1
|
Blengini CS, Tang S, Mendola RJ, Garrisi GJ, Swain JE, Schindler K. AURKA controls oocyte spindle assembly checkpoint and chromosome alignment by HEC1 phosphorylation. Life Sci Alliance 2025; 8:e202403146. [PMID: 40328643 PMCID: PMC12056248 DOI: 10.26508/lsa.202403146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
In human oocytes, meiosis I is error-prone, causing early miscarriages and developmental disorders. The Aurora protein kinases are key regulators of chromosome segregation in mitosis and meiosis, and their dysfunction is associated with aneuploidy. Oocytes express three Aurora kinase (AURK) proteins, but only AURKA is necessary and sufficient to support oocyte meiosis in mice. However, the unique molecular contributions to ensuring high egg quality of AURKA remain unclear. Here, using a combination of genetic and pharmacological approaches, we evaluated how AURKA phosphorylation regulates outer kinetochore function during oocyte meiosis. We found that the outer kinetochore protein Ndc80/HEC1 is constitutively phosphorylated at multiple residues by Aurora kinases during meiosis I, but that serine 69 is specifically phosphorylated by AURKA in mouse and human oocytes. We further show that serine 69 phosphorylation contributes to spindle assembly checkpoint activation and chromosome alignment during meiosis I. These results provide a fundamental mechanistic understanding of how AURKA regulates meiosis and kinetochore function to ensure meiosis I fidelity.
Collapse
Affiliation(s)
- Cecilia S Blengini
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| | - Shuang Tang
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| | - Robert J Mendola
- CCRM / Institute for Reproductive Medicine and Science (IRMS), Livingston, NJ, USA
| | - G John Garrisi
- CCRM / Institute for Reproductive Medicine and Science (IRMS), Livingston, NJ, USA
| | | | - Karen Schindler
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
2
|
Zheng H, Zhang Q, Liu X, Shi F, Yang F, Xiang S, Jiang H. Aurora-A condensation mediated by BuGZ aids its mitotic centrosome functions. iScience 2024; 27:109785. [PMID: 38746663 PMCID: PMC11090908 DOI: 10.1016/j.isci.2024.109785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/30/2023] [Accepted: 04/16/2024] [Indexed: 03/17/2025] Open
Abstract
Centrosomes composed of centrioles and the pericentriolar material (PCM), serve as the platform for microtubule polymerization during mitosis. Despite some centriole and PCM proteins have been reported to utilize liquid-liquid phase separation (LLPS) to perform their mitotic functions, whether and how centrosomal kinases exert the coacervation in mitosis is still unknown. Here we reveal that Aurora-A, one key centrosomal kinase in regulating centrosome formation and functions, undergoes phase separation in vitro or in centrosomes from prophase, mediated by the conserved positive-charged residues inside its intrinsic disordered region (IDR) and the intramolecular interaction between its N- and C-terminus. Aurora-A condensation affects centrosome maturation, separation, initial spindle formation from the spindle pole and its kinase activity. Moreover, BuGZ interacts with Aurora-A to enhance its LLPS and centrosome functions. Thus, we propose that Aurora-A collaborates with BuGZ to exhibit the property of LLPS in centrosomes to control its centrosome-dependent functions from prophase.
Collapse
Affiliation(s)
- Hui Zheng
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Qiaoqiao Zhang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Center for Cross-disciplinary Sciences, University of Science & Technology of China, School of Life Sciences, Hefei, China
| | - Fan Shi
- MOE Key Lab for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Center for Cross-disciplinary Sciences, University of Science & Technology of China, School of Life Sciences, Hefei, China
| | - Shengqi Xiang
- MOE Key Lab for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Hao Jiang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
3
|
Gupta D, Kumar M, Saifi S, Rawat S, Ethayathulla AS, Kaur P. A comprehensive review on role of Aurora kinase inhibitors (AKIs) in cancer therapeutics. Int J Biol Macromol 2024; 265:130913. [PMID: 38508544 DOI: 10.1016/j.ijbiomac.2024.130913] [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: 11/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Aurora kinases (AURKs) are a family of serine /threonine protein kinases that have a crucial role in cell cycle process mainly in the event of chromosomal segregation, centrosome maturation and cytokinesis. The family consists of three members including Aurora kinase A (AURK-A), Aurora kinase B (AURK-B) and Aurora kinase C (AURK-C). All AURKs contain a conserved kinase domain for their activity but differ in their cellular localization and functions. AURK-A and AURK-B are expressed mainly in somatic cells while the expression of AURK-C is limited to germ cells. AURK-A promotes G2 to M transition of cell cycle by controlling centrosome maturation and mitotic spindle assembly. AURK-B and AURK-C form the chromosome passenger complex (CPC) that ensures proper chromosomal alignments and segregation. Aberrant expression of AURK-A and AURK-B has been detected in several solid tumours and malignancies. Hence, they have become an attractive therapeutic target against cancer. The first part of this review focuses on AURKs structure, functions, subcellular localization, and their role in tumorigenesis. The review also highlights the functional and clinical impact of selective as well as pan kinase inhibitors. Currently, >60 compounds that target AURKs are in preclinical and clinical studies. The drawbacks of existing inhibitors like selectivity, drug resistance and toxicity have also been addressed. Since, majority of inhibitors are Aurora kinase inhibitor (AKI) type-1 that bind to the active (DFGin and Cin) conformation of the kinase, this information may be utilized to design highly selective kinase inhibitors that can be combined with other therapeutic agents for better clinical outcomes.
Collapse
Affiliation(s)
- Deepali Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Sana Saifi
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Shivani Rawat
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - A S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India.
| |
Collapse
|
4
|
Zeng Y, Ren X, Jin P, Zhang Y, Zhuo M, Wang J. Development of MPS1 Inhibitors: Recent Advances and Perspectives. J Med Chem 2023; 66:16484-16514. [PMID: 38095579 DOI: 10.1021/acs.jmedchem.3c00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Monopolar spindle kinase 1 (MPS1) plays a pivotal role as a dual-specificity kinase governing spindle assembly checkpoint activation and sister chromatid separation in mitosis. Its overexpression has been observed in various human malignancies. MPS1 reduces spindle assembly checkpoint sensitivity, allowing tumor cells with a high degree of aneuploidy to complete mitosis and survive. Thus, MPS1 has emerged as a promising candidate for cancer therapy. Despite the identification of numerous MPS1 inhibitors, only five have advanced to clinical trials with none securing FDA approval for cancer treatment. In this perspective, we provide a concise overview of the structural and functional characteristics of MPS1 by highlighting its relevance to cancer. Additionally, we explore the structure-activity relationships, selectivity, and pharmacokinetics of MPS1 inhibitors featuring diverse scaffolds. Moreover, we review the reported work on enhancing MPS1 inhibitor selectivity, offering valuable insights into the discovery of novel, highly potent small-molecule MPS1 inhibitors.
Collapse
Affiliation(s)
- Yangjie Zeng
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Pengyao Jin
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Yali Zhang
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Ming Zhuo
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Jubo Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
5
|
Kim S, Jun K, Kim YH, Jung KY, Oh JS, Kim JS. Endosulfine alpha maintains spindle pole integrity by recruiting Aurora A during mitosis. BMC Cancer 2023; 23:1263. [PMID: 38129815 PMCID: PMC10734108 DOI: 10.1186/s12885-023-11742-0] [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: 09/22/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The maintenance of spindle pole integrity is essential for spindle assembly and chromosome segregation during mitosis. However, the underlying mechanisms governing spindle pole integrity remain unclear. METHODS ENSA was inhibited by siRNA or MKI-2 treatment and its effect on cell cycle progression, chromosome alignment and microtubule alignment was observed by immunohistochemical staining and western blotting. PP2A-B55α knockdown by siRNA was performed to rescue the phenotype caused by ENSA inhibition. The interaction between ENSA and Aurora A was detected by in situ PLA. Furthermore, orthotopic implantation of 4Tl-luc cancer cells was conducted to confirm the consistency between the in vitro and in vivo relationship of the ENSA-Aurora A interaction. RESULTS During mitosis, p-ENSA is localized at the spindle poles, and the inhibition of ENSA results in mitotic defects, such as misaligned chromosomes, multipolar spindles, asymmetric bipolar spindles, and centrosome defects, with a delay in mitotic progression. Although the mitotic delay caused by ENSA inhibition was rescued by PP2A-B55α depletion, spindle pole defects persisted. Notably, we observed a interaction between ENSA and Aurora A during mitosis, and inhibition of ENSA reduced Aurora A expression at the mitotic spindle poles. Injecting MKI-2-sensitized tumors led to increased chromosomal instability and downregulation of the MASTL-ENSA-Aurora A pathway in an orthotopic breast cancer mouse model. CONCLUSIONS These findings provide novel insights into the regulation of spindle pole integrity by the MASTL-ENSA-Aurora A pathway during mitosis, highlighting the significance of ENSA in recruiting Aurora A to the spindle pole, independent of PP2A-B55α.
Collapse
Grants
- HN22C0173 Korea Drug Development Fund funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare
- HN22C0173 Korea Drug Development Fund funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare
- HN22C0173 Korea Drug Development Fund funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare
- HN22C0173 Korea Drug Development Fund funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare
- NRF-2020M2D9A2094153 the National Research Foundation of Korea
- NRF-2020M2D9A2094153 the National Research Foundation of Korea
- No.50531-2023 the Ministry of Science and ICT (MSIT), Republic of Korea
Collapse
Affiliation(s)
- Seul Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 215-4 Gongneung-Dong, Nowon-Ku, Seoul, 139706, Korea
| | - Kyoungho Jun
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 215-4 Gongneung-Dong, Nowon-Ku, Seoul, 139706, Korea
| | - Ye-Hyun Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 215-4 Gongneung-Dong, Nowon-Ku, Seoul, 139706, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34113, Korea
| | - Kwan-Young Jung
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea.
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 215-4 Gongneung-Dong, Nowon-Ku, Seoul, 139706, Korea.
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34113, Korea.
| |
Collapse
|
6
|
Inhibition of branched-chain alpha-keto acid dehydrogenase kinase augments the sensitivity of ovarian and breast cancer cells to paclitaxel. Br J Cancer 2023; 128:896-906. [PMID: 36526674 PMCID: PMC9977917 DOI: 10.1038/s41416-022-02095-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
CONTEXT Many cancer patients who initially respond to chemotherapy eventually develop chemoresistance, and to address this, we previously conducted a RNAi screen to identify genes contributing to resistance. One of the hits from the screen was branched-chain α-keto acid dehydrogenase kinase (BCKDK). BCKDK controls the metabolism of branched-chain amino acids (BCAAs) through phosphorylation and inactivation of the branched-chain α-keto acid dehydrogenase complex (BCKDH), thereby inhibiting catabolism of BCAAs. METHODS We measured the impact on paclitaxel sensitivity of inhibiting BCKDK in ovarian and breast cancer cell lines. RESULTS Inhibition of BCKDK using siRNA or two chemical inhibitors (BCKDKi) was synergistic with paclitaxel in both breast and ovarian cancer cells. BCKDKi reduced levels of BCAA and the addition of exogenous BCAA suppressed this synergy. BCKDKi inactivated the mTORC1-Aurora pathway, allowing cells to overcame M-phase arrest induced by paclitaxel. In some cases, cells almost completed cytokinesis, then reverted to a single cell, resulting in multinucleate cells. CONCLUSION BCKDK is an attractive target to augment the sensitivity of cancer cells to paclitaxel.
Collapse
|
7
|
Wang Q, Wang T, Liang S, Zhou L. Ox-LDL-Induced Vascular Smooth Muscle Cell Dysfunction Partly Depends on the Circ_0044073/miR-377-3p/AURKA Axis in Atherosclerosis. Int Heart J 2023; 64:252-262. [PMID: 37005319 DOI: 10.1536/ihj.22-148] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Atherosclerosis (AS) is the main reason for most cardiovascular diseases. Circular RNA hsa_circ_0044073 (circ_0044073) has been found to promote AS progression. However, the specific regulatory mechanism of circ_0044073 in AS progression remains unclear.In this study, oxidized low-density lipoprotein (Ox-LDL) -stimulated human vascular smooth muscle cells (VSMCs) were used as AS cell models. The expression changes of circ_0044073 in serum samples and Ox-LDL-stimulated human VSMCs were assessed via real-time quantitative polymerase chain reaction (RT-qPCR). Cell viability, proliferation, colony formation, migration, and invasion were assessed using 3- (4,5-Dimethylthiazol-2-yl) -2,5-Diphenyltetrazolium Bromide (MTT), 5-ethynyl-2'-deoxyuridine (EDU), colony formation, and transwell assays. Some protein levels were detected via Western blotting. The regulatory mechanism of circ_0044073 was predicted using bioinformatics analysis and validated by dual-luciferase reporter and RNA pull-down assays.We observed an overt increase in circ_0044073 expression in serum samples derived from AS patients and Ox-LDL-stimulated human VSMCs. Circ_0044073 was identified as a miR-377-3p sponge. Either circ_0044073 knockdown or miR-377-3p overexpression could impair Ox-LDL-induced human VSMC proliferation, migration, invasion, and inflammation. AURKA served as a miR-377-3p target, and circ_0044073 regulated AURKA expression by adsorbing miR-377-3p. Furthermore, AURKA overexpression partly reversed the effects of circ_0044073 inhibition on Ox-LDL-induced human VSMC proliferation, migration, invasion, and inflammation.Circ_0044073 promoted AS progression by elevating AURKA expression by functioning as a miR-377-3p sponge. Providing a proof-of-concept demonstration to support circ_0044073 might be a target for AS treatment.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Vascular Surgery, Taizhou University Affiliated Municipal Hospital
| | - Tao Wang
- Department of Vascular Surgery, Taizhou University Affiliated Municipal Hospital
| | - Siyuan Liang
- Department of Vascular Surgery, Taizhou University Affiliated Municipal Hospital
| | - Long Zhou
- Department of Vascular Surgery, Taizhou University Affiliated Municipal Hospital
| |
Collapse
|
8
|
Vichas A, Riley AK, Nkinsi NT, Kamlapurkar S, Parrish PCR, Lo A, Duke F, Chen J, Fung I, Watson J, Rees M, Gabel AM, Thomas JD, Bradley RK, Lee JK, Hatch EM, Baine MK, Rekhtman N, Ladanyi M, Piccioni F, Berger AH. Integrative oncogene-dependency mapping identifies RIT1 vulnerabilities and synergies in lung cancer. Nat Commun 2021; 12:4789. [PMID: 34373451 PMCID: PMC8352964 DOI: 10.1038/s41467-021-24841-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
CRISPR-based cancer dependency maps are accelerating advances in cancer precision medicine, but adequate functional maps are limited to the most common oncogenes. To identify opportunities for therapeutic intervention in other rarer subsets of cancer, we investigate the oncogene-specific dependencies conferred by the lung cancer oncogene, RIT1. Here, genome-wide CRISPR screening in KRAS, EGFR, and RIT1-mutant isogenic lung cancer cells identifies shared and unique vulnerabilities of each oncogene. Combining this genetic data with small-molecule sensitivity profiling, we identify a unique vulnerability of RIT1-mutant cells to loss of spindle assembly checkpoint regulators. Oncogenic RIT1M90I weakens the spindle assembly checkpoint and perturbs mitotic timing, resulting in sensitivity to Aurora A inhibition. In addition, we observe synergy between mutant RIT1 and activation of YAP1 in multiple models and frequent nuclear overexpression of YAP1 in human primary RIT1-mutant lung tumors. These results provide a genome-wide atlas of oncogenic RIT1 functional interactions and identify components of the RAS pathway, spindle assembly checkpoint, and Hippo/YAP1 network as candidate therapeutic targets in RIT1-mutant lung cancer.
Collapse
Affiliation(s)
- Athea Vichas
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amanda K Riley
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Naomi T Nkinsi
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shriya Kamlapurkar
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Phoebe C R Parrish
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - April Lo
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Fujiko Duke
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Jennifer Chen
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Iris Fung
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | | | - Matthew Rees
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Austin M Gabel
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - James D Thomas
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Robert K Bradley
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Emily M Hatch
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marina K Baine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Federica Piccioni
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Merck Research Laboratories, Boston, MA, USA
| | - Alice H Berger
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
9
|
Arslanhan MD, Rauniyar N, Yates JR, Firat-Karalar EN. Aurora Kinase A proximity map reveals centriolar satellites as regulators of its ciliary function. EMBO Rep 2021; 22:e51902. [PMID: 34169630 PMCID: PMC8339716 DOI: 10.15252/embr.202051902] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Aurora kinase A (AURKA) is a conserved kinase that plays crucial roles in numerous cellular processes. Although AURKA overexpression is frequent in human cancers, its pleiotropic functions and multifaceted regulation present challenges in its therapeutic targeting. Key to overcoming these challenges is to identify and characterize the full range of AURKA interactors, which are often weak and transient. Previous proteomic studies were limited in monitoring dynamic and non-mitotic AURKA interactions. Here, we generate the proximity interactome of AURKA in asynchronous cells, which consists of 440 proteins involving multiple biological processes and cellular compartments. Importantly, AURKA has extensive proximate and physical interactions to centriolar satellites, key regulators of the primary cilium. Loss-of-function experiments identify satellites as negative regulators of AURKA activity, abundance, and localization in quiescent cells. Notably, loss of satellites activates AURKA at the basal body, decreases centrosomal IFT88 levels, and causes ciliogenesis defects. Collectively, our results provide a resource for dissecting spatiotemporal regulation of AURKA and uncover its proteostatic regulation by satellites as a new mechanism for its ciliary functions.
Collapse
Affiliation(s)
- Melis D Arslanhan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Navin Rauniyar
- Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
10
|
Wang R, Ascanelli C, Abdelbaki A, Fung A, Rasmusson T, Michaelides I, Roberts K, Lindon C. Selective targeting of non-centrosomal AURKA functions through use of a targeted protein degradation tool. Commun Biol 2021; 4:640. [PMID: 34050235 PMCID: PMC8163823 DOI: 10.1038/s42003-021-02158-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
Targeted protein degradation tools are becoming a new therapeutic modality, allowing small molecule ligands to be reformulated as heterobifunctional molecules (PROteolysis Targeting Chimeras, PROTACs) that recruit ubiquitin ligases to targets of interest, leading to ubiquitination and destruction of the targets. Several PROTACs against targets of clinical interest have been described, but detailed descriptions of the cell biology modulated by PROTACs are missing from the literature. Here we describe the functional characterization of a PROTAC derived from AURKA inhibitor MLN8237 (alisertib). We demonstrate efficient and specific destruction of both endogenous and overexpressed AURKA by Cereblon-directed PROTACs. At the subcellular level, we find differential targeting of AURKA on the mitotic spindle compared to centrosomes. The phenotypic consequences of PROTAC treatment are therefore distinct from those mediated by alisertib, and in mitotic cells differentially regulate centrosome- and chromatin- based microtubule spindle assembly pathways. In interphase cells PROTAC-mediated clearance of non-centrosomal AURKA modulates the cytoplasmic role played by AURKA in mitochondrial dynamics, whilst the centrosomal pool is refractory to PROTAC-mediated clearance. Our results point to differential sensitivity of subcellular pools of substrate, governed by substrate conformation or localization-dependent accessibility to PROTAC action, a phenomenon not previously described for this new class of degrader compounds. Wang et al develop tools to target the mitotic regulator AURKA by synthesising PROTACs based on the inhibitor MLN8237. They find that the new PROTAC compound efficiently clears cytoplasmic and mitotic spindle-associated AURKA but does not eliminate AURKA activity from centrosomes, demonstrating the possibility of targeting subpopulations.
Collapse
Affiliation(s)
- Richard Wang
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Ahmed Abdelbaki
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Alex Fung
- Department of Pharmacology, University of Cambridge, Cambridge, UK.,University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Tim Rasmusson
- Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.,Bristol Myers Squibb, Cambridge, MA, USA
| | | | | | - Catherine Lindon
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Naik A, Decock J. Targeting of lactate dehydrogenase C dysregulates the cell cycle and sensitizes breast cancer cells to DNA damage response targeted therapy. Mol Oncol 2021; 16:885-903. [PMID: 34050611 PMCID: PMC8847988 DOI: 10.1002/1878-0261.13024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/21/2021] [Accepted: 05/27/2021] [Indexed: 12/31/2022] Open
Abstract
The cancer testis antigen (CTA) lactate dehydrogenase C (LDHC) is a promising anticancer target with tumor-specific expression and immunogenicity. Interrogation of breast cancer patient cohorts from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) indicate that upregulation of LDHC expression correlates with unfavorable prognosis. Although the role of LDHC is well characterized in spermatocytes, its role in tumors remains largely unknown. We investigated whether LDHC is involved in regulating genomic stability and whether it could be targeted to affect tumor cellular fitness. Silencing LDHC in four breast cancer cell lines significantly increased the presence of giant cells, nuclear aberrations, DNA damage, and apoptosis. LDHC-silenced cells demonstrated aberrant cell cycle progression with differential expression of cell cycle checkpoint and DNA damage response regulators. In addition, LDHC silencing-induced microtubule destabilization, culminating in increased mitotic catastrophe and reduced long-term survival. Notably, the clonogenicity of LDHC-silenced cells was further reduced by treatment with the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib and with the DNA-damaging drug cisplatin. This study supports the therapeutic potential of targeting LDHC to mitigate cancer cell survival and improve sensitivity to agents that cause DNA damage or inhibit its repair.
Collapse
Affiliation(s)
- Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
12
|
Nehlig A, Seiler C, Steblyanko Y, Dingli F, Arras G, Loew D, Welburn J, Prigent C, Barisic M, Nahmias C. Reciprocal regulation of Aurora kinase A and ATIP3 in the control of metaphase spindle length. Cell Mol Life Sci 2021; 78:1765-1779. [PMID: 32789689 PMCID: PMC11072152 DOI: 10.1007/s00018-020-03614-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/18/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022]
Abstract
Maintaining the integrity of the mitotic spindle in metaphase is essential to ensure normal cell division. We show here that depletion of microtubule-associated protein ATIP3 reduces metaphase spindle length. Mass spectrometry analyses identified the microtubule minus-end depolymerizing kinesin Kif2A as an ATIP3 binding protein. We show that ATIP3 controls metaphase spindle length by interacting with Kif2A and its partner Dda3 in an Aurora kinase A-dependent manner. In the absence of ATIP3, Kif2A and Dda3 accumulate at spindle poles, which is consistent with reduced poleward microtubule flux and shortening of the spindle. ATIP3 silencing also limits Aurora A localization to the poles. Transfection of GFP-Aurora A, but not kinase-dead mutant, rescues the phenotype, indicating that ATIP3 maintains Aurora A activity on the poles to control Kif2A targeting and spindle size. Collectively, these data emphasize the pivotal role of Aurora kinase A and its mutual regulation with ATIP3 in controlling spindle length.
Collapse
Affiliation(s)
- Anne Nehlig
- Inserm U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94800, Villejuif, France
- LabEx LERMIT, Université Paris Saclay, 92296, Châtenay-Malabry, France
- Institut Gustave Roussy, Inserm, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Université Paris-Saclay, 94800, Villejuif, France
| | - Cynthia Seiler
- Inserm U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94800, Villejuif, France
- LabEx LERMIT, Université Paris Saclay, 92296, Châtenay-Malabry, France
- Institut Gustave Roussy, Inserm, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Université Paris-Saclay, 94800, Villejuif, France
| | - Yulia Steblyanko
- Cell Division Laboratory, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Florent Dingli
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248, Paris Cedex 05, France
| | - Guillaume Arras
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248, Paris Cedex 05, France
| | - Damarys Loew
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248, Paris Cedex 05, France
| | - Julie Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Claude Prigent
- Institut de Génétique et Développement de Rennes (IGDR), Unité CNRS, UMR 6290, Université de Rennes, 35043, Rennes, France
| | - Marin Barisic
- Cell Division Laboratory, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Clara Nahmias
- Inserm U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94800, Villejuif, France.
- LabEx LERMIT, Université Paris Saclay, 92296, Châtenay-Malabry, France.
- Institut Gustave Roussy, Inserm, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Université Paris-Saclay, 94800, Villejuif, France.
| |
Collapse
|
13
|
Ganapathi RN, Norris EJ, Sutker AP, Klotz KE, Ganapathi MK. Targeting Aurora A Kinase (AAK) in Platinum-Resistant High Grade Serous Ovarian Cancer. Front Oncol 2020; 10:1354. [PMID: 32974133 PMCID: PMC7466726 DOI: 10.3389/fonc.2020.01354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/29/2020] [Indexed: 11/13/2022] Open
Abstract
Aurora A kinase (AAK) involved in G2-M transition is functionally involved in centrosome maturation and maintaining an active spindle assembly checkpoint. We tested the hypothesis that in platinum-taxane resistant high grade serous ovarian cancer (HGSOC) inhibition of AAK involved in G2-M transition would enhance the anti-tumor activity of cisplatin (CP) or paclitaxel (PT). Using HGSOC cell lines from platinum-taxane refractory patients that do not harbor BRCA1/2 mutations, we tested the anti-tumor activity of CP, or PT alone or in combination with the AAK inhibitor alisertib (AL). Treatment with CP for 3 h or PT for 6 h followed sequentially by AL for 48 h led to a significant decrease in cell survival (p < 0.001) compared to treatment with either drug alone in HGSOC cells but not in immortalized normal human ovarian surface epithelium or normal human fallopian tube secretory epithelium cells. The treatment with CP or PT followed by AL also led to a significant increase in reactive oxygen species (p < 0.05), apoptosis (p < 0.001) and accumulation of cells in G2/M that was accompanied by a modest increase in expression of AAK. Downregulation of AAK, but not aurora B kinase, with targeted siRNAs also significantly enhanced apoptosis by CP or PT, suggesting that AL specifically targeted AAK. In summary, in HGSOC without BRCA1/2 mutations, CP, or PT resistance can potentially be circumvented by sequential treatment with AL that inhibits AAK involved in G2-M transition.
Collapse
Affiliation(s)
- Ram N Ganapathi
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Eric J Norris
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Ashley P Sutker
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Kaitlin E Klotz
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Mahrukh K Ganapathi
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| |
Collapse
|
14
|
Abdelbaki A, Akman HB, Poteau M, Grant R, Gavet O, Guarguaglini G, Lindon C. AURKA destruction is decoupled from its activity at mitotic exit but is essential to suppress interphase activity. J Cell Sci 2020; 133:jcs243071. [PMID: 32393600 PMCID: PMC7328152 DOI: 10.1242/jcs.243071] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/19/2020] [Indexed: 12/22/2022] Open
Abstract
Activity of AURKA is controlled through multiple mechanisms including phosphorylation, ubiquitin-mediated degradation and allosteric interaction with TPX2. Activity peaks at mitosis, before AURKA is degraded during and after mitotic exit in a process strictly dependent on the APC/C coactivator FZR1. We used FZR1 knockout cells (FZR1KO) and a novel FRET-based AURKA biosensor to investigate how AURKA activity is regulated in the absence of destruction. We found that AURKA activity in FZR1KO cells dropped at mitotic exit as rapidly as in parental cells, despite absence of AURKA destruction. Unexpectedly, TPX2 was degraded normally in FZR1KO cells. Overexpression of an N-terminal TPX2 fragment sufficient for AURKA binding, but that is not degraded at mitotic exit, caused delay in AURKA inactivation. We conclude that inactivation of AURKA at mitotic exit is determined not by AURKA degradation but by degradation of TPX2 and therefore is dependent on CDC20 rather than FZR1. The biosensor revealed that FZR1 instead suppresses AURKA activity in interphase and is critically required for assembly of the interphase mitochondrial network after mitosis.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Ahmed Abdelbaki
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - H Begum Akman
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Marion Poteau
- Institut Gustave Roussy, UMR9019 - CNRS, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Rhys Grant
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Olivier Gavet
- Institut Gustave Roussy, UMR9019 - CNRS, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, CNR, Via degli Apuli 4, 00185 Roma, Italy
| | - Catherine Lindon
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| |
Collapse
|
15
|
Bertolin G, Tramier M. Insights into the non-mitotic functions of Aurora kinase A: more than just cell division. Cell Mol Life Sci 2020; 77:1031-1047. [PMID: 31562563 PMCID: PMC11104877 DOI: 10.1007/s00018-019-03310-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 02/02/2023]
Abstract
AURKA is a serine/threonine kinase overexpressed in several cancers. Originally identified as a protein with multifaceted roles during mitosis, improvements in quantitative microscopy uncovered several non-mitotic roles as well. In physiological conditions, AURKA regulates cilia disassembly, neurite extension, cell motility, DNA replication and senescence programs. In cancer-like contexts, AURKA actively promotes DNA repair, it acts as a transcription factor, promotes cell migration and invasion, and it localises at mitochondria to regulate mitochondrial dynamics and ATP production. Here we review the non-mitotic roles of AURKA, and its partners outside of cell division. In addition, we give an insight into how structural data and quantitative fluorescence microscopy allowed to understand AURKA activation and its interaction with new substrates, highlighting future developments in fluorescence microscopy needed to better understand AURKA functions in vivo. Last, we will recapitulate the most significant AURKA inhibitors currently in clinical trials, and we will explore how the non-mitotic roles of the kinase may provide new insights to ameliorate current pharmacological strategies against AURKA overexpression.
Collapse
Affiliation(s)
- Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000, Rennes, France.
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000, Rennes, France.
| |
Collapse
|
16
|
Cui P, Abbasi B, Lin D, Rui R, Ju S. Aurora A inhibition disrupts chromosome condensation and spindle assembly during the first embryonic division in pigs. Reprod Domest Anim 2020; 55:584-593. [PMID: 32053743 DOI: 10.1111/rda.13655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/10/2020] [Indexed: 11/29/2022]
Abstract
As common overexpression of Aurora A in various tumours, much attention has focused on its function in inducing cancer, and its value in cancer therapeutics, considerably less is known regarding its role in the first cleavage division of mammalian embryos. Here, we highlight an indispensable role of Aurora A during the first mitotic division progression of pig embryos just after meiosis. The expression and spatiotemporal localization of Aurora A were initially assessed in pig embryos during the first mitotic division by Western blot analysis and indirect immunofluorescent staining. Then, the potential role of Aurora A was further evaluated using a highly selective Aurora A inhibitor, MLN8054, during this mitotic progression in pig embryos. Aurora A was found to express and exhibit a specific dynamic intracellular localization pattern during the first mitotic division in pig embryos. Aurora A was diffused in the cytoplasm at the prophase stage, and then exhibited a dynamic intracellular localization which was tightly associated with the chromosome and spindle dynamics throughout subsequent mitotic phases. Inhibition of Aurora A by MLN8054 treatment led to the failure of the first cleavage, with the majority of embryos being arrested in prophase of the mitotic division. Further subcellular structure examination showed that Aurora A inhibition not only led to the failure of spindle microtubule assembly, but also resulted in severe defects in chromosome condensation, accompanied by an obvious decrease in p-TACC3(S558) expression during the prophase of the first mitosis. Together, these results illustrated that Aurora A is crucial for both spindle assembly and chromosome condensation during the first mitotic division in pig embryos, and that the regulation of Aurora A may be associated with its effects on p-TACC3(S558) expression.
Collapse
Affiliation(s)
- Panpan Cui
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Benazir Abbasi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Defeng Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Rong Rui
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shiqiang Ju
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
17
|
Tsuchiya Y, Byrne DP, Burgess SG, Bormann J, Baković J, Huang Y, Zhyvoloup A, Yu BYK, Peak-Chew S, Tran T, Bellany F, Tabor AB, Chan AE, Guruprasad L, Garifulin O, Filonenko V, Vonderach M, Ferries S, Eyers CE, Carroll J, Skehel M, Bayliss R, Eyers PA, Gout I. Covalent Aurora A regulation by the metabolic integrator coenzyme A. Redox Biol 2020; 28:101318. [PMID: 31546169 PMCID: PMC6812009 DOI: 10.1016/j.redox.2019.101318] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/14/2019] [Accepted: 09/01/2019] [Indexed: 12/12/2022] Open
Abstract
Aurora A kinase is a master mitotic regulator whose functions are controlled by several regulatory interactions and post-translational modifications. It is frequently dysregulated in cancer, making Aurora A inhibition a very attractive antitumor target. However, recently uncovered links between Aurora A, cellular metabolism and redox regulation are not well understood. In this study, we report a novel mechanism of Aurora A regulation in the cellular response to oxidative stress through CoAlation. A combination of biochemical, biophysical, crystallographic and cell biology approaches revealed a new and, to our knowledge, unique mode of Aurora A inhibition by CoA, involving selective binding of the ADP moiety of CoA to the ATP binding pocket and covalent modification of Cys290 in the activation loop by the thiol group of the pantetheine tail. We provide evidence that covalent CoA modification (CoAlation) of Aurora A is specific, and that it can be induced by oxidative stress in human cells. Oxidising agents, such as diamide, hydrogen peroxide and menadione were found to induce Thr 288 phosphorylation and DTT-dependent dimerization of Aurora A. Moreover, microinjection of CoA into fertilized mouse embryos disrupts bipolar spindle formation and the alignment of chromosomes, consistent with Aurora A inhibition. Altogether, our data reveal CoA as a new, rather selective, inhibitor of Aurora A, which locks this kinase in an inactive state via a "dual anchor" mechanism of inhibition that might also operate in cellular response to oxidative stress. Finally and most importantly, we believe that these novel findings provide a new rationale for developing effective and irreversible inhibitors of Aurora A, and perhaps other protein kinases containing appropriately conserved Cys residues.
Collapse
Affiliation(s)
- Yugo Tsuchiya
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK
| | - Dominic P Byrne
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Selena G Burgess
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Jenny Bormann
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Jovana Baković
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK
| | - Yueyang Huang
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK
| | - Alexander Zhyvoloup
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK
| | - Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK
| | - Sew Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Trang Tran
- Department of Chemistry, University College London, London, WC1E 6BT, UK
| | - Fiona Bellany
- Department of Chemistry, University College London, London, WC1E 6BT, UK
| | - Alethea B Tabor
- Department of Chemistry, University College London, London, WC1E 6BT, UK
| | - Aw Edith Chan
- Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | | | - Oleg Garifulin
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv 143, Ukraine
| | - Valeriy Filonenko
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv 143, Ukraine
| | - Matthias Vonderach
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Samantha Ferries
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Claire E Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK; Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - John Carroll
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Mark Skehel
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Richard Bayliss
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK.
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK; Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv 143, Ukraine.
| |
Collapse
|
18
|
Guo X, Dai X, Ni J, Ma X, Xue J, Wang X. Geraniin Differentially Modulates Chromosome Stability of Colon Cancer and Noncancerous Cells by Oppositely Regulating their Spindle Assembly Checkpoint. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:254-268. [PMID: 30403302 DOI: 10.1002/em.22265] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/20/2018] [Accepted: 11/01/2018] [Indexed: 06/08/2023]
Abstract
Geraniin has been reported to specifically induce apoptosis in multiple human cancers, but the underlying mechanism is poorly defined. The spindle assembly checkpoint (SAC) is a surveillance system to ensure high-fidelity chromosome segregation during mitosis. Weakening of SAC to enhance chromosome instability (CIN) can be therapeutic because very high levels of CIN are lethal. In this study, we have investigated the effects of geraniin on the SAC of colorectal cancer HCT116 cells and noncancerous colon epithelial CCD841 cells. We find that treatment of HCT116 cells with geraniin leads to dose-dependent decrease of cell proliferation, colony formation, and anchorage-independent growth. Geraniin is found to induce apoptosis in mitotic and postmitotic HCT116 cells. Furthermore, geraniin weakens the SAC function of HCT116 cells by decreasing the transcriptional expression of several SAC kinases (particularly Mad2 and Bub1), which in turn leads to premature anaphase entry, mitotic aberrations, and CIN in HCT116 cells. In contrast, the proliferation of CCD841 cells is slightly inhibited by geraniin. Even more interestingly, geraniin increases the transcriptional expression of several SAC kinases (e.g., Mad1 and BubR1) to strengthen SAC efficiency, which contributes to the reduction of mitotic aberrations and CIN in CCD841 cells. Altogether, our findings reveal that the SAC pathway in human colon cancer and noncancerous cell lineages responses oppositely to geraniin treatment, resulting CIN promotion and suppression, respectively. Specific abrogation of SAC to induce catastrophic CIN in HCT116 cells may account for the selective anticancer action of geraniin.. Environ. Mol. Mutagen. 60:254-268, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, China
| | - Xueqin Dai
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, China
| | - Juan Ni
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, China
| | - Xiaoling Ma
- China Gene Health Management Group, Ltd., Shanghai, China
| | - Jinglun Xue
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Xu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, China
| |
Collapse
|
19
|
Ruan W, Lim HH, Surana U. Mapping Mitotic Death: Functional Integration of Mitochondria, Spindle Assembly Checkpoint and Apoptosis. Front Cell Dev Biol 2019; 6:177. [PMID: 30687704 PMCID: PMC6335265 DOI: 10.3389/fcell.2018.00177] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/22/2018] [Indexed: 01/18/2023] Open
Abstract
Targeting the mitotic pathways of rapidly proliferating tumor cells has been an effective strategy in traditional cancer therapy. Chemotherapeutics such as taxanes and vinca alkaloids, which disrupt microtubule function, have enjoyed clinical success; however, the accompanying side effects, toxicity and multi drug resistance remain as serious concerns. The emerging classes of inhibitors targeting mitotic kinases and proteasome face their own set of challenges. It is hoped that elucidation of the regulatory interface between mitotic checkpoints, mitochondria and mitotic death will aid the development of more efficacious anti-mitotic agents and improved treatment protocols. The links between the spindle assembly checkpoint (SAC) and mitochondrial dynamics that control the progression of anti-mitotic agent-induced apoptosis have been under investigation for several years and the functional integration of these various signaling networks is now beginning to emerge. In this review, we highlight current research on the regulation of SAC, the death pathway and mitochondria with particular focus on their regulatory interconnections.
Collapse
Affiliation(s)
- Weimei Ruan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hong Hwa Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Uttam Surana
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Pharmacology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
20
|
Courthéoux T, Reboutier D, Vazeille T, Cremet JY, Benaud C, Vernos I, Prigent C. Microtubule nucleation during central spindle assembly requires NEDD1 phosphorylation on Serine 405 by Aurora A. J Cell Sci 2019; 132:jcs.231118. [DOI: 10.1242/jcs.231118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
During mitosis, the cell sequentially constructs two microtubule-based spindles to ensure faithful segregation of chromosomes. A bipolar spindle first pulls apart the sister chromatids, then a central spindle further separates them away. Although the assembly of the first spindle is well described, the assembly of the second remains poorly understood. We report here that the inhibition of Aurora A leads to an absence of the central spindle due to a lack of nucleation of microtubules in the midzone. In the absence of Aurora A, the HURP and NEDD1 proteins that are involved in nucleation of microtubules fail to concentrate in the midzone. HURP is an effector of RanGTP and NEDD1 serves as an anchor for the γTURC. Interestingly, Aurora A already phosphorylates them during assembly of the bipolar spindle. We show here that the expression of a NEDD1 isoform mimicking Aurora A phosphorylation is sufficient to restore microtubule nucleation in the midzone in a context of Aurora A inhibition. These results reveal a new control mechanism of nucleation of microtubules by Aurora A during assembly of the central spindle.
Collapse
Affiliation(s)
- Thibault Courthéoux
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - David Reboutier
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - Thibaut Vazeille
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jean-Yves Cremet
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - Christelle Benaud
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - Isabelle Vernos
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Claude Prigent
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| |
Collapse
|
21
|
Liu X, Li Y, Zhang X, Liu XY, Peng A, Chen Y, Meng L, Chen H, Zhang Y, Miao X, Zheng L, Huang K. Inhibition of kinesin family member 20B sensitizes hepatocellular carcinoma cell to microtubule-targeting agents by blocking cytokinesis. Cancer Sci 2018; 109:3450-3460. [PMID: 30191636 PMCID: PMC6215872 DOI: 10.1111/cas.13794] [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: 05/15/2018] [Revised: 08/09/2018] [Accepted: 09/05/2018] [Indexed: 01/08/2023] Open
Abstract
Kinesin family member 20B (KIF20B, also known as MPHOSPH1) is a kinesin protein that plays a critical role in cytokinesis. Previously, we and others have demonstrated the oncogenic role of KIF20B in several cancers; however, the exact mechanisms underlying its tumorigenic effects remain unclear. Herein, we showed overexpression of KIF20B in human hepatocellular carcinoma (HCC) and reported a negative correlation between KIF20B level and prognosis of patients. Mechanistically, reducing KIF20B blockades mitotic exit of HCC cells at telophase in a spindle assembly checkpoint independent way. Importantly, reducing KIF20B acts synergistically with three microtubule-associated agents (MTA) to p53- or p14ARF-dependently suppress p53-wt or p53-null HCC cells. In addition to taxol, reducing KIF20B also enhanced the toxicity of two chemotherapeutic drugs, hydroxycamptothecin and mitomycin C. In conclusion, we found a novel mechanism in that blocking cytokinesis by KIF20B inhibition increases the efficacy of MTA; our results thus suggested a dual-mitotic suppression approach against HCC by combining MTA with KIF20B inhibition, which simultaneously blocks mitosis at both metaphase and telophase.
Collapse
Affiliation(s)
- Xinran Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.,Centre for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan, China
| | - Yangkai Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Xin-Yuan Liu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Anlin Peng
- The Third Hospital of Wuhan, Wuhan, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Lijing Meng
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Miao
- Tongji School of Public Health, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zheng
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.,Centre for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan, China
| |
Collapse
|
22
|
First person – Thibault Courtheoux and Alghassimou Diallo. J Cell Sci 2018. [DOI: 10.1242/jcs.218446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ABSTRACT
First Person is a series of interviews with the first authors of a selection of papers published in Journal of Cell Science, helping early-career researchers promote themselves alongside their papers. Thibault Courtheoux and Alghassimou Diallo are joint first authors on ‘Aurora A kinase activity is required to maintain an active spindle assembly checkpoint during pro-metaphase’, published in Journal of Cell Science. Thibault is a postdoc and Alghassimou a PhD student in the lab of Dr Claude Prigent at IGDR-UMR, Université de Rennes, France, investigating cell division and mechanisms leading to chromosome instability and cancer.
Collapse
|