1
|
Arroyo M, Fernández-Mimbrera MA, Gollini E, Esteve-Codina A, Sánchez A, Marchal JA. TOP2A inhibition and its cellular effects related to cell cycle checkpoint adaptation pathway. Sci Rep 2025; 15:3831. [PMID: 39885205 PMCID: PMC11782647 DOI: 10.1038/s41598-025-87895-8] [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: 07/04/2024] [Accepted: 01/19/2025] [Indexed: 02/01/2025] Open
Abstract
In this study, we investigate the G2 checkpoint activated by chromosome entanglements, the so-called Decatenation Checkpoint (DC), which can be activated by TOP2A catalytic inhibition. Specifically, we focus on the spontaneous ability of cells to bypass or override this checkpoint, referred to as checkpoint adaptation. Some factors involved in adapting to this checkpoint are p53 and MCPH1. Using cellular models depleted of p53 or both p53 and MCPH1 in hTERT-RPE1 cells, we analyzed cell cycle dynamics and adaptation, segregation defects, apoptosis rate, and transcriptional changes related to prolonged exposure to TOP2A inhibitors. Our findings reveal that cell cycle dynamics are altered in MCPH1-depleted cells compared to control cells. We found that MCPH1 depletion can restore the robustness of the DC in a p53-negative background. Furthermore, this research highlights the differential effects of TOP2A poisons and catalytic inhibitors on cellular outcomes and transcriptional profiles. By examining the different mechanisms of TOP2A inhibition and their impact on cellular processes, this study contributes to a deeper understanding of the regulation and physiological implications of the DC and checkpoint adaptation in non-carcinogenic cell lines.
Collapse
Affiliation(s)
- Maria Arroyo
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, Darmstadt, Germany.
| | - M A Fernández-Mimbrera
- Departamento Biología Experimental, Universidad de Jaén, Paraje Las Lagunillas S/N E23071, Jaén, Spain
| | - E Gollini
- Departamento Biología Experimental, Universidad de Jaén, Paraje Las Lagunillas S/N E23071, Jaén, Spain
| | - A Esteve-Codina
- Centre Nacional d'Anàlisi Genòmica (CNAG), Baldiri Reixac 4, 08028, Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - A Sánchez
- Departamento Biología Experimental, Universidad de Jaén, Paraje Las Lagunillas S/N E23071, Jaén, Spain
| | - Juan Alberto Marchal
- Departamento Biología Experimental, Universidad de Jaén, Paraje Las Lagunillas S/N E23071, Jaén, Spain.
| |
Collapse
|
2
|
Wang X, Jin L, Zhang X, Li M, Zhu A, Zhang M, Fan H. Transcriptomic profiling and risk assessment in bladder cancer: Insights from copper death-related genes. Cell Signal 2024; 121:111237. [PMID: 38810861 DOI: 10.1016/j.cellsig.2024.111237] [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: 03/23/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND The study aimed to investigate the role of copper death-related genes (CRGs) in bladder cancer (BC) for improved prognosis assessment. METHODS Multi-omics techniques were utilized to analyze CRG expression in BC tissues from TCGA and GEO databases. Consensus clustering categorized patients into molecular subtypes based on clinical characteristics and immune cell infiltration. RESULTS An innovative risk assessment model identified eight critical genes associated with BC risk. In vitro and in vivo experiments validated LIPT1's significant impact on copper-induced cell death, proliferation, migration, and invasion in BC. CONCLUSION This multi-omics analysis elucidates the pivotal role of CRGs in BC progression, suggesting enhanced risk assessment through molecular subtype categorization and identification of key genes like LIPT1. Insights into these mechanisms offer the potential for improved diagnosis and treatment strategies for BC patients.
Collapse
Affiliation(s)
- Xu Wang
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China
| | - Long Jin
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China
| | - Xiaoyu Zhang
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China
| | - Mingyu Li
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China
| | - Ankang Zhu
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China
| | - Ming Zhang
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China
| | - Haitao Fan
- Department of Urology, The Second Hospital of Jilin University, Changchun 130022, PR China.
| |
Collapse
|
3
|
Soliman TN, Keifenheim D, Parker PJ, Clarke DJ. Cell cycle responses to Topoisomerase II inhibition: Molecular mechanisms and clinical implications. J Cell Biol 2023; 222:e202209125. [PMID: 37955972 PMCID: PMC10641588 DOI: 10.1083/jcb.202209125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
DNA Topoisomerase IIA (Topo IIA) is an enzyme that alters the topological state of DNA and is essential for the separation of replicated sister chromatids and the integrity of cell division. Topo IIA dysfunction activates cell cycle checkpoints, resulting in arrest in either the G2-phase or metaphase of mitosis, ultimately triggering the abscission checkpoint if non-disjunction persists. These events, which directly or indirectly monitor the activity of Topo IIA, have become of major interest as many cancers have deficiencies in Topoisomerase checkpoints, leading to genome instability. Recent studies into how cells sense Topo IIA dysfunction and respond by regulating cell cycle progression demonstrate that the Topo IIA G2 checkpoint is distinct from the G2-DNA damage checkpoint. Likewise, in mitosis, the metaphase Topo IIA checkpoint is separate from the spindle assembly checkpoint. Here, we integrate mechanistic knowledge of Topo IIA checkpoints with the current understanding of how cells regulate progression through the cell cycle to accomplish faithful genome transmission and discuss the opportunities this offers for therapy.
Collapse
Affiliation(s)
- Tanya N. Soliman
- Barts Cancer Institute, Queen Mary University London, London, UK
| | - Daniel Keifenheim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | | | - Duncan J. Clarke
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
4
|
Accurate Characterization of Bladder Cancer Cells with Intraoperative Flow Cytometry. Cancers (Basel) 2022; 14:cancers14215440. [PMID: 36358858 PMCID: PMC9656620 DOI: 10.3390/cancers14215440] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary Bladder cancer is a malignancy that predominantly affects male patients. Surgical treatment is the first option for clinical management and cancer cell characterization is critical for tumor margin detection and complete tumor removal. We developed a specialized intraoperative flow cytometry (iFC) methodology for bladder cancer cell detection. Our study, including 52 individuals, reveals that iFC is highly specific, sensitive and accurate in detecting cancer cells, based on the quantification of cell proliferation and the presence of tumor aneuploidy. The results of this study advocate further research on the utility of iFC as a next-generation malignancy evaluation technique during transurethral resections. Abstract Bladder cancer represents a major health issue. Transurethral resection is the first line treatment and an accurate assessment of tumor margins might warrant complete tumor removal. Genomic instability and proliferative potential are common hallmarks of cancer cells. We have previously demonstrated the utility of intraoperative flow cytometry (iFC), a next-generation margin evaluation methodology for assessment of DNA content, in the detection of several types of malignancy. In the current study we investigated the possible value of iFC in the characterization of bladder cancer during surgery. Samples from a population of 52 people with urothelial cancer were included in the study. The total time for iFC evaluation is 3–5 min per sample and included a two-step analysis, including DNA-index and Tumor-index calculation. First, DNA-index calculation revealed 24 hyperploid and one hypoploid tumor. Second, cell cycle analysis and Tumor-index calculation revealed that tumor samples are distinguished from normal cells based on their significantly higher proliferative potential. The standard for iFC evaluation was pathology assessment and revealed that our protocol exhibits an accuracy of 98% in defining the presence of cancer cells in a given sample. Our results support the further assessment of iFC value towards its use as a novel malignancy evaluation tool in transurethral resections.
Collapse
|
5
|
Rad54L promotes bladder cancer progression by regulating cell cycle and cell senescence. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:185. [PMID: 36071250 DOI: 10.1007/s12032-022-01751-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/13/2022] [Indexed: 10/14/2022]
Abstract
Bladder cancer (BCa) is the most prevalent cancer of the urinary system, but its pathogenesis is still poorly understood. Several reports have suggested that gene damage repair is highly correlated with tumor development and drug resistance, in which homologous recombination repair gene Rad54L seems to play an important role, through yet unclear mechanisms. Therefore, this study stratified cancer patients by Rad54L expression in BCa tissue, and high Rad54L expression was associated with a poor prognosis. Mechanistically, we demonstrate that high Rad54L expression promotes abnormal bladder tumor cell proliferation by changing the cell cycle and cell senescence. In addition, this study also suggests that Rad54L may be associated with p53, p21, and pRB in BCa tissue. In summary, this study exposes Rad54L as potential a prognostic biomarker and precision treatment target in BCa.
Collapse
|
6
|
Shi WW, Guan JZ, Long YP, Song Q, Xiong Q, Qin BY, Ma ZQ, Hu Y, Yang B. Integrative transcriptional characterization of cell cycle checkpoint genes promotes clinical management and precision medicine in bladder carcinoma. Front Oncol 2022; 12:915662. [PMID: 36033441 PMCID: PMC9404245 DOI: 10.3389/fonc.2022.915662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Background The aberrant regulation of cell cycle is significantly correlated with cancer carcinogenesis and progression, in which cell cycle checkpoints control phase transitions, cell cycle entry, progression, and exit. However, the integrative role of cell cycle checkpoint-related genes (CRGs) in bladder carcinoma (BC) remains unknown. Methods The transcriptomic data and clinical features of BC patients were downloaded from The Cancer Genome Atlas (TCGA), used to identify CRGs correlated with overall survival (OS) by univariate Cox regression analysis. Then, the multivariate and least absolute shrinkage and selection operator (LASSO) Cox regression analyses further developed a prognostic CRG signature, which was validated in three external datasets retrieved from Gene Expression Omnibus (GEO). The receiver operating characteristic curve (ROC) analysis was conducted for evaluating the performance of the CRG signature in prognosis prediction. RNA sequencing (RNA-Seq) was performed to explore the expression difference in the identified CRGs between tumor and normal tissue samples from 11 BC patients in the local cohort. Ultimately, genomic profiles and tumor microenvironment (TME), and the Genomics of Drug Sensitivity in Cancer (GDSC) were investigated to guide precision treatment for BC patients with different CRG features. Results The novel constructed 23-CRG prognostic signature could stratify BC patients into high-risk and low-risk groups with significantly different outcomes (median OS: 13.64 vs. 104.65 months). Notably, 19 CRGs were the first to be identified as being associated with BC progression. In three additional validation datasets (GSE13507, GSE31684, and GSE32548), higher CRG scores all indicated inferior survival, demonstrating the robust ability of the CRG signature in prognosis prediction. Moreover, the CRG signature as an independent prognostic factor had a robust and stable risk stratification for BC patients with different histological or clinical features. Then, a CRG signature-based nomogram with a better performance in prognostic prediction [concordance index (C-index): 0.76] was established. Functional enrichment analysis revealed that collagen-containing extracellular matrix (ECM), and ECM-related and MAPK signaling pathways were significantly associated with the signature. Further analysis showed that low-risk patients were characterized by particularly distinctive prevalence of FGFR3 (17.03% vs. 6.67%, p < 0.01) and POLE alterations (7.97% vs. 2.50%, p < 0.05), and enrichment of immune infiltrated cells (including CD8+ T cells, CD4+ naïve T cells, follicular helper T cells, Tregs, and myeloid dendritic cells). RNA-seq data in our local cohort supported the findings in the differentially expressed genes (DEGs) between tumor and normal tissue samples, and the difference in TME between high-risk and low-risk groups. Additionally, CRG signature score plus FGFR3 status divided BC patients into four molecular subtypes, with distinct prognosis, TME, and transcriptomic profiling of immune checkpoint genes. Of note, CRG signature score plus FGFR3 status could successfully distinguish BC patients who have a higher possibility of response to immunotherapy or chemotherapy drugs. Conclusions The CRG signature is a potent prognostic model for BC patients, and in combination with FGFR3 alterations, it had more practical capacity in the prediction of chemotherapy and immunotherapy response, helping guide clinical decision-making.
Collapse
Affiliation(s)
- Wei-Wei Shi
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Jing-Zhi Guan
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Ya-Ping Long
- School of Medicine, Nankai University, Tianjin, China
| | - Qi Song
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Qi Xiong
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Bo-Yu Qin
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhi-Qiang Ma
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yi Hu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Yi Hu, ; Bo Yang,
| | - Bo Yang
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Yi Hu, ; Bo Yang,
| |
Collapse
|
7
|
Lin CH, Chang HH, Lai CR, Wang HH, Tsai WC, Tsai YL, Changchien CY, Cheng YC, Wu ST, Chen Y. Fatty Acid Binding Protein 6 Inhibition Decreases Cell Cycle Progression, Migration and Autophagy in Bladder Cancers. Int J Mol Sci 2022; 23:ijms23042154. [PMID: 35216267 PMCID: PMC8878685 DOI: 10.3390/ijms23042154] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Bladder cancer (BC) has a high recurrence rate worldwide. The aim of this study was to evaluate the role of fatty acid binding protein 6 (FABP6) in proliferation and migration in human bladder cancer cells. Cell growth was confirmed by MTT and colony formation assay. Western blotting was used to explore protein expressions. Wound healing and Transwell assays were performed to evaluate the migration ability. A xenograft animal model with subcutaneous implantation of BC cells was generated to confirm the tumor progression. Knockdown of FABP6 reduced cell growth in low-grade TSGH-8301 and high-grade T24 cells. Cell cycle blockade was observed with the decrease of CDK2, CDK4, and Ki67 levels in FABP6-knockdown BC cells. Interestingly, knockdown of FBAP6 led to downregulation of autophagic markers and activation of AKT-mTOR signaling. The application of PI3K/AKT inhibitor decreased cell viability mediated by FABP6-knockdown additionally. Moreover, FABP6-knockdown reduced peroxisome proliferator-activated receptor γ and retinoid X receptor α levels but increased p-p65 expression. Knockdown of FABP6 also inhibited BC cell motility with focal adhesive complex reduction. Finally, shFABP6 combined with cisplatin suppressed tumor growth in vivo. These results provide evidence that FABP6 may be a potential target in BC cells progression.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- National Defense Medical Center, Department of Biology and Anatomy, Taipei 11490, Taiwan; (C.-H.L.); (H.-H.C.); (C.-R.L.); (C.-Y.C.); (Y.-C.C.)
| | - Hsin-Han Chang
- National Defense Medical Center, Department of Biology and Anatomy, Taipei 11490, Taiwan; (C.-H.L.); (H.-H.C.); (C.-R.L.); (C.-Y.C.); (Y.-C.C.)
| | - Chien-Rui Lai
- National Defense Medical Center, Department of Biology and Anatomy, Taipei 11490, Taiwan; (C.-H.L.); (H.-H.C.); (C.-R.L.); (C.-Y.C.); (Y.-C.C.)
| | - Hisao-Hsien Wang
- Department of Urology, Cheng Hsin General Hospital, Taipei 11490, Taiwan;
| | - Wen-Chiuan Tsai
- National Defense Medical Center, Department of Pathology, Tri-Service General Hospital, Taipei 11490, Taiwan; (W.-C.T.); (Y.-L.T.)
| | - Yu-Ling Tsai
- National Defense Medical Center, Department of Pathology, Tri-Service General Hospital, Taipei 11490, Taiwan; (W.-C.T.); (Y.-L.T.)
| | - Chih-Ying Changchien
- National Defense Medical Center, Department of Biology and Anatomy, Taipei 11490, Taiwan; (C.-H.L.); (H.-H.C.); (C.-R.L.); (C.-Y.C.); (Y.-C.C.)
- National Defense Medical Center, Department of Internal Medicine, Tri-Service General Hospital, Taipei 11490, Taiwan
| | - Yu-Chen Cheng
- National Defense Medical Center, Department of Biology and Anatomy, Taipei 11490, Taiwan; (C.-H.L.); (H.-H.C.); (C.-R.L.); (C.-Y.C.); (Y.-C.C.)
| | - Sheng-Tang Wu
- National Defense Medical Center, Division of Urology, Department of Surgery, Tri-Service General Hospital, Taipei 11490, Taiwan
- Correspondence: (S.-T.W.); (Y.C.); Tel.: +886-2-8792-3100 (ext. 18739) (Y.C.)
| | - Ying Chen
- National Defense Medical Center, Department of Biology and Anatomy, Taipei 11490, Taiwan; (C.-H.L.); (H.-H.C.); (C.-R.L.); (C.-Y.C.); (Y.-C.C.)
- Correspondence: (S.-T.W.); (Y.C.); Tel.: +886-2-8792-3100 (ext. 18739) (Y.C.)
| |
Collapse
|
8
|
Identification of Key Genes Associated with Progression and Prognosis of Bladder Cancer through Integrated Bioinformatics Analysis. Cancers (Basel) 2021; 13:cancers13235931. [PMID: 34885040 PMCID: PMC8656554 DOI: 10.3390/cancers13235931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Bladder cancer is a heterogeneous disease with high recurrence rates. The current prognostication depends on tumor stage and grade and there is a need for predictive biomarkers that can distinguish between progressive versus non-progressive disease. We have identified a 3-gene signature panel having prognostic value in bladder cancer, which could aid in clinical decision making. Abstract Bladder cancer prognosis remains dismal due to lack of appropriate biomarkers that can predict its progression. The study aims to identify novel prognostic biomarkers associated with the progression of bladder cancer by utilizing three Gene Expression Omnibus (GEO) datasets to screen differentially expressed genes (DEGs). A total of 1516 DEGs were identified between non-muscle invasive and muscle invasive bladder cancer specimens. To identify genes of prognostic value, we performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. A total of seven genes, including CDKN2A, CDC20, CTSV, FOXM1, MAGEA6, KRT23, and S100A9 were confirmed with strong prognostic values in bladder cancer and validated by qRT-PCR conducted in various human bladder cancer cells representing stage-specific disease progression. ULCAN, human protein atlas and The Cancer Genome Atlas datasets were used to confirm the predictive value of these genes in bladder cancer progression. Moreover, Kaplan–Meier analysis and Cox hazard ratio analysis were performed to determine the prognostic role of these genes. Univariate analysis performed on a validation set identified a 3-panel gene set viz. CDKN2A, CTSV and FOXM1 with 95.5% sensitivity and 100% specificity in predicting bladder cancer progression. In summary, our study screened and confirmed a 3-panel biomarker that could accurately predict the progression and prognosis of bladder cancer.
Collapse
|
9
|
Al-Harbi SA, Abdulrahman AO, Zamzami MA, Khan MI. Urolithins: The Gut Based Polyphenol Metabolites of Ellagitannins in Cancer Prevention, a Review. Front Nutr 2021; 8:647582. [PMID: 34164422 PMCID: PMC8215145 DOI: 10.3389/fnut.2021.647582] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/28/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer as a disease continues to ravage the world population without regard to sex, age, and race. Due to the growing number of cases worldwide, cancer exerts a significant negative impact on global health and the economy. Interestingly, chemotherapy has been used over the years as a therapeutic intervention against cancer. However, high cost, resistance, and toxic by-effects to treatment have overshadowed some of its benefits. In recent times, efforts have been ongoing in searching for anticancer therapeutics of plant origin, focusing on polyphenols. Urolithins are secondary polyphenol metabolites derived from the gut microbial action on ellagitannins and ellagic acid-rich foods such as pomegranate, berries, and nuts. Urolithins are emerging as a new class of anticancer compounds that can mediate their cancer-preventive activities through cell cycle arrest, aromatase inhibition, induction of apoptosis, tumor suppression, promotion of autophagy, and senescence, transcriptional regulation of oncogenes, and growth factor receptors. In this review, we discussed the growing shreds of evidence supporting these secondary phenolic metabolites' anticancer properties. Furthermore, we have pointed out some of the future directions needed to establish urolithins as anticancer agents.
Collapse
Affiliation(s)
- Sami A Al-Harbi
- Department of Chemistry, University College in Al-Jamoum, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Mazin A Zamzami
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Imran Khan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Downregulation of Cell Cycle and Checkpoint Genes by Class I HDAC Inhibitors Limits Synergism with G2/M Checkpoint Inhibitor MK-1775 in Bladder Cancer Cells. Genes (Basel) 2021; 12:genes12020260. [PMID: 33670166 PMCID: PMC7916885 DOI: 10.3390/genes12020260] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Since genes encoding epigenetic regulators are often mutated or deregulated in urothelial carcinoma (UC), they represent promising therapeutic targets. Specifically, inhibition of Class-I histone deacetylase (HDAC) isoenzymes induces cell death in UC cell lines (UCC) and, in contrast to other cancer types, cell cycle arrest in G2/M. Here, we investigated whether mutations in cell cycle genes contribute to G2/M rather than G1 arrest, identified the precise point of arrest and clarified the function of individual HDAC Class-I isoenzymes. Database analyses of UC tissues and cell lines revealed mutations in G1/S, but not G2/M checkpoint regulators. Using class I-specific HDAC inhibitors (HDACi) with different isoenzyme specificity (Romidepsin, Entinostat, RGFP966), cell cycle arrest was shown to occur at the G2/M transition and to depend on inhibition of HDAC1/2 rather than HDAC3. Since HDAC1/2 inhibition caused cell-type-specific downregulation of genes encoding G2/M regulators, the WEE1 inhibitor MK-1775 could not overcome G2/M checkpoint arrest and therefore did not synergize with Romidepsin inhibiting HDAC1/2. Instead, since DNA damage was induced by inhibition of HDAC1/2, but not of HDAC3, combinations between inhibitors of HDAC1/2 and of DNA repair should be attempted.
Collapse
|
11
|
Yang D, Ma Y, Zhao P, Ma J, He C. Systematic screening of protein-coding gene expression identified HMMR as a potential independent indicator of unfavorable survival in patients with papillary muscle-invasive bladder cancer. Biomed Pharmacother 2019; 120:109433. [PMID: 31568988 DOI: 10.1016/j.biopha.2019.109433] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022] Open
Abstract
Papillary and non-papillary are two histological patterns of bladder carcinogenesis and are considered as dual-track oncogenic pathways, which have different genetic alterations. The TCGA-bladder cancer (BLCA) database contains clinicopathological, genomic and survival data from over 400 muscle-invasive bladder cancer patients. In this study, using data from this database, we performed a systematic screening of gene expression to identify the protein-coding gene that might have prognostic value in papillary and non-papillary muscle-invasive bladder cancer (MIBC). The data of patients with primary MIBC in TCGA-BLCA was acquired from the UCSC Xena project (http://xena.ucsc.edu) for re-analysis. By setting |log2 fold change|≥2 and adjusted p value <0.01 as the screening criteria, we found 751 significantly dysregulated genes, including 183 overexpressed and 568 downregulated genes. HMMR was identified as a potential prognostic marker with unique expression. Multivariate analysis showed that its expression was an independent prognostic indicator of shorter progression-free survival (PFS) (HR: 1.400, 95%CI: 1.021-1.920, p = 0.037) in the papillary subtype. ENST00000393915.8 and ENST00000358715.3, two transcripts that contain all 18 exons and encode the full length of HMMR, were significantly upregulated in cancer tissues compared with normal bladder tissues. None of the 17 CpG sites in its DNA locus was relevant to HMMR expression. 26/403 (6.5%) MIBC cases had HMMR gene-level amplification, which was associated with upregulated HMMR expression compared with the copy-neutral and deletion groups. Gene set enrichment analysis (GSEA) in papillary MIBC found that the high HMMR expression group was associated with upregulated genes enriched in multiple gene sets with well-established role in BC development, including G2M checkpoint, E2 F Targets, Myc Targets V1, Myc Targets V2 and Glycolysis. Based on these findings, we infer that HMMR expression might be a specific prognostic marker in terms of PFS in papillary MIBC. DNA amplification might be an important mechanism of its elevation.
Collapse
Affiliation(s)
- Dong Yang
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yan Ma
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Pengcheng Zhao
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jing Ma
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Chaohong He
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
12
|
Deiss K, Lockwood N, Howell M, Segeren HA, Saunders RE, Chakravarty P, Soliman TN, Martini S, Rocha N, Semple R, Zalmas LP, Parker PJ. A genome-wide RNAi screen identifies the SMC5/6 complex as a non-redundant regulator of a Topo2a-dependent G2 arrest. Nucleic Acids Res 2019; 47:2906-2921. [PMID: 30590722 PMCID: PMC6451093 DOI: 10.1093/nar/gky1295] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 01/08/2023] Open
Abstract
The Topo2a-dependent arrest is associated with faithful segregation of sister chromatids and has been identified as dysfunctional in numerous tumour cell lines. This genome-protecting pathway is poorly understood and its characterization is of significant interest, potentially offering interventional opportunities in relation to synthetic lethal behaviours in arrest-defective tumours. Using the catalytic Topo2a inhibitor ICRF193, we have performed a genome-wide siRNA screen in arrest-competent, non-transformed cells, to identify genes essential for this arrest mechanism. In addition, we have counter-screened several DNA-damaging agents and demonstrate that the Topo2a-dependent arrest is genetically distinct from DNA damage checkpoints. We identify the components of the SMC5/6 complex, including the activity of the E3 SUMO ligase NSE2, as non-redundant players that control the timing of the Topo2a-dependent arrest in G2. We have independently verified the NSE2 requirement in fibroblasts from patients with germline mutations that cause severely reduced levels of NSE2. Through imaging Topo2a-dependent G2 arrested cells, an increased interaction between Topo2a and NSE2 is observed at PML bodies, which are known SUMOylation hotspots. We demonstrate that Topo2a is SUMOylated in an ICRF193-dependent manner by NSE2 at a novel non-canonical site (K1520) and that K1520 sumoylation is required for chromosome segregation but not the G2 arrest.
Collapse
Affiliation(s)
- Katharina Deiss
- Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nicola Lockwood
- Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Howell
- High Throughput Screening, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Hendrika Alida Segeren
- Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rebecca E Saunders
- High Throughput Screening, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Tanya N Soliman
- Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Silvia Martini
- Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nuno Rocha
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Robert Semple
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | | | - Peter J Parker
- Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- School of Cancer and Pharmaceutical Sciences King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
13
|
Brown A, Geiger H. Chromosome integrity checkpoints in stem and progenitor cells: transitions upon differentiation, pathogenesis, and aging. Cell Mol Life Sci 2018; 75:3771-3779. [PMID: 30066086 PMCID: PMC6154040 DOI: 10.1007/s00018-018-2891-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 07/22/2018] [Accepted: 07/25/2018] [Indexed: 01/30/2023]
Abstract
Loss of chromosome integrity is a major contributor to cancer. Checkpoints within the cell division cycle that facilitate the accuracy and outcome of chromosome segregation are thus critical pathways for preserving chromosome integrity and preventing chromosomal instability. The spindle assembly checkpoint, the decatenation checkpoint and the post-mitotic tetraploidy checkpoint ensure the appropriate establishment of the spindle apparatus, block mitotic entry upon entanglement of chromosomes or prevent further progression of post-mitotic cells that display massive spindle defects. Most of our knowledge on these mechanisms originates from studies conducted in yeast, cancer cell lines and differentiated cells. Considering that in many instances cancer derives from transformed stem and progenitor cells, our knowledge on these checkpoints in these cells just started to emerge. With this review, we provide a general overview of the current knowledge of these checkpoints in embryonic as well as in adult stem and progenitor cells with a focus on the hematopoietic system and outline common mis-regulations of their function associated with cancer and leukemia. Most cancers are aging-associated diseases. We will thus also discuss changes in the function and outcome of these checkpoints upon aging of stem and progenitor cells.
Collapse
Affiliation(s)
- Andreas Brown
- Institute of Molecular Medicine, Ulm University, Life Science Building N27, James Franck-Ring/Meyerhofstrasse, 89081, Ulm, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Life Science Building N27, James Franck-Ring/Meyerhofstrasse, 89081, Ulm, Germany.
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
14
|
Urolithins impair cell proliferation, arrest the cell cycle and induce apoptosis in UMUC3 bladder cancer cells. Invest New Drugs 2017. [PMID: 28631098 DOI: 10.1007/s10637-017-0483-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ellagitannins have been gaining attention as potential anticancer molecules. However, the low bioavailability of ellagitannins and their extensive metabolization in the gastrointestinal tract into ellagic acid and urolithins suggest that the health benefits of consuming ellagitannins rely on the direct effects of their metabolites. Recently, chemopreventive and chemotherapeutic activities were ascribed to urolithins. Nonetheless, there is still a need to screen and evaluate the selectivity of these molecules and to elucidate their cellular mechanisms of action. Therefore, this work focused on the antiproliferative effects of urolithins A, B and C and ellagic acid on different human tumor cell lines. The evaluation of cell viability and the determination of the half-maximal inhibitory concentrations indicated that the sensitivity to the studied urolithins varied markedly between the different cell lines, with the bladder cancer cells (UMUC3) being the most susceptible. In UMUC3 cells, urolithin A was the most active molecule, promoting cell cycle arrest at the G2/M checkpoint, increasing apoptotic cell death and inhibiting PI3K/Akt and MAPK signaling. Overall, the present study emphasizes the chemopreventive/chemotherapeutic potential of urolithins, highlighting the stronger effects of urolithin A and its potential to target transitional bladder cancer cells.
Collapse
|
15
|
Bower JJ, Vance LD, Psioda M, Smith-Roe SL, Simpson DA, Ibrahim JG, Hoadley KA, Perou CM, Kaufmann WK. Patterns of cell cycle checkpoint deregulation associated with intrinsic molecular subtypes of human breast cancer cells. NPJ Breast Cancer 2017; 3:9. [PMID: 28649649 PMCID: PMC5445620 DOI: 10.1038/s41523-017-0009-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/07/2017] [Indexed: 12/31/2022] Open
Abstract
Genomic instability is a hallmark of breast cancer, contributes to tumor heterogeneity, and influences chemotherapy resistance. Although Gap 2 and mitotic checkpoints are thought to prevent genomic instability, the role of these checkpoints in breast cancer is poorly understood. Here, we assess the Gap 2 and mitotic checkpoint functions of 24 breast cancer and immortalized mammary epithelial cell lines representing four of the six intrinsic molecular subtypes of breast cancer. We found that patterns of cell cycle checkpoint deregulation were associated with the intrinsic molecular subtype of breast cancer cell lines. Specifically, the luminal B and basal-like cell lines harbored two molecularly distinct Gap 2/mitosis checkpoint defects (impairment of the decatenation Gap 2 checkpoint and the spindle assembly checkpoint, respectively). All subtypes of breast cancer cell lines examined displayed aberrant DNA synthesis/Gap 2/mitosis progression and the basal-like and claudin-low cell lines exhibited increased percentages of chromatid cohesion defects. Furthermore, a decatenation Gap 2 checkpoint gene expression signature identified in the cell line panel correlated with clinical outcomes in breast cancer patients, suggesting that breast tumors may also harbor defects in decatenation Gap 2 checkpoint function. Taken together, these data imply that pharmacological targeting of signaling pathways driving these phenotypes may lead to the development of novel personalized treatment strategies for the latter two subtypes which currently lack targeted therapeutic options because of their triple negative breast cancer status.
Collapse
Affiliation(s)
- Jacquelyn J. Bower
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Leah D. Vance
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Matthew Psioda
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Stephanie L. Smith-Roe
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Dennis A. Simpson
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Joseph G. Ibrahim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Charles M. Perou
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - William K. Kaufmann
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
16
|
Xu P, Li J, Liu J, Wang J, Wu Z, Zhang X, Zhai Y. Mature adipocytes observed to undergo reproliferation and polyploidy. FEBS Open Bio 2017; 7:652-658. [PMID: 28469978 PMCID: PMC5407891 DOI: 10.1002/2211-5463.12207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/03/2017] [Accepted: 02/02/2017] [Indexed: 11/29/2022] Open
Abstract
Lipid‐filled mature adipocytes are important for the study of lipid metabolism and in the development of obesity, but whether they are capable of reproliferation is still controversial. Here, we monitored lipid droplet dynamics and adipocyte reproliferation in live, differentiated 3T3‐L1 cells using a phase‐contrast microscope in real time. Phase‐contrast microscopy achieves a similar visual effect in situ to that obtained using traditional dyes such as Oil Red O and BODIPY in vitro. Using this method, we captured the process that lipid droplets use for dynamic fusion in living cells. Unexpectedly, we acquired images of the moment that differentiated 3T3‐L1 cells containing lipid droplets entered mitosis. In addition, we observed some binucleated mature adipocytes. This information provides a better understanding of the adipocyte differentiation process.
Collapse
Affiliation(s)
- Pengfei Xu
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China
| | - Jiao Li
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China
| | - Jin Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China
| | - Jing Wang
- Department of Biology Science and Technology Baotou Teacher's College China
| | - Zekai Wu
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China
| | - Xiaotian Zhang
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry Institute of Cell Biology College of Life Sciences Beijing Normal University China
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development College of Life Sciences Beijing Normal University China.,Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry Institute of Cell Biology College of Life Sciences Beijing Normal University China
| |
Collapse
|
17
|
Jain CK, Majumder HK, Roychoudhury S. Natural Compounds as Anticancer Agents Targeting DNA Topoisomerases. Curr Genomics 2017; 18:75-92. [PMID: 28503091 PMCID: PMC5321768 DOI: 10.2174/1389202917666160808125213] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 12/14/2022] Open
Abstract
DNA topoisomerases are important cellular enzymes found in almost all types of living cells (eukaryotic and prokaryotic). These enzymes are essential for various DNA metabolic processes e.g. replication, transcription, recombination, chromosomal decatenation etc. These enzymes are important molecular drug targets and inhibitors of these enzymes are widely used as effective anticancer and antibacterial drugs. However, topoisomerase inhibitors have some therapeutic limitations and they exert serious side effects during cancer chemotherapy. Thus, development of novel anticancer topoisomerase inhibitors is necessary for improving cancer chemotherapy. Nature serves as a repertoire of structurally and chemically diverse molecules and in the recent years many DNA topoisomerase inhibitors have been identified from natural sources. The present review discusses anticancer properties and therapeutic importance of eighteen recently identified natural topoisomerase inhibitors (from the year 2009 to 2015). Structural characteristics of these novel inhibitors provide backbones for designing and developing new anticancer drugs.
Collapse
Affiliation(s)
- Chetan Kumar Jain
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Hemanta Kumar Majumder
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Susanta Roychoudhury
- Division of Research, Saroj Gupta Cancer Centre & Research Institute, M G Road, Thakurpukur, Kolkata-700 063, India
| |
Collapse
|
18
|
Spoerri L, Brooks K, Chia K, Grossman G, Ellis JJ, Dahmer-Heath M, Škalamera D, Pavey S, Burmeister B, Gabrielli B. A novel ATM-dependent checkpoint defect distinct from loss of function mutation promotes genomic instability in melanoma. Pigment Cell Melanoma Res 2016; 29:329-39. [PMID: 26854966 DOI: 10.1111/pcmr.12466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/03/2016] [Indexed: 11/29/2022]
Abstract
Melanomas have high levels of genomic instability that can contribute to poor disease prognosis. Here, we report a novel defect of the ATM-dependent cell cycle checkpoint in melanoma cell lines that promotes genomic instability. In defective cells, ATM signalling to CHK2 is intact, but the cells are unable to maintain the cell cycle arrest due to elevated PLK1 driving recovery from the arrest. Reducing PLK1 activity recovered the ATM-dependent checkpoint arrest, and over-expressing PLK1 was sufficient to overcome the checkpoint arrest and increase genomic instability. Loss of the ATM-dependent checkpoint did not affect sensitivity to ionizing radiation demonstrating that this defect is distinct from ATM loss of function mutations. The checkpoint defective melanoma cell lines over-express PLK1, and a significant proportion of melanomas have high levels of PLK1 over-expression suggesting this defect is a common feature of melanomas. The inability of ATM to impose a cell cycle arrest in response to DNA damage increases genomic instability. This work also suggests that the ATM-dependent checkpoint arrest is likely to be defective in a higher proportion of cancers than previously expected.
Collapse
Affiliation(s)
- Loredana Spoerri
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Kelly Brooks
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - KeeMing Chia
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Gavriel Grossman
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Jonathan J Ellis
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Mareike Dahmer-Heath
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Dubravka Škalamera
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Sandra Pavey
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Bryan Burmeister
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
- Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - Brian Gabrielli
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
19
|
PTEN stabilizes TOP2A and regulates the DNA decatenation. Sci Rep 2015; 5:17873. [PMID: 26657567 PMCID: PMC4674714 DOI: 10.1038/srep17873] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/06/2015] [Indexed: 11/12/2022] Open
Abstract
PTEN is a powerful tumor suppressor that antagonizes the cytoplasmic PI3K-AKT pathway and suppresses cellular proliferation. PTEN also plays a role in the maintenance of genomic stability in the nucleus. Here we report that PTEN facilitates DNA decatenation and controls a decatenation checkpoint. Catenations of DNA formed during replication are decatenated by DNA topoisomerase II (TOP2), and this process is actively monitored by a decatenation checkpoint in G2 phase. We found that PTEN deficient cells form ultra-fine bridges (UFBs) during anaphase and these bridges are generated as a result of insufficient decatenation. We show that PTEN is physically associated with a decatenation enzyme TOP2A and that PTEN influences its stability through OTUD3 deubiquitinase. In the presence of PTEN, ubiquitination of TOP2A is inhibited by OTUD3. Deletion or deficiency of PTEN leads to down regulation of TOP2A, dysfunction of the decatenation checkpoint and incomplete DNA decatenation in G2 and M phases. We propose that PTEN controls DNA decatenation to maintain genomic stability and integrity.
Collapse
|
20
|
Jain CK, Roychoudhury S, Majumder HK. Selective killing of G2 decatenation checkpoint defective colon cancer cells by catalytic topoisomerase II inhibitor. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1195-204. [PMID: 25746763 DOI: 10.1016/j.bbamcr.2015.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/09/2015] [Accepted: 02/25/2015] [Indexed: 12/16/2022]
Abstract
Cancer cells with defective DNA decatenation checkpoint can be selectively targeted by the catalytic inhibitors of DNA topoisomerase IIα (topo IIα) enzyme. Upon treatment with catalytic topo IIα inhibitors, cells with defective decatenation checkpoint fail to arrest their cell cycle in G2 phase and enter into M phase with catenated and under-condensed chromosomes resulting into impaired mitosis and eventually cell death. In the present work we analyzed decatenation checkpoint in five different colon cancer cell lines (HCT116, HT-29, Caco2, COLO 205 and SW480) and in one non-cancerous cell line (HEK293T). Four out of the five colon cancer cell lines i.e. HCT116, HT-29, Caco2, and COLO 205 were found to be compromised for the decatenation checkpoint function at different extents, whereas SW480 and HEK293T cell lines were found to be proficient for the checkpoint function. Upon treatment with ICRF193, decatenation checkpoint defective cell lines failed to arrest the cell cycle in G2 phase and entered into M phase without proper chromosomal decatenation, resulting into the formation of tangled mass of catenated and under-condensed chromosomes. Such cells underwent mitotic catastrophe and rapid apoptosis like cell death and showed higher sensitivity for ICRF193. Our study suggests that catalytic inhibitors of topoisomerase IIα are promising therapeutic agents for the treatment of colon cancers with defective DNA decatenation checkpoint.
Collapse
Affiliation(s)
- Chetan Kumar Jain
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India; Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Susanta Roychoudhury
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Hemanta Kumar Majumder
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
21
|
Brownlow N, Pike T, Zicha D, Collinson L, Parker PJ. Mitotic catenation is monitored and resolved by a PKCε-regulated pathway. Nat Commun 2014; 5:5685. [PMID: 25483024 PMCID: PMC4272242 DOI: 10.1038/ncomms6685] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022] Open
Abstract
Exit from mitosis is controlled by silencing of the spindle assembly checkpoint (SAC). It is important that preceding exit, all sister chromatid pairs are correctly bioriented, and that residual catenation is resolved, permitting complete sister chromatid separation in the ensuing anaphase. Here we determine that the metaphase response to catenation in mammalian cells operates through PKCε. The PKCε-controlled pathway regulates exit from the SAC only when mitotic cells are challenged by retained catenation and this delayed exit is characterized by BubR1-high and Mad2-low kinetochores. In addition, we show that this pathway is necessary to facilitate resolution of retained catenanes in mitosis. When delayed by catenation in mitosis, inhibition of PKCε results in premature entry into anaphase with PICH-positive strands and chromosome bridging. These findings demonstrate the importance of PKCε-mediated regulation in protection from loss of chromosome integrity in cells failing to resolve catenation in G2.
Collapse
Affiliation(s)
- Nicola Brownlow
- Protein Phosphorylation Laboratory, Cancer Research UK London
Research Institute, 44 Lincolns Inn Fields, London
WC2A 3LY, UK
| | - Tanya Pike
- Protein Phosphorylation Laboratory, Cancer Research UK London
Research Institute, 44 Lincolns Inn Fields, London
WC2A 3LY, UK
| | - Daniel Zicha
- Light Microscopy, Cancer Research UK London Research
Institute, London, WC2A 3LY, UK
| | - Lucy Collinson
- Electron Microscopy, Cancer Research UK London Research
Institute, London
WC2A 3LY, UK
| | - Peter J. Parker
- Protein Phosphorylation Laboratory, Cancer Research UK London
Research Institute, 44 Lincolns Inn Fields, London
WC2A 3LY, UK
- Division of Cancer Studies, King’s College London,
New Hunt’s House, Guy’s Campus, London
SE1 1UL, UK
| |
Collapse
|
22
|
Wong YH, Li CW, Chen BS. Evolution of network biomarkers from early to late stage bladder cancer samples. BIOMED RESEARCH INTERNATIONAL 2014; 2014:159078. [PMID: 25309904 PMCID: PMC4189772 DOI: 10.1155/2014/159078] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 11/29/2022]
Abstract
We use a systems biology approach to construct protein-protein interaction networks (PPINs) for early and late stage bladder cancer. By comparing the networks of these two stages, we find that both networks showed very significantly different mechanisms. To obtain the differential network structures between cancer and noncancer PPINs, we constructed cancer PPIN and noncancer PPIN network structures for the two bladder cancer stages using microarray data from cancer cells and their adjacent noncancer cells, respectively. With their carcinogenesis relevance values (CRVs), we identified 152 and 50 significant proteins and their PPI networks (network markers) for early and late stage bladder cancer by statistical assessment. To investigate the evolution of network biomarkers in the carcinogenesis process, primary pathway analysis showed that the significant pathways of early stage bladder cancer are related to ordinary cancer mechanisms, while the ribosome pathway and spliceosome pathway are most important for late stage bladder cancer. Their only intersection is the ubiquitin mediated proteolysis pathway in the whole stage of bladder cancer. The evolution of network biomarkers from early to late stage can reveal the carcinogenesis of bladder cancer. The findings in this study are new clues specific to this study and give us a direction for targeted cancer therapy, and it should be validated in vivo or in vitro in the future.
Collapse
Affiliation(s)
- Yung-Hao Wong
- Lab of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Wei Li
- Lab of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Bor-Sen Chen
- Lab of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
23
|
Brooks K, Chia KM, Spoerri L, Mukhopadhyay P, Wigan M, Stark M, Pavey S, Gabrielli B. Defective Decatenation Checkpoint Function Is a Common Feature of Melanoma. J Invest Dermatol 2014; 134:150-158. [DOI: 10.1038/jid.2013.264] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 04/17/2013] [Accepted: 04/30/2013] [Indexed: 12/16/2022]
|
24
|
Li XM, Yu C, Wang ZW, Zhang YL, Liu XM, Zhou D, Sun QY, Fan HY. DNA topoisomerase II is dispensable for oocyte meiotic resumption but is essential for meiotic chromosome condensation and separation in mice. Biol Reprod 2013; 89:118. [PMID: 24048577 DOI: 10.1095/biolreprod.113.110692] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
During mitosis, DNA topoisomerase II (TOP2) is required for sister chromatid separation. When TOP2 activity is inhibited, a decatenation checkpoint is activated by entangled chromatin. However, the functions of TOP2 in oocyte meiosis, particularly for homologous chromosome segregation during meiosis I, have not been investigated. In addition, it remains unknown if TOP2 inhibition activates a decatenation checkpoint at the G2/M transition in oocytes. In this study, we used mouse oocytes and specific inhibitors of TOP2 (ICRF-193 and etoposide) to investigate the role of TOP2 in meiosis. Our results indicated that an effective decatenation checkpoint did not exist in fully grown oocytes, as oocytes underwent the G2/M transition and reinitiated meiosis even when TOP2 activity was inhibited. However, oocytes treated with ICRF-193 had severe defects in chromosome condensation and homologous chromosome separation. Furthermore, condensed chromosomes failed to maintain their normal configurations in matured oocytes that were treated with ICRF-193. However, sister chromatid separation and subsequent chromosome decondensation during the exit from meiosis were not blocked by TOP2 inhibitors. These results indicated that TOP2 had a specific, crucial function in meiosis I. Thus, we identified important functions of TOP2 during oocyte maturation and provided novel insights into the decatenation checkpoint during meiosis.
Collapse
Affiliation(s)
- Xiao-Meng Li
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Chung KS, Choi HE, Shin JS, Cho YW, Choi JH, Cho WJ, Lee KT. 6,7-Dimethoxy-3-(3-methoxyphenyl)isoquinolin-1-amine induces mitotic arrest and apoptotic cell death through the activation of spindle assembly checkpoint in human cervical cancer cells. Carcinogenesis 2013; 34:1852-60. [DOI: 10.1093/carcin/bgt133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
26
|
Li BZ, Chen ZL, Shi SS, Feng XL, Tan XG, Zhou F, He J. Overexpression of Cdc25C predicts response to radiotherapy and survival in esophageal squamous cell carcinoma patients treated with radiotherapy followed by surgery. CHINESE JOURNAL OF CANCER 2013; 32:403-9. [PMID: 23470146 PMCID: PMC3845600 DOI: 10.5732/cjc.012.10233] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Biomarker identification is crucial for the selection of patients who might benefit from radiotherapy. To explore potential markers for response and prognosis in patients with locally advanced esophageal carcinoma treated with radiotherapy followed by surgery, we evaluated the expression of cell cycle checkpoint-related proteins Chk2, Cdc25C, and Cyclin D1. A total of 56 patients with locally advanced esophageal squamous cell carcinoma were treated with radiotherapy followed by surgery. Pretreatment tumor biopsy specimens were analyzed for Chk2, Cdc25C, and Cyclin D1 expression by immunohistochemistry. High expression of Chk2, Cyclin D1, and Cdc25C was observed in 44 (78.6%), 15 (26.8%), and 27 (48.2%) patients, respectively. The median survival was 16 months (range, 3–154 months), with a 5-year overall survival rate of 19.6%. Overexpression of Chk2 was associated with smoking (P = 0.021), overexpression of Cdc25C was associated with patient age (P = 0.033) and tumor length (P = 0.001), and overexpression of Cdc25C was associated with pathologic complete response (P = 0.038). Univariate analysis demonstrated that overexpression of Cdc25C and pathologic complete response was associated with better survival. In multivariate analysis, Cdc25C was the most significant independent predictor of better survival (P = 0.014) for patients treated with radiotherapy followed by surgery. Overexpression of Cdc25C was significantly associated with pathologic complete response and better survival of patients with locally advanced esophageal cancer treated with radiotherapy followed by surgery. These results suggest that Cdc25C may be a biomarker of treatment response and good prognosis for esophageal carcinoma patients. Thus, immunohistochemical staining of Cdc25C in a pretreatment specimen may be a useful method of identifying optimal treatment for patients with esophageal carcinoma.
Collapse
Affiliation(s)
- Bao-Zhong Li
- Departments of Thoracic Surgery, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
27
|
Kesseler KJ, Blinov ML, Elston TC, Kaufmann WK, Simpson DA. A predictive mathematical model of the DNA damage G2 checkpoint. J Theor Biol 2013; 320:159-69. [PMID: 23266715 PMCID: PMC3654547 DOI: 10.1016/j.jtbi.2012.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 11/21/2012] [Accepted: 12/10/2012] [Indexed: 12/21/2022]
Abstract
A predictive mathematical model of the transition from the G2 phase in the cell cycle to mitosis (M) was constructed from the known interactions of the proteins that are thought to play significant roles in the G2 to M transition as well as the DNA damage- induced G2 checkpoint. The model simulates the accumulation of active cyclin B1/Cdk1 (MPF) complexes in the nucleus to activate mitosis, the inhibition of this process by DNA damage, and transport of component proteins between cytoplasm and nucleus. Interactions in the model are based on activities of individual phospho-epitopes and binding sites of proteins involved in G2/M. Because tracking phosphoforms leads to combinatorial explosion, we employ a rule-based approach using the BioNetGen software. The model was used to determine the effects of depletion or over-expression of selected proteins involved in the regulation of the G2 to M transition in the presence and absence of DNA damage. Depletion of Plk1 delayed mitotic entry and recovery from the DNA damage-induced G2 arrest and over-expression of MPF attenuated the DNA damage-induced G2 delay. The model recapitulates the G2 delay observed in the biological response to varying levels of a DNA damage signal. The model produced the novel prediction that depletion of pkMyt1 results in an abnormal biological state in which G2 cells with DNA damage accumulate inactive nuclear MPF. Such a detailed model may prove useful for predicting DNA damage G2 checkpoint function in cancer and, therefore, sensitivity to cancer therapy.
Collapse
Affiliation(s)
- Kevin J. Kesseler
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, NC 27599-7255, USA
| | - Michael L. Blinov
- Center for Cell Analysis and Modeling, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-1507, USA
| | - Timothy C. Elston
- Department of Pharmacology, University of North Carolina at Chapel Hill,Chapel Hill, NC 27599-7260, USA
| | - William K. Kaufmann
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, NC 27599-7255, USA
| | - Dennis A. Simpson
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, NC 27599-7255, USA
| |
Collapse
|
28
|
Ou TT, Wang CJ, Lee YS, Wu CH, Lee HJ. Gallic acid induces G2/M phase cell cycle arrest via
regulating 14-3-3β release from Cdc25C and Chk2 activation in human bladder transitional carcinoma cells. Mol Nutr Food Res 2010; 54:1781-90. [DOI: 10.1002/mnfr.201000096] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
29
|
Bower JJ, Karaca GF, Zhou Y, Simpson DA, Cordeiro-Stone M, Kaufmann WK. Topoisomerase IIalpha maintains genomic stability through decatenation G(2) checkpoint signaling. Oncogene 2010; 29:4787-99. [PMID: 20562910 PMCID: PMC2928865 DOI: 10.1038/onc.2010.232] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Topoisomerase IIalpha (topoIIalpha) is an essential mammalian enzyme that topologically modifies DNA and is required for chromosome segregation during mitosis. Previous research suggests that inhibition of topoII decatenatory activity triggers a G(2) checkpoint response, which delays mitotic entry because of insufficient decatenation of daughter chromatids. Here we examine the effects of both topoIIalpha and topoIIbeta on decatenatory activity in cell extracts, DNA damage and decatenation G(2) checkpoint function, and the frequencies of p16(INK4A) allele loss and gain. In diploid human fibroblast lines, depletion of topoIIalpha by small-interfering RNA was associated with severely reduced decatenatory activity, delayed progression from G(2) into mitosis and insensitivity to G(2) arrest induced by the topoII catalytic inhibitor ICRF-193. Furthermore, interphase nuclei of topoIIalpha-depleted cells showed increased frequencies of losses and gains of the tumor suppressor genetic locus p16(INK4A). This study shows that the topoIIalpha protein is required for decatenation G(2) checkpoint function, and inactivation of decatenation and the decatenation G(2) checkpoint leads to abnormal chromosome segregation and genomic instability.
Collapse
Affiliation(s)
- J J Bower
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | | |
Collapse
|
30
|
Bower JJ, Zhou Y, Zhou T, Simpson DA, Arlander SJ, Paules RS, Cordeiro-Stone M, Kaufmann WK. Revised genetic requirements for the decatenation G2 checkpoint: the role of ATM. Cell Cycle 2010; 9:1617-28. [PMID: 20372057 DOI: 10.4161/cc.9.8.11470] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The decatenation G2 checkpoint is proposed to delay cellular progression from G2 into mitosis when intertwined daughter chromatids are insufficiently decatenated. Previous studies indicated that the ATM- and Rad3-related (ATR) checkpoint kinase, but not the ataxia telangiectasia-mutated (ATM) kinase, was required for decatenation G2 checkpoint function. Here, we show that the method used to quantify decatenation G2 checkpoint function can influence the identification of genetic requirements for the checkpoint. Normal human diploid fibroblast (NHDF) lines responded to the topoisomerase II (topo II) catalytic inhibitor ICRF-193 with a stringent G2 arrest and a reduction in the mitotic index. While siRNA-mediated depletion of ATR and CHEK1 increased the mitotic index in ICRF-193 treated NHDF lines, depletion of these proteins did not affect the mitotic entry rate, indicating that the decatenation G2 checkpoint was functional. These results suggest that ATR and CHEK1 are not required for the decatenation G2 checkpoint, but may influence mitotic exit after inhibition of topo II. A re-evaluation of ataxia telangiectasia (AT) cell lines using the mitotic entry assay indicated that ATM was required for the decatenation G2 checkpoint. Three NHDF cell lines responded to ICRF-193 with a mean 98% inhibition of the mitotic entry rate. Examination of the mitotic entry rates in AT fibroblasts upon treatment with ICRF-193 revealed a significantly attenuated decatenation G2 checkpoint response, with a mean 59% inhibition of the mitotic entry rate. In addition, a normal lymphoblastoid line exhibited a 95% inhibition of the mitotic entry rate after incubation with ICRF-193, whereas two AT lymphoblastoid lines displayed only 36% and 20% inhibition of the mitotic entry rate. Stable depletion of ATM in normal human fibroblasts with short hairpin RNA also attenuated decatenation G2 checkpoint function by an average of 40%. Western immunoblot analysis demonstrated that treatment with ICRF-193 induced ATM autophosphorylation and ATM-dependent phosphorylation of Ser15-p53 and Thr68 in Chk2, but no appreciable phosphorylation of Ser139-H2AX or Ser345-Chk1. The results suggest that inhibition of topo II induces ATM to phosphorylate selected targets that contribute to a G2 arrest independently of DNA damage.
Collapse
Affiliation(s)
- Jacquelyn J Bower
- Department of Pathology and Laboratory Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Polyploidy: Mechanisms and Cancer Promotion in Hematopoietic and Other Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 676:105-22. [DOI: 10.1007/978-1-4419-6199-0_7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Yu M, Ma M, Huang C, Yang H, Lai J, Yan S, Li L, Xiang M, Tan D. Correlation of expression of human arrest-defective-1 (hARD1) protein with breast cancer. Cancer Invest 2009; 27:978-83. [PMID: 19909012 DOI: 10.3109/07357900902769723] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human arrest-defective-1 (hARD1) was reported to be important in regulating cell cycle and promoting lung cancer cell proliferation. Here we have investigated the correlation between hARD1 and breast cancer. Analysis with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and flow cytometry (FCM) demonstrated that overexpression of hARD1 was associated with increased proliferation of MCF-7 cell, a human breast cancer cell line. Western blotting and immunohistochemical staining assay showed that hARD1 presented higher in breast cancer tissue than the adjacent tissue; accumulation of hARD1 protein was higher in 86% (37/43) of breast cancer, far more than noncancer samples. Our results suggest that hARD1 might play an important role in breast cancer carcinogenesis.
Collapse
Affiliation(s)
- Min Yu
- Laboratory of Biochemistry and Molecular Biology, School of Life Science, Yunnan University, Kunming, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Skladanowski A, Bozko P, Sabisz M. DNA structure and integrity checkpoints during the cell cycle and their role in drug targeting and sensitivity of tumor cells to anticancer treatment. Chem Rev 2009; 109:2951-73. [PMID: 19522503 DOI: 10.1021/cr900026u] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Andrzej Skladanowski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland.
| | | | | |
Collapse
|
34
|
Romanenko A, Kakehashi A, Morimura K, Wanibuchi H, Wei M, Vozianov A, Fukushima S. Urinary bladder carcinogenesis induced by chronic exposure to persistent low-dose ionizing radiation after Chernobyl accident. Carcinogenesis 2009; 30:1821-31. [PMID: 19643821 DOI: 10.1093/carcin/bgp193] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Urinary bladder urothelium as well as cells in the microenvironment of lamina propria (endothelial elements, fibroblasts and lymphocytes) demonstrate a number of responses to chronic persistent long-term, low-dose ionizing radiation (IR). Thus, oxidative stress occurs, accompanied by up-regulation of at least two signaling pathways (p38 mitogen-activated protein kinase and nuclear factor-kappaB cascades) and activation of growth factor receptors, in the bladder urothelium of people living in Cesium 137-contaminated areas of Ukraine, resulting in chronic inflammation and the development of proliferative atypical cystitis, so-called Chernobyl cystitis, which is considered a possible pre-neoplastic condition in humans. Furthermore, significant alterations in regulation of cell cycle transitions are associated with increased cell proliferation, along with up-regulated ubiquitination and sumoylation processes as well as inefficient DNA repair (base and nucleotide excision repair pathways) in the affected urothelium. The microenvironmental changes induced by chronic long-term, low-dose IR also appear to promote angiogenesis and remodeling of the extracellular matrix that could facilitate invasion as well as progression of pre-existing initiated cells to malignancy. Based on the available findings, new strategies have been developed for predicting and treatment of Chernobyl cystitis-a first step in urinary bladder carcinogenesis in humans.
Collapse
Affiliation(s)
- Alina Romanenko
- Department of Pathology, Institute of Urology, Academy of Medical Sciences of Ukraine, 9a, Yu. Kotzubinsky Street, 04053 Kiev, Ukraine
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
After DNA replication, sister chromatids must be untangled, or decatenated, before mitosis so that chromatids do not tear during anaphase. Topoisomerase IIalpha (Topo IIalpha) is the major decatenating enzyme. Topo IIalpha inhibitors prevent decatenation, causing cells to arrest during mitosis. Here we report that acute myeloid leukemia cells fail to arrest at the mitotic decatenation checkpoint, and their progression through this checkpoint is regulated by the DNA repair component Metnase (also termed SETMAR). Metnase contains a SET histone methylase and transposase nuclease domain, and is a component of the nonhomologous end-joining DNA double-strand break repair pathway. Metnase interacts with Topo IIalpha and enhances its decatenation activity. Here we show that multiple types of acute leukemia cells have an attenuated mitotic arrest when decatenation is inhibited and that in an acute myeloid leukemia (AML) cell line this is mediated by Metnase. Of further importance, Metnase permits continued proliferation of these AML cells even in the presence of the clinical Topo IIalpha inhibitor VP-16. In vitro, purified Metnase prevents VP-16 inhibition of Topo IIalpha decatenation of tangled DNA. Thus, Metnase expression levels may predict AML resistance to Topo IIalpha inhibitors, and Metnase is a potential therapeutic target for small molecule interference.
Collapse
|
36
|
Ho SY, Chen WC, Chiu HW, Lai CS, Guo HR, Wang YJ. Combination treatment with arsenic trioxide and irradiation enhances apoptotic effects in U937 cells through increased mitotic arrest and ROS generation. Chem Biol Interact 2009; 179:304-13. [DOI: 10.1016/j.cbi.2008.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 12/10/2008] [Accepted: 12/19/2008] [Indexed: 12/17/2022]
|
37
|
Wray J, Williamson EA, Royce M, Shaheen M, Beck BD, Lee SH, Nickoloff JA, Hromas R. Metnase mediates resistance to topoisomerase II inhibitors in breast cancer cells. PLoS One 2009; 4:e5323. [PMID: 19390626 PMCID: PMC2669129 DOI: 10.1371/journal.pone.0005323] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Accepted: 03/23/2009] [Indexed: 12/25/2022] Open
Abstract
DNA replication produces tangled, or catenated, chromatids, that must be decatenated prior to mitosis or catastrophic genomic damage will occur. Topoisomerase IIalpha (Topo IIalpha) is the primary decatenating enzyme. Cells monitor catenation status and activate decatenation checkpoints when decatenation is incomplete, which occurs when Topo IIalpha is inhibited by chemotherapy agents such as the anthracyclines and epididophyllotoxins. We recently demonstrated that the DNA repair component Metnase (also called SETMAR) enhances Topo IIalpha-mediated decatenation, and hypothesized that Metnase could mediate resistance to Topo IIalpha inhibitors. Here we show that Metnase interacts with Topo IIalpha in breast cancer cells, and that reducing Metnase expression significantly increases metaphase decatenation checkpoint arrest. Repression of Metnase sensitizes breast cancer cells to Topo IIalpha inhibitors, and directly blocks the inhibitory effect of the anthracycline adriamycin on Topo IIalpha-mediated decatenation in vitro. Thus, Metnase may mediate resistance to Topo IIalpha inhibitors, and could be a biomarker for clinical sensitivity to anthracyclines. Metnase could also become an important target for combination chemotherapy with current Topo IIalpha inhibitors, specifically in anthracycline-resistant breast cancer.
Collapse
Affiliation(s)
- Justin Wray
- Division of Hematology-Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, New Mexico, United States of America
| | - Elizabeth A. Williamson
- Division of Hematology-Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, New Mexico, United States of America
| | - Melanie Royce
- Division of Hematology-Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, New Mexico, United States of America
| | - Montaser Shaheen
- Division of Hematology-Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, New Mexico, United States of America
| | - Brian D. Beck
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Suk-Hee Lee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Jac A. Nickoloff
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Robert Hromas
- Division of Hematology-Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
38
|
Abstract
The nucleotide sequence of DNA is the repository of hereditary information. Yet, it is now clear that the DNA itself plays an active role in regulating the ability of the cell to extract its information. Basic biological processes, including control of gene transcription, faithful DNA replication and segregation, maintenance of the genome and cellular differentiation are subject to the conformational and topological properties of DNA in addition to the regulation imparted by the sequence itself. How do these DNA features manifest such striking effects and how does the cell regulate them? In this review, we describe how misregulation of DNA topology can lead to cellular dysfunction. We then address how cells prevent these topological problems. We close with a discussion on recent theoretical advances indicating that the topological problems, themselves, can provide the cues necessary for their resolution by type-2 topoisomerases.
Collapse
Affiliation(s)
- Zhirong Liu
- College of Chemistry and Molecular Engineering, and Center for Theoretical Biology, Peking University, Beijing 100871, China
| | | | | | | |
Collapse
|
39
|
Tang Y, Simoneau AR, Xie J, Shahandeh B, Zi X. Effects of the kava chalcone flavokawain A differ in bladder cancer cells with wild-type versus mutant p53. Cancer Prev Res (Phila) 2009; 1:439-51. [PMID: 19138991 DOI: 10.1158/1940-6207.capr-08-0165] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Flavokawain A is the predominant chalcone from kava extract. We have assessed the mechanisms of flavokawain A's action on cell cycle regulation. In a p53 wild-type, low-grade, and papillary bladder cancer cell line (RT4), flavokawain A increased p21/WAF1 and p27/KIP1, which resulted in a decrease in cyclin-dependent kinase-2 (CDK2) kinase activity and subsequent G(1) arrest. The increase of p21/WAF1 protein corresponded to an increased mRNA level, whereas p27/KIP1 accumulation was associated with the down-regulation of SKP2, which then increased the stability of the p27/KIP1 protein. The accumulation of p21/WAF1 and p27/KIP1 was independent of cell cycle position and thus not a result of the cell cycle arrest. In contrast, flavokawain A induced a G(2)-M arrest in six p53 mutant-type, high-grade bladder cancer cell lines (T24, UMUC3, TCCSUP, 5637, HT1376, and HT1197). Flavokawain A significantly reduced the expression of CDK1-inhibitory kinases, Myt1 and Wee1, and caused cyclin B1 protein accumulation leading to CDK1 activation in T24 cells. Suppression of p53 expression by small interfering RNA in RT4 cells restored Cdc25C expression and down-regulated p21/WAF1 expression, which allowed Cdc25C and CDK1 activation, which then led to a G(2)-M arrest and an enhanced growth-inhibitory effect by flavokawain A. Consistently, flavokawain A also caused a pronounced CDK1 activation and G(2)-M arrest in p53 knockout but not in p53 wild-type HCT116 cells. This selectivity of flavokawain A for inducing a G(2)-M arrest in p53-defective cells deserves further investigation as a new mechanism for the prevention and treatment of bladder cancer.
Collapse
Affiliation(s)
- Yaxiong Tang
- 1Department of Urology, University of California, Irvine, Orange, California 92868, USA
| | | | | | | | | |
Collapse
|
40
|
Sullivan KD, Mullen TE, Marzluff WF, Wagner EJ. Knockdown of SLBP results in nuclear retention of histone mRNA. RNA (NEW YORK, N.Y.) 2009; 15:459-72. [PMID: 19155325 PMCID: PMC2657014 DOI: 10.1261/rna.1205409] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 11/14/2008] [Indexed: 05/23/2023]
Abstract
Histone mRNAs are the only eukaryotic cellular mRNAs that are not polyadenylated. Synthesis of mature histone mRNA requires only a single processing reaction: an endonucleolytic cleavage between a conserved stem-loop and a purine-rich downstream element to form the 3' end. The stem-loop binding protein (SLBP) is required for processing, and following processing, histone mRNA is transported to the cytoplasm, where SLBP participates in translation of the histone mRNA and is also involved in regulation of histone mRNA degradation. Here we present an analysis of histone mRNA metabolism in cells with highly reduced levels of SLBP using RNA interference. Knocking down SLBP in U2OS cells results in a reduction in the rate of cell growth and an accumulation of cells in S-phase. Surprisingly, there is only a modest (twofold) decrease in histone mRNA levels. Much of histone mRNA in the SLBP knockdown cells is properly processed but is retained in the nucleus. The processed histone mRNA in SLBP knockdown cells is not rapidly degraded when DNA replication is inhibited. These results suggest a previously undescribed role for SLBP in histone mRNA export.
Collapse
Affiliation(s)
- Kelly D Sullivan
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
41
|
Kaufmann WK. Analysis of the topoisomerase II-dependent decatenation G2 checkpoint and checkpoint kinases in human cells. Methods Mol Biol 2009; 582:155-66. [PMID: 19763949 DOI: 10.1007/978-1-60761-340-4_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Inhibition of Topo II function using poisons and catalytic inhibitors triggers checkpoints that act to delay progression of G2 cells into mitosis. Topo II poisons induce Topo II-associated DNA double-strand breaks that activate ATM and the DNA damage G2 checkpoint. Topo II catalytic inhibitors do not induce DNA double-strand breaks but block decatenation of intertwined daughter chromatids. Complete decatenation before anaphase of mitosis is required for chromatid segregation. G2 cells appear to sense the degree of chromatid arm catenations and actively delay the onset of mitosis by sustaining the inhibition of mitosis-promoting factor (MPF) and polo-like kinase 1 (Plk-1) kinase activities that normally propel G2 cells into mitosis. This chapter details the methods for assay of decatenation G2 checkpoint function and checkpoint kinase activities.
Collapse
Affiliation(s)
- William K Kaufmann
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, US
| |
Collapse
|
42
|
Abstract
DNA topoisomerases are enzymes that alter the topology of DNA. They have important functions in DNA replication, transcription, Holliday junction dissolution, chromosome condensation, and sister chromatid separation. Deficiencies in these enzymes are associated with diseases that result from genome instability. The last 10-15 years has seen a great deal of exciting research in the field of topoisomerase. Here we discuss a selection of the new themes that have been recently introduced into the already large body of topoisomerase research.
Collapse
Affiliation(s)
- Duncan J Clarke
- Department of Genetics, University of Minnesota, Medical School, Minneapolis, MN, USA
| | | |
Collapse
|
43
|
Luo K, Yuan J, Chen J, Lou Z. Topoisomerase IIalpha controls the decatenation checkpoint. Nat Cell Biol 2008; 11:204-10. [PMID: 19098900 DOI: 10.1038/ncb1828] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 10/27/2008] [Indexed: 01/27/2023]
Abstract
Topoisomerase II (Topo II) is required to separate intertwined sister chromatids before chromosome segregation can occur in mitosis. However, it remains to be resolved whether Topo II has any role in checkpoint control. Here we report that when phosphorylated, Ser 1524 of Topo IIalpha acts as a binding site for the BRCT domain of MDC1 (mediator of DNA damage checkpoint protein-1), thereby recruiting MDC1 to chromatin. Although Topo IIalpha-MDC1 interaction is not required for checkpoint activation induced by DNA damage, it is required for activation of the decatenation checkpoint. Mutation of Ser 1524 results in a defective decatenation checkpoint. These results reveal an important role of Topo II in checkpoint activation and in the maintenance of genomic stability.
Collapse
Affiliation(s)
- Kuntian Luo
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
44
|
Wu YC, Yen WY, Yih LH. Requirement of a functional spindle checkpoint for arsenite-induced apoptosis. J Cell Biochem 2008; 105:678-87. [DOI: 10.1002/jcb.21861] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
45
|
Williamson EA, Rasila KK, Corwin LK, Wray J, Beck BD, Severns V, Mobarak C, Lee SH, Nickoloff JA, Hromas R. The SET and transposase domain protein Metnase enhances chromosome decatenation: regulation by automethylation. Nucleic Acids Res 2008; 36:5822-31. [PMID: 18790802 PMCID: PMC2566874 DOI: 10.1093/nar/gkn560] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Metnase is a human SET and transposase domain protein that methylates histone H3 and promotes DNA double-strand break repair. We now show that Metnase physically interacts and co-localizes with Topoisomerase IIα (Topo IIα), the key chromosome decatenating enzyme. Metnase promotes progression through decatenation and increases resistance to the Topo IIα inhibitors ICRF-193 and VP-16. Purified Metnase greatly enhanced Topo IIα decatenation of kinetoplast DNA to relaxed circular forms. Nuclear extracts containing Metnase decatenated kDNA more rapidly than those without Metnase, and neutralizing anti-sera against Metnase reversed that enhancement of decatenation. Metnase automethylates at K485, and the presence of a methyl donor blocked the enhancement of Topo IIα decatenation by Metnase, implying an internal regulatory inhibition. Thus, Metnase enhances Topo IIα decatenation, and this activity is repressed by automethylation. These results suggest that cancer cells could subvert Metnase to mediate clinically relevant resistance to Topo IIα inhibitors.
Collapse
Affiliation(s)
- Elizabeth A. Williamson
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kanwaldeep Kaur Rasila
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Lori Kwan Corwin
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Justin Wray
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Brian D. Beck
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Virginia Severns
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Charlotte Mobarak
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Suk-Hee Lee
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Jac A. Nickoloff
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Robert Hromas
- Division of Hematology–Oncology, Cancer Research and Treatment Center, Department of Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- *To whom correspondence should be addressed. Tel: +1 505 272 5837; Fax: +1 505 272 5865;
| |
Collapse
|
46
|
Fan J, Stanfield J, Guo Y, Karam JA, Frenkel E, Sun X, Hsieh JT. Effect of Trans-2,3-Dimethoxycinnamoyl Azide on Enhancing Antitumor Activity of Romidepsin on Human Bladder Cancer. Clin Cancer Res 2008; 14:1200-7. [DOI: 10.1158/1078-0432.ccr-07-1656] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Ye Y, Yang H, Grossman HB, Dinney C, Wu X, Gu J. Genetic variants in cell cycle control pathway confer susceptibility to bladder cancer. Cancer 2008; 112:2467-74. [PMID: 18361427 DOI: 10.1002/cncr.23472] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yuanqing Ye
- Department of Epidemiology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
48
|
Fan J, Wilson PF, Wong HK, Urbin SS, Thompson LH, Wilson DM. XRCC1 down-regulation in human cells leads to DNA-damaging agent hypersensitivity, elevated sister chromatid exchange, and reduced survival of BRCA2 mutant cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2007; 48:491-500. [PMID: 17603793 DOI: 10.1002/em.20312] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Previous studies using rodent cells indicate that a deficiency in XRCC1 results in reduced single-strand break repair, increased sensitivity to DNA-damaging agents, and elevated levels of sister chromatid exchange (SCE). Epidemiological studies have suggested an association of certain human XRCC1 polymorphisms with genetic instability and cancer susceptibility. However, investigations on the molecular functions of XRCC1 in human cells are limited. To determine the contributions of this nonenzymatic scaffold protein, we suppressed XRCC1 levels in several human cell lines using small interfering RNA (siRNA) technology. We report that XRCC1 down-regulation in HeLa cells leads to a concomitant decrease in the DNA ligase 3 protein level and an impaired nick ligation capacity. In addition, depletion of XRCC1 resulted in a significantly increased sensitivity to the alkylating agent methyl methanesulfonate and the thymidine base analog 5-hydroxymethyl-2'-deoxyuridine, a slightly increased sensitivity to ethyl methanesulfonate and 1,3-bis(2-chloroethyl)-1-nitrosourea, and no change in the response to camptothecin. We also discovered that a 70-80% reduction in XRCC1 protein leads to an elevated level of SCE in both HeLa cells and normal human fibroblasts, but does not affect chromosome aberrations in the diploid fibroblasts. Last, XRCC1 siRNA transfection led to an approximately 40% decrease in the survival of BRCA2-deficient cells, supporting a model whereby the accumulation of unrepaired SSBs leads to the accumulation of cytotoxic DNA double strand breaks following replication fork collapse in cells defective in homologous recombination.
Collapse
Affiliation(s)
- Jinshui Fan
- Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
49
|
Kaufmann WK, Nevis KR, Qu P, Ibrahim JG, Zhou T, Zhou Y, Simpson DA, Helms-Deaton J, Cordeiro-Stone M, Moore DT, Thomas NE, Hao H, Liu Z, Shields JM, Scott GA, Sharpless NE. Defective cell cycle checkpoint functions in melanoma are associated with altered patterns of gene expression. J Invest Dermatol 2007; 128:175-87. [PMID: 17597816 PMCID: PMC2753794 DOI: 10.1038/sj.jid.5700935] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Defects in DNA damage responses may underlie genetic instability and malignant progression in melanoma. Cultures of normal human melanocytes (NHMs) and melanoma lines were analyzed to determine whether global patterns of gene expression could predict the efficacy of DNA damage cell cycle checkpoints that arrest growth and suppress genetic instability. NHMs displayed effective G1 and G2 checkpoint responses to ionizing radiation-induced DNA damage. A majority of melanoma cell lines (11/16) displayed significant quantitative defects in one or both checkpoints. Melanomas with B-RAF mutations as a class displayed a significant defect in DNA damage G2 checkpoint function. In contrast the epithelial-like subtype of melanomas with wild-type N-RAS and B-RAF alleles displayed an effective G2 checkpoint but a significant defect in G1 checkpoint function. RNA expression profiling revealed that melanoma lines with defects in the DNA damage G1 checkpoint displayed reduced expression of p53 transcriptional targets, such as CDKN1A and DDB2, and enhanced expression of proliferation-associated genes, such as CDC7 and GEMININ. A Bayesian analysis tool was more accurate than significance analysis of microarrays for predicting checkpoint function using a leave-one-out method. The results suggest that defects in DNA damage checkpoints may be recognized in melanomas through analysis of gene expression.
Collapse
Affiliation(s)
- William K Kaufmann
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The decatenation checkpoint delays entry into mitosis until the chromosomes have been disentangled. Deficiency in or bypass of the decatenation checkpoint can cause chromosome breakage and nondisjunction during mitosis, which results in aneuploidy and chromosome rearrangements in the daughter cells. A deficiency in the decatenation checkpoint has been reported in lung and bladder cancer cell lines and may contribute to the accumulation of chromosome aberrations that commonly occur during tumour progression. A checkpoint deficiency has also been documented in cultured stem and progenitor cells, and cancer stem cells are likely to be derived from stem and progenitor cells that lack an effective decatenation checkpoint. An inefficient decatenation checkpoint is likely to be a source of the chromosome aberrations that are common features of most tumours, but an inefficient decatenation checkpoint in cancer stem cells could also provide a potential target for chemotherapy.
Collapse
Affiliation(s)
- M Damelin
- Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, 701 W. 168th St., New York, NY 10032, USA
| | - T H Bestor
- Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, 701 W. 168th St., New York, NY 10032, USA
- E-mail:
| |
Collapse
|