1
|
García-Vázquez N, González-Robles TJ, Lane E, Spasskaya D, Zhang Q, Kerzhnerman MA, Jeong Y, Collu M, Simoneschi D, Ruggles KV, Róna G, Kaisari S, Pagano M. Stabilization of GTSE1 by cyclin D1-CDK4/6-mediated phosphorylation promotes cell proliferation with implications for cancer prognosis. eLife 2025; 13:RP101075. [PMID: 40272409 PMCID: PMC12021411 DOI: 10.7554/elife.101075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
In healthy cells, cyclin D1 is expressed during the G1 phase of the cell cycle, where it activates CDK4 and CDK6. Its dysregulation is a well-established oncogenic driver in numerous human cancers. The cancer-related function of cyclin D1 has been primarily studied by focusing on the phosphorylation of the retinoblastoma (RB) gene product. Here, using an integrative approach combining bioinformatic analyses and biochemical experiments, we show that GTSE1 (G-Two and S phases expressed protein 1), a protein positively regulating cell cycle progression, is a previously unrecognized substrate of cyclin D1-CDK4/6 in tumor cells overexpressing cyclin D1 during G1 and subsequent phases. The phosphorylation of GTSE1 mediated by cyclin D1-CDK4/6 inhibits GTSE1 degradation, leading to high levels of GTSE1 across all cell cycle phases. Functionally, the phosphorylation of GTSE1 promotes cellular proliferation and is associated with poor prognosis within a pan-cancer cohort. Our findings provide insights into cyclin D1's role in cell cycle control and oncogenesis beyond RB phosphorylation.
Collapse
Affiliation(s)
- Nelson García-Vázquez
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Tania J González-Robles
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Medicine, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Ethan Lane
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Daria Spasskaya
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Qingyue Zhang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Marc A Kerzhnerman
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - YeonTae Jeong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Marta Collu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Kelly V Ruggles
- Department of Medicine, New York University Grossman School of MedicineNew YorkUnited States
| | - Gergely Róna
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural SciencesBudapestHungary
| | - Sharon Kaisari
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
2
|
García-Vázquez N, González-Robles TJ, Lane E, Spasskaya D, Zhang Q, Kerzhnerman M, Jeong Y, Collu M, Simoneschi D, Ruggles KV, Rona G, Kaisari S, Pagano M. Stabilization of GTSE1 by cyclin D1-CDK4/6-mediated phosphorylation promotes cell proliferation: relevance in cancer prognosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.26.600797. [PMID: 38979260 PMCID: PMC11230433 DOI: 10.1101/2024.06.26.600797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
In healthy cells, cyclin D1 is expressed during the G1 phase of the cell cycle, where it activates CDK4 and CDK6. Its dysregulation is a well-established oncogenic driver in numerous human cancers. The cancer-related function of cyclin D1 has been primarily studied by focusing on the phosphorylation of the retinoblastoma (RB) gene product. Here, using an integrative approach combining bioinformatic analyses and biochemical experiments, we show that GTSE1 (G-Two and S phases expressed protein 1), a protein positively regulating cell cycle progression, is a previously unrecognized substrate of cyclin D1-CDK4/6 in tumor cells overexpressing cyclin D1 during G1 and subsequent phases. The phosphorylation of GTSE1 mediated by cyclin D1-CDK4/6 inhibits GTSE1 degradation, leading to high levels of GTSE1 across all cell cycle phases. Functionally, the phosphorylation of GTSE1 promotes cellular proliferation and is associated with poor prognosis within a pan-cancer cohort. Our findings provide insights into cyclin D1's role in cell cycle control and oncogenesis beyond RB phosphorylation.
Collapse
Affiliation(s)
- Nelson García-Vázquez
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Tania J González-Robles
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Department of Medicine, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
| | - Ethan Lane
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Daria Spasskaya
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Qingyue Zhang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Marc Kerzhnerman
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - YeonTae Jeong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Marta Collu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Kelly V Ruggles
- Department of Medicine, New York University Grossman School of Medicine, NYC, NY, USA
| | - Gergely Rona
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Sharon Kaisari
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
| |
Collapse
|
3
|
Yan G, Li J, Gao X, Liu J, Feng G, Li Y, Zhou H. Comprehensive analysis of the diagnostic and therapeutic value, immune infiltration, and drug treatment mechanisms of GTSE1 in lung adenocarcinoma. Front Med (Lausanne) 2024; 11:1433601. [PMID: 39629227 PMCID: PMC11611587 DOI: 10.3389/fmed.2024.1433601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
Objective The aim of this investigation was to assess the diagnostic and therapeutic efficacy of G2 and S-phase expressed 1 (GTSE1) in lung adenocarcinoma (LUAD), while examining its impact on immune infiltration and drug treatment mechanisms. Methods This research involved examining the expression patterns and diagnostic accuracy of GTSE1 in LUAD using various databases and clinical samples. The databases utilized included Gene Expression Omnibus (GEO), Clinical Proteomic Tumor Analysis Consortium (CPTAC), and The Cancer Genome Atlas (TCGA). Both gene expression and protein levels were analyzed. Subsequently, the prognostic ability of GTSE1 was evaluated based on clinical follow-up data using methods such as using univariate, multivariate, and prognostic meta-analysis. Additionally, potential mechanisms of action of GTSE1 were explored through enrichment analysis. Furthermore, the correlation between GTSE1 expression and the tumor microenvironment, immune cell infiltration, and immune checkpoints was assessed using ESTIMATE and CIBERSORT algorithms. The effectiveness of chemotherapy and targeted therapy was predicted using the "pRophetic" R package, which analyzed gene expression data. Results Analysis of GEO data, CPTAC data, TCGA data, and clinical samples revealed increased levels of GTSE1 in LUAD tissues. Enhanced GTSE1 expression demonstrated excellent diagnostic accuracy and served as a significant prognostic indicator for LUAD patients. GTSE1 expression emerged as an independent predictive factor in both univariate and multivariate Cox regression analyses. Furthermore, functional enrichment analysis suggested a potential association between GTSE1 and the cell cycle, p53 signaling pathway, as well as ubiquitin-mediated protein degradation. High expression of GTSE1 was associated with increased immune cell infiltration and heightened sensitivity to a specific type of chemotherapy and targeted drugs. Conclusion Increased expression of GTSE1 in patients with LUAD showed significant diagnostic and prognostic significance. It was also associated with increased immune infiltration and an unfavorable response to targeted medication.
Collapse
Affiliation(s)
- Guanqiang Yan
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingxiao Li
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiang Gao
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun Liu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guiyu Feng
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yue Li
- Department of Guangxi Medical University, Nanning, China
| | - Huafu Zhou
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
4
|
Luppov D, Sorokin M, Zolotovskaya M, Sekacheva M, Suntsova M, Zakharova G, Buzdin A. Gene Expression and Pathway Activation Biomarkers of Breast Cancer Sensitivity to Taxanes. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1803-1822. [PMID: 39523117 DOI: 10.1134/s0006297924100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024]
Abstract
Taxanes are one of the most widely used classes of breast cancer (BC) therapeutics. Despite the long history of clinical usage, the molecular mechanisms of their action and cancer resistance are still not fully understood. Here we aimed to identify gene expression and molecular pathway activation biomarkers of BC sensitivity to taxane drugs paclitaxel and docetaxel. We used to our knowledge the biggest collection of clinically annotated publicly available literature BC gene expression data (12 datasets, n = 1250) and the experimental clinical BC cohort (n = 12). Seven literature datasets were used for biomarker discovery (n = 914), and the remaining five literature plus one experimental datasets (n = 336) - for the validation. We totally found 34 genes and 29 molecular pathways which could strongly discriminate good and poor responders to taxane treatments. The biomarker genes and pathways were associated with molecular processes related to formation of mitotic spindle and centromeres, and with the spindle assembly mitotic checkpoint. Furthermore, we created gene expression and pathway activation signatures predicting BC response to taxanes. These signatures were tested on the validation BC cohort and demonstrated strong biomarker potential reflected by mean AUC values of 0.76 and 0.77, respectively, which outperforms previously reported analogs. Taken together, these findings can deepen our understanding of mechanism of action of taxanes and potentially improve personalization of treatment in BC.
Collapse
Affiliation(s)
- Daniil Luppov
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
- Laboratory of Clinical and Genomic Bioinformatics, I. M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
| | - Maxim Sorokin
- Laboratory of Clinical and Genomic Bioinformatics, I. M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
- OmicsWay Corp., Walnut, CA, 91789, USA
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Marianna Zolotovskaya
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- Laboratory of Clinical and Genomic Bioinformatics, I. M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
| | - Marina Sekacheva
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Maria Suntsova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Galina Zakharova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Anton Buzdin
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
- Oncobox Ltd., Moscow, 141701, Russia
| |
Collapse
|
5
|
WANG C, XU J, LIU M, LIU J, HUANG Y, ZHOU L. [Relationship between GTSE1 and Cell Cycle and Potential Regulatory Mechanisms
in Lung Cancer Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:451-458. [PMID: 39026496 PMCID: PMC11258651 DOI: 10.3779/j.issn.1009-3419.2024.106.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Indexed: 07/20/2024]
Abstract
The regulation of the cell cycle is essential for maintaining normal cellular function, especially in the development of diseases such as lung cancer. The cell cycle consists of four major phases (G1, S, G2 and M phases), which are characterized by a series of precise molecular events to ensure proper cell proliferation and division. In lung cancer cells, cell cycle dysregulation can lead to disordered proliferation and increased invasiveness of cancer cells. G2 and S-phase expressed 1 (GTSE1) is a regulatory protein found in the cytoplasm of the cell, which plays a key role in the cell cycle distribution of a wide range of cancer cells and is involved in life processes such as cell proliferation and apoptosis. GTSE1 affects cell cycle progression by interacting with cyclin-dependent kinase inhibitor 1A (p21) and maintaining the stability of p21, which in turn inhibits the activity of cyclin-dependent kinase 1/2 (CDK1/2). In addition, GTSE1 is also involved in the regulation of tumor protein 53 (p53) signaling pathway. With the assistance of mouse double minute 2 homolog (MDM2), GTSE1 is able to transport p53 from the nucleus to the cytoplasm and promote its ubiquitination and degradation, thus affecting cell cycle and cell death-related signaling pathways. This paper reviews the expression of GTSE1 in lung cancer cells and its effects on lung cancer, as well as its potential mechanisms involved in cell cycle regulation.
.
Collapse
|
6
|
Ortiz NR, Guy N, Garcia YA, Sivils JC, Galigniana MD, Cox MB. Functions of the Hsp90-Binding FKBP Immunophilins. Subcell Biochem 2023; 101:41-80. [PMID: 36520303 DOI: 10.1007/978-3-031-14740-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The Hsp90 chaperone is known to interact with a diverse array of client proteins. However, in every case examined, Hsp90 is also accompanied by a single or several co-chaperone proteins. One class of co-chaperone contains a tetratricopeptide repeat (TPR) domain that targets the co-chaperone to the C-terminal region of Hsp90. Within this class are Hsp90-binding peptidylprolyl isomerases, most of which belong to the FK506-binding protein (FKBP) family. Despite the common association of FKBP co-chaperones with Hsp90, it is abundantly clear that the client protein influences, and is often influenced by, the particular FKBP bound to Hsp90. Examples include Xap2 in aryl hydrocarbon receptor complexes and FKBP52 in steroid receptor complexes. In this chapter, we discuss the known functional roles played by FKBP co-chaperones and, where possible, relate distinctive functions to structural differences between FKBP members.
Collapse
Affiliation(s)
- Nina R Ortiz
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Naihsuan Guy
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Yenni A Garcia
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Jeffrey C Sivils
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Mario D Galigniana
- Departamento de Química Biológica/IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires, Argentina
| | - Marc B Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, USA.
| |
Collapse
|
7
|
Wu F, Mao Y, Ma T, Wang X, Wei H, Wang T, Wang J, Zhang Y. CTPS1 inhibition suppresses proliferation and migration in colorectal cancer cells. Cell Cycle 2022; 21:2563-2574. [PMID: 35912542 PMCID: PMC9704378 DOI: 10.1080/15384101.2022.2105084] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Colorectal cancer (CRC) is now the third most prevalent tumor and one of the deadliest cancers worldwide, with an increasing prevalence every year. Therefore, we urgently need to understand the mechanisms regulating the progression of colorectal cancer and find potential diagnostic biomarkers. In this study, we performed an analysis using the TCGA and GEO databases to find a molecular biomarker for the diagnosis of CRC, namely CTPS1. The results of this analysis revealed that CTPS1 could promote tumor proliferation and metastasis. Furthermore, bioinformatics analysis revealed that CTPS1 promoted CRC progression through cell cycle and p53 pathways. Further investigation demonstrated that CTPS1 might be involved in the regulation of CCNB1, RRM2, GTSE1, CDK2 and CHEK2 genes. Moreover, PCR confirmed that CTPS1 regulated GTSE1 and CDK2 molecules. Then, western blot was used to verify that CTPS1 promoted the expression of GTSE1 and CDK2 by inhibiting the expression of p53. In summary, we identified an important diagnostic biomarker for CRC, namely CTPS1, and its importance was validated at the cellular level. These results suggest that CTPS1 could serve as a candidate biomarker for CRC and CTPS1 inhibitors may be a potential treatment for CRC.
Collapse
Affiliation(s)
- Fahong Wu
- Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Yudong Mao
- Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Tao Ma
- Department of Hematology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoli Wang
- Department of Obstetrics and Gynecology, Xiamen Third Hospital, Xiamen, China
| | - Hangzhi Wei
- Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Tianwei Wang
- Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Jia Wang
- Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Youcheng Zhang
- Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China,CONTACT Youcheng Zhang Department of General Surgery, Hepatic-biliary-pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, 730030, China
| |
Collapse
|
8
|
Bao Z, Cheng J, Zhu J, Ji S, Gu K, Zhao Y, Yu S, Meng Y. Using Weighted Gene Co-Expression Network Analysis to Identify Increased MND1 Expression as a Predictor of Poor Breast Cancer Survival. Int J Gen Med 2022; 15:4959-4974. [PMID: 35601002 PMCID: PMC9117423 DOI: 10.2147/ijgm.s354826] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 05/07/2022] [Indexed: 12/12/2022] Open
Abstract
Objective We used bioinformatics analysis to identify potential biomarker genes and their relationship with breast cancer (BC). Materials and Methods We used a weighted gene co-expression network analysis (WGCNA) to create a co-expression network based on the top 25% genes in the GSE24124, GSE33926, and GSE86166 datasets obtained from the Gene Expression Omnibus. We used the DAVID online platform to perform GO and KEGG pathway enrichment analyses and the Cytoscape CytoHubba plug-in to screen the potential genes. Then, we related the genes to prognostic values in BC using the Oncomine, GEPIA, and Kaplan–Meier Plotter databases. Findings were validated by immunohistochemical (IHC) staining in the Human Protein Atlas and the TCGA-BRCA cohort. LinkedOmics identified the interactive expressions of hub genes. We used UALCAN to evaluate the methylation levels of these hub genes. MethSurv and SurvivalMeth were used to assess the multilevel prognostic value. Finally, we assessed hub gene association with immune cell infiltration using TIMER. Results The mRNA levels of MKI67, UBE2C, GTSE1, CCNA2, and MND1 were significantly upregulated in BC, whereas ESR1, THSD4, TFF1, AGR2, and FOXA1 were significantly downregulated. The DNA methylation signature analysis showed a better prognosis in the low-risk group. Further subgroup analyses revealed that MND1 might serve as an independent risk factor for unfavorable BC prognosis. Additionally, MND1 expression levels positively correlate with the immune infiltration statuses of CD4+ T cells, CD8+ T cells, B cells, neutrophils, dendritic cells, and macrophages. Conclusion Our results indicate that the ten hub genes may be involved in BC’s carcinogenesis, development, or metastasis, and MND1 may be a potential biomarker and therapeutic target for BC.
Collapse
Affiliation(s)
- Zhaokang Bao
- Department of Oncology Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Jiale Cheng
- Department of Oncology Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Jiahao Zhu
- Department of Radiotherapy and Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People’s Republic of China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, The affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Ke Gu
- Department of Radiotherapy and Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People’s Republic of China
| | - Yutian Zhao
- Department of Radiotherapy and Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People’s Republic of China
| | - Shiyou Yu
- Department of Oncology Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - You Meng
- Department of Oncology Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
- Correspondence: You Meng, Department of Oncology Surgery, The affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, 16 West Baita Road, Suzhou, Jiangsu, People’s Republic of China, Email
| |
Collapse
|
9
|
Xie C, Xiang W, Shen H, Shen J. GTSE1 is possibly involved in the DNA damage repair and cisplatin resistance in osteosarcoma. J Orthop Surg Res 2021; 16:713. [PMID: 34876170 PMCID: PMC8650252 DOI: 10.1186/s13018-021-02859-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background G2 and S phase-expressed-1 (GTSE1) negatively regulates the tumor-suppressive protein p53 and is potentially correlated with chemoresistance of cancer cells. This study aims to explore the effect of GTSE1 on the DNA damage repair and cisplatin (CDDP) resistance in osteosarcoma (OS). Materials and methods Expression of GTSE1 in OS was predicted in bioinformatics system GEPIA and then validated in clinically obtained tissues and acquired cell lines using RT-qPCR, immunohistochemical staining, and western blot assays. Gain- and loss-of-function studies of GTSE1 were performed in MG-63 and 143B cells to examine its function in cell cycle progression, DNA replication, and CDDP resistance. Stably transfected MG-63 cells were administrated into mice, followed by CDDP treatment to detect the role of GTSE1 in CDDP resistance in vivo. Results GTSE1 was highly expressed in patients with OS and correlated with poor survival according to the bioinformatics predictions. Elevated GTSE1 expression was detected in OS tissues and cell lines. GTSE1 silencing reduced S/G2 transition and DNA replication, and it increased the CDDP sensitivity and decreased the expression of DNA repair-related biomarkers in MG-63 cells. GTSE1 overexpression in 143B cells led to inverse trends. In vivo, downregulation of GTSE1 strengthened the treating effect of CDDP and significantly repressed growth of xenograft tumors in nude mice. However, overexpression of GTSE1 blocked the anti-tumor effect of CDDP. Conclusion This study demonstrates that GTSE1 is possibly involved in the DNA damage repair and cisplatin resistance in OS.
Collapse
Affiliation(s)
- Chaofan Xie
- Department of Orthopaedic, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, Guangdong, People's Republic of China.,Department of Orthopaedic, The Eighth Affiliated Hospital of Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Wei Xiang
- Department of Orthopaedic, The Eighth Affiliated Hospital of Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518033, Guangdong, People's Republic of China
| | - Huiyong Shen
- Department of Orthopaedic, The Eighth Affiliated Hospital of Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518033, Guangdong, People's Republic of China.
| | - Jingnan Shen
- Department of Muscularskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510000, Guangdong, People's Republic of China.
| |
Collapse
|
10
|
Guo W, Zhu J, Zhu Y, Wang K. G2 and S phase-expressed-1 acts as a putative tumor promoter in cervical cancer by enhancing Wnt/β-catenin signaling via modulation of GSK-3β. ENVIRONMENTAL TOXICOLOGY 2021; 36:1628-1639. [PMID: 33974332 DOI: 10.1002/tox.23158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 06/12/2023]
Abstract
G2 and S phase-expressed-1 (GTSE1) is currently identified as a key regulator of carcinogenesis. However, the involvement of GTSE1 in cervical cancer is unclear. The aims of this work were to explore the relationship between GTSE1 and cervical cancer. Our data elucidated high GTSE1 expression in cervical cancer tissue, which predicted a poor prognosis in cervical cancer patients. GTSE1 knockdown had tumor-suppressive effects in cervical cancer cells by inhibiting cell proliferative and invasive abilities. GTSE1 knockdown decreased the level of phosphorylated glycogen synthase kinase-3β (GSK-3β) and active β-catenin, resulted in inactivation of Wnt/β-catenin signaling. Suppression of GSK-3β remarkably abolished the GTSE1-knockdown-induced inhibitory effects on Wnt/β-catenin signaling. Suppression of Wnt/β-catenin signaling abolished the GTSE1-overexpression-induced oncogenic effects. Notably, GTSE1 knockdown impeded the in vivo tumorigenicity of cervical cancer cells. In short, this work demonstrates that GTSE1 is overexpressed in cervical cancer and GTSE1 suppression exerts a tumor-inhibiting role in cervical cancer by down-regulating Wnt/β-catenin signaling. Our work underlines a crucial relevance between GTSE1 and cervical cancer progression and suggests GTSE1 as a promising therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Wenting Guo
- Obstetrics and Gynecology Department, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Jing Zhu
- Obstetrics and Gynecology Department, Yulin NO.2 Hospital, Yulin, China
| | - Yuan Zhu
- Gynecology Department, Maternal and Child Health Care Hospital of Zibo, Zibo City, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, China
- Oncology Department, Daxing Hospital, Xi'an, China
| |
Collapse
|
11
|
GTSE1 Facilitates the Malignant Phenotype of Lung Cancer Cells via Activating AKT/mTOR Signaling. ACTA ACUST UNITED AC 2021; 2021:5589532. [PMID: 34007784 PMCID: PMC8110388 DOI: 10.1155/2021/5589532] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/25/2022]
Abstract
The expression of G2 and S phase-expressed-1 (GTSE1) was upregulated in human cancer. However, its expression and roles in lung cancer have not been identified yet. In our study, we reported that GTSE1 expression was statistically higher in lung tissues than in the adjacent noncancerous tissues which might be a consequence of hypomethylation of the GTSE1 promoter. The upregulated expression of GTSE1 mRNA predicted the poorer survival of the lung patients. Ectopic expression of GTSE1 in lung cancer cells significantly increased while knockdown of GTSE1 decreased cell proliferation, cell migration, and cell invasion in H460 and A549 cells. Furthermore, knockdown of GTSE1 regulated the cell cycle and promoted cell apoptosis in H460 and A549 cells. Finally, we presented that GTSE1 was able to activate AKT/mTOR signaling in H460 and A549 cells. In conclusion, these results indicated that the overexpressed GTSE1 was involved in the progress of lung cancer by promoting proliferation migration and invasion and inhibiting apoptosis of lung cancer cells via activating AKT/mTOR signaling.
Collapse
|
12
|
Lei X, Du L, Zhang P, Ma N, Liang Y, Han Y, Qu B. Knockdown GTSE1 enhances radiosensitivity in non-small-cell lung cancer through DNA damage repair pathway. J Cell Mol Med 2020; 24:5162-5167. [PMID: 32202046 PMCID: PMC7205821 DOI: 10.1111/jcmm.15165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an important strategy for NSCLC. However, although a variety of comprehensive radiotherapy-based treatments have dominated the treatment of NSCLC, it cannot be avoided to overcome the growing radioresistance during radiotherapy. The purpose of this study was to elucidate the radiosensitizing effects of NSCLC via knockdown GTSE1 expression and its mechanism. Experiments were performed by using multiple NSCLC cells such as A549, H460 and H1299. Firstly, we found GTSE1 conferred to radioresistance via clonogenic assay and apoptosis assay. Then, we detected the level of DNA damage through comet assay and γH2AX foci, which we could clearly observe knockdown GTSE1 enhance DNA damage after IR. Furthermore, through using laser assay and detecting DNA damage repair early protein expression, we found radiation could induce GTSE1 recruited to DSB site and initiate DNA damage response. Our finding demonstrated that knockdown GTSE1 enhances radiosensitivity in NSCLC through DNA damage repair pathway. This novel observation may have therapeutic implications to improve therapeutic efficacy of radiation.
Collapse
Affiliation(s)
- Xiao Lei
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lehui Du
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pei Zhang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Na Ma
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanjie Liang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanan Han
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baolin Qu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
GTSE1, CDC20, PCNA, and MCM6 Synergistically Affect Regulations in Cell Cycle and Indicate Poor Prognosis in Liver Cancer. Anal Cell Pathol (Amst) 2019; 2019:1038069. [PMID: 32082966 PMCID: PMC7012210 DOI: 10.1155/2019/1038069] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
GTSE1 is well correlated with tumor progression; however, little is known regarding its role in liver cancer prognosis. By analyzing the hepatocellular carcinoma (HCC) datasets in GEO and TCGA databases, we showed that high expression of GTSE1 was correlated with advanced pathologic stage and poor prognosis of HCC patients. To investigate underlying molecular mechanism, we generated GTSE1 knockdown HCC cell line and explored the effects of GTSE1 deficiency in cell growth. Between GTSE1 knockdown and wild-type HCC cells, we identified 979 differentially expressed genes (520 downregulated and 459 upregulated genes) in the analysis of microarray-based gene expression profiling. Functional enrichment analysis of DEGs suggested that S phase was dysregulated without GTSE1 expression, which was further verified from flow cytometry analysis. Moreover, three other DEGs: CDC20, PCNA, and MCM6, were also found contributing to GTSE1-related cell cycle arrest and to be associated with poor overall survival of HCC patients. In conclusion, GTSE1, together with CDC20, PCNA, and MCM6, may synergistically promote adverse prognosis in HCC by activating cell cycle. Genes like GTSE1, CDC20, PCNA, and MCM6 may be promising prognostic molecular biomarkers in liver cancer.
Collapse
|
14
|
Lin F, Xie YJ, Zhang XK, Huang TJ, Xu HF, Mei Y, Liang H, Hu H, Lin ST, Luo FF, Lang YH, Peng LX, Qian CN, Huang BJ. GTSE1 is involved in breast cancer progression in p53 mutation-dependent manner. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:152. [PMID: 30961661 PMCID: PMC6454633 DOI: 10.1186/s13046-019-1157-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/29/2019] [Indexed: 12/24/2022]
Abstract
Background With the rapid development of the high throughput detection techniques, tumor-related Omics data has become an important source for studying the mechanism of tumor progression including breast cancer, one of the major malignancies worldwide. A previous study has shown that the G2 and S phase-expressed-1 (GTSE1) can act as an oncogene in several human cancers. However, its functional roles in breast cancer remain elusive. Method In this study, we analyzed breast cancer data downloaded from The Cancer Genome Atlas (TCGA) databases and other online database including the Oncomine, bc-GenExMiner and PROGgeneV2 database to identify the molecules contributing to the progression of breast cancer. The GTSE1 expression levels were investigated using qRT-PCR, immunoblotting and IHC. The biological function of GTSE1 in the growth, migration and invasion of breast cancer was examined in MDA-MB-231, MDA-MB-468 and MCF7 cell lines. The in vitro cell proliferative, migratory and invasive abilities were evaluated by MTS, colony formation and transwell assay, respectively. The role of GTSE1 in the growth and metastasis of breast cancer were revealed by in vivo investigation using BALB/c nude mice. Results We showed that the expression level of GTSE1 was upregulated in breast cancer specimens and cell lines, especially in triple negative breast cancer (TNBC) and p53 mutated breast cancer cell lines. Importantly, high GTSE1 expression was positively correlated with histological grade and poor survival. We demonstrated that GTSE1 could promote breast cancer cell growth by activating the AKT pathway and enhance metastasis by regulating the Epithelial-Mesenchymal transition (EMT) pathway. Furthermore, it could cause multidrug resistance in breast cancer cells. Interestingly, we found that GTSE1 could regulate the p53 function to alter the cell cycle distribution dependent on the mutation state of p53. Conclusion Our results reveal that GTSE1 played a key role in the progression of breast cancer, indicating that GTSE1 could serve as a novel biomarker to aid in the assessment of the prognosis of breast cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1157-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fen Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yu-Jie Xie
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Xin-Ke Zhang
- Department of pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Tie-Jun Huang
- Department of Nuclear Medicine, The Second People's Hospital of Shenzhen, Shenzhen, People's Republic of China
| | - Hong-Fa Xu
- Zhuhai Precision Medicine Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Yan Mei
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hu Liang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hao Hu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Si-Ting Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Fei-Fei Luo
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yan-Hong Lang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
15
|
Dong Z, Coates D, Liu Q, Sun H, Li C. Quantitative proteomic analysis of deer antler stem cells as a model of mammalian organ regeneration. J Proteomics 2019; 195:98-113. [PMID: 30641233 DOI: 10.1016/j.jprot.2019.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/25/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022]
Abstract
The ability to activate and regulate stem cells during wound healing and tissue/organ regeneration is a promising field which could bring innovative approaches to regenerative medicine. The regenerative capacity of invertebrates has been well documented, however in mammals, stem cells that drive organ regeneration are rare. Deer antler is unique in providing a mammalian model of complete organ regeneration based on stem cells. The present study investigated the differentially regulated proteins (DRPs) between different antler stem cell populations (n = 3) using 2D-DIGE. Western blotting was used to validate the proteomics results. Comparative proteomics resulted in protein profiles which were similar for the biological replicates but different between the cells derived from two different stem cell niches involved in antler growth/regeneration and cells derived from facial periosteum. Ninety-two up- and down-regulated proteins were identified by MALDI-TOF MS. The work indicates that the epithelial-mesenchymal transition process may participate in the initiation of antler regeneration including the first stage of scar-less wound healing. Cell mobility is also highly regulated during antler regeneration. Energy and nucleotide metabolism may however be less active in antler regeneration as compared to that in antler generation phase. These results provide new insights into the underlying mechanisms of stem cell-based regeneration of mammalian organs.
Collapse
Affiliation(s)
- Zhen Dong
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun 130112, China; Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Dawn Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Qingxiu Liu
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Hongmei Sun
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Chunyi Li
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun 130112, China; Changchun Sci-Tech University, Changchun 130600, China.
| |
Collapse
|
16
|
Stabilization of P/CAF, as a ubiquitin ligase toward MDM2, suppresses mitotic cell death through p53-p21 activation in HCT116 cells with SIRT2 suppression. Biochem Biophys Res Commun 2019; 508:230-236. [PMID: 30482390 DOI: 10.1016/j.bbrc.2018.11.136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 11/20/2018] [Indexed: 01/15/2023]
Abstract
We previously reported that the suppression of SIRT2, an NAD + -dependent protein deacetylases, induces p53 accumulation via degradation of p300 and the subsequent MDM2 degradation, eventually leading to apoptosis in HeLa cells. The present study identified a novel pathway of p53 accumulation by SIRT2 suppression in HCT116(p53+/+) cells in which SIRT2 suppression led to escape from mitotic cell death caused by spindle assembly checkpoint activation induced by microtubule inhibitors such as nocodazole but not apoptosis or G1 or G2 arrest. We found that SIRT2 interacts with P/CAF, a histone acetyltransferase, which also acts as a ubiquitin ligase against MDM2. SIRT2 suppression led to an increase of P/CAF acetylation and its stabilization followed by a decrease in MDM2 and activation of the p53-p21 pathway. Depression of mitotic cell death in HCT116(p53+/+) cells with SIRT2 suppression was released by suppression of P/CAF or p21. Thus, the P/CAF-MDM2-p53-p21 axis enables the escape from mitotic cell death and confers resistance to nocodazole in HCT116(p53+/+) cells with SIRT2 suppression. As SIRT2 has attracted attention as a potential target for cancer therapeutics for p53 regulation, the present study provides a molecular basis for the efficacy of SIRT2 for future cancer therapy based on p53 regulation. These findings also suggest an undesirable function of the SIRT2 suppression associated with activation of the p53-p21 pathway in the suppression of mitotic cell death caused by spindle assembly checkpoint activation.
Collapse
|
17
|
Xu T, Ma M, Chi Z, Si L, Sheng X, Cui C, Dai J, Yu S, Yan J, Yu H, Wu X, Tang H, Yu J, Kong Y, Guo J. High G2 and S-phase expressed 1 expression promotes acral melanoma progression and correlates with poor clinical prognosis. Cancer Sci 2018; 109:1787-1798. [PMID: 29660787 PMCID: PMC5989838 DOI: 10.1111/cas.13607] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/23/2018] [Accepted: 04/08/2018] [Indexed: 02/07/2023] Open
Abstract
G2 and S‐phase expressed 1 (GTSE1) regulates cell cycle progression in human cancers. However, its significance and mechanism of action in acral melanoma (AM) remain unknown. In the present study, we found that GTSE1 expression was upregulated in advanced stage/metastatic AM tissues and metastatic cell lines, and correlated with higher stage (P = .028) and poor disease‐free survival (DFS) in patients with AM (P = .003). Cox regression assays validated GTSE1 expression to be an independent prognostic factor of DFS for patients with AM (P = .004). Ectopic expression of GTSE1 enhanced primary AM cell proliferation, invasion, and migration. Loss‐of‐function in GTSE1 attenuated metastatic AM cell proliferation and metastatic ability in vitro and in vivo. We additionally observed that inhibition of migration and invasion occurred concomitantly with a GTSE1 knockdown‐mediated increase in E‐cadherin and decreases in N‐cadherin and Slug. We further showed that integrin subunit alpha 2 (ITGA2) interacts with GTSE1 and is a downstream effector of GTSE1. Further, ITGA2 levels were positively correlated with GTSE1 expression in human AM tissues. Ectopic ITGA2 expression rescued siGTSE1‐mediated inhibition of migration and invasion, thereby restoring epithelial‐to‐mesenchymal transition (EMT). In conclusion, GTSE1 expression promotes AM progression and correlates with clinical outcomes of patients with AM, and may represent a promising therapeutic target to suppress AM progression.
Collapse
Affiliation(s)
- Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Junya Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaowen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiayi Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
18
|
Tipton AR, Wren JD, Daum JR, Siefert JC, Gorbsky GJ. GTSE1 regulates spindle microtubule dynamics to control Aurora B kinase and Kif4A chromokinesin on chromosome arms. J Cell Biol 2017; 216:3117-3132. [PMID: 28821562 PMCID: PMC5626529 DOI: 10.1083/jcb.201610012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 04/20/2017] [Accepted: 07/12/2017] [Indexed: 12/24/2022] Open
Abstract
In mitosis, the dynamic assembly and disassembly of microtubules are critical for normal chromosome movement and segregation. Microtubule turnover varies among different mitotic spindle microtubules, dictated by their spatial distribution within the spindle. How turnover among the various classes of spindle microtubules is differentially regulated and the resulting significance of differential turnover for chromosome movement remains a mystery. As a new tactic, we used global microarray meta-analysis (GAMMA), a bioinformatic method, to identify novel regulators of mitosis, and in this study, we describe G2- and S phase-expressed protein 1 (GTSE1). GTSE1 is expressed exclusively in late G2 and M phase. From nuclear envelope breakdown until anaphase onset, GTSE1 binds preferentially to the most stable mitotic spindle microtubules and promotes their turnover. Cells depleted of GTSE1 show defects in chromosome alignment at the metaphase plate and in spindle pole integrity. These defects are coupled with an increase in the proportion of stable mitotic spindle microtubules. A consequence of this reduced microtubule turnover is diminished recruitment and activity of Aurora B kinase on chromosome arms. This decrease in Aurora B results in diminished binding of the chromokinesin Kif4A to chromosome arms.
Collapse
Affiliation(s)
- Aaron R Tipton
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - John R Daum
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Joseph C Siefert
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Gary J Gorbsky
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
19
|
GTSE1 promotes cell migration and invasion by regulating EMT in hepatocellular carcinoma and is associated with poor prognosis. Sci Rep 2017; 7:5129. [PMID: 28698581 PMCID: PMC5505986 DOI: 10.1038/s41598-017-05311-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
G2 and S phase-expressed-1 (GTSE1) regulates G1/S cell cycle transition. It was recently reported to be overexpressed in certain human cancers, but its significance and mechanism(s) in hepatocellular carcinoma (HCC) remain unknown. Here, we showed preferential GTSE1 upregulation in human HCC tissues and cell lines that positively correlated with Ki67. GTSE1 knockdown by short hairpin RNA resulted in deficient colony-forming ability and depleted capabilities of HCC cells to migrate and invade. Conversely, exogenous GTSE1 overexpression enhanced colony formation and stimulated HCC cell migration and invasion. Furthermore, GTSE1 silencing was associated with the downregulation of N-cadherin, β-catenin, and Snail, whereas GTSE1 overexpression caused the opposite effects. GTSE1 upregulated Snail via both transcription and protein degradation pathways. Additionally, GTSE1 modulated the sensitivity of HCC to 5-fluorouracil therapy. High GTSE1 correlates with chemo-resistance, while low GTSE1 increases drug sensitivity. Kaplan-Meier survival analysis indicated that high GTSE1 levels were significantly associated with poor overall survival. In conclusion, high expression of GTSE1 is commonly noted in HCC and is closely correlated with migration and invasion by epithelial-to-mesenchymal transition (EMT) modulation. Activated GTSE1 significantly interferes with chemotherapy efficacy and influences the probability of survival of patients with HCC. GTSE1 may thus represent a promising molecular target.
Collapse
|
20
|
Stelitano D, Peche LY, Dalla E, Monte M, Piazza S, Schneider C. GTSE1: a novel TEAD4-E2F1 target gene involved in cell protrusions formation in triple-negative breast cancer cell models. Oncotarget 2017; 8:67422-67438. [PMID: 28978043 PMCID: PMC5620183 DOI: 10.18632/oncotarget.18691] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 05/22/2017] [Indexed: 12/14/2022] Open
Abstract
GTSE1 over-expression has been reported as a potential marker for metastasis in various types of malignancies, including breast cancer. Despite this, the transcriptional regulation of this protein and the causes of its misregulation in tumors remain largely unknown. The aims of this work were to elucidate how GTSE1 is regulated at the transcriptional level and to clarify the mechanism underlying GTSE1-dependent cell functions in triple-negative breast cancer (TNBC). Here, we identified GTSE1 as a novel target gene of the TEAD4 transcription factor, highlighting a role for the YAP and TAZ coactivators in the transcriptional regulation of GTSE1. Moreover, we found that TEAD4 controls the formation of cell protrusions required for cell migration through GTSE1, unveiling a relevant effector role for this protein in the TEAD-dependent cellular functions and confirming TEAD4 role in promoting invasion and metastasis in breast cancer. Finally, we highlighted a role for the pRb-E2F1 pathway in the control of GTSE1 transcription and observed that treatment with drugs targeting the pRb-E2F1 or YAP/TAZ-TEAD pathways dramatically downregulated the expression levels of GTSE1 and of other genes involved in the formation of metastasis, suggesting their potential use in the treatment of TNBC.
Collapse
Affiliation(s)
- Debora Stelitano
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie (L.N.CIB), Trieste, Italy
| | - Leticia Yamila Peche
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie (L.N.CIB), Trieste, Italy
| | - Emiliano Dalla
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie (L.N.CIB), Trieste, Italy
| | - Martin Monte
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvano Piazza
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie (L.N.CIB), Trieste, Italy.,Bioinformatics Core facility, Centre for Integrative Biology, University of Trento (CIBIO), Trento, Italy
| | - Claudio Schneider
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie (L.N.CIB), Trieste, Italy.,Dipartimento di Scienze Biomediche e Biologiche (DSMB), Università degli Studi di Udine, Udine, Italy
| |
Collapse
|
21
|
Subhash VV, Tan SH, Tan WL, Yeo MS, Xie C, Wong FY, Kiat ZY, Lim R, Yong WP. GTSE1 expression represses apoptotic signaling and confers cisplatin resistance in gastric cancer cells. BMC Cancer 2015. [PMID: 26209226 PMCID: PMC4514980 DOI: 10.1186/s12885-015-1550-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background Platinum based therapy is commonly used in the treatment of advanced gastric cancer. However, resistance to chemotherapy is a major challenge that causes marked variation in individual response rate and survival rate. In this study, we aimed to identify the expression of GTSE1 and its correlation with cisplatin resistance in gastric cancer cells. Methods Methylation profiling was carried out in tissue samples from gastric cancer patients before undergoing neoadjuvent therapy using docetaxel, cisplatin and 5FU (DCX) and in gastric cancer cell lines. The correlation between GTSE1 expression and methylation in gastric cancer cells was determined by RT-PCR and MSP respectively. GTSE1 expression was knocked-down using shRNA’s and its effects on cisplatin cytotoxicity and cell survival were detected by MTS, proliferation and clonogenic survival assays. Additionally, the effect of GTSE1 knock down in drug induced apoptosis was determined by western blotting and apoptosis assays. Results GTSE1 exhibited a differential methylation index in gastric cancer patients and in cell lines that correlated with DCX treatment response and cisplatin sensitivity, respectively. In-vitro, GTSE1 expression showed a direct correlation with hypomethylation. Interestingly, Cisplatin treatment induced a dose dependent up regulation as well as nuclear translocation of GTSE1 expression in gastric cancer cells. Knock down of GTSE1 enhanced cisplatin cytotoxity and led to a significant reduction in cell proliferation and clonogenic survival. Also, loss of GTSE1 expression caused a significant increase in P53 mediated apoptosis in cisplatin treated cells. Conclusion Our study identifies GTSE1 as a biomarker for cisplatin resistance in gastric cancer cells. This study also suggests the repressive role of GTSE1 in cisplatin induced apoptosis and signifies its potential utility as a therapeutic target for better clinical management of gastric cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1550-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vinod Vijay Subhash
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| | - Shi Hui Tan
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Woei Loon Tan
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Mei Shi Yeo
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Chen Xie
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Foong Ying Wong
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Zee Ying Kiat
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Robert Lim
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| |
Collapse
|
22
|
Mazaira GI, Camisay MF, De Leo S, Erlejman AG, Galigniana MD. Biological relevance of Hsp90-binding immunophilins in cancer development and treatment. Int J Cancer 2015; 138:797-808. [PMID: 25754838 DOI: 10.1002/ijc.29509] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/17/2015] [Indexed: 12/14/2022]
Abstract
Immunophilins are a family of intracellular receptors for immunosuppressive drugs. Those immunophilins that are related to immunosuppression are the smallest proteins of the family, i.e., FKBP12 and CyPA, whereas the other members of the family have higher molecular weight because the show additional domains to the drug-binding site. Among these extra domains, the TPR-domain is perhaps the most relevant because it permits the interaction of high molecular weight immunophilins with the 90-kDa heat-shock protein, Hsp90. This essential molecular chaperone regulates the biological function of several protein-kinases, oncogenes, protein phosphatases, transcription factors and cofactors . Hsp90-binding immunophilins where first characterized due to their association with steroid receptors. They regulate the cytoplasmic transport and the subcellular localization of these and other Hsp90 client proteins, as well as transcriptional activity, cell proliferation, cell differentiation and apoptosis. Hsp90-binding immunophilins are frequently overexpressed in several types of cancers and play a key role in cell survival. In this article we analyze the most important biological actions of the best characterized Hsp90-binding immunophilins in both steroid receptor function and cancer development and discuss the potential use of these immunophilins for therapeutic purposes as potential targets of specific small molecules.
Collapse
Affiliation(s)
- Gisela I Mazaira
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - María F Camisay
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Sonia De Leo
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Alejandra G Erlejman
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Mario D Galigniana
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina.,Instituto De Biología Y Medicina Experimental-CONICET, Buenos Aires, Argentina
| |
Collapse
|
23
|
Romano S, D'Angelillo A, Romano MF. Pleiotropic roles in cancer biology for multifaceted proteins FKBPs. Biochim Biophys Acta Gen Subj 2015; 1850:2061-8. [PMID: 25592270 DOI: 10.1016/j.bbagen.2015.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND FK506 binding proteins (FKBP) are multifunctional proteins highly conserved across the species and abundantly expressed in the cell. In addition to a well-established role in immunosuppression, FKBPs modulate several signal transduction pathways in the cell, due to their isomerase activity and the capability to interact with other proteins, inducing changes in conformation and function of protein partners. Increasing literature data support the concept that FKBPs control cancer related pathways. SCOPE OF THE REVIEW The aim of the present article is to review current knowledge on FKBPs roles in regulation of key signaling pathways associated with cancer. MAJOR CONCLUSIONS Some family members appear to promote disease while others are protective against tumorigenesis. GENERAL SIGNIFICANCE FKBPs family proteins are expected to provide new biomarkers and small molecular targets, in the near future, increasing diagnostic and therapeutic opportunities in the cancer field. This article is part of a Special Issue entitled Proline-Directed Foldases: Cell Signaling Catalysts and Drug Targets.
Collapse
Affiliation(s)
- Simona Romano
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Anna D'Angelillo
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy; Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy.
| |
Collapse
|
24
|
Guy NC, Garcia YA, Sivils JC, Galigniana MD, Cox MB. Functions of the Hsp90-binding FKBP immunophilins. Subcell Biochem 2015; 78:35-68. [PMID: 25487015 DOI: 10.1007/978-3-319-11731-7_2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hsp90 functionally interacts with a broad array of client proteins, but in every case examined Hsp90 is accompanied by one or more co-chaperones. One class of co-chaperone contains a tetratricopeptide repeat domain that targets the co-chaperone to the C-terminal region of Hsp90. Within this class are Hsp90-binding peptidylprolyl isomerases, most of which belong to the FK506-binding protein (FKBP) family. Despite the common association of FKBP co-chaperones with Hsp90, it is now clear that the client protein influences, and is influenced by, the particular FKBP bound to Hsp90. Examples include Xap2 in aryl hydrocarbon receptor complexes and FKBP52 in steroid receptor complexes. In this chapter, we discuss the known functional roles played by FKBP co-chaperones and, where possible, relate distinctive functions to structural differences between FKBP members.
Collapse
Affiliation(s)
- Naihsuan C Guy
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 79968, El Paso, TX, USA,
| | | | | | | | | |
Collapse
|
25
|
Mazaira GI, Lagadari M, Erlejman AG, Galigniana MD. The Emerging Role of TPR-Domain Immunophilins in the Mechanism of Action of Steroid Receptors. NUCLEAR RECEPTOR RESEARCH 2014. [DOI: 10.11131/2014/101094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- G. I. Mazaira
- Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M. Lagadari
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - A. G. Erlejman
- Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M. D. Galigniana
- Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| |
Collapse
|
26
|
Neri S, Bourin P, Peyrafitte JA, Cattini L, Facchini A, Mariani E. Human adipose stromal cells (ASC) for the regeneration of injured cartilage display genetic stability after in vitro culture expansion. PLoS One 2013; 8:e77895. [PMID: 24205017 PMCID: PMC3810264 DOI: 10.1371/journal.pone.0077895] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/05/2013] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells are emerging as an extremely promising therapeutic agent for tissue regeneration due to their multi-potency, immune-modulation and secretome activities, but safety remains one of the main concerns, particularly when in vitro manipulation, such as cell expansion, is performed before clinical application. Indeed, it is well documented that in vitro expansion reduces replicative potential and some multi-potency and promotes cell senescence. Furthermore, during in vitro aging there is a decrease in DNA synthesis and repair efficiency thus leading to DNA damage accumulation and possibly inducing genomic instability. The European Research Project ADIPOA aims at validating an innovative cell-based therapy where autologous adipose stromal cells (ASCs) are injected in the diseased articulation to activate regeneration of the cartilage. The primary objective of this paper was to assess the safety of cultured ASCs. The maintenance of genetic integrity was evaluated during in vitro culture by karyotype and microsatellite instability analysis. In addition, RT-PCR array-based evaluation of the expression of genes related to DNA damage signaling pathways was performed. Finally, the senescence and replicative potential of cultured cells was evaluated by telomere length and telomerase activity assessment, whereas anchorage-independent clone development was tested in vitro by soft agar growth. We found that cultured ASCs do not show genetic alterations and replicative senescence during the period of observation, nor anchorage-independent growth, supporting an argument for the safety of ASCs for clinical use.
Collapse
Affiliation(s)
- Simona Neri
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopedic Institute, Bologna, Italy
- * E-mail:
| | - Philippe Bourin
- Etablissement Français du Sang Pyrénées Méditerranée (EFS-PM), Toulouse, France
- CSA21, Toulouse, France
| | - Julie-Anne Peyrafitte
- Etablissement Français du Sang Pyrénées Méditerranée (EFS-PM), Toulouse, France
- STROMALAB, UMR 5273 Centre national de la Recherche Scientifique (CNRS)/Université Paul Sabatier, U1031 Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Luca Cattini
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Andrea Facchini
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopedic Institute, Bologna, Italy
- Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy
| | - Erminia Mariani
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopedic Institute, Bologna, Italy
- Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy
| |
Collapse
|
27
|
Donley C, McClelland K, McKeen HD, Nelson L, Yakkundi A, Jithesh PV, Burrows J, McClements L, Valentine A, Prise KM, McCarthy HO, Robson T. Identification of RBCK1 as a novel regulator of FKBPL: implications for tumor growth and response to tamoxifen. Oncogene 2013; 33:3441-50. [PMID: 23912458 DOI: 10.1038/onc.2013.306] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 05/27/2013] [Accepted: 06/11/2013] [Indexed: 01/03/2023]
Abstract
FKBPL has been implicated in processes associated with cancer, including regulation of tumor growth and angiogenesis with high levels of FKBPL prognosticating for improved patient survival. Understanding how FKBPL levels are controlled within the cell is therefore critical. We have identified a novel role for RBCK1 as an FKBPL-interacting protein, which regulates FKBPL stability at the post-translational level via ubiquitination. Both RBCK1 and FKBPL are upregulated by 17-β-estradiol and interact within heat shock protein 90 chaperone complexes, together with estrogen receptor-α (ERα). Furthermore, FKBPL and RBCK1 associate with ERα at the promoter of the estrogen responsive gene, pS2, and regulate pS2 levels. MCF-7 clones stably overexpressing RBCK1 were shown to have reduced proliferation and increased levels of FKBPL and p21. Furthermore, these clones were resistant to tamoxifen therapy, suggesting that RBCK1 could be a predictive marker of response to endocrine therapy. RBCK1 knockdown using targeted small interfering RNA resulted in increased proliferation and increased sensitivity to tamoxifen treatment. Moreover, in support of our in vitro data, analysis of mRNA microarray data sets demonstrated that high levels of FKBPL and RBCK1 correlated with increased patient survival, whereas high RBCK1 predicted for a poor response to tamoxifen. Our findings support a role for RBCK1 in the regulation of FKBPL with important implications for estrogen receptor signaling, cell proliferation and response to endocrine therapy.
Collapse
Affiliation(s)
- C Donley
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - K McClelland
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - H D McKeen
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - L Nelson
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - A Yakkundi
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - P V Jithesh
- Liverpool Cancer Research UK Centre, University of Liverpool, Liverpool, UK
| | - J Burrows
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - L McClements
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - A Valentine
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - K M Prise
- Centre for Cancer Research and Cell Biology, Queen's University, Belfast, Northern Ireland
| | - H O McCarthy
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| | - T Robson
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Northern Ireland
| |
Collapse
|
28
|
Single-cell gene expression analysis reveals genetic associations masked in whole-tissue experiments. Nat Biotechnol 2013; 31:748-52. [PMID: 23873083 DOI: 10.1038/nbt.2642] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/14/2013] [Indexed: 12/17/2022]
Abstract
Gene expression in multiple individual cells from a tissue or culture sample varies according to cell-cycle, genetic, epigenetic and stochastic differences between the cells. However, single-cell differences have been largely neglected in the analysis of the functional consequences of genetic variation. Here we measure the expression of 92 genes affected by Wnt signaling in 1,440 single cells from 15 individuals to associate single-nucleotide polymorphisms (SNPs) with gene-expression phenotypes, while accounting for stochastic and cell-cycle differences between cells. We provide evidence that many heritable variations in gene function--such as burst size, burst frequency, cell cycle-specific expression and expression correlation/noise between cells--are masked when expression is averaged over many cells. Our results demonstrate how single-cell analyses provide insights into the mechanistic and network effects of genetic variability, with improved statistical power to model these effects on gene expression.
Collapse
|
29
|
Scolz M, Widlund PO, Piazza S, Bublik DR, Reber S, Peche LY, Ciani Y, Hubner N, Isokane M, Monte M, Ellenberg J, Hyman AA, Schneider C, Bird AW. GTSE1 is a microtubule plus-end tracking protein that regulates EB1-dependent cell migration. PLoS One 2012; 7:e51259. [PMID: 23236459 PMCID: PMC3517537 DOI: 10.1371/journal.pone.0051259] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/30/2012] [Indexed: 02/08/2023] Open
Abstract
The regulation of cell migration is a highly complex process that is often compromised when cancer cells become metastatic. The microtubule cytoskeleton is necessary for cell migration, but how microtubules and microtubule-associated proteins regulate multiple pathways promoting cell migration remains unclear. Microtubule plus-end binding proteins (+TIPs) are emerging as important players in many cellular functions, including cell migration. Here we identify a +TIP, GTSE1, that promotes cell migration. GTSE1 accumulates at growing microtubule plus ends through interaction with the EB1+TIP. The EB1-dependent +TIP activity of GTSE1 is required for cell migration, as well as for microtubule-dependent disassembly of focal adhesions. GTSE1 protein levels determine the migratory capacity of both nontransformed and breast cancer cell lines. In breast cancers, increased GTSE1 expression correlates with invasive potential, tumor stage, and time to distant metastasis, suggesting that misregulation of GTSE1 expression could be associated with increased invasive potential.
Collapse
Affiliation(s)
- Massimilano Scolz
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
| | - Per O. Widlund
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Silvano Piazza
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
| | - Debora Rosa Bublik
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
| | - Simone Reber
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Leticia Y. Peche
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
| | - Yari Ciani
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
| | - Nina Hubner
- Department of Molecular Cancer Research, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Mayumi Isokane
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Heidelberg, Germany
| | - Martin Monte
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
| | - Jan Ellenberg
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Heidelberg, Germany
| | - Anthony A. Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (AWB); (AAH); (CS)
| | - Claudio Schneider
- Laboratorio Nazionale The Interuniversity Consortium for Biotechnology, Area Science Park, Trieste, Italy
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
- * E-mail: (AWB); (AAH); (CS)
| | - Alexander W. Bird
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (AWB); (AAH); (CS)
| |
Collapse
|
30
|
The therapeutic and diagnostic potential of FKBPL; a novel anticancer protein. Drug Discov Today 2012; 17:544-8. [DOI: 10.1016/j.drudis.2012.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/05/2011] [Accepted: 01/09/2012] [Indexed: 11/22/2022]
|
31
|
Ching Chen S, Hseu YC, Sung JC, Chen CH, Chen LC, Chung KT. Induction of DNA damage signaling genes in benzidine-treated HepG2 cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2011; 52:664-72. [PMID: 21818781 DOI: 10.1002/em.20669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 06/20/2011] [Accepted: 06/21/2011] [Indexed: 05/10/2023]
Abstract
We examined genotoxicity and DNA damage response in HepG2 cells following exposure to benzidine. Using the Comet assay, we showed that benzidine (50-200 μM) induces DNA damage in HepG2 cells. DNA damage signaling pathway-based PCR arrays were used to investigate expression changes in genes involved in cell-cycle arrest, apoptosis, and DNA repair and showed upregulation of 23 genes and downregulation of one gene in benzidine-treated cells. Induction of G2/M arrest and apoptosis was confirmed at the protein level. Real-time PCR and Western blots were used to demonstrate the expression of select DNA repair-associated genes from the PCR array. Upregulation of the p53 protein in benzidine-treated cells suggests the induction of the p53 DNA damage signaling pathway. Collectively, DNA damage response genes induced by benzidine indicate recruitment complex molecular machinery involved in DNA repair, cell-cycle arrest, and potentially, activation of the apoptosis.
Collapse
Affiliation(s)
- Ssu Ching Chen
- Department of Life Science, National Central University, Chung-Li City, Taoyan Country, Taiwan, Republic of China.
| | | | | | | | | | | |
Collapse
|
32
|
Chung YS, Cho S, Ryou HJ, Jee HG, Choi JY, Yoon K, Choi HJ, Lee KE, Suh YJ, Oh SK, Youn YK. Is there a treatment advantage when paclitaxel and lovastatin are combined to dose anaplastic thyroid carcinoma cell lines? Thyroid 2011; 21:735-44. [PMID: 21568723 DOI: 10.1089/thy.2010.0304] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is the most aggressive type of thyroid carcinoma. The purpose of this study was to evaluate the combined cytotoxic effects of paclitaxel and lovastatin in ATC cell lines. METHODS ATC cells were treated with paclitaxel and lovastatin, separately or together, and the cytotoxicity of the compounds was determined by quantifying cell viability and apoptosis. We conducted an isobologram analysis to investigate the combined effect of the two drugs. RESULTS In 8505C cells, cellular viability was inhibited by lovastatin and paclitaxel in a concentration-dependent manner (p = 0.002 and p = 0.020, respectively). The IC(50) of lovastatin was 3.53 μM and that of paclitaxel was 5.98 nM. In BHT-101 cells, cellular viability was also inhibited in a concentration-dependent manner by lovastatin and paclitaxel (p = 0.020 and p = 0.032, respectively). The IC(50) of lovastatin was 17.13 μM and that of paclitaxel was 35.26 nM. In 8505C cells, paclitaxel and lovastatin alone induced apoptosis in a concentration-dependent manner. However, both an isobologram analysis on inhibition of viability and an analysis of apoptosis demonstrated antagonism between paclitaxel and lovastatin. In BHT-101 cells, however, neither drug had an apoptotic effect when used individually. There was a variable effect when used in combination, depending on the drug concentrations. CONCLUSIONS Paclitaxel and lovastatin were cytotoxic in two ATC cell lines and increased apoptosis in 8505C cells. However, in these cells, the combination of drugs resulted in antagonism that affected both the cytotoxicity of the compounds and the apoptosis of 8505C cells. The combination of paclitaxel and lovastatin did not enhance the treatment effect in ATC cell lines.
Collapse
Affiliation(s)
- Yoo Seung Chung
- Department of Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
McKeen HD, Brennan DJ, Hegarty S, Lanigan F, Jirstrom K, Byrne C, Yakkundi A, McCarthy HO, Gallagher WM, Robson T. The emerging role of FK506-binding proteins as cancer biomarkers: a focus on FKBPL. Biochem Soc Trans 2011; 39:663-8. [PMID: 21428958 DOI: 10.1042/bst0390663] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
FKBPs (FK506-binding proteins) have long been recognized as key regulators of the response to immunosuppressant drugs and as co-chaperones of steroid receptor complexes. More recently, evidence has emerged suggesting that this diverse protein family may also represent cancer biomarkers owing to their roles in cancer progression and response to treatment. FKBPL (FKBP-like) is a novel FKBP with roles in GR (glucocorticoid receptor), AR (androgen receptor) and ER (oestrogen receptor) signalling. FKBPL binds Hsp90 (heat-shock protein 90) and modulates translocation, transcriptional activation and phosphorylation of these steroid receptors. It has been proposed as a novel prognostic and predictive biomarker, where high levels predict for increased recurrence-free survival in breast cancer patients and enhanced sensitivity to endocrine therapy. Since this protein family has roles in a plethora of signalling pathways, its members represent novel prognostic markers and therapeutic targets for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Hayley D McKeen
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Microtubules are dynamic filamentous cytoskeletal proteins composed of tubulin and are an important therapeutic target in tumour cells. Agents that bind to microtubules have been part of the pharmacopoeia of anticancer therapy for decades and until the advent of targeted therapy, microtubules were the only alternative to DNA as a therapeutic target in cancer. The screening of a range of botanical species and marine organisms has yielded promising new antitubulin agents with novel properties. In the current search for novel microtubule-binding agents, enhanced tumour specificity, reduced neurotoxicity and insensitivity to chemoresistance mechanisms are the three main objectives.
Collapse
Affiliation(s)
- Charles Dumontet
- INSERM 590, Faculté Rockefeller, 8 Avenue Rockefeller, 69008 Lyon, France and Université Lyon 1, ISPB, Lyon, F-69003, France.
| | | |
Collapse
|