1
|
Eckardt NA, Avin-Wittenberg T, Bassham DC, Chen P, Chen Q, Fang J, Genschik P, Ghifari AS, Guercio AM, Gibbs DJ, Heese M, Jarvis RP, Michaeli S, Murcha MW, Mursalimov S, Noir S, Palayam M, Peixoto B, Rodriguez PL, Schaller A, Schnittger A, Serino G, Shabek N, Stintzi A, Theodoulou FL, Üstün S, van Wijk KJ, Wei N, Xie Q, Yu F, Zhang H. The lowdown on breakdown: Open questions in plant proteolysis. THE PLANT CELL 2024; 36:2931-2975. [PMID: 38980154 PMCID: PMC11371169 DOI: 10.1093/plcell/koae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/16/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
Proteolysis, including post-translational proteolytic processing as well as protein degradation and amino acid recycling, is an essential component of the growth and development of living organisms. In this article, experts in plant proteolysis pose and discuss compelling open questions in their areas of research. Topics covered include the role of proteolysis in the cell cycle, DNA damage response, mitochondrial function, the generation of N-terminal signals (degrons) that mark many proteins for degradation (N-terminal acetylation, the Arg/N-degron pathway, and the chloroplast N-degron pathway), developmental and metabolic signaling (photomorphogenesis, abscisic acid and strigolactone signaling, sugar metabolism, and postharvest regulation), plant responses to environmental signals (endoplasmic-reticulum-associated degradation, chloroplast-associated degradation, drought tolerance, and the growth-defense trade-off), and the functional diversification of peptidases. We hope these thought-provoking discussions help to stimulate further research.
Collapse
Affiliation(s)
| | - Tamar Avin-Wittenberg
- Department of Plant and Environmental Sciences, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Diane C Bassham
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Poyu Chen
- School of Biological Science and Technology, College of Science and Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Qian Chen
- Ministry of Agriculture and Rural Affairs Key Laboratory for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Jun Fang
- Section of Molecular Plant Biology, Department of Biology, University of Oxford, Oxford OX1 3RB, UK
| | - Pascal Genschik
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 12, rue du Général Zimmer, Strasbourg 67084, France
| | - Abi S Ghifari
- School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Angelica M Guercio
- Department of Plant Biology, College of Biological Sciences, University of California-Davis, Davis, CA 95616, USA
| | - Daniel J Gibbs
- School of Biosciences, University of Birmingham, Edgbaston B1 2RU, UK
| | - Maren Heese
- Department of Developmental Biology, University of Hamburg, Ohnhorststr. 18, Hamburg 22609, Germany
| | - R Paul Jarvis
- Section of Molecular Plant Biology, Department of Biology, University of Oxford, Oxford OX1 3RB, UK
| | - Simon Michaeli
- Department of Postharvest Sciences, Agricultural Research Organization (ARO), Volcani Institute, Rishon LeZion 7505101, Israel
| | - Monika W Murcha
- School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Sergey Mursalimov
- Department of Postharvest Sciences, Agricultural Research Organization (ARO), Volcani Institute, Rishon LeZion 7505101, Israel
| | - Sandra Noir
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 12, rue du Général Zimmer, Strasbourg 67084, France
| | - Malathy Palayam
- Department of Plant Biology, College of Biological Sciences, University of California-Davis, Davis, CA 95616, USA
| | - Bruno Peixoto
- Section of Molecular Plant Biology, Department of Biology, University of Oxford, Oxford OX1 3RB, UK
| | - Pedro L Rodriguez
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), Consejo Superior de Investigaciones Cientificas-Universidad Politecnica de Valencia, Valencia ES-46022, Spain
| | - Andreas Schaller
- Department of Plant Physiology and Biochemistry, Institute of Biology, University of Hohenheim, Stuttgart 70599, Germany
| | - Arp Schnittger
- Department of Developmental Biology, University of Hamburg, Ohnhorststr. 18, Hamburg 22609, Germany
| | - Giovanna Serino
- Department of Biology and Biotechnology, Sapienza Universita’ di Roma, p.le A. Moro 5, Rome 00185, Italy
| | - Nitzan Shabek
- Department of Plant Biology, College of Biological Sciences, University of California-Davis, Davis, CA 95616, USA
| | - Annick Stintzi
- Department of Plant Physiology and Biochemistry, Institute of Biology, University of Hohenheim, Stuttgart 70599, Germany
| | | | - Suayib Üstün
- Faculty of Biology and Biotechnology, Ruhr-University of Bochum, Bochum 44780, Germany
| | - Klaas J van Wijk
- Section of Plant Biology, School of Integrative Plant Sciences (SIPS), Cornell University, Ithaca, NY 14853, USA
| | - Ning Wei
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Qi Xie
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, the Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feifei Yu
- College of Grassland Science and Technology, China Agricultural University, Beijing 100083, China
| | - Hongtao Zhang
- Plant Sciences and the Bioeconomy, Rothamsted Research, Harpenden AL5 2JQ, UK
| |
Collapse
|
2
|
Xia X, Liu X, Xu Q, Gu J, Ling S, Liu Y, Li R, Zou M, Jiang S, Gao Z, Chen C, Liu S, Liu N. USP14 deficiency inhibits neointima formation following vascular injury via degradation of Skp2 protein. Cell Death Discov 2024; 10:295. [PMID: 38909015 PMCID: PMC11193710 DOI: 10.1038/s41420-024-02069-1] [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: 03/21/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024] Open
Abstract
Ubiquitin-proteasome system (UPS) is involved in vascular smooth muscle cell (VSMC) proliferation. Deubiquitinating enzymes (DUBs) have an essential role in the UPS-regulated stability of the substrate; however, the function of DUBs in intimal hyperplasia remains unclear. We screened DUBs to identify a protein responsible for regulating VSMC proliferation and identified USP14 protein that mediates cancer development, inflammation, and foam cell formation. USP14 promotes human aortic smooth muscle cell and A7r5 cell growth in vitro, and its inhibition or deficiency decreases the intimal area in the mice carotid artery ligation model. In addition, USP14 stabilizes Skp2 expression by decreasing its degradation, while Skp2 overexpression rescues USP14 loss-induced issues. The current findings suggested an essential role of USP14 in the pathology of vascular remodeling, deeming it a promising target for arterial restenosis therapy.
Collapse
Affiliation(s)
- Xiaohong Xia
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiaolin Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Qiong Xu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Jielei Gu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Sisi Ling
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Yajing Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Rongxue Li
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Min Zou
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Siqin Jiang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhiwei Gao
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Canshan Chen
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Shiming Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China.
| | - Ningning Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
3
|
Zheng L, Shen J, Chen Y, Lin J, Li P, Zhao X, Ren H, Sun Y, Wang Z. FBXO43 promotes cell cycle progression in cancer cells through stabilizing SKP2. Cancer Lett 2024; 591:216848. [PMID: 38604312 DOI: 10.1016/j.canlet.2024.216848] [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: 02/14/2024] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024]
Abstract
FBXO43 is a member of the FBXO subfamily of F-box proteins, known to be a regulatory hub during meiosis. A body of data showed that FBXO43 is overexpressed in a number of human cancers. However, whether and how FBXO43 affects cell cycle progression and growth of cancer cells remain elusive. In this study, we provide first piece of evidence, showing a pivotal role of FBXO43 in cell cycle progression and growth of cancer cells. Specifically, FBXO43 acts as a positive cell cycle regulator with an oncogenic activity in variety types of human cancer, including non-small cell lung cancer, hepatocellular carcinoma and sarcoma. Mechanistically, FBXO43 interacts with phosphorylated SKP2 induced by AKT1, leading to reduced SKP2 auto-ubiquitylation and subsequent proteasome degradation. Taken together, our study demonstrates that FBXO43 promotes cell cycle progression by stabilizing SKP2, and FBXO43 could serve as a potential anti-cancer target.
Collapse
Affiliation(s)
- Liyun Zheng
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiajia Shen
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Chen
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Lin
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Pengyu Li
- Qilu Hospital of Shan Dong University, Jinan, Shandong Province, China
| | - Xiaoli Zhao
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hangjiang Ren
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Sun
- Cancer Institute of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Hangzhou, China; Research Center for Life Science and Human Health, Beijing Institute of Zhejiang University, Hangzhou, China.
| | - Zhen Wang
- Department of Biochemistry, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Vasavan B, Das N, Kahnamouei P, Trombley C, Swan A. Skp2-Cyclin A Interaction Is Necessary for Mitotic Entry and Maintenance of Diploidy. J Mol Biol 2024; 436:168505. [PMID: 38423454 DOI: 10.1016/j.jmb.2024.168505] [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: 10/01/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Skp2, the substrate recognition component of the SCFSkp2 ubiquitin ligase, has been implicated in the targeted destruction of a number of key cell cycle regulators and the promotion of S-phase. One of its critical targets is the Cyclin dependent kinase (Cdk) inhibitor p27, and indeed the overexpression of Skp2 in a number of cancers is directly correlated with the premature degradation of p27. Skp2 was first identified as a protein that interacts with Cyclin A in transformed cells, but its role in this complex has remained unclear. In this paper, we demonstrate that Skp2 interacts with Cyclin A in Drosophila and is required to maintain Cyclin A levels and permit mitotic entry. Failure of mitotic entry in Skp2 mutant cells results in polyploidy. If these cells enter mitosis again they are unable to properly segregate their chromosomes, leading to checkpoint dependent cell cycle arrest or apoptosis. Thus, Skp2 is required for mitosis and for maintaining diploidy and genome stability.
Collapse
Affiliation(s)
- Biju Vasavan
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 2P1, Canada
| | - Nilanjana Das
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 2P1, Canada
| | - Paria Kahnamouei
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 2P1, Canada
| | - Chantelle Trombley
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 2P1, Canada
| | - Andrew Swan
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 2P1, Canada.
| |
Collapse
|
5
|
Feng Z, Yin J, Zhang Z, Chen Z, Huang L, Tang N, Wang K. O-GlcNAcylation of E3 ubiquitin ligase SKP2 promotes hepatocellular carcinoma proliferation. Oncogene 2024; 43:1149-1159. [PMID: 38396292 DOI: 10.1038/s41388-024-02977-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024]
Abstract
O-linked-β-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation) and ubiquitination are critical posttranslational modifications that regulate tumor development and progression. The continuous progression of the cell cycle is the fundamental cause of tumor proliferation. S-phase kinase-associated protein 2 (SKP2), an important E3 ubiquitin ligase, assumes a pivotal function in the regulation of the cell cycle. However, it is still unclear whether SKP2 is an effector of O-GlcNAcylation that affects tumor progression. In this study, we found that SKP2 interacted with O-GlcNAc transferase (OGT) and was highly O-GlcNAcylated in hepatocellular carcinoma (HCC). Mechanistically, the O-GlcNAcylation at Ser34 stabilized SKP2 by reducing its ubiquitination and degradation mediated by APC-CDH1. Moreover, the O-GlcNAcylation of SKP2 enhanced its binding ability with SKP1, thereby enhancing its ubiquitin ligase function. Consequently, SKP2 facilitated the transition from the G1-S phase of the cell cycle by promoting the ubiquitin degradation of cell cycle-dependent kinase inhibitors p27 and p21. Additionally, targeting the O-GlcNAcylation of SKP2 significantly suppressed the proliferation of HCC. Altogether, our findings reveal that O-GlcNAcylation, a novel posttranslational modification of SKP2, plays a crucial role in promoting HCC proliferation, and targeting the O-GlcNAcylation of SKP2 may become a new therapeutic strategy to impede the progression of HCC.
Collapse
Affiliation(s)
- Zhongqi Feng
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Jiaxin Yin
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Zhirong Zhang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Zhen Chen
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Luyi Huang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| | - Ni Tang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| | - Kai Wang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
6
|
Foy R, Lew KX, Saurin AT. The search for CDK4/6 inhibitor biomarkers has been hampered by inappropriate proliferation assays. NPJ Breast Cancer 2024; 10:19. [PMID: 38438376 PMCID: PMC10912267 DOI: 10.1038/s41523-024-00624-8] [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: 04/19/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
CDK4/6 inhibitors are effective at treating advanced HR+ /HER2- breast cancer, however biomarkers that can predict response are urgently needed. We demonstrate here that previous large-scale screens designed to identify which tumour types or genotypes are most sensitive to CDK4/6 inhibitors have misrepresented the responsive cell lines because of a reliance on metabolic proliferation assays. CDK4/6-inhibited cells arrest in G1 but continue to grow in size, thereby producing more mitochondria. We show that this growth obscures the arrest using ATP-based proliferation assays but not if DNA-based assays are used instead. Furthermore, lymphoma lines, previously identified as the most sensitive, simply appear to respond the best using ATP-based assays because they fail to overgrow during the G1 arrest. Similarly, the CDK4/6 inhibitor abemaciclib appears to inhibit proliferation better than palbociclib because it also restricts cellular overgrowth through off-target effects. DepMap analysis of screening data using reliable assay types, demonstrates that palbociclib-sensitive cell types are also sensitive to Cyclin D1, CDK4 and CDK6 knockout/knockdown, whereas the palbociclib-resistant lines are sensitive to Cyclin E1, CDK2 and SKP2 knockout/knockdown. Potential biomarkers of palbociclib-sensitive cells are increased expression of CCND1 and RB1, and reduced expression of CCNE1 and CDKN2A. Probing DepMap with similar data from metabolic assays fails to reveal these associations. Together, this demonstrates why CDK4/6 inhibitors, and any other anti-cancer drugs that arrest the cell cycle but permit continued cell growth, must now be re-screened against a wide-range of cell types using an appropriate proliferation assay. This would help to better inform clinical trials and to identify much needed biomarkers of response.
Collapse
Affiliation(s)
- Reece Foy
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Kah Xin Lew
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian T Saurin
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
7
|
Feng T, Wang P, Zhang X. Skp2: A critical molecule for ubiquitination and its role in cancer. Life Sci 2024; 338:122409. [PMID: 38184273 DOI: 10.1016/j.lfs.2023.122409] [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: 10/09/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
The ubiquitin-proteasome system (UPS) is a multi-step process that serves as the primary pathway for protein degradation within cells. UPS activity also plays a crucial role in regulating various life processes, including the cell cycle, signal transduction, DNA repair, and others. The F-box protein Skp2, a crucial member of the UPS, plays a central role in the development of various diseases. Skp2 controls cancer cell growth and drug resistance by ubiquitinating modifications to a variety of proteins. This review emphasizes the multifaceted role of Skp2 in a wide range of cancers and the mechanisms involved, highlighting the potential of Skp2 as a therapeutic target in cancer. Additionally, we describe the impactful influence exerted by Skp2 in various other diseases beyond cancer.
Collapse
Affiliation(s)
- Tianyang Feng
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China
| | - Ping Wang
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China
| | - Xiling Zhang
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China.
| |
Collapse
|
8
|
Zhang C, Pan G, Qin JJ. Role of F-box proteins in human upper gastrointestinal tumors. Biochim Biophys Acta Rev Cancer 2024; 1879:189035. [PMID: 38049014 DOI: 10.1016/j.bbcan.2023.189035] [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: 08/23/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Protein ubiquitination and degradation is an essential physiological process in almost all organisms. As the key participants in this process, the E3 ubiquitin ligases have been widely studied and recognized. F-box proteins, a crucial component of E3 ubiquitin ligases that regulates diverse biological functions, including cell differentiation, proliferation, migration, and apoptosis by facilitating the degradation of substrate proteins. Currently, there is an increasing focus on studying the role of F-box proteins in cancer. In this review, we present a comprehensive overview of the significant contributions of F-box proteins to the development of upper gastrointestinal tumors, highlighting their dual roles as both carcinogens and tumor suppressors. We delve into the molecular mechanisms underlying the involvement of F-box proteins in upper gastrointestinal tumors, exploring their interactions with specific substrates and their cross-talks with other key signaling pathways. Furthermore, we discuss the implications of F-box proteins in radiotherapy resistance in the upper gastrointestinal tract, emphasizing their potential as clinical therapeutic and prognostic targets. Overall, this review provides an up-to-date understanding of the intricate involvement of F-box proteins in human upper gastrointestinal tumors, offering valuable insights for the identification of prognostic markers and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Che Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guangzhao Pan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jiang-Jiang Qin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
9
|
Sun X, He L, Liu H, Thorne RF, Zeng T, Liu L, Zhang B, He M, Huang Y, Li M, Gao E, Ma M, Cheng C, Meng F, Lang C, Li H, Xiong W, Pan S, Ren D, Dang B, Yang Y, Wu M, Liu L. The diapause-like colorectal cancer cells induced by SMC4 attenuation are characterized by low proliferation and chemotherapy insensitivity. Cell Metab 2023; 35:1563-1579.e8. [PMID: 37543034 DOI: 10.1016/j.cmet.2023.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/12/2023] [Accepted: 07/12/2023] [Indexed: 08/07/2023]
Abstract
In response to adverse environmental conditions, embryonic development may reversibly cease, a process termed diapause. Recent reports connect this phenomenon with the non-genetic responses of tumors to chemotherapy, but the mechanisms involved are poorly understood. Here, we establish a multifarious role for SMC4 in the switching of colorectal cancer cells to a diapause-like state. SMC4 attenuation promotes the expression of three investment phase glycolysis enzymes increasing lactate production while also suppressing PGAM1. Resultant high lactate levels increase ABC transporter expression via histone lactylation, rendering tumor cells insensitive to chemotherapy. SMC4 acts as co-activator of PGAM1 transcription, and the coordinate loss of SMC4 and PGAM1 affects F-actin assembly, inducing cytokinesis failure and polyploidy, thereby inhibiting cell proliferation. These insights into the mechanisms underlying non-genetic chemotherapy resistance may have significant implications for the field, advancing our understanding of aerobic glycolysis functions in tumor and potentially informing future therapeutic strategies.
Collapse
Affiliation(s)
- Xuedan Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Lifang He
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China; Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230001 Anhui, China
| | - Hong Liu
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
| | - Rick Francis Thorne
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou, 450003 Henan, China
| | - Taofei Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Liu Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Bo Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Miao He
- Anhui Huaheng Biotechnology Co., Ltd., Hefei, 230001 Anhui, China
| | - Yabin Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Mingyue Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Enyi Gao
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan, China
| | - Mengyao Ma
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou, 450003 Henan, China
| | - Cheng Cheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Fanzheng Meng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Chuandong Lang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Hairui Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Wanxiang Xiong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Shixiang Pan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Delong Ren
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
| | - Bingyi Dang
- Henan Wild Animals Rescue Center, Henan Forestry Administration, Zhengzhou, 450040 Henan, China
| | - Yi Yang
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China
| | - Mian Wu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou, 450003 Henan, China.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, 230001 Anhui, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, 230001 Anhui, China.
| |
Collapse
|
10
|
Goodarzi V, Nouri S, Nassaj ZS, Bighash M, Abbasian S, Hagh RA. Long non coding RNAs reveal important pathways in childhood asthma: a future perspective. J Mol Histol 2023; 54:257-269. [PMID: 37537509 DOI: 10.1007/s10735-023-10131-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/04/2023] [Indexed: 08/05/2023]
Abstract
Asthma is a long-term inflammatory disease of the airways of the lungs refers changes that occur in conjunction with, or as a result of, chronic airway inflammation. Airway remodeling the subsequent of inflammation constitutes cellular and extracellular matrix changes in the wall airways, epithelial-to-mesenchymal-transition and airway smooth muscle cell proliferation. Diseases often begin in childhood and despite extensive research, causative pathogenic mechanisms still remain unclear. Transcriptome analysis of childhood asthma reveals distinct gene expression profiles of Long noncoding RNAs which have been reported to play a central regulatory role in various aspects of pathogenesis, clinical course and treatment of asthma. We briefly review current understanding of lnc-RNA dysregulation in children with asthma, focusing on their complex role in the inflammation, cell proliferation and remodeling of airway to guide future researches. We found that the lnc-RNAs increases activity of several oncogenes such c-Myc, Akt, and ERK and various signaling pathways such as MAPK (PI3K, Ras, JNK and p38), NF-κB and Wnt and crosstalk between these pathways by TGFβ, β-catenin, ERK and SKP2. Moreover, two different signal transduction pathways, Wnt and Notch1, can be activated by two lnc-RNAs through sponging the same miRNA for exacerbation cell proliferation.
Collapse
Affiliation(s)
- Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Shadi Nouri
- Arak University of Medical Sciences, Arak, Iran
| | - Zohre Saleh Nassaj
- Center for Health Related Social and Behavioral Sciences Research, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mansoureh Bighash
- Bachelor of Nursing, School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvn, Iran
| | - Sadegh Abbasian
- Department of Laboratory Science, School of Paramedical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | | |
Collapse
|
11
|
Rowland RJ, Heath R, Maskell D, Thompson RF, Ranson NA, Blaza JN, Endicott JA, Noble MEM, Salamina M. Cryo-EM structure of SKP1-SKP2-CKS1 in complex with CDK2-cyclin A-p27KIP1. Sci Rep 2023; 13:10718. [PMID: 37400515 PMCID: PMC10318019 DOI: 10.1038/s41598-023-37609-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/24/2023] [Indexed: 07/05/2023] Open
Abstract
p27KIP1 (cyclin-dependent kinase inhibitor 1B, p27) is a member of the CIP/KIP family of CDK (cyclin dependent kinase) regulators that inhibit cell cycle CDKs. p27 phosphorylation by CDK1/2, signals its recruitment to the SCFSKP2 (S-phase kinase associated protein 1 (SKP1)-cullin-SKP2) E3 ubiquitin ligase complex for proteasomal degradation. The nature of p27 binding to SKP2 and CKS1 was revealed by the SKP1-SKP2-CKS1-p27 phosphopeptide crystal structure. Subsequently, a model for the hexameric CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex was proposed by overlaying an independently determined CDK2-cyclin A-p27 structure. Here we describe the experimentally determined structure of the isolated CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex at 3.4 Å global resolution using cryogenic electron microscopy. This structure supports previous analysis in which p27 was found to be structurally dynamic, transitioning from disordered to nascent secondary structure on target binding. We employed 3D variability analysis to further explore the conformational space of the hexameric complex and uncovered a previously unidentified hinge motion centred on CKS1. This flexibility gives rise to open and closed conformations of the hexameric complex that we propose may contribute to p27 regulation by facilitating recognition with SCFSKP2. This 3D variability analysis further informed particle subtraction and local refinement approaches to enhance the local resolution of the complex.
Collapse
Affiliation(s)
- Rhianna J Rowland
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Richard Heath
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Daniel Maskell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Rebecca F Thompson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
- Life Sciences Electron Microscopy, Thermo Fisher Scientific, Leeds, UK
| | - Neil A Ranson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - James N Blaza
- Department of Chemistry, York Structural Biology Laboratory and York Biomedical Research Institute, University of York, Heslington, YO10 5DD, York, UK
| | - Jane A Endicott
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Martin E M Noble
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Marco Salamina
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
- Evotec (UK) Ltd., Milton, Abingdon, OX14 4RZ, UK.
| |
Collapse
|
12
|
Halperin R, Arnon L, Nasirov S, Friedensohn L, Gershinsky M, Telerman A, Friedman E, Bernstein-Molho R, Tirosh A. Germline CDKN1B variant type and site are associated with phenotype in MEN4. Endocr Relat Cancer 2023; 30:ERC-22-0174. [PMID: 36256846 DOI: 10.1530/erc-22-0174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Multiple endocrine neoplasia 4 (MEN4) is a rare multiglandular endocrine neoplasia syndrome clinically hallmarked by primary hyperparathyroidism (PHPT), pituitary adenoma (PitAd), and neuroendocrine tumors (NET), clinically overlapping MEN1. The underlying mutated gene - CDKN1B, encodes for the cell-cycle regulator p27. Possible genotype-phenotype correlations in MEN4 have not been thoroughly assessed. Prompted by the findings in three Israeli MEN4 kindreds, we performed a literature review on published and unpublished data from previously reported MEN4/CDKN1B cases. Univariate analysis analyzed time-dependent risks for developing PHPT, PitAd, or NET by variant type and position along the gene. Overall, 74 MEN4 cases were analyzed. PHPT risk was 53.4% by age 60 years (mean age at diagnosis age 50.6 ± 13.9 years), risk for PitAd was 23.2% and risk for NET was 16.2% (34.4 ± 21.4 and 52.9 ± 13.9 years, respectively). The frameshift variant p.Q107fs was the most common variant identified (4/41 (9.7%) kindreds). Patients with indels had higher risk for PHPT vs point mutations (log-rank, P = 0.029). Variants in codons 94-96 were associated with higher risk for PHPT (P < 0.001) and PitAd (P = 0.031). To conclude, MEN4 is clinically distinct from MEN1, with lower risk and older age for PHPT diagnosis. We report recurrent CDKN1B frameshift variants and possible genotype-phenotype correlations.
Collapse
Affiliation(s)
- Reut Halperin
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Liat Arnon
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Sapir Nasirov
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Limor Friedensohn
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Gershinsky
- Department of Endocrinology and Diabetes, Lady Davis Carmel Medical Center and Linn Medical Center and Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alona Telerman
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Eitan Friedman
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
- Personalized Preventive Genetics Center, Assuta Medical Center, Tel-Aviv, Israel
| | - Rinat Bernstein-Molho
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
- The Susanne Levy Gertner Oncogenetics Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Amit Tirosh
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
13
|
Song C, Zhang J, Liu X, Li M, Wang D, Kang Z, Yu J, Chen J, Pan H, Wang H, Li G, Huang H. PTEN loss promotes Warburg effect and prostate cancer cell growth by inducing FBP1 degradation. Front Oncol 2022; 12:911466. [PMID: 36237339 PMCID: PMC9552847 DOI: 10.3389/fonc.2022.911466] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Rationale Fructose-1,6-bisphosphatase (FBP1) is a tumor suppressor and a key enzyme negatively regulating Warburg effect in cancer. However, regulation of FBP1 protein expression and its exact role in prostate cancer (PCa) is largely unclear. Phosphatase and tensin homolog (PTEN) is one of the most frequently deleted tumor suppressor genes in human PCa. However, the role of PTEN loss in aberrant Warburg effect in cancer remains poorly understood. Methods Expression of PTEN and FBP1 was analyzed in several PCa cell lines and prostate tumor tissues in mice. Western blot (WB) and RT-PCR approaches were used to examine how PTEN regulates FBP1 expression. Co-immunoprecipitation (co-IP) and in vivo ubiquitination assays were used to define the regulatory mechanisms. A PCa xenograft model was employed to determine the impact of PTEN regulation of FBP1 on PCa growth in vivo. Result We demonstrated that in a manner dependent of PI3K/AKT signal pathway PTEN regulated FBP1 expression in various PCa cell lines and tumors in mice. We confirmed that this regulation took place at the protein level and was mediated by SKP2 E3 ubiquitin ligase. Mechanistically, we showed that serine 271 phosphorylation of FBP1 by cyclin-dependent kinases (CDKs) was essential for SKP2-mediated degradation of FBP1 protein induced by PTEN loss. Most importantly, we further showed that loss of PTEN expression enhanced Warburg effect and PCa growth in mice in a manner dependent, at least partially on FBP1 protein degradation. Conclusions Our results reveal a novel tumor-suppressive feature of PTEN in restraining FBP1 degradation and the Warburg effect. These results also suggest that prohibiting FBP1 protein degradation could be a viable therapeutic strategy for PTEN-deficient PCa.
Collapse
Affiliation(s)
- Changze Song
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Jianong Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Xiao Liu
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Meilu Li
- Department of Dermatology, The Second Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Dejie Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Zhijian Kang
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Jiaao Yu
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Jiuwei Chen
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Hongxin Pan
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Urological Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Honglei Wang
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Guangbin Li
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
- Department of Urology, Mayo Clinic College of Medicine, Rochester, MN, United States
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic College of Medicine, Rochester, MN, United States
- *Correspondence: Haojie Huang,
| |
Collapse
|
14
|
Sajid A, Saeed MS, Malik RM, Fazal S, Malik S, Kamal MA. Prediction of Secondary and Tertiary Structure and Docking of Rb1WT
And Rb1R661W Proteins. CURRENT BIOTECHNOLOGY 2022. [DOI: 10.2174/2211550111666220127100203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background:
Retinoblastoma, a malignancy occurring in the juvenile cells of the retina,
is responsible for light detection. It is one of the most emerging ra re childhood and infant cancer.
It is initiated by the mutation in Rb1, a first tumor suppressor gene located on chromosome 13q14.
Rb1 protein is responsible for cell cycle regulation.
Methods:
In our study, secondary and 3D-Structural predictions of Rb1WT and Rb1R661W were made
by comparative or homology modeling to find any structural change leading to the disruption in its
further interactions. Quality assurance of the structures was done by Ramachandran Plot for a stable
structure. Both the proteins were then applied by docking process with proteins of interest.
Results:
Secondary structure showed a number of mutations in helixes, β-Hairpins of Rb1R661W. The
major change was the loss of β-Hairpin loop, extension and shortening of helixes. 3D comparison
structure showed a change in the groove of Rb1R661W. Docking results, unlike RB1 WT, had different
and no interactions with some of the proteins of interest. This mutation in Rb1 protein had a deleterious
effect on the protein functionality.
Conclusion:
This study will help to design the appropriate therapy and also understand the mechanism
of disease of retinoblastoma, for researchers and pharmaceuticals.
Collapse
Affiliation(s)
- Aimen Sajid
- Capital University of Science and Technology, Islamabad, Pakistan
| | | | - Rabbiah Manzoor Malik
- Capital University of Science and Technology, Islamabad, Pakistan
- Wah Medical College, Wah Cantt, Pakistan
| | - Sahar Fazal
- Capital University of Science and Technology, Islamabad, Pakistan
| | - Shaukat Malik
- Capital University of Science and Technology, Islamabad, Pakistan
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- King Fahd Medical Research
Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee
Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| |
Collapse
|
15
|
Valizadeh M, Alimohammadi F, Azarm A, Pourtaghi Z, Derakhshan barjoei MM, Sabri H, Jafari A, Arabpour Z, Razavi P, Mokhtari M, Deravi N. Uses of soybean isoflavonoids in dentistry: A literature review. J Dent Sci 2021. [DOI: 10.1016/j.jds.2021.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
16
|
Kamptner AZM, Mayer CE, Sutterlüty H. Sprouty3, but Not Sprouty1, Expression Is Beneficial for the Malignant Potential of Osteosarcoma Cells. Int J Mol Sci 2021; 22:ijms222111944. [PMID: 34769378 PMCID: PMC8585105 DOI: 10.3390/ijms222111944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/21/2021] [Accepted: 10/31/2021] [Indexed: 11/16/2022] Open
Abstract
Sprouty proteins are widely accepted modulators of receptor tyrosine kinase-associated pathways and fulfill diversified roles in cancerogenesis dependent on the originating cells. In this study we detected a high expression of Sprouty3 in osteosarcoma-derived cells and addressed the question of whether Sprouty3 and Sprouty1 influence the malignant phenotype of this bone tumor entity. By using adenoviruses, the Sprouty proteins were expressed in two different cell lines and their influence on cellular behavior was assessed. Growth curve analyses and Scratch assays revealed that Sprouty3 accelerates cell proliferation and migration. Additionally, more colonies were grown in Soft agar if the cells express Sprouty3. In parallel, Sprouty1 had no significant effect on the measured endpoints of the study in osteosarcoma-derived cells. The promotion of the tumorigenic capacities in the presence of Sprouty3 coincided with an increased activation of signaling as measured by evaluating the phosphorylation of extracellular signal-regulated kinases (ERKs). Ectopic expression of a mutated Sprouty3 protein, in which the tyrosine necessary for its activation was substituted, resulted in inhibited migration of the treated cells. Our findings identify Sprouty3 as a candidate for a tumor promoter in osteosarcoma.
Collapse
|
17
|
Zhou H, Sun W, Zou J. Analysis of expression profiles and prognostic value of COP9 signalosome subunits for patients with head and neck squamous cell carcinoma. Oncol Lett 2021; 22:803. [PMID: 34630710 PMCID: PMC8477071 DOI: 10.3892/ol.2021.13064] [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: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has been associated with poor prognosis, due to its strong invasive ability and resistance to chemotherapy. Thus, there is an urgent requirement to identify effective biomarkers for the early diagnosis and prognostic evaluation of HNSCC. COP9 signalosome (COPS) regulates numerous cancer-associated biological processes in various malignancies. The aim of the present study was to investigate the association between COPS and HNSCC. The mRNA expression profiles of COPS in HNSCC were analyzed using UALCAN, Oncomine and UCSC Xena databases. The association between overall survival time in patients with HNSCC and the COPS genes was investigated using the Kaplan-Meier plotter database. The CERES score was obtained and evaluated to determine the importance of the COPS genes for survival of the HNSCC cell lines. Functional analysis for Gene Ontology and Gene Set Enrichment Analysis (GSEA) was performed using The Database for Annotation, Visualization and Integrated Discovery and GSEA software, respectively. After knocking down COPS5 and COPS6, cell Counting Kit-8 and wound healing assays were used to detect cell growth and migration of the CAL27 and SCC25 cell lines, respectively. Among the 10 COPS genes examined, most COPS subunits were upregulated in HNSCC samples compared with that in normal tissues, except for COPS9. Increased mRNA expression level of COPS5, COPS6, COPS7B, COPS8 and COPS9 was associated with TNM stage in patients with HNSCC. High mRNA expression level of COPS2, COPS5, COPS6, COPS7A, COPS7B, COPS8 and COPS9 had prognostic significance of patients with HNSCC. Knockdown of COPS5 and COPS6 inhibited cell growth and migration of the CAL27 and SCC25 cell lines. The results from the present study suggested that COPS subunits could be potential biomarkers in patients with HNSCC. COPS5 and COPS6 were important for cell survival and migration of the HNSCC cells.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Oral and Maxillofacial Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Wei Sun
- Department of Oral and Maxillofacial Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Jiaruan Zou
- Department of Oral and Maxillofacial Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
18
|
Gentric N, Genschik P, Noir S. Connections between the Cell Cycle and the DNA Damage Response in Plants. Int J Mol Sci 2021; 22:ijms22179558. [PMID: 34502465 PMCID: PMC8431409 DOI: 10.3390/ijms22179558] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/02/2022] Open
Abstract
Due to their sessile lifestyle, plants are especially exposed to various stresses, including genotoxic stress, which results in altered genome integrity. Upon the detection of DNA damage, distinct cellular responses lead to cell cycle arrest and the induction of DNA repair mechanisms. Interestingly, it has been shown that some cell cycle regulators are not only required for meristem activity and plant development but are also key to cope with the occurrence of DNA lesions. In this review, we first summarize some important regulatory steps of the plant cell cycle and present a brief overview of the DNA damage response (DDR) mechanisms. Then, the role played by some cell cycle regulators at the interface between the cell cycle and DNA damage responses is discussed more specifically.
Collapse
|
19
|
Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: a multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. J Hematol Oncol 2021; 14:121. [PMID: 34372899 PMCID: PMC8351444 DOI: 10.1186/s13045-021-01111-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022] Open
Abstract
MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and hematopoiesis. Due to the multi-functionality of MYC, its expression is regulated at multiple levels. Deregulation of this oncogene can give rise to a variety of cancers. In this review, MYC regulation and the mechanisms by which MYC adjusts cellular functions and its implication in hematologic malignancies are summarized. Further, we also discuss potential inhibitors of MYC that could be beneficial for treating hematologic malignancies.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression. Int J Mol Sci 2021; 22:ijms22115754. [PMID: 34072267 PMCID: PMC8198665 DOI: 10.3390/ijms22115754] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023] Open
Abstract
The cell cycle is a collection of events by which cellular components such as genetic materials and cytoplasmic components are accurately divided into two daughter cells. The cell cycle transition is primarily driven by the activation of cyclin-dependent kinases (CDKs), which activities are regulated by the ubiquitin-mediated proteolysis of key regulators such as cyclins, CDK inhibitors (CKIs), other kinases and phosphatases. Thus, the ubiquitin-proteasome system (UPS) plays a pivotal role in the regulation of the cell cycle progression via recognition, interaction, and ubiquitination or deubiquitination of key proteins. The illegitimate degradation of tumor suppressor or abnormally high accumulation of oncoproteins often results in deregulation of cell proliferation, genomic instability, and cancer occurrence. In this review, we demonstrate the diversity and complexity of the regulation of UPS machinery of the cell cycle. A profound understanding of the ubiquitination machinery will provide new insights into the regulation of the cell cycle transition, cancer treatment, and the development of anti-cancer drugs.
Collapse
|
21
|
Kelso S, Orlicky S, Beenstock J, Ceccarelli DF, Kurinov I, Gish G, Sicheri F. Bipartite binding of the N terminus of Skp2 to cyclin A. Structure 2021; 29:975-988.e5. [PMID: 33989513 DOI: 10.1016/j.str.2021.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/06/2021] [Accepted: 04/23/2021] [Indexed: 10/21/2022]
Abstract
Skp2 and cyclin A are cell-cycle regulators that control the activity of CDK2. Cyclin A acts as an activator and substrate recruitment factor of CDK2, while Skp2 mediates the ubiquitination and subsequent destruction of the CDK inhibitor protein p27. The N terminus of Skp2 can interact directly with cyclin A but is not required for p27 ubiquitination. To gain insight into this poorly understood interaction, we have solved the 3.2 Å X-ray crystal structure of the N terminus of Skp2 bound to cyclin A. The structure reveals a bipartite mode of interaction with two motifs in Skp2 recognizing two discrete surfaces on cyclin A. The uncovered binding mechanism allows for a rationalization of the inhibitory effect of Skp2 on CDK2-cyclin A kinase activity toward the RxL motif containing substrates and raises the possibility that other intermolecular regulators and substrates may use similar non-canonical modes of interaction for cyclin targeting.
Collapse
Affiliation(s)
- Susan Kelso
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, ON M5S 1A8, Canada
| | - Stephen Orlicky
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Jonah Beenstock
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Derek F Ceccarelli
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Igor Kurinov
- Department of Chemistry and Chemical Biology, Cornell University, NE-CAT, Argonne, IL 60439, USA
| | - Gerald Gish
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Frank Sicheri
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, ON M5S 1A8, Canada; Department of Biochemistry, University of Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
22
|
Hosoya Y, Ohkanda J. Intrinsically Disordered Proteins as Regulators of Transient Biological Processes and as Untapped Drug Targets. Molecules 2021; 26:2118. [PMID: 33917117 PMCID: PMC8067799 DOI: 10.3390/molecules26082118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) are critical players in the dynamic control of diverse cellular processes, and provide potential new drug targets because their dysregulation is closely related to many diseases. This review focuses on several medicinal studies that have identified low-molecular-weight inhibitors of IDPs. In addition, clinically relevant liquid-liquid phase separations-which critically involve both intermolecular interactions between IDPs and their posttranslational modification-are analyzed to understand the potential of IDPs as new drug targets.
Collapse
Affiliation(s)
| | - Junko Ohkanda
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano 399-4598, Japan;
| |
Collapse
|
23
|
A Sprouty4 Mutation Identified in Kallmann Syndrome Increases the Inhibitory Potency of the Protein towards FGF and Connected Processes. Int J Mol Sci 2021; 22:ijms22042145. [PMID: 33670044 PMCID: PMC7926442 DOI: 10.3390/ijms22042145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/03/2022] Open
Abstract
Kallmann syndrome is the result of innate genetic defects in the fibroblast growth factor (FGF) regulated signaling network causing diminished signal transduction. One of the rare mutations associated with the syndrome alters the Sprouty (Spry)4 protein by converting the serine at position 241 into a tyrosine. In this study, we characterize the tyrosine Spry4 mutant protein in the primary human embryonic lung fibroblasts WI-38 and osteosarcoma-derived cell line U2OS. As demonstrated in a cell signaling assay, Spry4 gains the capability of inhibiting FGF, but not epithelial growth factor (EGF)-induced signaling as a consequence of the tyrosine substitution. Additionally, migration of normal embryonic lung fibroblasts and osteosarcoma-derived cells is potently inhibited by the tyrosine Spry4 variant, while an effect of the wildtype Spry4 protein is hardly measureable. Concerning cell proliferation, the unaltered Spry4 protein is ineffective to influence the WI-38 cells, while the mutated Spry4 protein decelerates the cell doubling. In summary, these data emphasize that like the other mutations associated with Kallmann syndrome the described Spry4 mutation creates a hyperactive version of a selective inhibitory molecule and can thereby contribute to a weakened FGF signaling. Additionally, the study pinpoints a Spry4 variation expanding the applicability of Spry4 in a potential cancer therapy.
Collapse
|
24
|
Pan T, Qin Q, Nong C, Gao S, Wang L, Cai B, Zhang M, Wu C, Chen H, Li T, Xiong D, Li G, Wang S, Yan S. A novel WEE1 pathway for replication stress responses. NATURE PLANTS 2021; 7:209-218. [PMID: 33574575 DOI: 10.1038/s41477-021-00855-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/12/2021] [Indexed: 06/12/2023]
Abstract
DNA replication stress poses a severe threat to genome stability and is a hallmark of cancer as well as a target for cancer therapy. It is well known that the evolutionarily conserved protein kinase WEE1 regulates replication stress responses by directly phosphorylating and inhibiting the major cell cycle driver CDKs in many organisms. Here, we report a novel WEE1 pathway. We found that Arabidopsis WEE1 directly interacts with and phosphorylates the E3 ubiquitin ligase FBL17 that promotes the degradation of CDK inhibitors. The phosphorylated FBL17 is further polyubiquitinated and degraded, thereby leading to the accumulation of CDK inhibitors and the inhibition of CDKs. In strong support for this model, either loss of function of FBL17 or overexpression of CDK inhibitors suppresses the hypersensitivity of the wee1 mutant to replication stress. Intriguingly, human WEE1 also phosphorylates and destabilizes the FBL17 equivalent protein SKP2, indicating that this is a conserved mechanism. This study reveals that the WEE1-FBL17/SKP2-CKIs-CDKs axis is a molecular framework for replication stress responses, which may have clinical implications because the WEE1 inhibitor AZD1775 is currently in phase II clinical trial as an anticancer drug.
Collapse
Affiliation(s)
- Ting Pan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qi Qin
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Chubing Nong
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Shan Gao
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lili Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Bingcheng Cai
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ming Zhang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Chong Wu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Hanchen Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Tong Li
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Dan Xiong
- College of Informatics, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Guoliang Li
- College of Informatics, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Shui Wang
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Shunping Yan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.
| |
Collapse
|
25
|
Zhang W, Yu F, Yan C, Shao C, Gu P, Fu Y, Sun H, Fan X. PTEN Inhibition Accelerates Corneal Endothelial Wound Healing through Increased Endothelial Cell Division and Migration. Invest Ophthalmol Vis Sci 2021; 61:19. [PMID: 32667999 PMCID: PMC7425707 DOI: 10.1167/iovs.61.8.19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose To investigate the role of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) in the regulation of corneal endothelial cell (CECs) focusing on proliferation and migration, and to further evaluate the application of PTEN inhibitors in the treatment of corneal endothelial dysfunction in a rat model. Methods Expression of PTEN in human and rat corneal endothelium was determined by immunocytochemistry, western blotting, and ELISA. A small molecular inhibitor of PTEN, bpV(pic), was applied in the culture of human CEC cell line B4G12 and organ-cultured rat cornea in the presence of transforming growth factor beta 2 (TGF-β2). Cell cycle status was detected by flow cytometry and BrdU staining. Subcellular localization for endogenous p27Kip1 was detected by immunocytochemistry and western blotting. Moreover, exogenous transfected YFP-p27Kip1 was observed under a fluorescent microscope. Cell migration was examined with a wound scratch model and transwell invasion assay. Finally, bpV(pic) was intracamerally injected in a rat corneal endothelial injury model. The wound healing process was evaluated by slit lamp biomicroscopy, optical coherence tomography, histological and scanning electron microscope examination. Results The expression of PTEN in human corneal endothelium was higher compared with rat, which we speculate was mostly responsible for the relatively less proliferation capacity of human CEC than rat. PTEN inhibition by bpV(pic) could reverse TGF-β2-induced CEC G1-arrest by alleviating p27Kip1 nuclear accumulation and decreasing total p27Kip1 expression. In addition, bpV(pic) promoted CEC migration, which acted synergistically with TGF-β2. Finally, intracameral injection of bpV(pic) could promote corneal endothelial wound healing in a rat model. Conclusions Our study provided experimental basis for the development of therapeutic agent targeting on PTEN for the treatment of corneal endothelial dysfunction.
Collapse
|
26
|
Li M, Rui Y, Peng W, Hu J, Jiang A, Yang Z, Huang L. FIGNL1 promotes non‑small cell lung cancer cell proliferation. Int J Oncol 2021; 58:83-99. [PMID: 33367932 PMCID: PMC7721085 DOI: 10.3892/ijo.2020.5154] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/31/2020] [Indexed: 12/03/2022] Open
Abstract
Lung cancer is the most frequently diagnosed cancer and the leading cause of cancer‑associated mortality worldwide. In the present study, a novel molecular therapeutic target for lung cancer was investigated. The protein expression level of fidgetin‑like 1 (FIGNL1) in human lung cancer tissues was determined and its potential functions in the H1299 and A549 lung cancer cell lines was subsequently studied. In addition, the protein expression level of FIGNL1 in 109 lung cancer samples and corresponding para‑cancerous tissues was investigated, using immunohistochemical staining. RNA interference and overexpression of FIGNL1 was used to determine the role of FIGNL1 in regulating cell proliferation, and cDNA microarray analysis was performed to identify the potential regulatory pathways. Lastly, the potential role of FIGNL1 in regulating tumorigenesis in lungs and also the proliferation of lung cancer cells was investigated. Firstly, lung cancer tissues were found to express higher protein levels of FIGNL1 and was significantly associated with decreased cell proliferation, migration and invasion abilities, and enhanced cell death. Overexpression of FIGNL1 significantly promoted cell proliferation, including decreased arrest at the G1 phase of the cell cycle and apoptosis, as well as increased ability for fission and migration. These in vitro findings were consistent with the results of the cell‑line derived xenografts in BALB/c nude mice, where tumor growth was decreased when injected with cells transfected with shFIGNL1. Collectively, these results provide suggest that FIGNL1 is involved in cell growth and tumorigenesis.
Collapse
Affiliation(s)
- Miao Li
- Department of Respiratory Medicine, Anhui Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021
- Department of General Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000
| | - Yan Rui
- Department of Respiration and Critical Care Medicine, Anhui Provincial Key Laboratory of Clinical Basic Research on Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004
| | - Wenjia Peng
- Department of Epidemiology and Health Statistics, Bengbu Medical College, Bengbu, Anhui 233030
| | - Junfeng Hu
- Department of Respiration and Critical Care Medicine, Anhui Provincial Key Laboratory of Clinical Basic Research on Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004
| | - Anbang Jiang
- Department of Respiration and Critical Care Medicine, Anhui Provincial Key Laboratory of Clinical Basic Research on Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004
| | - Zeyu Yang
- Department of Respiration and Critical Care Medicine, Anhui Provincial Key Laboratory of Clinical Basic Research on Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004
| | - Linian Huang
- Department of Respiration and Critical Care Medicine, Anhui Provincial Key Laboratory of Clinical Basic Research on Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004
- Department of Respiratory Medicine, Anhui Provincial Hospital, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
27
|
Yamashita M, Owa T, Shiraishi R, Adachi T, Ichijo K, Taya S, Miyashita S, Hoshino M. The role of SCF Skp2 and SCF β-TrCP1/2 in the cerebellar granule cell precursors. Genes Cells 2020; 25:796-810. [PMID: 33020978 DOI: 10.1111/gtc.12813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/01/2022]
Abstract
A proper balance between proliferation and differentiation of cerebellar granule cell precursors (GCPs) is required for appropriate cerebellar morphogenesis. The Skp1-Cullin1-F-box (SCF) complex, an E3 ubiquitin ligase complex, is involved in polyubiquitination and subsequent degradation of various cell cycle regulators and transcription factors. However, it remains unknown how the SCF complex affects proliferation and differentiation of GCPs. In this study, we found that the scaffold protein Cullin1, and F-box proteins Skp2, β-TrCP1 and β-TrCP2 are expressed in the external granule layer (EGL). Knockdown of these molecules in the EGL showed that Cullin1, Skp2 and β-TrCP2 enhanced differentiation of GCPs. We also observed accumulation of cyclin-dependent kinase inhibitor p27 in GCPs when treated with a Cullin1 inhibitor or proteasome inhibitor. Furthermore, knockdown of p27 rescued enhancement of differentiation by Cullin1 knockdown. These results suggest that the SCF complex is involved in the maintenance of the proliferative state of GCPs through p27 degradation. In addition, inhibition of Cullin1 activity also prevented cell proliferation and enhanced accumulation of p27 in Daoy cells, a cell line derived from the sonic hedgehog subtype of medulloblastoma. This suggested that excess degradation of p27 through the SCF complex causes overproliferation of medulloblastoma cells.
Collapse
Affiliation(s)
- Mariko Yamashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Tomoo Owa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Ryo Shiraishi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of NCNP Brain Function and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, TMDU, Tokyo, Japan
| | - Toma Adachi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Life Science and Medical Bioscience, Graduate School of Advance Science and Engineering, TWIns, Waseda University, Tokyo, Japan
| | - Kentaro Ichijo
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichiro Taya
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Satoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
28
|
Razavipour SF, Harikumar KB, Slingerland JM. p27 as a Transcriptional Regulator: New Roles in Development and Cancer. Cancer Res 2020; 80:3451-3458. [PMID: 32341036 DOI: 10.1158/0008-5472.can-19-3663] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/25/2020] [Accepted: 04/21/2020] [Indexed: 11/16/2022]
Abstract
p27 binds and inhibits cyclin-CDK to arrest the cell cycle. p27 also regulates other processes including cell migration and development independent of its cyclin-dependent kinase (CDK) inhibitory action. p27 is an atypical tumor suppressor-deletion or mutational inactivation of the gene encoding p27, CDKN1B, is rare in human cancers. p27 is rarely fully lost in cancers because it can play both tumor suppressive and oncogenic roles. Until recently, the paradigm was that oncogenic deregulation results from either loss of growth restraint due to excess p27 proteolysis or from an oncogenic gain of function through PI3K-mediated C-terminal p27 phosphorylation, which disrupts the cytoskeleton to increase cell motility and metastasis. In cancers, C-terminal phosphorylation alters p27 protein-protein interactions and shifts p27 from CDK inhibitor to oncogene. Recent data indicate p27 regulates transcription and acts as a transcriptional coregulator of cJun. C-terminal p27 phosphorylation increases p27-cJun recruitment to and action on target genes to drive oncogenic pathways and repress differentiation programs. This review focuses on noncanonical, CDK-independent functions of p27 in migration, invasion, development, and gene expression, with emphasis on how transcriptional regulation by p27 illuminates its actions in cancer. A better understanding of how p27-associated transcriptional complexes are regulated might identify new therapeutic targets at the interface between differentiation and growth control.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Razavipour
- Breast Cancer Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Joyce M Slingerland
- Breast Cancer Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC.
| |
Collapse
|
29
|
Cai Z, Moten A, Peng D, Hsu CC, Pan BS, Manne R, Li HY, Lin HK. The Skp2 Pathway: A Critical Target for Cancer Therapy. Semin Cancer Biol 2020; 67:16-33. [PMID: 32014608 DOI: 10.1016/j.semcancer.2020.01.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 12/16/2022]
Abstract
Strictly regulated protein degradation by ubiquitin-proteasome system (UPS) is essential for various cellular processes whose dysregulation is linked to serious diseases including cancer. Skp2, a well characterized component of Skp2-SCF E3 ligase complex, is able to conjugate both K48-linked ubiquitin chains and K63-linked ubiquitin chains on its diverse substrates, inducing proteasome mediated proteolysis or modulating the function of tagged substrates respectively. Overexpression of Skp2 is observed in various human cancers associated with poor survival and adverse therapeutic outcomes, which in turn suggests that Skp2 engages in tumorigenic activity. To that end, the oncogenic properties of Skp2 are demonstrated by various genetic mouse models, highlighting the potential of Skp2 as a target for tackling cancer. In this article, we will describe the downstream substrates of Skp2 as well as upstream regulators for Skp2-SCF complex activity. We will further summarize the comprehensive oncogenic functions of Skp2 while describing diverse strategies and therapeutic platforms currently available for developing Skp2 inhibitors.
Collapse
Affiliation(s)
- Zhen Cai
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA.
| | - Asad Moten
- National Capital Consortium, Department of Defense, Washington DC, 20307, USA; Institute for Complex Systems, HealthNovations International, Houston, TX, 77089, USA; Center for Cancer Research, National Institutes of Health, Bethesda, MD, 20814, USA; Center on Genomics, Vulnerable Populations, and Health Disparities, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Danni Peng
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Che-Chia Hsu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Rajeshkumar Manne
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Hong-Yu Li
- University of Arkansas for Medical Sciences, College of Pharmacy, Division of Pharmaceutical Science, 200 South Cedar, Little Rock AR 72202, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA; Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
30
|
Feng L, Li J, Bu X, Zuo Y, Shen L, Qu X. BRAF V600E dictates cell survival via c-Myc-dependent induction of Skp2 in human melanoma. Biochem Biophys Res Commun 2020; 524:28-35. [PMID: 31980175 DOI: 10.1016/j.bbrc.2019.12.085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022]
Abstract
BRAFV600E mutation is frequently observed in melanoma, and contributes to tumor malignancy. Despite inhibition of BRAF causes a profound cell growth inhibition and a strong clinical benefit in BRAFV600E melanoma, acquired drug resistance is still the major hurdle. In this study, we demonstrate that BRAFV600E drives cell growth and glycolysis in melanoma cells but does so by a previously unappreciated mechanism that involves direct induction of Skp2. Skp2 is highly expressed in melanoma tissues and particularly in tissues with BRAFV600E mutation. The inhibition of BRAFV600E by either siRNA or inhibitor vemurafenib suppressed Skp2 expression and cell growth. Mechanistic study shows that BRAFV600E suppression of Skp2 is dependent on c-Myc transcription factor via specifically bounding to the E-box region on SKP2 promoter. Further, the overexpression of Skp2 resulted in a markedly increase in cell growth, cell cycle progression and glycolysis which were repressed by BRAFV600E inhibition. Supporting the biological significance, Skp2 is specifically correlated with poor patient outcome in BRAFV600E but did not in BRAFWT melanomas. Thus, as a downstream target of BRAFV600E, Skp2 is critical for responses to BRAF inhibition, indicating targeting Skp2 might be a promising strategy for the treatment of BRAFi resistant melanomas.
Collapse
Affiliation(s)
- Lin Feng
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jun Li
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xin Bu
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Zuo
- Affiliated Hospital of Xizang Minzu University, Xianyang, 712082, China
| | - Liangliang Shen
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Xuan Qu
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
31
|
Sprouty3 and Sprouty4, Two Members of a Family Known to Inhibit FGF-Mediated Signaling, Exert Opposing Roles on Proliferation and Migration of Glioblastoma-Derived Cells. Cells 2019; 8:cells8080808. [PMID: 31374860 PMCID: PMC6721513 DOI: 10.3390/cells8080808] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022] Open
Abstract
Dysregulation of receptor tyrosine kinase-induced pathways is a critical step driving the oncogenic potential of brain cancer. In this study, we investigated the role of two members of the Sprouty (Spry) family in brain cancer-derived cell lines. Using immunoblot analyses we found essential differences in the pattern of endogenous Spry3 and Spry4 expression. While Spry4 expression was mitogen-dependent and repressed in a number of cells from higher malignant brain cancers, Spry3 levels neither fluctuated in response to serum withdrawal nor were repressed in glioblastoma (GBM)-derived cell lines. In accordance to the well-known inhibitory role of Spry proteins in fibroblast growth factor (FGF)-mediated signaling, both Spry proteins were able to interfere with FGF-induced activation of the MAPK pathway although to a different extent. In response to serum solely, Spry4 exerts its role as a negative regulator of MAPK activation. Ectopic expression of Spry4 inhibited proliferation and migration of GBM-originated cells, positioning it as a tumor suppressor in brain cancer. In contrast, elevated Spry3 levels accelerated both proliferation and migration of these cell lines, while repression of Spry3 levels using shRNA caused a significant diminished growth and migration velocity rate of a GBM-derived cell line. This argues for a tumor-promoting function of Spry3 in GBMs. Based on these data we conclude that Spry3 and Spry4 fulfill different if not opposing roles within the cancerogenesis of brain malignancies.
Collapse
|
32
|
Dittmer J, Stütz A, Vanas V, Salhi J, Reisecker JM, Kral RM, Sutterlüty-Fall H. Spatial signal repression as an additional role of Sprouty2 protein variants. Cell Signal 2019; 62:109332. [PMID: 31154002 DOI: 10.1016/j.cellsig.2019.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 11/19/2022]
Abstract
Sprouty2 (Spry2) is a prominent member of a protein family with crucial functions in the modulation of signal transduction. One of its main actions is the repression of mitogen-activated protein kinase (MAPK) pathway in response to growth factor-induced signalling. A common single nucleotide polymorphism within the Spry2 gene creates two protein variants where a proline adjacent to the serine rich domain is converted to an additional serine. Both protein variants perform similar functions although their efficiency in fulfilling these tasks varies. In this report, we used biochemical fractionation methods as well as confocal microscopy to analyse quantitative and qualitative differences in the distribution of Spry2 variants. We found that Spry2 proteins localize not solely to the plasma membrane, but also to other membrane engulfed compartments like for example the Golgi apparatus. In these less dense organelles, predominantly slower migrating forms reside indicating that posttranslational modification contributes to the distribution profile of Spry2. However there is no significant difference in the distribution of the two variants. Additionally, we found that Spry2 could be found exclusively in membrane fractions irrespective of the mitogen availability and the phosphorylation status. Considering the interference of extracellular signal-regulated kinase (ERK) activation in the cytoplasm, both Spry2 variants inhibited the levels of phosphorylated ERK (pERK) significantly to a similar extent. In contrast, the induction profiles of pERK levels were completely different in the nuclei. Again, both Spry2 variants diminished the levels of pERK. While the proline variant lowered the activation throughout the observation period, the serine variant failed to interfere with immediate accumulation of nuclear pERK levels, but the signal duration was shortened. Since the extent of the pERK inhibition in the nuclei was drastically more pronounced than in the cytoplasm, we conclude that Spry2 - in addition to its known functions as a repressor of general ERK phosphorylation - functions as a spatial repressor of nucleic ERK activation. Accordingly, a dominant negative version of Spry2 was only able to enhance the pERK levels of serum-deprived cells in the cytosol, while in the nucleus the intensity of the pERK signal in response to serum addition was significantly increased.
Collapse
Affiliation(s)
- Jakob Dittmer
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Astrid Stütz
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Vanita Vanas
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jihen Salhi
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Johannes Manfred Reisecker
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Rosana Maria Kral
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Hedwig Sutterlüty-Fall
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
33
|
Bi H, Liu Y, Tian T, Xia T, Pu R, Zhang Y, Hu F, Zhao Y. A Propensity Score-adjusted Analysis of the Effects of Ubiquitin E3 Ligase Copy Number Variation in Peripheral Blood Leukocytes on Colorectal Cancer Risk. J Cancer 2019; 10:3291-3302. [PMID: 31289601 PMCID: PMC6603381 DOI: 10.7150/jca.29872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 05/07/2019] [Indexed: 12/29/2022] Open
Abstract
Background: The ubiquitin ligases E3 (E3s) plays a key role in the specific protein degradation in many carcinogenic biological processes. Colorectal cancer (CRC) development may be affected by the copy number variation (CNV) of E3s. Prior studies may have underestimated the impact of potential confounding factors' effects on the association between gene CNV and CRC risk, and CRC risk predictive model integrating gene CNV patterns is lacking. Our research sought to assess the genes CNVs of MDM2, SKP2, FBXW7, β-TRCP, and NEDD4-1 and CRC risk by using propensity score (PS) adjustment and developing models that integrate CNV patterns for CRC risk predictions. Methods: This study comprising 1036 participants used traditional regression and different PS techniques to adjust the confounding factors to evaluate the relationships between five gene CNVs and CRC risk, and to establish a CRC risk predictive model. The AUC was applied to evaluate the effect of the model. The categorical net reclassification improvement (NRI) and the integrated discrimination improvement (IDI) were analyzed to evaluate the discriminatory accuracy improvement among the models. Results: Compared to variable adjustment, the odds ratios (ORs) tended to be conservative and accurate with narrow confidence intervals (CIs) after PS adjustment. After PS adjustment, MDM2 amplification was related to increased CRC risk (Amp-pattern: OR = 8.684, 95% CI: 1.213-62.155, P = 0.031), whereas SKP2 deletion and the (del+amp) genotype were associated with reduced CRC risk (Del-pattern: OR = 0.323, 95% CI: 0.106-0.979, P = 0.046; Var-pattern: OR = 0.339, 95% CI: 0.135-0.854, P = 0.024). The predictive model integrating the gene CNV pattern could correctly reclassify 1.7% of the subjects. Conclusions: MDM2 amplification and SKP2 CNVs are associated with increased and decreased CRC risk, respectively; abnormal CNV-integrated model is more precise for predicting CRC risk. Further studies are needed to verify these encouraging outcomes.
Collapse
Affiliation(s)
- Haoran Bi
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Yupeng Liu
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Tian Tian
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Tingting Xia
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Rui Pu
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Yiwei Zhang
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Fulan Hu
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| | - Yashuang Zhao
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
34
|
Kawauchi T, Nabeshima YI. Growth Arrest Triggers Extra-Cell Cycle Regulatory Function in Neurons: Possible Involvement of p27 kip1 in Membrane Trafficking as Well as Cytoskeletal Regulation. Front Cell Dev Biol 2019; 7:64. [PMID: 31080801 PMCID: PMC6497764 DOI: 10.3389/fcell.2019.00064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/09/2019] [Indexed: 11/30/2022] Open
Abstract
Cell cycle regulation is essential for the development of multicellular organisms, but many cells in adulthood, including neurons, exit from cell cycle. Although cell cycle-related proteins are suppressed after cell cycle exit in general, recent studies have revealed that growth arrest triggers extra-cell cycle regulatory function (EXCERF) in some cell cycle proteins, such as p27(kip1), p57(kip2), anaphase-promoting complex/cyclosome (APC/C), and cyclin E. While p27 is known to control G1 length and cell cycle exit via inhibition of cyclin-dependent kinase (CDK) activities, p27 acquires additional cytoplasmic functions in growth-arrested neurons. Here, we introduce the EXCERFs of p27 in post-mitotic neurons, mainly focusing on its actin and microtubule regulatory functions. We also show that a small amount of p27 is associated with the Golgi apparatus positive for Rab6, p115, and GM130, but not endosomes positive for Rab5, Rab7, Rab8, Rab11, SNX6, or LAMTOR1. p27 is also colocalized with Dcx, a microtubule-associated protein. Based on these results, we discuss here the possible role of p27 in membrane trafficking and microtubule-dependent transport in post-mitotic cortical neurons. Collectively, we propose that growth arrest leads to two different fates in cell cycle proteins; either suppressing their expression or activating their EXCERFs. The latter group of proteins, including p27, play various roles in neuronal migration, morphological changes and axonal transport, whereas the re-activation of the former group of proteins in post-mitotic neurons primes for cell death.
Collapse
Affiliation(s)
- Takeshi Kawauchi
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yo-Ichi Nabeshima
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| |
Collapse
|
35
|
S-Like-Phase Cyclin-Dependent Kinases Stabilize the Epstein-Barr Virus BDLF4 Protein To Temporally Control Late Gene Transcription. J Virol 2019; 93:JVI.01707-18. [PMID: 30700607 DOI: 10.1128/jvi.01707-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/23/2019] [Indexed: 12/27/2022] Open
Abstract
Temporally controlled gene expression is necessary for the propagation of herpesviruses. To achieve this, herpesviruses encode several transcriptional regulators. In Epstein-Barr virus, BcRF1 associates with five viral proteins (BDLF4, BGLF3, BFRF2, BVLF1, and BDLF3.5) to form the viral late (L) gene regulatory complex, which is called the viral preinitiation complex (vPIC), on TATT-containing promoters. However, regulation of the vPIC has been largely unexplored. In this study, we performed two screens using a kinase inhibitor library and identified a series of cyclin-dependent kinase (CDK) inhibitors that downregulated the expression of L genes without any impact on viral DNA replication through destabilization of the BDLF4 protein. Knockdown of CDK2 by short hairpin RNA (shRNA) and proteasome inhibitor treatment showed that phosphorylation of the BDLF4 protein prevented ubiquitin-mediated degradation. Moreover, we demonstrated that cyclin A- and E-associated CDK2 complexes phosphorylated BDLF4 in vitro, and we identified several serine/threonine phosphorylation sites in BDLF4. Phosphoinactive and phosphomimic mutants revealed that phosphorylation at threonine 91 plays a role in stabilizing BDLF4. Therefore, our findings indicate that S-like-phase CDKs mediate the regulation of L gene expression through stabilization of the BDLF4 protein, which makes the temporal L gene expression system more robust.IMPORTANCE Late (L) genes represent more than one-third of the herpesvirus genome, suggesting that many of these genes are indispensable for the life cycle of the virus. With the exception of BCRF1, BDLF2, and BDLF3, Epstein-Barr virus L genes are transcribed by viral regulators, which are known as the viral preinitiation complex (vPIC) and the host RNA polymerase II complex. Because the vPIC is conserved in beta- and gammaherpesviruses, studying the control of viral L gene expression by the vPIC contributes to the development of drugs that specifically inhibit these processes in beta- and gammaherpesvirus infections/diseases. In this study, we demonstrated that CDK inhibitors induced destabilization of the vPIC component BDLF4, leading to a reduction in L gene expression and subsequent progeny production. Our findings suggest that CDK inhibitors may be a therapeutic option against beta- and gammaherpesviruses in combination with existing inhibitors of herpesvirus lytic replication, such as ganciclovir.
Collapse
|
36
|
Zhou W, Xu J, Tan M, Li H, Li H, Wei W, Sun Y. UBE2M Is a Stress-Inducible Dual E2 for Neddylation and Ubiquitylation that Promotes Targeted Degradation of UBE2F. Mol Cell 2019; 70:1008-1024.e6. [PMID: 29932898 DOI: 10.1016/j.molcel.2018.06.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/28/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022]
Abstract
UBE2M and UBE2F are two family members of neddylation E2 conjugating enzyme that, together with E3s, activate CRLs (Cullin-RING Ligases) by catalyzing cullin neddylation. However, whether and how two E2s cross-talk with each other are largely unknown. Here, we report that UBE2M is a stress-inducible gene subjected to cis-transactivation by HIF-1 and AP1, and MLN4924, a small molecule inhibitor of E1 NEDD8-activating enzyme (NAE), upregulates UBE2M via blocking degradation of HIF-1α and c-JUN. UBE2M is a dual E2 for targeted ubiquitylation and degradation of UBE2F, acting as a neddylation E2 to activate CUL3-Keap1 E3 under physiological conditions but as a ubiquitylation E2 for Parkin-DJ-1 E3 under stressed conditions. UBE2M-induced UBE2F degradation leads to CRL5 inactivation and subsequent NOXA accumulation to suppress the growth of lung cancer cells. Collectively, our study establishes a negative regulatory axis between two neddylation E2s with UBE2M ubiquitylating UBE2F, and two CRLs with CRL3 inactivating CRL5.
Collapse
Affiliation(s)
- Weihua Zhou
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Xu
- Department of Urology, the Second Affiliated Hospital, Third Military Medical University, Chongqing, PRC
| | - Mingjia Tan
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Haomin Li
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PRC; Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PRC
| | - Hua Li
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Yi Sun
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PRC.
| |
Collapse
|
37
|
García-Gutiérrez L, Delgado MD, León J. MYC Oncogene Contributions to Release of Cell Cycle Brakes. Genes (Basel) 2019; 10:E244. [PMID: 30909496 PMCID: PMC6470592 DOI: 10.3390/genes10030244] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Promotion of the cell cycle is a major oncogenic mechanism of the oncogene c-MYC (MYC). MYC promotes the cell cycle by not only activating or inducing cyclins and CDKs but also through the downregulation or the impairment of the activity of a set of proteins that act as cell-cycle brakes. This review is focused on the role of MYC as a cell-cycle brake releaser i.e., how MYC stimulates the cell cycle mainly through the functional inactivation of cell cycle inhibitors. MYC antagonizes the activities and/or the expression levels of p15, ARF, p21, and p27. The mechanism involved differs for each protein. p15 (encoded by CDKN2B) and p21 (CDKN1A) are repressed by MYC at the transcriptional level. In contrast, MYC activates ARF, which contributes to the apoptosis induced by high MYC levels. At least in some cells types, MYC inhibits the transcription of the p27 gene (CDKN1B) but also enhances p27's degradation through the upregulation of components of ubiquitin ligases complexes. The effect of MYC on cell-cycle brakes also opens the possibility of antitumoral therapies based on synthetic lethal interactions involving MYC and CDKs, for which a series of inhibitors are being developed and tested in clinical trials.
Collapse
Affiliation(s)
- Lucía García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
- Current address: Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
| | - María Dolores Delgado
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
| |
Collapse
|
38
|
Li Z, Qi DL, Singh HP, Zou Y, Shen B, Cobrinik D. A novel thyroid hormone receptor isoform, TRβ2-46, promotes SKP2 expression and retinoblastoma cell proliferation. J Biol Chem 2019; 294:2961-2969. [PMID: 30643022 DOI: 10.1074/jbc.ac118.006041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/07/2019] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma is a childhood retinal tumor that develops from cone photoreceptor precursors in response to inactivating RB1 mutations and loss of functional RB protein. The cone precursor's response to RB loss involves cell type-specific signaling circuitry that helps to drive tumorigenesis. One component of the cone precursor circuitry, the thyroid hormone receptor β2 (TRβ2), enables the aberrant proliferation of diverse RB-deficient cells in part by opposing the down-regulation of S-phase kinase-associated protein 2 (SKP2) by the more widely expressed and tumor-suppressive TRβ1. However, it is unclear how TRβ2 opposes TRβ1 to enable SKP2 expression and cell proliferation. Here, we show that in human retinoblastoma cells TRβ2 mRNA encodes two TRβ2 protein isoforms: a predominantly cytoplasmic 54-kDa protein (TRβ2-54) corresponding to the well-characterized full-length murine Trβ2 and an N-terminally truncated and exclusively cytoplasmic 46-kDa protein (TRβ2-46) that starts at Met-79. Whereas TRβ2 knockdown decreased SKP2 expression and impaired retinoblastoma cell cycle progression, re-expression of TRβ2-46 but not TRβ2-54 stabilized SKP2 and restored proliferation to an extent similar to that of ectopic SKP2 restoration. We conclude that TRβ2-46 is an oncogenic thyroid hormone receptor isoform that promotes SKP2 expression and SKP2-dependent retinoblastoma cell proliferation.
Collapse
Affiliation(s)
- Zhengke Li
- From The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, .,Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010.,Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, and
| | - Dong-Lai Qi
- From The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027
| | - Hardeep P Singh
- From The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027
| | - Yue Zou
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, and
| | - Binghui Shen
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - David Cobrinik
- From The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, .,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, and USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California 90033
| |
Collapse
|
39
|
Chen YT, Su YC, Kung JT. B Cell Development sans B Cell Receptor Responsiveness Due to Unfolded Protein Response–Triggered Mef2c Protein Degradation. THE JOURNAL OF IMMUNOLOGY 2018; 201:2885-2898. [DOI: 10.4049/jimmunol.1800685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/11/2018] [Indexed: 01/10/2023]
|
40
|
Meyer SE, Muench DE, Rogers AM, Newkold TJ, Orr E, O'Brien E, Perentesis JP, Doench JG, Lal A, Morris PJ, Thomas CJ, Lieberman J, McGlinn E, Aronow BJ, Salomonis N, Grimes HL. miR-196b target screen reveals mechanisms maintaining leukemia stemness with therapeutic potential. J Exp Med 2018; 215:2115-2136. [PMID: 29997117 PMCID: PMC6080909 DOI: 10.1084/jem.20171312] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 04/30/2018] [Accepted: 06/26/2018] [Indexed: 01/02/2023] Open
Abstract
We have shown that antagomiR inhibition of miRNA miR-21 and miR-196b activity is sufficient to ablate MLL-AF9 leukemia stem cells (LSC) in vivo. Here, we used an shRNA screening approach to mimic miRNA activity on experimentally verified miR-196b targets to identify functionally important and therapeutically relevant pathways downstream of oncogenic miRNA in MLL-r AML. We found Cdkn1b (p27Kip1) is a direct miR-196b target whose repression enhanced an embryonic stem cell-like signature associated with decreased leukemia latency and increased numbers of leukemia stem cells in vivo. Conversely, elevation of p27Kip1 significantly reduced MLL-r leukemia self-renewal, promoted monocytic differentiation of leukemic blasts, and induced cell death. Antagonism of miR-196b activity or pharmacologic inhibition of the Cks1-Skp2-containing SCF E3-ubiquitin ligase complex increased p27Kip1 and inhibited human AML growth. This work illustrates that understanding oncogenic miRNA target pathways can identify actionable targets in leukemia.
Collapse
MESH Headings
- Animals
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Cell Differentiation/genetics
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Survival/genetics
- Chromosomes, Human, Pair 11/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Cyclin-Dependent Kinases/metabolism
- Cyclins/metabolism
- Embryonic Stem Cells/metabolism
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice, Inbred C57BL
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oncogenes
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Sara E Meyer
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - David E Muench
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Andrew M Rogers
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Tess J Newkold
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Emily Orr
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Eric O'Brien
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - John P Perentesis
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Patrick J Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Edwina McGlinn
- EMBL Australia, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - H Leighton Grimes
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
41
|
Moonen L, D'Haese PC, Vervaet BA. Epithelial Cell Cycle Behaviour in the Injured Kidney. Int J Mol Sci 2018; 19:E2038. [PMID: 30011818 PMCID: PMC6073451 DOI: 10.3390/ijms19072038] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI), commonly caused by ischemia-reperfusion injury, has far-reaching health consequences. Despite the significant regenerative capacity of proximal tubular epithelium cells (PTCs), repair frequently fails, leading to the development of chronic kidney disease (CKD). In the last decade, it has been repeatedly demonstrated that dysregulation of the cell cycle can cause injured kidneys to progress to CKD. More precisely, severe AKI causes PTCs to arrest in the G1/S or G2/M phase of the cell cycle, leading to maladaptive repair and a fibrotic outcome. The mechanisms causing these arrests are far from known. The arrest might, at least partially, be attributed to DNA damage since activation of the DNA-damage response pathway leads to cell cycle arrest. Alternatively, cytokine signalling via nuclear factor kappa beta (NF-κβ) and p38-mitogen-activated protein kinase (p38-MAPK) pathways, and reactive oxygen species (ROS) can play a role independent of DNA damage. In addition, only a handful of cell cycle regulators (e.g., p53, p21) have been thoroughly studied during renal repair. Still, why and how PTCs decide to arrest their cell cycle and how this arrest can efficiently be overcome remain open and challenging questions. In this review we will discuss the evidence for cell cycle involvement during AKI and development of CKD together with putative therapeutic approaches.
Collapse
Affiliation(s)
- Lies Moonen
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Benjamin A Vervaet
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| |
Collapse
|
42
|
Mansour MA. Ubiquitination: Friend and foe in cancer. Int J Biochem Cell Biol 2018; 101:80-93. [PMID: 29864543 DOI: 10.1016/j.biocel.2018.06.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/05/2023]
Abstract
Dynamic modulation and posttranslational modification of proteins are tightly controlled biological processes that occur in response to physiological cues. One such dynamic modulation is ubiquitination, which marks proteins for degradation via the proteasome, altering their localization, affecting their activity, and promoting or interfering with protein interactions. Hence, ubiquitination is crucial for a plethora of physiological processes, including cell survival, differentiation and innate and adaptive immunity. Similar to kinases, components of the ubiquitination system are often deregulated, leading to a variety of diseases, such as cancer and neurodegenerative disorders. In a context-dependent manner, ubiquitination can regulate both tumor-suppressing and tumor-promoting pathways in cancer. This review outlines how components of the ubiquitination systems (e.g. E3 ligases and deubiquitinases) act as oncogenes or tumor suppressors according to the nature of their substrates. Furthermore, I interrogate how the current knowledge of the differential roles of ubiquitination in cancer lead to technical advances to inhibit or reactivate the components of the ubiquitination system accordingly.
Collapse
Affiliation(s)
- Mohammed A Mansour
- Institute of Cancer Sciences, University of Glasgow, United Kingdom; The CRUK Beatson Institute, Glasgow, Switchback Road, G61 1BD, United Kingdom; Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
43
|
Chou CF, Hsieh YH, Grubbs CJ, Atigadda VR, Mobley JA, Dummer R, Muccio DD, Eto I, Elmets CA, Garvey WT, Chang PL. The retinoid X receptor agonist, 9-cis UAB30, inhibits cutaneous T-cell lymphoma proliferation through the SKP2-p27kip1 axis. J Dermatol Sci 2018; 90:343-356. [PMID: 29599065 PMCID: PMC6329374 DOI: 10.1016/j.jdermsci.2018.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/15/2018] [Accepted: 03/08/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Bexarotene (Targretin®) is currently the only FDA approved retinoid X receptor (RXR) -selective agonist for the treatment of cutaneous T-cell lymphomas (CTCLs). The main side effects of bexarotene are hypothyroidism and elevation of serum triglycerides (TGs). The novel RXR ligand, 9-cis UAB30 (UAB30) does not elevate serum TGs or induce hypothyroidism in normal subjects. OBJECTIVES To assess preclinical efficacy and mechanism of action of UAB30 in the treatment of CTCLs and compare its action with bexarotene. METHODS With patient-derived CTCL cell lines, we evaluated UAB30 function in regulating growth, apoptosis, cell cycle check points, and cell cycle-related markers. RESULTS Compared to bexarotene, UAB30 had lower half maximal inhibitory concentration (IC50) values and was more effective in inhibiting the G1 cell cycle checkpoint. Both rexinoids increased the stability of the cell cycle inhibitor, p27kip1 protein, in part, through targeting components involved in the ubiquitination-proteasome system: 1) decreasing SKP2, a F-box protein that binds and targets p27kip1 for degradation by 26S proteasome and 2) suppressing 20S proteasome activity (cell line-dependent) through downregulation of PSMA7, a component of the 20S proteolytic complex in 26S proteasome. CONCLUSIONS UAB30 and bexarotene induce both early cell apoptosis and suppress cell proliferation. Inhibition of the G1 to S cell cycle transition by rexinoids is mediated, in part, through downregulation of SKP2 and/or 20S proteasome activity, leading to increased p27kip1 protein stability. Because UAB30 has minimal effect in elevating serum TGs and inducing hypothyroidism, it is potentially a better alternative to bexarotene for the treatment of CTCLs.
Collapse
Affiliation(s)
- Chu-Fang Chou
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Yu-Hua Hsieh
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Venkatram R Atigadda
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - James A Mobley
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Switzerland
| | - Donald D Muccio
- Department of Biochemistry, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Isao Eto
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Craig A Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Pi-Ling Chang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA; Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA.
| |
Collapse
|
44
|
Joshi V, Upadhyay A, Chhangani D, Amanullah A, Sharan RN, Mishra A. Gp78 involvement in cellular proliferation: Can act as a promising modulator for cell cycle regulatory proteins? J Cell Physiol 2018; 233:6352-6368. [PMID: 29741771 DOI: 10.1002/jcp.26618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 03/28/2018] [Indexed: 11/07/2022]
Abstract
In cells, protein synthesis and degradation are normal processes, which are tightly regulated by various cellular metabolic pathways. Cellular protein quality control (PQC) mechanisms always present a continuous and rigorous check over all intracellular proteins before they can participate in various cellular physiological processes with the help of PQC pathways like autophagy and ubiquitin proteasome system (UPS). The UPS employs few selective E3 ubiquitin ligases for the intracellular degradation of cyclin-dependent kinase inhibitor 1B (p27Kip1 ) that tightly controls cell cycle progression. But, the complex mechanistic interactions and the interplay between E3 ubiquitin ligases involved in the functional regulation as well as expression of p27 are not well known. Here, we demonstrate that cell surface glycoprotein Gp78, a putative E3 ubiquitin ligase, is involved in the stabilization of intracellular steady-state levels of p27. Transient overexpression of Gp78 increases the accumulation of p27 in cells in the form of massive inclusions like structures, which could be due to its cumulative increased stability in cells. We have also monitored how under stress condition, E3 ubiquitin ligase Gp78 regulates endogenous levels of p27 in cells. ER stress treatment generates a marginal increase in Gp78 endogenous levels, and this elevation effect was prominent for intracellular accumulation of p27 in cells. Taken together, our current findings suggest a valuable multifactorial regulatory mechanism and linkage of p27 with UPS pathway.
Collapse
Affiliation(s)
- Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Deepak Chhangani
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Rajesh N Sharan
- Radiation and Molecular Biology Unit, Department of Biochemistry, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| |
Collapse
|
45
|
Yokoda R, Nagalo BM, Arora M, Egan JB, Bogenberger JM, DeLeon TT, Zhou Y, Ahn DH, Borad MJ. Oncolytic virotherapy in upper gastrointestinal tract cancers. Oncolytic Virother 2018; 7:13-24. [PMID: 29616200 PMCID: PMC5870634 DOI: 10.2147/ov.s161397] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Upper gastrointestinal tract malignancies are among the most challenging cancers with regard to response to treatment and prognosis. Cancers of the esophagus, stomach, pancreas, liver, and biliary tree have dismal 5-year survival, and very modest improvements in this rate have been made in recent times. Oncolytic viruses are being developed to address these malignancies, with a focus on high safety profiles and low off-target toxicities. Each viral platform has evolved to enhance oncolytic potency and the clinical response to either single-agent viral therapy or combined viral treatment with radiotherapy and chemotherapy. A panel of genomic alterations, chimeric proteins, and pseudotyped capsids are the breakthroughs for vector success. This article revisits developments for each viral platform to each tumor type, in an attempt to achieve maximum tumor selectivity. From the bench to clinical trials, the scope of this review is to highlight the beginnings of translational oncolytic virotherapy research in upper gastrointestinal tract malignancies and provide a bioengineering perspective of the most promising platforms.
Collapse
Affiliation(s)
- Raquel Yokoda
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Bolni M Nagalo
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Mansi Arora
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Jan B Egan
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - James M Bogenberger
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Thomas T DeLeon
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Yumei Zhou
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Daniel H Ahn
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Mitesh J Borad
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ.,Department of Molecular Medicine, Center for Individualized Medicine, Mayo Clinic, Rochester, MN.,Department of Oncology, Mayo Clinic Cancer Center, Phoenix, AZ, USA
| |
Collapse
|
46
|
Abstract
We previously reported that p53 plays a role as a key regulator in the tetraploid G1 checkpoint, which is activated by actin damage-induced cytokinesis blockade and then prevents uncoupled DNA replication and nuclear division without cytokinesis. In this study, we investigated a role of Skp2, which targets CDK2 inhibitor p27/Kip1, in actin damage-induced tetraploid G1 arrest. Expression of Skp2 was reduced, but p27/Kip1 was increased, after actin damage-induced cytokinesis blockade. The role of Skp2 repression in tetraploid G1 arrest was investigated by analyzing the effects of ectopic expression of Skp2. After actin damage, ectopic expression of Skp2 resulted in DNA synthesis and accumulation of multinucleated cells, and ultimately, induction of apoptosis. These results suggest that Skp2 repression is important for sustaining tetraploid G1 arrest after cytokinesis blockade and is required to prevent uncoupled DNA replication and nuclear division without cytokinesis.
Collapse
Affiliation(s)
- Yongsam Jo
- Department of Microbiology, Dankook University College of Medicine, Cheonan 31116, Korea
| | - Deug Y Shin
- Department of Microbiology, Dankook University College of Medicine, Cheonan 31116, Korea
| |
Collapse
|
47
|
mTORC1 Inactivation Promotes Colitis-Induced Colorectal Cancer but Protects from APC Loss-Dependent Tumorigenesis. Cell Metab 2018; 27:118-135.e8. [PMID: 29275959 DOI: 10.1016/j.cmet.2017.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/21/2017] [Accepted: 11/15/2017] [Indexed: 01/17/2023]
Abstract
Dietary habits that can induce inflammatory bowel disease (IBD) are major colorectal cancer (CRC) risk factors, but mechanisms linking nutrients, IBD, and CRC are unknown. Using human data and mouse models, we show that mTORC1 inactivation-induced chromosomal instability impairs intestinal crypt proliferation and regeneration, CDK4/6 dependently. This triggers interleukin (IL)-6-associated reparative inflammation, inducing crypt hyper-proliferation, wound healing, and CRC. Blocking IL-6 signaling or reactivating mTORC1 reduces inflammation-induced CRC, so mTORC1 activation suppresses tumorigenesis in IBD. Conversely, mTORC1 inactivation is beneficial in APC loss-dependent CRC. Thus, IL-6 blockers or protein-rich-diet-linked mTORC1 activation may prevent IBD-associated CRC. However, abolishing mTORC1 can mitigate CRC in predisposed patients with APC mutations. Our work reveals mTORC1 oncogenic and tumor-suppressive roles in intestinal epithelium and avenues to optimized and personalized therapeutic regimens for CRC.
Collapse
|
48
|
Composition and Regulation of the Cellular Repertoire of SCF Ubiquitin Ligases. Cell 2017; 171:1326-1339.e14. [PMID: 29103612 DOI: 10.1016/j.cell.2017.10.016] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/17/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
SCF (Skp1-Cullin-F-box) ubiquitin ligases comprise several dozen modular enzymes that have diverse roles in biological regulation. SCF enzymes share a common catalytic core containing Cul1⋅Rbx1, which is directed toward different substrates by a variable substrate receptor (SR) module comprising 1 of 69 F-box proteins bound to Skp1. Despite the broad cellular impact of SCF enzymes, important questions remain about the architecture and regulation of the SCF repertoire, including whether SRs compete for Cul1 and, if so, how this competition is managed. Here, we devise methods that preserve the in vivo assemblages of SCF complexes and apply quantitative mass spectrometry to perform a census of these complexes (the "SCFome") in various states. We show that Nedd8 conjugation and the SR exchange factor Cand1 have a profound effect on shaping the SCFome. Together, these factors enable rapid remodeling of SCF complexes to promote biased assembly of SR modules bound to substrate.
Collapse
|
49
|
Craveiro M, Cretenet G, Mongellaz C, Matias MI, Caron O, de Lima MCP, Zimmermann VS, Solary E, Dardalhon V, Dulić V, Taylor N. Resveratrol stimulates the metabolic reprogramming of human CD4 + T cells to enhance effector function. Sci Signal 2017; 10:10/501/eaal3024. [PMID: 29042482 DOI: 10.1126/scisignal.aal3024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The polyphenol resveratrol activates the deacetylase Sirt1, resulting in various antioxidant, chemoprotectant, neuroprotective, cardioprotective, and anti-inflammatory properties. We found that at high concentrations of resveratrol, human CD4+ T cells showed defective antigen receptor signaling and arrest at the G1 stage of the cell cycle, whereas at low concentrations, cells were readily activated and exhibited enhanced Sirt1 deacetylase activity. Nevertheless, low-dose resveratrol rapidly stimulated genotoxic stress in the T cells, which resulted in engagement of a DNA damage response pathway that depended on the kinase ATR [ataxia telangiectasia-mutated (ATM) and Rad3-related], but not ATM, and subsequently in premitotic cell cycle arrest. The concomitant activation of p53 was coupled to the expression of gene products that regulate cell metabolism, leading to a metabolic reprogramming that was characterized by decreased glycolysis, increased glutamine consumption, and a shift to oxidative phosphorylation. These alterations in the bioenergetic homeostasis of CD4+ T cells resulted in enhanced effector function, with both naïve and memory CD4+ T cells secreting increased amounts of the inflammatory cytokine interferon-γ. Thus, our data highlight the wide range of metabolic adaptations that CD4+ T lymphocytes undergo in response to genomic stress.
Collapse
Affiliation(s)
- Marco Craveiro
- IGMM, CNRS, Université de Montpellier, Montpellier, France.,CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | - Maria I Matias
- IGMM, CNRS, Université de Montpellier, Montpellier, France
| | - Olivier Caron
- INSERM U1170, Gustave Roussy Cancer Center, Villejuif, France.,Faculty of Medicine, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | | | | | - Eric Solary
- INSERM U1170, Gustave Roussy Cancer Center, Villejuif, France.,Faculty of Medicine, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | | | | | - Naomi Taylor
- IGMM, CNRS, Université de Montpellier, Montpellier, France.
| |
Collapse
|
50
|
Wang L, Zhang R, You X, Zhang H, Wei S, Cheng T, Cao Q, Wang Z, Chen Y. The steady-state level of CDK4 protein is regulated by antagonistic actions between PAQR4 and SKP2 and involved in tumorigenesis. J Mol Cell Biol 2017; 9:409-421. [DOI: 10.1093/jmcb/mjx028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/04/2017] [Indexed: 01/26/2023] Open
Affiliation(s)
- Lin Wang
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Zhang
- Cancer Molecular Diagnostic Core Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xue You
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Huanhuan Zhang
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Siying Wei
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tingting Cheng
- Department of Clinical Medicine, Tongji University, Shanghai, China
| | - Qianqian Cao
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenzhen Wang
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|