1
|
Azadegan C, Santoro J, Whetstine JR. CONNECTING THE DOTS: EPIGENETIC REGULATION OF EXTRACHROMOSOMAL AND INHERITED DNA AMPLIFICATIONS. J Biol Chem 2025:108454. [PMID: 40154613 DOI: 10.1016/j.jbc.2025.108454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025] Open
Abstract
DNA amplification has intrigued scientists for decades. Since its discovery, significant progress has been made in understanding the mechanisms promoting DNA amplification and their associated function(s). While DNA copy gains were once thought to be regulated purely by stochastic processes, recent findings have revealed the important role of epigenetic modifications in driving these amplifications and their integration into the genome. Furthermore, advances in genomic technology have enabled detailed characterization of these genomic events in terms of size, structure, formation, and regulation. This review highlights how our understanding of DNA amplifications has evolved over time, tracing its trajectory from initial discovery to the contemporary landscape. We describe how recent discoveries have started to uncover how these genomic events occur by controlled biological processes rather than stochastic mechanisms, presenting opportunities for therapeutic modulation.
Collapse
Affiliation(s)
- Chloe Azadegan
- Drexel University, College of Medicine, Philadelphia, PA, 19111; Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111
| | - John Santoro
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111
| | - Johnathan R Whetstine
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111.
| |
Collapse
|
2
|
Chen T, Meng J, Yu K, Huang T, Zhao J. Chromatin Licensing and DNA Replication Factor 1 (CDT1) Is a Potential Prognostic Biomarker Involved in the Malignant Biological Behavior of Glioma. ACS Pharmacol Transl Sci 2024; 7:3131-3143. [PMID: 39416957 PMCID: PMC11475523 DOI: 10.1021/acsptsci.4c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
Glioma is the primary malignant tumor with the highest incidence rate in the adult central nervous system. The application of bioinformatics methods to analyze the RNA sequences of multiple gliomas revealed that the CDT1 gene has a significant impact on the cell cycle of glioma cells. Subsequently, we comprehensively and systematically investigated the expression of CDT1 in gliomas through bioinformatics analysis, clinical tissue specimens, and in vitro functional experiments. Our study is the first to report the expression of CDT1 in glioma. Our findings demonstrate that CDT1 plays a crucial role in the proliferation and invasion of glioma. Additionally, our bioinformatics analysis identified several other genes and signaling pathways that are dysregulated in multifocal gliomas, providing potential targets for further research and drug development.
Collapse
Affiliation(s)
- Tiange Chen
- Department
of Neurosurgery, Hainan General Hospital/Hainan
Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| | - Jiawei Meng
- Department
of Laboratory Medicine, The Third Xiangya
Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ke Yu
- Department
of Laboratory Medicine, The Third Xiangya
Hospital, Central South University, Changsha, Hunan 410013, China
| | - Tianxiang Huang
- Department
of Neurosurgery, and National Clinical Research Center of Geriatric
Disorders, Xiangya Hospital, Central South
University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Jiannong Zhao
- Department
of Neurosurgery, Hainan General Hospital/Hainan
Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| |
Collapse
|
3
|
Lin BQ, Chen F, Gu L, Wu ZX, Ye J, Zhang L, Huang BJ, Yu ZY, Lai GX, Lan XP, Zhao H, Liu W. CDT1, transcriptionally regulated by E2F2, promotes lung adenocarcinoma progression. Heliyon 2024; 10:e36557. [PMID: 39262963 PMCID: PMC11388403 DOI: 10.1016/j.heliyon.2024.e36557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
CDT1, a gene that shows excessive expression in various malignancies, functions as a pivotal regulator of replication licensing. In this study, we observed a positive correlation in expression between CDT1 and E2F2 among patients with lung adenocarcinoma (LUAD). Our findings substantiated that E2F2 directly interacted with the promoter region of CDT1, as confirmed by ChIP-qPCR assays, and depletion of E2F2 resulted in a downregulation of CDT1 expression in LUAD cell lines by gene interference technology. Furthermore, we identified an upregulation of CDT1 mRNA level in Chinese LUAD samples. Notably, in the loss-of-function assays, depletion of CDT1 in LUAD cell lines inhibited cell proliferation, migration, and invasion. Concurrently, it promoted cell apoptosis and induced G0/G1 phase arrest using MTT, flow cytometry, and Transwell assays, reinforcing its role as an oncogene.Furthermore, enhanced tumor ablation was determined in a CDT1-downregulated LUAD tumor-bearing nude mouse model. Collectively, our results strongly suggest that E2F2 positively regulates CDT1 expression and actively participates in the progression of lung adenocarcinoma, thereby providing valuable insights into identifying novel therapeutic targets for LUAD treatment.
Collapse
Affiliation(s)
- Bao-Quan Lin
- Cardio-Thoracic Surgery Department, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| | - Feng Chen
- Fuzong Teaching Hospital of Fujian University of Traditional Chinese Medicine (The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army), Fuzhou, Fujian, 350025, China
| | - Lei Gu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zai-Xin Wu
- Medical Service Management Office, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| | - Jia Ye
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital of Fujian University of Traditional Chinese Medicine (The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army), Fuzhou, Fujian, 350025, China
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| | - Lei Zhang
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital of Fujian University of Traditional Chinese Medicine (The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army), Fuzhou, Fujian, 350025, China
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| | - Bing-Jing Huang
- Department of Respiratory and Critical Care Medicine, Xiamen Haicang Hospital, No. 89 Haiyu Road, Xiamen, Fujian, 361026, China
| | - Zong-Yang Yu
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital of Fujian University of Traditional Chinese Medicine (The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army), Fuzhou, Fujian, 350025, China
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| | - Guo-Xiang Lai
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital of Fujian University of Traditional Chinese Medicine (The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army), Fuzhou, Fujian, 350025, China
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| | - Xiao-Peng Lan
- Institute for Laboratory Medicine, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fujian Medical University, Fuzhou, Fujian, 350025, China
| | - Hu Zhao
- Department of General Surgery, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian, 350025, China
- Department of General Surgery, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian, 350025, China
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital of Fujian University of Traditional Chinese Medicine (The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army), Fuzhou, Fujian, 350025, China
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army, Fuzhou, Fujian, 350025, China
| |
Collapse
|
4
|
Falbo L, Técher H, Sannino V, Robusto M, Fagà G, Pezzimenti F, Romeo F, Colombo LG, Vultaggio S, Fancelli D, Monzani S, Cecatiello V, Pasqualato S, Varasi M, Mercurio C, Costanzo V. A high-throughput screening identifies MCM chromatin loading inhibitors targeting cells with increased replication origins. iScience 2024; 27:110567. [PMID: 39184446 PMCID: PMC11342271 DOI: 10.1016/j.isci.2024.110567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/25/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Replication origin assembly is a pivotal step in chromosomal DNA replication. In this process, the ORC complex binds DNA and, together with the CDC6 and CDT1, promotes the loading of the MCM helicase. Chemicals targeting origin assembly might be useful to sensitize highly proliferative cancer cells. However, identifying such compounds is challenging due to the multistage nature of this process. Here, using Xenopus laevis egg extract we set up a high-throughput screening to isolate MCM chromatin loading inhibitors, which led to the identification of NSC-95397 as a powerful inhibitor of replication origin assembly that targets CDC6 protein and promotes its degradation. Using systems developed to test selective drug-induced lethality we show that NSC-95397 triggers cell death both in human cells and Xenopus embryos that have higher proliferative ability. These findings demonstrate the effectiveness of molecules disrupting DNA replication processes in targeting hyperproliferating cells, highlighting their potential as anti-cancer molecules.
Collapse
Affiliation(s)
- Lucia Falbo
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, 20133 Milan, Italy
| | - Hervé Técher
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Vincenzo Sannino
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Michela Robusto
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giovanni Fagà
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Francesco Romeo
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, 20133 Milan, Italy
| | | | | | - Daniele Fancelli
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Silvia Monzani
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Valentina Cecatiello
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Sebastiano Pasqualato
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Mario Varasi
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Ciro Mercurio
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Vincenzo Costanzo
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, 20133 Milan, Italy
| |
Collapse
|
5
|
Liao L, Han W, Shen Y, Shen G. Comprehensive analysis of aberrantly methylated differentially expressed genes and validation of CDC6 in melanoma. J Cancer Res Clin Oncol 2024; 150:362. [PMID: 39052109 PMCID: PMC11272740 DOI: 10.1007/s00432-024-05851-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Skin Cutaneous Melanoma (SKCM) is a highly aggressive malignant tumor with a significant increase in mortality upon metastasis. The molecular mechanisms driving melanoma progression remain largely unclear. Recent studies have highlighted the importance of epigenetic alterations, especially DNA methylation, in melanoma development. This study aims to identify and analyze methylation-regulated differentially expressed genes (MeDEGs) in genome-wide profiles between primary and metastatic melanoma. METHODS Gene expression profiling datasets GSE8401 and gene methylation profiling datasets GSE86355 were collected from the GEO database. Differentially expressed genes (DEGs) and differentially methylated genes (DMGs) were systematically identified. Integration of DEGs and DMGs yielded a set of MeDEGs, which subsequently underwent functional enrichment analysis. The protein-protein interaction (PPI) network was constructed using STRING and visualized using Cytoscape software. Survival analysis was used to select prognostic hub genes. In addition, 37 SKCM and 37 normal skin tissues from the First Affiliated Hospital of Soochow University (FAHSU) were collected for immunohistochemical (IHC) staining and evaluation. Furthermore, DNA methylation patterns of CDC6 were analyzed. To validate these findings, SKCM cell cultures were utilized to elucidate the expression and behavioral characteristics of CDC6. Additionally, gene set enrichment analysis (GSEA) and immune infiltration analysis were conducted for CDC6. RESULTS In our study, we discovered 120 hypomethylated-upregulated genes and 212 hypermethylated-downregulated genes. The hypomethylated-upregulated genes were notably associated with biological processes such as spindle assembly checkpoint signaling, mitotic spindle assembly, and negative regulation of mitotic metaphase/anaphase transition. Our pathway analysis revealed significant enrichment in pathways related to dilated cardiomyopathy, amino sugar metabolism, progesterone-mediated oocyte maturation, and chemical carcinogenesis. Conversely, hypermethylated-downregulated genes were found to be enriched in processes like epidermis development, keratinocyte differentiation, and skin development. Additionally, pathway analysis highlighted associations with estrogen signaling, Staphylococcus aureus infection, axon guidance, and arachidonic acid metabolism. Following the establishment of PPI networks and survival analysis, we identified 11 prognostic hub genes: CCNA2, CDC6, CDCA3, CKS2, DTL, HJURP, KRT5, KRT14, KRT15, KRT16, and NEK2. Notably, among the 11 hub genes, our findings indicate that CDC6 plays a pivotal role in enhancing the proliferation, migration, and invasion capabilities of melanoma cells in vitro. CONCLUSIONS Our comprehensive genomic analyses reveal that genes with aberrant methylation exhibit differential expression during the transition from primary to metastatic melanoma. The identified genes, especially CDC6, which plays a crucial role in enhancing melanoma cell proliferation, migration, and invasion, provide valuable insights into potential methylation-based biomarkers. These findings could contribute significantly to advancing precision medicine in SKCM.
Collapse
Affiliation(s)
- Li Liao
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215000, China
- Department of Cosmetic Dermatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, China
| | - Wei Han
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377, Munich, Germany
- Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Yue Shen
- Department of Cosmetic Dermatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, China
| | - Guoliang Shen
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215000, China.
| |
Collapse
|
6
|
Cui J, Liu X, Shang Q, Sun S, Chen S, Dong J, Zhu Y, Liu L, Xia Y, Wang Y, Xiang L, Fan B, Zhan J, Zhou Y, Chen P, Zhao R, Liu X, Xing N, Wu D, Shi B, Zou Y. Deubiquitination of CDC6 by OTUD6A promotes tumour progression and chemoresistance. Mol Cancer 2024; 23:86. [PMID: 38685067 PMCID: PMC11057083 DOI: 10.1186/s12943-024-01996-y] [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: 12/04/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND CDC6 is an oncogenic protein whose expression level fluctuates during the cell cycle. Although several E3 ubiquitin ligases responsible for the ubiquitin-mediated proteolysis of CDC6 have been identified, the deubiquitination pathway for CDC6 has not been investigated. METHODS The proteome-wide deubiquitinase (DUB) screening was used to identify the potential regulator of CDC6. Immunofluorescence, protein half-life and deubiquitination assays were performed to determine the protein stability of CDC6. Gain- and loss-of-function experiments were implemented to analyse the impacts of OUTD6A-CDC6 axis on tumour growth and chemosensitivity in vitro. N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced conditional Otud6a knockout (CKO) mouse model and tumour xenograft model were performed to analyse the role of OTUD6A-CDC6 axis in vivo. Tissue specimens were used to determine the association between OTUD6A and CDC6. RESULTS OTUD6A interacts with, depolyubiquitinates and stabilizes CDC6 by removing K6-, K33-, and K48-linked polyubiquitination. Moreover, OTUD6A promotes cell proliferation and decreases sensitivity to chemotherapy by upregulating CDC6. CKO mice are less prone to BCa tumorigenesis induced by BBN, and knockdown of OTUD6A inhibits tumour progression in vivo. Furthermore, OTUD6A protein level has a positive correlation with CDC6 protein level, and high protein levels of OTUD6A and CDC6 are associated with poor prognosis in patients with bladder cancer. CONCLUSIONS We reveal an important yet missing piece of novel DUB governing CDC6 stability. In addition, our findings propose a model for the OTUD6A-CDC6 axis that provides novel insights into cell cycle and chemosensitivity regulation, which may become a potential biomarker and promising drug target for cancer treatment.
Collapse
Affiliation(s)
- Jianfeng Cui
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaochen Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
- Department of Clinical laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Qinghong Shang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Shuna Sun
- Department of Dermatology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Jianping Dong
- Department of Urology, Shouguang People's Hospital, Weifang, Shandong, 262750, China
| | - Yaofeng Zhu
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Liu
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Yangyang Xia
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Yong Wang
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Lu Xiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Bowen Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Jiafeng Zhan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Yadi Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Renchang Zhao
- Department of Thoracic Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaofei Liu
- Departement of Breast and Thyroid Surgery, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Dalei Wu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China.
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China.
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
7
|
Liu Y, Han T, Xu Z, Wu J, Zhou J, Guo J, Miao R, Xing Y, Ge D, Bai Y, Hu D. CDC45 promotes the stemness and metastasis in lung adenocarcinoma by affecting the cell cycle. J Transl Med 2024; 22:335. [PMID: 38589907 PMCID: PMC11000299 DOI: 10.1186/s12967-024-05038-5] [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: 10/30/2023] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
OBJECTIVE This study aimed to assess the functions of cell division cycle protein 45 (CDC45) in Non-small cell lung cancer (NSCLC) cancer and its effects on stemness and metastasis. METHODS Firstly, differentially expressed genes related to lung cancer metastasis and stemness were screened by differential analysis and lasso regression. Then, in vitro, experiments such as colony formation assay, scratch assay, and transwell assay were conducted to evaluate the impact of CDC45 knockdown on the proliferation and migration abilities of lung cancer cells. Western blotting was used to measure the expression levels of related proteins and investigate the regulation of CDC45 on the cell cycle. Finally, in vivo model with subcutaneous injection of lung cancer cells was performed to verify the effect of CDC45 on tumor growth. RESULTS This study identified CDC45 as a key gene potentially influencing tumor stemness and lymph node metastasis. Knockdown of CDC45 not only suppressed the proliferation and migration abilities of lung cancer cells but also caused cell cycle arrest at the G2/M phase. Further analysis revealed a negative correlation between CDC45 and cell cycle-related proteins, stemness-related markers, and tumor mutations. Mouse experiments confirmed that CDC45 knockdown inhibited tumor growth. CONCLUSION As a novel regulator of stemness, CDC45 plays a role in regulating lung cancer cell proliferation, migration, and cell cycle. Therefore, CDC45 may serve as a potential target for lung cancer treatment and provide a reference for further mechanistic research and therapeutic development.
Collapse
Affiliation(s)
- Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Zhi Xu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
| | - Jing Wu
- Joint Research Center for Occupational Medicine and Health of IHM, School of Medicine, Anhui University of Science and Technology, Huainan, People's Republic of China.
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Rui Miao
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
| | - Yingru Xing
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
- Department of Clinical Laboratory, Anhui Zhongke Gengjiu Hospital, Hefei, People's Republic of China
| | - Deyong Ge
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China.
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, 232001, People's Republic of China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, People's Republic of China.
| |
Collapse
|
8
|
Wu H, Zhu P, Shu P, Zhang S. Screening and verification of hub genes in esophageal squamous cell carcinoma by integrated analysis. Sci Rep 2024; 14:6894. [PMID: 38519533 PMCID: PMC10959922 DOI: 10.1038/s41598-024-57320-7] [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: 11/04/2023] [Accepted: 03/17/2024] [Indexed: 03/25/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors. However, the mechanisms underlying ESCC tumorigenesis have not been fully elucidated. Thus, we aimed to determine the key genes involved in ESCC tumorigenesis. The following bioinformatics analyses were performed: identification of differentially expressed genes (DEGs); gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis; integrated analysis of the protein-protein interaction network and Gene Expression Profiling Interactive Analysis database for validation of hub genes. Finally, western blotting and qPCR were used to explore the expression of cell division cycle 6 (CDC6) in ESCC cell lines. Immunohistochemistry analysis of ESCC samples from patients and matched clinical characteristics was used to determine the effects of CDC6. A total of 494 DEGs were identified, and functional enrichment was mainly focused on cell cycle and DNA replication. Biological pathway analysis of the hub genes was closely related to the cell cycle. We found that CDC6 was upregulated in ESCC cell lines and patient tissues and was related to the clinicopathological characteristics of ESCC. In conclusion, this study identified hub genes and crucial biological pathways related to ESCC tumorigenesis and integrated analyses indicated that CDC6 may be a novel diagnostic and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Hongqiang Wu
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China
| | - Peiyao Zhu
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China
| | - Peng Shu
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China
| | - Shuguang Zhang
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang, 110001, China.
| |
Collapse
|
9
|
Shen M, Zhang Y, Tang L, Fu Q, Zhang J, Xu Y, Zeng H, Li Y. CDC6, a key replication licensing factor, is overexpressed and confers poor prognosis in diffuse large B-cell lymphoma. BMC Cancer 2023; 23:978. [PMID: 37833632 PMCID: PMC10571299 DOI: 10.1186/s12885-023-11186-6] [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: 09/10/2022] [Accepted: 07/17/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Cell division cycle 6 (CDC6) is a key licensing factor in the assembly of pre-replicative complexes at origins of replication. The role of CDC6 in the pathogenesis of in diffuse larger B-cell lymphoma (DLBCL) remains unknown. We aim to investigate the effects of CDC6 on the proliferation, apoptosis and cell cycle regulation in DLBCL cells, delineate its underlying mechanism, and to correlate CDC6 expression with clinical characteristics and prognosis of patients with DLBCL. METHODS Initial bioinformatic analysis was performed to screen the potential role of CDC6 in DLBCL. Lentiviral constructs harboring CDC6 or shCDC6 was transfected to overexpress or knockdown CDC6 in SUDHL4 and OCI-LY7 cells. The cell proliferation was evaluated by CCK-8 assay, cell apoptosis was detected by Annexin-V APC/7-AAD double staining, and cell cycle was measured by flow cytometry. Real time quantitative PCR and western blot was used to characterize CDC6 expression and its downstream signaling pathways. The clinical data of DLBCL patients were retrospectively reviewed, the CDC6 expression in DLBCL or lymph node reactive hyperplasia tissues was evaluated by immunohistochemistry. RESULTS In silico data suggest that CDC6 overexpression is associated with inferior prognosis of DLBCL. We found that CDC6 overexpression increased SUDHL4 or OCI-LY7 cell proliferation, while knockdown of CDC6 inhibited cell proliferation in a time-dependent manner. Upon overexpression, CDC6 reduced cells in G1 phase and did not affect cell apoptosis; CDC6 knockdown led to significant cell cycle arrest in G1 phase and increase in cell apoptosis. Western blot showed that CDC6 inhibited the expression of INK4, E-Cadherin and ATR, accompanied by increased Bcl-2 and deceased Bax expression. The CDC6 protein was overexpressed DLBCL compared with lymph node reactive hyperplasia, and CDC6 overexpression was associated with non-GCB subtype, and conferred poor PFS and OS in patients with DLBCL. CONCLUSION CDC6 promotes cell proliferation and survival of DLBCL cells through regulation of G1/S cell cycle checkpoint and apoptosis. CDC6 is overexpressed and serves as a novel prognostic marker in DLBCL.
Collapse
Affiliation(s)
- Mingfang Shen
- Department of Hematology, the First Hospital of Jiaxing, 314001, Zhejiang, China
| | - Yunfeng Zhang
- Department of Hematology, the First Hospital of Jiaxing, 314001, Zhejiang, China
| | - Lun Tang
- Department of Hematology, the First Hospital of Jiaxing, 314001, Zhejiang, China
| | - Qinyan Fu
- Department of Hematology, the First Hospital of Jiaxing, 314001, Zhejiang, China
| | - Jiawei Zhang
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Zhejiang, China
| | - Yang Xu
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Zhejiang, China
| | - Hui Zeng
- Department of Hematology, the First Hospital of Jiaxing, 314001, Zhejiang, China.
| | - Yuan Li
- Department of Hematology, the First Hospital of Jiaxing, 314001, Zhejiang, China.
| |
Collapse
|
10
|
Mourkioti I, Polyzou A, Veroutis D, Theocharous G, Lagopati N, Gentile E, Stravokefalou V, Thanos DF, Havaki S, Kletsas D, Panaretakis T, Logothetis CJ, Stellas D, Petty R, Blandino G, Papaspyropoulos A, Gorgoulis VG. A GATA2-CDC6 axis modulates androgen receptor blockade-induced senescence in prostate cancer. J Exp Clin Cancer Res 2023; 42:187. [PMID: 37507762 PMCID: PMC10386253 DOI: 10.1186/s13046-023-02769-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Prostate cancer is a major cause of cancer morbidity and mortality in men worldwide. Androgen deprivation therapy (ADT) has proven effective in early-stage androgen-sensitive disease, but prostate cancer gradually develops into an androgen-resistant metastatic state in the vast majority of patients. According to our oncogene-induced model for cancer development, senescence is a major tumor progression barrier. However, whether senescence is implicated in the progression of early-stage androgen-sensitive to highly aggressive castration-resistant prostate cancer (CRPC) remains poorly addressed. METHODS Androgen-dependent (LNCaP) and -independent (C4-2B and PC-3) cells were treated or not with enzalutamide, an Androgen Receptor (AR) inhibitor. RNA sequencing and pathway analyses were carried out in LNCaP cells to identify potential senescence regulators upon treatment. Assessment of the invasive potential of cells and senescence status following enzalutamide treatment and/or RNAi-mediated silencing of selected targets was performed in all cell lines, complemented by bioinformatics analyses on a wide range of in vitro and in vivo datasets. Key observations were validated in LNCaP and C4-2B mouse xenografts. Senescence induction was assessed by state-of-the-art GL13 staining by immunocytochemistry and confocal microscopy. RESULTS We demonstrate that enzalutamide treatment induces senescence in androgen-sensitive cells via reduction of the replication licensing factor CDC6. Mechanistically, we show that CDC6 downregulation is mediated through endogenous activation of the GATA2 transcription factor functioning as a CDC6 repressor. Intriguingly, GATA2 levels decrease in enzalutamide-resistant cells, leading to CDC6 stabilization accompanied by activation of Epithelial-To-Mesenchymal Transition (EMT) markers and absence of senescence. We show that CDC6 loss is sufficient to reverse oncogenic features and induce senescence regardless of treatment responsiveness, thereby identifying CDC6 as a critical determinant of prostate cancer progression. CONCLUSIONS We identify a key GATA2-CDC6 signaling axis which is reciprocally regulated in enzalutamide-sensitive and -resistant prostate cancer environments. Upon acquired resistance, GATA2 repression leads to CDC6 stabilization, with detrimental effects in disease progression through exacerbation of EMT and abrogation of senescence. However, bypassing the GATA2-CDC6 axis by direct inhibition of CDC6 reverses oncogenic features and establishes senescence, thereby offering a therapeutic window even after acquiring resistance to therapy.
Collapse
Affiliation(s)
- Ioanna Mourkioti
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Polyzou
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Veroutis
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George Theocharous
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nefeli Lagopati
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emanuela Gentile
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vasiliki Stravokefalou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635, Athens, Greece
| | - Dimitris-Foivos Thanos
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Havaki
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Aghia Paraskevi, Greece
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dimitris Stellas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635, Athens, Greece
| | - Russell Petty
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Giovanni Blandino
- Department of Research, Oncogenomic and Epigenetic Unit, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Angelos Papaspyropoulos
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| | - Vassilis G Gorgoulis
- Department of Histology and Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- Biomedical Research Foundation, Academy of Athens, Athens, Greece.
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.
| |
Collapse
|
11
|
Yang M, Wang P, Liu T, Zou X, Xia Y, Li C, Wang X. High throughput sequencing revealed enhanced cell cycle signaling in SLE patients. Sci Rep 2023; 13:159. [PMID: 36599883 DOI: 10.1038/s41598-022-27310-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
The multi-system involvement and high heterogeneity of systemic lupus erythematosus (SLE) pose great challenges to its diagnosis and treatment. The purpose of the current study is to identify genes and pathways involved in the pathogenesis of SLE. High throughput sequencing was performed on the PBMCs from SLE patients. We conducted differential gene analysis, gene ontology (GO) analysis, kyoto encyclopedia of genes and genomes (KEGG) analysis, and quantitative real-time PCR (qRT-PCR) verification. Protein-protein interaction (PPI) analysis, alternative splicing analysis, and disease correlation analysis were conducted on some key pathogenic genes as well. Furthermore, si-CDC6 was used for transfection and cell proliferation was monitored using a cell counting kit-8 (CCK-8) assay. We identified 2495 differential genes (1494 upregulated and 1001 downregulated) in SLE patients compared with healthy controls. The significantly upregulated genes were enriched in the biological process-related GO terms of the cell cycle, response to stress, and chromosome organization. KEGG enrichment analysis revealed 7 significantly upregulated pathways including SLE, alcoholism, viral carcinogenesis, cell cycle, proteasome, malaria, and transcriptional misregulation in cancer. We successfully verified some differential genes on the SLE pathway and the cell cycle pathway. CDC6, a key gene in the cell cycle pathway, had remarkably higher MXE alternative splicing events in SLE patients than that in controls, which may explain its significant upregulation in SLE patients. We found that CDC6 participates in the pathogenesis of many proliferation-related diseases and its levels are positively correlated with the severity of SLE. Knockdown of CDC6 suppressed the proliferation of Hela cells and PBMCs from SLE patients in vitro. We identified SLE-related genes and their alternative splicing events. The cell cycle pathway and the cell cycle-related biological processes are over-activated in SLE patients. We revealed a higher incidence of MXE events of CDC6, which may lead to its high expression in SLE patients. Upregulated cell cycle signaling and CDC6 may be related to the hyperproliferation and pathogenesis of SLE.
Collapse
Affiliation(s)
- Mingyue Yang
- Laboratory for Tumor Immunology, Translational Medicine Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Peisong Wang
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Tao Liu
- Department of Rheumatology and Immunology, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaojuan Zou
- Department of Rheumatology and Immunology, First Hospital of Jilin University, Changchun, 130021, China
| | - Ying Xia
- Laboratory for Tumor Immunology, Translational Medicine Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Chenxu Li
- Laboratory for Tumor Immunology, Translational Medicine Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaosong Wang
- Laboratory for Tumor Immunology, Translational Medicine Department, First Hospital of Jilin University, Changchun, 130021, China.
- Institute of Translational Medicine, First Hospital of Jilin University, No.519 Dongminzhu Street, Changchun, 130021, China.
| |
Collapse
|
12
|
Li X, Abdel-Maksoud MA, Iqbal I, Mubarak A, Farrag MA, Haris M, Alghamdi S, Ain QU, Almekhlafi S. Deciphering cervical cancer-associated biomarkers by integrated multi-omics approach. Am J Transl Res 2022; 14:8843-8861. [PMID: 36628250 PMCID: PMC9827308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/13/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Cervical Squamous Cell Carcinoma (CESC) is one of the most fatal female malignancies, and the underlying molecular mechanisms governing this disease have not been fully explored. In this research, we planned to conduct the analysis of Gene Expression Omnibus (GEO) cervical squamous cell carcinoma microarray datasets by a detailed in silico approach and to explore some novel biomarkers of CESC. METHODS The top commonly differentially expressed genes (DEGs) from the GSE138080 and GSE113942 datasets were analyzed by Limma package-based GEO2R tool. The protein-protein interaction (PPI) network of the DEGs was drawn through Search Tool for the Retrieval of Interacting Genes (STRING), and top 6 hub genes were obtained from Cytoscape. Expression analysis and validation of hub genes expression in CESC samples and cell lines were done using UALCAN, OncoDB, GENT2, and HPA. Additionally, cBioPortal, Gene set enrichment analysis (GSEA) tool, Kaplan-Meier (KM) plotter, ShinyGO, and DGIdb databases were also used to check some important values of hub genes in CESC. RESULTS Out of 79 DEGs, the minichromosome maintenance complex component 4 (MCM4), nucleolar and spindle-associated protein 1 (NUSAP1), cell division cycle associated 5 (CDCA5), cell division cycle 45 (CDC45), denticleless E3 ubiquitin protein ligase homolog (DTL), and chromatin licensing and DNA replication factor 1 (CDT1) genes were regarded as hub genes in CESC. Further analysis revealed that the expressions of all these hub genes were significantly elevated in CESC cell lines and samples of diverse clinical attributes. In this study, we also documented some important correlations between hub genes and some other diverse measures, including DNA methylation, genetic alterations, and Overall Survival (OS). Last, we also identify hub genes associated ceRNA network and 31 important chemotherapeutic drugs. CONCLUSION Through detailed in silico methodology, we identified 6 hub genes, including MCM4, NUSAP1, CDCA5, CDC45, DTL, and CDT1, which are likely to be associated with CESC development and diagnosis.
Collapse
Affiliation(s)
- Xuhong Li
- Department of Gynaecology and Obstetrics, Shanghai Eighth People’s HospitalShanghai, China
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Iqra Iqbal
- Azra Naheed Medical CollegeLahore, Pakistan
| | - Ayman Mubarak
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Mohamed A Farrag
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Muhammad Haris
- Department of Anatomy, Institute of Basic Medical Sciences, Khyber Medical UniversityPeshawar, Pakistan
| | - Sumaiah Alghamdi
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Qurat Ul Ain
- Anhui Provincial Hospital, Division of Life Science and Medicine, University of Science and Technology ChinaHefei, Anhui, China
| | - Sally Almekhlafi
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
13
|
Yan Z, Liu Q, Cao Z, Wang J, Zhang H, Liu J, Zou L. Multi-omics integration reveals a six-malignant cell maker gene signature for predicting prognosis in high-risk neuroblastoma. Front Neuroinform 2022; 16:1034793. [DOI: 10.3389/fninf.2022.1034793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
BackgroundNeuroblastoma is the most common extracranial solid tumor of childhood, arising from the sympathetic nervous system. High-risk neuroblastoma (HRNB) remains a major therapeutic challenge with low survival rates despite the intensification of therapy. This study aimed to develop a malignant-cell marker gene signature (MMGS) that might serve as a prognostic indicator in HRNB patients.MethodsMulti-omics datasets, including mRNA expression (single-cell and bulk), DNA methylation, and clinical information of HRNB patients, were used to identify prognostic malignant cell marker genes. MMGS was established by univariate Cox analysis, LASSO, and stepwise multivariable Cox regression analysis. Kaplan–Meier (KM) curve and time-dependent receiver operating characteristic curve (tROC) were used to evaluate the prognostic value and performance of MMGS, respectively. MMGS further verified its reliability and accuracy in the independent validation set. Finally, the characteristics of functional enrichment, tumor immune features, and inflammatory activity between different MMGS risk groups were also investigated.ResultsWe constructed a prognostic model consisting of six malignant cell maker genes (MAPT, C1QTNF4, MEG3, NPW, RAMP1, and CDT1), which stratified patients into ultra-high-risk (UHR) and common-high-risk (CHR) group. Patients in the UHR group had significantly worse overall survival (OS) than those in the CHR group. MMGS was verified as an independent predictor for the OS of HRNB patients. The area under the curve (AUC) values of MMGS at 1-, 3-, and 5-year were 0.78, 0.693, and 0.618, respectively. Notably, functional enrichment, tumor immune features, and inflammatory activity analyses preliminarily indicated that the poor prognosis in the UHR group might result from the dysregulation of the metabolic process and immunosuppressive microenvironment.ConclusionThis study established a novel six-malignant cell maker gene prognostic model that can be used to predict the prognosis of HRNB patients, which may provide new insight for the treatment and personalized monitoring of HRNB patients.
Collapse
|
14
|
Karantzelis N, Petropoulos M, De Marco V, Egan DA, Fish A, Christodoulou E, Will DW, Lewis JD, Perrakis A, Lygerou Z, Taraviras S. Small Molecule Inhibitor Targeting CDT1/Geminin Protein Complex Promotes DNA Damage and Cell Death in Cancer Cells. Front Pharmacol 2022; 13:860682. [PMID: 35548337 PMCID: PMC9083542 DOI: 10.3389/fphar.2022.860682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/30/2022] [Indexed: 01/18/2023] Open
Abstract
DNA replication initiation requires the loading of MCM2-7 complexes at the origins of replication during G1. Replication licensing renders chromatin competent for DNA replication and its tight regulation is essential to prevent aberrant DNA replication and genomic instability. CDT1 is a critical factor of licensing and its activity is controlled by redundant mechanisms, including Geminin, a protein inhibitor of CDT1. Aberrant CDT1 and Geminin expression have been shown to promote tumorigenesis in vivo and are also evident in multiple human tumors. In this study, we developed an in vitro AlphaScreen™ high-throughput screening (HTS) assay for the identification of small-molecule inhibitors targeting the CDT1/Geminin protein complex. Biochemical characterization of the most potent compound, AF615, provided evidence of specific, dose-dependent inhibition of Geminin binding to CDT1 both in-vitro and in cells. Moreover, compound AF615 induces DNA damage, inhibits DNA synthesis and reduces viability selectively in cancer cell lines, and this effect is CDT1-dependent. Taken together, our data suggest that AF615 may serve as a useful compound to elucidate the role of CDT1/Geminin protein complex in replication licensing and origin firing as well as a scaffold for further medicinal chemistry optimisation.
Collapse
Affiliation(s)
| | - Michalis Petropoulos
- Department of General Biology, Medical School, University of Patras, Patras, Greece
| | - Valeria De Marco
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - David A Egan
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alexander Fish
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - David W Will
- Chemical Biology Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Joe D Lewis
- Chemical Biology Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Anastassis Perrakis
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Zoi Lygerou
- Department of General Biology, Medical School, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
15
|
Shen Y, Ye H, Zhang D, Yang M, Ji Y, Tang L, Zhu X, Yuan L. The role of exosomal CDC6 in the hirudin-mediated suppression of the malignant phenotype of bladder cancer cells. Gene 2022; 821:146269. [PMID: 35150820 DOI: 10.1016/j.gene.2022.146269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/22/2021] [Accepted: 01/13/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Bladder cancer is a malignant tumor characterized by high recurrence and persistence due to the limited therapies that are currently available. Hirudin exerts a strong anticancer effect on several tumors. Thus, it is urgent to explore the biological function of hirudin in bladder cancer and the role of bladder cancer-derived exosomes in tumor inhibition. METHODS First, a network pharmacology analysis was performed to explore the relationships among hirudin, bladder cancer, and exosomes. Then, the effects of hirudin were examined by CCK-8 assay, flow cytometry, Transwell assay, and tumorigenic ability experiments in vitro. Exosomes derived from cells were identified with transmission electron microscopy, fluorescence labeling, and Western blotting and collected for further microarray analysis. Only CDC6 expression and mRNA abundance in hirudin-treated cells and exosomes was subjected to further validation using quantitative PCR and Western blotting. RESULTS Through network analysis, we found that hirudin affected bladder cancer, and this effect was related to exosomes. Our studies verified the effects of hirudin by revealing that hirudin inhibits malignant processes of bladder cancer cells in vitro, such as invasion, metastasis, and apoptosis. Similarly, the oncogenic effects of bladder cancer-derived exosomes were successfully isolated and identified. Via microarray assessment of the exosomes, we identified 600 differential mRNAs, of which the expression of the core target CDC6 was found to be significantly different in both The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. We further confirmed that hirudin suppresses CDC6 expression mRNA abundance in both cells and exosomes. CONCLUSION Hirudin was able to decrease the expression of CDC6 in bladder cancer cells and exosomes, which effectively repressed the malignant processes of bladder cancer cells.
Collapse
Affiliation(s)
- Yang Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Hesong Ye
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Dongjian Zhang
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ming Yang
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Yuanyuan Ji
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Longlong Tang
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Xudong Zhu
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Lin Yuan
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
16
|
Zampetidis CP, Galanos P, Angelopoulou A, Zhu Y, Polyzou A, Karamitros T, Kotsinas A, Lagopati N, Mourkioti I, Mirzazadeh R, Polyzos A, Garnerone S, Mizi A, Gusmao EG, Sofiadis K, Gál Z, Larsen DH, Pefani DE, Demaria M, Tsirigos A, Crosetto N, Maya-Mendoza A, Papaspyropoulos A, Evangelou K, Bartek J, Papantonis A, Gorgoulis VG. A recurrent chromosomal inversion suffices for driving escape from oncogene-induced senescence via subTAD reorganization. Mol Cell 2021; 81:4907-4923.e8. [PMID: 34793711 DOI: 10.1016/j.molcel.2021.10.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 07/14/2021] [Accepted: 10/16/2021] [Indexed: 12/12/2022]
Abstract
Oncogene-induced senescence (OIS) is an inherent and important tumor suppressor mechanism. However, if not removed timely via immune surveillance, senescent cells also have detrimental effects. Although this has mostly been attributed to the senescence-associated secretory phenotype (SASP) of these cells, we recently proposed that "escape" from the senescent state is another unfavorable outcome. The mechanism underlying this phenomenon remains elusive. Here, we exploit genomic and functional data from a prototypical human epithelial cell model carrying an inducible CDC6 oncogene to identify an early-acquired recurrent chromosomal inversion that harbors a locus encoding the circadian transcription factor BHLHE40. This inversion alone suffices for BHLHE40 activation upon CDC6 induction and driving cell cycle re-entry of senescent cells, and malignant transformation. Ectopic overexpression of BHLHE40 prevented induction of CDC6-triggered senescence. We provide strong evidence in support of replication stress-induced genomic instability being a causative factor underlying "escape" from oncogene-induced senescence.
Collapse
Affiliation(s)
- Christos P Zampetidis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiotis Galanos
- Genome Integrity Group, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark.
| | - Andriani Angelopoulou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Yajie Zhu
- Translational Epigenetics Group, Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Aikaterini Polyzou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Timokratis Karamitros
- Unit of Bioinformatics and Applied Genomics, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Athanassios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nefeli Lagopati
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioanna Mourkioti
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Reza Mirzazadeh
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Stockholm, Sweden
| | - Alexandros Polyzos
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Silvano Garnerone
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Stockholm, Sweden
| | - Athanasia Mizi
- Translational Epigenetics Group, Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Eduardo G Gusmao
- Translational Epigenetics Group, Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Konstantinos Sofiadis
- Translational Epigenetics Group, Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Zita Gál
- Nucleolar Stress and Disease Group, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Dorthe H Larsen
- Nucleolar Stress and Disease Group, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | | | - Marco Demaria
- University of Groningen (RUG), European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), 9713 AV Groningen, the Netherlands
| | | | - Nicola Crosetto
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Stockholm, Sweden
| | - Apolinar Maya-Mendoza
- DNA Replication and Cancer Group, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Angelos Papaspyropoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Jiri Bartek
- Genome Integrity Group, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Stockholm, Sweden.
| | - Argyris Papantonis
- Translational Epigenetics Group, Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, University of Manchester, M20 4GJ Manchester, UK; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK.
| |
Collapse
|
17
|
Andriessen A, Bongiovanni L, Driedonks TAP, van Liere E, Seijger A, Hegeman CV, van Nimwegen SA, Galac S, Westendorp B, Nolte-'t Hoen ENM, de Bruin A. CDC6: A novel canine tumour biomarker detected in circulating extracellular vesicles. Vet Comp Oncol 2021; 20:381-392. [PMID: 34743398 PMCID: PMC9299066 DOI: 10.1111/vco.12781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022]
Abstract
Circulating nucleic acids and extracellular vesicles (EV) represent novel biomarkers to diagnose cancer. The non‐invasive nature of these so‐called liquid biopsies provides an attractive alternative to tissue biopsy‐based cancer diagnostics. This study aimed to investigate if circulating cell cycle‐related E2F target transcripts can be used to diagnose tumours in canine tumour patients with different types of tumours. Furthermore, we assessed if these mRNAs are localised within circulating EV. We isolated total RNA from the plasma of 20 canine tumour patients and 20 healthy controls. Four E2F target genes (CDC6, DHFR, H2AFZ and ATAD2) were selected based on the analysis of published data of tumour samples available in public databases. We performed reverse transcription and quantitative real‐time PCR to analyse the plasma levels of selected E2F target transcripts. All four E2F target transcripts were detectable in the plasma of canine tumour patients. CDC6 mRNA levels were significantly higher in the plasma of canine tumour patients compared to healthy controls. A subset of canine tumour patient and healthy control plasma samples (n = 7) were subjected to size exclusion chromatography in order to validate association of the E2F target transcripts to circulating EV. For CDC6, EV analysis enhanced their detectability compared to total plasma analysis. In conclusion, our study reveals circulating CDC6 as a promising non‐invasive biomarker to diagnose canine tumours.
Collapse
Affiliation(s)
- Anneloes Andriessen
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Laura Bongiovanni
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tom A P Driedonks
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Elsbeth van Liere
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anne Seijger
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Charlotte V Hegeman
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sebastiaan A van Nimwegen
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sara Galac
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Bart Westendorp
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Esther N M Nolte-'t Hoen
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Alain de Bruin
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
18
|
Cai C, Zhang Y, Hu X, Hu W, Yang S, Qiu H, Chu T. CDT1 Is a Novel Prognostic and Predictive Biomarkers for Hepatocellular Carcinoma. Front Oncol 2021; 11:721644. [PMID: 34631549 PMCID: PMC8497762 DOI: 10.3389/fonc.2021.721644] [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: 06/07/2021] [Accepted: 09/03/2021] [Indexed: 01/11/2023] Open
Abstract
Objective Hepatocellular carcinoma (HCC) is one of the most common malignant tumors endangering human health and life in the 21st century. Chromatin licensing and DNA replication factor 1 (CDT1) is an important regulator of DNA replication licensing, which is essential for initiation of DNA replication. CDT1 overexpression in several human cancers reportedly leads to abnormal cell replication, activates DNA damage checkpoints, and predisposes malignant transformation. However, the abnormal expression of CDT1 in HCC and its diagnostic and prognostic value remains to be elucidated. Methods TCGA, ONCOMINE, UALCAN, HCCDB, HPA, Kaplan-Meier plotter, STRING, GEPIA, GeneMANIA, and TIMER were conducted for bioinformatics analysis. CDT1 protein expression was evaluated by immunohistochemistry in HCC tissues through a tissue microarray. qRT-PCR, western blot and a cohort of functional experiments were performed for in vitro validation. Results In this study, we discovered remarkably upregulated transcription of CDT1 in HCC samples relative to normal liver samples through bioinformatic analysis, which was further verified in clinical tissue microarray samples and in vitro experiments. Moreover, the transcriptional level of CDT1 in HCC samples was positively associated with clinical parameters such as clinical tumor stage. Survival, logistic regression, and Cox regression analyses revealed the significant clinical prognostic value of CDT1 expression in HCC. The receiver operating characteristic curve and nomogram analysis results demonstrated the strong predictive ability of CDT1 in HCC. Kyoto Encyclopedia of Genes and Genomes and gene set enrichment analyses indicated that CDT1 was mainly associated with the cell cycle, DNA repair, and DNA replication. We further demonstrated the significant correlation between CDT1 and minichromosome maintenance (MCM) family genes, revealing abnormal expression and prognostic significance of MCMs in HCC. Immune infiltration analysis indicated that CDT1 was significantly associated with immune cell subsets and affected the survival of HCC patients. Finally, knockdown of CDT1 decreased, whereas overexpression of CDT1 promoted the proliferation, migration, invasion of HCC cells in vitro. Conclusions Our study findings demonstrate the potential diagnostic and prognostic significance of CDT1 expression in HCC, and elucidate the potential molecular mechanism underlying its role in promoting the occurrence and development of liver cancer. These results may provide new opportunities and research paths for targeted therapies in HCC.
Collapse
Affiliation(s)
- Chenhui Cai
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xu Hu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Sizhen Yang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Qiu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tongwei Chu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
19
|
Shoji H, Takahari D, Hara H, Nagashima K, Adachi J, Boku N. A Phase I study of pevonedistat plus capecitabine plus oxaliplatin in patients with advanced gastric cancer refractory to platinum (NCCH-1811). Future Sci OA 2021; 7:FSO721. [PMID: 34258028 PMCID: PMC8256327 DOI: 10.2144/fsoa-2021-0023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023] Open
Abstract
Based on synergistic anti-tumor effects between blockades of NEDD8 activating enzyme and a platinum in preclinical studies, this Phase I study is designed to investigate the safety and tolerability of pevonedistat in combination with capecitabine plus oxaliplatin as third-line or later treatment in patients with unresectable advanced/recurrent gastric cancer who were previously treated with fluoropyrimidines and platinum (cisplatin or oxaliplatin) as the first-line treatment and paclitaxel (including nab-paclitaxel) as the second-line treatment. The aim of this trial is to determine the recommended dose of pevonedistat and to see its pharmacokinetics in combination with capecitabine plus oxaliplatin in the dose-finding part and explore its efficacy and safety in the expansion part. Trial registration number: jRCT2031190020 (jRCTs: the Japan Registry of Clinical Trials).
Collapse
Affiliation(s)
- Hirokazu Shoji
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Daisuke Takahari
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-550, Japan
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, 780 Komuro, Inamachi, Kitaadachi-gun, Saitama, 362-0806, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Jun Adachi
- Laboratory of Clinical & Analytical Chemistry, National Institute of Biomedical Innovation, Health & Nutrition, Ibaraki, Osaka, 567-0085, Japan.,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health & Nutrition, Ibaraki, Osaka, 567-0085, Japan
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
20
|
Rapsomaniki MA, Maxouri S, Nathanailidou P, Garrastacho MR, Giakoumakis NN, Taraviras S, Lygeros J, Lygerou Z. In silico analysis of DNA re-replication across a complete genome reveals cell-to-cell heterogeneity and genome plasticity. NAR Genom Bioinform 2021; 3:lqaa112. [PMID: 33554116 PMCID: PMC7846089 DOI: 10.1093/nargab/lqaa112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/15/2020] [Accepted: 01/20/2021] [Indexed: 01/06/2023] Open
Abstract
DNA replication is a complex and remarkably robust process: despite its inherent uncertainty, manifested through stochastic replication timing at a single-cell level, multiple control mechanisms ensure its accurate and timely completion across a population. Disruptions in these mechanisms lead to DNA re-replication, closely connected to genomic instability and oncogenesis. Here, we present a stochastic hybrid model of DNA re-replication that accurately portrays the interplay between discrete dynamics, continuous dynamics and uncertainty. Using experimental data on the fission yeast genome, model simulations show how different regions respond to re-replication and permit insight into the key mechanisms affecting re-replication dynamics. Simulated and experimental population-level profiles exhibit a good correlation along the genome, robust to model parameters, validating our approach. At a single-cell level, copy numbers of individual loci are affected by intrinsic properties of each locus, in cis effects from adjoining loci and in trans effects from distant loci. In silico analysis and single-cell imaging reveal that cell-to-cell heterogeneity is inherent in re-replication and can lead to genome plasticity and a plethora of genotypic variations.
Collapse
Affiliation(s)
- Maria Anna Rapsomaniki
- Department of Biology, School of Medicine, University of Patras, 26500 Rio Patras, Greece
| | - Stella Maxouri
- Department of Biology, School of Medicine, University of Patras, 26500 Rio Patras, Greece
| | - Patroula Nathanailidou
- Department of Biology, School of Medicine, University of Patras, 26500 Rio Patras, Greece
| | | | | | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, 26500 Rio Patras, Greece
| | - John Lygeros
- Automatic Control Laboratory, ETH Zurich, 8092 Zurich, Switzerland
| | - Zoi Lygerou
- Department of Biology, School of Medicine, University of Patras, 26500 Rio Patras, Greece
| |
Collapse
|
21
|
Xu H, Huang J, Hua S, Liang L, He X, Zhan M, Lu L, Chu J. Interactome analysis of gene expression profiles identifies CDC6 as a potential therapeutic target modified by miR-215-5p in hepatocellular carcinoma. Int J Med Sci 2020; 17:2926-2940. [PMID: 33173413 PMCID: PMC7646103 DOI: 10.7150/ijms.51145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/07/2020] [Indexed: 01/11/2023] Open
Abstract
Background: Illustrating the pathogenesis of hepatocellular carcinoma (HCC) pathogenesis as well as identifying specific biomarkers are of great significance. Methods: The original CEL files were obtain from Gene Expression Omnibus, then affymetrix package was used to preprocess the CEL files, the function of DEGs were investigated by multiple bioinformatics approach. Finally, typical HCC cell lines and tissue samples were using to validate the role of CDC6 in vitro. Bioinformatics software was used to predict potential microRNA of CDC6. Luciferase assay was used to verify the interactions between CDC6 and microRNA. Results: A total of 445 DEGs were identified in HCC tissues based on two GEO datasets. GSEA results showed that the significant enriched gene sets were only associated with cell cycle signaling pathway. In the co-expression analysis, there were 370 hub genes from the blue modules were screened. We integrated DEGs, hub genes, TCGA cohort and GSEA analyses to further obtain 10 upregulated genes for validation. These genes were overexpressed in HCC tissues and negatively associated with overall and disease-free survival in HCC patients and related to immune cell infiltration in HCC microenvironments. Finally, Cell Division Cycle 6 (CDC6) was highlighted as one of the most probable genes among the 10 candidates participating in cancer process. The expression of CDC6 either in public datasets and HCC tissues sample were commonly high than the non-cancerous counterpart. Furthermore, we recognized that miR-215-5p, could directly bind to the 3'UTR of CDC6. In addition, CDC6 promoted proliferation via regulation of G1 phase checkpoint and was negative regulated by miR-215-5p to involve in the proliferation of HCC. Conclusion: Our study suggested that CDC6 served as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Hongfa Xu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Jianwen Huang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Shengni Hua
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Linjun Liang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Xu He
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Meixiao Zhan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Jing Chu
- Department of Urology, Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| |
Collapse
|
22
|
Xiong G, Diao D, Lu D, Liu X, Liu Z, Mai S, Feng S, Dong X, Cai K. Circular RNA circNELL2 Acts as the Sponge of miR-127-5p to Promote Esophageal Squamous Cell Carcinoma Progression. Onco Targets Ther 2020; 13:9245-9255. [PMID: 32982311 PMCID: PMC7502390 DOI: 10.2147/ott.s247847] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction Owing to its involvement in both the initiation and progression of various cancers, aberrant circular RNA (circRNA) expression has been researched extensively in the recent times. In the present study, we aim to investigate the effect of a novel circRNA has_circ_0025933 (circNELL2) in the progression of esophageal squamous cell carcinoma (ESCC). Materials and Methods Sanger sequencing and the detection of circNELL2 level after RNase R or actinomycin D treatment were performed to identify the existence of cirNELL2 in ESCC cells. WST, EDU staining and colony-formation assay were used to assess the proliferation while transwell assay was used to evaluate the migration of ESCC cells. Luciferase assay, RNA pull down and the FISH assay were performed to verify the interaction between circNELL2 and miR-127-5p as well as miR-127-5p and CDC6. Xenograft model was carried out to evaluate the effect of circNELL2 in vivo. Results circNELL2 was proved to exist in ESCC cells. The up-regulated expression of circNELL2 in the clinical ESCC specimens was also verified. Next, function studies suggested that circNELL2 knockdown inhibited the proliferation of ESCC cells in vitro and in vivo, while circNELL2 overexpression promotes that of ESCC cells. Besides, this study mechanically predicted and verified the target miR of circNELL2, which is miR-127-5p. It was found that miR-127-5p was capable of reversing the effect of circNELL2 on ESCC cells. Moreover, miR-127-5p was also found to target CDC6 to participate in the regulation of cell phenotype. Discussion circNELL2 promoted the progression of ESCC cells via sponging miR-127-5p, and it has the potential to serve as a novel prognostic and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Gang Xiong
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Dingwei Diao
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Di Lu
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Xiguang Liu
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Zhaoguo Liu
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Shijie Mai
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Siyang Feng
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Xiaoying Dong
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| | - Kaican Cai
- Department of Thoracic Surgery, Southern Medical University Nanfang Hospital, Guangzhou City, Guangdong Province 510515, People's Republic of China
| |
Collapse
|
23
|
Menzel J, Tatman P, Black JC. Isolation and analysis of rereplicated DNA by Rerep-Seq. Nucleic Acids Res 2020; 48:e58. [PMID: 32239215 PMCID: PMC7261181 DOI: 10.1093/nar/gkaa197] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/05/2020] [Accepted: 03/16/2020] [Indexed: 01/31/2023] Open
Abstract
Changes in gene copy number contribute to genomic instability, the onset and progression of cancer, developmental abnormalities and adaptive potential. The origins of gene amplifications have remained elusive; however, DNA rereplication has been implicated as a source of gene amplifications. The inability to determine which sequences are rereplicated and under what conditions have made it difficult to determine the validity of the proposed models. Here we present Rerep-Seq, a technique that selectively enriches for rereplicated DNA in preparation for analysis by DNA sequencing that can be applied to any species. We validated Rerep-Seq by simulating DNA rereplication in yeast and human cells. Using Rerep-Seq, we demonstrate that rereplication induced in Saccharomyces cerevisiae by deregulated origin licensing is non-random and defined by broad domains that span multiple replication origins and topological boundaries.
Collapse
Affiliation(s)
- Johannes Menzel
- University of Colorado Anschutz Medical Campus, Department of Pharmacology, 12800 E 19th Ave, Aurora, CO 80045, USA.,University of Colorado Anschutz Medical Campus, Molecular Biology Graduate Program, 12800 E 19th Ave, Aurora, CO 80045, USA
| | - Philip Tatman
- University of Colorado Anschutz Medical Campus, Department of Pharmacology, 12800 E 19th Ave, Aurora, CO 80045, USA.,University of Colorado Anschutz Medical Campus, Medical Scientist Training Program, 12800 E 19th Ave, Aurora, CO 80045, USA
| | - Joshua C Black
- University of Colorado Anschutz Medical Campus, Department of Pharmacology, 12800 E 19th Ave, Aurora, CO 80045, USA.,University of Colorado Anschutz Medical Campus, Molecular Biology Graduate Program, 12800 E 19th Ave, Aurora, CO 80045, USA.,University of Colorado Anschutz Medical Campus, Medical Scientist Training Program, 12800 E 19th Ave, Aurora, CO 80045, USA
| |
Collapse
|
24
|
VanGenderen C, Harkness TAA, Arnason TG. The role of Anaphase Promoting Complex activation, inhibition and substrates in cancer development and progression. Aging (Albany NY) 2020; 12:15818-15855. [PMID: 32805721 PMCID: PMC7467358 DOI: 10.18632/aging.103792] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
The Anaphase Promoting Complex (APC), a multi-subunit ubiquitin ligase, facilitates mitotic and G1 progression, and is now recognized to play a role in maintaining genomic stability. Many APC substrates have been observed overexpressed in multiple cancer types, such as CDC20, the Aurora A and B kinases, and Forkhead box M1 (FOXM1), suggesting APC activity is important for cell health. We performed BioGRID analyses of the APC coactivators CDC20 and CDH1, which revealed that at least 69 proteins serve as APC substrates, with 60 of them identified as playing a role in tumor promotion and 9 involved in tumor suppression. While these substrates and their association with malignancies have been studied in isolation, the possibility exists that generalized APC dysfunction could result in the inappropriate stabilization of multiple APC targets, thereby changing tumor behavior and treatment responsiveness. It is also possible that the APC itself plays a crucial role in tumorigenesis through its regulation of mitotic progression. In this review the connections between APC activity and dysregulation will be discussed with regards to cell cycle dysfunction and chromosome instability in cancer, along with the individual roles that the accumulation of various APC substrates may play in cancer progression.
Collapse
Affiliation(s)
- Cordell VanGenderen
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Troy Anthony Alan Harkness
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Terra Gayle Arnason
- Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.,Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
25
|
Lim N, Townsend PA. Cdc6 as a novel target in cancer: Oncogenic potential, senescence and subcellular localisation. Int J Cancer 2020; 147:1528-1534. [PMID: 32010971 PMCID: PMC7496346 DOI: 10.1002/ijc.32900] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/22/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
Cdc6 is a key replication licencing factor with a pivotal role in regulating the process of DNA replication, rendering it an important investigatory focus in tumourigenesis. Indeed, Cdc6 overexpression has been found to be a feature in certain tumours and has been associated as an early event in malignancies. With a focus on pancreatic cancer, there are evidence of its convergence in downstream pathways implicated in major genetic alterations found in pancreatic cancer, primarily KRAS. There is also data of its direct influence on protumourigenic processes as a transcriptional regulator, repressing the key tumour suppressor loci CDH1 (E‐Cadherin) and influencing epithelial to mesenchymal transition (EMT). Moreover, gene amplification of Cdc6 as well as of E2F (an upstream regulator of Cdc6) have also been found to be a key feature in tumours overexpressing Cdc6, further highlighting this event as a potential driver of tumourigenesis. In this review, we summarise the evidence for the role of Cdc6 overexpression in cancer, specifically that of pancreatic cancer. More importantly, we recapitulate the role of Cdc6 as part of the DNA damage response and on senescence—an important antitumour barrier—in the context of pancreatic cancer. Finally, recent emerging observations suggest that the potential of the subcellular localisation of Cdc6 in inducing senescence. In this regard, we speculate and hypothesise potentially exploitable mechanisms in the context of inducing senescence via a novel pathway involving cytoplasmic retention of Cdc6 and Cyclin E.
Collapse
Affiliation(s)
- Nicholas Lim
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester Cancer Research Centre, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, United Kingdom
| | - Paul A Townsend
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester Cancer Research Centre, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, United Kingdom.,Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
26
|
Machine learning and data mining frameworks for predicting drug response in cancer: An overview and a novel in silico screening process based on association rule mining. Pharmacol Ther 2019; 203:107395. [DOI: 10.1016/j.pharmthera.2019.107395] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
|
27
|
Petropoulos M, Champeris Tsaniras S, Taraviras S, Lygerou Z. Replication Licensing Aberrations, Replication Stress, and Genomic Instability. Trends Biochem Sci 2019; 44:752-764. [PMID: 31054805 DOI: 10.1016/j.tibs.2019.03.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/24/2019] [Accepted: 03/27/2019] [Indexed: 01/07/2023]
Abstract
Strict regulation of DNA replication is of fundamental significance for the maintenance of genome stability. Licensing of origins of DNA replication is a critical event for timely genome duplication. Errors in replication licensing control lead to genomic instability across evolution. Here, we present accumulating evidence that aberrant replication licensing is linked to oncogene-induced replication stress and poses a major threat to genome stability, promoting tumorigenesis. Oncogene activation can lead to defects in where along the genome and when during the cell cycle licensing takes place, resulting in replication stress. We also discuss the potential of replication licensing as a specific target for novel anticancer therapies.
Collapse
Affiliation(s)
- Michalis Petropoulos
- Department of Biology, School of Medicine, University of Patras, Patras 26504, Greece
| | | | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, Patras 26504, Greece.
| | - Zoi Lygerou
- Department of Biology, School of Medicine, University of Patras, Patras 26504, Greece.
| |
Collapse
|
28
|
DNA Rereplication Is Susceptible to Nucleotide-Level Mutagenesis. Genetics 2019; 212:445-460. [PMID: 31028114 PMCID: PMC6553831 DOI: 10.1534/genetics.119.302194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022] Open
Abstract
The initiation of eukaryotic DNA replication at replication origins is tightly regulated to prevent re-initiation and re-replication within each cell cycle. This regulation is critical for genome stability as re-replication is an extremely potent inducer... The sources of genome instability, a hallmark of cancer, remain incompletely understood. One potential source is DNA rereplication, which arises when the mechanisms that prevent the reinitiation of replication origins within a single cell cycle are compromised. Using the budding yeast Saccharomyces cerevisiae, we previously showed that DNA rereplication is extremely potent at inducing gross chromosomal alterations and that this arises in part because of the susceptibility of rereplication forks to break. Here, we examine the ability of DNA rereplication to induce nucleotide-level mutations. During normal replication these mutations are restricted by three overlapping error-avoidance mechanisms: the nucleotide selectivity of replicative polymerases, their proofreading activity, and mismatch repair. Using lys2InsEA14, a frameshift reporter that is poorly proofread, we show that rereplication induces up to a 30× higher rate of frameshift mutations and that this mutagenesis is due to passage of the rereplication fork, not secondary to rereplication fork breakage. Rereplication can also induce comparable rates of frameshift and base-substitution mutations in a more general mutagenesis reporter CAN1, when the proofreading activity of DNA polymerase ε is inactivated. Finally, we show that the rereplication-induced mutagenesis of both lys2InsEA14 and CAN1 disappears in the absence of mismatch repair. These results suggest that mismatch repair is attenuated during rereplication, although at most sequences DNA polymerase proofreading provides enough error correction to mitigate the mutagenic consequences. Thus, rereplication can facilitate nucleotide-level mutagenesis in addition to inducing gross chromosomal alterations, broadening its potential role in genome instability.
Collapse
|
29
|
Mughal MJ, Mahadevappa R, Kwok HF. DNA replication licensing proteins: Saints and sinners in cancer. Semin Cancer Biol 2018; 58:11-21. [PMID: 30502375 DOI: 10.1016/j.semcancer.2018.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/08/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
DNA replication is all-or-none process in the cell, meaning, once the DNA replication begins it proceeds to completion. Hence, to achieve maximum control of DNA replication, eukaryotic cells employ a multi-subunit initiator protein complex known as "pre-replication complex or DNA replication licensing complex (DNA replication LC). This complex involves multiple proteins which are origin-recognition complex family proteins, cell division cycle-6, chromatin licensing and DNA replication factor 1, and minichromosome maintenance family proteins. Higher-expression of DNA replication LC proteins appears to be an early event during development of cancer since it has been a common hallmark observed in a wide variety of cancers such as oesophageal, laryngeal, pulmonary, mammary, colorectal, renal, urothelial etc. However, the exact mechanisms leading to the abnormally high expression of DNA replication LC have not been clearly deciphered. Increased expression of DNA replication LC leads to licensing and/or firing of multiple origins thereby inducing replication stress and genomic instability. Therapeutic approaches where the reduction in the activity of DNA replication LC was achieved either by siRNA or shRNA techniques, have shown increased sensitivity of cancer cell lines towards the anti-cancer drugs such as cisplatin, 5-Fluorouracil, hydroxyurea etc. Thus, the expression level of DNA replication LC within the cell determines a cell's fate thereby creating a paradox where DNA replication LC acts as both "Saint" and "Sinner". With a potential to increase sensitivity to chemotherapy drugs, DNA replication LC proteins have prospective clinical importance in fighting cancer. Hence, in this review, we will shed light on importance of DNA replication LC with an aim to use DNA replication LC in diagnosis and prognosis of cancer in patients as well as possible therapeutic targets for cancer therapy.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Ravikiran Mahadevappa
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau.
| |
Collapse
|
30
|
Aygun N, Altungoz O. MYCN is amplified during S phase, and c‑myb is involved in controlling MYCN expression and amplification in MYCN‑amplified neuroblastoma cell lines. Mol Med Rep 2018; 19:345-361. [PMID: 30483774 PMCID: PMC6297758 DOI: 10.3892/mmr.2018.9686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 10/03/2018] [Indexed: 01/08/2023] Open
Abstract
Neuroblastoma derived from primitive sympathetic neural precursors is a common type of solid tumor in infants. MYCN proto-oncogene bHLH transcription factor (MYCN) amplification and 1p36 deletion are important factors associated with the poor prognosis of neuroblastoma. Expression levels of MYCN and c-MYB proto-oncogene transcription factor (c-myb) decline during the differentiation of neuroblastoma cells; E2F transcription factor 1 (E2F1) activates the MYCN promoter. However, the underlying mechanism of MYCN overexpression and amplification requires further investigation. In the present study, potential c-Myb target genes, and the effect of c-myb RNA interference (RNAi) on MYCN expression and amplification were investigated in MYCN-amplified neuroblastoma cell lines. The mRNA expression levels and MYCN gene copy number in five neuroblastoma cell lines were determined by quantitative polymerase chain reaction. In addition, variations in potential target gene expression and MYCN gene copy number between pre- and post-c-myb RNAi treatment groups in MYCN-amplified Kelly, IMR32, SIMA and MHH-NB-11 cell lines, normalized to those of non-MYCN-amplified SH-SY5Y, were examined. To determine the associations between gene expression levels and chromosomal aberrations, MYCN amplification and 1p36 alterations in interphases/metaphases were analyzed using fluorescence in situ hybridization. Statistical analyses revealed correlations between 1p36 alterations and the expression of c-myb, MYB proto-oncogene like 2 (B-myb) and cyclin dependent kinase inhibitor 1A (p21). Additionally, the results of the present study also demonstrated that c-myb may be associated with E2F1 and L3MBTL1 histone methyl-lysine binding protein (L3MBTL1) expression, and that E2F1 may contribute to MYCN, B-myb, p21 and chromatin licensing and DNA replication factor 1 (hCdt1) expression, but to the repression of geminin (GMNN). On c-myb RNAi treatment, L3MBTL1 expression was silenced, while GMNN was upregulated, indicating G2/M arrest. In addition, MYCN gene copy number increased following treatment with c-myb RNAi. Notably, the present study also reported a 43.545% sequence identity between upstream of MYCN and Drosophila melanogaster amplification control element 3, suggesting that expression and/or amplification mechanisms of developmentally-regulated genes may be evolutionarily conserved. In conclusion, c-myb may be associated with regulating MYCN expression and amplification. c-myb, B-myb and p21 may also serve a role against chromosome 1p aberrations. Together, it was concluded that MYCN gene is amplified during S phase, potentially via a replication-based mechanism.
Collapse
Affiliation(s)
- Nevim Aygun
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| | - Oguz Altungoz
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| |
Collapse
|
31
|
Gorgoulis VG, Pefani D, Pateras IS, Trougakos IP. Integrating the DNA damage and protein stress responses during cancer development and treatment. J Pathol 2018; 246:12-40. [PMID: 29756349 PMCID: PMC6120562 DOI: 10.1002/path.5097] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 04/16/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
Abstract
During evolution, cells have developed a wide spectrum of stress response modules to ensure homeostasis. The genome and proteome damage response pathways constitute the pillars of this interwoven 'defensive' network. Consequently, the deregulation of these pathways correlates with ageing and various pathophysiological states, including cancer. In the present review, we highlight: (1) the structure of the genome and proteome damage response pathways; (2) their functional crosstalk; and (3) the conditions under which they predispose to cancer. Within this context, we emphasize the role of oncogene-induced DNA damage as a driving force that shapes the cellular landscape for the emergence of the various hallmarks of cancer. We also discuss potential means to exploit key cancer-related alterations of the genome and proteome damage response pathways in order to develop novel efficient therapeutic modalities. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of MedicineNational and Kapodistrian University of AthensAthensGreece
- Biomedical Research Foundation of the Academy of AthensAthensGreece
- Faculty of Biology, Medicine and HealthUniversity of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Dafni‐Eleftheria Pefani
- CRUK/MRC Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Ioannis S Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
32
|
Champeris Tsaniras S, Villiou M, Giannou AD, Nikou S, Petropoulos M, Pateras IS, Tserou P, Karousi F, Lalioti ME, Gorgoulis VG, Patmanidi AL, Stathopoulos GT, Bravou V, Lygerou Z, Taraviras S. Geminin ablation in vivo enhances tumorigenesis through increased genomic instability. J Pathol 2018; 246:134-140. [PMID: 29952003 DOI: 10.1002/path.5128] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/29/2018] [Accepted: 06/01/2018] [Indexed: 11/09/2022]
Abstract
Geminin, a DNA replication licensing inhibitor, ensures faithful DNA replication in vertebrates. Several studies have shown that geminin depletion in vitro results in rereplication and DNA damage, whereas increased expression of geminin has been observed in human cancers. However, conditional inactivation of geminin during embryogenesis has not revealed any detectable DNA replication defects. In order to examine its role in vivo, we conditionally inactivated geminin in the murine colon and lung, and assessed chemically induced carcinogenesis. We show here that mice lacking geminin develop a significantly higher number of tumors and bear a larger tumor burden than sham-treated controls in urethane-induced lung and azoxymethane/dextran sodium sulfate-induced colon carcinogenesis. Survival is also significantly reduced in mice lacking geminin during lung carcinogenesis. A significant increase in the total number and grade of lesions (hyperplasias, adenomas, and carcinomas) was also confirmed by hematoxylin and eosin staining. Moreover, increased genomic aberrations, identified by increased ATR and γH2AX expression, was detected with immunohistochemistry analysis. In addition, we analyzed geminin expression in human colon cancer, and found increased expression, as well as a positive correlation with ATM/ATR levels and a non-monotonic association with γH2AX. Taken together, our data demonstrate that geminin acts as a tumor suppressor by safeguarding genome stability, whereas its overexpression is also associated with genomic instability. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Maria Villiou
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Anastassios D Giannou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Patras, Greece
| | - Sofia Nikou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | | | - Ioannis S Pateras
- Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Paraskevi Tserou
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Foteini Karousi
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Maria-Eleni Lalioti
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Vassilis G Gorgoulis
- Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Patras, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Zoi Lygerou
- Department of Biology, Medical School, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
33
|
Yao L, Chen J, Wu X, Jia S, Meng A. Zebrafish cdc6 hypomorphic mutation causes Meier-Gorlin syndrome-like phenotype. Hum Mol Genet 2018; 26:4168-4180. [PMID: 28985365 PMCID: PMC5886151 DOI: 10.1093/hmg/ddx305] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/26/2017] [Indexed: 11/13/2022] Open
Abstract
Cell Division Cycle 6 (Cdc6) is a component of pre-replicative complex (preRC) forming on DNA replication origins in eukaryotes. Recessive mutations in ORC1, ORC4, ORC6, CDT1 or CDC6 of the preRC in human cause Meier-Gorlin syndrome (MGS) that is characterized by impaired post-natal growth, short stature and microcephaly. However, vertebrate models of MGS have not been reported. Through N-ethyl-N-nitrosourea mutagenesis and Cas9 knockout, we generate several cdc6 mutant lines in zebrafish. Loss-of-function mutations of cdc6, as manifested by cdc6tsu4305 and cdc6tsu7cd mutants, lead to embryonic lethality due to cell cycle arrest at the S phase and extensive apoptosis. Embryos homozygous for a cdc6 hypomorphic mutation, cdc6tsu21cd, develop normally during embryogenesis. Later on, compared with their wild-type (WT) siblings, cdc6tsu21cd mutant fish show growth retardation, and their body weight and length in adulthood are greatly reduced, which resemble human MGS. Surprisingly, cdc6tsu21cd mutant fish become males with a short life and fail to mate with WT females, suggesting defective reproduction. Overexpression of Cdc6 mutant forms, which mimic human CDC6(T323R) mutation found in a MGS patient, in zebrafish cdc6tsu4305 mutant embryos partially represses cell death phenotype, suggesting that the human CDC6(T323R) mutation is a hypomorph. cdc6tsu21cd mutant fish will be useful to detect more tissue defects and develop medical treatment strategies for MGS patients.
Collapse
Affiliation(s)
- Likun Yao
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing Chen
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaotong Wu
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shunji Jia
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Anming Meng
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
34
|
AAV-Mediated angiotensin 1-7 overexpression inhibits tumor growth of lung cancer in vitro and in vivo. Oncotarget 2018; 8:354-363. [PMID: 27861149 PMCID: PMC5352125 DOI: 10.18632/oncotarget.13396] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/11/2016] [Indexed: 11/25/2022] Open
Abstract
Ang-(1-7) inhibits lung cancer cell growth both in vitro and in vivo. However, the molecular mechanism of action is unclear and also the rapid degradation of Ang-(1-7) in vivo limits its clinical application. Here, we have demonstrated that Ang- (1-7) inhibits lung cancer cell growth by interrupting pre-replicative complex assembly and restrains epithelial-mesenchymal transition via Cdc6 inhibition. Furthermore, we constructed a mutant adeno-associated viral vector AAV8 (Y733F) that produced stable and high efficient Ang-(1-7) expression in a xenograft tumor model. The results show that AAV8-mediated Ang-(1-7) over-expression can remarkably suppress tumor growth in vivo by down-regulating Cdc6 and anti-angiogenesis. Ang-(1-7) over-expression via the AAV8 method may be a promising strategy for lung cancer treatment.
Collapse
|
35
|
Chen S, Chen X, Xie G, He Y, Yan D, Zheng D, Li S, Fu X, Li Y, Pang X, Hu Z, Li H, Tan W, Li J. Cdc6 contributes to cisplatin-resistance by activation of ATR-Chk1 pathway in bladder cancer cells. Oncotarget 2018; 7:40362-40376. [PMID: 27246979 PMCID: PMC5130013 DOI: 10.18632/oncotarget.9616] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/08/2016] [Indexed: 12/22/2022] Open
Abstract
High activation of DNA damage response is implicated in cisplatin (CDDP) resistance which presents as a serious obstacle for bladder cancer treatment. Cdc6 plays an important role in the malignant progression of tumor. Here, we reported that Cdc6 expression is up-regulated in bladder cancer tissues and is positively correlated to high tumor grade. Cdc6 depletion can attenuate the malignant properties of bladder cancer cells, including DNA replication, migration and invasion. Furthermore, higher levels of chromatin-binding Cdc6 and ATR were detected in CDDP-resistant bladder cancer cells than in the parent bladder cancer cells. Intriguingly, down-regulation of Cdc6 can enhance sensitivity to CDDP both in bladder cancer cells and CDDP-resistant bladder cancer cells. Cdc6 depletion abrogates S phase arrest caused by CDDP, leading to aberrant mitosis by inactivating ATR-Chk1-Cdc25C pathway. Our results indicate that Cdc6 may be a promising target for overcoming CDDP resistance in bladder cancer.
Collapse
Affiliation(s)
- Sansan Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinglu Chen
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Gui'e Xie
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yue He
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Daoyu Yan
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Dianpeng Zheng
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shi Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyang Fu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yeping Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiang Pang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinlong Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Komseli ES, Pateras IS, Krejsgaard T, Stawiski K, Rizou SV, Polyzos A, Roumelioti FM, Chiourea M, Mourkioti I, Paparouna E, Zampetidis CP, Gumeni S, Trougakos IP, Pefani DE, O’Neill E, Gagos S, Eliopoulos AG, Fendler W, Chowdhury D, Bartek J, Gorgoulis VG. A prototypical non-malignant epithelial model to study genome dynamics and concurrently monitor micro-RNAs and proteins in situ during oncogene-induced senescence. BMC Genomics 2018; 19:37. [PMID: 29321003 PMCID: PMC5763532 DOI: 10.1186/s12864-017-4375-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Senescence is a fundamental biological process implicated in various pathologies, including cancer. Regarding carcinogenesis, senescence signifies, at least in its initial phases, an anti-tumor response that needs to be circumvented for cancer to progress. Micro-RNAs, a subclass of regulatory, non-coding RNAs, participate in senescence regulation. At the subcellular level micro-RNAs, similar to proteins, have been shown to traffic between organelles influencing cellular behavior. The differential function of micro-RNAs relative to their subcellular localization and their role in senescence biology raises concurrent in situ analysis of coding and non-coding gene products in senescent cells as a necessity. However, technical challenges have rendered in situ co-detection unfeasible until now. METHODS In the present report we describe a methodology that bypasses these technical limitations achieving for the first time simultaneous detection of both a micro-RNA and a protein in the biological context of cellular senescence, utilizing the new commercially available SenTraGorTM compound. The method was applied in a prototypical human non-malignant epithelial model of oncogene-induced senescence that we generated for the purposes of the study. For the characterization of this novel system, we applied a wide range of cellular and molecular techniques, as well as high-throughput analysis of the transcriptome and micro-RNAs. RESULTS This experimental setting has three advantages that are presented and discussed: i) it covers a "gap" in the molecular carcinogenesis field, as almost all corresponding in vitro models are fibroblast-based, even though the majority of neoplasms have epithelial origin, ii) it recapitulates the precancerous and cancerous phases of epithelial tumorigenesis within a short time frame under the light of natural selection and iii) it uses as an oncogenic signal, the replication licensing factor CDC6, implicated in both DNA replication and transcription when over-expressed, a characteristic that can be exploited to monitor RNA dynamics. CONCLUSIONS Consequently, we demonstrate that our model is optimal for studying the molecular basis of epithelial carcinogenesis shedding light on the tumor-initiating events. The latter may reveal novel molecular targets with clinical benefit. Besides, since this method can be incorporated in a wide range of low, medium or high-throughput image-based approaches, we expect it to be broadly applicable.
Collapse
Affiliation(s)
- Eirini-Stavroula Komseli
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
| | - Ioannis S. Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
| | - Thorbjørn Krejsgaard
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3c, DK-2200 Copenhagen, Denmark
| | - Konrad Stawiski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 15 Mazowiecka St. 92-215, Lodz, Poland
| | - Sophia V. Rizou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
| | - Alexander Polyzos
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St, GR-11527 Athens, Greece
| | - Fani-Marlen Roumelioti
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St, GR-11527 Athens, Greece
| | - Maria Chiourea
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St, GR-11527 Athens, Greece
| | - Ioanna Mourkioti
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
| | - Eleni Paparouna
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
| | - Christos P. Zampetidis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National & Kapodistrian University of Athens, GR-15784 Athens, Greece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National & Kapodistrian University of Athens, GR-15784 Athens, Greece
| | - Dafni-Eleftheria Pefani
- CRUK/MRC Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Eric O’Neill
- CRUK/MRC Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Sarantis Gagos
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St, GR-11527 Athens, Greece
| | - Aristides G. Eliopoulos
- Department of Biology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research & Technology-Hellas, GR-70013 Heraklion, Crete Greece
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 15 Mazowiecka St. 92-215, Lodz, Poland
- Department of Radiation Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215 USA
| | - Dipanjan Chowdhury
- Department of Radiation Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215 USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115 USA
| | - Jiri Bartek
- Genome Integrity Unit, Danish Cancer Society Research Centre, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínská, 1333/5, 779 00 Olomouc, Czech Republic
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, SE-171 77 Stockholm, Sweden
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National & Kapodistrian University of Athens, 75 Mikras Asias St, GR-11527 Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St, GR-11527 Athens, Greece
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Wilmslow Road, Manchester, M20 4QL UK
| |
Collapse
|
37
|
Agarwal S, Varma D. Targeting mitotic pathways for endocrine-related cancer therapeutics. Endocr Relat Cancer 2017; 24:T65-T82. [PMID: 28615236 PMCID: PMC5557717 DOI: 10.1530/erc-17-0080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/14/2017] [Indexed: 12/21/2022]
Abstract
A colossal amount of basic research over the past few decades has provided unprecedented insights into the highly complex process of cell division. There is an ever-expanding catalog of proteins that orchestrate, participate and coordinate in the exquisite processes of spindle formation, chromosome dynamics and the formation and regulation of kinetochore microtubule attachments. Use of classical microtubule poisons has still been widely and often successfully used to combat a variety of cancers, but their non-selective interference in other crucial physiologic processes necessitate the identification of novel druggable components specific to the cell cycle/division pathway. Considering cell cycle deregulation, unscheduled proliferation, genomic instability and chromosomal instability as a hallmark of tumor cells, there lies an enormous untapped terrain that needs to be unearthed before a drug can pave its way from bench to bedside. This review attempts to systematically summarize the advances made in this context so far with an emphasis on endocrine-related cancers and the avenues for future progress to target mitotic mechanisms in an effort to combat these dreadful cancers.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Cell and Molecular BiologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dileep Varma
- Department of Cell and Molecular BiologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
38
|
Couto PP, Bastos-Rodrigues L, Schayek H, Melo FM, Lisboa RGC, Miranda DM, Vilhena A, Bale AE, Friedman E, De Marco L. Spectrum of germline mutations in smokers and non-smokers in Brazilian non-small-cell lung cancer (NSCLC) patients. Carcinogenesis 2017; 38:1112-1118. [DOI: 10.1093/carcin/bgx089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
39
|
Zhang JH, He YL, Zhu R, Du W, Xiao JH. Deregulated expression of Cdc6 as BCR/ABL-dependent survival factor in chronic myeloid leukemia cells. Tumour Biol 2017. [PMID: 28639894 DOI: 10.1177/1010428317713394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Chronic myeloid leukemia is characterized by the presence of the reciprocal translocation t(9;22) and the BCR/ABL oncogene. The BCR/ABL oncogene activates multiple signaling pathways and involves the dysregulation of oncogenes during the progression of chronic myeloid leukemia. The cell division cycle protein 6, an essential regulator of DNA replication, is elevated in some human cancer cells. However, the expression of cell division cycle protein 6 in chronic myeloid leukemia and the underlying regulatory mechanism remain to be elucidated. In this study, our data showed that cell division cycle protein 6 expression was significantly upregulated in primary chronic myeloid leukemia cells and the chronic myeloid leukemia cell line K562 cells, as compared to the normal bone marrow mononuclear cells. BCR/ABL kinase inhibitor STI571 or BCR/ABL small interfering RNA could significantly downregulate cell division cycle protein 6 messenger RNA expression in K562 cells. Moreover, phosphoinositide 3-kinase/AKT pathway inhibitor LY294002 and Janus kinase/signal transducer and activator of transcription pathway inhibitor AG490 could downregulate cell division cycle protein 6 expression in K562 cells, but not RAS/mitogen-activated protein kinase pathway inhibitor PD98059 had such effect. Cell division cycle protein 6 gene silencing by small interfering RNA effectively resulted in decrease of proliferation, increase of apoptosis, and arrest of cell cycle in K562 cells. These findings have demonstrated that cell division cycle protein 6 overexpression may contribute to the high proliferation and low apoptosis in chronic myeloid leukemia cells and can be regulated by BCR/ABL signal transduction through downstream phosphoinositide 3-kinase/Akt and Janus kinase/signal transducer and activator of transcription pathways, suggesting cell division cycle protein 6 as a potential therapeutic target in chronic myeloid leukemia.
Collapse
Affiliation(s)
- Jia-Hua Zhang
- 1 Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yan-Li He
- 1 Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Rui Zhu
- 2 Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Wen Du
- 1 Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Jun-Hua Xiao
- 3 Department of Pharmacology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
40
|
Abstract
Replication forks encounter obstacles that must be repaired or bypassed to complete chromosome duplication before cell division. Proteomic analysis of replication forks suggests that the checkpoint and repair machinery travels with unperturbed forks, implying that they are poised to respond to stalling and collapse. However, impaired fork progression still generates aberrations, including repeat copy number instability and chromosome rearrangements. Deregulated origin firing also causes fork instability if a newer fork collides with an older one, generating double-strand breaks (DSBs) and partially rereplicated DNA. Current evidence suggests that multiple mechanisms are used to repair rereplication damage, yet these can have deleterious consequences for genome integrity.
Collapse
|
41
|
Gamell C, Gulati T, Levav-Cohen Y, Young RJ, Do H, Pilling P, Takano E, Watkins N, Fox SB, Russell P, Ginsberg D, Monahan BJ, Wright G, Dobrovic A, Haupt S, Solomon B, Haupt Y. Reduced abundance of the E3 ubiquitin ligase E6AP contributes to decreased expression of the INK4/ARF locus in non-small cell lung cancer. Sci Signal 2017; 10:10/461/eaaf8223. [PMID: 28074012 DOI: 10.1126/scisignal.aaf8223] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor suppressor p16INK4a, one protein encoded by the INK4/ARF locus, is frequently absent in multiple cancers, including non-small cell lung cancer (NSCLC). Whereas increased methylation of the encoding gene (CDKN2A) accounts for its loss in a third of patients, no molecular explanation exists for the remainder. We unraveled an alternative mechanism for the silencing of the INK4/ARF locus involving the E3 ubiquitin ligase and transcriptional cofactor E6AP (also known as UBE3A). We found that the expression of three tumor suppressor genes encoded in the INK4/ARF locus (p15INK4b, p16INK4a, and p19ARF) was decreased in E6AP-/- mouse embryo fibroblasts. E6AP induced the expression of the INK4/ARF locus at the transcriptional level by inhibiting CDC6 transcription, a gene encoding a key repressor of the locus. Luciferase assays revealed that E6AP inhibited CDC6 expression by reducing its E2F1-dependent transcription. Chromatin immunoprecipitation analysis indicated that E6AP reduced the amount of E2F1 at the CDC6 promoter. In a subset of NSCLC samples, an E6AP-low/CDC6-high/p16INK4a-low protein abundance profile correlated with low methylation of the gene encoding p16INK4a (CDKN2A) and poor patient prognosis. These findings define a previously unrecognized tumor-suppressive role for E6AP in NSCLC, reveal an alternative silencing mechanism of the INK4/ARF locus, and reveal E6AP as a potential prognostic marker in NSCLC.
Collapse
Affiliation(s)
- Cristina Gamell
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia
| | - Twishi Gulati
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia
| | - Yaara Levav-Cohen
- The Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| | - Richard J Young
- Molecular Therapeutics and Biomarkers Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Hongdo Do
- Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria 3084, Australia
| | - Pat Pilling
- Biomedical Manufacturing Program, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria 3169, Australia
| | - Elena Takano
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Neil Watkins
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Prudence Russell
- Department of Anatomical Pathology, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Doron Ginsberg
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Brendon J Monahan
- Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 2555, Australia
| | - Gavin Wright
- Department of Surgery, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Alex Dobrovic
- Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria 3084, Australia
| | - Sue Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia
| | - Ben Solomon
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia.,Molecular Therapeutics and Biomarkers Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Ygal Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3800, Australia.,Department of Pathology, University of Melbourne, Victoria 3800, Australia
| |
Collapse
|
42
|
Sang Y, Yan F, Ren X. The role and mechanism of CRL4 E3 ubiquitin ligase in cancer and its potential therapy implications. Oncotarget 2016; 6:42590-602. [PMID: 26460955 PMCID: PMC4767455 DOI: 10.18632/oncotarget.6052] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/23/2015] [Indexed: 12/21/2022] Open
Abstract
CRLs (Cullin-RING E3 ubiquitin ligases) are the largest E3 ligase family in eukaryotes, which ubiquitinate a wide range of substrates involved in cell cycle regulation, signal transduction, transcriptional regulation, DNA damage response, genomic integrity, tumor suppression and embryonic development. CRL4 E3 ubiquitin ligase, as one member of CRLs family, consists of a RING finger domain protein, cullin4 (CUL4) scaffold protein and DDB1–CUL4 associated substrate receptors. The CUL4 subfamily includes two members, CUL4A and CUL4B, which share extensively sequence identity and functional redundancy. Aberrant expression of CUL4 has been found in a majority of tumors. Given the significance of CUL4 in cancer, understanding its detailed aspects of pathogenesis of human malignancy would have significant value for the treatment of cancer. Here, the work provides an overview to address the role of CRL4 E3 ubiquitin ligase in cancer development and progression, and discuss the possible mechanisms of CRL4 ligase involving in many cellular processes associated with tumor. Finally, we discuss its potential value in cancer therapy.
Collapse
Affiliation(s)
- Youzhou Sang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Fan Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
43
|
Galanos P, Vougas K, Walter D, Polyzos A, Maya-Mendoza A, Haagensen EJ, Kokkalis A, Roumelioti FM, Gagos S, Tzetis M, Canovas B, Igea A, Kanavakis E, Kletsas D, Roninson I, Garbis SD, Nebreda A, Thanos D, Townsend P, Blow JJ, Sørensen CS, Bartek J, Gorgoulis VG. Chronic p53-independent p21 expression causes genomic instability by deregulating replication licensing. Nat Cell Biol 2016; 18:777-89. [PMID: 27323328 PMCID: PMC6535144 DOI: 10.1038/ncb3378] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/19/2016] [Indexed: 12/17/2022]
Abstract
The cyclin-dependent kinase inhibitor p21(WAF1/CIP1) (p21) is a cell-cycle checkpoint effector and inducer of senescence, regulated by p53. Yet, evidence suggests that p21 could also be oncogenic, through a mechanism that has so far remained obscure. We report that a subset of atypical cancerous cells strongly expressing p21 showed proliferation features. This occurred predominantly in p53-mutant human cancers, suggesting p53-independent upregulation of p21 selectively in more aggressive tumour cells. Multifaceted phenotypic and genomic analyses of p21-inducible, p53-null, cancerous and near-normal cellular models showed that after an initial senescence-like phase, a subpopulation of p21-expressing proliferating cells emerged, featuring increased genomic instability, aggressiveness and chemoresistance. Mechanistically, sustained p21 accumulation inhibited mainly the CRL4-CDT2 ubiquitin ligase, leading to deregulated origin licensing and replication stress. Collectively, our data reveal the tumour-promoting ability of p21 through deregulation of DNA replication licensing machinery-an unorthodox role to be considered in cancer treatment, since p21 responds to various stimuli including some chemotherapy drugs.
Collapse
Affiliation(s)
- Panagiotis Galanos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Konstantinos Vougas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - David Walter
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Alexander Polyzos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Emma J. Haagensen
- Centre for Gene Regulation & Expression, College of Life Sciences, University of Dundee, Dundee, UK
| | - Antonis Kokkalis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Sarantis Gagos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Tzetis
- Department of Medical Genetics, Medical School, University of Athens, Athens, Greece
| | - Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Ana Igea
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Emanuel Kanavakis
- Department of Medical Genetics, Medical School, University of Athens, Athens, Greece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biology, National Centre for Scientific Research ‘Demokritos’, Athens, Greece
| | - Igor Roninson
- Cancer Center, Ordway Research Institute, Albany, NY, USA
| | - Spiros D. Garbis
- Cancer and Clinical Experimental Science Units, Faculty of Medicine, Institute for Life Sciences, Center for Proteome Research, University of Southampton, Southampton, UK
| | - Angel Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Dimitris Thanos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Paul Townsend
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Centre for Cellular Metabolism, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - J. Julian Blow
- Centre for Gene Regulation & Expression, College of Life Sciences, University of Dundee, Dundee, UK
| | | | - Jiri Bartek
- Genome Integrity Unit, Danish Cancer Society Research Centre, Copenhagen, Denmark
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Centre for Cellular Metabolism, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
44
|
Logotheti S, Khoury N, Vlahopoulos SA, Skourti E, Papaevangeliou D, Liloglou T, Gorgoulis V, Budunova I, Kyriakopoulos AM, Zoumpourlis V. N-bromotaurine surrogates for loss of antiproliferative response and enhances cisplatin efficacy in cancer cells with impaired glucocorticoid receptor. Transl Res 2016; 173:58-73.e2. [PMID: 27063960 DOI: 10.1016/j.trsl.2016.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 03/09/2016] [Accepted: 03/12/2016] [Indexed: 11/15/2022]
Abstract
Glucocorticoids (GCs) are frequently used in anticancer combination regimens; however, their continuous use adds selective pressure on cancer cells to develop GC-resistance via impairment of the glucocorticoid receptor (GR), therefore creating a need for GC-alternatives. Based on the drug repurposing approach and the commonalities between inflammation and neoplasia, drugs that are either in late-stage clinical trials and/or already marketed for GC-refractory inflammatory diseases could be evaluated as GC-substitutes in the context of cancer. Advantageously, unlike new molecular entities currently being de novo developed to restore GC-responsiveness of cancer cells, such drugs have documented safety and efficacy profile, which overall simplifies their introduction in clinical cancer trials. In this study, we estimated the potential of a well-established, multistage, cell line-based, mouse skin carcinogenesis model to be exploited as an initial screening tool for unveiling covert GC-substitutes. First, we categorized the cell lines of this model to GC-sensitive and GC-resistant, in correlation with their corresponding GR status, localization, and functionality. We found that GC-resistance starts in papilloma stages, due to a dysfunctional GR, which is overexpressed, DNA binding-competent, but transactivation-incompetent in papilloma, squamous, and spindle stages of the model. Then, aided by this tool, we evaluated the ability of N-bromotaurine, a naturally occurring, small-molecule, nonsteroid anti-inflammatory drug which is under consideration for use interchangeably/in replacement to GCs in skin inflammations, to restore antiproliferative response of GC-resistant cancer cells. Unlike GCs, N-bromotaurine inhibited cell-cycle progression in GC-resistant cancer cells and efficiently synergized with cisplatin, thus indicating a potential to be exploited instead of GCs against cancer.
Collapse
Affiliation(s)
- Stella Logotheti
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece; Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Nikolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Spiros A Vlahopoulos
- Horemio Research Institute, First Department of Pediatrics, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elena Skourti
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Dimitra Papaevangeliou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Triantafyllos Liloglou
- University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Liverpool, UK
| | - Vassilis Gorgoulis
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, Ill, USA
| | | | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece.
| |
Collapse
|
45
|
Plasma membrane/cell wall perturbation activates a novel cell cycle checkpoint during G1 in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 2016; 113:6910-5. [PMID: 27274080 DOI: 10.1073/pnas.1523824113] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cellular wound healing or the repair of plasma membrane/cell wall damage (plasma membrane damage) occurs frequently in nature. Although various cellular perturbations, such as DNA damage, spindle misalignment, and impaired daughter cell formation, are monitored by cell cycle checkpoint mechanisms in budding yeast, whether plasma membrane damage is monitored by any of these checkpoints remains to be addressed. Here, we define the mechanism by which cells sense membrane damage and inhibit DNA replication. We found that the inhibition of DNA replication upon plasma membrane damage requires GSK3/Mck1-dependent degradation of Cdc6, a component of the prereplicative complex. Furthermore, the CDK inhibitor Sic1 is stabilized in response to plasma membrane damage, leading to cell integrity maintenance in parallel with the Mck1-Cdc6 pathway. Cells defective in both Cdc6 degradation and Sic1 stabilization failed to grow in the presence of plasma membrane damage. Taking these data together, we propose that plasma membrane damage triggers G1 arrest via Cdc6 degradation and Sic1 stabilization to promote the cellular wound healing process.
Collapse
|
46
|
Petrakis TG, Komseli ES, Papaioannou M, Vougas K, Polyzos A, Myrianthopoulos V, Mikros E, Trougakos IP, Thanos D, Branzei D, Townsend P, Gorgoulis VG. Exploring and exploiting the systemic effects of deregulated replication licensing. Semin Cancer Biol 2016; 37-38:3-15. [PMID: 26707000 DOI: 10.1016/j.semcancer.2015.12.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/10/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
Maintenance and accurate propagation of the genetic material are key features for physiological development and wellbeing. The replication licensing machinery is crucial for replication precision as it ensures that replication takes place once per cell cycle. Thus, the expression status of the components comprising the replication licensing apparatus is tightly regulated to avoid re-replication; a form of replication stress that leads to genomic instability, a hallmark of cancer. In the present review we discuss the mechanistic basis of replication licensing deregulation, which leads to systemic effects, exemplified by its role in carcinogenesis and a variety of genetic syndromes. In addition, new insights demonstrate that above a particular threshold, the replication licensing factor Cdc6 acts as global transcriptional regulator, outlining new lines of exploration. The role of the putative replication licensing factor ChlR1/DDX11, mutated in the Warsaw Breakage Syndrome, in cancer is also considered. Finally, future perspectives focused on the potential therapeutic advantage by targeting replication licensing factors, and particularly Cdc6, are discussed.
Collapse
Affiliation(s)
- Theodoros G Petrakis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Eirini-Stavroula Komseli
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Marilena Papaioannou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Kostas Vougas
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | | | - Emmanuel Mikros
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Athens, Greece
| | - Dimitris Thanos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dana Branzei
- FIRC (Fondazione Italiana per la Ricerca sul Cancro) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Paul Townsend
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece; Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
47
|
Karavias D, Maroulis I, Papadaki H, Gogos C, Kakkos S, Karavias D, Bravou V. Overexpression of CDT1 Is a Predictor of Poor Survival in Patients with Hepatocellular Carcinoma. J Gastrointest Surg 2016; 20:568-79. [PMID: 26408331 DOI: 10.1007/s11605-015-2960-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/16/2015] [Indexed: 01/31/2023]
Abstract
BACKGROUND Genomic instability is a common feature in hepatocellular carcinoma. Deregulation of replication licensing factors has been shown to trigger DNA damage response contributing to genomic instability. Overexpression of DNA replication licensing factors chromatin licensing and DNA replication factor 1 (CDT1) and minichromosome maintenance complex component 7 (MCM7) has been previously reported in several human cancers. The aim of the present study was to evaluate the expression and prognostic significance of CDT1 and MCM7 in association with DNA damage response markers and p53 in patients with hepatocellular carcinoma. METHODS Expression of CDT1, MCM7, p-H2A histone family member X (H2AX), phospho-ataxia telangiectasia-mutated (ATM)/ataxia telangiectasia rad3-related (ATR) substrate, and p53 was evaluated by immunohistochemistry on formalin-fixed paraffin-embedded surgical specimens from 111 patients who underwent hepatectomy for hepatocellular carcinoma. Statistical analysis was performed to evaluate associations between the studied proteins, clinicopathological parameters, and patient survival. RESULTS CDT1 expression correlated with p-H2AX (p = 0.038), while MCM7 correlated with p-H2AX and phospho-ATM/ATR substrate (p < 0.001). Increased CDT1 expression was associated with higher tumor grade (p = 0.006) and tumor-node-metastasis (TNM) stage (p = 0.033). High CDT1 expression correlated significantly with reduced overall survival (60.8 and 26.5 % vs 82.8 and 53.0 %, for low CDT1 expression, at 2 and 5 years, respectively, p = 0.012) and was identified by multivariate analysis as an independent predictor of poor overall survival (p = 0.049). CONCLUSIONS Overexpression of CDT1 and MCM7 in hepatocellular carcinoma correlates with DNA damage response, and CDT1 overexpression is a significant prognostic biomarker in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Dimitrios Karavias
- Department of Surgery, University Hospital of Patras, Rio, 26500, Greece.
| | - Ioannis Maroulis
- Department of Surgery, University Hospital of Patras, Rio, 26500, Greece
| | - Helen Papadaki
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Rio, Greece
| | - Charalambos Gogos
- Department of Internal Medicine, University Hospital of Patras, Rio, Greece
| | - Stavros Kakkos
- Department of Vascular Surgery, University Hospital of Patras, Rio, Greece
| | | | - Vasiliki Bravou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Rio, Greece
| |
Collapse
|
48
|
Excess Cdt1 inhibits nascent strand elongation by repressing the progression of replication forks in Xenopus egg extracts. Biochem Biophys Res Commun 2016; 470:405-410. [DOI: 10.1016/j.bbrc.2016.01.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 11/22/2022]
|
49
|
Wang Y, Han R, Chen Z, Fu M, Kang J, Li K, Li L, Chen H, He Y. A transcriptional miRNA-gene network associated with lung adenocarcinoma metastasis based on the TCGA database. Oncol Rep 2016; 35:2257-69. [PMID: 26781266 DOI: 10.3892/or.2016.4560] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 11/20/2015] [Indexed: 11/05/2022] Open
Abstract
Lung adenocarcinoma is the most common subtype of non-small cell lung cancer (NSCLC), leading to the largest number of cancer-related deaths worldwide. The high mortality rate may be attributed to the delay of detection. Therefore, it is of great importance to explore the mechanism of lung adenocarcinoma metastasis and the strategy to block metastasis of the disease. We searched and downloaded mRNA and miRNA expression data and clinical data from The Cancer Genome Atlas (TCGA) database to identify differences in mRNA and miRNA expression of primary tumor tissues from lung adenocarcinoma that did and did not metastasize. In addition, combined with bioinformatic prediction, we constructed an miRNA-target gene regulatory network. Finally, we employed RT-qPCR to validate the bioinformatic approach by determining the expression of 10 significantly differentially expressed genes which were also putative targets of several dysregulated miRNAs. RT-qPCR results indicated that the bioinformatic approach in our study was acceptable. Our data suggested that some of the genes including PKM2, STRAP and FLT3, may participate in the pathology of lung adenocarcinoma metastasis and could be applied as potential markers or therapeutic targets for lung adenocarcinoma.
Collapse
Affiliation(s)
- Yubo Wang
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Rui Han
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Zhaojun Chen
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Ming Fu
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Jun Kang
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Kunlin Li
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Li Li
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Hengyi Chen
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Yong He
- Department of Respiratory Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
50
|
Expression Analysis of Genes Involved in the RB/E2F Pathway in Astrocytic Tumors. PLoS One 2015; 10:e0137259. [PMID: 26317630 PMCID: PMC4552853 DOI: 10.1371/journal.pone.0137259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/13/2015] [Indexed: 02/08/2023] Open
Abstract
Astrocytic gliomas, which are derived from glial cells, are considered the most common primary neoplasias of the central nervous system (CNS) and are histologically classified as low grade (I and II) or high grade (III and IV). Recent studies have shown that astrocytoma formation is the result of the deregulation of several pathways, including the RB/E2F pathway, which is commonly deregulated in various human cancers via genetic or epigenetic mechanisms. On the basis of the assumption that the study of the mechanisms controlling the INK4/ARF locus can help elucidate the molecular pathogenesis of astrocytic tumors, identify diagnostic and prognostic markers, and help select appropriate clinical treatments, the present study aimed to evaluate and compare methylation patterns using bisulfite sequencing PCR and evaluate the gene expression profile using real-time PCR in the genes CDKN2A, CDKN2B, CDC6, Bmi-1, CCND1, and RB1 in astrocytic tumors. Our results indicate that all the evaluated genes are not methylated independent of the tumor grade. However, the real-time PCR results indicate that these genes undergo progressive deregulation as a function of the tumor grade. In addition, the genes CDKN2A, CDKN2B, and RB1 were underexpressed, whereas CDC6, Bmi-1, and CCND1 were overexpressed; the increase in gene expression was significantly associated with decreased patient survival. Therefore, we propose that the evaluation of the expression levels of the genes involved in the RB/E2F pathway can be used in the monitoring of patients with astrocytomas in clinical practice and for the prognostic indication of disease progression.
Collapse
|