1
|
Li X, Wang Z, Oakley G, Wang L, Lanzel E, Buchakjian M, Peng A. Targeting Aurora A to Overcome Cisplatin Resistance in Head and Neck Cancer. J Dent Res 2025; 104:531-540. [PMID: 40017056 PMCID: PMC12000625 DOI: 10.1177/00220345241309624] [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] [Indexed: 03/01/2025] Open
Abstract
Cisplatin-based chemotherapy is a cornerstone treatment for advanced recurrent head and neck squamous cell carcinoma (HNSCC). However, the effectiveness of the treatment is often hindered by intrinsic and acquired resistance and associated toxicity, highlighting a pressing unmet clinical need. Here, our compound screening identified Aurora kinase inhibitors, particularly those targeting Aurora A kinase, as potential agents to sensitize resistant HNSCC cells to cisplatin. While Aurora kinases are well-established regulators of mitosis, their precise role in cisplatin resistance is largely unknown, given that cisplatin confers toxicity primarily in cells undergoing DNA replication. We confirmed that depletion of Aurora A or its activators enhanced cisplatin response in resistant HNSCC cells. Analyses of a comprehensive database and locally treated HNSCC patient samples revealed compelling associations between Aurora A overexpression/activation and cisplatin resistance, tumor recurrence, and poor patient survival. Pharmacologic inhibition of Aurora A effectively synergized with cisplatin treatment in cellular assays and a syngeneic mouse tumor model of HNSCC. Mechanistically, Aurora A inhibition enhanced apoptosis induction after cisplatin treatment, particularly in S-phase cells; induced replication stress; and suppressed the repair of cisplatin-induced DNA crosslinking. Taken together, our findings shed light on important functions of Aurora A kinase beyond mitotic regulation. The multifaceted roles of Aurora A suggest its potential as a prime anticancer drug target. Given the ongoing investigations into numerous Aurora inhibitors for cancer therapy, exploring their clinical applications in HNSCC, especially in combination with platinum drugs, may hold significant promise.
Collapse
Affiliation(s)
- X. Li
- Department of Biomedical Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Z. Wang
- Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, NE, USA
| | - G.G. Oakley
- Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, NE, USA
| | - L. Wang
- Department of Biomedical Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - E.A. Lanzel
- Department of Oral Pathology, Radiology, & Medicine, College of Dentistry, The University of Iowa, Iowa City, IA, USA
| | - M.R. Buchakjian
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - A. Peng
- Department of Biomedical Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NA, USA
| |
Collapse
|
2
|
Sun M, Wang Y, Xin G, Yang B, Jiang Q, Zhang C. NuSAP regulates microtubule flux and Kif2A localization to ensure accurate chromosome congression. J Cell Biol 2024; 223:e202108070. [PMID: 38117947 PMCID: PMC10733630 DOI: 10.1083/jcb.202108070] [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: 08/13/2021] [Revised: 10/10/2023] [Accepted: 11/26/2023] [Indexed: 12/22/2023] Open
Abstract
Precise chromosome congression and segregation requires the proper assembly of a steady-state metaphase spindle, which is dynamic and maintained by continuous microtubule flux. NuSAP is a microtubule-stabilizing and -bundling protein that promotes chromosome-dependent spindle assembly. However, its function in spindle dynamics remains unclear. Here, we demonstrate that NuSAP regulates the metaphase spindle length control. Mechanistically, NuSAP facilitates kinetochore capture and spindle assembly by promoting Eg5 binding to microtubules. It also prevents excessive microtubule depolymerization through interaction with Kif2A, which reduces Kif2A spindle-pole localization. NuSAP is phosphorylated by Aurora A at Ser-240 during mitosis, and this phosphorylation promotes its interaction with Kif2A on the spindle body and reduces its localization with the spindle poles, thus maintaining proper spindle microtubule flux. NuSAP knockout resulted in the formation of shorter spindles with faster microtubule flux and chromosome misalignment. Taken together, we uncover that NuSAP participates in spindle assembly, dynamics, and metaphase spindle length control through the regulation of microtubule flux and Kif2A localization.
Collapse
Affiliation(s)
- Mengjie Sun
- The Academy for Cell and Life Health, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Yao Wang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Guangwei Xin
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Biying Yang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Qing Jiang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Chuanmao Zhang
- The Academy for Cell and Life Health, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
3
|
Fan YW, Lu IC, Hsu MY, Kuo WT, Wu SY, Lan SH, Wang PY, Chen CY, Liu HS, Su CL. Synthetic lethality in human bladder cancer cells by curcumin via concurrent Aurora A inhibition and autophagy induction. J Nutr Biochem 2023; 121:109438. [PMID: 37666476 DOI: 10.1016/j.jnutbio.2023.109438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 01/24/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
Combination therapies to induce mixed-type cell death and synthetic lethality have the potential to overcome drug resistance in cancer. In this study, we demonstrated that the curcumin-enhanced cytotoxicity of cisplatin/carboplatin in combination with gemcitabine was associated with Aurora A suppression-mediated G2/M arrest, and thus apoptosis, as well as MEK/ERK-mediated autophagy in human bladder cancer cells. Animal study data confirmed that curcumin combined with cisplatin/gemcitabine reduced tumorigenesis of xenograft in mice and this phenomenon was associated with elevated expressions of p-ERK and reduced p-Aurora A in tumors. Gene analyses using data repositories further revealed that reduced Aurora A expression alone did not significantly elevate the sensitivity of human bladder carcinoma cells to these anticancer drugs. Unlike other major cancer types, human bladder urothelial carcinoma tissue coexpressed higher AURKA and lower MAP1LC3B than normal tissue, and reduced Aurora A and induction of autophagy have been clinically associated with a better prognosis in patients with early but not advanced stage bladder cancer. Therefore, our results suggest that treatment strategies can utilize the synthetic lethal pair to concurrently suppress oncogenic Aurora A and induce autophagy by coadministrating curcumin with anticancer drugs for early-stage bladder cancer with high expression of Aurora A.
Collapse
Affiliation(s)
- Ya-Wen Fan
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei, Taiwan
| | - I-Ching Lu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Man-Yuan Hsu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wan-Ting Kuo
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shan-Ying Wu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Hui Lan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pao-Yuan Wang
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-Ying Chen
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Infectious Disease and Signaling Research Center, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei, Taiwan; Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan.
| |
Collapse
|
4
|
Lin CI, Chen ZC, Chen CH, Chang YH, Lee TC, Tang TT, Yu TW, Yang CM, Tsai MC, Huang CC, Yang TW, Lin CC, Wang RH, Chiou GY, Jong YJ, Chao JI. Co-inhibition of Aurora A and Haspin kinases enhances survivin blockage and p53 induction for mitotic catastrophe and apoptosis in human colorectal cancer. Biochem Pharmacol 2022; 206:115289. [PMID: 36241092 DOI: 10.1016/j.bcp.2022.115289] [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: 07/07/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is a leading cause and mortality worldwide. Aurora A and haspin kinases act pivotal roles in mitotic progression. However, the blockage of Aurora A and Haspin for CRC therapy is still unclear. Here we show that the Haspin and p-H3T3 protein levels were highly expressed in CRC tumor tissues of clinical patients. Overexpression of Haspin increased the protein levels of p-H3T3 and survivin in human CRC cells; conversely, the protein levels of p-H3T3 and survivin were decreased by the Haspin gene knockdown. Moreover, the gene knockdown of Aurora A induced abnormal chromosome segregation, mitotic catastrophe, and cell growth inhibition. Combined targeted by co-treatment of CHR6494, a Haspin inhibitor, and MLN8237, an Aurora A inhibitor, enhanced apoptosis and CRC tumor inhibition. MLN8237 and CHR6494 induced abnormal chromosome segregation and mitotic catastrophe. Meanwhile, MLN8237 and CHR6494 inhibited survivin protein levels but conversely induced p53 protein expression. Ectopic survivin expression by transfection with a survivin-expressed vector resisted the cell death in the MLN8237- and CHR6494-treated cells. In contrast, the existence of functional p53 increased the apoptotic levels by treatment with MLN8237 and CHR6494. Co-treatment of CHR6494 and MLN8237 enhanced the blockage of human CRC xenograft tumors in nude mice. Taken together, co-inhibition of Aurora A and Haspin enhances survivin inhibition, p53 pathway induction, mitotic catastrophe, apoptosis and tumor inhibition that may provide a potential strategy for CRC therapy.
Collapse
Affiliation(s)
- Chien-I Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zan-Chu Chen
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Chien-Hung Chen
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yun-Hsuan Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tsai-Chia Lee
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tsai-Tai Tang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tzu-Wei Yu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Chih-Man Yang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Ming-Chang Tsai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chi-Chou Huang
- Division of Colon and Rectum, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tzu-Wei Yang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Che Lin
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Rou-Hsin Wang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Guang-Yuh Chiou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yuh-Jyh Jong
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Departments of Pediatrics and Laboratory Medicine, and Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Jui-I Chao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Center For Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
5
|
Inchanalkar S, Balasubramanian N. Adhesion-growth factor crosstalk regulates AURKB activation and ERK signalling in re-adherent fibroblasts. J Biosci 2021. [DOI: 10.1007/s12038-021-00164-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
6
|
Bowling FZ, Frohman MA, Airola MV. Structure and regulation of human phospholipase D. Adv Biol Regul 2021; 79:100783. [PMID: 33495125 DOI: 10.1016/j.jbior.2020.100783] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
Mammalian phospholipase D (PLD) generates phosphatidic acid, a dynamic lipid secondary messenger involved with a broad spectrum of cellular functions including but not limited to metabolism, migration, and exocytosis. As a promising pharmaceutical target, the biochemical properties of PLD have been well characterized. This has led to the recent crystal structures of human PLD1 and PLD2, the development of PLD specific pharmacological inhibitors, and the identification of cellular regulators of PLD. In this review, we discuss the PLD1 and PLD2 structures, PLD inhibition by small molecules, and the regulation of PLD activity by effector proteins and lipids.
Collapse
Affiliation(s)
- Forrest Z Bowling
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Michael A Frohman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Michael V Airola
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
7
|
Pterostilbene Sensitizes Cisplatin-Resistant Human Bladder Cancer Cells with Oncogenic HRAS. Cancers (Basel) 2020; 12:cancers12102869. [PMID: 33036162 PMCID: PMC7650649 DOI: 10.3390/cancers12102869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/26/2020] [Accepted: 10/02/2020] [Indexed: 12/28/2022] Open
Abstract
Simple Summary RAS oncoproteins are considered undruggable cancer targets. Nearly 15% of cases of bladder cancer have a mutation of HRAS. The active HRAS contributes to the tumor progression and the risk of recurrence. Using our novel gene expression screening platform, pterostilbene was identified to sensitize cisplatin-resistant bladder cancer cells with HRAS alterations via RAS-related autophagy and cell senescence pathways, suggesting a potentially chemotherapeutic role of pterostilbene for cisplatin treatment of human bladder cancer with oncogenic HRAS. Pterostilbene is a safe and readily available food ingredient in edible plants worldwide. Exploiting the principle of combination therapy on pterostilbene-enhanced biosensitivity to identify undruggable molecular targets for cancer therapy may have a great possibility to overcome the cisplatin resistance of bladder cancer. Our data make HRAS a good candidate for modulation by pterostilbene for targeted cancer therapy in combination with conventional chemotherapeutic agents cisplatin plus gemcitabine. Abstract Analysis of various public databases revealed that HRAS gene mutation frequency and mRNA expression are higher in bladder urothelial carcinoma. Further analysis revealed the roles of oncogenic HRAS, autophagy, and cell senescence signaling in bladder cancer cells sensitized to the anticancer drug cisplatin using the phytochemical pterostilbene. A T24 cell line with the oncogenic HRAS was chosen for further experiments. Indeed, coadministration of pterostilbene increased stronger cytotoxicity on T24 cells compared to HRAS wild-type E7 cells, which was paralleled by neither elevated apoptosis nor induced cell cycle arrest, but rather a marked elevation of autophagy and cell senescence in T24 cells. Pterostilbene-induced autophagy in T24 cells was paralleled by inhibition of class I PI3K/mTOR/p70S6K as well as activation of MEK/ERK (a RAS target) and class III PI3K pathways. Pterostilbene-induced cell senescence on T24 cells was paralleled by increased pan-RAS and decreased phospho-RB expression. Coadministration of PI3K class III inhibitor 3-methyladenine or MEK inhibitor U0126 suppressed pterostilbene-induced autophagy and reversed pterostilbene-enhanced cytotoxicity, but did not affect pterostilbene-elevated cell senescence in T24 cells. Animal study data confirmed that pterostilbene enhanced cytotoxicity of cisplatin plus gemcitabine. These results suggest a therapeutic application of pterostilbene in cisplatin-resistant bladder cancer with oncogenic HRAS.
Collapse
|
8
|
Wimbish RT, DeLuca JG. Hec1/Ndc80 Tail Domain Function at the Kinetochore-Microtubule Interface. Front Cell Dev Biol 2020; 8:43. [PMID: 32161753 PMCID: PMC7054225 DOI: 10.3389/fcell.2020.00043] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 12/28/2022] Open
Abstract
Successful mitotic cell division is critically dependent on the formation of correct attachments between chromosomes and spindle microtubules. Microtubule attachments are mediated by kinetochores, which are large proteinaceous structures assembled on centromeric chromatin of mitotic chromosomes. These attachments must be sufficiently stable to transduce force; however, the strength of these attachments are also tightly regulated to ensure timely, error-free progression through mitosis. The highly conserved, kinetochore-associated NDC80 complex is a core component of the kinetochore-microtubule attachment machinery in eukaryotic cells. A small, disordered region within the Hec1 subunit of the NDC80 complex – the N-terminal “tail” domain – has been actively investigated during the last decade due to its roles in generating and regulating kinetochore-microtubule attachments. In this review, we discuss the role of the NDC80 complex, and specifically the Hec1 tail domain, at the kinetochore-microtubule interface, and how recent studies provide a more unified view of Hec1 tail domain function.
Collapse
Affiliation(s)
- Robert T Wimbish
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
9
|
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
|
10
|
Using the Pleiotropic Characteristics of Curcumin to Validate the Potential Application of a Novel Gene Expression Screening Platform. Nutrients 2019; 11:nu11061397. [PMID: 31234318 PMCID: PMC6627093 DOI: 10.3390/nu11061397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
Curcumin is a polyphenol derived from curcumin longa that exhibits anticancer and anti-inflammatory properties. The consumption of foods at supernutritional levels to obtain health benefits may paradoxically result in negative health outcomes. In the present study, multiple targeting characteristics of curcumin were analyzed using our gene expression screening system, which utilized the gene expression signatures of curcumin from human hepatocellular carcinoma and colorectal cancer cells to query gene expression databases and effectively identify the molecular actions of curcumin. In agreement with prediction, curcumin inhibited NF-κB and Aurora-A, and induced G2/M arrest and apoptosis. Curcumin-suppressed NF-κB was identified through inhibition of PLCG1, PIK3R1, and MALT1 in the CD4-T-cell-receptor-signaling NF-κB cascade pathway. The results suggest that our novel gene expression screening platform is an effective method of rapidly identifying unknown biological functions and side effects of compounds with potential nutraceutical benefits.
Collapse
|
11
|
Okabe S, Tauchi T, Tanaka Y, Ohyashiki K. Therapeutic targeting of Aurora A kinase in Philadelphia chromosome-positive ABL tyrosine kinase inhibitor-resistant cells. Oncotarget 2018; 9:32496-32506. [PMID: 30197758 PMCID: PMC6126699 DOI: 10.18632/oncotarget.25985] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/29/2018] [Indexed: 11/25/2022] Open
Abstract
Abelson murine leukemia viral oncogene homolog (ABL) tyrosine kinase inhibitors (TKIs) have been shown to be effective for treatment of chronic myeloid leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia patients. However, resistance to ABL TKIs can develop as a result of breakpoint cluster region-ABL point mutations. Aurora kinases regulate many processes associated with mitosis. In this study, we investigated whether inhibiting Aurora kinase can reduce the viability of Ph+ leukemia cells. Treatment with the Aurora kinase A inhibitor alisertib blocked Ph+ leukemia cell proliferation and Aurora kinase A phosphorylation; it also induced G2/M-phase arrest and increased the intracellular levels of reactive oxygen species. Combined treatment of Ph+ cells with ABL TKIs and alisertib was cytotoxic, with the fraction of senescent cells increasing in a time- and dose-dependent manner. Aurora A gene silencing suppressed cell proliferation and enhanced ABL TKI efficacy. In a mouse xenograft model, co-administration of ponatinib and alisertib enhanced survival and reduced tumor size; moreover, the treatments were well tolerated by the animals. These results indicate that inhibiting Aurora kinase can enhance the cytotoxic effects of ABL TKIs and is, therefore, an effective therapeutic strategy against ABL TKI-resistant cells, including those with the T315I mutation.
Collapse
Affiliation(s)
- Seiichi Okabe
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Tetsuzo Tauchi
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Yuko Tanaka
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Kazuma Ohyashiki
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
12
|
Abstract
Mitosis is controlled by reversible protein phosphorylation involving specific kinases and phosphatases. A handful of major mitotic protein kinases, such as the cyclin B-CDK1 complex, the Aurora kinases, and Polo-like kinase 1 (PLK1), cooperatively regulate distinct mitotic processes. Research has identified proteins and mechanisms that integrate these kinases into signaling cascades that guide essential mitotic events. These findings have important implications for our understanding of the mechanisms of mitotic regulation and may advance the development of novel antimitotic drugs. We review collected evidence that in vertebrates, the Aurora kinases serve as catalytic subunits of distinct complexes formed with the four scaffold proteins Bora, CEP192, INCENP, and TPX2, which we deem "core" Aurora cofactors. These complexes and the Aurora-PLK1 cascades organized by Bora, CEP192, and INCENP control crucial aspects of mitosis and all pathways of spindle assembly. We compare the mechanisms of Aurora activation in relation to the different spindle assembly pathways and draw a functional analogy between the CEP192 complex and the chromosomal passenger complex that may reflect the coevolution of centrosomes, kinetochores, and the actomyosin cleavage apparatus. We also analyze the roles and mechanisms of Aurora-PLK1 signaling in the cell and centrosome cycles and in the DNA damage response.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, Saint-Petersburg 197758, Russian Federation.
| | | |
Collapse
|
13
|
Inchanalkar S, Deshpande NU, Kasherwal V, Jayakannan M, Balasubramanian N. Polymer Nanovesicle-Mediated Delivery of MLN8237 Preferentially Inhibits Aurora Kinase A To Target RalA and Anchorage-Independent Growth in Breast Cancer Cells. Mol Pharm 2018; 15:3046-3059. [PMID: 29863884 DOI: 10.1021/acs.molpharmaceut.8b00163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The small GTPase RalA is a known mediator of anchorage-independent growth in cancers and is differentially regulated by adhesion and aurora kinase A (AURKA). Hence, inhibiting AURKA offers a means of specifically targeting RalA (over RalB) in cancer cells. MLN8237 (alisertib) is a known inhibitor of aurora kinases; its specificity for AURKA, however, is compromised by its poor solubility and transport across the cell membrane. A polymer nanovesicle platform is used for the first time to deliver and differentially inhibit AURKA in cancer cells. For this purpose, polysaccharide nanovesicles made from amphiphilic dextran were used as nanocarriers to successfully administer MLN8237 (VMLN) in cancer cells in 2D and 3D microenvironments. These nanovesicles (<200 nm) carry the drug in their intermembrane space with up to 85% of it released by the action of esterase enzyme(s). Lysotracker experiments reveal the polymer nanovesicles localize in the lysosomal compartment of the cell, where they are enzymatically targeted and MLN released in a controlled manner. Rhodamine B fluorophore trapped in the nanovesicles hydrophilic core (VMLN+RhB) allows us to visualize its uptake and localization in cells in a 2D and 3D microenvironment. In breast cancer, MCF-7 cells VMLN inhibits AURKA significantly better than the free drug at low concentrations (0.02-0.04 μM). This ensures that the drug in VMLN at these concentrations can specifically inhibit up to 94% of endogenous AURKA without affecting AURKB. This targeting of AURKA causes the downstream differential inhibition of active RalA (but not RalB). Free MLN8237 at similar concentrations and conditions failed to affect RalA activation. VMLN-mediated inhibition of RalA, in turn, disrupts the anchorage-independent growth of MCF-7 cells supporting a role for the AURKA-RalA crosstalk in mediating the same. These studies not only identify the polysaccharide nanovesicle to be an improved way to efficiently deliver low concentrations of MLN8237 to inhibit AURKA but, in doing so, also help reveal a role for AURKA and its crosstalk with RalA in anchorage-independent growth of MCF-7 cells.
Collapse
|
14
|
DeLuca JG. Aurora A Kinase Function at Kinetochores. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2018; 82:91-99. [PMID: 29700233 DOI: 10.1101/sqb.2017.82.034991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
One of the most important regulatory aspects of chromosome segregation is the ability of kinetochores to precisely control their attachment strength to spindle microtubules. Central to this regulation is Aurora B, a mitotic kinase that phosphorylates kinetochore substrates to promote microtubule turnover. A critical target of Aurora B is the kinetochore protein Ndc80/Hec1, which is a component of the NDC80 complex, the primary force-transducing link between kinetochores and microtubules. Although Aurora B is regarded as the "master regulator" of kinetochore-microtubule attachment, it is becoming clear that this kinase is not solely responsible for phosphorylating Hec1 and other kinetochore substrates to facilitate microtubule turnover. In particular, there is growing evidence that Aurora A kinase, whose activities at spindle poles have been extensively described, has additional roles at kinetochores in regulating the kinetochore-microtubule interface.
Collapse
Affiliation(s)
- Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523-1870
| |
Collapse
|
15
|
Dissection of Protein Kinase Pathways in Live Cells Using Photoluminescent Probes: Surveillance or Interrogation? CHEMOSENSORS 2018. [DOI: 10.3390/chemosensors6020019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Hamze-Komaiha O, Sarr S, Arlot-Bonnemains Y, Samuel D, Gassama-Diagne A. SHIP2 Regulates Lumen Generation, Cell Division, and Ciliogenesis through the Control of Basolateral to Apical Lumen Localization of Aurora A and HEF 1. Cell Rep 2017; 17:2738-2752. [PMID: 27926875 DOI: 10.1016/j.celrep.2016.11.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/05/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022] Open
Abstract
Lumen formation during epithelial morphogenesis requires the creation of a luminal space at cell interfaces named apical membrane-initiation sites (AMISs). This is dependent upon integrated signaling from mechanical and biochemical cues, vesicle trafficking, cell division, and processes tightly coupled to ciliogenesis. Deciphering relationships between polarity determinants and lumen or cilia generation remains a fundamental issue. Here, we report that Src homology 2 domain-containing inositol 5-phosphatase 2 (SHIP2), a basolateral determinant of polarity, regulates RhoA-dependent actin contractility and cell division to form AMISs. SHIP2 regulates mitotic spindle alignment. SHIP2 is expressed in G1 phase, whereas Aurora A kinase is enriched in mitosis. SHIP2 binds Aurora A kinase and the scaffolding protein HEF1 and promotes their basolateral localization at the expense of their luminal expression connected with cilia resorption. Furthermore, SHIP2 expression increases cilia length. Thus, our findings offer new insight into the relationships among basolateral proteins, lumen generation, and ciliogenesis.
Collapse
Affiliation(s)
- Ola Hamze-Komaiha
- Université Paris-Sud, 91400 Orsay, France; Unité 1193, 94800 Villejuif, France
| | - Sokavuth Sarr
- Université Paris-Sud, 91400 Orsay, France; Unité 1193, 94800 Villejuif, France
| | | | - Didier Samuel
- Université Paris-Sud, 91400 Orsay, France; Unité 1193, 94800 Villejuif, France; AP-HP Hôpital Paul Brousse, Centre Hépato-Biliaire, 94800 Villejuif, France
| | - Ama Gassama-Diagne
- Université Paris-Sud, 91400 Orsay, France; Unité 1193, 94800 Villejuif, France.
| |
Collapse
|
17
|
DeVaul N, Koloustroubis K, Wang R, Sperry AO. A novel interaction between kinase activities in regulation of cilia formation. BMC Cell Biol 2017; 18:33. [PMID: 29141582 PMCID: PMC5688660 DOI: 10.1186/s12860-017-0149-5] [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: 07/26/2017] [Accepted: 11/01/2017] [Indexed: 01/08/2023] Open
Abstract
Background The primary cilium is an extension of the cell membrane that encloses a microtubule-based axoneme. Primary cilia are essential for transmission of environmental cues that determine cell fate. Disruption of primary cilia function is the molecular basis of numerous developmental disorders. Despite their biological importance, the mechanisms governing their assembly and disassembly are just beginning to be understood. Cilia growth and disassembly are essential events when cells exit and reenter into the cell cycle. The kinases never in mitosis-kinase 2 (Nek2) and Aurora A (AurA) act to depolymerize cilia when cells reenter the cell cycle from G0. Results Coexpression of either kinase with its kinase dead companion [AurA with kinase dead Nek2 (Nek2 KD) or Nek2 with kinase dead AurA (AurA KD)] had different effects on cilia depending on whether cilia are growing or shortening. AurA and Nek2 are individually able to shorten cilia when cilia are growing but both are required when cilia are being absorbed. The depolymerizing activity of each kinase is increased when coexpressed with the kinase dead version of the other kinase but only when cilia are assembling. Additionally, the two kinases act additively when cilia are assembling but not disassembling. Inhibition of AurA increases cilia number while inhibition of Nek2 significantly stimulates cilia length. The complex functional relationship between the two kinases reflects their physical interaction. Further, we identify a role for a PP1 binding protein, PPP1R42, in inhibiting Nek2 and increasing ciliation of ARPE-19 cells. Conclusion We have uncovered a novel functional interaction between Nek2 and AurA that is dependent on the growth state of cilia. This differential interdependence reflects opposing regulation when cilia are growing or shortening. In addition to interaction between the kinases to regulate ciliation, the PP1 binding protein PPP1R42 directly inhibits Nek2 independent of PP1 indicating another level of regulation of this kinase. In summary, we demonstrate a complex interplay between Nek2 and AurA kinases in regulation of ciliation in ARPE-19 cells. Electronic supplementary material The online version of this article (10.1186/s12860-017-0149-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole DeVaul
- Laboratory of Biochemistry and Genetics, National Institute of Diabetics and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katerina Koloustroubis
- Anatomy and Cell Biology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - Rong Wang
- Anatomy and Cell Biology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - Ann O Sperry
- Anatomy and Cell Biology, East Carolina University, Brody School of Medicine, Greenville, NC, USA.
| |
Collapse
|
18
|
Vo TTL, Park JH, Seo JH, Lee EJ, Choi H, Bae SJ, Le H, An S, Lee HS, Wee HJ, Kim KW. ARD1-mediated aurora kinase A acetylation promotes cell proliferation and migration. Oncotarget 2017; 8:57216-57230. [PMID: 28915666 PMCID: PMC5593637 DOI: 10.18632/oncotarget.19332] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/30/2017] [Indexed: 11/25/2022] Open
Abstract
Aurora kinase A (AuA) is a prerequisite for centrosome maturation, separation, and mitotic spindle assembly, thus, it is essential for cell cycle regulation. Overexpression of AuA is implicated in poor prognosis of many types of cancer. However, the regulatory mechanisms underlying the functions of AuA are still not fully understood. Here, we report that AuA colocalizes with arrest defective protein 1 (ARD1) acetyltransferase during cell division and cell migration. Additionally, AuA is acetylated by ARD1 at lysine residues at positions 75 and 125. The double mutations at K75/K125 abolished the kinase activity of AuA. Moreover, the double mutant AuA exhibited diminished ability to promote cell proliferation and cell migration. Mechanistic studies revealed that AuA acetylation at K75/K125 promoted cell proliferation via activation of cyclin E/CDK2 and cyclin B1. In addition, AuA acetylation stimulated cell migration by activating the p38/AKT/MMP-2 pathway. Our findings indicate that ARD1-mediated acetylation of AuA enhances cell proliferation and migration, and probably contributes to cancer development.
Collapse
Affiliation(s)
- Tam Thuy Lu Vo
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Ji-Hyeon Park
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Ji Hae Seo
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu 42601, Korea
| | - Eun Ji Lee
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hoon Choi
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Sung-Jin Bae
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hoang Le
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Sunho An
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hye Shin Lee
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hee-Jun Wee
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Kyu-Won Kim
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.,Crop Biotechnology Institute, GreenBio Science and Technology, Seoul National University, Pyeongchang 25354, Korea
| |
Collapse
|
19
|
Fu J, Bian M, Xin G, Deng Z, Luo J, Guo X, Chen H, Wang Y, Jiang Q, Zhang C. TPX2 phosphorylation maintains metaphase spindle length by regulating microtubule flux. J Cell Biol 2016; 210:373-83. [PMID: 26240182 PMCID: PMC4523612 DOI: 10.1083/jcb.201412109] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
TPX2 is phosphorylated by Aurora A and is essential for normal microtubule flux on the metaphase spindle. A steady-state metaphase spindle maintains constant length, although the microtubules undergo intensive dynamics. Tubulin dimers are incorporated at plus ends of spindle microtubules while they are removed from the minus ends, resulting in poleward movement. Such microtubule flux is regulated by the microtubule rescue factors CLASPs at kinetochores and depolymerizing protein Kif2a at the poles, along with other regulators of microtubule dynamics. How microtubule polymerization and depolymerization are coordinated remains unclear. Here we show that TPX2, a microtubule-bundling protein and activator of Aurora A, plays an important role. TPX2 was phosphorylated by Aurora A during mitosis. Its phospho-null mutant caused short metaphase spindles coupled with low microtubule flux rate. Interestingly, phosphorylation of TPX2 regulated its interaction with CLASP1 but not Kif2a. The effect of its mutant in shortening the spindle could be rescued by codepletion of CLASP1 and Kif2a that abolished microtubule flux. Together we propose that Aurora A–dependent TPX2 phosphorylation controls mitotic spindle length through regulating microtubule flux.
Collapse
Affiliation(s)
- Jingyan Fu
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Minglei Bian
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guangwei Xin
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Zhaoxuan Deng
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jia Luo
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Xiao Guo
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hao Chen
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yao Wang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
20
|
Garrido G, Vernos I. Non-centrosomal TPX2-Dependent Regulation of the Aurora A Kinase: Functional Implications for Healthy and Pathological Cell Division. Front Oncol 2016; 6:88. [PMID: 27148480 PMCID: PMC4831974 DOI: 10.3389/fonc.2016.00088] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/29/2016] [Indexed: 01/09/2023] Open
Abstract
Aurora A has been extensively characterized as a centrosomal kinase with essential functions during cell division including centrosome maturation and separation and spindle assembly. However, Aurora A localization is not restricted to the centrosomes and compelling evidence support the existence of specific mechanisms of activation and functions for non-centrosomal Aurora A in the dividing cell. It has been now well established that spindle assembly involves an acentrosomal RanGTP-dependent pathway that triggers microtubule assembly and organization in the proximity of the chromosomes whether centrosomes are present or not. The mechanism involves the regulation of a number of NLS-containing proteins, generically called SAFS (Spindle Assembly Factors) that exert their functions upon release from karyopherins by RanGTP. One of them, the nuclear protein TPX2 interacts with and activates Aurora A upon release from importins by RanGTP. This basic mechanism triggers the activation of Aurora A in the proximity of the chromosomes potentially translating the RanGTP signaling gradient centered on the chromosome into an Aurora A phosphorylation network. Here, we will review our current knowledge on the RanGTP-dependent TPX2 activation of Aurora A away from centrosomes: from the mechanism of activation and its functional consequences on the kinase stability and regulation to its roles in spindle assembly and cell division. We will then focus on the substrates of the TPX2-activated Aurora A having a role in microtubule nucleation, stabilization, and organization. Finally, we will briefly discuss the implications of the use of Aurora A inhibitors in anti-tumor therapies in the light of its functional interaction with TPX2.
Collapse
Affiliation(s)
- Georgina Garrido
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
21
|
Weiderhold KN, Fadri-Moskwik M, Pan J, Nishino M, Chuang C, Deeraksa A, Lin SH, Yu-Lee LY. Dynamic Phosphorylation of NudC by Aurora B in Cytokinesis. PLoS One 2016; 11:e0153455. [PMID: 27074040 PMCID: PMC4830538 DOI: 10.1371/journal.pone.0153455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
Nuclear distribution protein C (NudC) is a mitotic regulator that plays a role in cytokinesis. However, how NudC is regulated during cytokinesis remains unclear. Here, we show that NudC is phosphorylated by Aurora B, a kinase critical for cell abscission. NudC is co-localized with Aurora B at the midbody and co-immunoprecipitated with Aurora B in mitosis. Inhibition of Aurora B by ZM447439 reduced NudC phosphorylation, suggesting that NudC is an Aurora B substrate in vivo. We identified T40 on NudC as an Aurora B phosphorylation site. NudC depletion resulted in cytokinesis failure with a dramatic elongation of the intercellular bridge between daughter cells, sustained Aurora B activity at the midbody, and reduced cell abscission. These cytokinetic defects can be rescued by the ectopic expression of wild-type NudC. Reconstitution with T40A phospho-defective NudC was found to rescue the cytokinesis defect. In contrast, reconstitution with the T40D phospho-mimetic NudC was inefficient in supporting the completion of cytokinesis. These results suggest that that dynamic phosphorylation of NudC by Aurora B regulates cytokinesis.
Collapse
Affiliation(s)
- Kimberly N. Weiderhold
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Maria Fadri-Moskwik
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jing Pan
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michiya Nishino
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Carol Chuang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Arpaporn Deeraksa
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Li-Yuan Yu-Lee
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine, Section of Allergy Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
22
|
Diaz RJ, Golbourn B, Faria C, Picard D, Shih D, Raynaud D, Leadly M, MacKenzie D, Bryant M, Bebenek M, Smith CA, Taylor MD, Huang A, Rutka JT. Mechanism of action and therapeutic efficacy of Aurora kinase B inhibition in MYC overexpressing medulloblastoma. Oncotarget 2016; 6:3359-74. [PMID: 25739120 PMCID: PMC4413659 DOI: 10.18632/oncotarget.3245] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 12/24/2014] [Indexed: 12/31/2022] Open
Abstract
Medulloblastoma comprises four molecular subgroups of which Group 3 medulloblastoma is characterized by MYC amplification and MYC overexpression. Lymphoma cells expressing high levels of MYC are susceptible to apoptosis following treatment with inhibitors of mitosis. One of the key regulatory kinases involved in multiple stages of mitosis is Aurora kinase B. We hypothesized that medulloblastoma cells that overexpress MYC would be uniquely sensitized to the apoptotic effects of Aurora B inhibition. The specific inhibition of Aurora kinase B was achieved in MYC-overexpressing medulloblastoma cells with AZD1152-HQPA. MYC overexpression sensitized medulloblastoma cells to cell death upon Aurora B inhibition. This process was found to be independent of endoreplication. Using both flank and intracranial cerebellar xenografts we demonstrate that tumors formed from MYC-overexpressing medulloblastoma cells show a response to Aurora B inhibition including growth impairment and apoptosis induction. Lastly, we show the distribution of AZD1152-HQPA within the mouse brain and the ability to inhibit intracranial tumor growth and prolong survival in mice bearing tumors formed from MYC-overexpressing medulloblastoma cells. Our results suggest the potential for therapeutic application of Aurora kinase B inhibitors in the treatment of Group 3 medulloblastoma.
Collapse
Affiliation(s)
- Roberto Jose Diaz
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Brian Golbourn
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Claudia Faria
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Daniel Picard
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - David Shih
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Denis Raynaud
- Analytical Facility for Bioactive Molecules, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Leadly
- Analytical Facility for Bioactive Molecules, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Danielle MacKenzie
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Melissa Bryant
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Matthew Bebenek
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Christian A Smith
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - Michael D Taylor
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Annie Huang
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | - James T Rutka
- The Hospital for Sick Children. Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Chou EJ, Hung LY, Tang CJC, Hsu WB, Wu HY, Liao PC, Tang TK. Phosphorylation of CPAP by Aurora-A Maintains Spindle Pole Integrity during Mitosis. Cell Rep 2016; 14:2975-87. [PMID: 26997271 DOI: 10.1016/j.celrep.2016.02.085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/26/2016] [Accepted: 02/23/2016] [Indexed: 02/01/2023] Open
Abstract
CPAP is required for centriole elongation during S/G2 phase, but the role of CPAP in mitosis is incompletely understood. Here, we show that CPAP maintains spindle pole integrity through its phosphorylation by Aurora-A during mitosis. Depletion of CPAP induced a prolonged delay in mitosis, pericentriolar material (PCM) dispersion, and multiple mitotic abnormalities. Further studies demonstrated that CPAP directly interacts with and is phosphorylated by Aurora-A at serine 467 during mitosis. Interestingly, the dispersal of the PCM was effectively rescued by ectopic expression of wild-type CPAP or a phospho-mimic CPAP-S467D mutant, but not a non-phosphorylated CPAP-S467A mutant. Finally, we found that CPAP-S467D has a low affinity for microtubule binding but a high affinity for PCM proteins. Together, our results support a model wherein CPAP is required for proper mitotic progression, and phosphorylation of CPAP by Aurora-A is essential for maintaining spindle pole integrity.
Collapse
Affiliation(s)
- En-Ju Chou
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Liang-Yi Hung
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chieh-Ju C Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Wen-Bin Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Hsin-Yi Wu
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pao-Chi Liao
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tang K Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
24
|
Al-Bataineh MM, Alzamora R, Ohmi K, Ho PY, Marciszyn AL, Gong F, Li H, Hallows KR, Pastor-Soler NM. Aurora kinase A activates the vacuolar H+-ATPase (V-ATPase) in kidney carcinoma cells. Am J Physiol Renal Physiol 2016; 310:F1216-28. [PMID: 26911844 DOI: 10.1152/ajprenal.00061.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/19/2016] [Indexed: 11/22/2022] Open
Abstract
Extracellular proton-secreting transport systems that contribute to extracellular pH include the vacuolar H(+)-ATPase (V-ATPase). This pump, which mediates ATP-driven transport of H(+) across membranes, is involved in metastasis. We previously showed (Alzamora R, Thali RF, Gong F, Smolak C, Li H, Baty CJ, Bertrand CA, Auchli Y, Brunisholz RA, Neumann D, Hallows KR, Pastor-Soler NM. J Biol Chem 285: 24676-24685, 2010) that V-ATPase A subunit phosphorylation at Ser-175 is important for PKA-induced V-ATPase activity at the membrane of kidney intercalated cells. However, Ser-175 is also located within a larger phosphorylation consensus sequence for Aurora kinases, which are known to phosphorylate proteins that contribute to the pathogenesis of metastatic carcinomas. We thus hypothesized that Aurora kinase A (AURKA), overexpressed in aggressive carcinomas, regulates the V-ATPase in human kidney carcinoma cells (Caki-2) via Ser-175 phosphorylation. We found that AURKA is abnormally expressed in Caki-2 cells, where it binds the V-ATPase A subunit in an AURKA phosphorylation-dependent manner. Treatment with the AURKA activator anacardic acid increased V-ATPase expression and activity at the plasma membrane of Caki-2 cells. In addition, AURKA phosphorylates the V-ATPase A subunit at Ser-175 in vitro and in Caki-2 cells. Immunolabeling revealed that anacardic acid induced marked membrane accumulation of the V-ATPase A subunit in transfected Caki-2 cells. However, anacardic acid failed to induce membrane accumulation of a phosphorylation-deficient Ser-175-to-Ala (S175A) A subunit mutant. Finally, S175A-expressing cells had decreased migration in a wound-healing assay compared with cells expressing wild-type or a phospho-mimetic Ser-175-to-Asp (S175D) mutant A subunit. We conclude that AURKA activates the V-ATPase in kidney carcinoma cells via phosphorylation of Ser-175 in the V-ATPase A subunit. This regulation contributes to kidney carcinoma V-ATPase-mediated extracellular acidification and cell migration.
Collapse
Affiliation(s)
- Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Rodrigo Alzamora
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Kazuhiro Ohmi
- Department of Medicine, USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of USC, Los Angeles, California
| | - Pei-Yin Ho
- Department of Medicine, USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of USC, Los Angeles, California
| | - Allison L Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Fan Gong
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Hui Li
- Department of Medicine, USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of USC, Los Angeles, California
| | - Kenneth R Hallows
- Department of Medicine, USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of USC, Los Angeles, California
| | - Núria M Pastor-Soler
- Department of Medicine, USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of USC, Los Angeles, California
| |
Collapse
|
25
|
Weimer AK, Demidov D, Lermontova I, Beeckman T, Van Damme D. Aurora Kinases Throughout Plant Development. TRENDS IN PLANT SCIENCE 2016; 21:69-79. [PMID: 26616196 DOI: 10.1016/j.tplants.2015.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/10/2015] [Accepted: 10/02/2015] [Indexed: 06/05/2023]
Abstract
Aurora kinases are evolutionarily conserved key mitotic determinants in all eukaryotes. Yeasts contain a single Aurora kinase, whereas multicellular eukaryotes have at least two functionally diverged members. The involvement of Aurora kinases in human cancers has provided an in-depth mechanistic understanding of their roles throughout cell division in animal and yeast models. By contrast, understanding Aurora kinase function in plants is only starting to emerge. Nevertheless, genetic, cell biological, and biochemical approaches have revealed functional diversification between the plant Aurora kinases and suggest a role in formative (asymmetric) divisions, chromatin modification, and genome stability. This review provides an overview of the accumulated knowledge on the function of plant Aurora kinases as well as some major challenges for the future.
Collapse
Affiliation(s)
- Annika K Weimer
- Department of Plant Systems Biology, Flanders Institute for Biotechnology (VIB), 9052 Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Dmitri Demidov
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Stadt Seeland, 06466 Germany
| | - Inna Lermontova
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Stadt Seeland, 06466 Germany
| | - Tom Beeckman
- Department of Plant Systems Biology, Flanders Institute for Biotechnology (VIB), 9052 Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Daniël Van Damme
- Department of Plant Systems Biology, Flanders Institute for Biotechnology (VIB), 9052 Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
26
|
de Groot CO, Hsia JE, Anzola JV, Motamedi A, Yoon M, Wong YL, Jenkins D, Lee HJ, Martinez MB, Davis RL, Gahman TC, Desai A, Shiau AK. A Cell Biologist's Field Guide to Aurora Kinase Inhibitors. Front Oncol 2015; 5:285. [PMID: 26732741 PMCID: PMC4685510 DOI: 10.3389/fonc.2015.00285] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/03/2015] [Indexed: 01/19/2023] Open
Abstract
Aurora kinases are essential for cell division and are frequently misregulated in human cancers. Based on their potential as cancer therapeutics, a plethora of small molecule Aurora kinase inhibitors have been developed, with a subset having been adopted as tools in cell biology. Here, we fill a gap in the characterization of Aurora kinase inhibitors by using biochemical and cell-based assays to systematically profile a panel of 10 commercially available compounds with reported selectivity for Aurora A (MLN8054, MLN8237, MK-5108, MK-8745, Genentech Aurora Inhibitor 1), Aurora B (Hesperadin, ZM447439, AZD1152-HQPA, GSK1070916), or Aurora A/B (VX-680). We quantify the in vitro effect of each inhibitor on the activity of Aurora A alone, as well as Aurora A and Aurora B bound to fragments of their activators, TPX2 and INCENP, respectively. We also report kinome profiling results for a subset of these compounds to highlight potential off-target effects. In a cellular context, we demonstrate that immunofluorescence-based detection of LATS2 and histone H3 phospho-epitopes provides a facile and reliable means to assess potency and specificity of Aurora A versus Aurora B inhibition, and that G2 duration measured in a live imaging assay is a specific readout of Aurora A activity. Our analysis also highlights variation between HeLa, U2OS, and hTERT-RPE1 cells that impacts selective Aurora A inhibition. For Aurora B, all four tested compounds exhibit excellent selectivity and do not significantly inhibit Aurora A at effective doses. For Aurora A, MK-5108 and MK-8745 are significantly more selective than the commonly used inhibitors MLN8054 and MLN8237. A crystal structure of an Aurora A/MK-5108 complex that we determined suggests the chemical basis for this higher specificity. Taken together, our quantitative biochemical and cell-based analyses indicate that AZD1152-HQPA and MK-8745 are the best current tools for selectively inhibiting Aurora B and Aurora A, respectively. However, MK-8745 is not nearly as ideal as AZD1152-HQPA in that it requires high concentrations to achieve full inhibition in a cellular context, indicating a need for more potent Aurora A-selective inhibitors. We conclude with a set of “good practice” guidelines for the use of Aurora inhibitors in cell biology experiments.
Collapse
Affiliation(s)
- Christian O de Groot
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Judy E Hsia
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - John V Anzola
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Amir Motamedi
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Michelle Yoon
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Yao Liang Wong
- Laboratory of Chromosome Biology, Ludwig Institute for Cancer Research, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - David Jenkins
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Hyun J Lee
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Mallory B Martinez
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Robert L Davis
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Timothy C Gahman
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| | - Arshad Desai
- Laboratory of Chromosome Biology, Ludwig Institute for Cancer Research, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research , La Jolla, CA , USA
| |
Collapse
|
27
|
Rebelo S, Santos M, Martins F, da Cruz e Silva EF, da Cruz e Silva OA. Protein phosphatase 1 is a key player in nuclear events. Cell Signal 2015; 27:2589-98. [DOI: 10.1016/j.cellsig.2015.08.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 12/17/2022]
|
28
|
Polat AN, Karayel Ö, Giese SH, Harmanda B, Sanal E, Hu CK, Renard BY, Özlü N. Phosphoproteomic Analysis of Aurora Kinase Inhibition in Monopolar Cytokinesis. J Proteome Res 2015; 14:4087-98. [PMID: 26270265 DOI: 10.1021/acs.jproteome.5b00645] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cytokinesis is the last step of the cell cycle that requires coordinated activities of the microtubule cytoskeleton, actin cytoskeleton, and membrane compartments. Aurora B kinase is one of the master regulatory kinases that orchestrate multiple events during cytokinesis. To reveal targets of the Aurora B kinase, we combined quantitative mass spectrometry with chemical genetics. Using the quantitative proteomic approach, SILAC (stable isotope labeling with amino acids in cell culture), we analyzed the phosphoproteome of monopolar cytokinesis upon VX680- or AZD1152-mediated aurora kinase inhibition. In total, our analysis quantified over 20 000 phosphopeptides in response to the Aurora-B kinase inhibition; 246 unique phosphopeptides were significantly down-regulated and 74 were up-regulated. Our data provide a broad analysis of downstream effectors of Aurora kinase and offer insights into how Aurora kinase regulates cytokinesis.
Collapse
Affiliation(s)
- Ayse Nur Polat
- Department of Molecular Biology and Genetics, Koç University , Fen Fakultesi, Sariyer, Istanbul 34450, Turkey
| | - Özge Karayel
- Department of Molecular Biology and Genetics, Koç University , Fen Fakultesi, Sariyer, Istanbul 34450, Turkey
| | - Sven H Giese
- Research Group Bioinformatics (NG 4), Robert Koch-Institute , Postfach 65 02 61, D-13302 Berlin, Germany.,Department of Bioanalytics, Institute of Biotechnology, Technische Universität Berlin , Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Büşra Harmanda
- Department of Molecular Biology and Genetics, Koç University , Fen Fakultesi, Sariyer, Istanbul 34450, Turkey
| | - Erdem Sanal
- Department of Molecular Biology and Genetics, Koç University , Fen Fakultesi, Sariyer, Istanbul 34450, Turkey
| | - Chi-Kuo Hu
- Department of Genetics, Stanford University, School of Medicine , 291 Campus Drive, Li Ka Shing Building, Stanford, California 94305, United States
| | - Bernhard Y Renard
- Research Group Bioinformatics (NG 4), Robert Koch-Institute , Postfach 65 02 61, D-13302 Berlin, Germany
| | - Nurhan Özlü
- Department of Molecular Biology and Genetics, Koç University , Fen Fakultesi, Sariyer, Istanbul 34450, Turkey
| |
Collapse
|
29
|
Yang XH, Li M, Wang B, Zhu W, Desgardin A, Onel K, de Jong J, Chen J, Chen L, Cunningham JM. Systematic computation with functional gene-sets among leukemic and hematopoietic stem cells reveals a favorable prognostic signature for acute myeloid leukemia. BMC Bioinformatics 2015; 16:97. [PMID: 25887548 PMCID: PMC4376348 DOI: 10.1186/s12859-015-0510-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 02/24/2015] [Indexed: 12/16/2022] Open
Abstract
Background Genes that regulate stem cell function are suspected to exert adverse effects on prognosis in malignancy. However, diverse cancer stem cell signatures are difficult for physicians to interpret and apply clinically. To connect the transcriptome and stem cell biology, with potential clinical applications, we propose a novel computational “gene-to-function, snapshot-to-dynamics, and biology-to-clinic” framework to uncover core functional gene-sets signatures. This framework incorporates three function-centric gene-set analysis strategies: a meta-analysis of both microarray and RNA-seq data, novel dynamic network mechanism (DNM) identification, and a personalized prognostic indicator analysis. This work uses complex disease acute myeloid leukemia (AML) as a research platform. Results We introduced an adjustable “soft threshold” to a functional gene-set algorithm and found that two different analysis methods identified distinct gene-set signatures from the same samples. We identified a 30-gene cluster that characterizes leukemic stem cell (LSC)-depleted cells and a 25-gene cluster that characterizes LSC-enriched cells in parallel; both mark favorable-prognosis in AML. Genes within each signature significantly share common biological processes and/or molecular functions (empirical p = 6e-5 and 0.03 respectively). The 25-gene signature reflects the abnormal development of stem cells in AML, such as AURKA over-expression. We subsequently determined that the clinical relevance of both signatures is independent of known clinical risk classifications in 214 patients with cytogenetically normal AML. We successfully validated the prognosis of both signatures in two independent cohorts of 91 and 242 patients respectively (log-rank p < 0.0015 and 0.05; empirical p < 0.015 and 0.08). Conclusion The proposed algorithms and computational framework will harness systems biology research because they efficiently translate gene-sets (rather than single genes) into biological discoveries about AML and other complex diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0510-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinan Holly Yang
- Department of Pediatrics, and Comer Children's Hospital, Section of Hematology/Oncology, The University of Chicago, 900 East 57th Street, KCBD Room 5121, Chicago, Illinois, 60637, USA.
| | - Meiyi Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Bin Wang
- Department of Pediatrics, and Comer Children's Hospital, Section of Hematology/Oncology, The University of Chicago, 900 East 57th Street, KCBD Room 5121, Chicago, Illinois, 60637, USA.
| | - Wanqi Zhu
- Laboratory Schools, The University of Chicago, Chicago, USA.
| | - Aurelie Desgardin
- Department of Pediatrics, and Comer Children's Hospital, Section of Hematology/Oncology, The University of Chicago, 900 East 57th Street, KCBD Room 5121, Chicago, Illinois, 60637, USA.
| | - Kenan Onel
- Department of Pediatrics, and Comer Children's Hospital, Section of Hematology/Oncology, The University of Chicago, 900 East 57th Street, KCBD Room 5121, Chicago, Illinois, 60637, USA.
| | - Jill de Jong
- Department of Pediatrics, and Comer Children's Hospital, Section of Hematology/Oncology, The University of Chicago, 900 East 57th Street, KCBD Room 5121, Chicago, Illinois, 60637, USA.
| | - Jianjun Chen
- Department of Medicine, The University of Chicago, Chicago, USA.
| | - Luonan Chen
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - John M Cunningham
- Department of Pediatrics, and Comer Children's Hospital, Section of Hematology/Oncology, The University of Chicago, 900 East 57th Street, KCBD Room 5121, Chicago, Illinois, 60637, USA.
| |
Collapse
|
30
|
Korrodi-Gregório L, Esteves SLC, Fardilha M. Protein phosphatase 1 catalytic isoforms: specificity toward interacting proteins. Transl Res 2014; 164:366-91. [PMID: 25090308 DOI: 10.1016/j.trsl.2014.07.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/26/2014] [Accepted: 07/01/2014] [Indexed: 01/21/2023]
Abstract
The coordinated and reciprocal action of serine-threonine protein kinases and protein phosphatases produces transitory phosphorylation, a fundamental regulatory mechanism for many biological processes. Phosphoprotein phosphatase 1 (PPP1), a major serine-threonine phosphatase, in particular, is ubiquitously distributed and regulates a broad range of cellular functions, including glycogen metabolism, cell cycle progression, and muscle relaxation. PPP1 has evolved effective catalytic machinery but in vitro lacks substrate specificity. In vivo, its specificity is achieved not only by the existence of different PPP1 catalytic isoforms, but also by binding of the catalytic moiety to a large number of regulatory or targeting subunits. Here, we will address exhaustively the existence of diverse PPP1 catalytic isoforms and the relevance of their specific partners and consequent functions.
Collapse
Affiliation(s)
- Luís Korrodi-Gregório
- Laboratório de Transdução de Sinais, Departamento de Biologia, Secção Autónoma de Ciências de Saúde, Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal
| | - Sara L C Esteves
- Laboratório de Transdução de Sinais, Departamento de Biologia, Secção Autónoma de Ciências de Saúde, Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal
| | - Margarida Fardilha
- Laboratório de Transdução de Sinais, Departamento de Biologia, Secção Autónoma de Ciências de Saúde, Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal.
| |
Collapse
|
31
|
Jung Y, Shin SY, Yong Y, Jung H, Ahn S, Lee YH, Lim Y. Plant-Derived Flavones as Inhibitors of Aurora B Kinase and Their Quantitative Structure-Activity Relationships. Chem Biol Drug Des 2014; 85:574-85. [DOI: 10.1111/cbdd.12445] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 09/12/2014] [Accepted: 09/30/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Yearam Jung
- Division of Bioscience and Biotechnology; BMIC; Konkuk University; Seoul 143-701 Korea
| | - Soon Young Shin
- Department of Biological Sciences; Konkuk University; Seoul 143-701 Korea
| | - Yeonjoong Yong
- Division of Bioscience and Biotechnology; BMIC; Konkuk University; Seoul 143-701 Korea
| | - Hyeryoung Jung
- Division of Bioscience and Biotechnology; BMIC; Konkuk University; Seoul 143-701 Korea
| | - Seunghyun Ahn
- Division of Bioscience and Biotechnology; BMIC; Konkuk University; Seoul 143-701 Korea
| | - Young Han Lee
- Department of Biological Sciences; Konkuk University; Seoul 143-701 Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology; BMIC; Konkuk University; Seoul 143-701 Korea
| |
Collapse
|
32
|
Neumayer G, Belzil C, Gruss OJ, Nguyen MD. TPX2: of spindle assembly, DNA damage response, and cancer. Cell Mol Life Sci 2014; 71:3027-47. [PMID: 24556998 PMCID: PMC11114040 DOI: 10.1007/s00018-014-1582-7] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 10/25/2022]
Abstract
For more than 15 years, TPX2 has been studied as a factor critical for mitosis and spindle assembly. These functions of TPX2 are attributed to its Ran-regulated microtubule-associated protein properties and to its control of the Aurora A kinase. Overexpressed in cancers, TPX2 is being established as marker for the diagnosis and prognosis of malignancies. During interphase, TPX2 resides preferentially in the nucleus where its function had remained elusive until recently. The latest finding that TPX2 plays a role in amplification of the DNA damage response, combined with the characterization of TPX2 knockout mice, open new perspectives to understand the biology of this protein. This review provides an historic overview of the discovery of TPX2 and summarizes its cytoskeletal and signaling roles with relevance to cancer therapies. Finally, the review aims to reconcile discrepancies between the experimental and pathological effects of TPX2 overexpression and advances new roles for compartmentalized TPX2.
Collapse
Affiliation(s)
- Gernot Neumayer
- Department of Clinical Neurosciences, Department of Cell Biology and Anatomy, Department of Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada,
| | | | | | | |
Collapse
|
33
|
Zorba A, Buosi V, Kutter S, Kern N, Pontiggia F, Cho YJ, Kern D. Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2. eLife 2014; 3:e02667. [PMID: 24867643 PMCID: PMC4032492 DOI: 10.7554/elife.02667] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We elucidate the molecular mechanisms of two distinct activation strategies (autophosphorylation and TPX2-mediated activation) in human Aurora A kinase. Classic allosteric activation is in play where either activation loop phosphorylation or TPX2 binding to a conserved hydrophobic groove shifts the equilibrium far towards the active conformation. We resolve the controversy about the mechanism of autophosphorylation by demonstrating intermolecular autophosphorylation in a long-lived dimer by combining X-ray crystallography with functional assays. We then address the allosteric activation by TPX2 through activity assays and the crystal structure of a domain-swapped dimer of dephosphorylated Aurora A and TPX21−25. While autophosphorylation is the key regulatory mechanism in the centrosomes in the early stages of mitosis, allosteric activation by TPX2 of dephosphorylated Aurora A could be at play in the spindle microtubules. The mechanistic insights into autophosphorylation and allosteric activation by TPX2 binding proposed here, may have implications for understanding regulation of other protein kinases. DOI:http://dx.doi.org/10.7554/eLife.02667.001 The kinase, Aurora A, is a human protein that is needed for cells to divide normally. Kinases are enzymes that control other proteins by adding phosphate groups to these proteins; however, like other kinases, Aurora A must first be activated or ‘switched on’ before it can do this. Aurora A kinase can be switched on in two ways: by having a phosphate group added to its ‘activation loop’; or by binding to another protein called TPX2. Also like other kinases, Aurora A can self-activate, but the details of this process are not understood. Does a single Aurora A kinase add a phosphate group to its own activation loop, or does one Aurora A kinase activate a second? Furthermore, it is not clear how binding to TPX2 can activate an Aurora A kinase without adding a phosphate group to the activation loop. Zorba, Buosi et al. now show that Aurora A kinases that have been activated in different ways—via the addition of a phosphate group or binding to TPX2—are equally good at adding phosphate groups to other proteins. Zorba, Buosi et al. also worked out the three-dimensional shapes of the kinases activated in these two ways—since many proteins change shape when they are switched on—and found that they were also the same. Finally, it was shown that self-activation involves two Aurora A kinases binding to each other, and one kinase adding a phosphate group to the other, rather than a single kinase adding a phosphate group to itself. Since other protein kinases can be activated in similar ways to Aurora A, the findings of Zorba, Buosi et al. might also help us to understand how other protein kinases can be switched ‘on’ or ‘off’. And, as mutations in Aurora A have been linked to the development of cancer, uncovering how this kinase is controlled could help efforts to design new drugs to treat this disease. DOI:http://dx.doi.org/10.7554/eLife.02667.002
Collapse
Affiliation(s)
- Adelajda Zorba
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Vanessa Buosi
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Steffen Kutter
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Nadja Kern
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Francesco Pontiggia
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Young-Jin Cho
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Dorothee Kern
- Department of Biochemistry, Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| |
Collapse
|
34
|
Kumar A, Rajendran V, Sethumadhavan R, Purohit R. Computational investigation of cancer-associated molecular mechanism in Aurora A (S155R) mutation. Cell Biochem Biophys 2014; 66:787-96. [PMID: 23412841 DOI: 10.1007/s12013-013-9524-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Centrosomes are the key-regulating element of cell cycle progression. Aberrations in their functional mechanism lead to several cancer-related disorders. Aurora A protein is a centrosome-associated protein that regulates the centriole duplication and its abberations are associated with multiple cases of aneuploidy and cancer-related disorders. S155R mutation in Aurora A is reported to induce cancer like phenotype and disrupt its binding with TPX2 protein. In this study, we have demonstrated the structural consequences of Aurora A S155R mutation and the atomic changes that influenced the loss of TPX2-binding affinity. Docking and molecular dynamics simulation results suggested significant loss in atomic contacts between mutant Aurora A and TPX2 protein. Further, we observed a notable changes in conformation of mutant Aurora A-TPX2 docked complex as compared to the native. Loss of binding affinity rendered the TPX2 domain free which then induced unfolding in its coiled region and enabled the overall expansion of mutant complex as compared to the native. The significant outcomes obtained from this study will facilitate in future cancer researches and in developing the potent drug therapies.
Collapse
Affiliation(s)
- Ambuj Kumar
- Bioinformatics Division, School of Bio Sciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | | | | | | |
Collapse
|
35
|
Ke CS, Liu HS, Yen CH, Huang GC, Cheng HC, Huang CYF, Su CL. Curcumin-induced Aurora-A suppression not only causes mitotic defect and cell cycle arrest but also alters chemosensitivity to anticancer drugs. J Nutr Biochem 2014; 25:526-39. [PMID: 24613085 DOI: 10.1016/j.jnutbio.2014.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 12/29/2013] [Accepted: 01/08/2014] [Indexed: 01/19/2023]
Abstract
Overexpression of oncoprotein Aurora-A increases drug resistance and promotes lung metastasis of breast cancer cells. Curcumin is an active anticancer compound in turmeric and curry. Here we observed that Aurora-A protein and kinase activity were reduced in curcumin-treated human breast chemoresistant nonmetastatic MCF-7 and highly metastatic cancer MDA-MB-231 cells. Curcumin acts in a similar manner to Aurora-A small interfering RNA (siRNA), resulting in monopolar spindle formation, S and G2/M arrest, and cell division reduction. Ectopic Aurora-A extinguished the curcumin effects. The anticancer effects of curcumin were enhanced by Aurora-A siRNA and produced additivity and synergism effects in cell division and monopolar phenotype, respectively. Combination treatment with curcumin overrode the chemoresistance to four Food and Drug Administration (FDA)-approved anticancer drugs (ixabepilone, cisplatin, vinorelbine, or everolimus) in MDA-MB-231 cells, which was characterized by a decrease in cell viability and the occurrence of an additivity or synergy effect. Ectopic expression of Aurora-A attenuated curcumin-enhanced chemosensitivity to these four tested drugs. A similar benefit of curcumin was observed in MCF-7 cells treated with ixabepilone, the primary systemic therapy to patients with invasive breast cancer (stages IIA-IIIB) before surgery. Antagonism effect was observed when MCF-7 cells were treated with curcumin plus cisplatin, vinorelbine or everolimus. Curcumin-induced enhancement in chemosensitivity was paralleled by significant increases (additivity or synergy effect) in apoptosis and cell cycle arrest at S and G2/M phases, the consequences of Aurora-A inhibition. These results suggest that a combination of curcumin with FDA-approved anticancer drugs warrants further assessment with a view to developing a novel clinical treatment for breast cancer.
Collapse
Affiliation(s)
- Ching-Shiun Ke
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Center of Infectious Disease and Signaling Research Center, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Cheng-Hsin Yen
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan
| | - Guan-Cheng Huang
- Division of Hemato-oncology, Department of Internal Medicine, Yuan's General Hospital, Kaohsiung 802, Taiwan
| | - Hung-Chi Cheng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi-Ying F Huang
- Institute of BioPharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan
| | - Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan.
| |
Collapse
|
36
|
Silva VC, Cassimeris L. Stathmin and microtubules regulate mitotic entry in HeLa cells by controlling activation of both Aurora kinase A and Plk1. Mol Biol Cell 2013; 24:3819-31. [PMID: 24152729 PMCID: PMC3861079 DOI: 10.1091/mbc.e13-02-0108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 09/18/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022] Open
Abstract
Depletion of stathmin, a microtubule (MT) destabilizer, delays mitotic entry by ∼4 h in HeLa cells. Stathmin depletion reduced the activity of CDC25 and its upstream activators, Aurora A and Plk1. Chemical inhibition of both Aurora A and Plk1 was sufficient to delay mitotic entry by 4 h, while inhibiting either kinase alone did not cause a delay. Aurora A and Plk1 are likely regulated downstream of stathmin, because the combination of stathmin knockdown and inhibition of Aurora A and Plk1 was not additive and again delayed mitotic entry by 4 h. Aurora A localization to the centrosome required MTs, while stathmin depletion spread its localization beyond that of γ-tubulin, indicating an MT-dependent regulation of Aurora A activation. Plk1 was inhibited by excess stathmin, detected in in vitro assays and cells overexpressing stathmin-cyan fluorescent protein. Recruitment of Plk1 to the centrosome was delayed in stathmin-depleted cells, independent of MTs. It has been shown that depolymerizing MTs with nocodazole abrogates the stathmin-depletion induced cell cycle delay; in this study, depolymerization with nocodazole restored Plk1 activity to near normal levels, demonstrating that MTs also contribute to Plk1 activation. These data demonstrate that stathmin regulates mitotic entry, partially via MTs, to control localization and activation of both Aurora A and Plk1.
Collapse
Affiliation(s)
- Victoria C. Silva
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| | - Lynne Cassimeris
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| |
Collapse
|
37
|
Chen TC, Liu YW, Huang YH, Yeh YC, Chou TY, Wu YC, Wu CC, Chen YR, Cheng HC, Lu PJ, Lai JM, Huang CYF. Protein phosphorylation profiling using an in situ proximity ligation assay: phosphorylation of AURKA-elicited EGFR-Thr654 and EGFR-Ser1046 in lung cancer cells. PLoS One 2013; 8:e55657. [PMID: 23520446 PMCID: PMC3592865 DOI: 10.1371/journal.pone.0055657] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 01/03/2013] [Indexed: 01/01/2023] Open
Abstract
The epidermal growth factor receptor (EGFR), which is up-regulated in lung cancer, involves the activation of mitogenic signals and triggers multiple signaling cascades. To dissect these EGFR cascades, we used 14 different phospho-EGFR antibodies to quantify protein phosphorylation using an in situ proximity ligation assay (in situ PLA). Phosphorylation at EGFR-Thr654 and -Ser1046 was EGF-dependent in the wild-type (WT) receptor but EGF-independent in a cell line carrying the EGFR-L858R mutation. Using a ProtoAarray™ containing ∼5000 recombinant proteins on the protein chip, we found that AURKA interacted with the EGFR-L861Q mutant. Moreover, overexpression of EGFR could form a complex with AURKA, and the inhibitors of AURKA and EGFR decreased EGFR-Thr654 and -Ser1046 phosphorylation. Immunohistochemical staining of stage I lung adenocarcinoma tissues demonstrated a positive correlation between AURKA expression and phosphorylation of EGFR at Thr654 and Ser1046 in EGFR-mutant specimens, but not in EGFR-WT specimens. The interplay between EGFR and AURKA provides an explanation for the difference in EGF dependency between EGFR-WT and EGFR-mutant cells and may provide a new therapeutic strategy for lung cancer patients carrying EGFR mutations.
Collapse
Affiliation(s)
- Tzu-Chi Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Wen Liu
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yei-Hsuan Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chen Yeh
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Teh-Ying Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Chung Wu
- Division of Thoracic Surgery, Department of Surgery, Veterans General Hospital, Taipei, Taiwan
| | - Chun-Chi Wu
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Yi-Rong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Hui-Chuan Cheng
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Jin-Mei Lai
- Department of Life Science, Fu-Jen Catholic University, Taipei, Taiwan
| | - Chi-Ying F. Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
38
|
Carvalho TG, Doerig C, Reininger L. Nima- and Aurora-related kinases of malaria parasites. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:1336-45. [PMID: 23462523 DOI: 10.1016/j.bbapap.2013.02.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 02/14/2013] [Indexed: 10/27/2022]
Abstract
Completion of the life cycle of malaria parasite requires a succession of developmental stages which vary greatly with respect to proliferation status, implying a tightly regulated control of the parasite's cell cycle, which remains to be understood at the molecular level. Progression of the eukaryotic cell cycle is controlled by members of mitotic kinase of the families CDK (cyclin-dependent kinases), Aurora, Polo and NIMA. Plasmodium parasites possess cyclin-dependent protein kinases and cyclins, which strongly suggests that some of the principles underlying cell cycle control in higher eukaryotes also operate in this organism. However, atypical features of Plasmodium cell cycle organization and important divergences in the composition of the cell cycle machinery suggest the existence of regulatory mechanisms that are at variance with those of higher eukaryotes. This review focuses on several recently described Plasmodium protein kinases related to the NIMA and Aurora kinase families and discusses their functional involvement in parasite's biology. Given their demonstrated essential roles in the erythrocytic asexual cycle and/or sexual stages, these enzymes represent novel potential drug targets for antimalarial intervention aiming at inhibiting parasite replication and/or blocking transmission of the disease. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases (2012).
Collapse
Affiliation(s)
- Teresa Gil Carvalho
- Department of Microbiology, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | | | | |
Collapse
|
39
|
The role of NEDD1 phosphorylation by Aurora A in chromosomal microtubule nucleation and spindle function. Curr Biol 2012; 23:143-9. [PMID: 23273898 DOI: 10.1016/j.cub.2012.11.046] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 09/28/2012] [Accepted: 11/21/2012] [Indexed: 01/05/2023]
Abstract
Chromatin directs de novo microtubule (MT) nucleation in dividing cells by generating a gradient of GTP-bound Ran protein (RanGTP) that controls the activity of a number of spindle assembly factors (SAFs). It is now well established that these MTs are essential for the assembly of a functional bipolar spindle. Although it has been shown that RanGTP-dependent MT nucleation requires γ-tubulin and a number of RanGTP-regulated proteins, the mechanism involved is still poorly understood. We previously showed that the mitotic kinase Aurora A, which is activated in a RanGTP-dependent manner in mitotic cells, has a role in this pathway. Here we show that Aurora A interacts with and phosphorylates the γTURC adaptor protein NEDD1 at a single residue, Ser405. Ser405 phosphorylation is not required for centrosomal MT nucleation but is critical for MT nucleation in the vicinity of the chromosomes in mitotic cells. Moreover, it is essential for RanGTP aster formation and chromatin-driven MT assembly in Xenopus egg extracts. Our data suggest that one important function of Aurora A in mitotic cells is to promote MT nucleation around the chromatin by phosphorylating NEDD1, and thereby to promote functional spindle assembly.
Collapse
|
40
|
Hsueh KW, Fu SL, Chang CB, Chang YL, Lin CH. A novel Aurora-A-mediated phosphorylation of p53 inhibits its interaction with MDM2. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012. [PMID: 23201157 DOI: 10.1016/j.bbapap.2012.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE Crosstalk between Aurora-A kinase and p53 has been proposed. While the genetic amplification of Aurora-A has been observed in many human cancers, how p53 is regulated by Aurora-A remains ambiguous. In this study, Aurora-A-mediated phosphorylation of p53 was analyzed by mass spectrometry in order to identify a new phosphorylation site. Subsequently, the functional consequences of such phosphorylation were examined. EXPERIMENTAL DESIGN In vitro phosphorylation of p53 by Aurora-A was performed and the phosphorylated protein was then digested with trypsin and enriched for phosphopeptides by immobilized metal affinity chromatography. Subsequently, a combination of β-elimination and Michael addition was applied to the phosphopeptides in order to facilitate the identification of phosphorylation sites by MS. The functional consequences of the novel phosphorylation of p53 on the protein-protein interactions, protein stability and transactivation activity were then examined using co-immunoprecipitation, Western blotting and reporter assays. RESULTS Ser-106 of p53 was identified as a novel site phosphorylated by Aurora-A. A serine-to-alanine mutation at this site was found to attenuate Aurora-A-mediated phosphorylation in vitro. In addition, phosphate-sensitive Phos-tag SDS-PAGE was used to confirm that the Ser-106 of p53 is in vivo phosphorylated by Aurora-A. Finally, co-immunoprecipitation studies suggested that Ser-106 phosphorylation of p53 decreases its interaction with MDM2 and prolongs the half-life of p53. CONCLUSIONS The inhibition of the interaction between p53 and MDM2 by a novel Aurora-A-mediated p53 phosphorylation was identified in this study and this provides important information for further investigations into the interaction between p53 and Aurora-A in terms of cancer biology.
Collapse
Affiliation(s)
- Kai-Wei Hsueh
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
41
|
Alghamdi WM, Gaskell SJ, Barber J. Detection of low-abundance protein phosphorylation by selective 18O labeling and mass spectrometry. Anal Chem 2012; 84:7384-92. [PMID: 22876816 DOI: 10.1021/ac301038u] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Reversible phosphorylation regulates the majority of intracellular networking and pathways. The study of this widely explored post-translational modification is usually challenged by low stoichiometric levels of modification. Many approaches have been developed to overcome this problem and to achieve rigorous characterization of protein phosphorylation. We describe a method for enhanced detection of low-abundance protein phosphorylation that uses selective introduction of (18)O label into phosphorylation sites with H(2)(18)O and mass spectrometric detection. The method was applied to introduce (18)O label into bacterially expressed Aurora A kinase phosphorylation sites and resulted in the representation of phosphorylated peptides as doublets or triplets according to the number of phosphate groups. A total of 28 phosphopeptides were observed by this method.
Collapse
Affiliation(s)
- Waleed M Alghamdi
- The Michael Barber Centre for Mass Spectrometry, Manchester Interdisciplinary Biocentre (MIB), Manchester, United Kingdom
| | | | | |
Collapse
|
42
|
Okazaki K, Nakayama N, Nariai Y, Nakayama K, Miyazaki K, Maruyama R, Kato H, Kosugi S, Urano T, Sakashita G. Nuclear localization signal in a cancer-related transcriptional regulator protein NAC1. Carcinogenesis 2012; 33:1854-62. [PMID: 22665369 DOI: 10.1093/carcin/bgs193] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nucleus accumbens-associated protein 1 (NAC1) might have potential oncogenic properties and participate in regulatory networks for pluripotency. Although NAC1 is described as a transcriptional regulator, the nuclear import machinery of NAC1 remains unclear. We found, using a point mutant, that dimer formation was not committed to the nuclear localization of NAC1 and, using deletion mutants, that the amino-terminal half of NAC1 harbored a potential nuclear localization signal (NLS). Wild type, but not mutants of this region, alone was sufficient to drive the importation of green fluorescent protein (GFP) into the nucleus. Bimax1, a synthetic peptide that blocks the importin α/β pathway, impaired nuclear localization of NAC1 in cells. We also used the binding properties of importin to demonstrate that this region is an NLS. Furthermore, the transcriptional regulator function of NAC1 was dependent on its nuclear localization activity in cells. Taken together, these results show that the region with a bipartite motif constitutes a functional nuclear import sequence in NAC1 that is independent of NAC1 dimer formation. The identification of an NAC1 NLS thus clarifies the mechanism through which NAC1 translocates to the nucleus to regulate the transcription of genes involved in oncogenicity and pluripotency.
Collapse
Affiliation(s)
- Kosuke Okazaki
- Department of Biochemistry, Shimane University School of Medicine, Izumo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Inoko A, Matsuyama M, Goto H, Ohmuro-Matsuyama Y, Hayashi Y, Enomoto M, Ibi M, Urano T, Yonemura S, Kiyono T, Izawa I, Inagaki M. Trichoplein and Aurora A block aberrant primary cilia assembly in proliferating cells. ACTA ACUST UNITED AC 2012; 197:391-405. [PMID: 22529102 PMCID: PMC3341160 DOI: 10.1083/jcb.201106101] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The trichoplein–AurA pathway must suppress primary cilia assembly in order for cells to exit G1. The primary cilium is an antenna-like organelle that modulates differentiation, sensory functions, and signal transduction. After cilia are disassembled at the G0/G1 transition, formation of cilia is strictly inhibited in proliferating cells. However, the mechanisms of this inhibition are unknown. In this paper, we show that trichoplein disappeared from the basal body in quiescent cells, whereas it localized to mother and daughter centrioles in proliferating cells. Exogenous expression of trichoplein inhibited primary cilia assembly in serum-starved cells, whereas ribonucleic acid interference–mediated depletion induced primary cilia assembly upon cultivation with serum. Trichoplein controlled Aurora A (AurA) activation at the centrioles predominantly in G1 phase. In vitro analyses confirmed that trichoplein bound and activated AurA directly. Using trichoplein mutants, we demonstrate that the suppression of primary cilia assembly by trichoplein required its ability not only to localize to centrioles but also to bind and activate AurA. Trichoplein or AurA knockdown also induced G0/G1 arrest, but this phenotype was reversed when cilia formation was prevented by simultaneous knockdown of IFT-20. These data suggest that the trichoplein–AurA pathway is required for G1 progression through a key role in the continuous suppression of primary cilia assembly.
Collapse
Affiliation(s)
- Akihito Inoko
- Division of Biochemistry, Aichi Cancer Center Research Institute, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Meehan M, Parthasarathi L, Moran N, Jefferies CA, Foley N, Lazzari E, Murphy D, Ryan J, Ortiz B, Fabius AWM, Chan TA, Stallings RL. Protein tyrosine phosphatase receptor delta acts as a neuroblastoma tumor suppressor by destabilizing the aurora kinase A oncogene. Mol Cancer 2012; 11:6. [PMID: 22305495 PMCID: PMC3395855 DOI: 10.1186/1476-4598-11-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 02/05/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Protein tyrosine phosphatase receptor delta (PTPRD) is a member of a large family of protein tyrosine phosphatases which negatively regulate tyrosine phosphorylation. Neuroblastoma is a major childhood cancer arising from precursor cells of the sympathetic nervous system which is known to acquire deletions and alterations in the expression patterns of PTPRD, indicating a potential tumor suppressor function for this gene. The molecular mechanism, however, by which PTPRD renders a tumor suppressor effect in neuroblastoma is unknown. RESULTS As a molecular mechanism, we demonstrate that PTPRD interacts with aurora kinase A (AURKA), an oncogenic protein that is over-expressed in multiple forms of cancer, including neuroblastoma. Ectopic up-regulation of PTPRD in neuroblastoma dephosphorylates tyrosine residues in AURKA resulting in a destabilization of this protein culminating in interfering with one of AURKA's primary functions in neuroblastoma, the stabilization of MYCN protein, the gene of which is amplified in approximately 25 to 30% of high risk neuroblastoma. CONCLUSIONS PTPRD has a tumor suppressor function in neuroblastoma through AURKA dephosphorylation and destabilization and a downstream destabilization of MYCN protein, representing a novel mechanism for the function of PTPRD in neuroblastoma.
Collapse
Affiliation(s)
- Maria Meehan
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ritchey L, Ottman R, Roumanos M, Chakrabarti R. A functional cooperativity between Aurora A kinase and LIM kinase1: implication in the mitotic process. Cell Cycle 2012; 11:296-309. [PMID: 22214762 DOI: 10.4161/cc.11.2.18734] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aurora kinase A (Aur-A), a mitotic kinase, regulates initiation of mitosis through centrosome separation and proper assembly of bipolar spindles. LIM kinase 1 (LIMK1), a modulator of actin and microtubule dynamics, is involved in the mitotic process through inactivating phosphorylation of cofilin. Phosphorylated LIMK1 is recruited to the centrosomes during early prophase, where it colocalizes with γ-tubulin. Here, we report a novel functional cooperativity between Aur-A and LIMK1 through mutual phosphorylation. LIMK1 is recruited to the centrosomes during early prophase and then to the spindle poles, where it colocalizes with Aur-A. Aur-A physically associates with LIMK1 and activates it through phosphorylation, which is important for its centrosomal and spindle pole localization. Aur-A also acts as a substrate of LIMK1, and the function of LIMK1 is important for its specific localization and regulation of spindle morphology. Taken together, the novel molecular interaction between these two kinases and their regulatory roles on one another's function may provide new insight on the role of Aur-A in manipulation of actin and microtubular structures during spindle formation.
Collapse
Affiliation(s)
- Lisa Ritchey
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | | | | | | |
Collapse
|
46
|
Liu HS, Ke CS, Cheng HC, Huang CYF, Su CL. Curcumin-induced mitotic spindle defect and cell cycle arrest in human bladder cancer cells occurs partly through inhibition of aurora A. Mol Pharmacol 2011; 80:638-46. [PMID: 21757545 DOI: 10.1124/mol.111.072512] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Curcumin, an active compound in turmeric and curry, has been proven to induce tumor apoptosis and inhibit tumor proliferation, invasion, angiogenesis, and metastasis via modulating numerous targets in various types of cancer cells. Aurora A is a mitosis-related serine-threonine kinase and plays important roles in diverse human cancers. However, the effect of curcumin on Aurora A has not been reported. In this study, Aurora A promoter activity and mRNA expression were inhibited in curcumin-treated human bladder cancer T24 cells, suggesting that Aurora A is regulated at the transcription level. We also found that curcumin preferentially inhibited the growth of T24 cells, which show a higher proliferation rate, invasion activity, and expression level of Aurora A compared with that of human immortalized uroepithelial E7cells. Furthermore, inhibition of phosphorylation of Aurora A and its downstream target histone H3 accompanied by the formation of monopolar spindle, induction of G(2)/M phase arrest, and reduction in cell division in response to curcumin were detected in T24 cells. These curcumin-induced phenomena were similar to those using Aurora A small interfering RNA and were attenuated by ectopic expression of Aurora A. Therefore, the antitumor mechanism of curcumin is Aurora A-related, which further supports the application of curcumin in treatments of human cancers.
Collapse
Affiliation(s)
- Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
47
|
Xiao L, Chen Y, Ji M, Volle DJ, Lewis RE, Tsai MY, Dong J. KIBRA protein phosphorylation is regulated by mitotic kinase aurora and protein phosphatase 1. J Biol Chem 2011; 286:36304-15. [PMID: 21878642 DOI: 10.1074/jbc.m111.246850] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent genetic studies in Drosophila identified Kibra as a novel regulator of the Hippo pathway, which controls tissue growth and tumorigenesis by inhibiting cell proliferation and promoting apoptosis. The cellular function and regulation of human KIBRA remain largely unclear. Here, we show that KIBRA is a phosphoprotein and that phosphorylation of KIBRA is regulated in a cell cycle-dependent manner with the highest level of phosphorylated KIBRA detected in mitosis. We further demonstrate that the mitotic kinases Aurora-A and -B phosphorylate KIBRA both in vitro and in vivo. We identified the highly conserved Ser(539) as the primary phosphorylation site for Aurora kinases. Moreover, we found that wild-type, but not catalytically inactive, protein phosphatase 1 (PP1) associates with KIBRA. PP1 dephosphorylated Aurora-phosphorylated KIBRA. KIBRA depletion impaired the interaction between Aurora-A and PP1. We also show that KIBRA associates with neurofibromatosis type 2/Merlin in a Ser(539) phosphorylation-dependent manner. Phosphorylation of KIBRA on Ser(539) plays a role in mitotic progression. Our results suggest that KIBRA is a physiological substrate of Aurora kinases and reveal a new avenue between KIBRA/Hippo signaling and the mitotic machinery.
Collapse
Affiliation(s)
- Ling Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Cheung CHA, Lin WH, Hsu JTA, Hour TC, Yeh TK, Ko S, Lien TW, Coumar MS, Liu JF, Lai WY, Shiao HY, Lee TR, Hsieh HP, Chang JY. BPR1K653, a novel Aurora kinase inhibitor, exhibits potent anti-proliferative activity in MDR1 (P-gp170)-mediated multidrug-resistant cancer cells. PLoS One 2011; 6:e23485. [PMID: 21887256 PMCID: PMC3160846 DOI: 10.1371/journal.pone.0023485] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 07/18/2011] [Indexed: 12/30/2022] Open
Abstract
Background Over-expression of Aurora kinases promotes the tumorigenesis of cells. The aim of this study was to determine the preclinical profile of a novel pan-Aurora kinase inhibitor, BPR1K653, as a candidate for anti-cancer therapy. Since expression of the drug efflux pump, MDR1, reduces the effectiveness of various chemotherapeutic compounds in human cancers, this study also aimed to determine whether the potency of BPR1K653 could be affected by the expression of MDR1 in cancer cells. Principal Findings BPR1K653 specifically inhibited the activity of Aurora-A and Aurora-B kinase at low nano-molar concentrations in vitro. Anti-proliferative activity of BPR1K653 was evaluated in various human cancer cell lines. Results of the clonogenic assay showed that BPR1K653 was potent in targeting a variety of cancer cell lines regardless of the tissue origin, p53 status, or expression of MDR1. At the cellular level, BPR1K653 induced endo-replication and subsequent apoptosis in both MDR1-negative and MDR1-positive cancer cells. Importantly, it showed potent activity against the growth of xenograft tumors of the human cervical carcinoma KB and KB-derived MDR1-positive KB-VIN10 cells in nude mice. Finally, BPR1K653 also exhibited favorable pharmacokinetic properties in rats. Conclusions and Significance BPR1K653 is a novel potent anti-cancer compound, and its potency is not affected by the expression of the multiple drug resistant protein, MDR1, in cancer cells. Therefore, BPR1K653 is a promising anti-cancer compound that has potential for the management of various malignancies, particularly for patients with MDR1-related drug resistance after prolonged chemotherapeutic treatments.
Collapse
Affiliation(s)
- Chun Hei Antonio Cheung
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan R.O.C.
| | - Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
| | - John Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
| | - Tzyh-Chyuan Hour
- Institute of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan R.O.C.
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
| | - Shengkai Ko
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
| | - Tzu-Wen Lien
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, India
| | - Jin-Fen Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan R.O.C.
| | - Wen-Yang Lai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan R.O.C.
| | - Hui-Yi Shiao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
| | - Tian-Ren Lee
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan R.O.C.
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan R.O.C.
- * E-mail: (JYC); (HPH)
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan R.O.C.
- Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan R.O.C.
- * E-mail: (JYC); (HPH)
| |
Collapse
|
49
|
Aurora-A phosphorylates hnRNPK and disrupts its interaction with p53. FEBS Lett 2011; 585:2671-5. [PMID: 21821029 DOI: 10.1016/j.febslet.2011.07.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 07/07/2011] [Accepted: 07/21/2011] [Indexed: 01/18/2023]
Abstract
Amplification of Aurora-A, encoding a cell cycle-regulating kinase, has been reported in human cancers. Although Aurora-A is known to directly phosphorylate and down-regulate p53, the detailed mechanism remains unclear. Here we show that Aurora-A phosphorylates hnRNPK, a transcriptional coactivator of p53, on serine 379. This phosphorylation does not affect the post-transcriptional activity or cellular localization of hnRNPK, but disrupts its interaction with p53. Inverse correlation between Aurora-A activity and hnRNPK-p53 interaction was further demonstrated in DNA-damaged cells. Our results provide an alternative mechanism, whereby via phosphorylating hnRNPK Aurora-A participates in regulating p53 activity during DNA damage.
Collapse
|
50
|
Kettenbach AN, Schweppe DK, Faherty BK, Pechenick D, Pletnev AA, Gerber SA. Quantitative phosphoproteomics identifies substrates and functional modules of Aurora and Polo-like kinase activities in mitotic cells. Sci Signal 2011; 4:rs5. [PMID: 21712546 PMCID: PMC3808085 DOI: 10.1126/scisignal.2001497] [Citation(s) in RCA: 421] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mitosis is a process involving a complex series of events that require careful coordination. Protein phosphorylation by a small number of kinases, in particular Aurora A, Aurora B, the cyclin-dependent kinase-cyclin complex Cdk1/cyclinB, and Polo-like kinase 1 (Plk1), orchestrates almost every step of cell division, from entry into mitosis to cytokinesis. To discover more about the functions of Aurora A, Aurora B, and kinases of the Plk family, we mapped mitotic phosphorylation sites to these kinases through the combined use of quantitative phosphoproteomics and selective targeting of kinase activities by small-molecule inhibitors. Using this integrated approach, we connected 778 phosphorylation sites on 562 proteins with these enzymes in cells arrested in mitosis. By connecting the kinases to protein complexes, we associated these kinases with functional modules. In addition to predicting previously unknown functions, this work establishes additional substrate-recognition motifs for these kinases and provides an analytical template for further use in dissecting kinase signaling events in other areas of cellular signaling and systems biology.
Collapse
Affiliation(s)
- Arminja N. Kettenbach
- Department of Genetics, Dartmouth Medical School, Lebanon, NH 03756, USA
- Norris Cotton Cancer Center, Lebanon, NH 03756, USA
| | - Devin K. Schweppe
- Department of Genetics, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Brendan K. Faherty
- Department of Genetics, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Dov Pechenick
- Department of Genetics, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Alexandre A. Pletnev
- Norris Cotton Cancer Center, Lebanon, NH 03756, USA
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Scott A. Gerber
- Department of Genetics, Dartmouth Medical School, Lebanon, NH 03756, USA
- Norris Cotton Cancer Center, Lebanon, NH 03756, USA
| |
Collapse
|