1
|
Liu B, Lu T, Ding M, Zhou X, Jiang Y, Shang J, Sun W, Hu S, Wang X, Zhou X. Targeting TTK Inhibits Tumorigenesis of T-Cell Lymphoma Through Dephosphorylating p38α and Activating AMPK/mTOR Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413990. [PMID: 39836493 PMCID: PMC11905054 DOI: 10.1002/advs.202413990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/16/2024] [Indexed: 01/23/2025]
Abstract
T-cell lymphoma (TCL) is a group of non-Hodgkin's lymphoma with high heterogeneity and unfavorable prognosis. Current standard treatments have demonstrated limited efficacy in improving the outcomes for TCL patients. Therefore, identification of novel drug targets is urgently needed to improve the prognosis of TCL patients. Through multi-omics analysis, aberrant expression of threonine tyrosine kinase (TTK) in TCL is identified. High expression of TTK is closely associated with poor prognosis in TCL patients. Targeting TTK through gene knockdown exerts anti-tumor effects in vitro and in vivo, including inhibiting the cell proliferation, inducing G2/M phase arrest, enhancing DNA damage and cell apoptosis. Mechanically, p38α is identified as the potential phosphorylation substrate of TTK through phosphoproteomic quantification and motif prediction. Furthermore, inhibition of TTK suppresses activation of p38α through dephosphorylating it at Thr180/Tyr182, thereby promoting the activation of AMPK/mTOR pathway. In addition, targeting TTK enhances the autophagy in TCL cells through dephosphorylating p38α. CFI-402257, a specific inhibitor of TTK, is found to exhibit anti-tumor effects and exerted synergistic efficacy with PI3K inhibitor, Duvelisib, in TCL. The study shows that TTK contributes to the development of TCL by regulating p38α-mediated AMPK/mTOR pathway. CFI-402257 is expected to be a promising strategy for TCL treatment.
Collapse
Affiliation(s)
- Bingyu Liu
- Department of HematologyShandong Provincial HospitalCheeloo College of MedicineShandong UniversityJinanShandong250021China
| | - Tiange Lu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
| | - Mengfei Ding
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
| | - Xiaoli Zhou
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
| | - Yujie Jiang
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
| | - Juanjuan Shang
- Department of HematologyShandong Provincial HospitalCheeloo College of MedicineShandong UniversityJinanShandong250021China
| | - Wenyue Sun
- Department of HematologyShandong Provincial HospitalCheeloo College of MedicineShandong UniversityJinanShandong250021China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
| | - Xin Wang
- Department of HematologyShandong Provincial HospitalCheeloo College of MedicineShandong UniversityJinanShandong250021China
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandong250021China
| | - Xiangxiang Zhou
- Department of HematologyShandong Provincial HospitalCheeloo College of MedicineShandong UniversityJinanShandong250021China
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021China
| |
Collapse
|
2
|
Calheiros-Lobo M, Silva JPN, Pinto B, Monteiro L, Silva PMA, Bousbaa H. Exploring the Therapeutic Implications of Co-Targeting the EGFR and Spindle Assembly Checkpoint Pathways in Oral Cancer. Pharmaceutics 2024; 16:1196. [PMID: 39339232 PMCID: PMC11435222 DOI: 10.3390/pharmaceutics16091196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Head and neck cancer (HNC), the sixth most common cancer worldwide, is increasing in incidence, with oral squamous cell carcinoma (OSCC) as the predominant subtype. OSCC mainly affects middle-aged to elderly males, often occurring on the posterior lateral border of the tongue, leading to significant disfigurement and functional impairments, such as swallowing and speech difficulties. Despite advancements in understanding OSCC's genetic and epigenetic variations, survival rates for advanced stages remain low, highlighting the need for new treatment options. Primary treatment includes surgery, often combined with radiotherapy (RT) and chemotherapy (CT). Cetuximab-based chemotherapy, targeting the overexpressed epidermal growth factor receptor (EGFR) in 80-90% of HNCs, is commonly used but correlates with poor prognosis. Additionally, monopolar spindle 1 (MPS1), a spindle assembly checkpoint (SAC) component, is a significant target due to its role in genomic fidelity during mitosis and its overexpression in several cancers. This review explores EGFR and MPS1 as therapeutic targets in HNC, analyzing their molecular mechanisms and the effects of their inhibition on cancer cells. It also highlights the promise of combinatorial approaches, such as microtubule-targeting agents (MTAs) and antimitotic agents, in improving HNC therapies, patient outcomes, and survival rates.
Collapse
Affiliation(s)
- Mafalda Calheiros-Lobo
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (M.C.-L.); (J.P.N.S.); (B.P.); (L.M.)
| | - João P. N. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (M.C.-L.); (J.P.N.S.); (B.P.); (L.M.)
| | - Bárbara Pinto
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (M.C.-L.); (J.P.N.S.); (B.P.); (L.M.)
| | - Luís Monteiro
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (M.C.-L.); (J.P.N.S.); (B.P.); (L.M.)
- Medicine and Oral Surgery Department, University Institute of Health Sciences—CESPU (IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Patrícia M. A. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (M.C.-L.); (J.P.N.S.); (B.P.); (L.M.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Hassan Bousbaa
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (M.C.-L.); (J.P.N.S.); (B.P.); (L.M.)
| |
Collapse
|
3
|
Liang C, Zhou Y, Xin L, Kang K, Tian L, Zhang D, Li H, Zhao Q, Gao H, Shi Z. Hijacking monopolar spindle 1 (MPS1) for various cancer types by small molecular inhibitors: Deep insights from a decade of research and patents. Eur J Med Chem 2024; 273:116504. [PMID: 38795520 DOI: 10.1016/j.ejmech.2024.116504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/28/2024]
Abstract
Monopolar spindle 1 (MPS1) has garnered significant attention due to its pivotal role in regulating the cell cycle. Anomalous expression and hyperactivation of MPS1 have been associated with the onset and advancement of diverse cancers, positioning it as a promising target for therapeutic interventions. This review focuses on MPS1 small molecule inhibitors from the past decade, exploring design strategies, structure-activity relationships (SAR), safety considerations, and clinical performance. Notably, we propose prospects for MPS1 degraders based on proteolysis targeting chimeras (PROTACs), as well as reversible covalent bonding as innovative MPS1 inhibitor design strategies. The objective is to provide valuable information for future development and novel perspectives on potential MPS1 inhibitors.
Collapse
Affiliation(s)
- Chengyuan Liang
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an, 710021, China; Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China.
| | - Ying Zhou
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an, 710021, China; Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China
| | - Liang Xin
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an, 710021, China; Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China
| | - Kairui Kang
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an, 710021, China; Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China
| | - Lei Tian
- Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China; College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science& Technology, Xi'an, 710021, China
| | - Dezhu Zhang
- Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China; Shaanxi Panlong Pharmaceutical Group Co., Ltd., Xi'an, 710025, China
| | - Han Li
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an, 710021, China; Shaanxi Pioneer Biotech Co., Ltd., Xi'an, 710082, China
| | - Qianqian Zhao
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an, 710021, China; Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China
| | - Hong Gao
- Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Research of Xi'an, Xi'an, 710021, China; Shaanxi Pioneer Biotech Co., Ltd., Xi'an, 710082, China
| | - Zhenfeng Shi
- Department of Urology Surgery Center, The People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, 830002, China
| |
Collapse
|
4
|
Naik A, Lattab B, Qasem H, Decock J. Cancer testis antigens: Emerging therapeutic targets leveraging genomic instability in cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200768. [PMID: 38596293 PMCID: PMC10876628 DOI: 10.1016/j.omton.2024.200768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cancer care has witnessed remarkable progress in recent decades, with a wide array of targeted therapies and immune-based interventions being added to the traditional treatment options such as surgery, chemotherapy, and radiotherapy. However, despite these advancements, the challenge of achieving high tumor specificity while minimizing adverse side effects continues to dictate the benefit-risk balance of cancer therapy, guiding clinical decision making. As such, the targeting of cancer testis antigens (CTAs) offers exciting new opportunities for therapeutic intervention of cancer since they display highly tumor specific expression patterns, natural immunogenicity and play pivotal roles in various biological processes that are critical for tumor cellular fitness. In this review, we delve deeper into how CTAs contribute to the regulation and maintenance of genomic integrity in cancer, and how these mechanisms can be exploited to specifically target and eradicate tumor cells. We review the current clinical trials targeting aforementioned CTAs, highlight promising pre-clinical data and discuss current challenges and future perspectives for future development of CTA-based strategies that exploit tumor genomic instability.
Collapse
Affiliation(s)
- Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Boucif Lattab
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Hanan Qasem
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
5
|
Zeng Y, Ren X, Jin P, Zhang Y, Zhuo M, Wang J. Development of MPS1 Inhibitors: Recent Advances and Perspectives. J Med Chem 2023; 66:16484-16514. [PMID: 38095579 DOI: 10.1021/acs.jmedchem.3c00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Monopolar spindle kinase 1 (MPS1) plays a pivotal role as a dual-specificity kinase governing spindle assembly checkpoint activation and sister chromatid separation in mitosis. Its overexpression has been observed in various human malignancies. MPS1 reduces spindle assembly checkpoint sensitivity, allowing tumor cells with a high degree of aneuploidy to complete mitosis and survive. Thus, MPS1 has emerged as a promising candidate for cancer therapy. Despite the identification of numerous MPS1 inhibitors, only five have advanced to clinical trials with none securing FDA approval for cancer treatment. In this perspective, we provide a concise overview of the structural and functional characteristics of MPS1 by highlighting its relevance to cancer. Additionally, we explore the structure-activity relationships, selectivity, and pharmacokinetics of MPS1 inhibitors featuring diverse scaffolds. Moreover, we review the reported work on enhancing MPS1 inhibitor selectivity, offering valuable insights into the discovery of novel, highly potent small-molecule MPS1 inhibitors.
Collapse
Affiliation(s)
- Yangjie Zeng
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Pengyao Jin
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Yali Zhang
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Ming Zhuo
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Jubo Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
6
|
Zhang X, Huang L, Sun J, Liu J, Zong Y, Wan L, Yang X, Yan X, Zhang Y, Zhao R, Liu J, Zhong H, Wei C, Yang X, Tai Y, Han Y, Wang Y. Monopolar spindle 1 contributes to tamoxifen resistance in breast cancer through phosphorylation of estrogen receptor α. Breast Cancer Res Treat 2023; 202:595-606. [PMID: 37695401 DOI: 10.1007/s10549-023-07098-5] [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: 04/10/2023] [Accepted: 08/06/2023] [Indexed: 09/12/2023]
Abstract
PURPOSE The overexpression of mitotic kinase monopolar spindle 1 (Mps1) has been identified in many tumor types, and targeting Mps1 for tumor therapy has shown great promise in multiple preclinical cancer models. However, the role played by Mps1 in tamoxifen (TAM) resistance in breast cancer has never been reported. METHODS The sensitivity of breast cancer cells to tamoxifen was analysed in colony formation assays and wound healing assays. Enhanced transactivational activity of estrogen receptor α (ERα) led by Mps1 overexpression was determined by luciferase assays. The interaction between Mps1 and ERα was verified by co-immunoprecipitation and proximity ligation assay. Phosphorylation of ERα by Mps1 was detected by in vitro kinase assay and such phosphorylation process in vivo was proven by co-immunoprecipitation. The potential phosphorylation site(s) of ERα were analyzed by mass spectrometry. RESULTS Mps1 determines the sensitivity of breast cancer cells to tamoxifen treatment. Mps1 overexpression rendered breast cancer cells more resistant to tamoxifen, while an Mps1 inhibitor or siMps1 oligos enabled cancer cells to overcome tamoxifen resistance. Mechanistically, Mps1 interacted with estrogen receptor α and stimulated its transactivational activity in a kinase activity-dependent manner. Mps1 was critical for ERα phosphorylation at Thr224 amino acid site. Importantly, Mps1 failed to enhance the transactivational activity of the ERα-T224A mutant. CONCLUSION Mps1 contributes to tamoxifen resistance in breast cancer and is a potential therapeutic that can overcome tamoxifen resistance in breast cancer.
Collapse
Affiliation(s)
- Xuemiao Zhang
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, The Training Site for Postgraduates of Jinzhou Medical University, Jinzhou, 121001, China
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jing Sun
- China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Jialong Liu
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yulong Zong
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaopan Yang
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xue Yan
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yanhong Zhang
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Ruzhou Zhao
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jing Liu
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaoli Yang
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, The Training Site for Postgraduates of Jinzhou Medical University, Jinzhou, 121001, China
- Department of Clinical Laboratory, The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Clinical School of the Third Medical Center of Chinese PLA General Hospital, Anhui Medical University, Hefei, 230032, China
| | - Yanhong Tai
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100166, China
| | - Yue Han
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yanhai Wang
- Department of Clinical Laboratory, Huhhot First Hospital, Huhhot, 010030, China.
| |
Collapse
|
7
|
Zheng J, Miao F, Wang Z, Ma Y, Lin Z, Chen Y, Kong X, Wang Y, Zhuang A, Wu T, Li W. Identification of MDM2 as a prognostic and immunotherapeutic biomarker in a comprehensive pan-cancer analysis: A promising target for breast cancer, bladder cancer and ovarian cancer immunotherapy. Life Sci 2023; 327:121832. [PMID: 37276911 DOI: 10.1016/j.lfs.2023.121832] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND The murine double minute 2 (MDM2) gene is a crucial factor in the development and progression of various cancer types. Multiple rigorous scientific studies have consistently shown its involvement in tumorigenesis and cancer progression in a wide range of cancer types. However, a comprehensive analysis of the role of MDM2 in human cancer has yet to be conducted. METHODS We used various databases, including TIMER2.0, TCGA, GTEx and STRING, to analyze MDM2 expression and its correlation with clinical outcomes, interacting genes and immune cell infiltration. We also investigated the association of MDM2 with immune checkpoints and performed gene enrichment analysis using DAVID tools. RESULTS The pan-cancer MDM2 analysis found that MDM2 expression and mutation status were observably different in 25 types of cancer tissue compared with healthy tissues, and prognosis analysis showed that there was a significant correlation between MDM2 expression and patient prognosis. Furthermore, correlation analysis showed that MDM2 expression was correlated with tumor mutational burden, microsatellite instability and drug sensitivity in certain cancer types. We found that there was an association between MDM2 expression and immune cell infiltration across cancer types, and MDM2 inhibitors might enhance the effect of immunotherapy on breast cancer, bladder cancer and ovarian cancer. CONCLUSIONS The first systematic pan-cancer analysis of MDM2 was conducted, and it demonstrated that MDM2 was a reliable prognostic biomarker and was closely related to cancer immunity, providing a potential immunotherapeutic target for breast cancer, bladder cancer and ovarian cancer.
Collapse
Affiliation(s)
- Jialiang Zheng
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Fenglin Miao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhao Wang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yuan Ma
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhenhang Lin
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yaqin Chen
- Nursing Department of Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Xu Kong
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yue Wang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Aobo Zhuang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ting Wu
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Wengang Li
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
8
|
Jiao Y, Li S, Gong J, Zheng K, Xie Y. Comprehensive analysis of the expression and prognosis for RAI2: A promising biomarker in breast cancer. Front Oncol 2023; 13:1134149. [PMID: 37064084 PMCID: PMC10090471 DOI: 10.3389/fonc.2023.1134149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
IntroductionRetinoic acid-induced 2 (RAI2) was initially related to cell differentiation and induced by retinoic acid. RAI2 has been identified as an emerging tumor suppressor in breast cancer and colorectal cancer.MethodsIn this study, we performed systematic analyses of RAI2 in breast cancer. Meta-analysis and Kaplan-Meier survival curves were applied to identify the survival prediction potential of RAI2. Moreover, the association between RAI2 expression and the abundance of six tumor-infiltrating immune cells was investigated by TIMER, including B cells, CD8+ T cells, CD4+ T cells, B cells, dendritic cells, neutrophils, and macrophages. The expression profiles of high and low RAI2 mRNA levels in GSE7390 were compared to identify differentially expressed genes (DEGs) and the biological function of these DEGs was analyzed by R software, which was further proved in GSE7390.ResultsOur results showed that the normal tissues had more RAI2 expression than breast cancer tissues. Patients with high RAI2 expression were related to a favorable prognosis and more immune infiltrates. A total of 209 DEGs and 182 DEGs were identified between the expression profiles of high and low RAI2 mRNA levels in the GSE7390 and GSE21653 databases, respectively. Furthermore, Gene Ontology (GO) enrichment indicated that these DEGs from two datasets were both mainly distributed in “biological processes” (BP), including “organelle fission” and “nuclear division”. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis demonstrated that these DEGs from two datasets were both significantly enriched in the “cell cycle”. Common hub genes between the DEGs in GSE7390 and GSE21653 were negatively associated with RAI2 expression, including CCNA2, MAD2L1, MELK, CDC20, and CCNB2.DiscussionsThese results above suggested that RAI2 might play a pivotal role in preventing the initiation and progression of breast cancer. The present study may contribute to understanding the molecular mechanisms of RAI2 and enriching biomarkers to predict patient prognosis in breast cancer.
Collapse
Affiliation(s)
- Ying Jiao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Shiyu Li
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Juejun Gong
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Zheng
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Ya Xie
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Ya Xie,
| |
Collapse
|
9
|
Jinteng L, Peitao X, Wenhui Y, Guiwen Y, Feng Y, Xiaojun X, Zepeng S, Jiajie L, Yunshu C, Zhaoqiang Z, Yipeng Z, Zhikun L, Pei F, Qian C, Dateng L, Zhongyu X, Yanfeng W, Huiyong S. BMAL1-TTK-H2Bub1 loop deficiency contributes to impaired BM-MSC-mediated bone formation in senile osteoporosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:568-585. [PMID: 36910712 PMCID: PMC9996134 DOI: 10.1016/j.omtn.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
During the aging process, the reduced osteogenic differentiation of bone marrow mesenchymal stem cells (BM-MSCs) results in decreased bone formation, which contributes to senile osteoporosis. Previous studies have confirmed that interrupted circadian rhythm plays an indispensable role in age-related disease. However, the mechanism underlying the impaired osteogenic differentiation of BM-MSCs during aging and its relationship with interrupted circadian rhythm remains unclear. In this study, we confirmed that the circadian rhythm was interrupted in aging mouse skeletal systems. The level of the core rhythm component BMAL1 but not that of CLOCK in the osteoblast lineage was decreased in senile osteoporotic specimens from both human and mouse. BMAL1 targeted TTK as a circadian-controlled gene to phosphorylate MDM2 and regulate H2Bub1 level, while H2Bub1 in turn regulated the expression of BMAL1. The osteogenic capacity of BM-MSCs was maintained by a positive loop formed by BMAL1-TTK-MDM2-H2Bub1. Furthermore, we demonstrated that using bone-targeting recombinant adeno-associated virus 9 (rAAV9) to enhance Bmal1 or Ttk might have a therapeutic effect on senile osteoporosis and delays bone repair in aging mice. In summary, our study indicated that targeting the BMAL1-TTK-MDM2-H2Bub1 axis via bone-targeting rAAV9 might be a promising strategy for the treatment of senile osteoporosis.
Collapse
Affiliation(s)
- Li Jinteng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Xu Peitao
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Yu Wenhui
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Ye Guiwen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Ye Feng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Xu Xiaojun
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Su Zepeng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Lin Jiajie
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Che Yunshu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Zhang Zhaoqiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Zeng Yipeng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Li Zhikun
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Feng Pei
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Cao Qian
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Li Dateng
- Department of Statistical Science, Southern Methodist University, Dallas, TX, USA
| | - Xie Zhongyu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Wu Yanfeng
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| | - Shen Huiyong
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, P.R. China
| |
Collapse
|
10
|
Wei S, Zhang J, Shi R, Yu Z, Chen X, Wang H. Identification of an integrated kinase-related prognostic gene signature associated with tumor immune microenvironment in human uterine corpus endometrial carcinoma. Front Oncol 2022; 12:944000. [PMID: 36158685 PMCID: PMC9491090 DOI: 10.3389/fonc.2022.944000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
In the worldwide, uterine corpus endometrial carcinoma (UCEC) is the sixth most common malignancy in women, and the number of women diagnosed is increasing. Kinase plays an important role in the occurrence and development of malignant tumors. However, the research about kinase in endometrial cancer is still unclear. Here, we first downloaded the gene expression data of 552 UCEC patients and 23 healthy endometrial tissues from The Cancer Genome Atlas (TCGA), obtained 538 kinase-related genes from the previous literature, and calculated 67 differentially expressed kinases. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were referenced to identify multiple important biological functions and signaling pathways related to 67 differentially expressed kinases. Using univariate Cox regression and Least absolute shrinkage and selection operator (LASSO), seven kinases (ALPK2, CAMKV, TTK, PTK6, MAST1, CIT, and FAM198B) were identified to establish a prognostic model of endometrial cancer. Then, patients were divided into high- and low-risk groups based on risk scores. Receiver operating characteristic (ROC) curves were plotted to evaluate that the model had a favorable predictive ability. Kaplan–Meier survival analysis suggested that high-risk groups experienced worse overall survival than low-risk groups. qRT-PCR and ISH assays confirmed the consistency between predicted candidate genes and real sample contents. CIBERSORT algorithm and ssGSEA were adopted to investigate the relationship between this signature and tumor immune microenvironment, and revealed that in low- and high-risk groups, the types of tumor-infiltrating immune cells and the immune cell-related functions were significantly different. In summary, a seven-gene signature risk model has been constructed, and could accurately predict the prognosis of UCEC, which may offer ideas and breakthrough points to the kinase-associated development of UCEC.
Collapse
Affiliation(s)
- Sitian Wei
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Shi
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhicheng Yu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingwei Chen
- Department of Industrial engineering, Tsinghua University, Beijing, China
| | - Hongbo Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hongbo Wang,
| |
Collapse
|
11
|
Erol A. Genotoxicity-Stimulated and CYLD-Driven Malignant Transformation. Cancer Manag Res 2022; 14:2339-2356. [PMID: 35958947 PMCID: PMC9362849 DOI: 10.2147/cmar.s373557] [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: 05/07/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Oxidative stress, which can cause DNA damage, can both activate TNF-R1 directly in the absence of TNF stimulation and phosphorylate c-Abl, thus promoting its cytoplasmic translocation. Persistent cytoplasmic localization of c-Abl has been associated with cellular transformation. c-Abl phosphorylates OTULIN at tyrosine 56, thereby disrupting its relationship with LUBAC. OTULIN-released LUBAC interacts with SPATA2 and is recruited to the TNF-R1sc, facilitating SPATA2-CYLD interaction. All these interactions are required for the activation of IKKβ to stimulate NF-κB transcriptional activity following genotoxic stress. IKKβ also induces the critical phosphorylation of CYLD at serine 568 to increase its deubiquitinating (DUB) activity required for the termination of signaling cascades. Contrary to the widespread belief that CYLD is an absolute tumor suppressor, CYLD initiates and terminates NF-κB activity by alternately using its oncoprotein and tumor suppressor activities, respectively. If IKKβ fails to achieve the DUB activity-inducing phosphorylation at serine 568, CYLD would operate in a sustained mode of oncogenic activity. The resulting dysregulated NF-κB activation and other accompanying pathologies will disrupt cellular homeostasis in favor of transformation.
Collapse
Affiliation(s)
- Adnan Erol
- Independent Researcher, Istanbul, Turkey
| |
Collapse
|
12
|
Luna-Maldonado F, Andonegui-Elguera MA, Díaz-Chávez J, Herrera LA. Mitotic and DNA Damage Response Proteins: Maintaining the Genome Stability and Working for the Common Good. Front Cell Dev Biol 2021; 9:700162. [PMID: 34966733 PMCID: PMC8710681 DOI: 10.3389/fcell.2021.700162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Cellular function is highly dependent on genomic stability, which is mainly ensured by two cellular mechanisms: the DNA damage response (DDR) and the Spindle Assembly Checkpoint (SAC). The former provides the repair of damaged DNA, and the latter ensures correct chromosome segregation. This review focuses on recently emerging data indicating that the SAC and the DDR proteins function together throughout the cell cycle, suggesting crosstalk between both checkpoints to maintain genome stability.
Collapse
Affiliation(s)
- Fernando Luna-Maldonado
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, México City, Mexico
| | - Marco A. Andonegui-Elguera
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, México City, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, México City, Mexico
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, México City, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
13
|
Zhang Z, Liang ZC, Liang XY, Zhang QH, Wang YJ, Zhang JH, De Liu S. Physarum polycephalum macroplasmodium exhibits countermeasures against TiO 2 nanoparticle toxicity: A physiological, biochemical, transcriptional, and metabolic perspective. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 279:116936. [PMID: 33773179 DOI: 10.1016/j.envpol.2021.116936] [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: 10/14/2020] [Revised: 03/02/2021] [Accepted: 03/10/2021] [Indexed: 06/12/2023]
Abstract
Concerns about the environmental and human health implications of TiO2 nanoparticles (nTiO2) are growing with their increased use in consumer and industrial products. Investigations of the underlying molecular mechanisms of nTiO2 tolerance in organisms will assist in countering nTiO2 toxicity. In this study, the countermeasures exhibited by the slime mold Physarum polycephalum macroplasmodium against nTiO2 toxicity were investigated from a physiological, transcriptional, and metabolic perspective. The results suggested that the countermeasures against nTiO2 exposure include gene-associated metabolic rearrangements in cellular pathways involved in amino acid, carbohydrate, and nucleic acid metabolism. Gene-associated nonmetabolic rearrangements involve processes such as DNA repair, DNA replication, and the cell cycle, and occur mainly when macroplasmodia are exposed to inhibitory doses of nTiO2. Interestingly, the growth of macroplasmodia and mammal cells was significantly restored by supplementation with a combination of responsive metabolites identified by metabolome analysis. Taken together, we report a novel model organism for the study of nTiO2 tolerance and provide insights into countermeasures taken by macroplasmodia in response to nTiO2 toxicity. Furthermore, we also present an approach to mitigate the effects of nTiO2 toxicity in cells by metabolic intervention.
Collapse
Affiliation(s)
- Zhi Zhang
- School of Food Science/School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China; Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Zhi Cheng Liang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Xiu Yi Liang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Qing Hai Zhang
- School of Food Science/School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Ya Jie Wang
- School of Food Science/School of Public Health/the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Jian Hua Zhang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Shi De Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
14
|
Fang Q, Chen XL, Zhang L, Li YB, Sun TZ, Yang CX, Chang JF, Yang XM, Sun F. The essential roles of Mps1 in spermatogenesis and fertility in mice. Cell Death Dis 2021; 12:531. [PMID: 34031364 PMCID: PMC8144579 DOI: 10.1038/s41419-021-03815-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022]
Abstract
Monopolar spindle 1 (MPS1), which plays a critical role in somatic mitosis, has also been revealed to be essential for meiosis I in oocytes. Spermatogenesis is an important process involving successive mitosis and meiosis, but the function of MPS1 in spermatogenesis remains unclear. Here, we generated Mps1 conditional knockout mice and found that Ddx4-cre-driven loss of Mps1 in male mice resulted in depletion of undifferentiated spermatogonial cells and subsequently of differentiated spermatogonia and spermatocytes. In addition, Stra8-cre-driven ablation of Mps1 in male mice led to germ cell loss and fertility reduction. Spermatocytes lacking Mps1 have blocked at the zygotene-to-pachytene transition in the prophase of meiosis I, which may be due to decreased H2B ubiquitination level mediated by MDM2. And the expression of many meiotic genes was decreased, while that of apoptotic genes was increased. Moreover, we also detected increased apoptosis in spermatocytes with Mps1 knockout, which may have been the reason why germ cells were lost. Taken together, our findings indicate that MPS1 is required for mitosis of gonocytes and spermatogonia, differentiation of undifferentiated spermatogonia, and progression of meiosis I in spermatocytes.
Collapse
Affiliation(s)
- Qiang Fang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xue-Lin Chen
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lei Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Ya-Bin Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Tian-Zeng Sun
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chen-Xin Yang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jian-Feng Chang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiao-Mei Yang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Feng Sun
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
15
|
Klein AM, de Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about p53. Genes Dev 2021; 35:575-601. [PMID: 33888565 PMCID: PMC8091979 DOI: 10.1101/gad.347872.120] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, Klein et al. discuss the p53-independent roles of MDM2 and MDMX. First, they review the structural and functional features of MDM2 and MDMX proteins separately and together that could be relevant to their p53-independent activities. Following this, they summarize how these two proteins are regulated and how they can function in cells that lack p53. Most well studied as proteins that restrain the p53 tumor suppressor protein, MDM2 and MDMX have rich lives outside of their relationship to p53. There is much to learn about how these two proteins are regulated and how they can function in cells that lack p53. Regulation of MDM2 and MDMX, which takes place at the level of transcription, post-transcription, and protein modification, can be very intricate and is context-dependent. Equally complex are the myriad roles that these two proteins play in cells that lack wild-type p53; while many of these independent outcomes are consistent with oncogenic transformation, in some settings their functions could also be tumor suppressive. Since numerous small molecules that affect MDM2 and MDMX have been developed for therapeutic outcomes, most if not all designed to prevent their restraint of p53, it will be essential to understand how these diverse molecules might affect the p53-independent activities of MDM2 and MDMX.
Collapse
Affiliation(s)
- Alyssa M Klein
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, New York 10032, USA
| | | | - Divya Venkatesh
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
16
|
Lahalle A, Lacroix M, De Blasio C, Cissé MY, Linares LK, Le Cam L. The p53 Pathway and Metabolism: The Tree That Hides the Forest. Cancers (Basel) 2021; 13:cancers13010133. [PMID: 33406607 PMCID: PMC7796211 DOI: 10.3390/cancers13010133] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The p53 pathway is a major tumor suppressor pathway that prevents the propagation of abnormal cells by regulating DNA repair, cell cycle progression, cell death, or senescence. The multiple cellular processes regulated by p53 were more recently extended to the control of metabolism, and many studies support the notion that perturbations of p53-associated metabolic activities are linked to cancer development. Converging lines of evidence support the notion that, in addition to p53, other key components of this molecular cascade are also important regulators of metabolism. Here, we illustrate the underestimated complexity of the metabolic network controlled by the p53 pathway and show how its perturbation contributes to human diseases including cancer, aging, and metabolic diseases. Abstract The p53 pathway is functionally inactivated in most, if not all, human cancers. The p53 protein is a central effector of numerous stress-related molecular cascades. p53 controls a safeguard mechanism that prevents accumulation of abnormal cells and their transformation by regulating DNA repair, cell cycle progression, cell death, or senescence. The multiple cellular processes regulated by p53 were more recently extended to the control of metabolism and many studies support the notion that perturbations of p53-associated metabolic activities are linked to cancer development, as well as to other pathophysiological conditions including aging, type II diabetes, and liver disease. Although much less documented than p53 metabolic activities, converging lines of evidence indicate that other key components of this tumor suppressor pathway are also involved in cellular metabolism through p53-dependent as well as p53-independent mechanisms. Thus, at least from a metabolic standpoint, the p53 pathway must be considered as a non-linear pathway, but the complex metabolic network controlled by these p53 regulators and the mechanisms by which their activities are coordinated with p53 metabolic functions remain poorly understood. In this review, we highlight some of the metabolic pathways controlled by several central components of the p53 pathway and their role in tissue homeostasis, metabolic diseases, and cancer.
Collapse
Affiliation(s)
- Airelle Lahalle
- Université de Montpellier, F-34090 Montpellier, France; (A.L.); (M.L.); (C.D.B.); (L.K.L.)
- IRCM, Institut de Recherche en Cancérologie de Montpellier, F-34298 Montpellier, France
- ICM, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1194, F-24298 Montpellier, France
- Equipe Labellisée Ligue Contre le Cancer, F-75013 Paris, France
| | - Matthieu Lacroix
- Université de Montpellier, F-34090 Montpellier, France; (A.L.); (M.L.); (C.D.B.); (L.K.L.)
- IRCM, Institut de Recherche en Cancérologie de Montpellier, F-34298 Montpellier, France
- ICM, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1194, F-24298 Montpellier, France
- Equipe Labellisée Ligue Contre le Cancer, F-75013 Paris, France
| | - Carlo De Blasio
- Université de Montpellier, F-34090 Montpellier, France; (A.L.); (M.L.); (C.D.B.); (L.K.L.)
- IRCM, Institut de Recherche en Cancérologie de Montpellier, F-34298 Montpellier, France
- ICM, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1194, F-24298 Montpellier, France
- Equipe Labellisée Ligue Contre le Cancer, F-75013 Paris, France
| | - Madi Y. Cissé
- Department of Molecular Metabolism, Harvard, T.H Chan School of Public Health, Boston, MA 02115, USA;
| | - Laetitia K. Linares
- Université de Montpellier, F-34090 Montpellier, France; (A.L.); (M.L.); (C.D.B.); (L.K.L.)
- IRCM, Institut de Recherche en Cancérologie de Montpellier, F-34298 Montpellier, France
- ICM, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1194, F-24298 Montpellier, France
| | - Laurent Le Cam
- Université de Montpellier, F-34090 Montpellier, France; (A.L.); (M.L.); (C.D.B.); (L.K.L.)
- IRCM, Institut de Recherche en Cancérologie de Montpellier, F-34298 Montpellier, France
- ICM, Institut Régional du Cancer de Montpellier, F-34298 Montpellier, France
- INSERM, Institut National de la Santé et de la Recherche Médicale, U1194, F-24298 Montpellier, France
- Equipe Labellisée Ligue Contre le Cancer, F-75013 Paris, France
- Correspondence:
| |
Collapse
|
17
|
TTK, CDC25A, and ESPL1 as Prognostic Biomarkers for Endometrial Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4625123. [PMID: 33282948 PMCID: PMC7685798 DOI: 10.1155/2020/4625123] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/19/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022]
Abstract
Objective Endometrial cancer (EC) is one of the most common malignant gynaecological tumours worldwide. This study was aimed at identifying EC prognostic genes and investigating the molecular mechanisms of these genes in EC. Methods Two mRNA datasets of EC were downloaded from the Gene Expression Omnibus (GEO). The GEO2R tool and Draw Venn Diagram were used to identify differentially expressed genes (DEGs) between normal endometrial tissues and EC tissues. Then, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). Next, the protein-protein interactions (PPIs) of these DEGs were determined by the Search Tool for the Retrieval of Interacting Genes (STRING) tool and Cytoscape with Molecular Complex Detection (MCODE). Furthermore, Kaplan-Meier survival analysis was performed by UALCAN to verify genes associated with significantly poor prognosis. Next, Gene Expression Profiling Interactive Analysis (GEPIA) was used to verify the expression levels of these selected genes. Additionally, a reanalysis of the KEGG pathways was performed to understand the potential biological functions of selected genes. Finally, the associations between these genes and clinical features were analysed based on TCGA cancer genomic datasets for EC. Results In EC tissues, compared with normal endometrial tissues, 147 of 249 DEGs were upregulated and 102 were downregulated. A total of 64 upregulated genes were assembled into a PPI network. Next, 14 genes were found to be both associated with significantly poor prognosis and highly expressed in EC tissues. Reanalysis of the KEGG pathways found that three of these genes were enriched in the cell cycle pathway. TTK, CDC25A, and ESPL1 showed higher expression in cancers with late stage and higher tumour grade. Conclusion In summary, through integrated bioinformatics approaches, we found three significant prognostic genes of EC, which might be potential therapeutic targets for EC patients.
Collapse
|
18
|
Huang M, Huang Y, Guo J, Yu L, Chang Y, Wang X, Luo J, Huang Y, Tu Z, Lu X, Xu Y, Zhang Z, Zhang Z, Ding K. Pyrido[2, 3-d]pyrimidin-7(8H)-ones as new selective orally bioavailable Threonine Tyrosine Kinase (TTK) inhibitors. Eur J Med Chem 2020; 211:113023. [PMID: 33248853 DOI: 10.1016/j.ejmech.2020.113023] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/08/2020] [Accepted: 11/10/2020] [Indexed: 01/12/2023]
Abstract
A series of pyrido [2, 3-d]pyrimidin-7(8H)-ones were designed and synthesized as new selective orally bioavailable Threonine Tyrosine Kinase (TTK) inhibitors. One of the representative compounds, 5o, exhibited strong binding affinity with a Kd value of 0.15 nM, but was significantly less potent against a panel of 402 wild-type kinases at 100 nM. The compound also potently inhibited the kinase activity of TTK with an IC50 value of 23 nM, induced chromosome missegregation and aneuploidy, and suppressed proliferation of a panel of human cancer cell lines with low μM IC50 values. Compound 5o demonstrated good oral pharmacokinetic properties with a bioavailability value of 45.3% when administered at a dose of 25 mg/kg in rats. Moreover, a combination therapy of 5o with paclitaxel displayed promising in vivo efficacy against the HCT-116 human colon cancer xenograft model in nude mice with a Tumor Growth Inhibition (TGI) value of 78%. Inhibitor 5o may provide a new research tool for further validating therapeutic potential of TTK inhibition.
Collapse
Affiliation(s)
- Minhao Huang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China; University of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | - Yongjun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Jing Guo
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Lei Yu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Yu Chang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xiaolu Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Jinfeng Luo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | - Yanhui Huang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | - Zhengchao Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Yong Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510530, China
| | - Zhimin Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China.
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China.
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China.
| |
Collapse
|
19
|
Ju JQ, Li XH, Pan MH, Xu Y, Xu Y, Sun MH, Sun SC. Mps1 controls spindle assembly, SAC, and DNA repair in the first cleavage of mouse early embryos. J Cell Biochem 2020; 122:290-300. [PMID: 33025669 DOI: 10.1002/jcb.29858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/28/2020] [Accepted: 09/14/2020] [Indexed: 11/08/2022]
Abstract
Monopolar spindle-1 (Mps1) is a critical interphase regulator that also involves into the spindle assembly checkpoint for the cell cycle control in both mitosis and meiosis. However, the functions of Mps1 during mouse early embryo development is still unclear. In this study, we reported the important roles of Mps1 in the first cleavage of mouse embryos. Our data indicated that the loss of Mps1 activity caused precocious cleavage of zygotes to 2-cell embryos; however, prolonged culture disturbed the early embryo development to the blastocyst. We found that the spindle organization was disrupted after Mps1 inhibition, and the chromosomes were misaligned in the first cleavage. Moreover, the kinetochore-microtubule attachment was lost and Aurora B failed to accumulate to the kinetochores, indicating that the spindle assembly checkpoint (SAC) was activated. Furthermore, the inhibition of Mps1 activity resulted in an increase of DNA damage, which further induced oxidative stress, showing with positive γ-H2A.X signal and increased reactive oxygen species level. Ultimately, irreparable DNA damage and oxidative stress-activated apoptosis and autophagy, which was confirmed by the positive Annexin-V signal and increased autophagosomes. Taken together, our data indicated that Mps1 played important roles in the control of SAC and DNA repair during mouse early embryo development.
Collapse
Affiliation(s)
- Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiao-Han Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yi Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yao Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ming-Hong Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
20
|
Jin N, Lera RF, Yan RE, Guo F, Oxendine K, Horner VL, Hu Y, Wan J, Mattison RJ, Weaver BA, Burkard ME. Chromosomal instability upregulates interferon in acute myeloid leukemia. Genes Chromosomes Cancer 2020; 59:627-638. [PMID: 32557940 DOI: 10.1002/gcc.22880] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 01/20/2023] Open
Abstract
Chromosome instability (CIN) generates genetic and karyotypic diversity that is common in hematological malignancies. Low to moderate levels of CIN are well tolerated and can promote cancer proliferation. However, high levels of CIN are lethal. Thus, CIN may serve both as a prognostic factor to predict clinical outcome and as a predictive biomarker. A retrospective study was performed to evaluate CIN in acute myeloid leukemia (AML). Chromosome mis-segregation frequency was correlated with clinical outcome in bone marrow core biopsy specimens from 17 AML cases. Additionally, we induced chromosome segregation errors in AML cell lines with AZ3146, an inhibitor of the Mps1 mitotic checkpoint kinase, to quantify the phenotypic effects of high CIN. We observed a broad distribution of chromosome mis-segregation frequency in AML bone marrow core specimens. High CIN correlated with complex karyotype in AML, as expected, although there was no clear survival effect. In addition to CIN, experimentally inducing chromosome segregation errors by Mps1 inhibition in AML cell lines causes DNA damage, micronuclei formation, and upregulation of interferon stimulated genes. High levels of CIN appear to be immunostimulatory, suggesting an opportunity to combine mitotic checkpoint inhibitors with immunotherapy in treatment of AML.
Collapse
Affiliation(s)
- Ning Jin
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA
| | - Robert F Lera
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Rachel E Yan
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Fen Guo
- Department of Pathology and Laboratory Medicine, Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, Wisconsin, USA
| | - Kim Oxendine
- Department of Pathology and Laboratory Medicine, Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, Wisconsin, USA
| | - Vanessa L Horner
- Department of Pathology and Laboratory Medicine, Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, Wisconsin, USA
| | - Yang Hu
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA.,Medical Scientist Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Jun Wan
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA.,Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, USA.,Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Ryan J Mattison
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA
| | - Beth A Weaver
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA.,Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, USA
| | - Mark E Burkard
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
21
|
Huang H, Yang Y, Zhang W, Liu X, Yang G. TTK regulates proliferation and apoptosis of gastric cancer cells through the Akt-mTOR pathway. FEBS Open Bio 2020; 10:1542-1549. [PMID: 32530571 PMCID: PMC7396433 DOI: 10.1002/2211-5463.12909] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/07/2020] [Accepted: 06/05/2020] [Indexed: 01/17/2023] Open
Abstract
TTK (also known as Mps1) is the core component of the spindle assembly checkpoint, which ensures proper distribution of chromosomes to daughter cells to maintain genome integrity and to balance growth and division. However, the function of TTK in tumorigenesis has not been extensively studied, especially in relation to the development of gastric cancer. In this study, survival and tumor recurrence data related to TTK expression level in gastric cancer patients were collected and analyzed. We observed that TTK expression was negatively correlated with survival and tumor recurrence in vivo. TTK was also upregulated in gastric cancer cells and was observed to be essential for the proliferation and survival of gastric cancer cells. Knockdown of TTK inhibited proliferation and increased apoptosis. Furthermore, we report that TTK regulates the proliferation and apoptosis of tumor cells through the Akt‐mTOR pathway. Knockdown of TTK inhibited activation of Akt‐mTOR signaling. In summary, our data indicate that TTK is involved in the regulation of gastric cancer proliferation and apoptosis.
Collapse
Affiliation(s)
- Hongxia Huang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yadong Yang
- Institute of Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Wenyuan Zhang
- Institute of Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Xinzhu Liu
- Institute of Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Geng Yang
- Institute of Bioengineering, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
22
|
Zhang L, Jiang B, Zhu N, Tao M, Jun Y, Chen X, Wang Q, Luo C. Mitotic checkpoint kinase Mps1/TTK predicts prognosis of colon cancer patients and regulates tumor proliferation and differentiation via PKCα/ERK1/2 and PI3K/Akt pathway. Med Oncol 2019; 37:5. [PMID: 31720873 DOI: 10.1007/s12032-019-1320-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/04/2019] [Indexed: 01/09/2023]
Abstract
Mps1/TTK plays an important role in development of many tumors. The purpose of the present study was designed to investigate the role of TTK in colon cancer. We analyzed TTK and colon cancer in the GEO database, colon cancer tissues and normal tissues were collected to verify the results by immunohistochemistry. We detected the TTK expression in the colon cancer cell lines, and overexpressed or silenced TTK expression in colon cancer cell lines. GEO database showed that the expression of TTK was higher in the colon cancer tissues than normal tissues, higher level of TTK shows unfavourable prognosis in colon patients. Furthermore, high differentiation of colon shows the lower expression of TTK. The higher expression of TTK links with the high microsatellite status. However, the expression of TTK has no significant difference among the different stages of colon cancer patients, and has no significant relationship with recurrence or relapse. Here, we also report that the differential expression of TTK in colon cancer cells alters the intrinsic negative regulation of cell proliferation and differentiation, resulting in the difference of proliferation and differentiation capacity. TTK could activate the PKCα/ERK1/2 to influence the proliferation and inactivate the PI3K/AKT pathway to inhibition the expression of MUC2 and TFF3 that related to the differentiation of colon cells. In conclusions, TTK promote the colon cancer cell proliferation via activation of PKCα/ERK1/2 and inhibit the differentiation via inactivation of PI3K/Akt pathway. TTK inhibition may be the potential therapeutic pathway for the treatment of colon cancer.
Collapse
Affiliation(s)
- Li Zhang
- Department of Central Laboratory and Huai'an Key Laboratory of Esophageal Cancer Biobank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Baofei Jiang
- Department of Gastrointestinal Surgery, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huaian, 223300, Jiangsu, China
| | - Ni Zhu
- Department of Microbiology, Hubei University of Science and Technology, Xianning, 437100, Hubei, China
| | - Mingyue Tao
- Department of Central Laboratory and Huai'an Key Laboratory of Esophageal Cancer Biobank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Yali Jun
- Department of Central Laboratory and Huai'an Key Laboratory of Esophageal Cancer Biobank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Xiaofei Chen
- Department of Central Laboratory and Huai'an Key Laboratory of Esophageal Cancer Biobank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, 223300, China.
| | - Qilong Wang
- Department of Central Laboratory and Huai'an Key Laboratory of Esophageal Cancer Biobank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, 223300, China.
| | - Chao Luo
- Department of Central Laboratory and Huai'an Key Laboratory of Esophageal Cancer Biobank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
23
|
Molecular design and anticancer activities of small-molecule monopolar spindle 1 inhibitors: A Medicinal chemistry perspective. Eur J Med Chem 2019; 175:247-268. [DOI: 10.1016/j.ejmech.2019.04.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 11/21/2022]
|
24
|
Masnadi-Shirazi M, Maurya MR, Pao G, Ke E, Verma IM, Subramaniam S. Time varying causal network reconstruction of a mouse cell cycle. BMC Bioinformatics 2019; 20:294. [PMID: 31142274 PMCID: PMC6542064 DOI: 10.1186/s12859-019-2895-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022] Open
Abstract
Background Biochemical networks are often described through static or time-averaged measurements of the component macromolecules. Temporal variation in these components plays an important role in both describing the dynamical nature of the network as well as providing insights into causal mechanisms. Few methods exist, specifically for systems with many variables, for analyzing time series data to identify distinct temporal regimes and the corresponding time-varying causal networks and mechanisms. Results In this study, we use well-constructed temporal transcriptional measurements in a mammalian cell during a cell cycle, to identify dynamical networks and mechanisms describing the cell cycle. The methods we have used and developed in part deal with Granger causality, Vector Autoregression, Estimation Stability with Cross Validation and a nonparametric change point detection algorithm that enable estimating temporally evolving directed networks that provide a comprehensive picture of the crosstalk among different molecular components. We applied our approach to RNA-seq time-course data spanning nearly two cell cycles from Mouse Embryonic Fibroblast (MEF) primary cells. The change-point detection algorithm is able to extract precise information on the duration and timing of cell cycle phases. Using Least Absolute Shrinkage and Selection Operator (LASSO) and Estimation Stability with Cross Validation (ES-CV), we were able to, without any prior biological knowledge, extract information on the phase-specific causal interaction of cell cycle genes, as well as temporal interdependencies of biological mechanisms through a complete cell cycle. Conclusions The temporal dependence of cellular components we provide in our model goes beyond what is known in the literature. Furthermore, our inference of dynamic interplay of multiple intracellular mechanisms and their temporal dependence on one another can be used to predict time-varying cellular responses, and provide insight on the design of precise experiments for modulating the regulation of the cell cycle. Electronic supplementary material The online version of this article (10.1186/s12859-019-2895-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maryam Masnadi-Shirazi
- Department of Electrical and Computer Engineering and Bioengineering, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Mano R Maurya
- Department of Bioengineering and San Diego Supercomputer center, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Gerald Pao
- Salk institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Eugene Ke
- Salk institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Inder M Verma
- Salk institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Shankar Subramaniam
- Department of Bioengineering, Departments of Computer Science and Engineering, Cellular and Molecular Medicine, and the Graduate Program in Bioinformatics, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.
| |
Collapse
|
25
|
Yang H, Zhang F, Huang CJ, Liao J, Han Y, Hao P, Chu Y, Lu X, Li W, Yu H, Kang J. Mps1 regulates spindle morphology through MCRS1 to promote chromosome alignment. Mol Biol Cell 2019; 30:1060-1068. [PMID: 30785839 PMCID: PMC6724509 DOI: 10.1091/mbc.e18-09-0546] [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] [Indexed: 01/30/2023] Open
Abstract
Accurate partitioning of chromosomes during mitosis is essential for genetic stability and requires the assembly of the dynamic mitotic spindle and proper kinetochore–microtubule attachment. The spindle assembly checkpoint (SAC) monitors the incompleteness and errors in kinetochore–microtubule attachment and delays anaphase. The SAC kinase Mps1 regulates the recruitment of downstream effectors to unattached kinetochores. Mps1 also actively promotes chromosome alignment during metaphase, but the underlying mechanism is not completely understood. Here, we show that Mps1 regulates chromosome alignment through MCRS1, a spindle assembly factor that controls the dynamics of the minus end of kinetochore microtubules. Mps1 binds and phosphorylates MCRS1. This mechanism enables KIF2A localization to the minus end of spindle microtubules. Thus, our study reveals a novel role of Mps1 in regulating the dynamics of the minus end of microtubules and expands the functions of Mps1 in genome maintenance.
Collapse
Affiliation(s)
- Hongdan Yang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Fengxia Zhang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Ching-Jung Huang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Jun Liao
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Ying Han
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Piliang Hao
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Youjun Chu
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Xiaoai Lu
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Wenshu Li
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Hongtao Yu
- Howard Hughes Medical Institute, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jungseog Kang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China.,NYU-ECNU Center for Computational Chemistry, New York University at Shanghai, Shanghai 200062, China
| |
Collapse
|
26
|
Ronis MJ, Gomez-Acevedo H, Shankar K, Sharma N, Blackburn M, Singhal R, Mercer KE, Badger TM. EB 2017 Article: Soy protein isolate feeding does not result in reproductive toxicity in the pre-pubertal rat testis. Exp Biol Med (Maywood) 2019; 243:695-707. [PMID: 29763383 DOI: 10.1177/1535370218771333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The isoflavone phytoestrogens found in the soy protein isolate used in soy infant formulas have been shown to have estrogenic actions in the developing male reproductive tract resulting in reproductive toxicity. However, few studies have examined potential estrogenicity of soy protein isolate as opposed to that of pure isoflavones. In this study, we fed weanling male Sprague-Dawley rats a semi-purified diet with casein or soy protein isolate as the sole protein source from postnatal day 21 to 33. Additional groups were fed casein or soy protein isolate and treated s.c. with 10 µg/kg/d estradiol via osmotic minipump. Estradiol treatment reduced testis, prostate weights, and serum androgen concentrations ( P < 0.05). Soy protein isolate had no effect. Estradiol up-regulated 489 and down-regulated 1237 testicular genes >1.5-fold ( P < 0.05). In contrast, soy protein isolate only significantly up-regulated expression of 162 genes and down-regulated 16 genes. The top 30 soy protein isolate-up-regulated genes shared 93% concordance with estradiol up-regulated genes. There was little overlap between soy protein isolate down-regulated genes and those down-regulated by estradiol treatment. Functional annotation analysis revealed significant differences in testicular biological processes affected by estradiol or soy protein isolate. Estradiol had major actions on genes involved in reproductive processes including down-regulation of testicular steroid synthesis and expression of steroid receptor activated receptor (Star) and cytochrome P450 17α-hydroxylase/(Cyp17a1). In contrast, soy protein isolate primarily affected pathways associated with macromolecule modifications including ubiquitination and histone methylation. Our results indicate that rather than acting as a weak estrogen in the developing testis, soy protein isolate appears to act as a selective estrogen receptor modulator with little effect on reproductive processes. Impact statement Soy protein isolate (SPI) is the sole protein used to make soy-based infant formulas. SPI contains phytoestrogens, which are structurally similar to estradiol. These phytoestrogens, daidzein, genistein, and equol, fit the definition of endocrine-disrupting compounds, and at high concentrations, have estrogenic actions resulting in reproductive toxicity in the developing male, when provided as isolated chemicals. However, few animal studies have examined the potential estrogenicity of SPI as opposed to pure isoflavones. In this study, SPI feeding did not elicit an estrogenic response in the testis nor any adverse outcomes including reduced testicular growth, or androgen production during early development in rats when compared to those receiving estradiol. These findings are consistent with emerging data showing no differences in reproductive development in males and female children that received breast milk, cow's milk formula, or soy infant formula during the postnatal feeding period.
Collapse
Affiliation(s)
- Martin Jj Ronis
- 1 Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center - New Orleans, LA 70112, USA
| | - Horacio Gomez-Acevedo
- 2 Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kartik Shankar
- 3 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | - Neha Sharma
- 4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | | | - Rohit Singhal
- 4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | - Kelly E Mercer
- 3 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | - Thomas M Badger
- 3 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| |
Collapse
|
27
|
Zhu D, Xu S, Deyanat-Yazdi G, Peng SX, Barnes LA, Narla RK, Tran T, Mikolon D, Ning Y, Shi T, Jiang N, Raymon HK, Riggs JR, Boylan JF. Synthetic Lethal Strategy Identifies a Potent and Selective TTK and CLK1/2 Inhibitor for Treatment of Triple-Negative Breast Cancer with a Compromised G 1-S Checkpoint. Mol Cancer Ther 2018; 17:1727-1738. [PMID: 29866747 DOI: 10.1158/1535-7163.mct-17-1084] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/13/2018] [Accepted: 05/08/2018] [Indexed: 11/16/2022]
Abstract
Historically, phenotypic-based drug discovery has yielded a high percentage of novel drugs while uncovering new tumor biology. CC-671 was discovered using a phenotypic screen for compounds that preferentially induced apoptosis in triple-negative breast cancer cell lines while sparing luminal breast cancer cell lines. Detailed in vitro kinase profiling shows CC-671 potently and selectively inhibits two kinases-TTK and CLK2. Cellular mechanism of action studies demonstrate that CC-671 potently inhibits the phosphorylation of KNL1 and SRp75, direct TTK and CLK2 substrates, respectively. Furthermore, CC-671 causes mitotic acceleration and modification of pre-mRNA splicing leading to apoptosis, consistent with cellular TTK and CLK inhibition. Correlative analysis of genomic and potency data against a large panel of breast cancer cell lines identifies breast cancer cells with a dysfunctional G1-S checkpoint as more sensitive to CC-671, suggesting synthetic lethality between G1-S checkpoint and TTK/CLK2 inhibition. Furthermore, significant in vivo CC-671 efficacy was demonstrated in two cell line-derived and one patient tumor-derived xenograft models of triple-negative breast cancer (TNBC) following weekly dosing. These findings are the first to demonstrate the unique inhibitory combination activity of a dual TTK/CLK2 inhibitor that preferably kills TNBC cells and shows synthetic lethality with a compromised G1-S checkpoint in breast cancer cell lines. On the basis of these data, CC-671 was moved forward for clinical development as a potent and selective TTK/CLK2 inhibitor in a subset of patients with TNBC. Mol Cancer Ther; 17(8); 1727-38. ©2018 AACR.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Oncology Research, Celgene Corporation, San Diego, California.
| | - Shuichan Xu
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | | | - Sophie X Peng
- Department of Pharmacology, Celgene Corporation, San Diego, California
| | - Leo A Barnes
- Department of Pharmacology, Celgene Corporation, San Diego, California
| | | | - Tam Tran
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | - David Mikolon
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | - Yuhong Ning
- Informatics and Knowledge Utilization Department, Celgene Corporation, San Diego, California
| | - Tao Shi
- Informatics and Knowledge Utilization Department, Celgene Corporation, San Diego, California
| | - Ning Jiang
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | - Heather K Raymon
- Department of Pharmacology, Celgene Corporation, San Diego, California
| | - Jennifer R Riggs
- Department of Chemistry, Celgene Corporation, San Diego, California
| | - John F Boylan
- Department of Oncology Research, Celgene Corporation, San Diego, California
| |
Collapse
|
28
|
Galal SA, Khairat SHM, Ali HI, Shouman SA, Attia YM, Ali MM, Mahmoud AE, Abdel-Halim AH, Fyiad AA, Tabll A, El-Shenawy R, El Abd YS, Ramdan R, El Diwani HI. Part II: New candidates of pyrazole-benzimidazole conjugates as checkpoint kinase 2 (Chk2) inhibitors. Eur J Med Chem 2018; 144:859-873. [PMID: 29316526 DOI: 10.1016/j.ejmech.2017.12.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/12/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023]
Abstract
The development of checkpoint kinase 2 (Chk2) inhibitors for the treatment of cancer has been an ongoing and attractive objective in drug discovery. In this study, twenty-one feasible pyrazole-benzimidazole conjugates were synthesized to study their effect against Chk2 activity using Checkpoint Kinase Assay. The antitumor activity of these compounds was investigated using SRB assay. A potentiation effect of the synthesized Chk2 inhibitors was also investigated using the genotoxic anticancer drugs cisplatin and doxorubicin on breast carcinoma, (ER+) cell line (MCF-7). In vivo Chk2 and antitumor activities of 8d as a single-agent, and in combination with doxorubicin, were evaluated in breast cancer bearing animals induced by N-methylnitrosourea. The effect of 8d alone and in combination with doxorubicin was also studied on cell-cycle phases of MCF-7 cells using flow cytometry analysis. The results revealed their potencies as Chk2 inhibitors with IC50 ranges from 9.95 to 65.07 nM. Generally the effect of cisplatin or doxorubicin was potentiated by the effect of most of the compounds that were studied. The in vivo results indicated that the combination of 8d and doxorubicin inhibited checkpoint kinase activity more than either doxorubicin or 8d alone. There was a positive correlation between checkpoint kinase inhibition and the improvement observed in histopathological features. Single dose treatment with doxorubicin or 8d produced S phase cell cycle arrest whereas their combination created cell cycle arrest at G2/M from 8% in case of doxorubicin to 51% in combination. Gold molecular modelling studies displayed a high correlation to the biological results.
Collapse
Affiliation(s)
- Shadia A Galal
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt.
| | - Sarah H M Khairat
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| | - Hamed I Ali
- Department of Pharmaceutical Sciences, Texas A&M University Irma Lerma Rangel College of Pharmacy Kingsville, TX 78363, United States; Department of Pharmaceutical Chemistry, College of Pharmacy, Helwan University, Cairo, Egypt
| | - Samia A Shouman
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| | - Yasmin M Attia
- Department of Cancer Biology, National Cancer Institute, Cairo University, Egypt
| | - Mamdouh M Ali
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Abeer E Mahmoud
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Abeer H Abdel-Halim
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Amal A Fyiad
- Department of Biochemistry, Division of Genetic Engineering and Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Ashraf Tabll
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Reem El-Shenawy
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Yasmine S El Abd
- Department of Microbial Biotechnology, Division of Genetic Engineering & Biotechnology, National Research Centre, 12622, Cairo, Egypt
| | - Raghda Ramdan
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium; Department of Basic Medical Sciences, Physiology Group, Ghent University, Ghent, Belgium
| | - Hoda I El Diwani
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, 12622, Egypt
| |
Collapse
|
29
|
Chen F, Szymanski EP, Olivier KN, Liu X, Tettelin H, Holland SM, Duggal P. Whole-Exome Sequencing Identifies the 6q12-q16 Linkage Region and a Candidate Gene, TTK, for Pulmonary Nontuberculous Mycobacterial Disease. Am J Respir Crit Care Med 2017; 196:1599-1604. [PMID: 28777004 DOI: 10.1164/rccm.201612-2479oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Pulmonary nontuberculous mycobacterial disease (PNTM) often affects white postmenopausal women, with a tall and lean body habitus and higher rates of scoliosis, pectus excavatum, mitral valve prolapse, and mutations in the CFTR gene. These clinical features and the familial clustering of the disease suggest an underlying genetic mechanism. OBJECTIVES To map the genes associated with PNTM, whole-exome sequencing was conducted in 12 PNTM families and 57 sporadic cases recruited at the National Institutes of Health Clinical Center during 2001-2013. METHODS We performed a variant-level and a gene-level parametric linkage analysis on nine PNTM families (16 affected and 20 unaffected) as well as a gene-level association analysis on nine PNTM families and 55 sporadic cases. MEASUREMENTS AND MAIN RESULTS The genome-wide variant-level linkage analysis using 4,328 independent common variants identified a 20-cM region on chromosome 6q12-6q16 (heterogeneity logarithm of odds score = 3.9), under a recessive disease model with 100% penetrance and a risk allele frequency of 5%. All genes on chromosome 6 were then tested in the gene-level linkage analysis, using the collapsed haplotype pattern method. The TTK protein kinase gene (TTK) on chromosome 6q14.1 was the most significant (heterogeneity logarithm of odds score = 3.38). In addition, the genes MAP2K4, RCOR3, KRT83, IFNLR1, and SLC29A1 were associated with PNTM in our gene-level association analysis. CONCLUSIONS The TTK gene encodes a protein kinase that is essential for mitotic checkpoints and the DNA damage response. TTK and other genetic loci identified in our study may contribute to the increased susceptibility to NTM infection and its progression to pulmonary disease.
Collapse
Affiliation(s)
- Fei Chen
- 1 Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Eva P Szymanski
- 2 Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, Maryland
| | - Kenneth N Olivier
- 3 Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland; and
| | - Xinyue Liu
- 4 Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hervé Tettelin
- 4 Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Steven M Holland
- 2 Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, Maryland
| | - Priya Duggal
- 1 Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
30
|
Riggs JR, Nagy M, Elsner J, Erdman P, Cashion D, Robinson D, Harris R, Huang D, Tehrani L, Deyanat-Yazdi G, Narla RK, Peng X, Tran T, Barnes L, Miller T, Katz J, Tang Y, Chen M, Moghaddam MF, Bahmanyar S, Pagarigan B, Delker S, LeBrun L, Chamberlain PP, Calabrese A, Canan SS, Leftheris K, Zhu D, Boylan JF. The Discovery of a Dual TTK Protein Kinase/CDC2-Like Kinase (CLK2) Inhibitor for the Treatment of Triple Negative Breast Cancer Initiated from a Phenotypic Screen. J Med Chem 2017; 60:8989-9002. [PMID: 28991472 DOI: 10.1021/acs.jmedchem.7b01223] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Triple negative breast cancer (TNBC) remains a serious unmet medical need with discouragingly high relapse rates. We report here the synthesis and structure-activity relationship (SAR) of a novel series of 2,4,5-trisubstituted-7H-pyrrolo[2,3-d]pyrimidines with potent activity against TNBC tumor cell lines. These compounds were discovered from a TNBC phenotypic screen and possess a unique dual inhibition profile targeting TTK (mitotic exit) and CLK2 (mRNA splicing). Design and optimization, driven with a TNBC tumor cell assay, identified potent and selective compounds with favorable in vitro and in vivo activity profiles and good iv PK properties. This cell-based driven SAR produced compounds with strong single agent in vivo efficacy in multiple TNBC xenograft models without significant body weight loss. These data supported the nomination of CC-671 into IND-enabling studies as a single agent TNBC therapy.
Collapse
Affiliation(s)
- Jennifer R Riggs
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Mark Nagy
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Jan Elsner
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Paul Erdman
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Dan Cashion
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Dale Robinson
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Roy Harris
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Dehua Huang
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Lida Tehrani
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Gordafaried Deyanat-Yazdi
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Rama Krishna Narla
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Xiaohui Peng
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Tam Tran
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Leo Barnes
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Terra Miller
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Jason Katz
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Yang Tang
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Ming Chen
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Mehran F Moghaddam
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Sogole Bahmanyar
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Barbra Pagarigan
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Silvia Delker
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Laurie LeBrun
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Philip P Chamberlain
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Andrew Calabrese
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Stacie S Canan
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Katerina Leftheris
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - Dan Zhu
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| | - John F Boylan
- Celgene Corporation , 10300 Campus Pointe Drive, Suite 100, San Diego, California 92121, United States
| |
Collapse
|
31
|
Funk LC, Zasadil LM, Weaver BA. Living in CIN: Mitotic Infidelity and Its Consequences for Tumor Promotion and Suppression. Dev Cell 2017; 39:638-652. [PMID: 27997823 DOI: 10.1016/j.devcel.2016.10.023] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Errors in chromosome segregation during mitosis have been recognized as a hallmark of tumor cells since the late 1800s, resulting in the long-standing hypothesis that mitotic abnormalities drive tumorigenesis. Recent work has shown that mitotic defects can promote tumors, suppress them, or do neither, depending on the rate of chromosome missegregation. Here we discuss the causes of chromosome missegregation, their effects on tumor initiation and progression, and the evidence that increasing the rate of chromosome missegregation may be an effective chemotherapeutic strategy.
Collapse
Affiliation(s)
- Laura C Funk
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lauren M Zasadil
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Beth A Weaver
- Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Avenue, 6109 WIMR I, Madison, WI 53705-2275, USA.
| |
Collapse
|
32
|
Riscal R, Schrepfer E, Arena G, Cissé MY, Bellvert F, Heuillet M, Rambow F, Bonneil E, Sabourdy F, Vincent C, Ait-Arsa I, Levade T, Thibaut P, Marine JC, Portais JC, Sarry JE, Le Cam L, Linares LK. Chromatin-Bound MDM2 Regulates Serine Metabolism and Redox Homeostasis Independently of p53. Mol Cell 2016; 62:890-902. [PMID: 27264869 DOI: 10.1016/j.molcel.2016.04.033] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 03/07/2016] [Accepted: 04/27/2016] [Indexed: 12/20/2022]
Abstract
The mouse double minute 2 (MDM2) oncoprotein is recognized as a major negative regulator of the p53 tumor suppressor, but growing evidence indicates that its oncogenic activities extend beyond p53. Here, we show that MDM2 is recruited to chromatin independently of p53 to regulate a transcriptional program implicated in amino acid metabolism and redox homeostasis. Identification of MDM2 target genes at the whole-genome level highlights an important role for ATF3/4 transcription factors in tethering MDM2 to chromatin. MDM2 recruitment to chromatin is a tightly regulated process that occurs during oxidative stress and serine/glycine deprivation and is modulated by the pyruvate kinase M2 (PKM2) metabolic enzyme. Depletion of endogenous MDM2 in p53-deficient cells impairs serine/glycine metabolism, the NAD(+)/NADH ratio, and glutathione (GSH) recycling, impacting their redox state and tumorigenic potential. Collectively, our data illustrate a previously unsuspected function of chromatin-bound MDM2 in cancer cell metabolism.
Collapse
Affiliation(s)
- Romain Riscal
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France
| | - Emilie Schrepfer
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France
| | - Giuseppe Arena
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France
| | - Madi Y Cissé
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France
| | - Floriant Bellvert
- INSA, UPS, INP, Université de Toulouse, 135 Avenue de Rangueil, 31 077 Toulouse, France; INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Maud Heuillet
- INSA, UPS, INP, Université de Toulouse, 135 Avenue de Rangueil, 31 077 Toulouse, France; INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Florian Rambow
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, 3000 Leuven, Belgium; Laboratory for Molecular Cancer Biology, Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Eric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128 Station Centre-Ville, Montreal, QC H3C 3J7, Canada
| | - Frédérique Sabourdy
- Laboratoire de Biochimie Métabolique, IFB, CHU Purpan, 31059 Toulouse, France; INSERM UMR 1037, CRCT, Université Paul Sabatier Toulouse-III, 31062 Toulouse, France
| | - Charles Vincent
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France
| | - Imade Ait-Arsa
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, IFB, CHU Purpan, 31059 Toulouse, France; INSERM UMR 1037, CRCT, Université Paul Sabatier Toulouse-III, 31062 Toulouse, France
| | - Pierre Thibaut
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128 Station Centre-Ville, Montreal, QC H3C 3J7, Canada; Department of Chemistry, Université de Montréal, P.O. Box 6128 Station Centre-Ville, Montreal, QC H3C 3J7, Canada
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, 3000 Leuven, Belgium; Laboratory for Molecular Cancer Biology, Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Jean-Charles Portais
- INSA, UPS, INP, Université de Toulouse, 135 Avenue de Rangueil, 31 077 Toulouse, France; INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Jean-Emmanuel Sarry
- INSERM UMR 1037, CRCT, Université Paul Sabatier Toulouse-III, 31062 Toulouse, France
| | - Laurent Le Cam
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France.
| | - Laetitia K Linares
- IRCM, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier, France; INSERM, U1194, 34298 Montpellier, France; Université de Montpellier, 34298 Montpellier, France; Institut Régional du Cancer Montpellier, 34298 Montpellier, France.
| |
Collapse
|
33
|
Tandle A, Camphausen K. Mitotic Protein Kinase 1: Role in Spindle Assembly Checkpoint Revisited. JOURNAL OF CANCER CLINICAL TRIALS 2016; 1:111. [PMID: 27885363 PMCID: PMC5119518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Anita Tandle
- Corresponding author: Anita Tandle, Radiation Oncology Branch, National Cancer Institute Building 10, Room B3B100, Bethesda, MD 20892, USA, Tel: 301-443-4402; Fax: 301-480-1434;
| | | |
Collapse
|