1
|
Doobin DJ, Helmer P, Carabalona A, Bertipaglia C, Vallee RB. The Role of Nde1 phosphorylation in interkinetic nuclear migration and neural migration during cortical development. Mol Biol Cell 2024; 35:ar129. [PMID: 39167527 PMCID: PMC11481692 DOI: 10.1091/mbc.e24-05-0217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/18/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
Nde1 is a cytoplasmic dynein regulatory protein with important roles in vertebrate brain development. One noteworthy function is in the nuclear oscillatory behavior in neural progenitor cells, the control and mechanism of which remain poorly understood. Nde1 contains multiple phosphorylation sites for the cell cycle-dependent protein kinase CDK1, though the function of these sites is not well understood. To test their role in brain development, we expressed phosphorylation-state mutant forms of Nde1 in embryonic rat brains using in utero electroporation. We find that Nde1 T215 and T243 phosphomutants block apical interkinetic nuclear migration (INM) and, consequently, mitosis in radial glial progenitor cells. Another Nde1 phosphomutant at T246 also interfered with mitotic entry without affecting INM, suggesting a more direct role for Nde1 T246 in mitotic regulation. We also found that the Nde1 S214F mutation, which is associated with schizophrenia, inhibits Cdk5 phosphorylation at an adjacent residue which causes alterations in neuronal lamination. These results together identify important new roles for Nde1 phosphorylation in neocortical development and disease, and represent the first evidence for Nde1 phosphorylation roles in INM and neuronal lamination.
Collapse
Affiliation(s)
| | - Paige Helmer
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY
| | - Aurelie Carabalona
- Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | | | - Richard B. Vallee
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
2
|
Courchesne E, Taluja V, Nazari S, Aamodt CM, Pierce K, Duan K, Stophaeros S, Lopez L, Barnes CC, Troxel J, Campbell K, Wang T, Hoekzema K, Eichler EE, Nani JV, Pontes W, Sanchez SS, Lombardo MV, de Souza JS, Hayashi MAF, Muotri AR. Embryonic origin of two ASD subtypes of social symptom severity: the larger the brain cortical organoid size, the more severe the social symptoms. Mol Autism 2024; 15:22. [PMID: 38790065 PMCID: PMC11127428 DOI: 10.1186/s13229-024-00602-8] [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: 12/05/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Social affective and communication symptoms are central to autism spectrum disorder (ASD), yet their severity differs across toddlers: Some toddlers with ASD display improving abilities across early ages and develop good social and language skills, while others with "profound" autism have persistently low social, language and cognitive skills and require lifelong care. The biological origins of these opposite ASD social severity subtypes and developmental trajectories are not known. METHODS Because ASD involves early brain overgrowth and excess neurons, we measured size and growth in 4910 embryonic-stage brain cortical organoids (BCOs) from a total of 10 toddlers with ASD and 6 controls (averaging 196 individual BCOs measured/subject). In a 2021 batch, we measured BCOs from 10 ASD and 5 controls. In a 2022 batch, we tested replicability of BCO size and growth effects by generating and measuring an independent batch of BCOs from 6 ASD and 4 control subjects. BCO size was analyzed within the context of our large, one-of-a-kind social symptom, social attention, social brain and social and language psychometric normative datasets ranging from N = 266 to N = 1902 toddlers. BCO growth rates were examined by measuring size changes between 1- and 2-months of organoid development. Neurogenesis markers at 2-months were examined at the cellular level. At the molecular level, we measured activity and expression of Ndel1; Ndel1 is a prime target for cell cycle-activated kinases; known to regulate cell cycle, proliferation, neurogenesis, and growth; and known to be involved in neuropsychiatric conditions. RESULTS At the BCO level, analyses showed BCO size was significantly enlarged by 39% and 41% in ASD in the 2021 and 2022 batches. The larger the embryonic BCO size, the more severe the ASD social symptoms. Correlations between BCO size and social symptoms were r = 0.719 in the 2021 batch and r = 0. 873 in the replication 2022 batch. ASD BCOs grew at an accelerated rate nearly 3 times faster than controls. At the cell level, the two largest ASD BCOs had accelerated neurogenesis. At the molecular level, Ndel1 activity was highly correlated with the growth rate and size of BCOs. Two BCO subtypes were found in ASD toddlers: Those in one subtype had very enlarged BCO size with accelerated rate of growth and neurogenesis; a profound autism clinical phenotype displaying severe social symptoms, reduced social attention, reduced cognitive, very low language and social IQ; and substantially altered growth in specific cortical social, language and sensory regions. Those in a second subtype had milder BCO enlargement and milder social, attention, cognitive, language and cortical differences. LIMITATIONS Larger samples of ASD toddler-derived BCO and clinical phenotypes may reveal additional ASD embryonic subtypes. CONCLUSIONS By embryogenesis, the biological bases of two subtypes of ASD social and brain development-profound autism and mild autism-are already present and measurable and involve dysregulated cell proliferation and accelerated neurogenesis and growth. The larger the embryonic BCO size in ASD, the more severe the toddler's social symptoms and the more reduced the social attention, language ability, and IQ, and the more atypical the growth of social and language brain regions.
Collapse
Affiliation(s)
- Eric Courchesne
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA.
| | - Vani Taluja
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Sanaz Nazari
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Caitlin M Aamodt
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, Gilman Drive, La Jolla, CA, 92093, USA
| | - Karen Pierce
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Kuaikuai Duan
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Sunny Stophaeros
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Linda Lopez
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Cynthia Carter Barnes
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Jaden Troxel
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Kathleen Campbell
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, 8110 La Jolla Shores Dr., La Jolla, CA, 92037, USA
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing, 100191, China
- Neuroscience Research Institute, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing, 100191, China
| | - Kendra Hoekzema
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Joao V Nani
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, Gilman Drive, La Jolla, CA, 92093, USA
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Wirla Pontes
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, Gilman Drive, La Jolla, CA, 92093, USA
| | - Sandra Sanchez Sanchez
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, Gilman Drive, La Jolla, CA, 92093, USA
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Janaina S de Souza
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, Gilman Drive, La Jolla, CA, 92093, USA
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Alysson R Muotri
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, Gilman Drive, La Jolla, CA, 92093, USA.
- Rady Children's Hospital, Center for Academic Research and Training in Anthropogeny (CARTA), Archealization Center (ArchC), Kavli Institute for Brain and Mind, La Jolla, CA, USA.
| |
Collapse
|
3
|
Zheng H, Zhang Q, Liu X, Shi F, Yang F, Xiang S, Jiang H. Aurora-A condensation mediated by BuGZ aids its mitotic centrosome functions. iScience 2024; 27:109785. [PMID: 38746663 PMCID: PMC11090908 DOI: 10.1016/j.isci.2024.109785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/30/2023] [Accepted: 04/16/2024] [Indexed: 03/17/2025] Open
Abstract
Centrosomes composed of centrioles and the pericentriolar material (PCM), serve as the platform for microtubule polymerization during mitosis. Despite some centriole and PCM proteins have been reported to utilize liquid-liquid phase separation (LLPS) to perform their mitotic functions, whether and how centrosomal kinases exert the coacervation in mitosis is still unknown. Here we reveal that Aurora-A, one key centrosomal kinase in regulating centrosome formation and functions, undergoes phase separation in vitro or in centrosomes from prophase, mediated by the conserved positive-charged residues inside its intrinsic disordered region (IDR) and the intramolecular interaction between its N- and C-terminus. Aurora-A condensation affects centrosome maturation, separation, initial spindle formation from the spindle pole and its kinase activity. Moreover, BuGZ interacts with Aurora-A to enhance its LLPS and centrosome functions. Thus, we propose that Aurora-A collaborates with BuGZ to exhibit the property of LLPS in centrosomes to control its centrosome-dependent functions from prophase.
Collapse
Affiliation(s)
- Hui Zheng
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Qiaoqiao Zhang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Center for Cross-disciplinary Sciences, University of Science & Technology of China, School of Life Sciences, Hefei, China
| | - Fan Shi
- MOE Key Lab for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Center for Cross-disciplinary Sciences, University of Science & Technology of China, School of Life Sciences, Hefei, China
| | - Shengqi Xiang
- MOE Key Lab for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Hao Jiang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
4
|
Saatci O, Sahin O. TACC3: a multi-functional protein promoting cancer cell survival and aggressiveness. Cell Cycle 2023; 22:2637-2655. [PMID: 38197196 PMCID: PMC10936615 DOI: 10.1080/15384101.2024.2302243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
TACC3 is the most oncogenic member of the transforming acidic coiled-coil domain-containing protein (TACC) family. It is one of the major recruitment factors of distinct multi-protein complexes. TACC3 is localized to spindles, centrosomes, and nucleus, and regulates key oncogenic processes, including cell proliferation, migration, invasion, and stemness. Recently, TACC3 inhibition has been identified as a vulnerability in highly aggressive cancers, such as cancers with centrosome amplification (CA). TACC3 has spatiotemporal functions throughout the cell cycle; therefore, targeting TACC3 causes cell death in mitosis and interphase in cancer cells with CA. In the clinics, TACC3 is highly expressed and associated with worse survival in multiple cancers. Furthermore, TACC3 is a part of one of the most common fusions of FGFR, FGFR3-TACC3 and is important for the oncogenicity of the fusion. A detailed understanding of the regulation of TACC3 expression, its key partners, and molecular functions in cancer cells is vital for uncovering the most vulnerable tumors and maximizing the therapeutic potential of targeting this highly oncogenic protein. In this review, we summarize the established and emerging interactors and spatiotemporal functions of TACC3 in cancer cells, discuss the potential of TACC3 as a biomarker in cancer, and therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
5
|
Gcap14 is a microtubule plus-end-tracking protein coordinating microtubule-actin crosstalk during neurodevelopment. Proc Natl Acad Sci U S A 2023; 120:e2214507120. [PMID: 36795749 PMCID: PMC9974511 DOI: 10.1073/pnas.2214507120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Regulation of microtubule dynamics is required to properly control various steps of neurodevelopment. In this study, we identified granule cell antiserum-positive 14 (Gcap14) as a microtubule plus-end-tracking protein and as a regulator of microtubule dynamics during neurodevelopment. Gcap14 knockout mice exhibited impaired cortical lamination. Gcap14 deficiency resulted in defective neuronal migration. Moreover, nuclear distribution element nudE-like 1 (Ndel1), an interacting partner of Gcap14, effectively corrected the downregulation of microtubule dynamics and the defects in neuronal migration caused by Gcap14 deficiency. Finally, we found that the Gcap14-Ndel1 complex participates in the functional link between microtubule and actin filament, thereby regulating their crosstalks in the growth cones of cortical neurons. Taken together, we propose that the Gcap14-Ndel1 complex is fundamental for cytoskeletal remodeling during neurodevelopmental processes such as neuronal processes elongation and neuronal migration.
Collapse
|
6
|
Peng L, He Y, Wang W, Chu Y, Lin Q, Rui R, Li Q, Ju S. PAK1 Is Involved in the Spindle Assembly during the First Meiotic Division in Porcine Oocytes. Int J Mol Sci 2023; 24:ijms24021123. [PMID: 36674642 PMCID: PMC9866149 DOI: 10.3390/ijms24021123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
P21-activated kinase 1 (PAK1), as a member of the PAK family, has been implicated in various functions during somatic mitosis; however, less is known about its role during oocyte meiosis. Herein, we highlight the indispensable role of PAK1 in regulating spindle assembly and cell cycle progression during the first meiotic division of porcine oocytes. First, we found that the activated PAK1 expressed dynamically, and its subcellular localization was tightly associated with the spindle dynamics during meiosis in porcine oocytes. Specific inhibition of PAK1 activity by inhibitor targeting PAK1 activation-3 (IPA-3) led to impaired extrusion of the first polar body (PB1); with most of the IPA-3-treated oocytes arrested at germinal vesicle breakdown (GVBD) and subjected to failure of bipolar spindle formation. However, the adverse effects caused by IPA-3 on oocytes could be restored by reducing disulfide bonds between PAK1 and IPA-3 with dithiothreitol (DTT) treatment. Furthermore, the co-immunoprecipitation assay revealed that PAK1 interacted directly with Aurora A and transforming acidic coiled coil 3 (TACC3), providing an additional explanation for the similar localization of Aurora A and activated PAK1. Additionally, inhibiting the activity of PAK1 decreased the expression of p-Aurora A and p-TACC3; however, the reduced activity of Aurora A and TACC3 could be restored by DTT. In conclusion, PAK1 plays a crucial role in the proper assembly of the spindle during the first meiotic division of porcine oocytes, and the regulation of PAK1 is associated with its effects on p-Aurora A and p-TACC3 expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qiao Li
- Correspondence: (Q.L.); (S.J.)
| | | |
Collapse
|
7
|
Janczyk PŁ, Żyłkiewicz E, De Hoyos H, West T, Matson DR, Choi WC, Young HMR, Derewenda ZS, Stukenberg PT. Aurora A phosphorylates Ndel1 to reduce the levels of Mad1 and NuMA at spindle poles. Mol Biol Cell 2023; 34:br1. [PMID: 36350697 PMCID: PMC9816647 DOI: 10.1091/mbc.e21-09-0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
Dynein inactivates the spindle assembly checkpoint (SAC) by transporting checkpoint proteins away from kinetochores toward spindle poles in a process known as "stripping." We find that inhibition of Aurora A kinase, which is localized to spindle poles, enables the accumulation of the spindle checkpoint activator Mad1 at poles where it is normally absent. Aurora kinases phosphorylate the dynein activator NudE neurodevelopment protein 1 like 1 (Ndel1) on Ser285 and Mad1 accumulates at poles when Ndel1 is replaced by a nonphosphorylatable mutant in human cells. The pole focusing protein NuMA, transported to poles by dynein, also accumulates at poles in cells harboring a mutant Ndel1. Phosphorylation of Ndel1 on Ser285 is required for robust spindle checkpoint activity and regulates the poles of asters in Xenopus extracts. Our data suggest that dynein/SAC complexes that are generated at kinetochores and then transported directionally toward poles on microtubules are inhibited by Aurora A before they reach spindle poles. These data suggest that Aurora A generates a spatial signal at spindle poles that controls dynein transport and spindle function.
Collapse
Affiliation(s)
- Paweł Ł. Janczyk
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Eliza Żyłkiewicz
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Henry De Hoyos
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Thomas West
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Daniel R. Matson
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Won-Chan Choi
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Heather M. Raimer Young
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Zygmunt S. Derewenda
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - P. Todd Stukenberg
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903
| |
Collapse
|
8
|
Zhao Z, Wang H, Kang N, Wang Z, Hou X, Hu L, Qie S, Guo J, Wei S, Ruan X, Zheng X. Aurora kinase a promotes the progression of papillary thyroid carcinoma by activating the mTORC2-AKT signalling pathway. Cell Biosci 2022; 12:195. [PMID: 36471438 PMCID: PMC9721059 DOI: 10.1186/s13578-022-00934-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Treatment failure is the main cause of death from papillary thyroid carcinoma (PTC). It is urgent to look for new intervention targets and to develop new therapies for treating PTC. Aurora-A kinase (AURKA) functionally regulates cell mitosis and is closely related to the occurrence and development of a variety of tumours. However, the expression and potential functions of AURKA in PTC remain largely elusive. RESULTS Clinicopathologically, AURKA is highly expressed in PTC tissues compared to normal tissues and is correlated with lymph node metastasis, TNM stage and patient prognosis. Biologically, AURKA functions as an oncoprotein to promote the proliferation and migration of PTC cells. Mechanistically, AURKA directly binds to SIN1 and compromises CUL4B-based E3 ligase-mediated ubiquitination and subsequent degradation of SIN1, leading to hyperactivation of the mTORC2-AKT pathway in PTC cells. CONCLUSIONS We found that AURKA plays critical roles in regulating the progression of PTC by activating the mTORC2-AKT pathway, highlighting the potential of targeting AURKA to treat PTC.
Collapse
Affiliation(s)
- Zewei Zhao
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Huijuan Wang
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Ning Kang
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Zhongyu Wang
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Xiukun Hou
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Linfei Hu
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Shuo Qie
- grid.411918.40000 0004 1798 6427Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Jianping Guo
- grid.412615.50000 0004 1803 6239Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510275 Guangdong China
| | - Songfeng Wei
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Xianhui Ruan
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Xiangqian Zheng
- grid.411918.40000 0004 1798 6427Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| |
Collapse
|
9
|
Torisawa T, Kimura A. Sequential accumulation of dynein and its regulatory proteins at the spindle region in the Caenorhabditis elegans embryo. Sci Rep 2022; 12:11740. [PMID: 35817834 PMCID: PMC9273622 DOI: 10.1038/s41598-022-15042-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/16/2022] [Indexed: 11/09/2022] Open
Abstract
Cytoplasmic dynein is responsible for various cellular processes during the cell cycle. The mechanism by which its activity is regulated spatially and temporarily inside the cell remains elusive. There are various regulatory proteins of dynein, including dynactin, NDEL1/NUD-2, and LIS1. Characterizing the spatiotemporal localization of regulatory proteins in vivo will aid understanding of the cellular regulation of dynein. Here, we focused on spindle formation in the Caenorhabditis elegans early embryo, wherein dynein and its regulatory proteins translocated from the cytoplasm to the spindle region upon nuclear envelope breakdown (NEBD). We found that (i) a limited set of dynein regulatory proteins accumulated in the spindle region, (ii) the spatial localization patterns were distinct among the regulators, and (iii) the regulatory proteins did not accumulate in the spindle region simultaneously but sequentially. Furthermore, the accumulation of NUD-2 was unique among the regulators. NUD-2 started to accumulate before NEBD (pre-NEBD accumulation), and exhibited the highest enrichment compared to the cytoplasmic concentration. Using a protein injection approach, we revealed that the C-terminal helix of NUD-2 was responsible for pre-NEBD accumulation. These findings suggest a fine temporal control of the subcellular localization of regulatory proteins.
Collapse
Affiliation(s)
- Takayuki Torisawa
- Cell Architecture Laboratory, National Institute of Genetics, Mishima, Japan.,Department of Genetics, The Graduate University for Advanced Studies, Sokendai, Mishima, Japan
| | - Akatsuki Kimura
- Cell Architecture Laboratory, National Institute of Genetics, Mishima, Japan. .,Department of Genetics, The Graduate University for Advanced Studies, Sokendai, Mishima, Japan.
| |
Collapse
|
10
|
Centrosome Defects in Hematological Malignancies: Molecular Mechanisms and Therapeutic Insights. BLOOD SCIENCE 2022; 4:143-151. [DOI: 10.1097/bs9.0000000000000127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022] Open
|
11
|
Garrott SR, Gillies JP, DeSantis ME. Nde1 and Ndel1: Outstanding Mysteries in Dynein-Mediated Transport. Front Cell Dev Biol 2022; 10:871935. [PMID: 35493069 PMCID: PMC9041303 DOI: 10.3389/fcell.2022.871935] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Cytoplasmic dynein-1 (dynein) is the primary microtubule minus-end directed molecular motor in most eukaryotes. As such, dynein has a broad array of functions that range from driving retrograde-directed cargo trafficking to forming and focusing the mitotic spindle. Dynein does not function in isolation. Instead, a network of regulatory proteins mediate dynein’s interaction with cargo and modulate dynein’s ability to engage with and move on the microtubule track. A flurry of research over the past decade has revealed the function and mechanism of many of dynein’s regulators, including Lis1, dynactin, and a family of proteins called activating adaptors. However, the mechanistic details of two of dynein’s important binding partners, the paralogs Nde1 and Ndel1, have remained elusive. While genetic studies have firmly established Nde1/Ndel1 as players in the dynein transport pathway, the nature of how they regulate dynein activity is unknown. In this review, we will compare Ndel1 and Nde1 with a focus on discerning if the proteins are functionally redundant, outline the data that places Nde1/Ndel1 in the dynein transport pathway, and explore the literature supporting and opposing the predominant hypothesis about Nde1/Ndel1’s molecular effect on dynein activity.
Collapse
Affiliation(s)
- Sharon R. Garrott
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - John P. Gillies
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Morgan E. DeSantis
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Morgan E. DeSantis,
| |
Collapse
|
12
|
Yan Z, Shi Q, Liu X, Li J, Ahire V, Zhang S, Zhang J, Yang D, Allen TD. The phytochemical, corynoline, diminishes Aurora kinase B activity to induce mitotic defect and polyploidy. Pharmacotherapy 2022; 147:112645. [DOI: 10.1016/j.biopha.2022.112645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/01/2022] [Accepted: 01/12/2022] [Indexed: 01/21/2023]
|
13
|
Arslanhan MD, Rauniyar N, Yates JR, Firat-Karalar EN. Aurora Kinase A proximity map reveals centriolar satellites as regulators of its ciliary function. EMBO Rep 2021; 22:e51902. [PMID: 34169630 PMCID: PMC8339716 DOI: 10.15252/embr.202051902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Aurora kinase A (AURKA) is a conserved kinase that plays crucial roles in numerous cellular processes. Although AURKA overexpression is frequent in human cancers, its pleiotropic functions and multifaceted regulation present challenges in its therapeutic targeting. Key to overcoming these challenges is to identify and characterize the full range of AURKA interactors, which are often weak and transient. Previous proteomic studies were limited in monitoring dynamic and non-mitotic AURKA interactions. Here, we generate the proximity interactome of AURKA in asynchronous cells, which consists of 440 proteins involving multiple biological processes and cellular compartments. Importantly, AURKA has extensive proximate and physical interactions to centriolar satellites, key regulators of the primary cilium. Loss-of-function experiments identify satellites as negative regulators of AURKA activity, abundance, and localization in quiescent cells. Notably, loss of satellites activates AURKA at the basal body, decreases centrosomal IFT88 levels, and causes ciliogenesis defects. Collectively, our results provide a resource for dissecting spatiotemporal regulation of AURKA and uncover its proteostatic regulation by satellites as a new mechanism for its ciliary functions.
Collapse
Affiliation(s)
- Melis D Arslanhan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Navin Rauniyar
- Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
14
|
Mou PK, Yang EJ, Shi C, Ren G, Tao S, Shim JS. Aurora kinase A, a synthetic lethal target for precision cancer medicine. Exp Mol Med 2021; 53:835-847. [PMID: 34050264 PMCID: PMC8178373 DOI: 10.1038/s12276-021-00635-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/01/2023] Open
Abstract
Recent advances in high-throughput sequencing technologies and data science have facilitated the development of precision medicine to treat cancer patients. Synthetic lethality is one of the core methodologies employed in precision cancer medicine. Synthetic lethality describes the phenomenon of the interplay between two genes in which deficiency of a single gene does not abolish cell viability but combined deficiency of two genes leads to cell death. In cancer treatment, synthetic lethality is leveraged to exploit the dependency of cancer cells on a pathway that is essential for cell survival when a tumor suppressor is mutated. This approach enables pharmacological targeting of mutant tumor suppressors that are theoretically undruggable. Successful clinical introduction of BRCA-PARP synthetic lethality in cancer treatment led to additional discoveries of novel synthetic lethal partners of other tumor suppressors, including p53, PTEN, and RB1, using high-throughput screening. Recent work has highlighted aurora kinase A (AURKA) as a synthetic lethal partner of multiple tumor suppressors. AURKA is a serine/threonine kinase involved in a number of central biological processes, such as the G2/M transition, mitotic spindle assembly, and DNA replication. This review introduces synthetic lethal interactions between AURKA and its tumor suppressor partners and discusses the potential of AURKA inhibitors in precision cancer medicine.
Collapse
Affiliation(s)
- Pui Kei Mou
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Eun Ju Yang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Changxiang Shi
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guowen Ren
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Shishi Tao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Joong Sup Shim
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
15
|
Blengini CS, Ibrahimian P, Vaskovicova M, Drutovic D, Solc P, Schindler K. Aurora kinase A is essential for meiosis in mouse oocytes. PLoS Genet 2021; 17:e1009327. [PMID: 33901174 PMCID: PMC8102010 DOI: 10.1371/journal.pgen.1009327] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/06/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
The Aurora protein kinases are well-established regulators of spindle building and chromosome segregation in mitotic and meiotic cells. In mouse oocytes, there is significant Aurora kinase A (AURKA) compensatory abilities when the other Aurora kinase homologs are deleted. Whether the other homologs, AURKB or AURKC can compensate for loss of AURKA is not known. Using a conditional mouse oocyte knockout model, we demonstrate that this compensation is not reciprocal because female oocyte-specific knockout mice are sterile, and their oocytes fail to complete meiosis I. In determining AURKA-specific functions, we demonstrate that its first meiotic requirement is to activate Polo-like kinase 1 at acentriolar microtubule organizing centers (aMTOCs; meiotic spindle poles). This activation induces fragmentation of the aMTOCs, a step essential for building a bipolar spindle. We also show that AURKA is required for regulating localization of TACC3, another protein required for spindle building. We conclude that AURKA has multiple functions essential to completing MI that are distinct from AURKB and AURKC.
Collapse
Affiliation(s)
- Cecilia S. Blengini
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- Human Genetics Institute of New Jersey; Piscataway, New Jersey, United States of America
| | - Patricia Ibrahimian
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Michaela Vaskovicova
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - David Drutovic
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Petr Solc
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Karen Schindler
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- Human Genetics Institute of New Jersey; Piscataway, New Jersey, United States of America
| |
Collapse
|
16
|
Du R, Huang C, Liu K, Li X, Dong Z. Targeting AURKA in Cancer: molecular mechanisms and opportunities for Cancer therapy. Mol Cancer 2021; 20:15. [PMID: 33451333 PMCID: PMC7809767 DOI: 10.1186/s12943-020-01305-3] [Citation(s) in RCA: 295] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Aurora kinase A (AURKA) belongs to the family of serine/threonine kinases, whose activation is necessary for cell division processes via regulation of mitosis. AURKA shows significantly higher expression in cancer tissues than in normal control tissues for multiple tumor types according to the TCGA database. Activation of AURKA has been demonstrated to play an important role in a wide range of cancers, and numerous AURKA substrates have been identified. AURKA-mediated phosphorylation can regulate the functions of AURKA substrates, some of which are mitosis regulators, tumor suppressors or oncogenes. In addition, enrichment of AURKA-interacting proteins with KEGG pathway and GO analysis have demonstrated that these proteins are involved in classic oncogenic pathways. All of this evidence favors the idea of AURKA as a target for cancer therapy, and some small molecules targeting AURKA have been discovered. These AURKA inhibitors (AKIs) have been tested in preclinical studies, and some of them have been subjected to clinical trials as monotherapies or in combination with classic chemotherapy or other targeted therapies.
Collapse
Affiliation(s)
- Ruijuan Du
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China.
| | - Chuntian Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China.,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China. .,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China. .,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China. .,College of medicine, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
17
|
Wang Y, Yang Y, Gan Z, Zhao C, Lv C, Zhang Y, Zhao X. Role of AURKA in the hypothalamus-pituitary-testicular axis in Tibetan sheep from Tianzhu. Gen Comp Endocrinol 2021; 300:113617. [PMID: 32950578 DOI: 10.1016/j.ygcen.2020.113617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 11/20/2022]
Abstract
The mitosis-associated protein aurora kinase A (AURKA) regulates the maturation of germ cells. We have previously reported using transcriptome analysis that AURKA is expressed in yak testes. Although Tibetan sheep possess an immense economic value, their reproductive rate is low. Herein, the expression and functions of AURKA in the hypothalamus-pituitary-testicular (HPT) axis in Tibetan sheep from Tianzhu were investigated. The cDNA sequence of sheep AURKA was cloned and bioinformatics techniques were used to predict its structure. Tissue expression of AURKA was determined by qPCR, immunoblotting, immunostaining, and immunohistochemistry. The AURKA coding sequence was found to be 1218 bp in length, encoding a 405-amino acid polypeptide chain. Furthermore, the highest sequence similarity of AURKA with the corresponding sequence in other species was seen in goat and cattle; the least degree of similarity was seen in the domestic cat. In addition, AURKA expression was elevated in the testes compared to that in the hypothalamus and pituitary (p < 0.01). Moreover, AURKA was mainly localized in the hypothalamic paraventricular nucleus (magnocellular), chromophobe cells of the pituitary, and spermatogenic cells of the testis. These results indicated that AURKA might participate in sheep reproductive regulation, thus providing a reference for the study of AURKA function in the reproductive process of Tibetan sheep from Tianzhu.
Collapse
Affiliation(s)
- Yuanyuan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yang Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Ze Gan
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Caiying Zhao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Chen Lv
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China.
| |
Collapse
|
18
|
Ong JY, Bradley MC, Torres JZ. Phospho-regulation of mitotic spindle assembly. Cytoskeleton (Hoboken) 2020; 77:558-578. [PMID: 33280275 PMCID: PMC7898546 DOI: 10.1002/cm.21649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/08/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022]
Abstract
The assembly of the bipolar mitotic spindle requires the careful orchestration of a myriad of enzyme activities like protein posttranslational modifications. Among these, phosphorylation has arisen as the principle mode for spatially and temporally activating the proteins involved in early mitotic spindle assembly processes. Here, we review key kinases, phosphatases, and phosphorylation events that regulate critical aspects of these processes. We highlight key phosphorylation substrates that are important for ensuring the fidelity of centriole duplication, centrosome maturation, and the establishment of the bipolar spindle. We also highlight techniques used to understand kinase-substrate relationships and to study phosphorylation events. We conclude with perspectives on the field of posttranslational modifications in early mitotic spindle assembly.
Collapse
Affiliation(s)
- Joseph Y Ong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Michelle C Bradley
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
19
|
Kapoor S, Kotak S. Centrosome Aurora A gradient ensures single polarity axis in C. elegans embryos. Biochem Soc Trans 2020; 48:1243-1253. [PMID: 32597472 PMCID: PMC7616972 DOI: 10.1042/bst20200298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 01/31/2023]
Abstract
Cellular asymmetries are vital for generating cell fate diversity during development and in stem cells. In the newly fertilized Caenorhabditis elegans embryo, centrosomes are responsible for polarity establishment, i.e. anterior-posterior body axis formation. The signal for polarity originates from the centrosomes and is transmitted to the cell cortex, where it disassembles the actomyosin network. This event leads to symmetry breaking and the establishment of distinct domains of evolutionarily conserved PAR proteins. However, the identity of an essential component that localizes to the centrosomes and promotes symmetry breaking was unknown. Recent work has uncovered that the loss of Aurora A kinase (AIR-1 in C. elegans and hereafter referred to as Aurora A) in the one-cell embryo disrupts stereotypical actomyosin-based cortical flows that occur at the time of polarity establishment. This misregulation of actomyosin flow dynamics results in the occurrence of two polarity axes. Notably, the role of Aurora A in ensuring a single polarity axis is independent of its well-established function in centrosome maturation. The mechanism by which Aurora A directs symmetry breaking is likely through direct regulation of Rho-dependent contractility. In this mini-review, we will discuss the unconventional role of Aurora A kinase in polarity establishment in C. elegans embryos and propose a refined model of centrosome-dependent symmetry breaking.
Collapse
Affiliation(s)
- Sukriti Kapoor
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), 560012 Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), 560012 Bangalore, India
| |
Collapse
|
20
|
He Z, Mei L, Connell M, Maxwell CA. Hyaluronan Mediated Motility Receptor (HMMR) Encodes an Evolutionarily Conserved Homeostasis, Mitosis, and Meiosis Regulator Rather than a Hyaluronan Receptor. Cells 2020; 9:cells9040819. [PMID: 32231069 PMCID: PMC7226759 DOI: 10.3390/cells9040819] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
Hyaluronan is an extracellular matrix component that absorbs water in tissues and engages cell surface receptors, like Cluster of Differentiation 44 (CD44), to promote cellular growth and movement. Consequently, CD44 demarks stem cells in normal tissues and tumor-initiating cells isolated from neoplastic tissues. Hyaluronan mediated motility receptor (HMMR, also known as RHAMM) is another one of few defined hyaluronan receptors. HMMR is also associated with neoplastic processes and its role in cancer progression is often attributed to hyaluronan-mediated signaling. But, HMMR is an intracellular, microtubule-associated, spindle assembly factor that localizes protein complexes to augment the activities of mitotic kinases, like polo-like kinase 1 and Aurora kinase A, and control dynein and kinesin motor activities. Expression of HMMR is elevated in cells prior to and during mitosis and tissues with detectable HMMR expression tend to be highly proliferative, including neoplastic tissues. Moreover, HMMR is a breast cancer susceptibility gene product. Here, we briefly review the associations between HMMR and tumorigenesis as well as the structure and evolution of HMMR, which identifies Hmmr-like gene products in several insect species that do not produce hyaluronan. This review supports the designation of HMMR as a homeostasis, mitosis, and meiosis regulator, and clarifies how its dysfunction may promote the tumorigenic process and cancer progression.
Collapse
Affiliation(s)
- Zhengcheng He
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
| | - Lin Mei
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
| | - Christopher A. Maxwell
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital, Vancouver, BC V5Z 4H4, Canada
- Correspondence: ; Tel.: +1-6048752000 (ext. 4691)
| |
Collapse
|
21
|
Woo Y, Kim SJ, Suh BK, Kwak Y, Jung HJ, Nhung TTM, Mun DJ, Hong JH, Noh SJ, Kim S, Lee A, Baek ST, Nguyen MD, Choe Y, Park SK. Sequential phosphorylation of NDEL1 by the DYRK2-GSK3β complex is critical for neuronal morphogenesis. eLife 2019; 8:e50850. [PMID: 31815665 PMCID: PMC6927744 DOI: 10.7554/elife.50850] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/08/2019] [Indexed: 12/20/2022] Open
Abstract
Neuronal morphogenesis requires multiple regulatory pathways to appropriately determine axonal and dendritic structures, thereby to enable the functional neural connectivity. Yet, however, the precise mechanisms and components that regulate neuronal morphogenesis are still largely unknown. Here, we newly identified the sequential phosphorylation of NDEL1 critical for neuronal morphogenesis through the human kinome screening and phospho-proteomics analysis of NDEL1 from mouse brain lysate. DYRK2 phosphorylates NDEL1 S336 to prime the phosphorylation of NDEL1 S332 by GSK3β. TARA, an interaction partner of NDEL1, scaffolds DYRK2 and GSK3β to form a tripartite complex and enhances NDEL1 S336/S332 phosphorylation. This dual phosphorylation increases the filamentous actin dynamics. Ultimately, the phosphorylation enhances both axonal and dendritic outgrowth and promotes their arborization. Together, our findings suggest the NDEL1 phosphorylation at S336/S332 by the TARA-DYRK2-GSK3β complex as a novel regulatory mechanism underlying neuronal morphogenesis.
Collapse
Affiliation(s)
- Youngsik Woo
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Soo Jeong Kim
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Bo Kyoung Suh
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Yongdo Kwak
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Hyun-Jin Jung
- Korea Brain Research InstituteDaeguRepublic of Korea
| | - Truong Thi My Nhung
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Dong Jin Mun
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Ji-Ho Hong
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Su-Jin Noh
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Seunghyun Kim
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Ahryoung Lee
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Seung Tae Baek
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryCanada
- Department of Clinical Neurosciences, Cumming School of MedicineUniversity of CalgaryCalgaryCanada
- Department of Cell Biology and Anatomy, Cumming School of MedicineUniversity of CalgaryCalgaryCanada
- Department of Biochemistry and Molecular Biology, Cumming School of MedicineUniversity of CalgaryCalgaryCanada
| | | | - Sang Ki Park
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| |
Collapse
|
22
|
Wynne CL, Vallee RB. Cdk1 phosphorylation of the dynein adapter Nde1 controls cargo binding from G2 to anaphase. J Cell Biol 2018; 217:3019-3029. [PMID: 29930206 PMCID: PMC6122996 DOI: 10.1083/jcb.201707081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 04/06/2018] [Accepted: 05/23/2018] [Indexed: 12/28/2022] Open
Abstract
Cytoplasmic dynein is involved in diverse cell cycle-dependent functions regulated by several accessory factors, including Nde1 and Ndel1. Little is known about the role of these proteins in dynein cargo binding, and less is known about their cell cycle--dependent dynein regulation. Using Nde1 RNAi, mutant cDNAs, and a phosphorylation site-specific antibody, we found a specific association of phospho-Nde1 with the late G2-M nuclear envelope and prophase to anaphase kinetochores, comparable to the pattern for the Nde1 interactor CENP-F. Phosphomutant-Nde1 associated only with prometaphase kinetochores and showed weaker CENP-F binding in in vitro assays. Nde1 RNAi caused severe delays in mitotic progression, which were substantially rescued by both phosphomimetic and phosphomutant Nde1. Expression of a dynein-binding-deficient Nde1 mutant reduced kinetochore dynein by half, indicating a major role for Nde1 in kinetochore dynein recruitment. These results establish CENP-F as the first well-characterized Nde1 cargo protein, and reveal phosphorylation control of Nde1 cargo binding throughout a substantial fraction of the cell cycle.
Collapse
Affiliation(s)
- Caitlin L Wynne
- Pathology and Cell Biology, Columbia University, New York, NY
| | | |
Collapse
|
23
|
Ding ZM, Huang CJ, Jiao XF, Wu D, Huo LJ. The role of TACC3 in mitotic spindle organization. Cytoskeleton (Hoboken) 2017; 74:369-378. [PMID: 28745816 DOI: 10.1002/cm.21388] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 07/04/2017] [Accepted: 07/21/2017] [Indexed: 12/31/2022]
Abstract
TACC3 regulates spindle organization during mitosis and also regulates centrosome-mediated microtubule nucleation by affecting γ-Tubulin ring complexes. In addition, it interacts with different proteins (such as ch-TOG, clathrin and Aurora-A) to function in mitotic spindle assembly and stability. By forming the TACC3/ch-TOG complex, TACC3 acts as a plus end-tracking protein to promote microtubule elongation. The TACC3/ch-TOG/clathrin complex is formed to stabilize kinetochore fibers by crosslinking adjacent microtubules. Furthermore, the phosphorylation of TACC3 by Aurora-A is important for the formation of TACC3/ch-TOG/clathrin and its recruitment to kinetochore fibers. Recently, the aberrant expression of TACC3 in a variety of human cancers has been linked with mitotic defects. Thus, in this review, we will discuss our current understanding of the biological roles of TACC3 in mitotic spindle organization.
Collapse
Affiliation(s)
- Zhi-Ming Ding
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Chun-Jie Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Xiao-Fei Jiao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Di Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Li-Jun Huo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| |
Collapse
|
24
|
Bradshaw NJ, Hayashi MAF. NDE1 and NDEL1 from genes to (mal)functions: parallel but distinct roles impacting on neurodevelopmental disorders and psychiatric illness. Cell Mol Life Sci 2017; 74:1191-1210. [PMID: 27742926 PMCID: PMC11107680 DOI: 10.1007/s00018-016-2395-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023]
Abstract
NDE1 (Nuclear Distribution Element 1, also known as NudE) and NDEL1 (NDE-Like 1, also known as NudEL) are the mammalian homologues of the fungus nudE gene, with important and at least partially overlapping roles for brain development. While a large number of studies describe the various properties and functions of these proteins, many do not directly compare the similarities and differences between NDE1 and NDEL1. Although sharing a high degree structural similarity and multiple common cellular roles, each protein presents several distinct features that justify their parallel but also unique functions. Notably both proteins have key binding partners in dynein, LIS1 and DISC1, which impact on neurodevelopmental and psychiatric illnesses. Both are implicated in schizophrenia through genetic and functional evidence, with NDE1 also strongly implicated in microcephaly, as well as other neurodevelopmental and psychiatric conditions through copy number variation, while NDEL1 possesses an oligopeptidase activity with a unique potential as a biomarker in schizophrenia. In this review, we aim to give a comprehensive overview of the various cellular roles of these proteins in a "bottom-up" manner, from their biochemistry and protein-protein interactions on the molecular level, up to the consequences for neuronal differentiation, and ultimately to their importance for correct cortical development, with direct consequences for the pathophysiology of neurodevelopmental and mental illness.
Collapse
Affiliation(s)
- Nicholas J Bradshaw
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany.
| | - Mirian A F Hayashi
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, Brazil
| |
Collapse
|
25
|
Schneider MA, Christopoulos P, Muley T, Warth A, Klingmueller U, Thomas M, Herth FJF, Dienemann H, Mueller NS, Theis F, Meister M. AURKA, DLGAP5, TPX2, KIF11 and CKAP5: Five specific mitosis-associated genes correlate with poor prognosis for non-small cell lung cancer patients. Int J Oncol 2017; 50:365-372. [PMID: 28101582 PMCID: PMC5238780 DOI: 10.3892/ijo.2017.3834] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/05/2016] [Indexed: 11/08/2022] Open
Abstract
The growth of a tumor depends to a certain extent on an increase in mitotic events. Key steps during mitosis are the regulated assembly of the spindle apparatus and the separation of the sister chromatids. The microtubule-associated protein Aurora kinase A phosphorylates DLGAP5 in order to correctly segregate the chromatids. Its activity and recruitment to the spindle apparatus is regulated by TPX2. KIF11 and CKAP5 control the correct arrangement of the microtubules and prevent their degradation. In the present study, we investigated the role of these five molecules in non-small cell lung cancer (NSCLC). We analyzed the expression of the five genes in a large cohort of NSCLC patients (n=362) by quantitative real-time PCR. Each of the genes was highly overexpressed in the tumor tissues compared to corresponding normal lung tissue. The correlation of the expression of the individual genes depended on the histology. An increased expression of AURKA, DLGAP5, TPX2, KIF11 and CKAP5 was associated with poor overall survival (P=0.001–0.065). AURKA was a significant prognostic marker using multivariate analyses (P=0.006). Immunofluorescence studies demonstrated that the five mitosis-associated proteins co-localized with the spindle apparatus during cell division. Taken together, our data demonstrate that the expression of the mitosis-associated genes AURKA, DLGAP5, TPX2, KIF11 and CKAP5 is associated with the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Marc A Schneider
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Arne Warth
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Ursula Klingmueller
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Felix J F Herth
- Department of Pneumology and Critical Care Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Hendrik Dienemann
- Department of Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Nikola S Mueller
- Cellular Dynamics and Cell Patterning, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Fabian Theis
- Cellular Dynamics and Cell Patterning, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Michael Meister
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Fathi AT, Wander SA, Blonquist TM, Brunner AM, Amrein PC, Supko J, Hermance NM, Manning AL, Sadrzadeh H, Ballen KK, Attar EC, Graubert TA, Hobbs G, Joseph C, Perry AM, Burke M, Silver R, Foster J, Bergeron M, Ramos AY, Som TT, Fishman KM, McGregor KL, Connolly C, Neuberg DS, Chen YB. Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia. Haematologica 2016; 102:719-727. [PMID: 28034990 PMCID: PMC5395112 DOI: 10.3324/haematol.2016.158394] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/26/2016] [Indexed: 12/17/2022] Open
Abstract
Aberrant expression of aurora kinase A is implicated in the genesis of various
neoplasms, including acute myeloid leukemia. Alisertib, an aurora A kinase
inhibitor, has demonstrated efficacy as monotherapy in trials of myeloid
malignancy, and this efficacy appears enhanced in combination with conventional
chemotherapies. In this phase I, dose-escalation study, newly diagnosed patients
received conventional induction with cytarabine and idarubicin, after which
alisertib was administered for 7 days. Dose escalation occurred
via cohorts. Patients could then receive up to four cycles
of consolidation, incorporating alisertib, and thereafter alisertib maintenance
for up to 12 months. Twenty-two patients were enrolled. One dose limiting
toxicity occurred at dose level 2 (prolonged thrombocytopenia), and the
recommended phase 2 dose was established at 30mg twice daily. Common
therapy-related toxicities included cytopenias and mucositis. Only three
(14%) patients had persistent disease at mid-cycle, requiring
“5+2” reinduction. The composite remission rate (complete
remission and complete remission with incomplete neutrophil recovery) was
86% (nineteen of twenty-two patients; 90% CI
68–96%). Among those over age 65 and those with high-risk
disease (secondary acute leukemia or cytogenetically high-risk disease), the
composite remission rate was 88% and 100%, respectively. The
median follow up was 13.5 months. Of those treated at the recommended phase 2
dose, the 12-month overall survival and progression-free survival were
62% (90% CI 33–81%) and 42% (90%
CI 17–65%), respectively. Alisertib is well tolerated when
combined with induction chemotherapy in acute myeloid leukemia, with a promising
suggestion of efficacy. (clinicaltrials.gov Identifier:01779843).
Collapse
Affiliation(s)
- Amir T Fathi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Seth A Wander
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | | | - Andrew M Brunner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Philip C Amrein
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Jeffrey Supko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Nicole M Hermance
- Worcester Polytechnic Institute, Department of Biology, Worcester, MA, USA
| | - Amity L Manning
- Worcester Polytechnic Institute, Department of Biology, Worcester, MA, USA
| | - Hossein Sadrzadeh
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Karen K Ballen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Eyal C Attar
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Timothy A Graubert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Gabriela Hobbs
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Christelle Joseph
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Ashley M Perry
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Meghan Burke
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Regina Silver
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Julia Foster
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Meghan Bergeron
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Aura Y Ramos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Tina T Som
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Kaitlyn M Fishman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Kristin L McGregor
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Christine Connolly
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Donna S Neuberg
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Yi-Bin Chen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| |
Collapse
|
27
|
Liu SZ, Wei ZF, Meng XQ, Han XY, Cheng D, Zhong T, Zhang TL, Wang ZB. Exposure to Aroclor-1254 impairs spindle assembly during mouse oocyte maturation. ENVIRONMENTAL TOXICOLOGY 2016; 31:1652-1662. [PMID: 26174069 DOI: 10.1002/tox.22169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 06/17/2015] [Accepted: 06/27/2015] [Indexed: 06/04/2023]
Abstract
Polychlorinated biphenyls (PCBs), as typical environmental estrogen disruptors, are a structurally-related group of halogenated aromatic hydrocarbons that are composed of 209 isomers and present as a mixture in the environment. PCBs congener with different numbers and positions of chlorine atoms substituted on the biphenyl moiety. Aroclor-1254 is a mixture of more than 60 PCB congeners. Previous studies have provided the evidence that PCBs have severe negative effects on reproductive functions, but the effects of PCBs on spindle assembly during mouse oocyte maturation in vitro have not been reported. In the present study, female ICR mouse immature oocytes were cultured in M2 medium with 1 and 10 μg mL-1 Aroclor-1254 separately in vitro. The percentage of germinal vesicle breakdown (GVBD) and the first polar body extrusion were recorded. The results showed no significant difference in the percentage of GVBD or the first polar body extrusion between control oocytes and Aroclor-1254-treated oocytes. Further studies showed that the normal localization of γ-tubulin and Aurora-A kinase was interfered and α-tubulin assembling into spindle was affected when mouse oocytes were exposed to Aroclor-1254. The length of spindle from 10 μg mL-1 Aroclor-1254-treated oocytes was longer than that from control oocytes, and the spindle area in the Aroclor-1254-treated groups were decreased. Furthermore, the percentage of DNA damage in cumulus cells revealed an increase after exposed to Aroclor-1254. These results will provide the important reference for the prevention of reproductive disorders caused by PCBs. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1652-1662, 2016.
Collapse
Affiliation(s)
- Shu-Zhen Liu
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, Jinan, China.
| | - Ze-Feng Wei
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Xiao-Qian Meng
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, Jinan, China
| | - Xiao-Ying Han
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, Jinan, China
| | - Dong Cheng
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Tao Zhong
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, Jinan, China
| | - Tian-Liang Zhang
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
28
|
Hong JH, Kwak Y, Woo Y, Park C, Lee SA, Lee H, Park SJ, Suh Y, Suh BK, Goo BS, Mun DJ, Sanada K, Nguyen MD, Park SK. Regulation of the actin cytoskeleton by the Ndel1-Tara complex is critical for cell migration. Sci Rep 2016; 6:31827. [PMID: 27546710 PMCID: PMC4992831 DOI: 10.1038/srep31827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/27/2016] [Indexed: 11/16/2022] Open
Abstract
Nuclear distribution element-like 1 (Ndel1) plays pivotal roles in diverse biological processes and is implicated in the pathogenesis of multiple neurodevelopmental disorders. Ndel1 function by regulating microtubules and intermediate filaments; however, its functional link with the actin cytoskeleton is largely unknown. Here, we show that Ndel1 interacts with TRIO-associated repeat on actin (Tara), an actin-bundling protein, to regulate cell movement. In vitro wound healing and Boyden chamber assays revealed that Ndel1- or Tara-deficient cells were defective in cell migration. Moreover, Tara overexpression induced the accumulation of Ndel1 at the cell periphery and resulted in prominent co-localization with F-actin. This redistribution of Ndel1 was abolished by deletion of the Ndel1-interacting domain of Tara, suggesting that the altered peripheral localization of Ndel1 requires a physical interaction with Tara. Furthermore, co-expression of Ndel1 and Tara in SH-SY5Y cells caused a synergistic increase in F-actin levels and filopodia formation, suggesting that Tara facilitates cell movement by sequestering Ndel1 at peripheral structures to regulate actin remodeling. Thus, we demonstrated that Ndel1 interacts with Tara to regulate cell movement. These findings reveal a novel role of the Ndel1-Tara complex in actin reorganization during cell movement.
Collapse
Affiliation(s)
- Ji-Ho Hong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Yongdo Kwak
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Cana Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Seol-Ae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Haeryun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Sung Jin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Kamon Sanada
- Molecular Genetics Research Laboratory, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, University of Calgary, Calgary T2N 4N1, Canada
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| |
Collapse
|
29
|
Zhou DS, Wang HB, Zhou ZG, Zhang YJ, Zhong Q, Xu L, Huang YH, Yeung SC, Chen MS, Zeng MS. TACC3 promotes stemness and is a potential therapeutic target in hepatocellular carcinoma. Oncotarget 2016. [PMID: 26219398 PMCID: PMC4695177 DOI: 10.18632/oncotarget.4643] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transforming acidic coiled-coil protein 3 (TACC3) is essential for cell mitosis and transcriptional functions. In the present study, we first demonstrated that both TACC3 protein and mRNA levels were elevated in HCC tissue samples compared with non-cancerous tissue biopsies according to western blot analyses, immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR) assays. Moreover, high TACC3 expression was positively correlated with poor overall survival (OS) and disease-free survival (DFS) (p < 0.001). Using HCC cell lines, we then demonstrated that either TACC3 knockdown or treatment with the potential TACC3 inhibitor KHS101 suppressed cell growth and sphere formation as well as the expression of stem cell transcription factors, including Bmi1, c-Myc and Nanog. Silencing TACC3 may suppress the Wnt/β-catenin and PI3K/AKT signaling pathways, which regulate cancer stem cell-like characteristics. Taken together, these data suggest that TACC3 is enriched in HCC and that TACC3 down-regulation inhibits the proliferation, clonogenicity, and cancer stem cell-like phenotype of HCC cells. KHS101, a TACC3 inhibitor, may serve as a novel therapeutic agent for HCC patients with tumors characterized by high TACC3 expression.
Collapse
Affiliation(s)
- Dong-Sheng Zhou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China.,Shandong Provincial Qianfoshan Hospital, Jinan, P. R. China
| | - Hong-Bo Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhong-Guo Zhou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Yao-Jun Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Qian Zhong
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Li Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Yue-Hua Huang
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Sai-Ching Yeung
- Department of General Internal Medicine, Ambulatory Treatment and Emergency Care, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min-Shan Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Mu-Sheng Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| |
Collapse
|
30
|
Barretta ML, Spano D, D'Ambrosio C, Cervigni RI, Scaloni A, Corda D, Colanzi A. Aurora-A recruitment and centrosomal maturation are regulated by a Golgi-activated pool of Src during G2. Nat Commun 2016; 7:11727. [PMID: 27242098 PMCID: PMC4895030 DOI: 10.1038/ncomms11727] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/25/2016] [Indexed: 02/02/2023] Open
Abstract
The Golgi apparatus is composed of stacks of cisternae laterally connected by tubules to form a ribbon-like structure. At the onset of mitosis, the Golgi ribbon is broken down into discrete stacks, which then undergo further fragmentation. This ribbon cleavage is required for G2/M transition, which thus indicates that a ‘Golgi mitotic checkpoint' couples Golgi inheritance with cell cycle transition. We previously showed that the Golgi-checkpoint regulates the centrosomal recruitment of the mitotic kinase Aurora-A; however, how the Golgi unlinking regulates this recruitment was unknown. Here we show that, in G2, Aurora-A recruitment is promoted by activated Src at the Golgi. Our data provide evidence that Src and Aurora-A interact upon Golgi ribbon fragmentation; Src phosphorylates Aurora-A at tyrosine 148 and this specific phosphorylation is required for Aurora-A localization at the centrosomes. This process, pivotal for centrosome maturation, is a fundamental prerequisite for proper spindle formation and chromosome segregation. The Golgi mitotic checkpoint couples Golgi inheritance with cell cycle transition, and regulates centrosomal recruitment of the mitotic kinase Aurora-A. Here the authors show that upon Golgi ribbon fragmentation in G2, Src phosphorylates Aurora-A at the Golgi, driving its localization to the centrosomes.
Collapse
Affiliation(s)
- Maria Luisa Barretta
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Daniela Spano
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Chiara D'Ambrosio
- Proteomics and Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, ISPAAM, National Research Council (CNR), Via Argine 1085, 80147 Naples, Italy
| | - Romina Ines Cervigni
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Andrea Scaloni
- Proteomics and Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, ISPAAM, National Research Council (CNR), Via Argine 1085, 80147 Naples, Italy
| | - Daniela Corda
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Antonino Colanzi
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy
| |
Collapse
|
31
|
Tuccilli C, Baldini E, Prinzi N, Morrone S, Sorrenti S, Filippini A, Catania A, Alessandrini S, Rendina R, Coccaro C, D'Armiento M, Ulisse S. Preclinical testing of selective Aurora kinase inhibitors on a medullary thyroid carcinoma-derived cell line. Endocrine 2016. [PMID: 26215279 DOI: 10.1007/s12020-015-0700-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Deregulated expression of the Aurora kinases (Aurora-A, B, and C) is thought to be involved in cell malignant transformation and genomic instability in several cancer types. Over the last decade, a number of small-molecule inhibitors of Aurora kinases have been developed, which have proved to efficiently restrain malignant cell growth and tumorigenicity. Regarding medullary thyroid carcinoma (MTC), we previously showed the efficacy of a pan-Aurora kinase inhibitor (MK-0457) in impairing growth and survival of the MTC-derived cell line TT. In the present study, we sought to establish if one of the Aurora kinases might represent a preferential target for MTC therapy. The effects of selective inhibitors of Aurora-A (MLN8237) and Aurora-B (AZD1152) were analyzed on TT cell proliferation, apoptosis, cell cycle, and ploidy. The two inhibitors reduced TT cell proliferation in a time- and dose-dependent manner, with IC50 of 19.0 ± 2.4 nM for MLN8237 and 401.6 ± 44.1 nM for AZD1152. Immunofluorescence experiments confirmed that AZD1152 inhibited phosphorylation of histone H3 (Ser10) by Aurora-B, while it did not affect Aurora-A autophosphorylation. MLN8237 inhibited Aurora-A autophosphorylation as expected, but at concentrations required to achieve the maximum antiproliferative effects it also abolished H3 (Ser10) phosphorylation. Cytofluorimetry experiments showed that both inhibitors induced accumulation of cells in G2/M phase and increased the subG0/G1 fraction and polyploidy. Finally, both inhibitors triggered apoptosis. We demonstrated that inhibition of either Aurora-A or Aurora-B has antiproliferative effects on TT cells, and thus it would be worthwhile to further investigate the therapeutical potential of Aurora kinase inhibitors in MTC treatment.
Collapse
Affiliation(s)
- Chiara Tuccilli
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Enke Baldini
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Natalie Prinzi
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Stefania Morrone
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Salvatore Sorrenti
- Department of Surgical Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Angelo Filippini
- Department of Surgical Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Antonio Catania
- Department of Surgical Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Stefania Alessandrini
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Roberta Rendina
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Carmela Coccaro
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Massimino D'Armiento
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy
| | - Salvatore Ulisse
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161, Rome, Italy.
| |
Collapse
|
32
|
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
|
33
|
Reboutier D, Benaud C, Prigent C. Aurora A's Functions During Mitotic Exit: The Guess Who Game. Front Oncol 2015; 5:290. [PMID: 26734572 PMCID: PMC4685928 DOI: 10.3389/fonc.2015.00290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/07/2015] [Indexed: 11/24/2022] Open
Abstract
Until recently, the knowledge of Aurora A kinase functions during mitosis was limited to pre-metaphase events, particularly centrosome maturation, G2/M transition, and mitotic spindle assembly. However, an involvement of Aurora A in post-metaphase events was also suspected, but not clearly demonstrated due to the technical difficulty to perform the appropriate experiments. Recent developments of both an analog-specific version of Aurora A and small molecule inhibitors have led to the first demonstration that Aurora A is required for the early steps of cytokinesis. As in pre-metaphase, Aurora A plays diverse functions during anaphase, essentially participating in astral microtubules dynamics and central spindle assembly and functioning. The present review describes the experimental systems used to decipher new functions of Aurora A during late mitosis and situate these functions into the context of cytokinesis mechanisms.
Collapse
Affiliation(s)
- David Reboutier
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Christelle Benaud
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Claude Prigent
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| |
Collapse
|
34
|
Tsunematsu T, Arakaki R, Yamada A, Ishimaru N, Kudo Y. The Non-Canonical Role of Aurora-A in DNA Replication. Front Oncol 2015; 5:187. [PMID: 26380219 PMCID: PMC4548192 DOI: 10.3389/fonc.2015.00187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/05/2015] [Indexed: 12/04/2022] Open
Abstract
Aurora-A is a well-known mitotic kinase that regulates mitotic entry, spindle formation, and chromosome maturation as a canonical role. During mitosis, Aurora-A protein is stabilized by its phosphorylation at Ser51 via blocking anaphase-promoting complex/cyclosome-mediated proteolysis. Importantly, overexpression and/or hyperactivation of Aurora-A is involved in tumorigenesis via aneuploidy and genomic instability. Recently, the novel function of Aurora-A for DNA replication has been revealed. In mammalian cells, DNA replication is strictly regulated for preventing over-replication. Pre-replication complex (pre-RC) formation is required for DNA replication as an initiation step occurring at the origin of replication. The timing of pre-RC formation depends on the protein level of geminin, which is controlled by the ubiquitin–proteasome pathway. Aurora-A phosphorylates geminin to prevent its ubiquitin-mediated proteolysis at the mitotic phase to ensure proper pre-RC formation and ensuing DNA replication. In this review, we introduce the novel non-canonical role of Aurora-A in DNA replication.
Collapse
Affiliation(s)
- Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Akiko Yamada
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| |
Collapse
|
35
|
Chou CH, Loh JK, Yang MC, Lin CC, Hong MC, Cho CL, Chou AK, Wang CH, Lieu AS, Howng SL, Hsu CM, Hong YR. AIBp regulates mitotic entry and mitotic spindle assembly by controlling activation of both Aurora-A and Plk1. Cell Cycle 2015; 14:2764-76. [PMID: 26114227 PMCID: PMC4614063 DOI: 10.1080/15384101.2015.1066536] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/16/2015] [Accepted: 06/23/2015] [Indexed: 10/23/2022] Open
Abstract
We previously reported that Aurora-A and the hNinein binding protein AIBp facilitate centrosomal structure maintenance and contribute to spindle formation. Here, we report that AIBp also interacts with Plk1, raising the possibility of functional similarity to Bora, which subsequently promotes Aurora-A-mediated Plk1 activation at Thr210 as well as Aurora-A activation at Thr288. In kinase assays, AIBp acts not only as a substrate but also as a positive regulator of both Aurora-A and Plk1. However, AIBp functions as a negative regulator to block phosphorylation of hNinein mediated by Aurora-A and Plk1. These findings suggest a novel AIBp-dependent regulatory machinery that controls mitotic entry. Additionally, knockdown of hNinein caused failure of AIBp to target the centrosome, whereas depletion of AIBp did not affect the localization of hNinein and microtubule nucleation. Notably, knockdown of AIBp in HeLa cells impaired both Aurora-A and Plk1 kinase, resulting in phenotypes with multiple spindle pole formation and chromosome misalignment. Our data show that depletion of AIBp results in the mis-localization of TACC3 and ch-TOG, but not CEP192 and CEP215, suggesting that loss of AIBp dominantly affects the Aurora-A substrate to cause mitotic aberrations. Collectively, our data demonstrate that AIBp contributes to mitotic entry and bipolar spindle assembly and may partially control localization, phosphorylation, and activation of both Aurora-A and Plk1 via hNinein during mitotic progression.
Collapse
Affiliation(s)
- Chia-Hua Chou
- Department of Biochemistry; Faculty of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
- Department of Biological Sciences; National Sun Yat-Sen University; Kaohsiung, Taiwan
| | - Joon-Khim Loh
- Department of Surgery; Kaohsiung Municipal Hsiao-Kang Hospital; Kaohsiung Medical University; Kaohsiung, Taiwan
- Graduate Institute of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
- Department of Neurosurgery; Kaohsiung Medical University Hospital; Kaohsiung, Taiwan
| | - Ming-Chang Yang
- Department of Biological Sciences; National Sun Yat-Sen University; Kaohsiung, Taiwan
- Laboratory of Medical Research; Center of Education and Faculty Development; Kaohsiung Armed Forces General Hospital; Kaohsiung, Taiwan
| | - Ching-Chih Lin
- Department of Biochemistry; Faculty of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
| | - Ming-Chang Hong
- Department of Biochemistry; Faculty of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
| | - Chung-Lung Cho
- Department of Biological Sciences; National Sun Yat-Sen University; Kaohsiung, Taiwan
| | - An-Kuo Chou
- Department of Anesthesiology; Kaohsiung Chang Gung Memorial Hospital and College of Medicine; Chang Gung University; Kaohsiung, Taiwan
| | - Chi-Huei Wang
- Department of Biotechnology; Kaohsiung Medical University; Kaohsiung, Taiwan
| | - Ann-Shung Lieu
- Graduate Institute of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
- Department of Neurosurgery; Kaohsiung Medical University Hospital; Kaohsiung, Taiwan
| | - Shen-Long Howng
- Graduate Institute of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
- Department of Neurosurgery; Kaohsiung Medical University Hospital; Kaohsiung, Taiwan
| | - Ching-Mei Hsu
- Department of Biological Sciences; National Sun Yat-Sen University; Kaohsiung, Taiwan
| | - Yi-Ren Hong
- Department of Biochemistry; Faculty of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
- Department of Biological Sciences; National Sun Yat-Sen University; Kaohsiung, Taiwan
- Graduate Institute of Medicine; Kaohsiung Medical University; Kaohsiung, Taiwan
| |
Collapse
|
36
|
Arthur AL, Yang SZ, Abellaneda AM, Wildonger J. Dendrite arborization requires the dynein cofactor NudE. J Cell Sci 2015; 128:2191-201. [PMID: 25908857 PMCID: PMC4450295 DOI: 10.1242/jcs.170316] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/10/2015] [Indexed: 12/28/2022] Open
Abstract
The microtubule-based molecular motor dynein is essential for proper neuronal morphogenesis. Dynein activity is regulated by cofactors, and the role(s) of these cofactors in shaping neuronal structure are still being elucidated. Using Drosophila melanogaster, we reveal that the loss of the dynein cofactor NudE results in abnormal dendrite arborization. Our data show that NudE associates with Golgi outposts, which mediate dendrite branching, suggesting that NudE normally influences dendrite patterning by regulating Golgi outpost transport. Neurons lacking NudE also have increased microtubule dynamics, reflecting a change in microtubule stability that is likely to also contribute to abnormal dendrite growth and branching. These defects in dendritogenesis are rescued by elevating levels of Lis1, another dynein cofactor that interacts with NudE as part of a tripartite complex. Our data further show that the NudE C-terminus is dispensable for dendrite morphogenesis and is likely to modulate NudE activity. We propose that a key function of NudE is to enhance an interaction between Lis1 and dynein that is crucial for motor activity and dendrite architecture.
Collapse
Affiliation(s)
- Ashley L Arthur
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sihui Z Yang
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Allison M Abellaneda
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA Biochemistry Scholars Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
37
|
DBZ regulates cortical cell positioning and neurite development by sustaining the anterograde transport of Lis1 and DISC1 through control of Ndel1 dual-phosphorylation. J Neurosci 2015; 35:2942-58. [PMID: 25698733 DOI: 10.1523/jneurosci.5029-13.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cell positioning and neuronal network formation are crucial for proper brain function. Disrupted-in-Schizophrenia 1 (DISC1) is anterogradely transported to the neurite tips, together with Lis1, and functions in neurite extension via suppression of GSK3β activity. Then, transported Lis1 is retrogradely transported and functions in cell migration. Here, we show that DISC1-binding zinc finger protein (DBZ), together with DISC1, regulates mouse cortical cell positioning and neurite development in vivo. DBZ hindered Ndel1 phosphorylation at threonine 219 and serine 251. DBZ depletion or expression of a double-phosphorylated mimetic form of Ndel1 impaired the transport of Lis1 and DISC1 to the neurite tips and hampered microtubule elongation. Moreover, application of DISC1 or a GSK3β inhibitor rescued the impairments caused by DBZ insufficiency or double-phosphorylated Ndel1 expression. We concluded that DBZ controls cell positioning and neurite development by interfering with Ndel1 from disproportionate phosphorylation, which is critical for appropriate anterograde transport of the DISC1-complex.
Collapse
|
38
|
Cep68 can be regulated by Nek2 and SCF complex. Eur J Cell Biol 2015; 94:162-72. [PMID: 25704143 DOI: 10.1016/j.ejcb.2015.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 01/01/2023] Open
Abstract
Centrosome cohesion maintains centrosomes in close proximity until mitosis, when cell cycle-dependent regulatory signaling events dissolve cohesion and promote centrosome separation in preparation for bipolar spindle assembly at mitosis. Cohesion is regulated by the antagonistic activities of the mitotic NIMA-related kinase 2 (Nek2), protein phosphatase 1, the cohesion fiber components rootletin, centrosomal Nek2-associated protein 1 (C-Nap1) and Cep68. The centrosomal protein Cep68 is essential for centrosome cohesion and dissociates from centrosomes at the onset of mitosis. Here, our cell line studies show the C-terminal 300-400 amino acids of Cep68 are necessary to localize Cep68 to interphase centrosomes while C-terminal 400-500 amino acids might regulate Cep68 dissociation from centrosomes at mitotic onset. In addition, Nek2 was demonstrated to phosphorylate Cep68 in vivo and this phosphorylation appears to promote Cep68 degradation in mitosis. We further show that the SCF complex destroys Cep68 at mitosis through recognition by the beta-Trcp F box component of SCF. Together, the findings provide a new insight into the control of centrosome separation by Cep68 during mitosis.
Collapse
|
39
|
Abstract
The centrosome was discovered in the late 19th century when mitosis was first described. Long recognized as a key organelle of the spindle pole, its core component, the centriole, was realized more than 50 or so years later also to comprise the basal body of the cilium. Here, we chart the more recent acquisition of a molecular understanding of centrosome structure and function. The strategies for gaining such knowledge were quickly developed in the yeasts to decipher the structure and function of their distinctive spindle pole bodies. Only within the past decade have studies with model eukaryotes and cultured cells brought a similar degree of sophistication to our understanding of the centrosome duplication cycle and the multiple roles of this organelle and its component parts in cell division and signaling. Now as we begin to understand these functions in the context of development, the way is being opened up for studies of the roles of centrosomes in human disease.
Collapse
Affiliation(s)
- Jingyan Fu
- Cancer Research UK Cell Cycle Genetics Group, Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Iain M Hagan
- Cancer Research UK Manchester Institute, University of Manchester, Withington, Manchester M20 4BX, United Kingdom
| | - David M Glover
- Cancer Research UK Cell Cycle Genetics Group, Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| |
Collapse
|
40
|
Li Z, Sun Y, Chen X, Squires J, Nowroozizadeh B, Liang C, Huang J. p53 Mutation Directs AURKA Overexpression via miR-25 and FBXW7 in Prostatic Small Cell Neuroendocrine Carcinoma. Mol Cancer Res 2014; 13:584-91. [PMID: 25512615 DOI: 10.1158/1541-7786.mcr-14-0277-t] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Prostatic small cell neuroendocrine carcinoma (SCNC) is a rare but aggressive form of prostate cancer that is negative for androgen receptor (AR) and not responsive to hormonal therapy. The molecular etiology of this prostate cancer variant is not well understood; however, mutation of the p53 (TP53) tumor suppressor in prostate neuroendocrine cells inactivates the IL8-CXCR2-p53 pathway that normally inhibits cellular proliferation, leading to the development of SCNC. SCNC also overexpresses Aurora kinase A (AURKA) which is considered to be a viable therapeutic target. Therefore, the relationship of these two molecular events was studied, and we show that p53 mutation leads to increased expression of miR-25 and downregulation of the E3 ubiquitin ligase FBXW7, resulting in elevated levels of Aurora kinase A. This study demonstrates an intracellular pathway by which p53 mutation leads to Aurora kinase A expression, which is critically important for the rapid proliferation and aggressive behavior of prostatic SCNC. IMPLICATIONS The pathogenesis of prostatic SCNC involves a p53 and Aurora Kinase A signaling mechanism, both potentially targetable pathways.
Collapse
Affiliation(s)
- Zhen Li
- Department of Pathology and Urology, Jonsson Comprehensive Cancer Center and Broad Center of Regenerative Medicine and Stem Cell Research, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Yin Sun
- Department of Pathology and Urology, Jonsson Comprehensive Cancer Center and Broad Center of Regenerative Medicine and Stem Cell Research, UCLA David Geffen School of Medicine, Los Angeles, California.
| | - Xufeng Chen
- Department of Pathology and Urology, Jonsson Comprehensive Cancer Center and Broad Center of Regenerative Medicine and Stem Cell Research, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Jill Squires
- Department of Pathology and Urology, Jonsson Comprehensive Cancer Center and Broad Center of Regenerative Medicine and Stem Cell Research, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Behdokht Nowroozizadeh
- Department of Pathology and Urology, Jonsson Comprehensive Cancer Center and Broad Center of Regenerative Medicine and Stem Cell Research, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiaoti Huang
- Department of Pathology and Urology, Jonsson Comprehensive Cancer Center and Broad Center of Regenerative Medicine and Stem Cell Research, UCLA David Geffen School of Medicine, Los Angeles, California.
| |
Collapse
|
41
|
Shagisultanova E, Dunbrack RL, Golemis EA. Issues in interpreting the in vivo activity of Aurora-A. Expert Opin Ther Targets 2014; 19:187-200. [PMID: 25384454 DOI: 10.1517/14728222.2014.981154] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Based on its role as a mitotic regulatory kinase, overexpressed and associated with aneuploidy in cancer, small-molecule inhibitors have been developed for Aurora-A (AURKA) kinase. In preclinical and clinical assessments, these agents have shown efficacy in inducing stable disease or therapeutic response. In optimizing the use of Aurora-A inhibitors, it is critical to have robust capacity to measure the kinase activity of Aurora-A in tumors. AREAS COVERED We provide an overview of molecular mechanisms of mitotic and non-mitotic activation of Aurora-A kinase, and interaction of Aurora-A with its regulatory partners. Typically, Aurora-A activity is measured by use of phospho-antibodies targeting an autophosphorylated T288 epitope. However, recent studies have identified alternative means of Aurora-A activation control, including allosteric regulation by partners, phosphorylation on alternative activating residues (S51, S98), dephosphorylation on inhibitory sites (S342) and T288 phosphorylation by alternative kinases such as Pak enzymes. Additional work has shown that the relative abundance of Aurora-A partners can affect the activity of Aurora-A inhibitors, and that Aurora-A activation also occurs in interphase cells. EXPERT OPINION Taken together, this work suggests the need for comprehensive analysis of Aurora-A activity and expression of Aurora-A partners in order to stratify patients for likely therapeutic response.
Collapse
Affiliation(s)
- Elena Shagisultanova
- Fox Chase Cancer Center, Department of Medical Oncology , Philadelphia, PA 19111 , USA
| | | | | |
Collapse
|
42
|
Aradottir M, Reynisdottir ST, Stefansson OA, Jonasson JG, Sverrisdottir A, Tryggvadottir L, Eyfjord JE, Bodvarsdottir SK. Aurora A is a prognostic marker for breast cancer arising in BRCA2 mutation carriers. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2014; 1:33-40. [PMID: 27499891 PMCID: PMC4858119 DOI: 10.1002/cjp2.6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/07/2014] [Indexed: 12/20/2022]
Abstract
Overexpression of the Aurora A kinase has been shown to have prognostic value in breast cancer. Previously, we showed a significant association between AURKA gene amplification and BRCA2 mutation in breast cancer. The aim of this study was to assess the prognostic impact of Aurora A overexpression on breast cancer arising in BRCA2 mutation carriers. Aurora A expression was evaluated by immunohistochemistry on breast tumour tissue microarrays from 107 BRCA2 999del5 mutation carriers and 284 of sporadic origin. Prognostic value of Aurora A nuclear staining was estimated in relation to clinical markers and adjuvant treatment, using multivariate Cox's proportional hazards ratio regression model. BRCA2 wild‐type allele loss was measured by TaqMan in BRCA2 mutated tumour samples. All statistical tests were two sided. Multivariate analysis of breast cancer‐specific survival, including proliferative markers and treatment, indicated independent prognostic value of Aurora A nuclear staining for BRCA2 mutation carriers (hazards ratio = 7.06; 95% confidence interval = 1.23–40.6; p = 0.028). Poor breast cancer‐specific survival of BRCA2 mutation carriers was found to be significantly associated with combined Aurora A nuclear expression and BRCA2 wild type allele loss in tumours (p < 0.001). Multivariate analysis indicated independent prognostic value of both positive Aurora A nuclear staining (hazards ratio = 10.09; 95% confidence interval = 1.19–85.4, p = 0.034) and BRCA2 wild type allele loss (hazards ratio = 9.63; 95% confidence interval = 1.81–51.0, p = 0.008) for BRCA2 mutation carriers. Aurora A nuclear expression was found to be a significant prognostic marker for BRCA2 mutation carriers, independent of clinical parameters and adjuvant treatment. Our conclusion is that treatment benefits for BRCA2 mutation carriers and sporadic breast cancer patients with Aurora A positive tumours may be enhanced by giving attention to Aurora A targeted treatment.
Collapse
Affiliation(s)
- Margret Aradottir
- Cancer Research Laboratory, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland Reykjavik Iceland
| | - Sigridur T Reynisdottir
- Cancer Research Laboratory, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland Reykjavik Iceland
| | - Olafur A Stefansson
- Cancer Research Laboratory, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland Reykjavik Iceland
| | - Jon G Jonasson
- Faculty of MedicineSchool of Health Sciences, University of IcelandReykjavikIceland; Icelandic Cancer RegistryIcelandic Cancer SocietyReykjavikIceland; Department of PathologyNational University HospitalReykjavikIceland
| | | | - Laufey Tryggvadottir
- Faculty of MedicineSchool of Health Sciences, University of IcelandReykjavikIceland; Icelandic Cancer RegistryIcelandic Cancer SocietyReykjavikIceland
| | - Jorunn E Eyfjord
- Cancer Research Laboratory, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland Reykjavik Iceland
| | - Sigridur K Bodvarsdottir
- Cancer Research Laboratory, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland Reykjavik Iceland
| |
Collapse
|
43
|
Aurora A kinase modulates actin cytoskeleton through phosphorylation of Cofilin: Implication in the mitotic process. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2719-29. [PMID: 25090971 DOI: 10.1016/j.bbamcr.2014.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/22/2022]
Abstract
Aurora A kinase regulates early mitotic events through phosphorylation and activation of a variety of proteins. Specifically, Aur-A is involved in centrosomal separation and formation of mitotic spindles in early prophase. The effect of Aur-A on mitotic spindles is mediated by the modulation of microtubule dynamics and association with microtubule binding proteins. In this study we show that Aur-A exerts its effects on spindle organization through the regulation of the actin cytoskeleton. Aurora A phosphorylates Cofilin at multiple sites including S(3) resulting in the inactivation of its actin depolymerizing function. Aur-A interacts with Cofilin in early mitotic phases and regulates its phosphorylation status. Cofilin phosphorylation follows a dynamic pattern during the progression of prophase to metaphase. Inhibition of Aur-A activity induced a delay in the progression of prophase to metaphase. Aur-A inhibitor also disturbed the pattern of Cofilin phosphorylation, which correlated with the mitotic delay. Our results establish a novel function of Aur-A in the regulation of actin cytoskeleton reorganization, through Cofilin phosphorylation during early mitotic stages.
Collapse
|
44
|
Adaptors for disorders of the brain? The cancer signaling proteins NEDD9, CASS4, and PTK2B in Alzheimer's disease. Oncoscience 2014; 1:486-503. [PMID: 25594051 PMCID: PMC4278314 DOI: 10.18632/oncoscience.64] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/23/2014] [Indexed: 12/19/2022] Open
Abstract
No treatment strategies effectively limit the progression of Alzheimer's disease (AD), a common and debilitating neurodegenerative disorder. The absence of viable treatment options reflects the fact that the pathophysiology and genotypic causes of the disease are not well understood. The advent of genome-wide association studies (GWAS) has made it possible to broadly investigate genotypic alterations driving phenotypic occurrences. Recent studies have associated single nucleotide polymorphisms (SNPs) in two paralogous scaffolding proteins, NEDD9 and CASS4, and the kinase PTK2B, with susceptibility to late-onset AD (LOAD). Intriguingly, NEDD9, CASS4, and PTK2B have been much studied as interacting partners regulating oncogenesis and metastasis, and all three are known to be active in the brain during development and in cancer. However, to date, the majority of studies of these proteins have emphasized their roles in the directly cancer relevant processes of migration and survival signaling. We here discuss evidence for roles of NEDD9, CASS4 and PTK2B in additional processes, including hypoxia, vascular changes, inflammation, microtubule stabilization and calcium signaling, as potentially relevant to the pathogenesis of LOAD. Reciprocally, these functions can better inform our understanding of the action of NEDD9, CASS4 and PTK2B in cancer.
Collapse
|
45
|
Zheng F, Li T, Jin DY, Syrovatkina V, Scheffler K, Tran PT, Fu C. Csi1p recruits alp7p/TACC to the spindle pole bodies for bipolar spindle formation. Mol Biol Cell 2014; 25:2750-60. [PMID: 25057016 PMCID: PMC4161510 DOI: 10.1091/mbc.e14-03-0786] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The spindle pole body (SPB) localization of the fission yeast Schizosaccharomyces pombe TACC orthologue alp7p depends on the SPB protein csi1p. Compromised interaction between csi1p and alp7p delays bipolar spindle formation and leads to abnormal chromosome segregation. Accurate chromosome segregation requires timely bipolar spindle formation during mitosis. The transforming acidic coiled-coil (TACC) family proteins and the ch-TOG family proteins are key players in bipolar spindle formation. They form a complex to stabilize spindle microtubules, mainly dependent on their localization to the centrosome (the spindle pole body [SPB] in yeast). The molecular mechanism underlying the targeting of the TACC–ch-TOG complex to the centrosome remains unclear. Here we show that the fission yeast Schizosaccharomyces pombe TACC orthologue alp7p is recruited to the SPB by csi1p. The csi1p-interacting region lies within the conserved TACC domain of alp7p, and the carboxyl-terminal domain of csi1p is responsible for interacting with alp7p. Compromised interaction between csi1p and alp7p impairs the localization of alp7p to the SPB during mitosis, thus delaying bipolar spindle formation and leading to anaphase B lagging chromosomes. Hence our study establishes that csi1p serves as a linking molecule tethering spindle-stabilizing factors to the SPB for promoting bipolar spindle assembly.
Collapse
Affiliation(s)
- Fan Zheng
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong, China HKU-Shenzhen Institute of Research and Innovation, University of Hong Kong, Shenzhen, China
| | - Tianpeng Li
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong, China HKU-Shenzhen Institute of Research and Innovation, University of Hong Kong, Shenzhen, China
| | - Dong-Yan Jin
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Kathleen Scheffler
- Institut Curie, Centre National de la Recherche Scientifique, Paris 75005, France
| | - Phong T Tran
- Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 10104 Institut Curie, Centre National de la Recherche Scientifique, Paris 75005, France
| | - Chuanhai Fu
- Department of Biochemistry, University of Hong Kong, Pokfulam, Hong Kong, China HKU-Shenzhen Institute of Research and Innovation, University of Hong Kong, Shenzhen, China
| |
Collapse
|
46
|
Chen H, Mohan P, Jiang J, Nemirovsky O, He D, Fleisch MC, Niederacher D, Pilarski LM, Lim CJ, Maxwell CA. Spatial regulation of Aurora A activity during mitotic spindle assembly requires RHAMM to correctly localize TPX2. Cell Cycle 2014; 13:2248-61. [PMID: 24875404 DOI: 10.4161/cc.29270] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Construction of a mitotic spindle requires biochemical pathways to assemble spindle microtubules and structural proteins to organize these microtubules into a bipolar array. Through a complex with dynein, the receptor for hyaluronan-mediated motility (RHAMM) cross-links mitotic microtubules to provide structural support, maintain spindle integrity, and correctly orient the mitotic spindle. Here, we locate RHAMM to sites of microtubule assembly at centrosomes and non-centrosome sites near kinetochores and demonstrate that RHAMM is required for the activation of Aurora kinase A. Silencing of RHAMM delays the kinetics of spindle assembly, mislocalizes targeting protein for XKlp2 (TPX2), and attenuates the localized activation of Aurora kinase A with a consequent reduction in mitotic spindle length. The RHAMM-TPX2 complex requires a C-terminal basic leucine zipper in RHAMM and a domain that includes the nuclear localization signal in TPX2. Together, our findings identify RHAMM as a critical regulator for Aurora kinase A signaling and suggest that RHAMM ensures bipolar spindle assembly and mitotic progression through the integration of biochemical and structural pathways.
Collapse
Affiliation(s)
- Helen Chen
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| | - Pooja Mohan
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| | - Jihong Jiang
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| | - Oksana Nemirovsky
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| | - Daniel He
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| | - Markus C Fleisch
- Department of Gynaecology and Obstetrics; University Hospital Düsseldorf; Heinrich-Heine University; Düsseldorf, Germany
| | - Dieter Niederacher
- Department of Gynaecology and Obstetrics; University Hospital Düsseldorf; Heinrich-Heine University; Düsseldorf, Germany
| | - Linda M Pilarski
- Department of Oncology; University of Alberta and Cross Cancer Institute; Edmonton, Alberta, Canada
| | - C James Lim
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| | - Christopher A Maxwell
- Department of Pediatrics; Child and Family Research Institute; University of British Columbia; Vancouver, British Columbia, Canada
| |
Collapse
|
47
|
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: 102] [Impact Index Per Article: 9.3] [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
|
48
|
Pei Z, Lang B, Fragoso YD, Shearer KD, Zhao L, Mccaffery PJA, Shen S, Ding YQ, McCaig CD, Collinson JM. The expression and roles of Nde1 and Ndel1 in the adult mammalian central nervous system. Neuroscience 2014; 271:119-36. [PMID: 24785679 PMCID: PMC4048543 DOI: 10.1016/j.neuroscience.2014.04.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/08/2014] [Accepted: 04/18/2014] [Indexed: 11/01/2022]
Abstract
Mental and neurological illnesses affect one in four people. While genetic linkage analyses have shown an association of nuclear distribution factor E (NDE1, or NudE) and its ohnolog NDE-like 1 (NDEL1, or Nudel) with mental disorders, the cellular mechanisms remain unclear. In the present study, we have demonstrated that Nde1 and Ndel1 are differentially localised in the subventricular zone (SVZ) of the forebrain and the subgranular zone (SGZ) of the hippocampus, two regions where neurogenesis actively occurs in the adult brain. Nde1, but not Ndel1, is localized to putative SVZ stem cells, and to actively dividing progenitors of the SGZ. The influence of these proteins on neural stem cell differentiation was investigated by overexpression in a hippocampal neural stem cell line, HCN-A94. Increasing Nde1 expression in this neural stem cell line led to increased neuronal differentiation while decreasing levels of astroglial differentiation. In primary cultured neurons and astrocytes, Nde1 and Ndel1 were found to have different but comparable subcellular localizations. In addition, we have shown for the first time that Nde1 is heterogeneously distributed in cortical astrocytes of human brains. Our data indicate that Nde1 and Ndel1 have distinct but overlapping distribution patterns in mouse brain and cultured nerve cells. They may function differently and therefore their dosage changes may contribute to some aspects of mental disorders.
Collapse
Affiliation(s)
- Z Pei
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - B Lang
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom.
| | - Y D Fragoso
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom; Department of Neurology, Medical Faculty, Universidade Metropolitana de Santos, Sao Paulo, Brazil
| | - K D Shearer
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - L Zhao
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - P J A Mccaffery
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - S Shen
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom; Regenerative Medicine Institute, School of Medicine, NUI Galway, Galway, Ireland
| | - Y Q Ding
- Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - C D McCaig
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - J M Collinson
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom.
| |
Collapse
|
49
|
Kimura H, Miki Y, Nakanishi A. Centrosomes at M phase act as a scaffold for the accumulation of intracellular ubiquitinated proteins. Cell Cycle 2014; 13:1928-37. [PMID: 24743317 DOI: 10.4161/cc.28896] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Centrosome size varies considerably during the cell cycle; it is greatest during metaphase, partly because of pericentriolar matrix recruitment and an increase in microtubule-organizing activity. However, the mechanism of centrosome maturation during M phase is poorly defined. In the present study, we identified and quantified centrosomal proteins during S and M phases using stable isotope labeling by amino acids in cell culture (SILAC) coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS). We identified 991 proteins, of which 310 and 325 proteins were upregulated during S and M phases, respectively. Ubiquitinated proteins containing K48- and K63-linked polyubiquitin chains accumulated in the centrosomes during M phase, although 26S proteasome activity in the centrosomes did not markedly differ between S and M phases. Conversely, cytoplasmic dynein, which transports ubiquitinated proteins to the centrosomes, increased 2-fold in the centrosomes during M phase relative to S phase. Furthermore, PYR-41, a ubiquitin E1 inhibitor, reduced centrosome size during metaphase, causing increased aneuploidy. RNA interference suppression of Ecm29, which inhibits proteasome activity, decreased the accumulation of ubiquitinated proteins in the centrosomes. These results show that accumulation of ubiquitinated proteins promotes centrosome maturation during M phase and further suggest a novel function of centrosomes as a scaffold temporarily gathering intracellular ubiquitinated proteins.
Collapse
Affiliation(s)
- Hitomi Kimura
- Department of Molecular Genetics; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Bunkyo-ku, Tokyo, Japan
| | - Yoshio Miki
- Department of Molecular Genetics; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Bunkyo-ku, Tokyo, Japan; Department of Genetic Diagnosis; The Cancer Institute; Japanese Foundation for Cancer Research; Koto-ku, Tokyo, Japan
| | - Akira Nakanishi
- Department of Molecular Genetics; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
50
|
Goldenson B, Crispino JD. The aurora kinases in cell cycle and leukemia. Oncogene 2014; 34:537-45. [PMID: 24632603 PMCID: PMC4167158 DOI: 10.1038/onc.2014.14] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/14/2014] [Accepted: 01/21/2014] [Indexed: 12/14/2022]
Abstract
The Aurora kinases, which include Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC), are serine/threonine kinases required for the control of mitosis (AURKA and AURKB) and meiosis (AURKC). Since their discovery nearly 20 years ago, Aurora kinases have been studied extensively in cell and cancer biology. Several early studies found that Aurora kinases are amplified and overexpressed at the transcript and protein level in various malignancies, including several types of leukemia. These discoveries and others provided a rationale for the development of small-molecule inhibitors of Aurora kinases as leukemia therapies. The first generation of Aurora kinase inhibitors did not fare well in clinical trials, owing to poor efficacy and high toxicity. However, the creation of second-generation, highly selective Aurora kinase inhibitors has increased the enthusiasm for targeting these proteins in leukemia. This review will describe the functions of each Aurora kinase, summarize their involvement in leukemia and discuss inhibitor development and efficacy in leukemia clinical trials.
Collapse
Affiliation(s)
- B Goldenson
- Division of Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | - J D Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, IL, USA
| |
Collapse
|