1
|
Shi D, Yang Z, Cai Y, Li H, Lin L, Wu D, Zhang S, Guo Q. Research advances in the molecular classification of gastric cancer. Cell Oncol (Dordr) 2024; 47:1523-1536. [PMID: 38717722 PMCID: PMC11466988 DOI: 10.1007/s13402-024-00951-9] [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] [Accepted: 04/13/2024] [Indexed: 06/27/2024] Open
Abstract
Gastric cancer (GC) is a malignant tumor with one of the lowest five-year survival rates. Traditional first-line treatment regimens, such as platinum drugs, have limited therapeutic efficacy in treating advanced GC and significant side effects, greatly reducing patient quality of life. In contrast, trastuzumab and other immune checkpoint inhibitors, such as nivolumab and pembrolizumab, have demonstrated consistent and reliable efficacy in treating GC. Here, we discuss the intrinsic characteristics of GC from a molecular perspective and provide a comprehensive review of classification and treatment advances in the disease. Finally, we suggest several strategies based on the intrinsic molecular characteristics of GC to aid in overcoming clinical challenges in the development of precision medicine and improve patient prognosis.
Collapse
Affiliation(s)
- Dike Shi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road, Hangzhou, 310009, China
| | - Zihan Yang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yanna Cai
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongbo Li
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lele Lin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road, Hangzhou, 310009, China
| | - Dan Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road, Hangzhou, 310009, China
| | - Shengyu Zhang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Qingqu Guo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road, Hangzhou, 310009, China.
| |
Collapse
|
2
|
Devillers R, Dos Santos A, Destombes Q, Laplante M, Elowe S. Recent insights into the causes and consequences of chromosome mis-segregation. Oncogene 2024; 43:3139-3150. [PMID: 39278989 DOI: 10.1038/s41388-024-03163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
Mitotic cells face the challenging task of ensuring accurate and equal segregation of their duplicated, condensed chromosomes between the nascent daughter cells. Errors in the process result in chromosome missegregation, a significant consequence of which is the emergence of aneuploidy-characterized by an imbalance in chromosome number-and the associated phenomenon of chromosome instability (CIN). Aneuploidy and CIN are common features of cancer, which leverages them to promote genome heterogeneity and plasticity, thereby facilitating rapid tumor evolution. Recent research has provided insights into how mitotic errors shape cancer genomes by inducing both numerical and structural chromosomal changes that drive tumor initiation and progression. In this review, we survey recent findings regarding the mitotic causes and consequences of aneuploidy. We discuss new findings into the types of chromosome segregation errors that lead to aneuploidy and novel pathways that protect genome integrity during mitosis. Finally, we describe new developments in our understanding of the immediate consequences of chromosome mis-segregation on the genome stability of daughter cells.
Collapse
Affiliation(s)
- Romain Devillers
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Alexsandro Dos Santos
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Quentin Destombes
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Mathieu Laplante
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Sabine Elowe
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada.
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada.
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada.
- Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
3
|
Hosea R, Hillary S, Naqvi S, Wu S, Kasim V. The two sides of chromosomal instability: drivers and brakes in cancer. Signal Transduct Target Ther 2024; 9:75. [PMID: 38553459 PMCID: PMC10980778 DOI: 10.1038/s41392-024-01767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 04/02/2024] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and is associated with tumor cell malignancy. CIN triggers a chain reaction in cells leading to chromosomal abnormalities, including deviations from the normal chromosome number or structural changes in chromosomes. CIN arises from errors in DNA replication and chromosome segregation during cell division, leading to the formation of cells with abnormal number and/or structure of chromosomes. Errors in DNA replication result from abnormal replication licensing as well as replication stress, such as double-strand breaks and stalled replication forks; meanwhile, errors in chromosome segregation stem from defects in chromosome segregation machinery, including centrosome amplification, erroneous microtubule-kinetochore attachments, spindle assembly checkpoint, or defective sister chromatids cohesion. In normal cells, CIN is deleterious and is associated with DNA damage, proteotoxic stress, metabolic alteration, cell cycle arrest, and senescence. Paradoxically, despite these negative consequences, CIN is one of the hallmarks of cancer found in over 90% of solid tumors and in blood cancers. Furthermore, CIN could endow tumors with enhanced adaptation capabilities due to increased intratumor heterogeneity, thereby facilitating adaptive resistance to therapies; however, excessive CIN could induce tumor cells death, leading to the "just-right" model for CIN in tumors. Elucidating the complex nature of CIN is crucial for understanding the dynamics of tumorigenesis and for developing effective anti-tumor treatments. This review provides an overview of causes and consequences of CIN, as well as the paradox of CIN, a phenomenon that continues to perplex researchers. Finally, this review explores the potential of CIN-based anti-tumor therapy.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sumera Naqvi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
4
|
Yang D, He Y, Li R, Huang Z, Zhou Y, Shi Y, Deng Z, Wu J, Gao Y. Histone H3K79 methylation by DOT1L promotes Aurora B localization at centromeres in mitosis. Cell Rep 2023; 42:112885. [PMID: 37494186 DOI: 10.1016/j.celrep.2023.112885] [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: 01/27/2023] [Revised: 05/23/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023] Open
Abstract
Centromere localization of the chromosome passenger complex (CPC) is paramount for achieving accurate sister chromosome segregation in mitosis. Although it has been widely recognized that the recruitment of CPC is directly regulated by two histone codes, phosphorylation of histone H3 at threonine 3 (H3T3ph) and phosphorylation of histone H2A at threonine 120 (H2AT120ph), the regulation of CPC localization by other histone codes remains elusive. We show that dysfunction of disruptor of telomeric silencing 1 like (DOT1L) leads to mislocation of the CPC in prometaphase, caused by disturbing the level of H3T3ph and its reader Survivin. This cascade is initiated by over-dephosphorylation of H3T3ph mediated by the phosphatase RepoMan-PP1, whose scaffold RepoMan translocalizes to chromosomes, while the level of H3K79me2/3 is diminished. Together, our findings uncover a biological function of DOT1L and H3K79 methylation in mitosis and give insight into how genomic stability is coordinated by different histone codes.
Collapse
Affiliation(s)
- Dan Yang
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yanji He
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Renyan Li
- Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing 401120, China
| | - Zhenting Huang
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yong Zhou
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yingxu Shi
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zhongliang Deng
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jingxian Wu
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yanfei Gao
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
5
|
Cheng A, Xu T, You W, Wang T, Zhang D, Guo H, Zhang H, Pan X, Wang Y, Liu L, Zhang K, Shi J, Yao X, Guo J, Yang Z. A mitotic NADPH upsurge promotes chromosome segregation and tumour progression in aneuploid cancer cells. Nat Metab 2023; 5:1141-1158. [PMID: 37349486 DOI: 10.1038/s42255-023-00832-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/26/2023] [Indexed: 06/24/2023]
Abstract
Redox metabolites have been observed to fluctuate through the cell cycle in cancer cells, but the functional impacts of such metabolic oscillations remain unknown. Here, we uncover a mitosis-specific nicotinamide adenine dinucleotide phosphate (NADPH) upsurge that is essential for tumour progression. Specifically, NADPH is produced by glucose 6-phosphate dehydrogenase (G6PD) upon mitotic entry, which neutralizes elevated reactive oxygen species (ROS) and prevents ROS-mediated inactivation of mitotic kinases and chromosome missegregation. Mitotic activation of G6PD depends on the phosphorylation of its co-chaperone protein BAG3 at threonine 285, which results in dissociation of inhibitory BAG3. Blocking BAG3T285 phosphorylation induces tumour suppression. A mitotic NADPH upsurge is present in aneuploid cancer cells with high levels of ROS, while nearly unobservable in near-diploid cancer cells. High BAG3T285 phosphorylation is associated with worse prognosis in a cohort of patients with microsatellite-stable colorectal cancer. Our study reveals that aneuploid cancer cells with high levels of ROS depend on a G6PD-mediated NADPH upsurge in mitosis to protect them from ROS-induced chromosome missegregation.
Collapse
Affiliation(s)
- Aoxing Cheng
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weiyi You
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ting Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dongming Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huimin Guo
- Center for Biological Technology, Anhui Agricultural University, Hefei, China
| | - Haiyan Zhang
- Core Facility Centre for Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin Pan
- National Center of Biomedical Analysis of China, Beijing, China
| | - Yucai Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Liu Liu
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jue Shi
- Center for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Guo
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Zhenye Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
6
|
Hu SY, Qian JX, Yang SY, Andriani L, Liao L, Deng L, Huang MY, Zhang YL, Zhang FL, Shao ZM, Li DQ. Destabilization of microrchidia family CW-type zinc finger 2 via the cyclin-dependent kinase 1-chaperone-mediated autophagy pathway promotes mitotic arrest and enhances cancer cellular sensitivity to microtubule-targeting agents. Clin Transl Med 2023; 13:e1210. [PMID: 36967563 PMCID: PMC10040724 DOI: 10.1002/ctm2.1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/29/2023] [Accepted: 02/15/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Microtubule-targeing agents (MTAs), such as paclitaxel (PTX) and vincristine (VCR), kill cancer cells through activtion of the spindle assembly checkpoint (SAC) and induction of mitotic arrest, but the development of resistance poses significant clinical challenges. METHODS Immunoblotting and RT-qPCR were used to investigate potential function and related mechanism of MORC2. Flow cytometry analyses were carried out to determine cell cycle distribution and apoptosis. The effect of MORC2 on cellular sensitivity to PTX and VCR was determined by immunoblotting, flow cytometry, and colony formation assays. Immunoprecipitation assays and immunofluorescent staining were utilized to investigate protein-protein interaction and protein co-localization. RESULTS Here, we identified microrchidia family CW-type zinc finger 2 (MORC2), a poorly characterized oncoprotein, as a novel regulator of SAC activation, mitotic progression, and resistance of cancer cells to PTX and VCR. Mechanically, PTX and VCR activate cyclin-dependent kinase 1, which in turn induces MORC2 phosphorylation at threonine 717 (T717) and T733. Phosphorylated MORC2 enhances its interation with HSPA8 and LAMP2A, two essential components of the chaperone-mediated autophagy (CMA) mechinery, resulting in its autophagic degradation. Degradation of MORC2 during mitosis leads to SAC activation through stabilizing anaphase promoting complex/cyclosome activator protein Cdc20 and facilitating mitotic checkpoint complex assembly, thus contributing to mitotic arrest induced by PTX and VCR. Notably, knockdown of MORC2 promotes mitotic arrest induced by PTX and VCR and enhances the sensitivity of cancer cells to PTX and VCR. CONCLUSIONS Collectively, these findings unveil a previously unrecognized function and regulatory mechanism of MORC2 in mitotic progression and resistance of cancer cells to MTAs. These results also provide a new clue for developing combined treatmentstrategy by targeting MORC2 in combination with MTAs against human cancer.
Collapse
Affiliation(s)
- Shu-Yuan Hu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Xian Qian
- Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shao-Ying Yang
- Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lisa Andriani
- Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Liao
- Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Deng
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min-Ying Huang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yin-Ling Zhang
- Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Lin Zhang
- Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Min Shao
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Da-Qiang Li
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Guo S, Lei X, Chang Y, Zhao J, Wang J, Dong X, Liu Q, Zhang Z, Wang L, Yi D, Ma L, Li Q, Zhang Y, Ding J, Liang C, Li X, Guo F, Wang J, Cen S. SARS-CoV-2 hijacks cellular kinase CDK2 to promote viral RNA synthesis. Signal Transduct Target Ther 2022; 7:400. [PMID: 36575184 PMCID: PMC9793359 DOI: 10.1038/s41392-022-01239-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/30/2022] [Accepted: 10/24/2022] [Indexed: 12/28/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has devastated global health. Identifying key host factors essential for SARS-CoV-2 RNA replication is expected to unravel cellular targets for the development of broad-spectrum antiviral drugs which have been quested for the preparedness of future viral outbreaks. Here, we have identified host proteins that associate with nonstructural protein 12 (nsp12), the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 using a mass spectrometry (MS)-based proteomic approach. Among the candidate factors, CDK2 (Cyclin-dependent kinase 2), a member of cyclin-dependent kinases, interacts with nsp12 and causes its phosphorylation at T20, thus facilitating the assembly of the RdRp complex consisting of nsp12, nsp7 and nsp8 and promoting efficient synthesis of viral RNA. The crucial role of CDK2 in viral RdRp function is further supported by our observation that CDK2 inhibitors potently impair viral RNA synthesis and SARS-CoV-2 infection. Taken together, we have discovered CDK2 as a key host factor of SARS-CoV-2 RdRp complex, thus serving a promising target for the development of SARS-CoV-2 RdRp inhibitors.
Collapse
Affiliation(s)
- Saisai Guo
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaobo Lei
- grid.506261.60000 0001 0706 7839NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan Chang
- grid.411609.b0000 0004 1758 4735Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jianyuan Zhao
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Wang
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojing Dong
- grid.506261.60000 0001 0706 7839NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qian Liu
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zixiong Zhang
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lidan Wang
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dongrong Yi
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ling Ma
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Quanjie Li
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongxin Zhang
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiwei Ding
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chen Liang
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC Canada
| | - Xiaoyu Li
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fei Guo
- grid.506261.60000 0001 0706 7839NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianwei Wang
- grid.506261.60000 0001 0706 7839NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shan Cen
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China ,grid.506261.60000 0001 0706 7839CAMS Key Laboratory of Antiviral Drug Research, Chinese Academy of Medical Sciences & Peking Union Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Roy S, Saha S, Dhar D, Chakraborty P, Singha Roy K, Mukherjee C, Gupta A, Bhattacharyya S, Roy A, Sengupta S, Roychoudhury S, Nath S. Molecular crosstalk between CUEDC2 and ERα influences the clinical outcome by regulating mitosis in breast cancer. Cancer Gene Ther 2022; 29:1697-1706. [PMID: 35732909 DOI: 10.1038/s41417-022-00494-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/13/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023]
Abstract
Development of endocrine resistance in hormone-receptor-positive (HR+ve) subtype and lack of definitive target in triple-negative subtype challenge breast cancer management. Contributing to such endocrine resistance is a protein called CUEDC2. It degrades hormone receptors, estrogen receptor-α (ERα) and progesterone receptor. Higher level of CUEDC2 in ERα+ve breast cancer corresponded to poorer disease prognosis. It additionally influences mitotic progression. However, the crosstalk of these two CUEDC2-driven functions in the outcome of breast cancer remained elusive. We showed that CUEDC2 degrades ERα during mitosis, utilising the mitotic-ubiquitination-machinery. We elucidated the importance of mitosis-specific phosphorylation of CUEDC2 in this process. Furthermore, upregulated CUEDC2 overrode mitotic arrest, increasing aneuploidy. Finally, recruiting a prospective cohort of breast cancer, we found significantly upregulated CUEDC2 in HR-ve cases. Moreover, individuals with higher CUEDC2 levels showed a poorer progression-free-survival. Together, our data confirmed that CUEDC2 up-regulation renders ERα+ve malignancies to behave essentially as HR-ve tumors with the prevalence of aneuploidy. This study finds CUEDC2 as a potential prognostic marker and a therapeutic target in the clinical management of breast cancer.
Collapse
Affiliation(s)
- Stuti Roy
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India
| | - Suryendu Saha
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India
| | - Debanil Dhar
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India
| | - Puja Chakraborty
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India
| | - Kumar Singha Roy
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | | | - Arnab Gupta
- Department of Surgery, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India
| | - Samir Bhattacharyya
- Department of Surgery, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India
| | - Anup Roy
- Department of Pathology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | | | - Susanta Roychoudhury
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India.,CSIR-Indian Institute of Chemical Biology, CN-06, CN Block, Sector V, Kolkata, India
| | - Somsubhra Nath
- Department of Basic and Translational Research, Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, India.
| |
Collapse
|
9
|
Bai S, Sun L, Wang X, Wang SM, Luo ZQ, Wang Y, Jin QW. Recovery from spindle checkpoint-mediated arrest requires a novel Dnt1-dependent APC/C activation mechanism. PLoS Genet 2022; 18:e1010397. [PMID: 36108046 PMCID: PMC9514617 DOI: 10.1371/journal.pgen.1010397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/27/2022] [Accepted: 08/24/2022] [Indexed: 11/19/2022] Open
Abstract
The activated spindle assembly checkpoint (SAC) potently inhibits the anaphase-promoting complex/cyclosome (APC/C) to ensure accurate chromosome segregation at anaphase. Early studies have recognized that the SAC should be silenced within minutes to enable rapid APC/C activation and synchronous segregation of chromosomes once all kinetochores are properly attached, but the underlying silencers are still being elucidated. Here, we report that the timely silencing of SAC in fission yeast requires dnt1+, which causes severe thiabendazole (TBZ) sensitivity and increased rate of lagging chromosomes when deleted. The absence of Dnt1 results in prolonged inhibitory binding of mitotic checkpoint complex (MCC) to APC/C and attenuated protein levels of Slp1Cdc20, consequently slows the degradation of cyclin B and securin, and eventually delays anaphase entry in cells released from SAC activation. Interestingly, Dnt1 physically associates with APC/C upon SAC activation. We propose that this association may fend off excessive and prolonged MCC binding to APC/C and help to maintain Slp1Cdc20 stability. This may allow a subset of APC/C to retain activity, which ensures rapid anaphase onset and mitotic exit once SAC is inactivated. Therefore, our study uncovered a new player in dictating the timing and efficacy of APC/C activation, which is actively required for maintaining cell viability upon recovery from the inhibition of APC/C by spindle checkpoint.
Collapse
Affiliation(s)
- Shuang Bai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Li Sun
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shuang-min Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhou-qing Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- * E-mail: (ZL); (YW); (QJ)
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- * E-mail: (ZL); (YW); (QJ)
| | - Quan-wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- * E-mail: (ZL); (YW); (QJ)
| |
Collapse
|
10
|
Deng DJ, Wang X, Yue KY, Wang Y, Jin QW. Analysis of the potential role of fission yeast PP2A in spindle assembly checkpoint inactivation. FASEB J 2022; 36:e22524. [PMID: 36006032 DOI: 10.1096/fj.202101884r] [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: 12/07/2021] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022]
Abstract
As a surveillance mechanism, the activated spindle assembly checkpoint (SAC) potently inhibits the E3 ubiquitin ligase APC/C (anaphase-promoting complex/cyclosome) to ensure accurate chromosome segregation. Although the protein phosphatase 2A (PP2A) has been proposed to be both, directly and indirectly, involved in spindle assembly checkpoint inactivation in mammalian cells, whether it is similarly operating in the fission yeast Schizosaccharomycer pombe has never been demonstrated. Here, we investigated whether fission yeast PP2A is involved in SAC silencing by following the rate of cyclin B (Cdc13) destruction at SPBs during the recovery phase in nda3-KM311 cells released from the inhibition of APC/C by the activated spindle checkpoint. The timing of the SAC inactivation is only slightly delayed when two B56 regulatory subunits (Par1 and Par2) of fission yeast PP2A are absent. Overproduction of individual PP2A subunits either globally in the nda3-KM311 arrest-and-release system or locally in the synthetic spindle checkpoint activation system only slightly suppresses the SAC silencing defects in PP1 deletion (dis2Δ) cells. Our study thus demonstrates that the fission yeast PP2A is not a key regulator actively involved in SAC inactivation.
Collapse
Affiliation(s)
- Da-Jie Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Kai-Ye Yue
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Quan-Wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
11
|
La Manna S, De Benedictis I, Marasco D. Proteomimetics of Natural Regulators of JAK-STAT Pathway: Novel Therapeutic Perspectives. Front Mol Biosci 2022; 8:792546. [PMID: 35047557 PMCID: PMC8762217 DOI: 10.3389/fmolb.2021.792546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
The JAK-STAT pathway is a crucial cellular signaling cascade, including an intricate network of Protein-protein interactions (PPIs) responsible for its regulation. It mediates the activities of several cytokines, interferons, and growth factors and transduces extracellular signals into transcriptional programs to regulate cell growth and differentiation. It is essential for the development and function of both innate and adaptive immunities, and its aberrant deregulation was highlighted in neuroinflammatory diseases and in crucial mechanisms for tumor cell recognition and tumor-induced immune escape. For its involvement in a multitude of biological processes, it can be considered a valuable target for the development of drugs even if a specific focus on possible side effects associated with its inhibition is required. Herein, we review the possibilities to target JAK-STAT by focusing on its natural inhibitors as the suppressor of cytokine signaling (SOCS) proteins. This protein family is a crucial checkpoint inhibitor in immune homeostasis and a valuable target in immunotherapeutic approaches to cancer and immune deficiency disorders.
Collapse
Affiliation(s)
| | | | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
12
|
Mitotic phosphorylation of tumor suppressor DAB2IP maintains spindle assembly checkpoint and chromosomal stability through activating PLK1-Mps1 signal pathway and stabilizing mitotic checkpoint complex. Oncogene 2022; 41:489-501. [PMID: 34775484 PMCID: PMC8782720 DOI: 10.1038/s41388-021-02106-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 11/09/2022]
Abstract
Chromosomal instability (CIN) is a driving force for cancer development. The most common causes of CIN include the dysregulation of the spindle assembly checkpoint (SAC), which is a surveillance mechanism that prevents premature chromosome separation during mitosis by targeting anaphase-promoting complex/cyclosome (APC/C). DAB2IP is frequently silenced in advanced prostate cancer (PCa) and is associated with aggressive phenotypes of PCa. Our previous study showed that DAB2IP activates PLK1 and functions in mitotic regulation. Here, we report the novel mitotic phosphorylation of DAB2IP by Cdks, which mediates DAB2IP's interaction with PLK1 and the activation of the PLK1-Mps1 pathway. DAB2IP interacts with Cdc20 in a phosphorylation-independent manner. However, the phosphorylation of DAB2IP inhibits the ubiquitylation of Cdc20 in response to SAC, and blocks the premature release of the APC/C-MCC. The PLK1-Mps1 pathway plays an important role in mitotic checkpoint complex (MCC) assembly. It is likely that DAB2IP acts as a scaffold to aid PLK1-Mps1 in targeting Cdc20. Depletion or loss of the Cdks-mediated phosphorylation of DAB2IP destabilizes the MCC, impairs the SAC, and increases chromosome missegregation and subsequent CIN, thus contributing to tumorigenesis. Collectively, these results demonstrate the mechanism of DAB2IP in SAC regulation and provide a rationale for targeting the SAC to cause lethal CIN against DAB2IP-deficient aggressive PCa, which exhibits a weak SAC.
Collapse
|
13
|
Cheng A, Jiang Y, Wang T, Yu F, Ishrat I, Zhang D, Ji X, Chen M, Xiao W, Li Q, Zhang K, Niu G, Shi J, Pan Y, Yang Z, Guo J. Energy restriction causes metaphase delay and chromosome mis-segregation in cancer cells. Cell Cycle 2021; 20:1195-1208. [PMID: 34048314 DOI: 10.1080/15384101.2021.1930679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
ATP metabolism during mitosis needs to be coordinated with numerous energy-demanding activities, especially in cancer cells whose metabolic pathways are reprogramed to sustain rapid proliferation in a nutrient-deficient environment. Although strategies targeting the energy metabolic pathways have shown therapeutic efficacy in preclinical cancer models, how normal cells and cancer cells differentially respond to energy shortage is unclear. In this study, using time-lapse microscopy, we found that cancer cells displayed unique mitotic phenotypes in a dose-dependent manner upon decreasing ATP (i.e. energy) supply. When reduction in ATP concentration was moderate, chromosome movements in mitosis were barely affected, while the metaphase-anaphase transition was significantly prolonged due to reduced tension between the sister-kinetochores, which delayed the satisfaction of the spindle assembly checkpoint. Further reduction in ATP concentration led to a decreased level of Aurora-B at the centromere, resulting in increased chromosome mis-segregation after metaphase delay. In contrast to cancer cells, ATP restriction in non-transformed cells induced cell cycle arrest in interphase, rather than causing mitotic defects. In addition, data mining of cancer patient database showed a correlation between signatures of energy production and chromosomal instability possibly resulted from mitotic defects. Together, these results reveal that energy restriction induces differential responses in normal and cancer cells, with chromosome mis-segregation only observed in cancer cells. This points to targeting energy metabolism as a potentially cancer-selective therapeutic strategy.
Collapse
Affiliation(s)
- Aoxing Cheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ya Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ting Wang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fazhi Yu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Iqra Ishrat
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dongming Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaoyang Ji
- Joint Turing-Darwin Laboratory of Phil Rivers Technology Ltd. And Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.,Department of Computational Biology, Phil Rivers Technology Ltd, Beijing, China
| | - Minhua Chen
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weihua Xiao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qing Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Gang Niu
- Joint Turing-Darwin Laboratory of Phil Rivers Technology Ltd. And Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.,Department of Computational Biology, Phil Rivers Technology Ltd, Beijing, China.,West Institute of Computing Technology, Chinese Academy of Sciences, Chongqing, China
| | - Jue Shi
- Center for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Yueyin Pan
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhenye Yang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,University of Science and Technology of China, Hefei, China
| | - Jing Guo
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
14
|
Bodrug T, Welsh KA, Hinkle M, Emanuele MJ, Brown NG. Intricate Regulatory Mechanisms of the Anaphase-Promoting Complex/Cyclosome and Its Role in Chromatin Regulation. Front Cell Dev Biol 2021; 9:687515. [PMID: 34109183 PMCID: PMC8182066 DOI: 10.3389/fcell.2021.687515] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023] Open
Abstract
The ubiquitin (Ub)-proteasome system is vital to nearly every biological process in eukaryotes. Specifically, the conjugation of Ub to target proteins by Ub ligases, such as the Anaphase-Promoting Complex/Cyclosome (APC/C), is paramount for cell cycle transitions as it leads to the irreversible destruction of cell cycle regulators by the proteasome. Through this activity, the RING Ub ligase APC/C governs mitosis, G1, and numerous aspects of neurobiology. Pioneering cryo-EM, biochemical reconstitution, and cell-based studies have illuminated many aspects of the conformational dynamics of this large, multi-subunit complex and the sophisticated regulation of APC/C function. More recent studies have revealed new mechanisms that selectively dictate APC/C activity and explore additional pathways that are controlled by APC/C-mediated ubiquitination, including an intimate relationship with chromatin regulation. These tasks go beyond the traditional cell cycle role historically ascribed to the APC/C. Here, we review these novel findings, examine the mechanistic implications of APC/C regulation, and discuss the role of the APC/C in previously unappreciated signaling pathways.
Collapse
Affiliation(s)
- Tatyana Bodrug
- Department of Biochemistry and Biophysics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kaeli A Welsh
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Megan Hinkle
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Michael J Emanuele
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Nicholas G Brown
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
15
|
Zhang L, Liu Q, Liu KW, Qin ZY, Zhu GX, Shen LT, Zhang N, Liu BY, Che LR, Li JY, Wang T, Wen LZ, Liu KJ, Guo Y, Yin XR, Wang XW, Zhou ZH, Xiao HL, Cui YH, Bian XW, Lan CH, Chen D, Wang B. SHARPIN stabilizes β-catenin through a linear ubiquitination-independent manner to support gastric tumorigenesis. Gastric Cancer 2021; 24:402-416. [PMID: 33159601 DOI: 10.1007/s10120-020-01138-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Aberrant activation of Wnt/β-catenin signaling by dysregulated post-translational protein modifications, especially ubiquitination is causally linked to cancer development and progression. Although Lys48-linked ubiquitination is known to regulate Wnt/β-catenin signaling, it remains largely obscure how other types of ubiquitination, such as linear ubiquitination governs its signaling activity. METHODS The expression and regulatory mechanism of linear ubiquitin chain assembly complex (LUBAC) on Wnt/β-catenin signaling was examined by immunoprecipitation, western blot and immunohistochemical staining. The ubiquitination status of β-catenin was detected by ubiquitination assay. The impacts of SHARPIN, a core component of LUBAC on malignant behaviors of gastric cancer cells were determined by various functional assays in vitro and in vivo. RESULTS Unlike a canonical role in promoting linear ubiquitination, SHARPIN specifically interacts with β-catenin to maintain its protein stability. Mechanistically, SHARPIN competes with the E3 ubiquitin ligase β-Trcp1 for β-catenin binding, thereby decreasing β-catenin ubiquitination levels to abolish its proteasomal degradation. Importantly, SHARPIN is required for invasiveness and malignant growth of gastric cancer cells in vitro and in vivo, a function that is largely dependent on its binding partner β-catenin. In line with these findings, elevated expression of SHARPIN in gastric cancer tissues is associated with disease malignancy and correlates with β-catenin expression levels. CONCLUSIONS Our findings reveal a novel molecular link connecting linear ubiquitination machinery and Wnt/β-catenin signaling via SHARPIN-mediated stabilization of β-catenin. Targeting the linear ubiquitination-independent function of SHARPIN could be exploited to inhibit the hyperactive β-catenin signaling in a subset of human gastric cancers.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Qin Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Ke-Wei Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Zhong-Yi Qin
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Guang-Xi Zhu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Li-Ting Shen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Ni Zhang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Bi-Ying Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Lin-Rong Che
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Jin-Yang Li
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Tao Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Liang-Zhi Wen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Kai-Jun Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Yan Guo
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Xin-Ru Yin
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Xing-Wei Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Zhi-Hua Zhou
- Department of Pathology, The 904 Hospital of People Liberation Army, Wuxi, People's Republic of China
| | - Hua-Liang Xiao
- Department of Pathology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chun-Hui Lan
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China.
| | - Dongfeng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China.
| | - Bin Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China. .,Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
16
|
Liang J, Niu Z, Zhang B, Yu X, Zheng Y, Wang C, Ren H, Wang M, Ruan B, Qin H, Zhang X, Gu S, Sai X, Tai Y, Gao L, Ma L, Chen Z, Huang H, Wang X, Sun Q. p53-dependent elimination of aneuploid mitotic offspring by entosis. Cell Death Differ 2021; 28:799-813. [PMID: 33110215 PMCID: PMC7862607 DOI: 10.1038/s41418-020-00645-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/12/2020] [Indexed: 12/31/2022] Open
Abstract
Entosis was proposed to promote aneuploidy and genome instability by cell-in-cell mediated engulfment in tumor cells. We reported here, in epithelial cells, that entosis coupled with mitotic arrest functions to counteract genome instability by targeting aneuploid mitotic progenies for engulfment and elimination. We found that the formation of cell-in-cell structures associated with prolonged mitosis, which was sufficient to induce entosis. This process was controlled by the tumor suppressor p53 (wild-type) that upregulates Rnd3 expression in response to DNA damages associated with prolonged metaphase. Rnd3-compartmentalized RhoA activities accumulated during prolonged metaphase to drive cell-in-cell formation. Remarkably, this prolonged mitosis-induced entosis selectively targets non-diploid progenies for internalization, blockade of which increased aneuploidy. Thus, our work uncovered a heretofore unrecognized mechanism of mitotic surveillance for entosis, which eliminates newly born abnormal daughter cells in a p53-dependent way, implicating in the maintenance of genome integrity.
Collapse
Affiliation(s)
- Jianqing Liang
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Zubiao Niu
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - Bo Zhang
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, 10 TIEYI Road, Beijing, 100038, China
| | - Xiaochen Yu
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - You Zheng
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - Chenxi Wang
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - He Ren
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, 10 TIEYI Road, Beijing, 100038, China
| | - Manna Wang
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
- Institute of Molecular Immunology, Southern Medical University, Guangzhou, 510515, China
| | - Banzhan Ruan
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - Hongquan Qin
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
- Institute of Molecular Immunology, Southern Medical University, Guangzhou, 510515, China
| | - Xin Zhang
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
- Department of Pediatric Hematology and Oncology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Songzhi Gu
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - Xiaoyong Sai
- National Clinic Center of Geriatric & the State Key Laboratory of Kidney, the Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanhong Tai
- The 307 Hospital, 8 Dongda Street, Beijing, 100071, China
| | - Lihua Gao
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - Li Ma
- Institute of Molecular Immunology, Southern Medical University, Guangzhou, 510515, China
| | - Zhaolie Chen
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China
| | - Hongyan Huang
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, 10 TIEYI Road, Beijing, 100038, China.
| | - Xiaoning Wang
- National Clinic Center of Geriatric & the State Key Laboratory of Kidney, the Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qiang Sun
- Laboratory of Cell Engineering, Institute of Biotechnology, 20 Dongda Street, Beijing, 100071, China.
| |
Collapse
|
17
|
Xu S, Huang S, Li D, Zou Q, Yuan Y, Yang Z. Comparison of ADAM19 and CUEDC2 expression in EHCC and their clinicopathological significance. Biomark Med 2020; 14:1573-1584. [PMID: 32960074 DOI: 10.2217/bmm-2020-0321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: To evaluate the expression and clinicopathological significance of a disintegrin and metalloproteinases 19 (ADAM19) CUE domain containing protein 2 (CUEDC2) in extrahepatic cholangiocarcinoma (EHCC). Materials & methods: Immunostaining of ADAM19 and CUEDC2 was performed by EnVision immunohistochemistry in benign and malignant biliary tract tissues. Result: The expression of ADAM19 and CUEDC2 were significantly higher in EHCC (p < 0.05). ADAM19 expression was positive correlated with CUEDC2 expression in EHCC (p < 0.05). The overall survival time of those with positive expression of ADAM19 and CUEDC2 was lower (p < 0.001). Both positive expression of ADAM19 and CUEDC2 were independent prognostic factors in EHCC. Conclusion: ADAM19 and CUEDC2 have a positive correlation to the pathogenesis and dismal prognosis in EHCC.
Collapse
Affiliation(s)
- Shu Xu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Shengfu Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Daiqiang Li
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Qiong Zou
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, PR China
| | - Yuan Yuan
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, PR China
| | - Zhulin Yang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| |
Collapse
|
18
|
VanGenderen C, Harkness TAA, Arnason TG. The role of Anaphase Promoting Complex activation, inhibition and substrates in cancer development and progression. Aging (Albany NY) 2020; 12:15818-15855. [PMID: 32805721 PMCID: PMC7467358 DOI: 10.18632/aging.103792] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
The Anaphase Promoting Complex (APC), a multi-subunit ubiquitin ligase, facilitates mitotic and G1 progression, and is now recognized to play a role in maintaining genomic stability. Many APC substrates have been observed overexpressed in multiple cancer types, such as CDC20, the Aurora A and B kinases, and Forkhead box M1 (FOXM1), suggesting APC activity is important for cell health. We performed BioGRID analyses of the APC coactivators CDC20 and CDH1, which revealed that at least 69 proteins serve as APC substrates, with 60 of them identified as playing a role in tumor promotion and 9 involved in tumor suppression. While these substrates and their association with malignancies have been studied in isolation, the possibility exists that generalized APC dysfunction could result in the inappropriate stabilization of multiple APC targets, thereby changing tumor behavior and treatment responsiveness. It is also possible that the APC itself plays a crucial role in tumorigenesis through its regulation of mitotic progression. In this review the connections between APC activity and dysregulation will be discussed with regards to cell cycle dysfunction and chromosome instability in cancer, along with the individual roles that the accumulation of various APC substrates may play in cancer progression.
Collapse
Affiliation(s)
- Cordell VanGenderen
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Troy Anthony Alan Harkness
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Terra Gayle Arnason
- Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.,Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
19
|
Shi YX, He YJ, Zhou Y, Li HK, Yang D, Li RY, Deng ZL, Gao YF. LSD1 negatively regulates autophagy in myoblast cells by driving PTEN degradation. Biochem Biophys Res Commun 2020; 522:924-930. [DOI: 10.1016/j.bbrc.2019.11.182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023]
|
20
|
Wang A, Li J, Zhou T, Li T, Cai H, Shi H, Liu A. CUEDC2 Contributes to Cisplatin-Based Chemotherapy Resistance in Ovarian Serious Carcinoma by Regulating p38 MAPK Signaling. J Cancer 2019; 10:1800-1807. [PMID: 31205536 PMCID: PMC6547988 DOI: 10.7150/jca.29889] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/07/2019] [Indexed: 12/22/2022] Open
Abstract
Chemoresistance remains an obstacle to the successful treatment of ovarian carcinoma. CUE domain-containing 2 (CUEDC2) plays critical roles in tumor genesis and overexpresses in many solid cancers, including ovarian serous carcinoma. In previous study, we found that overexpression of CUEDC2 might be a promising biomarker to evaluate the progression and to predict likely relapse of serous ovarian carcinoma. In present study, we found that higher expression of CUEDC2 was associated with higher resistance to cisplatin. The overall survival (OS) and disease-free survival time (DFS) of patients with cisplatin resistant was shorter than that of those with cisplatin sensitive, respectively, and the cisplatin sensitivity was independent predictor of a shorter OS time and DFS time. Knockdown of CUEDC2 by small interfering RNA enhanced the cisplatin sensitivity of serous ovarian carcinoma cells in SKOV3 cell lines. Furthermore, the phosphorylation of p38 MAPK were obviously increased after CUEDC2 knockdown, while p38 MAPK signaling contributes to cell growth and cell apoptosis. Our data suggest that CUEDC2 takes part in cisplatin-based chemotherapy resistance by regulating p38 MAPK signaling. And CUEDC2 is a promising biomarker and therapeutic target of cisplatin resistance in ovarian serous carcinoma.
Collapse
Affiliation(s)
- Aichun Wang
- Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China.,Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Jinhang Li
- Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Tao Zhou
- National Center of Biomedical Analysis, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Tao Li
- National Center of Biomedical Analysis, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Hong Cai
- National Center of Biomedical Analysis, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Huaiyin Shi
- Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Aijun Liu
- Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| |
Collapse
|
21
|
Targeting SOCS Proteins to Control JAK-STAT Signalling in Disease. Trends Pharmacol Sci 2019; 40:298-308. [PMID: 30948191 DOI: 10.1016/j.tips.2019.03.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/03/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
Defective regulation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling pathway in cancers, haematological diseases, and chronic inflammatory conditions highlights its clinical significance. While several biologic and small molecule therapeutics targeting this pathway have been developed, these have several limitations. Therefore, there is a need to identify new targets for intervention. Suppressor of cytokine signalling (SOCS) proteins are a family of inducible inhibitors of cytokine receptors that activate the JAK-STAT pathway. Here we propose that newly identified mechanisms controlling SOCS function could be exploited to develop molecularly targeted drugs with unique modes of action to inhibit JAK-STAT signalling in disease.
Collapse
|
22
|
AMPK-mediated activation of MCU stimulates mitochondrial Ca 2+ entry to promote mitotic progression. Nat Cell Biol 2019; 21:476-486. [PMID: 30858581 DOI: 10.1038/s41556-019-0296-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022]
Abstract
The capacity of cells to alter bioenergetics in response to the demands of various biological processes is essential for normal physiology. The coordination of energy sensing and production with highly energy-demanding cellular processes, such as cell division, is poorly understood. Here, we show that a cell cycle-dependent mitochondrial Ca2+ transient connects energy sensing to mitochondrial activity for mitotic progression. The mitochondrial Ca2+ uniporter (MCU) mediates a rapid mitochondrial Ca2+ transient during mitosis. Inhibition of mitochondrial Ca2+ transients via MCU depletion causes spindle checkpoint-dependent mitotic delay. Cellular ATP levels drop during early mitosis, and the mitochondrial Ca2+ transients boost mitochondrial respiration to restore energy homeostasis. This is achieved through mitosis-specific MCU phosphorylation and activation by the mitochondrial translocation of energy sensor AMP-activated protein kinase (AMPK). Our results establish a critical role for AMPK- and MCU-dependent mitochondrial Ca2+ signalling in mitosis and reveal a mechanism of mitochondrial metabolic adaptation to acute cellular energy stress.
Collapse
|
23
|
Hu S, Hu Z, Li S, He C, Wu Y, Teng D, Yan Y, Li H, Xing X, Zou G, Li Y, Yang Y, Wang Y, Du X. Expression of CUEDC2 in colorectal cancer with different invasion and migration abilities. J Int Med Res 2019; 47:905-914. [PMID: 30651016 PMCID: PMC6381460 DOI: 10.1177/0300060518813072] [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] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To investigate CUE domain containing 2 ( CUEDC2) expression in colorectal cancer with different invasion and migration abilities. METHODS Fresh colon cancer tissues, obtained from patients with or without lymph node metastasis who were treated at the Department of General Surgery, Chinese People's Liberation Army General Hospital, and SW620 and HT29 colorectal cancer cell lines, were analysed for CUEDC2 expression. RESULTS Real-time polymerase chain reaction showed significantly higher CUEDC2 mRNA levels in colon cancer tissue from patients with ( n = 8) versus without ( n = 8) lymph node metastasis, and in SW620 versus HT29 cells. Western blots revealed significantly higher CUEDC2 protein levels in colon cancer tissues from patients with versus without lymph node metastasis, and in SW620 versus HT29 cells. Colorectal cancer tissues from patients with lymph node metastasis showed more intense immunohistochemical staining and moderate staining of cell nuclei and cytoplasm versus less intense/weak staining in tissues from patients without lymph node metastasis. CONCLUSIONS CUEDC2 is highly expressed in colorectal cancer tissues and colorectal cancer cell lines with high invasion and migration ability. CUEDC2 may be involved in promoting invasion and metastasis in colorectal cancer.
Collapse
Affiliation(s)
- Shidong Hu
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China.,*These authors contributed equally to this work
| | - Zilong Hu
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China.,*These authors contributed equally to this work
| | - Songyan Li
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China.,*These authors contributed equally to this work
| | - Changzheng He
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Youjun Wu
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Da Teng
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yang Yan
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hao Li
- 2 Team of Aviation Medical Examination, Chinese People's Liberation Army Air Force General Hospital, Beijing, China
| | - Xiaowei Xing
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guijun Zou
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuxuan Li
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yu Yang
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yufeng Wang
- 3 Department of Hospital Management, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaohui Du
- 1 Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
24
|
Wu M, Chang Y, Hu H, Mu R, Zhang Y, Qin X, Duan X, Li W, Tu H, Zhang W, Wang G, Han Q, Li A, Zhou T, Iwai K, Zhang X, Li H. LUBAC controls chromosome alignment by targeting CENP-E to attached kinetochores. Nat Commun 2019; 10:273. [PMID: 30655516 PMCID: PMC6336796 DOI: 10.1038/s41467-018-08043-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 12/07/2018] [Indexed: 11/30/2022] Open
Abstract
Faithful chromosome segregation requires proper chromosome congression at prometaphase and dynamic maintenance of the aligned chromosomes at metaphase. Chromosome missegregation can result in aneuploidy, birth defects and cancer. The kinetochore-bound KMN network and the kinesin motor CENP-E are critical for kinetochore-microtubule attachment and chromosome stability. The linear ubiquitin chain assembly complex (LUBAC) attaches linear ubiquitin chains to substrates, with well-established roles in immune response. Here, we identify LUBAC as a key player of chromosome alignment during mitosis. LUBAC catalyzes linear ubiquitination of the kinetochore motor CENP-E, which is specifically required for the localization of CENP-E at attached kinetochores, but not unattached ones. KNL1 acts as a receptor of linear ubiquitin chains to anchor CENP-E at attached kinetochores in prometaphase and metaphase. Thus, linear ubiquitination promotes chromosome congression and dynamic chromosome alignment by coupling the dynamic kinetochore microtubule receptor CENP-E to the static one, the KMN network. During cell division, faithful chromosome segregation requires proper chromosome congression and dynamic maintenance of the aligned chromosomes. Here, the authors find that LUBAC promotes dynamic chromosome congression and alignment by targeting kinetochore motor CENP-E to the KMN network.
Collapse
Affiliation(s)
- Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Yan Chang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Huaibin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Rui Mu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yucheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Xuanhe Qin
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Xiaotao Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China
| | - Weihua Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Haiqing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Weina Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Guang Wang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Qiuying Han
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Ailing Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Xuemin Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China.
| | - Huiyan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China. .,School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
25
|
Wu QY, Zhu YY, Liu Y, Wei F, Tong YX, Cao J, Zhou P, Niu MS, Li ZY, Zeng LY, Li F, Xu KL. CUEDC2, a novel interacting partner of the SOCS1 protein, plays important roles in the leukaemogenesis of acute myeloid leukaemia. Cell Death Dis 2018; 9:774. [PMID: 29991678 PMCID: PMC6039501 DOI: 10.1038/s41419-018-0812-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/14/2018] [Accepted: 06/21/2018] [Indexed: 01/06/2023]
Abstract
Downregulation of suppressor of cytokine signalling-1 (SOCS1) is one of the vital reasons for JAK1-STAT3 pathway activation in acute myeloid leukaemia (AML). CUE domain-containing 2 (CUEDC2) was a novel interacting partner of SOCS1 and a positive correlation between the expression of CUEDC2 and SOCS1 was confirmed in primary AML cells and AML cell lines without SOCS1 promoter methylation. We aimed to explore roles of CUEDC2 in regulating ubiquitin-mediated degradation of SOCS1 in the leukaemogenesis of AML.According to in vitro experiments, CUEDC2 overexpression increased the level of SOCS1 protein, suppressed JAK1-STAT3 pathway activation. The suppression of this pathway inhibited AML cells' proliferation by causing G1 arrest and enhanced AML cells' sensitivity to cytarabine and idarubicin. Similarity, downregulation of CUEDC2 produced opposite results. Knockout or low expression of CUEDC2 in mouse or AML patients displayed lower overall survival and event-free survival rates, compared with these mouse and AML patients had high-CUEDC2 expression. Mechanistic studies revealed that CUEDC2 overexpression attenuated SOCS1 ubiquitination, facilitated its stabilisation by enhancing SOCS1, Elongin C and Cullin-2 (CUL2) interactions, thus inhibited JAK1-STAT3 pathway and leukaemogenesis of AML. Therefore, our novel findings indicated that CUEDC2 interacted with SOCS1 to suppress SOCS1's ubiquitin-mediated degradation, JAK1-STAT3 pathway activation and leukaemogenesis of AML.
Collapse
Affiliation(s)
- Qing-Yun Wu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan-Yuan Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Liu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fang Wei
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Xue Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiang Cao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ping Zhou
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ming-Shan Niu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhen-Yu Li
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ling-Yu Zeng
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Feng Li
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| | - Kai-Lin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
26
|
Liu Y, Jiang P, Wang G, Liu X, Luo S. Downregulation of RFX1 predicts poor prognosis of patients with small hepatocellular carcinoma. Eur J Surg Oncol 2018; 44:1087-1093. [PMID: 29764705 DOI: 10.1016/j.ejso.2018.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/08/2018] [Accepted: 04/18/2018] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Regulatory factor X1 (RFX1) deletion has been reported to be correlated with poor prognosis of some types of cancer. The present study aimed to investigate the prognostic value of RFX1 in HCC, especially in small hepatocellular carcinoma. METHODS Immunohistochemical assay was used to investigate RFX1 expression in 221 HCC tissues and another validation cohort of 71 small HCC samples. We also performed in vitro experiments to investigate if RFX1 regulated invasive capacity of HCC cells and expression of epithelial-mesenchymal transition (EMT) markers. RESULTS We found that RFX1 expression was significantly lower in HCC tissues compared to the corresponding non-tumor tissues. Further survival analysis suggested that the downregulation of RFX1 correlated with poor prognosis and a high recurrence risk in HCC patients, particularly in small HCC patients. Furthermore, another validation cohort of small HCC samples confirmed that downregulation of RFX1 in HCC tissues predicted high recurrence risk and poor prognosis for early stage HCC patients. In vitro studies suggested that knocking down RFX1 facilitated HCC cell invasion, while overexpression of RFX1 reduced the invasion of HCC cells. Western blot assays also indicated that RFX1 regulated expression of some EMT markers. Knocking down RFX1 decreased E-cadherin and increased vimentin expression, while RFX1 overexpression enhanced E-cadherin and decreased vimentin expression. CONCLUSIONS Our study demonstrated that RFX1 downregulation is a new predictive marker of high recurrence risk and poor prognosis of HCC; It has potential to help guide treatment for postoperative HCC patients, especially for small HCC patients.
Collapse
Affiliation(s)
- Yingjun Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Peng Jiang
- Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Gangcheng Wang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xiaonyong Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Suxia Luo
- Department of Internal Medicine, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
27
|
Jin BF, Yang F, Ying XM, Gong L, Hu SF, Zhao Q, Liao YD, Chen KZ, Li T, Tai YH, Cao Y, Li X, Huang Y, Zhan XY, Qin XH, Wu J, Chen S, Guo SS, Zhang YC, Chen J, Shen DH, Sun KK, Chen L, Li WH, Li AL, Wang N, Xia Q, Wang J, Zhou T. Signaling protein signature predicts clinical outcome of non-small-cell lung cancer. BMC Cancer 2018; 18:259. [PMID: 29510676 PMCID: PMC5840771 DOI: 10.1186/s12885-018-4104-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 02/06/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) is characterized by abnormalities of numerous signaling proteins that play pivotal roles in cancer development and progression. Many of these proteins have been reported to be correlated with clinical outcomes of NSCLC. However, none of them could provide adequate accuracy of prognosis prediction in clinical application. METHODS A total of 384 resected NSCLC specimens from two hospitals in Beijing (BJ) and Chongqing (CQ) were collected. Using immunohistochemistry (IHC) staining on stored formalin-fixed paraffin-embedded (FFPE) surgical samples, we examined the expression levels of 75 critical proteins on BJ samples. Random forest algorithm (RFA) and support vector machines (SVM) computation were applied to identify protein signatures on 2/3 randomly assigned BJ samples. The identified signatures were tested on the remaining BJ samples, and were further validated with CQ independent cohort. RESULTS A 6-protein signature for adenocarcinoma (ADC) and a 5-protein signature for squamous cell carcinoma (SCC) were identified from training sets and tested in testing sets. In independent validation with CQ cohort, patients can also be divided into high- and low-risk groups with significantly different median overall survivals by Kaplan-Meier analysis, both in ADC (31 months vs. 87 months, HR 2.81; P < 0.001) and SCC patients (27 months vs. not reached, HR 9.97; P < 0.001). Cox regression analysis showed that both signatures are independent prognostic indicators and outperformed TNM staging (ADC: adjusted HR 3.07 vs. 2.43, SCC: adjusted HR 7.84 vs. 2.24). Particularly, we found that only the ADC patients in high-risk group significantly benefited from adjuvant chemotherapy (P = 0.018). CONCLUSIONS Both ADC and SCC protein signatures could effectively stratify the prognosis of NSCLC patients, and may support patient selection for adjuvant chemotherapy.
Collapse
Affiliation(s)
- Bao-Feng Jin
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Fan Yang
- Department of Thoracic Surgery, People’s Hospital, Peking University, Beijing, 100044 China
| | - Xiao-Min Ying
- Computational Medicine Laboratory, Beijing Institute of Basic Medical Sciences, Beijing, 100850 China
| | - Lin Gong
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Shuo-Feng Hu
- Computational Medicine Laboratory, Beijing Institute of Basic Medical Sciences, Beijing, 100850 China
| | - Qing Zhao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Yi-Da Liao
- Department of Thoracic Surgery, People’s Hospital, Peking University, Beijing, 100044 China
| | - Ke-Zhong Chen
- Department of Thoracic Surgery, People’s Hospital, Peking University, Beijing, 100044 China
| | - Teng Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Yan-Hong Tai
- The 90th Hospital of Jinan, Jinan, 250031 China
- Department of Pathology, The 307th Hospital of Chinese PLA, Beijing, 100071 China
| | - Yuan Cao
- The 90th Hospital of Jinan, Jinan, 250031 China
| | - Xiao Li
- Department of Thoracic Surgery, People’s Hospital, Peking University, Beijing, 100044 China
| | - Yan Huang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Xiao-Yan Zhan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Xuan-He Qin
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Jin Wu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Shuai Chen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Sai-Sai Guo
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Yu-Cheng Zhang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Jing Chen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Dan-Hua Shen
- Department of Pathology, People’s Hospital, Peking University, Beijing, 100044 China
| | - Kun-Kun Sun
- Department of Pathology, People’s Hospital, Peking University, Beijing, 100044 China
| | - Lu Chen
- Institute of Pathology, Southwest Cancer Center, Southwest Hospital, Chongqing, 400038 China
| | - Wei-Hua Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Na Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Qing Xia
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| | - Jun Wang
- Department of Thoracic Surgery, People’s Hospital, Peking University, Beijing, 100044 China
| | - Tao Zhou
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing, 100850 China
| |
Collapse
|
28
|
Wei R, Li B, Guo J, Li M, Zhu R, Yang X, Gao R. Smurf1 targets Securin for ubiquitin-dependent degradation and regulates the metaphase-to-anaphase transition. Cell Signal 2017; 38:60-66. [DOI: 10.1016/j.cellsig.2017.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 11/25/2022]
|
29
|
Gilberto S, Peter M. Dynamic ubiquitin signaling in cell cycle regulation. J Cell Biol 2017; 216:2259-2271. [PMID: 28684425 PMCID: PMC5551716 DOI: 10.1083/jcb.201703170] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/11/2017] [Accepted: 05/25/2017] [Indexed: 12/16/2022] Open
Abstract
Gilberto and Peter discuss the role of ubiquitylation in the regulation of DNA replication and mitosis. The cell division cycle is driven by a collection of enzymes that coordinate DNA duplication and separation, ensuring that genomic information is faithfully and perpetually maintained. The activity of the effector proteins that perform and coordinate these biological processes oscillates by regulated expression and/or posttranslational modifications. Ubiquitylation is a cardinal cellular modification and is long known for driving cell cycle transitions. In this review, we emphasize emerging concepts of how ubiquitylation brings the necessary dynamicity and plasticity that underlie the processes of DNA replication and mitosis. New studies, often focusing on the regulation of chromosomal proteins like DNA polymerases or kinetochore kinases, are demonstrating that ubiquitylation is a versatile modification that can be used to fine-tune these cell cycle events, frequently through processes that do not involve proteasomal degradation. Understanding how the increasing variety of identified ubiquitin signals are transduced will allow us to develop a deeper mechanistic perception of how the multiple factors come together to faithfully propagate genomic information. Here, we discuss these and additional conceptual challenges that are currently under study toward understanding how ubiquitin governs cell cycle regulation.
Collapse
Affiliation(s)
- Samuel Gilberto
- Department of Biology, Institute of Biochemistry, Swiss Federal Institute of Technology, Zurich, Switzerland.,Molecular Life Science PhD Program, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Matthias Peter
- Department of Biology, Institute of Biochemistry, Swiss Federal Institute of Technology, Zurich, Switzerland
| |
Collapse
|
30
|
Shin J, Mishra V, Glasgow E, Zaidi S, Ohshiro K, Chitti B, Kapadia AA, Rana N, Mishra L, Deng CX, Rao S, Mishra B. PRAJA is overexpressed in glioblastoma and contributes to neural precursor development. Genes Cancer 2017; 8:640-649. [PMID: 28966725 PMCID: PMC5620009 DOI: 10.18632/genesandcancer.151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022] Open
Abstract
PRAJA, a RING-H2 E3 ligase, is abundantly expressed in brain tissues such as the cerebellum and frontal cortex, amongst others, and more specifically in neural progenitor cells as well as in multiple cancers that include glioblastomas. However, the specific role that Praja plays in neural development and gliomas remains unclear. In this investigation, we performed bioinformatic analyses to examine Praja1 and Praja2 expression across 29 cancer types, and observed raised levels of Praja1 and Praja2 in gliomas with an inverse relationship between Praja1 and apoptotic genes and Praja substrates such as Smad3. We analyzed the role of Praja in the developing brain through loss of function studies, using morpholinos targeting Praja1 in embryonic zebrafish, and observed that Praja1 is expressed prominently in regions enriched with neural precursor cell subtypes. Antisense Praja morpholinos resulted in multiple embryonic defects including delayed neural development likely through increased apoptosis. Further studies revealed high levels of Cdk1 with loss of Praja1 in TGF-β or insulin treated cells, supporting the link between Praja1 and cell cycle regulation. In summary, these studies underscore Praja's role in mammalian brain development and Praja1 deregulation may lead to gliomas possibly through the regulation of cell cycle and/or apoptosis.
Collapse
Affiliation(s)
- Joshua Shin
- University of Virginia, Charlottesville, VA, USA
| | - Viveka Mishra
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric Glasgow
- Department of Molecular Oncology, Georgetown University, Washington DC, USA
| | - Sobia Zaidi
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC, USA
| | - Kazufumi Ohshiro
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC, USA
| | - Bhargava Chitti
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Amee A. Kapadia
- John Hopkins University, Department of Chemical and Biomolecular Engineering, Baltimore, MD, USA
| | | | - Lopa Mishra
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC, USA
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Shuyun Rao
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC, USA
| | - Bibhuti Mishra
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC, USA
| |
Collapse
|
31
|
Li F, Tang C, Jin D, Guan L, Wu Y, Liu X, Wu X, Wu QY, Gao D. CUEDC2 suppresses glioma tumorigenicity by inhibiting the activation of STAT3 and NF-κB signaling pathway. Int J Oncol 2017; 51:115-127. [PMID: 28534933 PMCID: PMC5467786 DOI: 10.3892/ijo.2017.4009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/27/2017] [Indexed: 12/23/2022] Open
Abstract
CUEDC2, a CUE domain containing 2 protein, plays critical roles in many biological processes, such as cell cycle, inflammation and tumorigenesis. However, whether CUEDC2 was involved in tumorigenesis of glioma and the possible mechanism remains to be elucidated. In the present study, our results implied that the expression of CUEDC2 was lower in the glioma tissue and glioma cell lines than that of normal tissue and asctrocyte cells. Downregulation of endogenous CUEDC2 in glioma U251 cell lines by RNAi promoted the tumor cells proliferation, migration, invasion and glioma neurosphere formation, while, overexpression of CUEDC2 showed the opposite effect. Further studies showed that overexpression of CUEDC2 suppressed the activation and nuclear translocation of phosphorylated-STAT3 (p-STAT3) but the level of p-STAT3 increased after interfering with the expression of CUEDC2. Moreover, CUEDC2 expression has an inhibitory effect on the activation of NF-κB. Thus, our studies suggested that the decreased expression of CUEDC2 in glioma led to the activation of transcription factor STAT3 and NF-κB signaling pathway which may be related to the tumorigenicity in glioma.
Collapse
Affiliation(s)
- Feng Li
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Chuanxi Tang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Dan Jin
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Li Guan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Yue Wu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xinfeng Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xiuxiang Wu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Qing Yun Wu
- Laboratory of Transplantation and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Dianshuai Gao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
32
|
Wang JC, Zhu Y, Wu L, Dong E. Progress in Pharmacological Sciences in China. Mol Pharmacol 2017; 92:188-192. [PMID: 28404616 DOI: 10.1124/mol.116.108167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 04/04/2017] [Indexed: 11/22/2022] Open
Abstract
Pharmacology is the science that investigates the interactions between organisms and drugs and their mechanisms. Pharmacology plays a translational role in modern medicine, bridging basic research and the clinic. With its economy booming, China has invested an enormous amount of financial and human resources in pharmacological research in the recent decade. As a result, major breakthroughs have been achieved in both basic and clinical research, with the discovery of many potential drug targets and biomarkers that has made a sizable contribution to the overall advancement of pharmacological sciences. In this article, we review recent research efforts and representative scientific achievements and discuss future challenges and directions for the pharmacological sciences in China.
Collapse
Affiliation(s)
- Jian-Cheng Wang
- Department of Health Sciences, National Natural Science Foundation of China (J.-C.W., Y.Z., L.W., E.D.) and School of Pharmaceutical Sciences, Peking University (J.-C.W.), Beijing, China
| | - Yuangui Zhu
- Department of Health Sciences, National Natural Science Foundation of China (J.-C.W., Y.Z., L.W., E.D.) and School of Pharmaceutical Sciences, Peking University (J.-C.W.), Beijing, China
| | - Lei Wu
- Department of Health Sciences, National Natural Science Foundation of China (J.-C.W., Y.Z., L.W., E.D.) and School of Pharmaceutical Sciences, Peking University (J.-C.W.), Beijing, China
| | - Erdan Dong
- Department of Health Sciences, National Natural Science Foundation of China (J.-C.W., Y.Z., L.W., E.D.) and School of Pharmaceutical Sciences, Peking University (J.-C.W.), Beijing, China
| |
Collapse
|
33
|
Wei R, Guo J, Li M, Yang X, Zhu R, Huang H, Li K, Zhang L, Gao R. Smurf1 controls S phase progression and tumorigenesis through Wee1 degradation. FEBS Lett 2017; 591:1150-1158. [DOI: 10.1002/1873-3468.12624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Rongfei Wei
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Jing Guo
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
- Department of Inorganic Non-metallic Materials; School of Materials Science and Engineering; University of Science and Technology Beijing; China
| | - Mengyuan Li
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Xingjiu Yang
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Ruimin Zhu
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Hao Huang
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Kejuan Li
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics; Beijing Proteome Research Center; Beijing Institute of Radiation Medicine; Collaborative Innovation Center for Cancer Medicine; Beijing China
| | - Ran Gao
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| |
Collapse
|
34
|
Zhong X, Tian S, Zhang X, Diao X, Dong F, Yang J, Li Z, Sun L, Wang L, He X, Wu G, Hu X, Wang L, Song L, Zhang H, Pan X, Li A, Gao P. CUE domain-containing protein 2 promotes the Warburg effect and tumorigenesis. EMBO Rep 2017; 18:809-825. [PMID: 28325773 DOI: 10.15252/embr.201643617] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/05/2017] [Accepted: 02/15/2017] [Indexed: 12/12/2022] Open
Abstract
Cancer progression depends on cellular metabolic reprogramming as both direct and indirect consequence of oncogenic lesions; however, the underlying mechanisms are still poorly understood. Here, we report that CUEDC2 (CUE domain-containing protein 2) plays a vital role in facilitating aerobic glycolysis, or Warburg effect, in cancer cells. Mechanistically, we show that CUEDC2 upregulates the two key glycolytic proteins GLUT3 and LDHA via interacting with the glucocorticoid receptor (GR) or 14-3-3ζ, respectively. We further demonstrate that enhanced aerobic glycolysis is essential for the role of CUEDC2 to drive cancer progression. Moreover, using tissue microarray analysis, we show a correlation between the aberrant expression of CUEDC2, and GLUT3 and LDHA in clinical HCC samples, further demonstrating a link between CUEDC2 and the Warburg effect during cancer development. Taken together, our findings reveal a previously unappreciated function of CUEDC2 in cancer cell metabolism and tumorigenesis, illustrating how close oncogenic lesions are intertwined with metabolic alterations promoting cancer progression.
Collapse
Affiliation(s)
- Xiuying Zhong
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Shengya Tian
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xiang Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xinwei Diao
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Fangting Dong
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Jie Yang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Zhaoyong Li
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Linchong Sun
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Lin Wang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xiaoping He
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Gongwei Wu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xin Hu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Lihua Wang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Libing Song
- State Key Laboratory of Oncology in Southern China and Departments of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xin Pan
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Ailing Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Ping Gao
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
35
|
Yang X, Chen G, Li W, Peng C, Zhu Y, Yang X, Li T, Cao C, Pei H. Cervical Cancer Growth Is Regulated by a c-ABL-PLK1 Signaling Axis. Cancer Res 2017; 77:1142-1154. [PMID: 27899378 DOI: 10.1158/0008-5472.can-16-1378] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 10/15/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022]
Abstract
The nonreceptor tyrosine kinase c-ABL controls cell growth but its contributions in solid tumors are not fully understood. Here we report that the Polo-like kinase PLK1, an essential mitotic kinase regulator, is an important downstream effector of c-ABL in regulating the growth of cervical cancer. c-ABL interacted with and phosphorylated PLK1. Phosphorylation of PLK1 by c-ABL inhibited PLK1 ubiquitination and degradation and enhanced its activity, leading to cell-cycle progression and tumor growth. Both c-ABL and PLK1 were overexpressed in cervical carcinoma. Notably, PLK1 tyrosine phosphorylation correlated with patient survival in cervical cancer. In a murine xenograft model of human cervical cancer, combination treatment with c-ABL and PLK1 inhibitors yielded additive effects on tumor growth inhibition. Our findings highlight the c-ABL-PLK1 axis as a novel prognostic marker and treatment target for human cervical cancers. Cancer Res; 77(5); 1142-54. ©2016 AACR.
Collapse
Affiliation(s)
- Xu Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Gang Chen
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Beijing Institute of Biotechnology, Haidian District, Beijing, China
- Laboratory of Nuclear and Radiation Damage, The General Hospital of the PLA Rocket Force, Beijing, China
| | - Changmin Peng
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Industrial Microbiology Key Lab, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yue Zhu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiaoming Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Teng Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.
| | - Cheng Cao
- Beijing Institute of Biotechnology, Haidian District, Beijing, China.
| | - Huadong Pei
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China.
| |
Collapse
|
36
|
Aurora-A promotes the establishment of spindle assembly checkpoint by priming the Haspin-Aurora-B feedback loop in late G2 phase. Cell Discov 2017; 3:16049. [PMID: 28101375 PMCID: PMC5223110 DOI: 10.1038/celldisc.2016.49] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 11/22/2016] [Indexed: 12/11/2022] Open
Abstract
Aurora-A kinase functions mainly in centrosome maturation, separation and spindle formation. It has also been found to be amplified or overexpressed in a range of solid tumors, which is linked with tumor progression and poor prognosis. Importantly, Aurora-A inhibitors are being studied in a number of ongoing clinical trials. However, whether and how Aurora-A has a role in the regulation of the mitotic checkpoint is controversial. Additionally, the function of nuclear-accumulated Aurora-A in late G2 phase is not clear. Here we show that knockout, inhibition or blockade of the nuclear entry of Aurora-A severely decreased the centromere localization of Aurora-B and the phosphorylation of histone H3 threonine 3 (H3T3-ph) mediated by the kinase Haspin in late G2 phase. We further reveal that nuclear-accumulated Aurora-A phosphorylates Haspin at multiple sites at its N-terminus and that this promotes H3T3-ph and the rapid recruitment to the centromere of the chromosomal passenger complex. In addition, Aurora-A facilitates the association of Aurora-B with their common substrates: Haspin and Plk1. Notably, these functions of Aurora-A are mostly independent of Plk1. Thus we demonstrate that, in late G2 and prophase, Aurora-A phosphorylates Haspin to trigger the Haspin-H3T3-ph-Aurora-B positive feedback loop that supports the timely establishment of the chromosomal passenger complex and the mitotic checkpoint before spindle assembly.
Collapse
|
37
|
Abstract
The mitotic checkpoint is a specialized signal transduction pathway that contributes to the fidelity of chromosome segregation. The signaling of the checkpoint originates from defective kinetochore-microtubule interactions and leads to formation of the mitotic checkpoint complex (MCC), a highly potent inhibitor of the Anaphase Promoting Complex/Cyclosome (APC/C)—the E3 ubiquitin ligase essential for anaphase onset. Many important questions concerning the MCC and its interaction with APC/C have been intensively investigated and debated in the past 15 years, such as the exact composition of the MCC, how it is assembled during a cell cycle, how it inhibits APC/C, and how the MCC is disassembled to allow APC/C activation. These efforts have culminated in recently reported structure models for human MCC:APC/C supra-complexes at near-atomic resolution that shed light on multiple aspects of the mitotic checkpoint mechanisms. However, confusing statements regarding the MCC are still scattered in the literature, making it difficult for students and scientists alike to obtain a clear picture of MCC composition, structure, function and dynamics. This review will comb through some of the most popular concepts or misconceptions about the MCC, discuss our current understandings, present a synthesized model on regulation of CDC20 ubiquitination, and suggest a few future endeavors and cautions for next phase of MCC research.
Collapse
Affiliation(s)
- Song-Tao Liu
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Hang Zhang
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| |
Collapse
|
38
|
SUMOylated NKAP is essential for chromosome alignment by anchoring CENP-E to kinetochores. Nat Commun 2016; 7:12969. [PMID: 27694884 PMCID: PMC5064014 DOI: 10.1038/ncomms12969] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 08/19/2016] [Indexed: 01/29/2023] Open
Abstract
Chromosome alignment is required for accurate chromosome segregation. Chromosome misalignment can result in genomic instability and tumorigenesis. Here, we show that NF-κB activating protein (NKAP) is critical for chromosome alignment through anchoring CENP-E to kinetochores. NKAP knockdown causes chromosome misalignment and prometaphase arrest in human cells. NKAP dynamically localizes to kinetochores, and is required for CENP-E kinetochore localization. NKAP is SUMOylated predominantly in mitosis and the SUMOylation is needed for NKAP to bind CENP-E. A SUMOylation-deficient mutant of NKAP cannot support the localization of CENP-E on kinetochores or proper chromosome alignment. Moreover, Bub3 recruits NKAP to stabilize the binding of CENP-E to BubR1 at kinetochores. Importantly, loss of NKAP expression causes aneuploidy in cultured cells, and is observed in human soft tissue sarcomas. These findings indicate that NKAP is a novel and key regulator of mitosis, and its dysregulation might contribute to tumorigenesis by causing chromosomal instability. The kinetochore-bound motor CENP-E plays a critical role in chromosome alignment. Here, the authors show that NF-κB activating protein (NKAP) dynamically localises to kinetochores, is SUMOylated during mitosis, and this modification is required for NKAP to bind CENP-E and localise CENP-E to the kinetochore.
Collapse
|
39
|
CUEDC2 down-regulation is associated with tumor growth and poor prognosis in lung adenocarcinoma. Oncotarget 2016; 6:20685-96. [PMID: 26023733 PMCID: PMC4653035 DOI: 10.18632/oncotarget.3930] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 04/30/2015] [Indexed: 12/24/2022] Open
Abstract
CUE domain-containing 2 (CUEDC2) is a multi-functional protein, which regulates cell cycle, growth factor signaling and inflammation. We found that CUEDC2 was low in lung adenocarcinoma cell lines and lung adenocarcinoma tissues at both mRNA and protein levels. Low levels of CUEDC2 were correlated with a shorter survival time in patients with lung adenocarcinoma (p = 0.004). CUEDC2 expression was correlated with tumor T classification (P = 0.001) at clinical stage (P = 0.001) and tumor size (P = 0.033). Multivariate analysis suggested that CUEDC2 expression is an independent prognostic indicator for patients with lung adenocarcinoma. Ectopic expression of CUEDC2 decreased cell proliferation in vitro and inhibited tumor growth in nude mice in vivo. Knockdown of endogenous CUEDC2 by short hairpin RNAs (shRNAs) increased tumor growth. Inhibition of proliferation by CUEDC2 was associated with inactivation of the PI3K/Akt pathway, induction of p21 and down-regulation of cyclin D1. Our results suggest that decreased expression of CUEDC2 contributes to tumor growth in lung adenocarcinoma, leading to a poor clinical outcome.
Collapse
|
40
|
Jian Z, Liang B, Pan X, Xu G, Guo SS, Li T, Zhou T, Xiao YB, Li AL. CUEDC2 modulates cardiomyocyte oxidative capacity by regulating GPX1 stability. EMBO Mol Med 2016; 8:813-29. [PMID: 27286733 PMCID: PMC4931293 DOI: 10.15252/emmm.201506010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The irreversible loss of cardiomyocytes due to oxidative stress is the main cause of heart dysfunction following ischemia/reperfusion (I/R) injury and ageing-induced cardiomyopathy. Here, we report that CUEDC2, a CUE domain-containing protein, plays a critical role in oxidative stress-induced cardiac injury. Cuedc2(-/-) cardiomyocytes exhibited a greater resistance to oxidative stress-induced cell death. Loss of CUEDC2 enhanced the antioxidant capacity of cardiomyocytes, promoted reactive oxygen species (ROS) scavenging, and subsequently inhibited the redox-dependent activation of signaling pathways. Notably, CUEDC2 promoted E3 ubiquitin ligases tripartite motif-containing 33 (TRIM33)-mediated the antioxidant enzyme, glutathione peroxidase 1 (GPX1) ubiquitination, and proteasome-dependent degradation. Ablation of CUEDC2 upregulated the protein level of GPX1 in the heart significantly. Strikingly, in vivo, the infarct size of Cuedc2(-/-) heart was significantly decreased after I/R injury, and aged Cuedc2(-/-) mice preserved better heart function as the overall ROS levels in their hearts were significantly lower. Our results demonstrated a novel role of CUEDC2 in cardiomyocyte death regulation. Manipulating CUEDC2 level might be an attractive therapeutic strategy for promoting cardiomyocyte survival following oxidative stress-induced cardiac injury.
Collapse
Affiliation(s)
- Zhao Jian
- Institute of Cardiovascular Surgery, Xinqiao Hospital Third Military Medical University, Chongqing, China
| | - Bing Liang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| | - Xin Pan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| | - Guang Xu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| | - Sai-Sai Guo
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| | - Ting Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| | - Ying-Bin Xiao
- Institute of Cardiovascular Surgery, Xinqiao Hospital Third Military Medical University, Chongqing, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences National Center of Biomedical Analysis, Beijing, China
| |
Collapse
|
41
|
Wang YL, Chen H, Zhan YQ, Yin RH, Li CY, Ge CH, Yu M, Yang XM. EWSR1 regulates mitosis by dynamically influencing microtubule acetylation. Cell Cycle 2016; 15:2202-2215. [PMID: 27341063 DOI: 10.1080/15384101.2016.1200774] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
EWSR1, participating in transcription and splicing, has been identified as a translocation partner for various transcription factors, resulting in translocation, which in turn plays crucial roles in tumorigenesis. Recent studies have investigated the role of EWSR1 in mitosis. However, the effect of EWSR1 on mitosis is poorly understood. Here, we observed that depletion of EWSR1 resulted in cell cycle arrest in the mitotic phase, mainly due to an increase in the time from nuclear envelope breakdown to metaphase, resulting in a high percentage of unaligned chromosomes and multipolar spindles. We also demonstrated that EWSR1 is a spindle-associated protein that interacts with α-tubulin during mitosis. EWSR1 depletion increased the cold-sensitivity of spindle microtubules, and decreased the rate of spindle assembly. EWSR1 regulated the level of microtubule acetylation in the mitotic spindle; microtubule acetylation was rescued in EWSR1-depleted mitotic cells following suppression of HDAC6 activity by its specific inhibitor or siRNA treatment. In summary, these results suggest that EWSR1 regulates the acetylation of microtubules in a cell cycle-dependent manner through its dynamic location on spindle MTs, and may be a novel regulator for mitosis progress independent of its translocation.
Collapse
Affiliation(s)
- Yi-Long Wang
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Hui Chen
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Yi-Qun Zhan
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Rong-Hua Yin
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Chang-Yan Li
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Chang-Hui Ge
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Miao Yu
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| | - Xiao-Ming Yang
- a State Key Laboratory of Proteomics , Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , China
| |
Collapse
|
42
|
Abstract
The segregation of sister chromatids during mitosis is one of the most easily visualized, yet most remarkable, events during the life cycle of a cell. The accuracy of this process is essential to maintain ploidy during cell duplication. Over the past 20 years, substantial progress has been made in identifying components of both the kinetochore and the mitotic spindle that generate the force to move mitotic chromosomes. Additionally, we now have a reasonable, albeit incomplete, understanding of the molecular and biochemical events that are involved in establishing and dissolving sister-chromatid cohesion. However, it is less well-understood how this dissolution of cohesion occurs synchronously on all chromosomes at the onset of anaphase. At the centre of the action is the anaphase-promoting complex/cyclosome (APC/C), an E3 ubiquitin ligase that, in association with its activator cell-division cycle protein 20 homologue (Cdc20), is responsible for the destruction of securin. This leads to the activation of separase, a specialized protease that cleaves the kleisin-subunit of the cohesin complex, to relieve cohesion between sister chromatids. APC/C-Cdc20 is also responsible for the destruction of cyclin B and therefore inactivation of the cyclin B-cyclin-dependent kinase 1 (Cdk1). This latter event induces a change in the microtubule dynamics that results in the movement of sister chromatids to spindle poles (anaphase A), spindle elongation (anaphase B) and the onset of cytokinesis. In the present paper, we review the emerging evidence that multiple, spatially and temporally regulated feedback loops ensure anaphase onset is rapid, co-ordinated and irreversible.
Collapse
|
43
|
Wang A, Guo C, Sun Y, Lu L, Wang Y, Wang Q, Zhang Y, Zhang H, Wang L, Gu Y, Liu A. Overexpression of CUEDC2 Predicts Poor Prognosis in Ovarian Serous Carcinomas. J Cancer 2015; 6:542-7. [PMID: 26000046 PMCID: PMC4439940 DOI: 10.7150/jca.11420] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/22/2015] [Indexed: 12/22/2022] Open
Abstract
CUEDC2, a newly reported protein, plays critical roles in many biological processes, such as cell cycle, inflammation and tumorigenesis, however, its expression in ovarian serious carcinoma is still poorly understood. In this study, we performed an immunohistochemical study on 101 cases of ovarian serous carcinoma tissues to investigate whether CUEDC2 is a useful biomarker to evaluate the progression of ovarian serous carcinomas. The data showed that the overexpression of CUEDC2 was observed in 59.4% of ovarian serous carcinoma tissue samples and correlated with histopathological grade, patient age at diagnosis, FIGO stage and recurrence. To assess the clinical relevance of CUEDC2, we analyzed the survival follow-up information, the results showed that CUEDC2-positive expression was associated with a shorter disease-free survival time, the median disease-free survival time of CUEDC2-positive patients was 36.0 months compared with 53.9 months of CUEDC2-negative ones (Log-rank χ2=6.149, P=0.013). Collectively, our results suggested that CUEDC2 may be a promising biomarker to evaluate the progression of serous ovarian carcinoma and to predict likely relapse of ovarian serous carcinoma.
Collapse
Affiliation(s)
- Aichun Wang
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China ; 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Chao Guo
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yunfei Sun
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Lijuan Lu
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Yun Wang
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Qiong Wang
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yan Zhang
- 3. Department of Obsteristics and Gynecology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Hui Zhang
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Li Wang
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Yiqun Gu
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Aijun Liu
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| |
Collapse
|
44
|
Sivakumar S, Gorbsky GJ. Spatiotemporal regulation of the anaphase-promoting complex in mitosis. Nat Rev Mol Cell Biol 2015; 16:82-94. [PMID: 25604195 DOI: 10.1038/nrm3934] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The appropriate timing of events that lead to chromosome segregation during mitosis and cytokinesis is essential to prevent aneuploidy, and defects in these processes can contribute to tumorigenesis. Key mitotic regulators are controlled through ubiquitylation and proteasome-mediated degradation. The APC/C (anaphase-promoting complex; also known as the cyclosome) is an E3 ubiquitin ligase that has a crucial function in the regulation of the mitotic cell cycle, particularly at the onset of anaphase and during mitotic exit. Co-activator proteins, inhibitor proteins, protein kinases and phosphatases interact with the APC/C to temporally and spatially control its activity and thus ensure accurate timing of mitotic events.
Collapse
Affiliation(s)
- Sushama Sivakumar
- Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, Oklahoma 73104, USA
| | - Gary J Gorbsky
- Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
45
|
Varetti G, Pellman D, Gordon DJ. Aurea mediocritas: the importance of a balanced genome. Cold Spring Harb Perspect Biol 2014; 6:a015842. [PMID: 25237130 DOI: 10.1101/cshperspect.a015842] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Aneuploidy, defined as an abnormal number of chromosomes, is a hallmark of cancer. Paradoxically, aneuploidy generally has a negative impact on cell growth and fitness in nontransformed cells. In this work, we review recent progress in identifying how aneuploidy leads to genomic and chromosomal instability, how cells can adapt to the deleterious effects of aneuploidy, and how aneuploidy contributes to tumorigenesis in different genetic contexts. Finally, we also discuss how aneuploidy might be a target for anticancer therapies.
Collapse
Affiliation(s)
- Gianluca Varetti
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - David Pellman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115 Howard Hughes Medical Institute, Chevy Chase, Maryland 20815-6789
| | - David J Gordon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| |
Collapse
|
46
|
Wang K, Sturt-Gillespie B, Hittle JC, Macdonald D, Chan GK, Yen TJ, Liu ST. Thyroid hormone receptor interacting protein 13 (TRIP13) AAA-ATPase is a novel mitotic checkpoint-silencing protein. J Biol Chem 2014; 289:23928-37. [PMID: 25012665 DOI: 10.1074/jbc.m114.585315] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The mitotic checkpoint (or spindle assembly checkpoint) is a fail-safe mechanism to prevent chromosome missegregation by delaying anaphase onset in the presence of defective kinetochore-microtubule attachment. The target of the checkpoint is the E3 ubiquitin ligase anaphase-promoting complex/cyclosome. Once all chromosomes are properly attached and bioriented at the metaphase plate, the checkpoint needs to be silenced. Previously, we and others have reported that TRIP13 AAA-ATPase binds to the mitotic checkpoint-silencing protein p31(comet). Here we show that endogenous TRIP13 localizes to kinetochores. TRIP13 knockdown delays metaphase-to-anaphase transition. The delay is caused by prolonged presence of the effector for the checkpoint, the mitotic checkpoint complex, and its association and inhibition of the anaphase-promoting complex/cyclosome. These results suggest that TRIP13 is a novel mitotic checkpoint-silencing protein. The ATPase activity of TRIP13 is essential for its checkpoint function, and interference with TRIP13 abolished p31(comet)-mediated mitotic checkpoint silencing. TRIP13 overexpression is a hallmark of cancer cells showing chromosomal instability, particularly in certain breast cancers with poor prognosis. We suggest that premature mitotic checkpoint silencing triggered by TRIP13 overexpression may promote cancer development.
Collapse
Affiliation(s)
- Kexi Wang
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - James C Hittle
- the Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, and
| | - Dawn Macdonald
- the Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Gordon K Chan
- the Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Tim J Yen
- the Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, and
| | - Song-Tao Liu
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606,
| |
Collapse
|
47
|
Hopkins J, Hwang G, Jacob J, Sapp N, Bedigian R, Oka K, Overbeek P, Murray S, Jordan PW. Meiosis-specific cohesin component, Stag3 is essential for maintaining centromere chromatid cohesion, and required for DNA repair and synapsis between homologous chromosomes. PLoS Genet 2014; 10:e1004413. [PMID: 24992337 PMCID: PMC4081007 DOI: 10.1371/journal.pgen.1004413] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 04/19/2014] [Indexed: 11/18/2022] Open
Abstract
Cohesins are important for chromosome structure and chromosome segregation during mitosis and meiosis. Cohesins are composed of two structural maintenance of chromosomes (SMC1-SMC3) proteins that form a V-shaped heterodimer structure, which is bridged by a α-kleisin protein and a stromal antigen (STAG) protein. Previous studies in mouse have shown that there is one SMC1 protein (SMC1β), two α-kleisins (RAD21L and REC8) and one STAG protein (STAG3) that are meiosis-specific. During meiosis, homologous chromosomes must recombine with one another in the context of a tripartite structure known as the synaptonemal complex (SC). From interaction studies, it has been shown that there are at least four meiosis-specific forms of cohesin, which together with the mitotic cohesin complex, are lateral components of the SC. STAG3 is the only meiosis-specific subunit that is represented within all four meiosis-specific cohesin complexes. In Stag3 mutant germ cells, the protein level of other meiosis-specific cohesin subunits (SMC1β, RAD21L and REC8) is reduced, and their localization to chromosome axes is disrupted. In contrast, the mitotic cohesin complex remains intact and localizes robustly to the meiotic chromosome axes. The instability of meiosis-specific cohesins observed in Stag3 mutants results in aberrant DNA repair processes, and disruption of synapsis between homologous chromosomes. Furthermore, mutation of Stag3 results in perturbation of pericentromeric heterochromatin clustering, and disruption of centromere cohesion between sister chromatids during meiotic prophase. These defects result in early prophase I arrest and apoptosis in both male and female germ cells. The meiotic defects observed in Stag3 mutants are more severe when compared to single mutants for Smc1β, Rec8 and Rad21l, however they are not as severe as the Rec8, Rad21l double mutants. Taken together, our study demonstrates that STAG3 is required for the stability of all meiosis-specific cohesin complexes. Furthermore, our data suggests that STAG3 is required for structural changes of chromosomes that mediate chromosome pairing and synapsis, DNA repair and progression of meiosis.
Collapse
Affiliation(s)
- Jessica Hopkins
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Grace Hwang
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Justin Jacob
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Nicklas Sapp
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Rick Bedigian
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Kazuhiro Oka
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Paul Overbeek
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Steve Murray
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Philip W. Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
48
|
Chen Y, Wang SX, Mu R, Luo X, Liu ZS, Liang B, Zhuo HL, Hao XP, Wang Q, Fang DF, Bai ZF, Wang QY, Wang HM, Jin BF, Gong WL, Zhou T, Zhang XM, Xia Q, Li T. Dysregulation of the miR-324-5p-CUEDC2 axis leads to macrophage dysfunction and is associated with colon cancer. Cell Rep 2014; 7:1982-93. [PMID: 24882011 DOI: 10.1016/j.celrep.2014.05.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/07/2014] [Accepted: 05/05/2014] [Indexed: 12/12/2022] Open
Abstract
CUEDC2, a CUE-domain-containing protein, modulates inflammation, but its involvement in tumorigenesis is still poorly understood. Here, we report that CUEDC2 is a key regulator of macrophage function and critical for protection against colitis-associated tumorigenesis. CUEDC2 expression is dramatically upregulated during macrophage differentiation, and CUEDC2 deficiency results in excessive production of proinflammatory cytokines. The level of CUEDC2 in macrophages is modulated by miR- 324-5p. We find that Cuedc2 KO mice are more susceptible to dextran-sodium-sulfate-induced colitis, and macrophage transplantation results suggest that the increased susceptibility results from the dysfunction of macrophages lacking CUEDC2. Furthermore, we find that Cuedc2 KO mice are more prone to colitis-associated cancer. Importantly, CUEDC2 expression is almost undetectable in macrophages in human colon cancer, and this decreased CUEDC2 expression is associated with high levels of interleukin-4 and miR-324-5p. Thus, CUEDC2 plays a crucial role in modulating macrophage function and is associated with both colitis and colon tumorigenesis.
Collapse
Affiliation(s)
- Yuan Chen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Shao-Xin Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; Department of Digestive, Navy General Hospital, 6 Fucheng Road, Beijing 100048, China
| | - Rui Mu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Xue Luo
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Zhao-Shan Liu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Bing Liang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Hai-Long Zhuo
- Department of Transfusion Medicine, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Xiao-Peng Hao
- Department of General Surgery, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Qiong Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Di-Feng Fang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Zhao-Fang Bai
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Qian-Yi Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - He-Mei Wang
- Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing 100850, China
| | - Bao-Feng Jin
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Wei-Li Gong
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Xue-Min Zhang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Qing Xia
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China.
| | - Tao Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China.
| |
Collapse
|
49
|
Gao B, Wang Y, Qiu Y, Xia W, Meng X, Lu M, Zhang W, Qiao X, Zhang Y, Xue D. Bioinformatics screening regarding herbal components that targetedly regulate the function of tumour-associated macrophages. Oncol Rep 2014; 32:309-17. [PMID: 24859918 DOI: 10.3892/or.2014.3217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/17/2014] [Indexed: 11/06/2022] Open
Abstract
As an important component of tumour stroma, tumour-associated macrophages (TAMs) promote tumour development and progression. Herbs have been increasingly used in anticancer therapies due to their wide-ranging anticancer effects and minor side-effects. However, no herb-based treatments targeting TAMs have yet been proposed. To address this issue, screening using modular analysis bioinformatics techniques found 6 core functional modules for TAMs that contain 46 total genes. Moreover, 15 potential new anticancer drugs that regulate the genes in the 6 core modules were identified through bioinformatics techniques and Fisher's exact test. Our results provide a new research avenue for targeting TAMs in anticancer therapies.
Collapse
Affiliation(s)
- Bo Gao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yingmei Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yan Qiu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weiliang Xia
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xianzhi Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ming Lu
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Weihui Zhang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xin Qiao
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Yingmei Zhang
- Central Laboratory, The First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
50
|
Wang Y, Jin F, Higgins R, McKnight K. The current view for the silencing of the spindle assembly checkpoint. Cell Cycle 2014; 13:1694-701. [PMID: 24776751 DOI: 10.4161/cc.29027] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Chromosome bipolar attachment is achieved when sister kinetochores are attached by microtubules emanating from opposite spindle poles, and this process is essential for faithful chromosome segregation during anaphase. A fundamental question in cell biology is how cells ensure that chromosome segregation only occurs after bipolar attachment. It is well documented that unattached kinetochores activate the spindle assembly checkpoint (SAC) to delay chromosome segregation. Therefore, the silencing of the SAC is thought to trigger anaphase onset, but how correct chromosome attachment is coupled with SAC silencing and the subsequent anaphase onset is poorly understood. The establishment of chromosome bipolar attachment not only results in the occupancy of kinetochores by microtubules but also applies tension on sister kinetochores. A long-standing debate is whether the kinetochore attachment (occupancy) or the tension silences the SAC. Recent work in budding yeast reveals the SAC silencing network SSN that prevents SAC silencing prior to tension generation at kinetochores. Therefore, this signaling pathway ensures that SAC silencing and the subsequent anaphase onset occur only after chromosome bipolar attachment applies tension on chromosomes. This review will summarize the recent advances in the understanding of the SAC silencing process.
Collapse
Affiliation(s)
- Yanchang Wang
- Department of Biomedical Sciences; College of Medicine; Florida State University; Tallahassee, FL USA
| | - Fengzhi Jin
- Department of Biomedical Sciences; College of Medicine; Florida State University; Tallahassee, FL USA
| | - Ryan Higgins
- Department of Biomedical Sciences; College of Medicine; Florida State University; Tallahassee, FL USA
| | - Kelly McKnight
- Department of Biomedical Sciences; College of Medicine; Florida State University; Tallahassee, FL USA
| |
Collapse
|