kinase, DNA-activated, catalytic polypeptide (PRKDC) is a critical component of DNA repair machinery and its dysregulated expression has been observed in various cancer types or premalignant cells. However, its role in prostate cancer (PCa) development and its prognostic significance in PCa is unknown. METHODS The mRNA and protein levels of PRKDC were analyzed in 15 pairs of PCa and benign prostatic hyperplasia tissues as well as PCa cell lines by quantitative real-time polymerase chain reaction and Western blot, respectively. Small interfering RNA and short hairpin RNA-mediated knockdown of PRKDC, followed by cell proliferation, colony formation, and soft agar assays were performed. Xenograft mouse model was used to evaluate in vivo effects of PRKDC knockdown. The association between PRKDC expression and clinicopathologic features was assessed by χ2 tests. Kaplan-Meier analysis was performed to investigate the association between PRKDC expression and overall survival. Cox proportional hazards regression models were used to examine the prognostic significance of PRKDC. RESULTS Expression of PRKDC mRNA and protein was notably higher in PCa tissues and PCa cell lines. Knockdown of PRKDC markedly reduced cell proliferation, colony formation efficiency, and soft agar growth in DU145 cells. Down-regulation of PRKDC inhibited tumor growth of DU145 xenografts and enhance mice survival. In addition, PRKDC expression in PCa was significantly associated with Gleason score (P = 0.01), tumor stage (P = 0.028), and distant metastasis (P = 0.025). Patients with PCa having higher PRKDC expression had substantially shorter survival than patients with lower PRKDC expression. CONCLUSION Down-regulation of PRKDC attenuates tumor progression in PCa. PRKDC may potentially be a prognostic biomarker in PCa.
Collapse
|
14
|
Ramzan Z, Nassri AB, Huerta S. Genotypic characteristics of resistant tumors to pre-operative ionizing radiation in rectal cancer. World J Gastrointest Oncol 2014; 6:194-210. [PMID: 25024812 PMCID: PMC4092337 DOI: 10.4251/wjgo.v6.i7.194] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/19/2014] [Accepted: 05/08/2014] [Indexed: 02/05/2023] Open
Abstract
Due to a wide range of clinical response in patients undergoing neo-adjuvant chemoradiation for rectal cancer it is essential to understand molecular factors that lead to the broad response observed in patients receiving the same form of treatment. Despite extensive research in this field, the exact mechanisms still remain elusive. Data raging from DNA-repair to specific molecules leading to cell survival as well as resistance to apoptosis have been investigated. Individually, or in combination, there is no single pathway that has become clinically applicable to date. In the following review, we describe the current status of various pathways that might lead to resistance to the therapeutic applications of ionizing radiation in rectal cancer.
Collapse
|
15
|
Perrot A, Pionneau C, Azar N, Baillou C, Lemoine FM, Leblond V, Merle-Béral H, Béné MC, Herbrecht R, Bahram S, Vallat L. Waldenström's macroglobulinemia harbors a unique proteome where Ku70 is severely underexpressed as compared with other B-lymphoproliferative disorders. Blood Cancer J 2012; 2:e88. [PMID: 22961060 PMCID: PMC3461705 DOI: 10.1038/bcj.2012.35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Waldenström's macroglobulinemia (WM) is a clonal B-cell lymphoproliferative disorder (LPD) of post-germinal center nature. Despite the fact that the precise molecular pathway(s) leading to WM remain(s) to be elucidated, a hallmark of the disease is the absence of the immunoglobulin heavy chain class switch recombination. Using two-dimensional gel electrophoresis, we compared proteomic profiles of WM cells with that of other LPDs. We were able to demonstrate that WM constitutes a unique proteomic entity as compared with chronic lymphocytic leukemia and marginal zone lymphoma. Statistical comparisons of protein expression levels revealed that a few proteins are distinctly expressed in WM in comparison with other LPDs. In particular we observed a major downregulation of the double strand repair protein Ku70 (XRCC6); confirmed at both the protein and RNA levels in an independent cohort of patients. Hence, we define a distinctive proteomic profile for WM where the downregulation of Ku70—a component of the non homologous end-joining pathway—might be relevant in disease pathophysiology.
Collapse
Affiliation(s)
- A Perrot
- 1] Laboratoire d'Immunogénétique Moléculaire Humaine, Centre de Recherche d'Immunologie et d'Hématologie. Faculté de Médecine, Université de Strasbourg, Strasbourg Cedex, France [2] Laboratoire Central d'Immunologie, Plateau Technique de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg Cedex, France [3] EA RHEM 4369, Laboratoire d'Immunologie, Faculté de médecine, Nancy-Université, Vandœuvre-lès-Nancy, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Roddam PL, Allan JM, Dring AM, Worrillow LJ, Davies FE, Morgan GJ. Non-homologous end-joining gene profiling reveals distinct expression patterns associated with lymphoma and multiple myeloma. Br J Haematol 2010; 149:258-62. [PMID: 20148879 DOI: 10.1111/j.1365-2141.2010.08088.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Repair of DNA strand breaks induced during lymphoid antigen receptor rearrangement involves non-homologous end-joining (NHEJ). We investigated NHEJ in the aetiology of lymphoproliferative disorders (LPDs) and the disease subtypes therein through real-time quantitative RT-PCR gene expression analysis. Lower expression of XRCC6 and MRE11A was observed in all tumours, with higher expression of both XRCC4 and RAD50 observed only in multiple myeloma (MM). Hierarchical clustering enabled tumours to be clearly distinguished from controls, and by morphological sub-type. We postulate this identifies targets worthy of investigation in the genetic predisposition, pathogenesis and prognosis of lymphoid malignancies.
Collapse
Affiliation(s)
- Philippa L Roddam
- The Children's Foundation Research Center, Le Bonheur Children's Medical Center, TN 38103, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Wang G, Wang S, Shen Q, Yin S, Li C, Li A, Li J, Zhou J, Liu Q. Polymorphisms in XRCC5, XRCC6, XRCC7 genes are involved in DNA double-strand breaks(DSBs) repair associated with the risk of acute myeloid leukemia(AML) in Chinese population. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/s1007-4376(09)60034-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
19
|
An J, Yang DY, Xu QZ, Zhang SM, Huo YY, Shang ZF, Wang Y, Wu DC, Zhou PK. DNA-dependent protein kinase catalytic subunit modulates the stability of c-Myc oncoprotein. Mol Cancer 2008; 7:32. [PMID: 18426604 PMCID: PMC2383926 DOI: 10.1186/1476-4598-7-32] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Accepted: 04/22/2008] [Indexed: 12/31/2022] Open
Abstract
Background C-Myc is a short-lived oncoprotein that is destroyed by ubiquitin-mediated proteolysis. Dysregulated accumulation of c-Myc commonly occurs in human cancers. Some of those cases with the dysregulated c-Myc protein accumulation are attributed to gene amplification or increased mRNA expression. However, the abnormal accumulation of c-Myc protein is also a common finding in human cancers with normal copy number and transcription level of c-Myc gene. It seems that the mechanistic dysregulation in the control of c-Myc protein stabilization is another important hallmark associated with c-Myc accumulation in cancer cells. Here we report a novel mechanistic pathway through which DNA-dependent protein kinase catalytic subunit (DNA-PKcs) modulates the stability of c-Myc protein. Results Firstly, siRNA-mediated silencing of DNA-PKcs strikingly downregulated c-Myc protein levels in HeLa and HepG2 cells, and simultaneously decreased cell proliferation. The c-Myc protein level in DNA-PKcs deficient human glioma M059J cells was also found much lower than that in DNA-PKcs efficient M059K cells. ATM deficiency does not affect c-Myc expression level. Silencing of DNA-PKcs in HeLa cells resulted in a decreased stability of c-Myc protein, which was associated the increasing of c-Myc phosphorylation on Thr58/Ser62 and ubiquitination level. Phosphorylation of Akt on Ser473, a substrate of DNA-PKcs was found decreased in DNA-PKcs deficient cells. As the consequence, the phosphorylation of GSK3 β on Ser9, a negatively regulated target of Akt, was also decreased, and which led to activation of GSK 3β and in turn phosphorylation of c-Myc on Thr58. Moreover, inhibition of GSK3 activity by LiCl or specific siRNA molecules rescued the downregulation of c-Myc mediated by silencing DNA-PKcs. Consistent with this depressed DNA-PKcs cell model, overexpressing DNA-PKcs in normal human liver L02 cells, by sub-chronically exposing to very low dose of carcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), increased c-Myc protein level, the phosphorylation of Akt and GSK3 β, as well as cell proliferation. siRNA-mediated silencing of DNA-PKcs in this cell model reversed above alterations to the original levels of L02 cells. Conclusion A suitable DNA-PKcs level in cells is necessary for maintaining genomic stability, while abnormal overexpression of DNA-PKcs may contribute to cell proliferation and even oncogenic transformation by stabilizing the c-Myc oncoprotein via at least the Akt/GSK3 pathway. Our results suggest DNA-PKcs a novel biological role beyond its DNA repair function.
Collapse
Affiliation(s)
- Jing An
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Activities of DNA-PK and Ku86, but not Ku70, may predict sensitivity to cisplatin in human gliomas. J Neurooncol 2008; 89:27-35. [PMID: 18415044 DOI: 10.1007/s11060-008-9592-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 03/28/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE This study was designed to investigate the relationship between activities of DNA-dependent protein kinase (DNA-PK), its subunits Ku86/Ku70, and sensitivities to cisplatin in human glioma samples. METHODS Thirty-six glioma samples from patients without prior treatment before neurosurgery were included in this study. The sensitivities to cisplatin as indicated by IC(50) (the inhibitory concentration leading to 50% cell death) were assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenytetrazolium (MTT) assay; activities of DNA-PK and Ku70/Ku86 were analyzed by SigmaTECT DNA-Dependent Protein Kinase Assay System and Ku70/Ku86 DNA Repair Kit, respectively. RESULTS Sensitivities to cisplatin correlated with the activities of DNA-PK/Ku86, but not with the Ku70 or other clinical parameters such as age, sex of the patients, pathological gradings of the tumors, or tumor size. The levels of DNA-PK activities also associated with pathological grading and Ku86, but not with other clinical parameters. The tumors of the patients who failed to respond to cisplatin-based chemotherapy tended to display higher activity levels of DNA-PK and Ku86. Furthermore, platinum-based chemotherapy did not result in significant changes of DNA-PK/Ku activities in four matched samples before and after chemotherapy. CONCLUSION Pretreatment determination of DNA-PK/Ku86 activities might be helpful in identifying patients who will actually benefit from platinum-based treatment.
Collapse
|
21
|
Yu ZJ, Xu QZ, Zhou LJ, Sui JL, Zhang SM, An J, Wang Y, Zhou PK. Over-expression of DNA-PKcs in hepatocarcinoma tissues and the antiproliferative effect mediated by down-regulating its expression with siRNA. Shijie Huaren Xiaohua Zazhi 2007; 15:3815-3821. [DOI: 10.11569/wcjd.v15.i36.3815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To characterize the difference in DNA-dependent protein kinase catalytic subunit (DNA-PKcs) expression between hepatocarcinoma tissues and non-tumor liver tissues, and to understand the biological significance of the altered expression of DNA-PKcs in cancer cells.
METHODS: DNA-PKcs expression was examined in 86 specimens of hepatocarcinoma and hepatic cholangiocarcinoma by immunohistochemistry analysis on tissue-arrays and ordinary pathological tissue slices. DNA-PKcs protein was detected in culture cells by Western blotting analysis. The siRNA was used to silence DNA-PKcs expression in HepG2 cells. Cell proliferation was evaluated by growth curve analysis, and radiosensitivity was tested using a colony-forming ability assay.
RESULTS: Tissue-array analysis showed that the proportions of cases with a DNA-PKcs expression level of < 25% (very low), 25%-50% (low), 51%-75% (medium) and > 75% (high) were 15%, 20%, 23.3% and 41.7%, respectively, among the 60 hepatocarcinoma tissue specimens. The proportions of the 64 normal liver tissues showing these DNA-PKcs expression levels were 68.7%, 10.9%, 12.6% and 7.8%, respectively. This result indicated that DNA-PKcs expression in cancer tissues was significantly higher than that in normal tissues (P = 0.0008). Immunohistochemistry analysis of ordinary pathological tissue slices also showed that DNA-PKcs levels in the hepatocarcinoma and hepatic cholangiocarcinoma tissues (total 26 specimens) were higher than those in the non-tumor liver tissues (P = 0.001). Moreover, the cultured human liver cancer cell lines HepG2, 7721 and 7402 also exhibited much higher levels of DNA-PKcs than normal liver LO2 cells. Silencing of DNA-PKcs mediated by siRNA resulted in not only an increased sensitivity of HepG2 cells to ionizing radiation, but also decreased proliferation HepG2 cells. We further demonstrated that the expression of c-Myc oncoprotein was also depressed following the down-regulation of DNA-PKcs.
CONCLUSION: DNA-PKcs is overexpressed in hepatocarcinoma tissues as well as cultured liver cancer cells. Targeting DNA-PKcs is an anticancer strategy acting through at least two mechanisms: directly decreasing the proliferation of cancer cells via, at least, depressing c-Myc oncoprotein; and sensitizing cancer cells to radiotherapy or chemotherapy with DNA-damaging agents.
Collapse
|
22
|
Abstract
Telomere length and telomerase activity are important factors in the pathobiology of human disease. Age-related diseases and premature ageing syndromes are characterized by short telomeres, which can compromise cell viability, whereas tumour cells can prevent telomere loss by aberrantly upregulating telomerase. Altered functioning of both telomerase and telomere-interacting proteins is present in some human premature ageing syndromes and in cancer, and recent findings indicate that alterations that affect telomeres at the level of chromatin structure might also have a role in human disease. These findings have inspired a number of potential therapeutic strategies that are based on telomerase and telomeres.
Collapse
Affiliation(s)
- Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), 28029 Madrid, Spain.
| |
Collapse
|