1
|
Nikhil K, Shah K. The significant others of aurora kinase a in cancer: combination is the key. Biomark Res 2024; 12:109. [PMID: 39334449 PMCID: PMC11438406 DOI: 10.1186/s40364-024-00651-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
AURKA is predominantly famous as an essential mitotic kinase. Recent findings have also established its critical role in a plethora of other biological processes including ciliogenesis, mitochondrial dynamics, neuronal outgrowth, DNA replication and cell cycle progression. AURKA overexpression in numerous cancers is strongly associated with poor prognosis and survival. Still no AURKA-targeted drug has been approved yet, partially because of the associated collateral toxicity and partly due to its limited efficacy as a single agent in a wide range of tumors. Mechanistically, AURKA overexpression allows it to phosphorylate numerous pathological substrates promoting highly aggressive oncogenic phenotypes. Our review examines the most recent advances in AURKA regulation and focuses on 33 such direct cancer-specific targets of AURKA and their associated oncogenic signaling cascades. One of the common themes that emerge is that AURKA is often involved in a feedback loop with its substrates, which could be the decisive factor causing its sustained upregulation and hyperactivation in cancer cells, an Achilles heel not exploited before. This dynamic interplay between AURKA and its substrates offers potential opportunities for targeted therapeutic interventions. By targeting these substrates, it may be possible to disrupt this feedback loop to effectively reverse AURKA levels, thereby providing a promising avenue for developing safer AURKA-targeted therapeutics. Additionally, exploring the synergistic effects of AURKA inhibition with its other oncogenic and/or tumor-suppressor targets could provide further opportunities for developing effective combination therapies against AURKA-driven cancers, thereby maximizing its potential as a critical drug target.
Collapse
Affiliation(s)
- Kumar Nikhil
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India.
| | - Kavita Shah
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
2
|
Afghah Z, Khan N, Datta G, Halcrow PW, Geiger JD, Chen X. Involvement of Endolysosomes and Aurora Kinase A in the Regulation of Amyloid β Protein Levels in Neurons. Int J Mol Sci 2024; 25:6200. [PMID: 38892390 PMCID: PMC11172969 DOI: 10.3390/ijms25116200] [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: 04/24/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Aurora kinase A (AURKA) is a serine/threonine-protein kinase that regulates microtubule organization during neuron migration and neurite formation. Decreased activity of AURKA was found in Alzheimer's disease (AD) brain samples, but little is known about the role of AURKA in AD pathogenesis. Here, we demonstrate that AURKA is expressed in primary cultured rat neurons, neurons from adult mouse brains, and neurons in postmortem human AD brains. AURKA phosphorylation, which positively correlates with its activity, is reduced in human AD brains. In SH-SY5Y cells, pharmacological activation of AURKA increased AURKA phosphorylation, acidified endolysosomes, decreased the activity of amyloid beta protein (Aβ) generating enzyme β-site amyloid precursor protein cleaving enzyme (BACE-1), increased the activity of the Aβ degrading enzyme cathepsin D, and decreased the intracellular and secreted levels of Aβ. Conversely, pharmacological inhibition of AURKA decreased AURKA phosphorylation, de-acidified endolysosomes, decreased the activity of cathepsin D, and increased intracellular and secreted levels of Aβ. Thus, reduced AURKA activity in AD may contribute to the development of intraneuronal accumulations of Aβ and extracellular amyloid plaque formation.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA; (Z.A.); (N.K.); (G.D.); (P.W.H.); (J.D.G.)
| |
Collapse
|
3
|
Lakkaniga NR, Wang Z, Xiao Y, Kharbanda A, Lan L, Li HY. Revisiting Aurora Kinase B: A promising therapeutic target for cancer therapy. Med Res Rev 2024; 44:686-706. [PMID: 37983866 DOI: 10.1002/med.21994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/28/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Cancer continues to be a major health concern globally, although the advent of targeted therapy has revolutionized treatment options. Aurora Kinase B is a serine-threonine kinase that has been explored as an oncology therapeutic target for more than two decades. Aurora Kinase B inhibitors show promising biological results in in-vitro and in-vivo experiments. However, there are no inhibitors approved yet for clinical use, primarily because of the side effects associated with Aurora B inhibitors. Several studies demonstrate that Aurora B inhibitors show excellent synergy with various chemotherapeutic agents, radiation therapy, and targeted therapies. This makes it an excellent choice as an adjuvant therapy to first-line therapies, which greatly improves the therapeutic window and side effect profile. Recent studies indicate the role of Aurora B in some deadly cancers with limited therapeutic options, like triple-negative breast cancer and glioblastoma. Herein, we review the latest developments in Aurora Kinase B targeted research, with emphasis on its potential as an adjuvant therapy and its role in some of the most difficult-to-treat cancers.
Collapse
Affiliation(s)
- Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | - Zhengyu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yao Xiao
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Anupreet Kharbanda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Li Lan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
4
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
5
|
Zhang XW, Li JY, Li L, Hu WQ, Tao Y, Gao WY, Ye ZN, Jia HY, Wang JN, Miao XK, Yang WL, Wang R, Mou LY. Neurokinin-1 receptor drives PKCɑ-AURKA/N-Myc signaling to facilitate the neuroendocrine progression of prostate cancer. Cell Death Dis 2023; 14:384. [PMID: 37385990 PMCID: PMC10310825 DOI: 10.1038/s41419-023-05894-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023]
Abstract
The widespread application of antiandrogen therapies has aroused a significant increase in the incidence of NEPC, a lethal form of the disease lacking efficient clinical treatments. Here we identified a cell surface receptor neurokinin-1 (NK1R) as a clinically relevant driver of treatment-related NEPC (tNEPC). NK1R expression increased in prostate cancer patients, particularly higher in metastatic prostate cancer and treatment-related NEPC, implying a relation with the progression from primary luminal adenocarcinoma toward NEPC. High NK1R level was clinically correlated with accelerated tumor recurrence and poor survival. Mechanical studies identified a regulatory element in the NK1R gene transcription ending region that was recognized by AR. AR inhibition enhanced the expression of NK1R, which mediated the PKCα-AURKA/N-Myc pathway in prostate cancer cells. Functional assays demonstrated that activation of NK1R promoted the NE transdifferentiation, cell proliferation, invasion, and enzalutamide resistance in prostate cancer cells. Targeting NK1R abrogated the NE transdifferentiation process and tumorigenicity in vitro and in vivo. These findings collectively characterized the role of NK1R in tNEPC progression and suggested NK1R as a potential therapeutic target.
Collapse
Affiliation(s)
- Xiao-Wei Zhang
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Jing-Yi Li
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Departemnt of Biochemistry and Molecular Biology, School of basic medical sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, P. R. China
| | - Lin Li
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Wen-Qian Hu
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Yan Tao
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Wen-Yan Gao
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Zi-Nuo Ye
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Hao-Yuan Jia
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Jia-Nan Wang
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Xiao-Kang Miao
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Wen-Le Yang
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China
| | - Rui Wang
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Ling-Yun Mou
- School of Life Science Lanzhou University, 222 TianShui South Road, Lanzhou, 730000, P. R. China.
- Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, P. R. China.
| |
Collapse
|
6
|
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
|
7
|
Zeppillo T, Schulmann A, Macciardi F, Hjelm BE, Föcking M, Sequeira PA, Guella I, Cotter D, Bunney WE, Limon A, Vawter MP. Functional impairment of cortical AMPA receptors in schizophrenia. Schizophr Res 2022; 249:25-37. [PMID: 32513544 PMCID: PMC7718399 DOI: 10.1016/j.schres.2020.03.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/14/2022]
Abstract
Clinical and preclinical studies suggest that some of the behavioral alterations observed in schizophrenia (SZ) may be mechanistically linked to synaptic dysfunction of glutamatergic signaling. Recent genetic and proteomic studies suggest alterations of cortical glutamate receptors of the AMPA-type (AMPARs), which are the predominant ligand-gated ionic channels of fast transmission at excitatory synapses. The impact of gene and protein alterations on the electrophysiological activity of AMPARs is not known in SZ. In this proof of principle work, using human postmortem brain synaptic membranes isolated from the dorsolateral prefrontal cortex (DLPFC), we combined electrophysiological analysis from microtransplanted synaptic membranes (MSM) with transcriptomic (RNA-Seq) and label-free proteomics data in 10 control and 10 subjects diagnosed with SZ. We observed in SZ a reduction in the amplitude of AMPARs currents elicited by kainate, an agonist of AMPARs that blocks the desensitization of the receptor. This reduction was not associated with protein abundance but with a reduction in kainate's potency to activate AMPARs. Electrophysiologically-anchored dataset analysis (EDA) was used to identify synaptosomal proteins that linearly correlate with the amplitude of the AMPARs responses, gene ontology functional annotations were then used to determine protein-protein interactions. Protein modules associated with positive AMPARs current increases were downregulated in SZ, while protein modules that were upregulated in SZ were associated with decreased AMPARs currents. Our results indicate that transcriptomic and proteomic alterations, frequently observed in the DLPFC in SZ, converge at the synaptic level producing a functional electrophysiological impairment of AMPARs.
Collapse
Affiliation(s)
- Tommaso Zeppillo
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch at Galveston, USA; Department of Life Sciences, University of Trieste, B.R.A.I.N., Centre for Neuroscience, Trieste, Italy
| | - Anton Schulmann
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA; Current address: National Institute of Mental Health, Human Genetics Branch, Bethesda, MD, USA
| | - Fabio Macciardi
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - Brooke E Hjelm
- Department of Translational Genomics, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, USA
| | | | - P Adolfo Sequeira
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - Ilaria Guella
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - David Cotter
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - William E Bunney
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - Agenor Limon
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch at Galveston, USA.
| | - Marquis P Vawter
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA.
| |
Collapse
|
8
|
Cacioppo R, Lindon C. Regulating the regulator: a survey of mechanisms from transcription to translation controlling expression of mammalian cell cycle kinase Aurora A. Open Biol 2022; 12:220134. [PMID: 36067794 PMCID: PMC9448500 DOI: 10.1098/rsob.220134] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/11/2022] [Indexed: 11/12/2022] Open
Abstract
Aurora Kinase A (AURKA) is a positive regulator of mitosis with a strict cell cycle-dependent expression pattern. Recently, novel oncogenic roles of AURKA have been uncovered that are independent of the kinase activity and act within multiple signalling pathways, including cell proliferation, survival and cancer stem cell phenotypes. For this, cellular abundance of AURKA protein is per se crucial and must be tightly fine-tuned. Indeed, AURKA is found overexpressed in different cancers, typically as a result of gene amplification or enhanced transcription. It has however become clear that impaired processing, decay and translation of AURKA mRNA can also offer the basis for altered AURKA levels. Accordingly, the involvement of gene expression mechanisms controlling AURKA expression in human diseases is increasingly recognized and calls for much more research. Here, we explore and create an integrated view of the molecular processes regulating AURKA expression at the level of transcription, post-transcription and translation, intercalating discussion on how impaired regulation underlies disease. Given that targeting AURKA levels might affect more functions compared to inhibiting the kinase activity, deeper understanding of its gene expression may aid the design of alternative and therapeutically more successful ways of suppressing the AURKA oncogene.
Collapse
Affiliation(s)
- Roberta Cacioppo
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
| | - Catherine Lindon
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
| |
Collapse
|
9
|
Zang J, Chen Y, Liu C, Lin S. Probing the Role of Aurora Kinase A Threonylation with Site-Specific Lysine Threonylation. ACS Chem Biol 2022; 18:674-678. [PMID: 35230082 DOI: 10.1021/acschembio.1c00682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protein post-translational modifications play central roles in regulating protein functions. Lysine threonylation is a newly discovered reversible post-translational modification. However, the biological effect of lysine threonylation on proteins remains largely elusive. Here we report a chemical biology approach for site-specific incorporation of Nε-threonyllysine into proteins with high efficiency and investigate the biological effect of lysine threonylation on Aurora kinase A. Using this unnatural amino acid mutagenesis approach, we find that threonylation of Lys162 of Aurora kinase A inhibits its kinase activity both in vitro and in vivo and that the inhibitory effect can be reversed by the deacetylase Sirtuin 3, which removes the threonylated group from the lysine. Additionally, threonylation of Aurora kinase A makes its substrate p53 more stable in the cell. Therefore, our study demonstrates that site-specific lysine threonylation is a powerful method for probing the biological effect of protein threonylation.
Collapse
Affiliation(s)
- Jia Zang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yulin Chen
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chao Liu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shixian Lin
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Bijian K, Wernic D, Nivedha AK, Su J, Lim FPL, Miron CE, Amzil H, Moitessier N, Alaoui-Jamali MA. Novel Aurora A and Protein Kinase C (α, β1, β2, and θ) Multitarget Inhibitors: Impact of Selenium Atoms on the Potency and Selectivity. J Med Chem 2022; 65:3134-3150. [PMID: 35167283 DOI: 10.1021/acs.jmedchem.1c01031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aurora kinases and protein kinase C (PKC) have been shown to be involved in different aspects of cancer progression. To date, no dual Aurora/PKC inhibitor with clinical efficacy and low toxicity is available. Here, we report the identification of compound 2e as a potent small molecule capable of selectively inhibiting Aurora A kinase and PKC isoforms α, β1, β2 and θ. Compound 2e demonstrated significant inhibition of the colony forming ability of metastatic breast cancer cells in vitro and metastasis development in vivo. In vitro kinase screening and molecular modeling studies revealed the critical role of the selenium-containing side chains within 2e, where selenium atoms were shown to significantly improve its selectivity and potency by forming additional interactions and modulating the protein dynamics. In comparison to other H-bonding heteroatoms such as sulfur, our studies suggested that these selenium atoms also confer more favorable PK properties.
Collapse
Affiliation(s)
- Krikor Bijian
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Dominik Wernic
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Anita K Nivedha
- Department of Chemistry, McGill University, Montréal, Québec H3A 0B8, Canada.,Molecular Forecaster, 7171 rue Frederick Banting, Saint Laurent, Quebec H4S 1Z9, Canada
| | - Jie Su
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, Quebec H3A 0B8, Canada
| | | | - Caitlin E Miron
- Department of Chemistry, McGill University, Montréal, Québec H3A 0B8, Canada
| | - Hind Amzil
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Nicolas Moitessier
- Department of Chemistry, McGill University, Montréal, Québec H3A 0B8, Canada
| | - Moulay A Alaoui-Jamali
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, Quebec H3A 0B8, Canada
| |
Collapse
|
11
|
Baltzer S, Bulatov T, Schmied C, Krämer A, Berger BT, Oder A, Walker-Gray R, Kuschke C, Zühlke K, Eichhorst J, Lehmann M, Knapp S, Weston J, von Kries JP, Süssmuth RD, Klussmann E. Aurora Kinase A Is Involved in Controlling the Localization of Aquaporin-2 in Renal Principal Cells. Int J Mol Sci 2022; 23:ijms23020763. [PMID: 35054947 PMCID: PMC8776063 DOI: 10.3390/ijms23020763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 02/01/2023] Open
Abstract
The cAMP-dependent aquaporin-2 (AQP2) redistribution from intracellular vesicles into the plasma membrane of renal collecting duct principal cells induces water reabsorption and fine-tunes body water homeostasis. However, the mechanisms controlling the localization of AQP2 are not understood in detail. Using immortalized mouse medullary collecting duct (MCD4) and primary rat inner medullary collecting duct (IMCD) cells as model systems, we here discovered a key regulatory role of Aurora kinase A (AURKA) in the control of AQP2. The AURKA-selective inhibitor Aurora-A inhibitor I and novel derivatives as well as a structurally different inhibitor, Alisertib, prevented the cAMP-induced redistribution of AQP2. Aurora-A inhibitor I led to a depolymerization of actin stress fibers, which serve as tracks for the translocation of AQP2-bearing vesicles to the plasma membrane. The phosphorylation of cofilin-1 (CFL1) inactivates the actin-depolymerizing function of CFL1. Aurora-A inhibitor I decreased the CFL1 phosphorylation, accounting for the removal of the actin stress fibers and the inhibition of the redistribution of AQP2. Surprisingly, Alisertib caused an increase in actin stress fibers and did not affect CFL1 phosphorylation, indicating that AURKA exerts its control over AQP2 through different mechanisms. An involvement of AURKA and CFL1 in the control of the localization of AQP2 was hitherto unknown.
Collapse
Affiliation(s)
- Sandrine Baltzer
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Timur Bulatov
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Christopher Schmied
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
- DKTK (German Translational Research Network), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| | - Benedict-Tilman Berger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
| | - Andreas Oder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Ryan Walker-Gray
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Christin Kuschke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Kerstin Zühlke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
- DKTK (German Translational Research Network), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt am Main, Germany
| | - John Weston
- JQuest Consulting, Carl-Orff-Weg 25, 65779 Kelkheim, Germany;
| | - Jens Peter von Kries
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Roderich D. Süssmuth
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence: ; Tel.: +49-30-9406-2596
| |
Collapse
|
12
|
Balmik AA, Chinnathambi S. Inter-relationship of Histone Deacetylase-6 with cytoskeletal organization and remodeling. Eur J Cell Biol 2022; 101:151202. [DOI: 10.1016/j.ejcb.2022.151202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
|
13
|
Akagawa R, Nabeshima YI, Kawauchi T. Alternative Functions of Cell Cycle-Related and DNA Repair Proteins in Post-mitotic Neurons. Front Cell Dev Biol 2021; 9:753175. [PMID: 34746147 PMCID: PMC8564117 DOI: 10.3389/fcell.2021.753175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Proper regulation of neuronal morphological changes is essential for neuronal migration, maturation, synapse formation, and high-order function. Many cytoplasmic proteins involved in the regulation of neuronal microtubules and the actin cytoskeleton have been identified. In addition, some nuclear proteins have alternative functions in neurons. While cell cycle-related proteins basically control the progression of the cell cycle in the nucleus, some of them have an extra-cell cycle-regulatory function (EXCERF), such as regulating cytoskeletal organization, after exit from the cell cycle. Our expression analyses showed that not only cell cycle regulators, including cyclin A1, cyclin D2, Cdk4/6, p21cip1, p27kip1, Ink4 family, and RAD21, but also DNA repair proteins, including BRCA2, p53, ATM, ATR, RAD17, MRE11, RAD9, and Hus1, were expressed after neurogenesis, suggesting that these proteins have alternative functions in post-mitotic neurons. In this perspective paper, we discuss the alternative functions of the nuclear proteins in neuronal development, focusing on possible cytoplasmic roles.
Collapse
Affiliation(s)
- Remi Akagawa
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| | - Yo-ichi Nabeshima
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| | - Takeshi Kawauchi
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Wakatsuki S, Araki T. Novel Molecular Basis for Synapse Formation: Small Non-coding Vault RNA Functions as a Riboregulator of MEK1 to Modulate Synaptogenesis. Front Mol Neurosci 2021; 14:748721. [PMID: 34630040 PMCID: PMC8498202 DOI: 10.3389/fnmol.2021.748721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Small non-coding vault RNAs (vtRNAs) have been described as a component of the vault complex, a hollow-and-barrel-shaped ribonucleoprotein complex found in most eukaryotes. It has been suggested that the function of vtRNAs might not be limited to simply maintaining the structure of the vault complex. Despite the increasing research on vtRNAs, little is known about their physiological functions. Recently, we have shown that murine vtRNA (mvtRNA) up-regulates synaptogenesis by activating the mitogen activated protein kinase (MAPK) signaling pathway. mvtRNA binds to and activates mitogen activated protein kinase 1 (MEK1), and thereby enhances MEK1-mediated extracellular signal-regulated kinase activation. Here, we introduce the regulatory mechanism of MAPK signaling in synaptogenesis by vtRNAs and discuss the possibility as a novel molecular basis for synapse formation.
Collapse
Affiliation(s)
- Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
15
|
Zhu L, Dai S, Lu D, Xu P, Chen L, Han Y, Zhong L, Chang L, Wu Q. Role of NDEL1 and VEGF/VEGFR-2 in Mouse Hippocampus After Status Epilepticus. ASN Neuro 2021; 12:1759091420926836. [PMID: 32423231 PMCID: PMC7238446 DOI: 10.1177/1759091420926836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Nuclear-distribution element-like 1 (NDEL1) is associated with the
proliferation and migration of neurons. Vascular endothelial growth
factor (VEGF) in combination with VEGF receptor-2 (VEGFR-2) regulates
the proliferation and migration of neurons. This study was performed
to explore undefined alterations in the expression levels of NDEL1 and
VEGF/VEGFR-2 within the hippocampus after status epilepticus (SE).
Following the creation of pilocarpine-induced epilepsy models using
adolescent male C57BL/6 mice, Western blotting and reverse
transcription quantitative polymerase chain reaction were applied to
assess the levels of NDEL1, VEGF, and VEGFR-2 expression in whole
hippocampi at 1, 2, 3, and 4 weeks post-SE, respectively.
Immunofluorescent labeling was also employed to detect the
colocalization of NDEL1 and VEGF in the hippocampus. Our results
indicated that NDEL1 and VEGF have similar patterns of upregulation
throughout the hippocampus. Upregulation of VEGFR-2 occurred only in
the early stages, and the expression decreased shortly afterward.
NDEL1 and VEGF were coexpressed in the cornu ammonis 3 pyramidal cell,
granular, and polymorph layers of the dentate gyrus in the
hippocampus. This study revealed that NDEL1, VEGF, and VEGFR-2 may
work together and are involved in the pathophysiology in the
hippocampus after SE.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Shujuan Dai
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Di Lu
- Biomedicine Engineering Research Centre, Kunming Medical University
| | - Puying Xu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Lu Chen
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Yanbing Han
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Lianmei Zhong
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Lvhua Chang
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University
| |
Collapse
|
16
|
Wakatsuki S, Takahashi Y, Shibata M, Adachi N, Numakawa T, Kunugi H, Araki T. Small noncoding vault RNA modulates synapse formation by amplifying MAPK signaling. J Cell Biol 2021; 220:211679. [PMID: 33439240 PMCID: PMC7809882 DOI: 10.1083/jcb.201911078] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 09/04/2020] [Accepted: 12/02/2020] [Indexed: 12/30/2022] Open
Abstract
The small noncoding vault RNA (vtRNA) is a component of the vault complex, a ribonucleoprotein complex found in most eukaryotes. Emerging evidence suggests that vtRNAs may be involved in the regulation of a variety of cellular functions when unassociated with the vault complex. Here, we demonstrate a novel role for vtRNA in synaptogenesis. Using an in vitro synapse formation model, we show that murine vtRNA (mvtRNA) promotes synapse formation by modulating the MAPK signaling pathway. mvtRNA is transported to the distal region of neurites as part of the vault complex. Interestingly, mvtRNA is released from the vault complex in the neurite by a mitotic kinase Aurora-A–dependent phosphorylation of MVP, a major protein component of the vault complex. mvtRNA binds to and activates MEK1 and thereby enhances MEK1-mediated ERK activation in neurites. These results suggest the existence of a regulatory mechanism of the MAPK signaling pathway by vtRNAs as a new molecular basis for synapse formation.
Collapse
Affiliation(s)
- Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoko Takahashi
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Megumi Shibata
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Naoki Adachi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo, Japan
| | - Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
17
|
Blazejewski SM, Bennison SA, Liu X, Toyo-Oka K. High-throughput kinase inhibitor screening reveals roles for Aurora and Nuak kinases in neurite initiation and dendritic branching. Sci Rep 2021; 11:8156. [PMID: 33854138 PMCID: PMC8047044 DOI: 10.1038/s41598-021-87521-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/31/2021] [Indexed: 12/25/2022] Open
Abstract
Kinases are essential regulators of a variety of cellular signaling processes, including neurite formation—a foundational step in neurodevelopment. Aberrant axonal sprouting and failed regeneration of injured axons are associated with conditions like traumatic injury, neurodegenerative disease, and seizures. Investigating the mechanisms underlying neurite formation will allow for identification of potential therapeutics. We used a kinase inhibitor library to screen 493 kinase inhibitors and observed that 45% impacted neuritogenesis in Neuro2a (N-2a) cells. Based on the screening, we further investigated the roles of Aurora kinases A, B, and C and Nuak kinases 1 and 2. The roles of Aurora and Nuak kinases have not been thoroughly studied in the nervous system. Inhibition or overexpression of Aurora and Nuak kinases in primary cortical neurons resulted in various neuromorphological defects, with Aurora A regulating neurite initiation, Aurora B and C regulating neurite initiation and elongation, all Aurora kinases regulating arborization, and all Nuak kinases regulating neurite initiation and elongation and arborization. Our high-throughput screening and analysis of Aurora and Nuak kinases revealed their functions and may contribute to the identification of therapeutics.
Collapse
Affiliation(s)
- Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
18
|
Sun N, Meng F, Zhao J, Li X, Li R, Han J, Chen X, Cheng W, Yang X, Kou Y, Zheng K, Yang J, Ikezoe T. Aurka deficiency in the intestinal epithelium promotes age-induced obesity via propionate-mediated AKT activation. Int J Biol Sci 2021; 17:1302-1314. [PMID: 33867847 PMCID: PMC8040479 DOI: 10.7150/ijbs.56477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
Aurora-A kinase, a serine/threonine mitotic kinase involved in mitosis, is overexpressed in several human cancers. A recent study showed that Aurora-A mediates glucose metabolism via SOX8/FOXK1 in ovarian cancer. However, the roles of Aurora-A in metabolic diseases remain unclear. This study found that Aurka loss in the intestinal epithelium promoted age-induced obesity and enlargement of lipid droplets in parallel with an increase in infiltrated macrophages in the white adipocyte tissue (WAT) of male mice. Moreover, loss of Aurka induced the expression of lipid metabolism regulatory genes, including acetyl-coenzyme A carboxylase 1 (Acc1), in association with an increase in the levels of p-AKT in the intestinal epithelium as well as WAT. Blockade of AKT activation reduced the expression of lipid metabolism regulatory genes. In subsequent experiments, we found that the Firmicutes abundance and the levels of short-chain fatty acids (SCFAs) in the gut were dramatically increased in Aurkaf/+;VillinCre/+ mice compared with Aurkaf/+ mice. Additionally, propionate increased the phosphorylation of AKT in vitro. These observations indicated that Aurka loss in the intestinal epithelium contributed to gut microbiota dysbiosis and higher levels of SCFAs, especially propionate, leading to AKT activation and lipid metabolism regulatory gene expression, which in turn promoted age-induced obesity.
Collapse
Affiliation(s)
- Na Sun
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Fandong Meng
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University
| | - Jie Zhao
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Xueqin Li
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Rongqing Li
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Jing Han
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Xin Chen
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Wanpeng Cheng
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Xiaoying Yang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Yanbo Kou
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University
| | - Kuiyang Zheng
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University.,National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Yang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Affiliated Hospital of Xuzhou Medical University.,Department of Pathogenic Biology and Immunology, Affiliated Hospital of Xuzhou Medical University.,National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Takayuki Ikezoe
- The Department of Hematology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
19
|
Kaarijärvi R, Kaljunen H, Ketola K. Molecular and Functional Links between Neurodevelopmental Processes and Treatment-Induced Neuroendocrine Plasticity in Prostate Cancer Progression. Cancers (Basel) 2021; 13:cancers13040692. [PMID: 33572108 PMCID: PMC7915380 DOI: 10.3390/cancers13040692] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Treatment-induced neuroendocrine prostate cancer (t-NEPC) is a subtype of castration-resistant prostate cancer (CRPC) which develops under prolonged androgen deprivation therapy. The mechanisms and pathways underlying the t-NEPC are still poorly understood and there are no effective treatments available. Here, we summarize the literature on the molecules and pathways contributing to neuroendocrine phenotype in prostate cancer in the context of their known cellular neurodevelopmental processes. We also discuss the role of tumor microenvironment in neuroendocrine plasticity, future directions, and therapeutic options under clinical investigation for neuroendocrine prostate cancer. Abstract Neuroendocrine plasticity and treatment-induced neuroendocrine phenotypes have recently been proposed as important resistance mechanisms underlying prostate cancer progression. Treatment-induced neuroendocrine prostate cancer (t-NEPC) is highly aggressive subtype of castration-resistant prostate cancer which develops for one fifth of patients under prolonged androgen deprivation. In recent years, understanding of molecular features and phenotypic changes in neuroendocrine plasticity has been grown. However, there are still fundamental questions to be answered in this emerging research field, for example, why and how do the prostate cancer treatment-resistant cells acquire neuron-like phenotype. The advantages of the phenotypic change and the role of tumor microenvironment in controlling cellular plasticity and in the emergence of treatment-resistant aggressive forms of prostate cancer is mostly unknown. Here, we discuss the molecular and functional links between neurodevelopmental processes and treatment-induced neuroendocrine plasticity in prostate cancer progression and treatment resistance. We provide an overview of the emergence of neurite-like cells in neuroendocrine prostate cancer cells and whether the reported t-NEPC pathways and proteins relate to neurodevelopmental processes like neurogenesis and axonogenesis during the development of treatment resistance. We also discuss emerging novel therapeutic targets modulating neuroendocrine plasticity.
Collapse
|
20
|
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: 304] [Impact Index Per Article: 76.0] [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
|
21
|
Wilkes OR, Moore AW. Distinct Microtubule Organizing Center Mechanisms Combine to Generate Neuron Polarity and Arbor Complexity. Front Cell Neurosci 2020; 14:594199. [PMID: 33328893 PMCID: PMC7711044 DOI: 10.3389/fncel.2020.594199] [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: 08/12/2020] [Accepted: 11/02/2020] [Indexed: 01/15/2023] Open
Abstract
Dendrite and axon arbor wiring patterns determine the connectivity and computational characteristics of a neuron. The identities of these dendrite and axon arbors are created by differential polarization of their microtubule arrays, and their complexity and pattern are generated by the extension and organization of these arrays. We describe how several molecularly distinct microtubule organizing center (MTOC) mechanisms function during neuron differentiation to generate and arrange dendrite and axon microtubules. The temporal and spatial organization of these MTOCs generates, patterns, and diversifies arbor wiring.
Collapse
Affiliation(s)
- Oliver R Wilkes
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, Japan.,Department of Cellular and Molecular Biology, Institute for Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Adrian W Moore
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, Japan
| |
Collapse
|
22
|
Aurora kinases and DNA damage response. Mutat Res 2020; 821:111716. [PMID: 32738522 DOI: 10.1016/j.mrfmmm.2020.111716] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/21/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
It is well established that Aurora kinases perform critical functions during mitosis. It has become increasingly clear that the Aurora kinases also perform a myriad of non-mitotic functions including DNA damage response. The available evidence indicates that inhibition Aurora kinase A (AURKA) may contribute to the G2 DNA damage checkpoint through AURKA's functions in PLK1 and CDC25B activation. Both AURKA and Aurora kinase B (AURKB) are also essential in mitotic DNA damage response that guard against DNA damage-induced chromosome segregation errors, including the control of abscission checkpoint and prevention of micronuclei formation. Dysregulation of Aurora kinases can trigger DNA damage in mitosis that is sensed in the subsequent G1 by a p53-dependent postmitotic checkpoint. Aurora kinases are themselves linked to the G1 DNA damage checkpoint through p53 and p73 pathways. Finally, several lines of evidence provide a connection between Aurora kinases and DNA repair and apoptotic pathways. Although more studies are required to provide a comprehensive picture of how cells respond to DNA damage, these findings indicate that both AURKA and AURKB are inextricably linked to pathways guarding against DNA damage. They also provide a rationale to support more detailed studies on the synergism between small-molecule inhibitors against Aurora kinases and DNA-damaging agents in cancer therapies.
Collapse
|
23
|
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
|
24
|
Tibbo AJ, Baillie GS. Phosphodiesterase 4B: Master Regulator of Brain Signaling. Cells 2020; 9:cells9051254. [PMID: 32438615 PMCID: PMC7291338 DOI: 10.3390/cells9051254] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Phosphodiesterases (PDEs) are the only superfamily of enzymes that have the ability to break down cyclic nucleotides and, as such, they have a pivotal role in neurological disease and brain development. PDEs have a modular structure that allows targeting of individual isoforms to discrete brain locations and it is often the location of a PDE that shapes its cellular function. Many of the eleven different families of PDEs have been associated with specific diseases. However, we evaluate the evidence, which suggests the activity from a sub-family of the PDE4 family, namely PDE4B, underpins a range of important functions in the brain that positions the PDE4B enzymes as a therapeutic target for a diverse collection of indications, such as, schizophrenia, neuroinflammation, and cognitive function.
Collapse
|
25
|
Kimata Y, Leturcq M, Aradhya R. Emerging roles of metazoan cell cycle regulators as coordinators of the cell cycle and differentiation. FEBS Lett 2020; 594:2061-2083. [PMID: 32383482 DOI: 10.1002/1873-3468.13805] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023]
Abstract
In multicellular organisms, cell proliferation must be tightly coordinated with other developmental processes to form functional tissues and organs. Despite significant advances in our understanding of how the cell cycle is controlled by conserved cell-cycle regulators (CCRs), how the cell cycle is coordinated with cell differentiation in metazoan organisms and how CCRs contribute to this process remain poorly understood. Here, we review the emerging roles of metazoan CCRs as intracellular proliferation-differentiation coordinators in multicellular organisms. We illustrate how major CCRs regulate cellular events that are required for cell fate acquisition and subsequent differentiation. To this end, CCRs employ diverse mechanisms, some of which are separable from those underpinning the conventional cell-cycle-regulatory functions of CCRs. By controlling cell-type-specific specification/differentiation processes alongside the progression of the cell cycle, CCRs enable spatiotemporal coupling between differentiation and cell proliferation in various developmental contexts in vivo. We discuss the significance and implications of this underappreciated role of metazoan CCRs for development, disease and evolution.
Collapse
Affiliation(s)
- Yuu Kimata
- School of Life Science and Technology, ShanghaiTech University, China
| | - Maïté Leturcq
- School of Life Science and Technology, ShanghaiTech University, China
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| |
Collapse
|
26
|
Bennison SA, Blazejewski SM, Smith TH, Toyo-Oka K. Protein kinases: master regulators of neuritogenesis and therapeutic targets for axon regeneration. Cell Mol Life Sci 2020; 77:1511-1530. [PMID: 31659414 PMCID: PMC7166181 DOI: 10.1007/s00018-019-03336-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/16/2019] [Accepted: 10/08/2019] [Indexed: 12/25/2022]
Abstract
Proper neurite formation is essential for appropriate neuronal morphology to develop and defects at this early foundational stage have serious implications for overall neuronal function. Neuritogenesis is tightly regulated by various signaling mechanisms that control the timing and placement of neurite initiation, as well as the various processes necessary for neurite elongation to occur. Kinases are integral components of these regulatory pathways that control the activation and inactivation of their targets. This review provides a comprehensive summary of the kinases that are notably involved in regulating neurite formation, which is a complex process that involves cytoskeletal rearrangements, addition of plasma membrane to increase neuronal surface area, coupling of cytoskeleton/plasma membrane, metabolic regulation, and regulation of neuronal differentiation. Since kinases are key regulators of these functions during neuromorphogenesis, they have high potential for use as therapeutic targets for axon regeneration after injury or disease where neurite formation is disrupted.
Collapse
Affiliation(s)
- Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
27
|
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: 53] [Impact Index Per Article: 10.6] [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
|
28
|
Bertolin G, Tramier M. Insights into the non-mitotic functions of Aurora kinase A: more than just cell division. Cell Mol Life Sci 2020; 77:1031-1047. [PMID: 31562563 PMCID: PMC11104877 DOI: 10.1007/s00018-019-03310-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 02/02/2023]
Abstract
AURKA is a serine/threonine kinase overexpressed in several cancers. Originally identified as a protein with multifaceted roles during mitosis, improvements in quantitative microscopy uncovered several non-mitotic roles as well. In physiological conditions, AURKA regulates cilia disassembly, neurite extension, cell motility, DNA replication and senescence programs. In cancer-like contexts, AURKA actively promotes DNA repair, it acts as a transcription factor, promotes cell migration and invasion, and it localises at mitochondria to regulate mitochondrial dynamics and ATP production. Here we review the non-mitotic roles of AURKA, and its partners outside of cell division. In addition, we give an insight into how structural data and quantitative fluorescence microscopy allowed to understand AURKA activation and its interaction with new substrates, highlighting future developments in fluorescence microscopy needed to better understand AURKA functions in vivo. Last, we will recapitulate the most significant AURKA inhibitors currently in clinical trials, and we will explore how the non-mitotic roles of the kinase may provide new insights to ameliorate current pharmacological strategies against AURKA overexpression.
Collapse
Affiliation(s)
- Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000, Rennes, France.
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000, Rennes, France.
| |
Collapse
|
29
|
Chau DDL, Lau KF. The roles of atypical protein kinase Cs (aPKCs) in the nervous system: targets for neuroregeneration? Neural Regen Res 2020; 15:1475-1476. [PMID: 31997808 PMCID: PMC7059589 DOI: 10.4103/1673-5374.274334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Dennis Dik-Long Chau
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kwok-Fai Lau
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
30
|
Structural basis for the design of allosteric inhibitors of the Aurora kinase A enzyme in the cancer chemotherapy. Biochim Biophys Acta Gen Subj 2020; 1864:129448. [DOI: 10.1016/j.bbagen.2019.129448] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022]
|
31
|
Bertolin G, Sizaire F, Déméautis C, Chapuis C, Mérola F, Erard M, Tramier M. Optimized FRET Pairs and Quantification Approaches To Detect the Activation of Aurora Kinase A at Mitosis. ACS Sens 2019; 4:2018-2027. [PMID: 31317736 DOI: 10.1021/acssensors.9b00486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Genetically encoded Förster's Resonance Energy Transfer (FRET) biosensors are indispensable tools to sense the spatiotemporal dynamics of signal transduction pathways. Investigating the crosstalk between different signaling pathways is becoming increasingly important to follow cell development and fate programs. To this end, FRET biosensors must be optimized to monitor multiple biochemical activities simultaneously and in single cells. In addition, their sensitivity must be increased to follow their activation even when the abundance of the biosensor is low. We describe here the development of a second generation of Aurora kinase A/AURKA biosensors. First, we adapt the original AURKA biosensor-GFP-AURKA-mCherry-to multiplex FRET by using dark acceptors as ShadowG or ShadowY. Then, we use the novel superYFP acceptor protein to measure FRET by 2-color Fluorescence Cross-Correlation Spectroscopy, in cytosolic regions where the abundance of AURKA is extremely low and undetectable with the original AURKA biosensor. These results pave the way to the use of FRET biosensors to follow AURKA activation in conjunction with substrate-based activity biosensors. In addition, they open up the possibility of tracking the activation of small pools of AURKA and its interaction with novel substrates, which would otherwise remain undetectable with classical biochemical approaches.
Collapse
Affiliation(s)
- Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000 Rennes, France
| | - Florian Sizaire
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000 Rennes, France
| | - Claire Déméautis
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000 Rennes, France
| | - Catherine Chapuis
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000 Rennes, France
| | - Fabienne Mérola
- Univ Paris Sud, CNRS, LCP (Laboratoire de Chimie Physique), UMR 800, F-91400 Orsay, France
| | - Marie Erard
- Univ Paris Sud, CNRS, LCP (Laboratoire de Chimie Physique), UMR 800, F-91400 Orsay, France
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000 Rennes, France
- Univ Rennes, BIOSIT, UMS CNRS 3480, US INSERM
018, F-35000 Rennes, France
| |
Collapse
|
32
|
Choi BKA, D’Onofrio PM, Shabanzadeh AP, Koeberle PD. Stabilization of primary cilia reduces abortive cell cycle re-entry to protect injured adult CNS neurons from apoptosis. PLoS One 2019; 14:e0220056. [PMID: 31369591 PMCID: PMC6675095 DOI: 10.1371/journal.pone.0220056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/08/2019] [Indexed: 01/09/2023] Open
Abstract
Abortive cell cycle (ACC) re-entry of apoptotic neurons is a recently characterized phenomenon that occurs after central nervous system (CNS) injury or over the course of CNS disease. Consequently, inhibiting cell cycle progression is neuroprotective in numerous CNS pathology models. Primary cilia are ubiquitous, centriole-based cellular organelles that prevent cell cycling, but their ability to modulate abortive cell cycle has not been described. Here, we show that neuronal cilia are ablated in-vitro and in-vivo following injury by hypoxia or optic nerve transection (ONT), respectively. Furthermore, forced cilia resorption sensitized neurons to these injuries and enhanced cell death. In contrast, pharmacological inhibition or shRNA knockdown of the proteins that disassemble the cilia increased neuron survival and decreased the phosphorylation of retinoblastoma (Rb), a master switch for cell cycle re-entry. Our findings show that the stabilization of neuronal primary cilia inhibits, at least transiently, apoptotic cell cycling, which has implications for future therapeutic strategies that halt or slow the progression of neurodegenerative diseases and acute CNS injuries.
Collapse
Affiliation(s)
- Brian K. A. Choi
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada
- Graduate Department of Rehabilitation Sciences, University of Toronto, Toronto, ON, Canada
| | - Philippe M. D’Onofrio
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada
- Graduate Department of Rehabilitation Sciences, University of Toronto, Toronto, ON, Canada
| | - Alireza P. Shabanzadeh
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University of Toronto, Toronto, ON, Canada
| | - Paulo D. Koeberle
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON, Canada
- Graduate Department of Rehabilitation Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Woo JAA, Liu T, Fang CC, Cazzaro S, Kee T, LePochat P, Yrigoin K, Penn C, Zhao X, Wang X, Liggett SB, Kang DE. Activated cofilin exacerbates tau pathology by impairing tau-mediated microtubule dynamics. Commun Biol 2019; 2:112. [PMID: 30911686 PMCID: PMC6430779 DOI: 10.1038/s42003-019-0359-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. While the accumulation of Aβ is pivotal to the etiology of AD, both the microtubule-associated protein tau (MAPT) and the F-actin severing protein cofilin are necessary for the deleterious effects of Aβ. However, the molecular link between tau and cofilin remains unclear. In this study, we found that cofilin competes with tau for direct microtubule binding in vitro, in cells, and in vivo, which inhibits tau-induced microtubule assembly. Genetic reduction of cofilin mitigates tauopathy and synaptic defects in Tau-P301S mice and movement deficits in tau transgenic C. elegans. The pathogenic effects of cofilin are selectively mediated by activated cofilin, as active but not inactive cofilin selectively interacts with tubulin, destabilizes microtubules, and promotes tauopathy. These results therefore indicate that activated cofilin plays an essential intermediary role in neurotoxic signaling that promotes tauopathy.
Collapse
Affiliation(s)
- Jung-A. A. Woo
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Tian Liu
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Cenxiao C. Fang
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Sara Cazzaro
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Teresa Kee
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Patrick LePochat
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Ksenia Yrigoin
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Courtney Penn
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Xingyu Zhao
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Xinming Wang
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Stephen B. Liggett
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - David E. Kang
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- James A. Haley Veteran’s Administration Hospital, Tampa, FL 33612 USA
| |
Collapse
|
34
|
Zhao P, Teng X, Tantirimudalige SN, Nishikawa M, Wohland T, Toyama Y, Motegi F. Aurora-A Breaks Symmetry in Contractile Actomyosin Networks Independently of Its Role in Centrosome Maturation. Dev Cell 2019; 48:631-645.e6. [DOI: 10.1016/j.devcel.2019.02.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/21/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022]
|
35
|
Kelliher MT, Saunders HA, Wildonger J. Microtubule control of functional architecture in neurons. Curr Opin Neurobiol 2019; 57:39-45. [PMID: 30738328 DOI: 10.1016/j.conb.2019.01.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 01/20/2023]
Abstract
Neurons are exquisitely polarized cells whose structure and function relies on microtubules. Microtubules in signal-receiving dendrites and signal-sending axons differ in their organization and microtubule-associated proteins. These differences, coupled with microtubule post-translational modifications, combine to locally regulate intracellular transport, morphology, and function. Recent discoveries provide new insight into the regulation of non-centrosomal microtubule arrays in neurons, the relationship between microtubule acetylation and mechanosensation, and the spatial patterning of microtubules that regulates motor activity and cargo delivery in axons and dendrites. Together, these new studies bring us closer to understanding how microtubule function is locally tuned to match the specialized tasks associated with signal reception and transmission.
Collapse
Affiliation(s)
- Michael T Kelliher
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Harriet Aj Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jill Wildonger
- Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
36
|
Abstract
Neurons are polarized cells with long branched axons and dendrites. Microtubule generation and organization machineries are crucial to grow and pattern these complex cellular extensions. Microtubule organizing centers (MTOCs) concentrate the molecular machinery for templating microtubules, stabilizing the nascent polymer, and organizing the resultant microtubules into higher-order structures. MTOC formation and function are well described at the centrosome, in the spindle, and at interphase Golgi; we review these studies and then describe recent results about how the machineries acting at these classic MTOCs are repurposed in the postmitotic neuron for axon and dendrite differentiation. We further discuss a constant tug-of-war interplay between different MTOC activities in the cell and how this process can be used as a substrate for transcription factor-mediated diversification of neuron types.
Collapse
Affiliation(s)
- Jason Y Tann
- Laboratory for Neurodiversity, RIKEN Centre for Brain Science, Saitama, Japan
| | - Adrian W Moore
- Laboratory for Neurodiversity, RIKEN Centre for Brain Science, Saitama, Japan.
| |
Collapse
|
37
|
Zhang W, Xing L, Xu L, Jin X, Du Y, Feng X, Liu S, Liu Q. Nudel involvement in the high-glucose-induced epithelial-mesenchymal transition of tubular epithelial cells. Am J Physiol Renal Physiol 2018; 316:F186-F194. [PMID: 30539652 DOI: 10.1152/ajprenal.00218.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nudel is a newly discovered factor related to cell migration. The tubular epithelial-mesenchymal transition (EMT) includes four steps: the loss of the adhesive properties of epithelial cells, the acquisition of a mesenchymal cell phenotype, the destruction of the tubular basal membrane, and the migration into the renal interstitium. The purpose of this study was to investigate the role of Nudel in the high-glucose-induced EMT of tubular epithelial cells. Human renal proximal tubular epithelial cells (HKCs) were treated with Nudel shRNA to clarify the role and mechanism of Nudel in tubular EMT induced by high glucose. We found that Nudel was expressed at a high level in high-glucose-stimulated HKCs, and the expression of Nudel was associated with the activation of signal transducer and activator of transcription 3. After transfection with Nudel shRNA, we detected the expression levels of E-cadherin, α-smooth muscle actin (α-SMA), and the Wiskott-Aldrich syndrome family of proteins (including WASP, N-WASP, WAVE1, WAVE2, and WAVE3) via assay. Cell migration was analyzed by the scratching method. The results showed that high glucose downregulated E-cadherin expression, upregulated α-SMA expression, and promoted the migration of HKCs. The expression levels of N-WASP, WAVE1, and WAVE2 were also elevated in HKCs treated with high glucose. All changes induced by high glucose were ameliorated by Nudel depletion. We conclude that Nudel participates in the transition and the migration of tubular epithelial cells via the regulation of WASP family proteins.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| | - Lingling Xing
- Department of Nephrology, The Second Affiliated Hospital of Hebei Medical University , Shijiazhuang, Hebei , China
| | - Lu Xu
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| | - Xiaoxue Jin
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| | - Yunxia Du
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| | - Xiaojuan Feng
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| | - Shuxia Liu
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| | - Qingjuan Liu
- Department of Pathology, Hebei Medical University, Key Laboratory of Kidney Diseases of Hebei Province , Shijiazhuang, Hebei , China
| |
Collapse
|
38
|
Gwee SSL, Radford RAW, Chow S, Syal MD, Morsch M, Formella I, Lee A, Don EK, Badrock AP, Cole NJ, West AK, Cheung SNS, Chung RS. Aurora kinase B regulates axonal outgrowth and regeneration in the spinal motor neurons of developing zebrafish. Cell Mol Life Sci 2018; 75:4269-4285. [PMID: 29468257 PMCID: PMC11105541 DOI: 10.1007/s00018-018-2780-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/10/2018] [Accepted: 02/15/2018] [Indexed: 01/23/2023]
Abstract
Aurora kinase B (AurkB) is a serine/threonine protein kinase with a well-characterised role in orchestrating cell division and cytokinesis, and is prominently expressed in healthy proliferating and cancerous cells. However, the role of AurkB in differentiated and non-dividing cells has not been extensively explored. Previously, we have described a significant upregulation of AurkB expression in cultured cortical neurons following an experimental axonal transection. This is somewhat surprising, as AurkB expression is generally associated only with dividing cells Frangini et al. (Mol Cell 51:647-661, 2013); Hegarat et al. (J Cell Biol 195:1103-1113, 2011); Lu et al. (J Biol Chem 283:31785-31790, 2008); Trakala et al. (Cell Cycle 12:1030-1041, 2014). Herein, we present the first description of a role for AurkB in terminally differentiated neurons. AurkB was prominently expressed within post-mitotic neurons of the zebrafish brain and spinal cord. The expression of AurkB varied during the development of the zebrafish spinal motor neurons. Utilising pharmacological and genetic manipulation to impair AurkB activity resulted in truncation and aberrant motor axon morphology, while overexpression of AurkB resulted in extended axonal outgrowth. Further pharmacological inhibition of AurkB activity in regenerating axons delayed their recovery following UV laser-mediated injury. Collectively, these results suggest a hitherto unreported role of AurkB in regulating neuronal development and axonal outgrowth.
Collapse
Affiliation(s)
- Serene S L Gwee
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia.
| | - Rowan A W Radford
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Sharron Chow
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Monisha D Syal
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Marco Morsch
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Isabel Formella
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Albert Lee
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Emily K Don
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Andrew P Badrock
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Nicholas J Cole
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia
| | - Adrian K West
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Steve N S Cheung
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
- School of Life and Environmental Sciences, Deakin University, Burwood, VIC, Australia
| | - Roger S Chung
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, NSW, Australia.
- School of Medicine, University of Tasmania, Hobart, TAS, Australia.
| |
Collapse
|
39
|
Willems E, Dedobbeleer M, Digregorio M, Lombard A, Lumapat PN, Rogister B. The functional diversity of Aurora kinases: a comprehensive review. Cell Div 2018; 13:7. [PMID: 30250494 PMCID: PMC6146527 DOI: 10.1186/s13008-018-0040-6] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
Aurora kinases are serine/threonine kinases essential for the onset and progression of mitosis. Aurora members share a similar protein structure and kinase activity, but exhibit distinct cellular and subcellular localization. AurA favors the G2/M transition by promoting centrosome maturation and mitotic spindle assembly. AurB and AurC are chromosome-passenger complex proteins, crucial for chromosome binding to kinetochores and segregation of chromosomes. Cellular distribution of AurB is ubiquitous, while AurC expression is mainly restricted to meiotically-active germ cells. In human tumors, all Aurora kinase members play oncogenic roles related to their mitotic activity and promote cancer cell survival and proliferation. Furthermore, AurA plays tumor-promoting roles unrelated to mitosis, including tumor stemness, epithelial-to-mesenchymal transition and invasion. In this review, we aim to understand the functional interplay of Aurora kinases in various types of human cells, including tumor cells. The understanding of the functional diversity of Aurora kinases could help to evaluate their relevance as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
- Estelle Willems
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium
| | - Matthias Dedobbeleer
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium
| | - Marina Digregorio
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium
| | - Arnaud Lombard
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium.,2Department of Neurosurgery, CHU of Liège, Liège, Belgium
| | - Paul Noel Lumapat
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium.,3Department of Neurology, CHU of Liège, Liège, Belgium
| | - Bernard Rogister
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium.,3Department of Neurology, CHU of Liège, Liège, Belgium
| |
Collapse
|
40
|
Bertolin G, Bulteau AL, Alves-Guerra MC, Burel A, Lavault MT, Gavard O, Le Bras S, Gagné JP, Poirier GG, Le Borgne R, Prigent C, Tramier M. Aurora kinase A localises to mitochondria to control organelle dynamics and energy production. eLife 2018; 7:38111. [PMID: 30070631 PMCID: PMC6140714 DOI: 10.7554/elife.38111] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022] Open
Abstract
Many epithelial cancers show cell cycle dysfunction tightly correlated with the overexpression of the serine/threonine kinase Aurora A (AURKA). Its role in mitotic progression has been extensively characterised, and evidence for new AURKA functions emerges. Here, we reveal that AURKA is located and imported in mitochondria in several human cancer cell lines. Mitochondrial AURKA impacts on two organelle functions: mitochondrial dynamics and energy production. When AURKA is expressed at endogenous levels during interphase, it induces mitochondrial fragmentation independently from RALA. Conversely, AURKA enhances mitochondrial fusion and ATP production when it is over-expressed. We demonstrate that AURKA directly regulates mitochondrial functions and that AURKA over-expression promotes metabolic reprogramming by increasing mitochondrial interconnectivity. Our work paves the way to anti-cancer therapeutics based on the simultaneous targeting of mitochondrial functions and AURKA inhibition. Structures called mitochondria power cells by turning oxygen and sugar into chemical energy. Each cell can have thousands of mitochondria, which work together to supply changing energy demands. They can fuse together or break apart, forming networks that change size and produce different amounts of energy. Getting the balance right is crucial; if energy levels are too low, the cell will not be able to grow and divide. If energy levels are too high, the cell can grow at a faster rate, which can contribute to the cell becoming cancerous. Although we know that mitochondria provide energy, it is not clear how they communicate to fine-tune the supply. Some clues come from cancer cells that seem dependent on their mitochondria for survival. In these cells, levels of a protein called AURKA are higher than normal. AURKA helps cells to divide, and it interacts with many different proteins. This complexity makes it difficult to work out exactly what AURKA does, but it is possible that it plays a role in energy supply. Bertolin et al. have now investigated whether mitochondria use AURKA to communicate inside human breast cancer cells. Tagging AURKA proteins with a fluorescent marker revealed that it accumulates inside mitochondria. Once it gets there, AURKA changes the shape of the mitochondria, which has dramatic effects on their capacity to produce energy. At normal levels, AURKA causes the mitochondria to fragment, breaking apart into smaller pieces. This maintains their energy output at a normal level. If AURKA levels are too high, the mitochondria fuse together and produce more energy. This means AURKA could help to fuel fast-growing cancer cells. Current drugs that aim to treat cancer by blocking the activity of AURKA show poor results. This is partly due to the fact that the protein has so many different roles in the cell. Finding that AURKA affects mitochondria is the first step in understanding one of its unknown roles. It also suggests the possibility of developing new drugs to change how mitochondria make energy in cancer cells that contain high levels of AURKA.
Collapse
Affiliation(s)
- Giulia Bertolin
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France
| | - Anne-Laure Bulteau
- ENS de Lyon, Lyon, France.,CNRS UMR 5242, Lyon, France.,INRA USC 1370, Lyon, France
| | - Marie-Clotilde Alves-Guerra
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Agnes Burel
- Microscopy Rennes Imaging Centre, SFR Biosit, UMS CNRS 3480- US INSERM 018, Université de Rennes, Rennes, France
| | - Marie-Thérèse Lavault
- Microscopy Rennes Imaging Centre, SFR Biosit, UMS CNRS 3480- US INSERM 018, Université de Rennes, Rennes, France
| | - Olivia Gavard
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Equipes labélisées Ligue Contre Le Cancer, Rennes, France.,Centre de recherche du CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Stephanie Le Bras
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France
| | - Jean-Philippe Gagné
- Centre de recherche du CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Guy G Poirier
- Centre de recherche du CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Roland Le Borgne
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Equipes labélisées Ligue Contre Le Cancer, Rennes, France
| | - Claude Prigent
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Equipes labélisées Ligue Contre Le Cancer, Rennes, France
| | - Marc Tramier
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Microscopy Rennes Imaging Centre, SFR Biosit, UMS CNRS 3480- US INSERM 018, Université de Rennes, Rennes, France
| |
Collapse
|
41
|
Hapak SM, Rothlin CV, Ghosh S. PAR3-PAR6-atypical PKC polarity complex proteins in neuronal polarization. Cell Mol Life Sci 2018; 75:2735-2761. [PMID: 29696344 PMCID: PMC11105418 DOI: 10.1007/s00018-018-2828-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/03/2018] [Accepted: 04/23/2018] [Indexed: 01/01/2023]
Abstract
Polarity is a fundamental feature of cells. Protein complexes, including the PAR3-PAR6-aPKC complex, have conserved roles in establishing polarity across a number of eukaryotic cell types. In neurons, polarity is evident as distinct axonal versus dendritic domains. The PAR3, PAR6, and aPKC proteins also play important roles in neuronal polarization. During this process, either aPKC kinase activity, the assembly of the PAR3-PAR6-aPKC complex or the localization of these proteins is regulated downstream of a number of signaling pathways. In turn, the PAR3, PAR6, and aPKC proteins control various effector molecules to establish neuronal polarity. Herein, we discuss the many signaling mechanisms and effector functions that have been linked to PAR3, PAR6, and aPKC during the establishment of neuronal polarity.
Collapse
Affiliation(s)
- Sophie M Hapak
- Department of Medicine, School of Medicine, University of Minnesota, 401 East River Parkway, Minneapolis, MN, 55455, USA.
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, 300 Cedar Street, New Haven, CT, 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, 300 George Street, New Haven, CT, 06511, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
42
|
NEK7 regulates dendrite morphogenesis in neurons via Eg5-dependent microtubule stabilization. Nat Commun 2018; 9:2330. [PMID: 29899413 PMCID: PMC5997995 DOI: 10.1038/s41467-018-04706-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/15/2018] [Indexed: 01/22/2023] Open
Abstract
Organization of microtubules into ordered arrays is best understood in mitotic systems, but remains poorly characterized in postmitotic cells such as neurons. By analyzing the cycling cell microtubule cytoskeleton proteome through expression profiling and targeted RNAi screening for candidates with roles in neurons, we have identified the mitotic kinase NEK7. We show that NEK7 regulates dendrite morphogenesis in vitro and in vivo. NEK7 kinase activity is required for dendrite growth and branching, as well as spine formation and morphology. NEK7 regulates these processes in part through phosphorylation of the kinesin Eg5/KIF11, promoting its accumulation on microtubules in distal dendrites. Here, Eg5 limits retrograde microtubule polymerization, which is inhibitory to dendrite growth and branching. Eg5 exerts this effect through microtubule stabilization, independent of its motor activity. This work establishes NEK7 as a general regulator of the microtubule cytoskeleton, controlling essential processes in both mitotic cells and postmitotic neurons. NEK7 is a kinase known for its role in mitotic spindle assembly, driving centrosome separation in prophase through regulation of the kinesin Eg5. Here, the authors show that NEK7 and Eg5 also control dendrite morphogenesis in postmitotic neurons.
Collapse
|
43
|
Genetics and mechanisms leading to human cortical malformations. Semin Cell Dev Biol 2018; 76:33-75. [DOI: 10.1016/j.semcdb.2017.09.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
|
44
|
Potential Role of Microtubule Stabilizing Agents in Neurodevelopmental Disorders. Int J Mol Sci 2017; 18:ijms18081627. [PMID: 28933765 PMCID: PMC5578018 DOI: 10.3390/ijms18081627] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/10/2017] [Accepted: 07/18/2017] [Indexed: 01/05/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) are characterized by neuroanatomical abnormalities indicative of corticogenesis disturbances. At the basis of NDDs cortical abnormalities, the principal developmental processes involved are cellular proliferation, migration and differentiation. NDDs are also considered “synaptic disorders” since accumulating evidence suggests that NDDs are developmental brain misconnection syndromes characterized by altered connectivity in local circuits and between brain regions. Microtubules and microtubule-associated proteins play a fundamental role in the regulation of basic neurodevelopmental processes, such as neuronal polarization and migration, neuronal branching and synaptogenesis. Here, the role of microtubule dynamics will be elucidated in regulating several neurodevelopmental steps. Furthermore, the correlation between abnormalities in microtubule dynamics and some NDDs will be described. Finally, we will discuss the potential use of microtubule stabilizing agents as a new pharmacological intervention for NDDs treatment.
Collapse
|
45
|
Vo TTL, Park JH, Seo JH, Lee EJ, Choi H, Bae SJ, Le H, An S, Lee HS, Wee HJ, Kim KW. ARD1-mediated aurora kinase A acetylation promotes cell proliferation and migration. Oncotarget 2017; 8:57216-57230. [PMID: 28915666 PMCID: PMC5593637 DOI: 10.18632/oncotarget.19332] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/30/2017] [Indexed: 11/25/2022] Open
Abstract
Aurora kinase A (AuA) is a prerequisite for centrosome maturation, separation, and mitotic spindle assembly, thus, it is essential for cell cycle regulation. Overexpression of AuA is implicated in poor prognosis of many types of cancer. However, the regulatory mechanisms underlying the functions of AuA are still not fully understood. Here, we report that AuA colocalizes with arrest defective protein 1 (ARD1) acetyltransferase during cell division and cell migration. Additionally, AuA is acetylated by ARD1 at lysine residues at positions 75 and 125. The double mutations at K75/K125 abolished the kinase activity of AuA. Moreover, the double mutant AuA exhibited diminished ability to promote cell proliferation and cell migration. Mechanistic studies revealed that AuA acetylation at K75/K125 promoted cell proliferation via activation of cyclin E/CDK2 and cyclin B1. In addition, AuA acetylation stimulated cell migration by activating the p38/AKT/MMP-2 pathway. Our findings indicate that ARD1-mediated acetylation of AuA enhances cell proliferation and migration, and probably contributes to cancer development.
Collapse
Affiliation(s)
- Tam Thuy Lu Vo
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Ji-Hyeon Park
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Ji Hae Seo
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu 42601, Korea
| | - Eun Ji Lee
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hoon Choi
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Sung-Jin Bae
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hoang Le
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Sunho An
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hye Shin Lee
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Hee-Jun Wee
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Kyu-Won Kim
- SNU-Harvard NeuroVascular Protection Research Center, College of Pharmacy and The Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.,Crop Biotechnology Institute, GreenBio Science and Technology, Seoul National University, Pyeongchang 25354, Korea
| |
Collapse
|
46
|
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
|
47
|
Ran-dependent TPX2 activation promotes acentrosomal microtubule nucleation in neurons. Sci Rep 2017; 7:42297. [PMID: 28205572 PMCID: PMC5304320 DOI: 10.1038/srep42297] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/05/2017] [Indexed: 01/07/2023] Open
Abstract
The microtubule (MT) cytoskeleton is essential for the formation of morphologically appropriate neurons. The existence of the acentrosomal MT organizing center in neurons has been proposed but its identity remained elusive. Here we provide evidence showing that TPX2 is an important component of this acentrosomal MT organizing center. First, neurite elongation is compromised in TPX2-depleted neurons. In addition, TPX2 localizes to the centrosome and along the neurite shaft bound to MTs. Depleting TPX2 decreases MT formation frequency specifically at the tip and the base of the neurite, and these correlate precisely with the regions where active GTP-bound Ran proteins are enriched. Furthermore, overexpressing the downstream effector of Ran, importin, compromises MT formation and neuronal morphogenesis. Finally, applying a Ran-importin signaling interfering compound phenocopies the effect of TPX2 depletion on MT dynamics. Together, these data suggest a model in which Ran-dependent TPX2 activation promotes acentrosomal MT nucleation in neurons.
Collapse
|
48
|
Blas-Rus N, Bustos-Morán E, Martín-Cófreces NB, Sánchez-Madrid F. Aurora-A shines on T cell activation through the regulation of Lck. Bioessays 2016; 39. [PMID: 27910998 DOI: 10.1002/bies.201600156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Different protein kinases control signaling emanating from the T cell receptor (TCR) during antigen-specific T cell activation. Mitotic kinases, e.g. Aurora-A, have been widely studied in the context of mitosis due to their role during microtubule (MT) nucleation, becoming critical regulators of cell cycle progression. We have recently described a specific role for Aurora-A kinase in antigenic T cell activation. Blockade of Aurora-A in T cells severely disrupts the dynamics of MTs and CD3ζ-bearing signaling vesicles during T cell activation. Furthermore, Aurora-A deletion impairs the activation of signaling molecules downstream of the TCR. Targeting Aurora-A disturbs the activation of Lck, which is one of the first signals that drive T cell activation in an antigen-dependent manner. This work describes possible models of regulation of Lck by Aurora-A during T cell activation. We also discuss possible roles for Aurora-A in other systems similar to the IS, and its putative functions in cell polarization.
Collapse
Affiliation(s)
- Noelia Blas-Rus
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain
| | - Eugenio Bustos-Morán
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Noa B Martín-Cófreces
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain.,Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
49
|
Cicenas J. The Aurora kinase inhibitors in cancer research and therapy. J Cancer Res Clin Oncol 2016; 142:1995-2012. [PMID: 26932147 DOI: 10.1007/s00432-016-2136-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/18/2016] [Indexed: 12/20/2022]
Abstract
Compounds that affect enzymatic function of kinases are valuable for the understanding of the complex biochemical processes in cells. Aurora kinases (AURKs) play a key role in the control of the mitosis. These kinases are frequently deregulated in different human cancers: overexpression, amplifications, translocations and deletions were reported in many cancer cell lines as well as patient tissues. These findings steered a rigorous hunt for small-molecule AURK inhibitors not only for research purposes as well as for therapeutic uses. In this review, we describe a number of AURK inhibitors and their use in cancer research and/or therapy. We hope to assist researchers and clinicians in deciding which inhibitor is most appropriate for their specific purpose. The review will also provide a broad overview of the clinical studies performed with some of these inhibitors (if such studies have been performed).
Collapse
Affiliation(s)
- Jonas Cicenas
- CALIPHO Group, Swiss Institute of Bioinformatics, CMU-1, rue Michel Servet, 1211, Geneva 4, Switzerland.
- MAP Kinase Resource, Melchiorstrasse 9, 3027, Bern, Switzerland.
- Proteomics Centre, Vilnius University Institute of Biochemistry, 08662, Vilnius, Lithuania.
| |
Collapse
|
50
|
Willems E, Lombard A, Dedobbeleer M, Goffart N, Rogister B. The Unexpected Roles of Aurora A Kinase in Gliobastoma Recurrences. Target Oncol 2016; 12:11-18. [DOI: 10.1007/s11523-016-0457-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|