1
|
Li Y, Tang X, Wang B, Chen M, Zheng J, Chang K. Current landscape of exosomal non-coding RNAs in prostate cancer: Modulators and biomarkers. Noncoding RNA Res 2024; 9:1351-1362. [PMID: 39247145 PMCID: PMC11380467 DOI: 10.1016/j.ncrna.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 09/10/2024] Open
Abstract
Prostate cancer (PCa) has the highest frequency of diagnosis among solid tumors and ranks second as the primary cause of cancer-related deaths. Non-coding RNAs (ncRNAs), such as microRNAs, long non-coding RNAs and circular RNAs, frequently exhibit dysregulation and substantially impact the biological behavior of PCa. Compared with circulating ncRNAs, ncRNAs loaded into exosomes are more stable because of protection by the lipid bilayer. Furthermore, exosomal ncRNAs facilitate the intercellular transfer of molecules and information. Increasing evidence suggests that exosomal ncRNAs hold promising potential in the progression, diagnosis and prognosis of PCa. This review aims to discuss the functions of exosomal ncRNAs in PCa, evaluate their possible applications as clinical biomarkers and therapeutic targets, and provide a comprehensive overview of the ncRNAs regulatory network in PCa. We also identified ncRNAs that can be utilized as biomarkers for diagnosis, staging, grading and prognosis assessment in PCa. This review offers researchers a fresh perspective on the functions of exosomal ncRNAs in PCa and provides additional options for its diagnosis, progression monitoring, and prognostic prediction.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, PR China
- School of Medicine, Chongqing University, Chongqing, 400030, PR China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Binpan Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, PR China
- School of Medicine, Chongqing University, Chongqing, 400030, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| |
Collapse
|
2
|
Cui J, Liu X, Shang Q, Sun S, Chen S, Dong J, Zhu Y, Liu L, Xia Y, Wang Y, Xiang L, Fan B, Zhan J, Zhou Y, Chen P, Zhao R, Liu X, Xing N, Wu D, Shi B, Zou Y. Deubiquitination of CDC6 by OTUD6A promotes tumour progression and chemoresistance. Mol Cancer 2024; 23:86. [PMID: 38685067 PMCID: PMC11057083 DOI: 10.1186/s12943-024-01996-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND CDC6 is an oncogenic protein whose expression level fluctuates during the cell cycle. Although several E3 ubiquitin ligases responsible for the ubiquitin-mediated proteolysis of CDC6 have been identified, the deubiquitination pathway for CDC6 has not been investigated. METHODS The proteome-wide deubiquitinase (DUB) screening was used to identify the potential regulator of CDC6. Immunofluorescence, protein half-life and deubiquitination assays were performed to determine the protein stability of CDC6. Gain- and loss-of-function experiments were implemented to analyse the impacts of OUTD6A-CDC6 axis on tumour growth and chemosensitivity in vitro. N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced conditional Otud6a knockout (CKO) mouse model and tumour xenograft model were performed to analyse the role of OTUD6A-CDC6 axis in vivo. Tissue specimens were used to determine the association between OTUD6A and CDC6. RESULTS OTUD6A interacts with, depolyubiquitinates and stabilizes CDC6 by removing K6-, K33-, and K48-linked polyubiquitination. Moreover, OTUD6A promotes cell proliferation and decreases sensitivity to chemotherapy by upregulating CDC6. CKO mice are less prone to BCa tumorigenesis induced by BBN, and knockdown of OTUD6A inhibits tumour progression in vivo. Furthermore, OTUD6A protein level has a positive correlation with CDC6 protein level, and high protein levels of OTUD6A and CDC6 are associated with poor prognosis in patients with bladder cancer. CONCLUSIONS We reveal an important yet missing piece of novel DUB governing CDC6 stability. In addition, our findings propose a model for the OTUD6A-CDC6 axis that provides novel insights into cell cycle and chemosensitivity regulation, which may become a potential biomarker and promising drug target for cancer treatment.
Collapse
Affiliation(s)
- Jianfeng Cui
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaochen Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
- Department of Clinical laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Qinghong Shang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Shuna Sun
- Department of Dermatology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Jianping Dong
- Department of Urology, Shouguang People's Hospital, Weifang, Shandong, 262750, China
| | - Yaofeng Zhu
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Liu
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Yangyang Xia
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Yong Wang
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Lu Xiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Bowen Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Jiafeng Zhan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Yadi Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Renchang Zhao
- Department of Thoracic Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaofei Liu
- Departement of Breast and Thyroid Surgery, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Dalei Wu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China.
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China.
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
Merjaneh N, Hajjar M, Lan YW, Kalinichenko VV, Kalin TV. The Promise of Combination Therapies with FOXM1 Inhibitors for Cancer Treatment. Cancers (Basel) 2024; 16:756. [PMID: 38398147 PMCID: PMC10886945 DOI: 10.3390/cancers16040756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Forkhead box M1 (FOXM1) is a transcription factor in the forkhead (FOX) family, which is required for cellular proliferation in normal and neoplastic cells. FOXM1 is highly expressed in many different cancers, and its expression is associated with a higher tumor stage and worse patient-related outcomes. Abnormally high expression of FOXM1 in cancers compared to normal tissue makes FOXM1 an attractive target for pharmacological inhibition. FOXM1-inhibiting agents and specific FOXM1-targeted small-molecule inhibitors have been developed in the lab and some of them have shown promising efficacy and safety profiles in mouse models. While the future goal is to translate FOXM1 inhibitors to clinical trials, potential synergistic drug combinations can maximize anti-tumor efficacy while minimizing off-target side effects. Hence, we discuss the rationale and efficacy of all previously studied drug combinations with FOXM1 inhibitors for cancer therapies.
Collapse
Affiliation(s)
- Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Mona Hajjar
- The Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA;
| | - Ying-Wei Lan
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| | - Vladimir V. Kalinichenko
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
- Division of Neonatology, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
| | - Tanya V. Kalin
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| |
Collapse
|
4
|
Choupani E, Madjd Z, Saraygord-Afshari N, Kiani J, Hosseini A. Combination of androgen receptor inhibitor enzalutamide with the CDK4/6 inhibitor ribociclib in triple negative breast cancer cells. PLoS One 2022; 17:e0279522. [PMID: 36548336 PMCID: PMC9779032 DOI: 10.1371/journal.pone.0279522] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer (BC) that currently lacks specific therapy options. Thus, chemotherapy continues to be the primary treatment, and developing novel targets is a top clinical focus. The androgen receptor (AR) has emerged as a therapeutic target in a subtype of TNBC, with substantial clinical benefits shown in various clinical studies. Numerous studies have shown that cancer is associated with changes in components of the cell cycle machinery. Although cell cycle cyclin-dependent kinase (CDK) 4/6 inhibitors are successful in the treatment of ER-positive BC, they are not helpful in the treatment of patients with TNBC. We investigated the possibility of combining CDK4/6 inhibitor(ribociclib) with AR inhibitor(enzalutamide) in the AR-positive TNBC cell line. Ribociclib showed an inhibitory effect in TNBC cells. Additionally, we found that enzalutamide reduced cell migration/invasion, clonogenic capacity, cell cycle progression, and cell growth in AR-positive cells. Enzalutamide therapy could increase the cytostatic impact of ribociclib in AR+ TNBC cells. Furthermore, dual inhibition of AR and CDK4/6 demonstrated synergy in an AR+ TNBC model compared to each treatment alone.
Collapse
Affiliation(s)
- Edris Choupani
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arshad Hosseini
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- * E-mail:
| |
Collapse
|
5
|
Ma N, Liu H, Wu Y, Yao M, Zhang B. Inhibition of N-Acetyltransferase 10 Suppresses the Progression of Prostate Cancer through Regulation of DNA Replication. Int J Mol Sci 2022; 23:ijms23126573. [PMID: 35743017 PMCID: PMC9223896 DOI: 10.3390/ijms23126573] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer suppression through the inhibition of N-acetyltransferase 10 (NAT10) by its specific inhibitor Remodelin has been demonstrated in a variety of human cancers. Here, we report the inhibitory effects of Remodelin on prostate cancer (PCa) cells and the possible associated mechanisms. The prostate cancer cell lines VCaP, LNCaP, PC3, and DU145 were used. The in vitro proliferation, migration, and invasion of cells were measured by a cell proliferation assay, colony formation, wound healing, and Transwell assays, respectively. In vivo tumor growth was analyzed by transplantation into nude mice. The inhibition of NAT10 by Remodelin not only suppressed growth, migration, and invasion in vitro, but also the in vivo cancer growth of prostate cancer cells. The involvement of NAT10 in DNA replication was assessed by EdU labeling, DNA spreading, iPOND, and ChIP-PCR assays. The inhibition of NAT10 by Remodelin slowed DNA replication. NAT10 was detected in the prereplication complex, and it could also bind to DNA replication origins. Furthermore, the interaction between NAT10 and CDC6 was analyzed by Co-IP. The altered expression of NAT10 was measured by immunofluorescence staining and Western blotting. Remodelin markedly reduced the levels of CDC6 and AR. The expression of NAT10 could be altered under either castration or noncastration conditions, and Remodelin still suppressed the growth of in vitro-induced castration-resistant prostate cancers. The analysis of a TCGA database revealed that the overexpression of NAT10, CDC6, and MCM7 in prostate cancers were correlated with the Gleason score and node metastasis. Our data demonstrated that Remodelin, an inhibitor of NAT10, effectively inhibits the growth of prostate cancer cells under either no castration or castration conditions, likely by impairing DNA replication.
Collapse
Affiliation(s)
| | | | | | | | - Bo Zhang
- Correspondence: ; Tel.: +86-10-82802627
| |
Collapse
|
6
|
Castaneda M, den Hollander P, Mani SA. Forkhead Box Transcription Factors: Double-Edged Swords in Cancer. Cancer Res 2022; 82:2057-2065. [PMID: 35315926 PMCID: PMC9258984 DOI: 10.1158/0008-5472.can-21-3371] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/12/2022] [Accepted: 03/14/2022] [Indexed: 01/07/2023]
Abstract
A plethora of treatment options exist for cancer therapeutics, but many are limited by side effects and either intrinsic or acquired resistance. The need for more effective targeted cancer treatment has led to the focus on forkhead box (FOX) transcription factors as possible drug targets. Forkhead factors such as FOXA1 and FOXM1 are involved in hormone regulation, immune system modulation, and disease progression through their regulation of the epithelial-mesenchymal transition. Forkhead factors can influence cancer development, progression, metastasis, and drug resistance. In this review, we discuss the various roles of forkhead factors in biological processes that support cancer as well as their function as pioneering factors and their potential as targetable transcription factors in the fight against cancer.
Collapse
Affiliation(s)
- Maria Castaneda
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sendurai A. Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Corresponding Author: Sendurai A. Mani, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Suite 910, Houston, TX 77030-3304. Phone: 713-792-9638; E-mail:
| |
Collapse
|
7
|
Mandigo AC, Shafi AA, McCann JJ, Yuan W, Laufer TS, Bogdan D, Gallagher L, Dylgjeri E, Semenova G, Vasilevskaya IA, Schiewer MJ, McNair CM, de Bono JS, Knudsen KE. Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer. Cancer Res 2022; 82:221-234. [PMID: 34625422 PMCID: PMC9397633 DOI: 10.1158/0008-5472.can-21-1159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/14/2021] [Accepted: 09/09/2021] [Indexed: 01/07/2023]
Abstract
The retinoblastoma tumor suppressor (RB) is a critical regulator of E2F-dependent transcription, controlling a multitude of protumorigenic networks including but not limited to cell-cycle control. Here, genome-wide assessment of E2F1 function after RB loss in isogenic models of prostate cancer revealed unexpected repositioning and cooperation with oncogenic transcription factors, including the major driver of disease progression, the androgen receptor (AR). Further investigation revealed that observed AR/E2F1 cooperation elicited novel transcriptional networks that promote cancer phenotypes, especially as related to evasion of cell death. These observations were reflected in assessment of human disease, indicating the clinical relevance of the AR/E2F1 cooperome in prostate cancer. Together, these studies reveal new mechanisms by which RB loss induces cancer progression and highlight the importance of understanding the targets of E2F1 function. SIGNIFICANCE: This study identifies that RB loss in prostate cancer drives cooperation between AR and E2F1 as coregulators of transcription, which is linked to the progression of advanced disease.
Collapse
Affiliation(s)
- Amy C Mandigo
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ayesha A Shafi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jennifer J McCann
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Wei Yuan
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Talya S Laufer
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Denisa Bogdan
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Lewis Gallagher
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Galina Semenova
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Irina A Vasilevskaya
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Matthew J Schiewer
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Chris M McNair
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Johann S de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Karen E Knudsen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Chen YX, Tan LM, Gong JP, Huang MS, Yin JY, Zhang W, Zhou HH, Liu ZQ. Response prediction biomarkers and drug combinations of PARP inhibitors in prostate cancer. Acta Pharmacol Sin 2021; 42:1970-1980. [PMID: 33589795 PMCID: PMC8632930 DOI: 10.1038/s41401-020-00604-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/20/2020] [Indexed: 01/31/2023] Open
Abstract
PARP inhibitors are a group of inhibitors targeting poly(ADP-ribose) polymerases (PARP1 or PARP2) involved in DNA repair and transcriptional regulation, which may induce synthetic lethality in BRCAness tumors. Systematic analyzes of genomic sequencing in prostate cancer show that ~10%-19% of patients with primary prostate cancer have inactivated DNA repair genes, with a notably higher proportion of 23%-27% in patients with metastatic castration-resistant prostate cancer (mCRPC). These characteristic genomic alterations confer possible vulnerability to PARP inhibitors in patients with mCRPC who benefit only modestly from other therapies. However, only a small proportion of patients with mCRPC shows sensitivity to PARP inhibitors, and these sensitive patients cannot be fully identified by existing response prediction biomarkers. In this review, we provide an overview of the potential response prediction biomarkers and synergistic combinations studied in the preclinical and clinical stages, which may expand the population of patients with prostate cancer who may benefit from PARP inhibitors.
Collapse
Affiliation(s)
- Yi-Xin Chen
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Li-Ming Tan
- Department of Pharmacy, The Second People's Hospital of Huaihua City, Huaihua, 418000, China
| | - Jian-Ping Gong
- Department of Pharmacy, The Second People's Hospital of Huaihua City, Huaihua, 418000, China
| | - Ma-Sha Huang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, China.
| |
Collapse
|
9
|
Gao Y, Liu Y, Liu Y, Peng Y, Yuan B, Fu Y, Qi X, Zhu Q, Cao T, Zhang S, Yin L, Li X. UHRF1 promotes androgen receptor-regulated CDC6 transcription and anti-androgen receptor drug resistance in prostate cancer through KDM4C-Mediated chromatin modifications. Cancer Lett 2021; 520:172-183. [PMID: 34265399 DOI: 10.1016/j.canlet.2021.07.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/26/2021] [Accepted: 07/08/2021] [Indexed: 12/30/2022]
Abstract
The UHRF1 and CDC6, oncogenes play critical roles in therapeutic resistance. In the present study, we found that UHRF1 mediates androgen receptor (AR)-regulated CDC6 transcription in prostate cancer cells. In prostate cancer tissues and cell lines, levels of UHRF1 and CDC6 were simultaneously upregulated, and this was associated with worse survival. UHRF1 silencing significantly promoted the cytotoxicity and anti-prostate cancer efficacy of bicalutamide in mouse xenografts by inhibiting CDC6 gene expression. UHRF1 promoted AR-regulated CDC6 transcription by binding to the CCAAT motif near the androgen response element (ARE) in the CDC6 promoter. We further found that UHRF1 promoted androgen-dependent chromatin occupancy of AR protein by recruiting the H3K9me2/3-specific demethyltransferase KDM4C and modifying the intense heterochromatin status. Altogether, we found for the first time that UHRF1 promotes AR-regulated CDC6 transcription through a novel chromatin modification mechanism and contributes to anti-AR drug resistance in prostate cancer. Targeting AR and UHRF1 simultaneously may be a novel and promising therapeutic modality for prostate cancer.
Collapse
Affiliation(s)
- Yingxue Gao
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Yijun Liu
- School of Basic Medicine, Changsha Medical University, China
| | - Youhong Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Yuchong Peng
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, China
| | - Bowen Yuan
- Department of Pathology, The Third Xiangya Hospital, Central South University, China
| | - Yuxin Fu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Xuli Qi
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Qianling Zhu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Tuoyu Cao
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Songwei Zhang
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, China; Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, China
| | - Linglong Yin
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, China
| | - Xiong Li
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, China; NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, China.
| |
Collapse
|
10
|
Piran M, Sepahi N, Moattari A, Rahimi A, Ghanbariasad A. Systems Biomedicine of Primary and Metastatic Colorectal Cancer Reveals Potential Therapeutic Targets. Front Oncol 2021; 11:597536. [PMID: 34249670 PMCID: PMC8263939 DOI: 10.3389/fonc.2021.597536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/31/2021] [Indexed: 12/23/2022] Open
Abstract
Colorectal cancer (CRC) is one of the major causes of cancer deaths across the world. Patients' survival at time of diagnosis depends mainly on stage of the tumor. Therefore, understanding the molecular mechanisms from low-grade to high-grade stages of cancer that lead to cellular migration from one tissue/organ to another tissue/organ is essential for implementing therapeutic approaches. To this end, we performed a unique meta-analysis flowchart by identifying differentially expressed genes (DEGs) between normal, primary (primary sites), and metastatic samples (Colorectal metastatic lesions in liver and lung) in some Test datasets. DEGs were employed to construct a protein-protein interaction (PPI) network. A smaller network containing 39 DEGs was then extracted from the PPI network whose nodes expression induction or suppression alone or in combination with each other would inhibit tumor progression or metastasis. These DEGs were then verified by gene expression profiling, survival analysis, and multiple Validation datasets. We suggested for the first time that downregulation of mitochondrial genes, including ETHE1, SQOR, TST, and GPX3, would help colorectal cancer cells to produce more energy under hypoxic conditions through mechanisms that are different from "Warburg Effect". Augmentation of given antioxidants and repression of P4HA1 and COL1A2 genes could be a choice of CRC treatment. Moreover, promoting active GSK-3β together with expression control of EIF2B would prevent EMT. We also proposed that OAS1 expression enhancement can induce the anti-cancer effects of interferon-gamma, while suppression of CTSH hinders formation of focal adhesions. ATF5 expression suppression sensitizes cancer cells to anchorage-dependent death signals, while LGALS4 induction recovers cell-cell junctions. These inhibitions and inductions would be another combinatory mechanism that inhibits EMT and cell migration. Furthermore, expression inhibition of TMPO, TOP2A, RFC3, GINS1, and CKS2 genes could prevent tumor growth. Besides, TRIB3 suppression would be a promising target for anti-angiogenic therapy. SORD is a poorly studied enzyme in cancer, found to be upregulated in CRC. Finally, TMEM131 and DARS genes were identified in this study whose roles have never been interrogated in any kind of cancer, neither as a biomarker nor curative target. All the mentioned mechanisms must be further validated by experimental wet-lab techniques.
Collapse
Affiliation(s)
- Mehran Piran
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
- Department of Bacteriology and Virology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Sepahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Rahimi
- Bioinformatics and Computational Biology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Ghanbariasad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
11
|
Kalathil D, John S, Nair AS. FOXM1 and Cancer: Faulty Cellular Signaling Derails Homeostasis. Front Oncol 2021; 10:626836. [PMID: 33680951 PMCID: PMC7927600 DOI: 10.3389/fonc.2020.626836] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Forkhead box transcription factor, FOXM1 is implicated in several cellular processes such as proliferation, cell cycle progression, cell differentiation, DNA damage repair, tissue homeostasis, angiogenesis, apoptosis, and redox signaling. In addition to being a boon for the normal functioning of a cell, FOXM1 turns out to be a bane by manifesting in several disease scenarios including cancer. It has been given an oncogenic status based on several evidences indicating its role in tumor development and progression. FOXM1 is highly expressed in several cancers and has also been implicated in poor prognosis. A comprehensive understanding of various aspects of this molecule has revealed its role in angiogenesis, invasion, migration, self- renewal and drug resistance. In this review, we attempt to understand various mechanisms underlying FOXM1 gene and protein regulation in cancer including the different signaling pathways, post-transcriptional and post-translational modifications. Identifying crucial molecules associated with these processes can aid in the development of potential pharmacological approaches to curb FOXM1 mediated tumorigenesis.
Collapse
Affiliation(s)
- Dhanya Kalathil
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Samu John
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Research Centre, University of Kerala, Thiruvananthapuram, India
| | - Asha S Nair
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Research Centre, University of Kerala, Thiruvananthapuram, India
| |
Collapse
|
12
|
MED19 alters AR occupancy and gene expression in prostate cancer cells, driving MAOA expression and growth under low androgen. PLoS Genet 2021; 17:e1008540. [PMID: 33513133 PMCID: PMC7875385 DOI: 10.1371/journal.pgen.1008540] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/10/2021] [Accepted: 01/04/2021] [Indexed: 11/19/2022] Open
Abstract
Androgen deprivation therapy (ADT) is a mainstay of prostate cancer treatment, given the dependence of prostate cells on androgen and the androgen receptor (AR). However, tumors become ADT-resistant, and there is a need to understand the mechanism. One possible mechanism is the upregulation of AR co-regulators, although only a handful have been definitively linked to disease. We previously identified the Mediator subunit MED19 as an AR co-regulator, and reported that MED19 depletion inhibits AR transcriptional activity and growth of androgen-insensitive LNCaP-abl cells. Therefore, we proposed that MED19 upregulation would promote AR activity and drive androgen-independent growth. Here, we show that stable overexpression of MED19 in androgen-dependent LNCaP cells promotes growth under conditions of androgen deprivation. To delineate the mechanism, we determined the MED19 and AR transcriptomes and cistromes in control and MED19-overexpressing LNCaP cells. We also examined genome-wide H3K27 acetylation. MED19 overexpression selectively alters AR occupancy, H3K27 acetylation, and gene expression. Under conditions of androgen deprivation, genes regulated by MED19 correspond to genes regulated by ELK1, a transcription factor that binds the AR N-terminus to induce select AR target gene expression and proliferation, and genomic sites occupied by MED19 and AR are enriched for motifs associated with ELK1. Strikingly, MED19 upregulates expression of monoamine oxidase A (MAOA), a factor that promotes prostate cancer growth. MAOA depletion reduces androgen-independent growth. MED19 and AR occupy the MAOA promoter, with MED19 overexpression enhancing AR occupancy and H3K27 acetylation. Furthermore, MED19 overexpression increases ELK1 occupancy at the MAOA promoter, and ELK1 depletion reduces MAOA expression and androgen-independent growth. This suggests that MED19 cooperates with ELK1 to regulate AR occupancy and H3K27 acetylation at MAOA, upregulating its expression and driving androgen independence in prostate cancer cells. This study provides important insight into the mechanisms of prostate cancer cell growth under low androgen, and underscores the importance of the MED19-MAOA axis in this process.
Collapse
|
13
|
Jorgez CJ, Seth A, Wilken N, Bournat JC, Chen CH, Lamb DJ. E2F1 regulates testicular descent and controls spermatogenesis by influencing WNT4 signaling. Development 2021; 148:dev191189. [PMID: 33441379 PMCID: PMC7823160 DOI: 10.1242/dev.191189] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022]
Abstract
Cryptorchidism is the most common urologic birth defect in men and is a predisposing factor of male infertility and testicular cancer, yet the etiology remains largely unknown. E2F1 microdeletions and microduplications contribute to cryptorchidism, infertility and testicular tumors. Although E2f1 deletion or overexpression in mice causes spermatogenic failure, the mechanism by which E2f1 influences testicular function is unknown. This investigation revealed that E2f1-null mice develop cryptorchidism with severe gubernacular defects and progressive loss of germ cells resulting in infertility and, in rare cases, testicular tumors. It was hypothesized that germ cell depletion resulted from an increase in WNT4 levels. To test this hypothesis, the phenotype of a double-null mouse model lacking both Wnt4 and E2f1 in germ cells was analyzed. Double-null mice are fertile. This finding indicates that germ cell maintenance is dependent on E2f1 repression of Wnt4, supporting a role for Wnt4 in germ cell survival. In the future, modulation of WNT4 expression in men with cryptorchidism and spermatogenic failure due to E2F1 copy number variations may provide a novel approach to improve their spermatogenesis and perhaps their fertility potential after orchidopexy.
Collapse
Affiliation(s)
- Carolina J Jorgez
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Abhishek Seth
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nathan Wilken
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Juan C Bournat
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ching H Chen
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dolores J Lamb
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Brady Urology Department, Center for Reproductive Genomics and Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
14
|
Youn Y, Lee JC, Kim J, Kim JH, Hwang JH. Cdc6 disruption leads to centrosome abnormalities and chromosome instability in pancreatic cancer cells. Sci Rep 2020; 10:16518. [PMID: 33020506 PMCID: PMC7536414 DOI: 10.1038/s41598-020-73474-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Cell division cycle 6 (Cdc6) plays key roles in regulating DNA replication, and activation and maintenance of cell cycle check points. In addition, Cdc6 exerts oncogenic properties via genomic instability associated with incomplete DNA replication. This study aimed to examine the effects of Cdc6 on pancreatic cancer (PC) cells. Our results showed that Cdc6 expression was higher in clinical PC specimens (based on analysis of the GEPIA database) and cell lines, and the high Cdc6 expression was associated with poorer survival in The Cancer Genome Atlas-PC cohort. In addition, Cdc6-depleted PC cells significantly inhibited cell proliferation and colony formation, delayed G2/M cell cycle progression, and increased expression of p-histone H3 and cyclin A2 levels. These observations could be explained by Cdc6 depletion leading to multipolar and split spindles via centrosome amplification and microtubule disorganization which eventually increases chromosome missegregation. Furthermore, Cdc6-depleted PC cells showed significantly increased apoptosis, which was consistent with increased caspase-9 and caspase-3 activation. Collectively, our results demonstrated that Cdc6-depleted PC cells are arrested in mitosis and eventually undergo cell death by induced multipolar spindles, centrosome aberrations, microtubule disorganization, and chromosome instability. In conclusion, Cdc6 may be a potential biomarker and therapeutic target for PC.
Collapse
Affiliation(s)
- Yuna Youn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jae Hyeong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea. .,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
15
|
Baicalin suppresses the cell cycle progression and proliferation of prostate cancer cells through the CDK6/FOXM1 axis. Mol Cell Biochem 2020; 469:169-178. [DOI: 10.1007/s11010-020-03739-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
|
16
|
Jin Y, Liang Z, Lou H. The Emerging Roles of Fox Family Transcription Factors in Chromosome Replication, Organization, and Genome Stability. Cells 2020; 9:cells9010258. [PMID: 31968679 PMCID: PMC7016735 DOI: 10.3390/cells9010258] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
The forkhead box (Fox) transcription factors (TFs) are widespread from yeast to humans. Their mutations and dysregulation have been linked to a broad spectrum of malignant neoplasias. They are known as critical players in DNA repair, metabolism, cell cycle control, differentiation, and aging. Recent studies, especially those from the simple model eukaryotes, revealed unexpected contributions of Fox TFs in chromosome replication and organization. More importantly, besides functioning as a canonical TF in cell signaling cascades and gene expression, Fox TFs can directly participate in DNA replication and determine the global replication timing program in a transcription-independent mechanism. Yeast Fox TFs preferentially recruit the limiting replication factors to a subset of early origins on chromosome arms. Attributed to their dimerization capability and distinct DNA binding modes, Fkh1 and Fkh2 also promote the origin clustering and assemblage of replication elements (replication factories). They can mediate long-range intrachromosomal and interchromosomal interactions and thus regulate the four-dimensional chromosome organization. The novel aspects of Fox TFs reviewed here expand their roles in maintaining genome integrity and coordinating the multiple essential chromosome events. These will inevitably be translated to our knowledge and new treatment strategies of Fox TF-associated human diseases including cancer.
Collapse
|
17
|
Kim YH, Byun YJ, Kim WT, Jeong P, Yan C, Kang HW, Kim YJ, Lee SC, Moon SK, Choi YH, Yun SJ, Kim WJ. CDC6 mRNA Expression Is Associated with the Aggressiveness of Prostate Cancer. J Korean Med Sci 2018; 33:e303. [PMID: 30450027 PMCID: PMC6236078 DOI: 10.3346/jkms.2018.33.e303] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 08/28/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cell division cycle 6 (CDC6) is an essential regulator of DNA replication and plays important roles in the activation and maintenance of the checkpoint mechanisms in the cell cycle. CDC6 has been associated with oncogenic activities in human cancers; however, the clinical significance of CDC6 in prostate cancer (PCa) remains unclear. Therefore, we investigated whether the CDC6 mRNA expression level is a diagnostic and prognostic marker in PCa. METHODS The study subjects included 121 PCa patients and 66 age-matched benign prostatic hyperplasia (BPH) patients. CDC6 expression was evaluated using real-time polymerase chain reaction and immunohistochemical (IH) staining, and then compared according to the clinicopathological characteristics of PCa. RESULTS CDC6 mRNA expression was significantly higher in PCa tissues than in BPH control tissues (P = 0.005). In addition, CDC6 expression was significantly higher in patients with elevated prostate-specific antigen (PSA) levels (> 20 ng/mL), a high Gleason score, and advanced stage than in those with low PSA levels, a low Gleason score, and earlier stage, respectively. Multivariate logistic regression analysis showed that high expression of CDC6 was significantly associated with advanced stage (≥ T3b) (odds ratio [OR], 3.005; confidence interval [CI], 1.212-7.450; P = 0.018) and metastasis (OR, 4.192; CI, 1.079-16.286; P = 0.038). Intense IH staining for CDC6 was significantly associated with a high Gleason score and advanced tumor stage including lymph node metastasis stage (linear-by-linear association, P = 0.044 and P = 0.003, respectively). CONCLUSION CDC6 expression is associated with aggressive clinicopathological characteristics in PCa. CDC6 may be a potential diagnostic and prognostic marker in PCa patients.
Collapse
Affiliation(s)
- Ye-Hwan Kim
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Young Joon Byun
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Won Tae Kim
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Pildu Jeong
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Chunri Yan
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
- Department of Preventive Medicine, Medical College, Yanbian University, Yanji, China
| | - Ho Won Kang
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Yong-June Kim
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Sang-Cheol Lee
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, Korea
| | - Yung-Hyun Choi
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan, Korea
| | - Seok Joong Yun
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University, College of Medicine, Cheongju, Korea
| |
Collapse
|
18
|
Yu X, Liu Y, Yin L, Peng Y, Peng Y, Gao Y, Yuan B, Zhu Q, Cao T, Xie B, Sun L, Chen Y, Gong Z, Qiu Y, Fan X, Li X. Radiation-promoted CDC6 protein stability contributes to radioresistance by regulating senescence and epithelial to mesenchymal transition. Oncogene 2018; 38:549-563. [PMID: 30158672 PMCID: PMC6345673 DOI: 10.1038/s41388-018-0460-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 07/11/2018] [Accepted: 07/24/2018] [Indexed: 12/15/2022]
Abstract
Ionizing radiation (IR) is a conventional cancer therapeutic, to which cancer cells develop radioresistance with exposure. The residual cancer cells after radiation treatment also have increased metastatic potential. The mechanisms by which cancer cells develop radioresistance and gain metastatic potential are still unknown. In this study acute IR exposure induced cancer cell senescence and apoptosis, but after long-term IR exposure, cancer cells exhibited radioresistance. The proliferation of radioresistant cells was retarded, and most cells were arrested in G0/G1 phase. The radioresistant cells simultaneously showed resistance to further IR-induced apoptosis, premature senescence, and epithelial to mesenchymal transformation (EMT). Acute IR exposure steadily elevated CDC6 protein levels due to the attenuation of ubiquitination, while CDC6 overexpression was observed in the radioresistant cells because the insufficiency of CDC6 phosphorylation blocked protein translocation from nucleus to cytoplasm, resulting in subcellular protein accumulation when the cells were arrested in G0/G1 phase. CDC6 ectopic overexpression in CNE2 cells resulted in apoptosis resistance, G0/G1 cell cycle arrest, premature senescence, and EMT, similar to the characteristics of radioresistant CNE2-R cells. Targeting CDC6 with siRNA promoted IR-induced senescence, sensitized cancer cells to IR-induced apoptosis, and reversed EMT. Furthermore, CDC6 depletion synergistically repressed the growth of CNE2-R xenografts when combined with IR. The study describes for the first time cell models for IR-induced senescence, apoptosis resistance, and EMT, three major mechanisms by which radioresistance develops. CDC6 is a novel radioresistance switch regulating senescence, apoptosis, and EMT. These studies suggest that CDC6highKI67low represents a new diagnostic marker of radiosensitivity, and CDC6 represents a new therapeutic target for cancer radiosensitization.
Collapse
Affiliation(s)
- Xiaohui Yu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youhong Liu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Linglong Yin
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongbo Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Yuchong Peng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxue Gao
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Bowen Yuan
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Qianling Zhu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Tuoyu Cao
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Bowen Xie
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Lunquan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Chen
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanzheng Qiu
- Department of Otorhinolaryngology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuegong Fan
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Li
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China. .,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
19
|
FOXM1 contributes to taxane resistance by regulating UHRF1-controlled cancer cell stemness. Cell Death Dis 2018; 9:562. [PMID: 29752436 PMCID: PMC5948215 DOI: 10.1038/s41419-018-0631-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/22/2018] [Accepted: 04/26/2018] [Indexed: 12/18/2022]
Abstract
Therapy-induced expansion of cancer stem cells (CSCs) has been identified as one of the most critical factors contributing to therapeutic resistance, but the mechanisms of this adaptation are not fully understood. UHRF1 is a key epigenetic regulator responsible for therapeutic resistance, and controls the self-renewal of stem cells. In the present study, taxane-resistant cancer cells were established and stem-like cancer cells were expanded. UHRF1 was overexpressed in the taxane-resistant cancer cells, which maintained CSC characteristics. UHRF1 depletion overcame taxane resistance in vitro and in vivo. Additionally, FOXM1 has been reported to play a role in therapeutic resistance and the self-renewal of CSCs. FOXM1 and UHRF1 are highly correlated in prostate cancer tissues and cells, FOXM1 regulates CSCs by regulating uhrf1 gene transcription in an E2F-independent manner, and FOXM1 protein directly binds to the FKH motifs at the uhrf1 gene promoter. This present study clarified a novel mechanism by which FOXM1 controls CSCs and taxane resistance through a UHRF1-mediated signaling pathway, and validated FOXM1 and UHRF1 as two potential therapeutic targets to overcome taxane resistance.
Collapse
|
20
|
Treatment with docetaxel in combination with Aneustat leads to potent inhibition of metastasis in a patient-derived xenograft model of advanced prostate cancer. Br J Cancer 2018; 118:802-812. [PMID: 29381682 PMCID: PMC5877435 DOI: 10.1038/bjc.2017.474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 01/09/2023] Open
Abstract
Background: Docetaxel used for first-line treatment of advanced prostate cancer (PCa) is only marginally effective. We previously showed, using the LTL-313H subrenal capsule patient-derived metastatic PCa xenograft model, that docetaxel combined with Aneustat (OMN54), a multivalent plant-derived therapeutic, led to marked synergistic tumour growth inhibition. Here, we investigated the effect of docetaxel+Aneustat on metastasis. Methods: C4-2 cells were incubated with docetaxel, Aneustat and docetaxel+Aneustat to assess effects on cell migration. The LTL-313H model, similarly treated, was analysed for effects on lung micro-metastasis and kidney invasion. The LTL-313H gene expression profile was compared with profiles of PCa patients (obtained from Oncomine) and subjected to IPA to determine involvement of cancer driver genes. Results: Docetaxel+Aneustat markedly inhibited C4-2 cell migration and LTL-313H lung micro-metastasis/kidney invasion. Oncomine analysis indicated that treatment with docetaxel+Aneustat was associated with improved patient outcome. The drug combination markedly downregulated expression of cancer driver genes such as FOXM1 (and FOXM1-target genes). FOXM1 overexpression reduced the anti-metastatic activity of docetaxel+Aneustat. Conclusions: Docetaxel+Aneustat can inhibit PCa tissue invasion and metastasis. This activity appears to be based on reduced expression of cancer driver genes such as FOXM1. Use of docetaxel+Aneustat may provide a new, more effective regimen for therapy of metastatic PCa.
Collapse
|
21
|
Zhao R, Wang Y, Zhang M, Gu X, Wang W, Tan J, Wei X, Jin N. Screening of potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles. Oncol Lett 2017; 14:5361-5369. [PMID: 29113170 PMCID: PMC5662906 DOI: 10.3892/ol.2017.6879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/23/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to analyze potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles. First, gene expression profiles GSE38241 and GSE3933 were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between prostate cancer and normal control samples were identified using the Linear Models for Microarray Data package. Pathway enrichment analysis of DEGs was performed using Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes. Furthermore, protein-protein interaction (PPI) networks of DEGs were constructed, on the basis of the Search Tool for the Retrieval of Interacting Genes/Proteins database. The Molecular Complex Detection was utilized to perform module analysis of the PPI networks. In addition, transcriptional regulatory networks were constructed on the basis of the associations between transcription factors (TFs) and target genes. A total of 529 DEGs were identified, including 129 upregulated genes that were primarily associated with to the cell cycle. Additionally, 400 downregulated genes were identified, which were principally enriched in the pathways associated with vascular smooth muscle contraction and focal adhesion. Cell Division Cycle Associated 8, Cell Division Cycle 45, Ubiquitin Conjugating Enzyme E2 C and Thymidine Kinase 1 were identified as hub genes in the upregulated sub-network. Furthermore, the upregulated TF E2F, and the downregulated TF Early Growth Response 1, were identified to be critical in the transcriptional regulatory networks. The identified DEGs and TFs may have critical roles in the progression of prostate cancer, and may be used as target molecules for treating prostate cancer.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yao Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Muchun Zhang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xinquan Gu
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Weihua Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Jiufeng Tan
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xin Wei
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ning Jin
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
22
|
FOXM1 promotes the progression of prostate cancer by regulating PSA gene transcription. Oncotarget 2017; 8:17027-17037. [PMID: 28199985 PMCID: PMC5370019 DOI: 10.18632/oncotarget.15224] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/09/2017] [Indexed: 12/30/2022] Open
Abstract
Androgen/AR is the primary contributor to prostate cancer (PCa) progression by regulating Prostate Specific Antigen (PSA) gene transcription. The disease inevitably evolves to androgen-independent (AI) status. Other mechanisms by which PSA is regulated and develops to AI have not yet been fully determined. FOXM1 is a cell proliferation-specific transcription factor highly expressed in PCa cells compared to non-malignant prostate epithelial cells, suggesting that the aberrant overexpression of FOXM1 contributes to PCa development. In addition to regulating AR gene transcription and cell cycle-regulatory genes, FOXM1 selectively regulates the gene transcription of KLK2 and PSA, typical androgen responsive genes. Screening the potential FOXM1-binding sites by ChIP-PCR, we found that FOXM1 directly binds to the FHK binding motifs in the PSA promoter/enhancer regions. AI C4-2 cells have more FOXM1 binding sites than androgen dependent LNCaP cells. The depletion of FOXM1 by small molecular inhibitors significantly improves the suppression of PSA gene transcription by the anti-AR agent Cadosax. This is the first report showing that FOXM1 promotes PCa progression by regulating PSA gene transcription, particularly in AI PCa cells. The combination of anti-AR agents and FOXM1 inhibitors has the potential to greatly improve therapy for late-stage PCa patients by suppressing PSA levels.
Collapse
|
23
|
The prognostic significance of Cdc6 and Cdt1 in breast cancer. Sci Rep 2017; 7:985. [PMID: 28428557 PMCID: PMC5430515 DOI: 10.1038/s41598-017-00998-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/21/2017] [Indexed: 01/03/2023] Open
Abstract
DNA replication is a critical step in cell proliferation. Overexpression of MCM2-7 genes correlated with poor prognosis in breast cancer patients. However, the roles of Cdc6 and Cdt1, which work with MCMs to regulate DNA replication, in breast cancers are largely unknown. In the present study, we have shown that the expression levels of Cdc6 and Cdt1 were both significantly correlated with an increasing number of MCM2-7 genes overexpression. Both Cdc6 and Cdt1, when expressed in a high level, alone or in combination, were significantly associated with poorer survival in the breast cancer patient cohort (n = 1441). In line with this finding, the expression of Cdc6 and Cdt1 was upregulated in breast cancer cells compared to normal breast epithelial cells. Expression of Cdc6 and Cdt1 was significantly higher in ER negative breast cancer, and was suppressed when ER signalling was inhibited either by tamoxifen in vitro or letrozole in human subjects. Importantly, breast cancer patients who responded to letrozole expressed significantly lower Cdc6 than those patients who did not respond. Our results suggest that Cdc6 is a potential prognostic marker and therapeutic target in breast cancer patients.
Collapse
|
24
|
Hou L, Chen M, Wang M, Cui X, Gao Y, Xing T, Li J, Deng S, Hu J, Yang H, Jiang J. Systematic analyses of key genes and pathways in the development of invasive breast cancer. Gene 2016; 593:1-12. [DOI: 10.1016/j.gene.2016.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/15/2016] [Accepted: 08/04/2016] [Indexed: 11/29/2022]
|
25
|
Chen C, Lu J, Yu Q, Xiao JR, Wei HF, Song XJ, Ge JB, Tao WD, Qian R, Yu XW, Zhao J. Expression of CDc6 after acute spinal cord injury in adult rats. Neuropeptides 2016; 56:59-67. [PMID: 26899166 DOI: 10.1016/j.npep.2016.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 01/27/2016] [Accepted: 02/01/2016] [Indexed: 01/19/2023]
Abstract
The cell division cycle 6 (CDc6) protein has been primarily investigated as a component of the pre-replicative complex for the initiation of DNA replication. Some studies have shown that CDc6 played a critical role in the development of human carcinoma. However, the expression and roles of CDc6 in the central nervous system remain unknown. We have performed an acute spinal cord injury (SCI) model in adult rats and investigated the dynamic changes of CDc6 expression in spinal cord. Western blot have found that CDc6 protein levels first significantly increase, reach a peak at day 3, and then gradually return to normal level at day 14 after SCI. Double immunofluorescence staining showed that CDc6 immunoreactivity was found in neurons, astrocytes, and microglia. Additionally, colocalization of CDc6/active caspase-3 has been detected in neurons and colocalization of CDc6/proliferating cell nuclear antigen has been detected in astrocytes and microglial. In vitro, CDc6 depletion by short interfering RNA inhibits astrocyte proliferation and reduces cyclin A and cyclin D1 protein levels. CDc6 knockdown also decreases neuronal apoptosis. We speculate that CDc6 might play crucial roles in CNS pathophysiology after SCI.
Collapse
Affiliation(s)
- Chen Chen
- Department of orthopedics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jian Lu
- Department of Neurology, Nantong Second People's Hospital, Nantong 226001, Jiangsu Province, China
| | - Qin Yu
- Department of Medical image, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jian-Ru Xiao
- Department of Orthopedics, Shanghai Changzheng Hospital, 200000 Shanghai, China
| | - Hai-Feng Wei
- Department of Orthopedics, Shanghai Changzheng Hospital, 200000 Shanghai, China
| | - Xin-jian Song
- Department of Neurology, Nantong Second People's Hospital, Nantong 226001, Jiangsu Province, China
| | - Jian-Bing Ge
- Department of Neurology, Nantong Second People's Hospital, Nantong 226001, Jiangsu Province, China
| | - Wei-Dong Tao
- Department of Neurology, Nantong Second People's Hospital, Nantong 226001, Jiangsu Province, China
| | - Rong Qian
- Department of orthopedics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao-Wei Yu
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 200233 Shanghai, China.
| | - Jian Zhao
- Department of Orthopedics, Shanghai Changzheng Hospital, 200000 Shanghai, China.
| |
Collapse
|
26
|
Distinct GAB2 signaling pathways are essential for myeloid and lymphoid transformation and leukemogenesis by BCR-ABL1. Blood 2016; 127:1803-13. [PMID: 26773044 DOI: 10.1182/blood-2015-06-653006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 01/04/2016] [Indexed: 11/20/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) directed against BCR-ABL1, the product of the Philadelphia (Ph) chromosome, have revolutionized treatment of patients with chronic myeloid leukemia (CML). However, acquired resistance to TKIs is a significant clinical problem in CML, and TKI therapy is much less effective against Ph(+)B-cell acute lymphoblastic leukemia (B-ALL). BCR-ABL1, via phosphorylated Tyr177, recruits the adapter GRB2-associated binding protein 2 (GAB2) as part of a GRB2/GAB2 complex. We showed previously that GAB2 is essential for BCR-ABL1-evoked myeloid transformation in vitro. Using a genetic strategy and mouse models of CML and B-ALL, we show here that GAB2 is essential for myeloid and lymphoid leukemogenesis by BCR-ABL1. In the mouse model, recipients of BCR-ABL1-transducedGab2(-/-)bone marrow failed to develop CML-like myeloproliferative neoplasia. Leukemogenesis was restored by expression of GAB2 but not by GAB2 mutants lacking binding sites for its effectors phosphatidylinositol 3-kinase (PI3K) or SRC homology 2-containing phosphotyrosine phosphatase 2 (SHP2). GAB2 deficiency also attenuated BCR-ABL1-induced B-ALL, but only the SHP2 binding site was required. The SHP2 and PI3K binding sites were differentially required for signaling downstream of GAB2. Hence, GAB2 transmits critical transforming signals from Tyr177 to PI3K and SHP2 for CML pathogenesis, whereas only the GAB2-SHP2 pathway is essential for lymphoid leukemogenesis. Given that GAB2 is dispensable for normal hematopoiesis, GAB2 and its effectors PI3K and SHP2 represent promising targets for therapy in Ph(+)hematologic neoplasms.
Collapse
|
27
|
High expression of CDC6 is associated with accelerated cell proliferation and poor prognosis of epithelial ovarian cancer. Pathol Res Pract 2015; 212:239-46. [PMID: 26920249 DOI: 10.1016/j.prp.2015.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/13/2015] [Accepted: 09/16/2015] [Indexed: 12/22/2022]
Abstract
Cell division cycle 6 (CDC6) is an essential regulator of DNA replication and plays important roles in the activation and maintenance of the checkpoint mechanisms in the cell cycle. CDC6 has been associated with the oncogenic activities in human cancers, but the biological function and clinical significance of CDC6 in EOC remain unclear. The aim of the present study is to examine the effect of CDC6 on epithelial ovarian cancer (EOC) cells proliferation. We found that CDC6 protein level was up-regulated in EOC tissues compared with the normal ovary tissues. CDC6 expression correlated significantly with FIGO stage (p<0.001), differentiation grade (p=0.002), ascites (p<0.001), malignant tumor cells in ascites (p=0.004), and lymph node status (p<0.001). In vitro, after the release of ovarian cancer cell line (HO8910) from serum starvation, the expression of CDC6, cyclinD1, and PCNA was up-regulated, whereas p16 expression was down-regulated. Furthermore, down-regulation of CDC6 in HO8910 cells decreased cell proliferation and colony formation. HO8910 cells transfected with sh CDC6#1 underwent G1 phase cell cycle arrest. Collectively, this study provides a novel regulatory signaling pathway of CDC6-regulated EOC growth and a new potential therapeutic target for EOC patients.
Collapse
|
28
|
Large-scale RNA-Seq Transcriptome Analysis of 4043 Cancers and 548 Normal Tissue Controls across 12 TCGA Cancer Types. Sci Rep 2015; 5:13413. [PMID: 26292924 PMCID: PMC4544034 DOI: 10.1038/srep13413] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 07/27/2015] [Indexed: 12/21/2022] Open
Abstract
The Cancer Genome Atlas (TCGA) has accrued RNA-Seq-based transcriptome data for more than 4000 cancer tissue samples across 12 cancer types, translating these data into biological insights remains a major challenge. We analyzed and compared the transcriptomes of 4043 cancer and 548 normal tissue samples from 21 TCGA cancer types, and created a comprehensive catalog of gene expression alterations for each cancer type. By clustering genes into co-regulated gene sets, we identified seven cross-cancer gene signatures altered across a diverse panel of primary human cancer samples. A 14-gene signature extracted from these seven cross-cancer gene signatures precisely differentiated between cancerous and normal samples, the predictive accuracy of leave-one-out cross-validation (LOOCV) were 92.04%, 96.23%, 91.76%, 90.05%, 88.17%, 94.29%, and 99.10% for BLCA, BRCA, COAD, HNSC, LIHC, LUAD, and LUSC, respectively. A lung cancer-specific gene signature, containing SFTPA1 and SFTPA2 genes, accurately distinguished lung cancer from other cancer samples, the predictive accuracy of LOOCV for TCGA and GSE5364 data were 95.68% and 100%, respectively. These gene signatures provide rich insights into the transcriptional programs that trigger tumorigenesis and metastasis, and many genes in the signature gene panels may be of significant value to the diagnosis and treatment of cancer.
Collapse
|
29
|
Cell cycle regulation of human DNA repair and chromatin remodeling genes. DNA Repair (Amst) 2015; 30:53-67. [PMID: 25881042 DOI: 10.1016/j.dnarep.2015.03.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 03/03/2015] [Accepted: 03/20/2015] [Indexed: 01/10/2023]
Abstract
Maintenance of a genome requires DNA repair integrated with chromatin remodeling. We have analyzed six transcriptome data sets and one data set on translational regulation of known DNA repair and remodeling genes in synchronized human cells. These data are available through our new database: www.dnarepairgenes.com. Genes that have similar transcription profiles in at least two of our data sets generally agree well with known protein profiles. In brief, long patch base excision repair (BER) is enriched for S phase genes, whereas short patch BER uses genes essentially equally expressed in all cell cycle phases. Furthermore, most genes related to DNA mismatch repair, Fanconi anemia and homologous recombination have their highest expression in the S phase. In contrast, genes specific for direct repair, nucleotide excision repair, as well as non-homologous end joining do not show cell cycle-related expression. Cell cycle regulated chromatin remodeling genes were most frequently confined to G1/S and S. These include e.g. genes for chromatin assembly factor 1 (CAF-1) major subunits CHAF1A and CHAF1B; the putative helicases HELLS and ATAD2 that both co-activate E2F transcription factors central in G1/S-transition and recruit DNA repair and chromatin-modifying proteins and DNA double strand break repair proteins; and RAD54L and RAD54B involved in double strand break repair. TOP2A was consistently most highly expressed in G2, but also expressed in late S phase, supporting a role in regulating entry into mitosis. Translational regulation complements transcriptional regulation and appears to be a relatively common cell cycle regulatory mechanism for DNA repair genes. Our results identify cell cycle phases in which different pathways have highest activity, and demonstrate that periodically expressed genes in a pathway are frequently co-expressed. Furthermore, the data suggest that S phase expression and over-expression of some multifunctional chromatin remodeling proteins may set up feedback loops driving cancer cell proliferation.
Collapse
|
30
|
Smith OK, Aladjem MI. Chromatin structure and replication origins: determinants of chromosome replication and nuclear organization. J Mol Biol 2014; 426:3330-41. [PMID: 24905010 DOI: 10.1016/j.jmb.2014.05.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/22/2014] [Accepted: 05/24/2014] [Indexed: 12/29/2022]
Abstract
The DNA replication program is, in part, determined by the epigenetic landscape that governs local chromosome architecture and directs chromosome duplication. Replication must coordinate with other biochemical processes occurring concomitantly on chromatin, such as transcription and remodeling, to insure accurate duplication of both genetic and epigenetic features and to preserve genomic stability. The importance of genome architecture and chromatin looping in coordinating cellular processes on chromatin is illustrated by two recent sets of discoveries. First, chromatin-associated proteins that are not part of the core replication machinery were shown to affect the timing of DNA replication. These chromatin-associated proteins could be working in concert, or perhaps in competition, with the transcriptional machinery and with chromatin modifiers to determine the spatial and temporal organization of replication initiation events. Second, epigenetic interactions are mediated by DNA sequences that determine chromosomal replication. In this review, we summarize recent findings and current models linking spatial and temporal regulation of the replication program with epigenetic signaling. We discuss these issues in the context of the genome's three-dimensional structure with an emphasis on events occurring during the initiation of DNA replication.
Collapse
Affiliation(s)
- Owen K Smith
- DNA Replication Group, Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Mirit I Aladjem
- DNA Replication Group, Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|