51
|
Vertii A, Hehnly H, Doxsey S. The Centrosome, a Multitalented Renaissance Organelle. Cold Spring Harb Perspect Biol 2016; 8:8/12/a025049. [PMID: 27908937 DOI: 10.1101/cshperspect.a025049] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The centrosome acts as a microtubule-organizing center (MTOC) from the G1 to G2 phases of the cell cycle; it can mature into a spindle pole during mitosis and/or transition into a cilium by elongating microtubules (MTs) from the basal body on cell differentiation or cell cycle arrest. New studies hint that the centrosome functions in more than MT organization. For instance, it has recently been shown that a specific substructure of the centrosome-the mother centriole appendages-are required for the recycling of endosomes back to the plasma membrane. This alone could have important implications for a renaissance in our understanding of the development of primary cilia, endosome recycling, and the immune response. Here, we review newly identified roles for the centrosome in directing membrane traffic, the immunological synapse, and the stress response.
Collapse
Affiliation(s)
- Anastassiia Vertii
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Heidi Hehnly
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Stephen Doxsey
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
52
|
Sakai S, Izumi H, Yoshiura Y, Nakayama Y, Yamaguchi T, Harada Y, Koi C, Kurata H, Morimoto Y. In vitro evaluation of a combination treatment involving anticancer agents and an aurora kinase B inhibitor. Oncol Lett 2016; 12:4263-4269. [PMID: 27895801 PMCID: PMC5104265 DOI: 10.3892/ol.2016.5156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 08/25/2016] [Indexed: 12/25/2022] Open
Abstract
Aurora kinase B (AURKB) inhibitors are regarded as potential molecular-targeting drugs for cancer therapy. The present study evaluated the cytotoxic effect of a combination of AZD1152-hQPA, an AURKB inhibitor, and various anticancer agents on the HeLa human cervical cancer cell line, as well as its cisplatin-resistant equivalent HCP4 cell line. It was demonstrated that AZD1152-hQPA had an antagonistic effect on the cytotoxicity of cisplatin, etoposide and doxorubicin, but had a synergistic effect on that of all-trans-retinoic acid (ATRA), Am80 and TAC-101, when tested on HeLa cells. Cisplatin, etoposide and doxorubicin were shown to increase the cellular expression of AURKB, while ATRA, Am80 and TAC-101 downregulated its expression. These results suggested that AURKB expression is regulated by these anticancer agents at the transcriptional level, and that the level of expression of AURKB may influence the cytotoxic effect of AZD1152-hQPA. Therefore, when using anticancer agents, decreasing the expression of AURKB using a molecular-targeting drug may be an optimal therapeutic strategy.
Collapse
Affiliation(s)
- Senna Sakai
- Department of Occupational Pneumology, Institute of Industrial Ecological Science, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
- Department of Bioscience and Bioinformatics, Biomedical Informatics R&D Center, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Hiroto Izumi
- Department of Occupational Pneumology, Institute of Industrial Ecological Science, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Yukiko Yoshiura
- Department of Occupational Pneumology, Institute of Industrial Ecological Science, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Yoshifumi Nakayama
- Department of Gastroenterological and General Surgery, Wakamatsu Hospital, University of Occupational and Environmental Health School of Medicine, Kitakyushu 808-0024, Japan
| | - Takahiro Yamaguchi
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Yoshikazu Harada
- Department of Dentistry and Oral Surgery, University Hospital, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Chiho Koi
- Department of Obstetrics and Gynecology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Biomedical Informatics R&D Center, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Yasuo Morimoto
- Department of Occupational Pneumology, Institute of Industrial Ecological Science, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| |
Collapse
|
53
|
Barisic M, Maiato H. Dynein prevents erroneous kinetochore-microtubule attachments in mitosis. Cell Cycle 2016; 14:3356-61. [PMID: 26397382 DOI: 10.1080/15384101.2015.1089369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Equal distribution of the genetic material during cell division relies on efficient congression of chromosomes to the metaphase plate. Prior to their alignment, the Dynein motor recruited to kinetochores transports a fraction of laterally-attached chromosomes along microtubules toward the spindle poles. By doing that, Dynein not only contributes to chromosome movements, but also prevents premature stabilization of end-on kinetochore-microtubule attachments. This is achieved by 2 parallel mechanisms: 1) Dynein-mediated poleward movement of chromosomes counteracts opposite polar-ejection forces (PEFs) on chromosome arms by the microtubule plus-end-directed motors chromokinesins. Otherwise, they could stabilize erroneous syntelic kinetochore-microtubule attachments and lead to the random ejection of chromosomes away from the spindle poles; and 2) By transporting chromosomes to the spindle poles, Dynein brings the former to the zone of highest Aurora A kinase activity, further destabilizing kinetochore-microtubule attachments. Thus, Dynein plays an important role in keeping chromosome segregation error-free by preventing premature stabilization of kinetochore-microtubule attachments near the spindle poles.
Collapse
Affiliation(s)
- Marin Barisic
- a Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular; Universidade do Porto ; Porto , Portugal.,b Instituto de Investigação e Inovação em Saúde - i3S; Universidade do Porto ; Portugal
| | - Helder Maiato
- a Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular; Universidade do Porto ; Porto , Portugal.,b Instituto de Investigação e Inovação em Saúde - i3S; Universidade do Porto ; Portugal.,c Cell Division Unit ; Department of Experimental Biology; Faculdade de Medicina; Universidade do Porto ; Porto , Portugal
| |
Collapse
|
54
|
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]
|
55
|
Ye AA, Torabi J, Maresca TJ. Aurora A Kinase Amplifies a Midzone Phosphorylation Gradient to Promote High-Fidelity Cytokinesis. THE BIOLOGICAL BULLETIN 2016; 231:61-72. [PMID: 27638695 PMCID: PMC5360107 DOI: 10.1086/689591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
During cytokinesis, aurora B kinase (ABK) relocalizes from centromeres to the spindle midzone, where it is thought to provide a spatial cue for cytokinesis. While global ABK inhibition in Drosophila S2 cells results in macro- and multi-nucleated large cells, mislocalization of midzone ABK (mABK) by depletion of Subito (Drosophila MKLP2) does not cause notable cytokinesis defects. Subito depletion was, therefore, used to investigate the contribution of other molecules and redundant pathways to cytokinesis in the absence of mABK. Inhibiting potential polar relaxation pathways via removal of centrosomes (CNN RNAi) or a kinetochore-based phosphatase gradient (Sds22 RNAi) did not result in cytokinesis defects on their own or in combination with loss of mABK. Disruption of aurora A kinase (AAK) activity resulted in midzone assembly defects, but did not significantly affect contractile ring positioning or cytokinesis. Live-cell imaging of a Förster resonance energy transfer (FRET)-based aurora kinase phosphorylation sensor revealed that midzone substrates were less phosphorylated in AAK-inhibited cells, despite the fact that midzone levels of active phosphorylated ABK (pABK) were normal. Interestingly, in the absence of mABK, an increased number of binucleated cells were observed following AAK inhibition. The data suggest that equatorial stimulation rather than polar relaxation mechanisms is the major determinant of contractile ring positioning and high-fidelity cytokinesis in Drosophila S2 cells. Furthermore, we propose that equatorial stimulation is mediated primarily by the delivery of factors to the cortex by noncentrosomal microtubules (MTs), as well as a midzone-derived phosphorylation gradient that is amplified by the concerted activities of mABK and a soluble pool of AAK.
Collapse
Affiliation(s)
- Anna A Ye
- Biology Department, and Molecular and Cellular Biology Graduate Group, University of Massachusetts, Amherst, Massachusetts, 01003
| | | | - Thomas J Maresca
- Biology Department, and Molecular and Cellular Biology Graduate Group, University of Massachusetts, Amherst, Massachusetts, 01003
| |
Collapse
|
56
|
van Heesbeen RGHP, Raaijmakers JA, Tanenbaum ME, Halim VA, Lelieveld D, Lieftink C, Heck AJR, Egan DA, Medema RH. Aurora A, MCAK, and Kif18b promote Eg5-independent spindle formation. Chromosoma 2016; 126:473-486. [PMID: 27354041 PMCID: PMC5509784 DOI: 10.1007/s00412-016-0607-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 06/19/2016] [Accepted: 06/21/2016] [Indexed: 11/28/2022]
Abstract
Inhibition of the microtubule (MT) motor protein Eg5 results in a mitotic arrest due to the formation of monopolar spindles, making Eg5 an attractive target for anti-cancer therapies. However, Eg5-independent pathways for bipolar spindle formation exist, which might promote resistance to treatment with Eg5 inhibitors. To identify essential components for Eg5-independent bipolar spindle formation, we performed a genome-wide siRNA screen in Eg5-independent cells (EICs). We find that the kinase Aurora A and two kinesins, MCAK and Kif18b, are essential for bipolar spindle assembly in EICs and in cells with reduced Eg5 activity. Aurora A promotes bipolar spindle assembly by phosphorylating Kif15, hereby promoting Kif15 localization to the spindle. In turn, MCAK and Kif18b promote bipolar spindle assembly by destabilizing the astral MTs. One attractive way to interpret our data is that, in the absence of MCAK and Kif18b, excessive astral MTs generate inward pushing forces on centrosomes at the cortex that inhibit centrosome separation. Together, these data suggest a novel function for astral MTs in force generation on spindle poles and how proteins involved in regulating microtubule length can contribute to bipolar spindle assembly.
Collapse
Affiliation(s)
| | - Jonne A Raaijmakers
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marvin E Tanenbaum
- Hubrecht Institute, The Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincentius A Halim
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daphne Lelieveld
- Cell Screening Core, Department of Cell Biology, Center for Molecular Medicine, University Medical Centre, Utrecht, The Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - David A Egan
- Cell Screening Core, Department of Cell Biology, Center for Molecular Medicine, University Medical Centre, Utrecht, The Netherlands
| | - René H Medema
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
57
|
Mross K, Richly H, Frost A, Scharr D, Nokay B, Graeser R, Lee C, Hilbert J, Goeldner RG, Fietz O, Scheulen ME. A phase I study of BI 811283, an Aurora B kinase inhibitor, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2016; 78:405-17. [PMID: 27349901 PMCID: PMC5080318 DOI: 10.1007/s00280-016-3095-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/15/2016] [Indexed: 12/15/2022]
Abstract
PURPOSE This phase I study investigated the maximum tolerated dose (MTD), safety, pharmacokinetics, pharmacodynamics, and antitumor activity of the Aurora B kinase inhibitor BI 811283 in patients with advanced solid tumors. METHODS BI 811283 was administered via 24-h infusion on Days 1 and 15 of a 4-week cycle (schedule A) or Day 1 of a 3-week cycle (schedule B) in a modified 3 + 3 dose-escalation design. Pharmacodynamic assessments included immunohistochemistry for phosphorylated histone H3 (pHH3) on skin biopsies to determine Aurora B kinase inhibition and plasma concentrations of caspase-cleaved CK-18 (apoptosis marker). RESULTS A total of 121 patients were treated. The MTDs of BI 811283 were 125 mg (schedule A) and 230 mg (schedule B). Dose-limiting toxicities were primarily hematological (febrile neutropenia and grade 4 neutropenia); the most common drug-related adverse effects included neutropenia, fatigue, leukopenia, nausea, alopecia, diarrhea, and decreased appetite. A trend toward a decrease in pHH3 was observed, with increasing BI 811283 doses, indicating target engagement; there was no consistent trend regarding caspase-cleaved CK-18 levels. No objective response was observed although 19 patients in each schedule achieved clinical benefit (stable disease). CONCLUSIONS BI 811283 demonstrated a generally manageable safety profile and disease stabilization in some patients. TRIAL REGISTRATION EudraCT No: 2007-000191-17, ClinicalTrials.gov Identifier: NCT00701324.
Collapse
Affiliation(s)
- Klaus Mross
- Department of Medical Oncology, Tumour Biology Center, Breisacherstrasse 117, 79106, Freiburg, Germany. .,, Waldhofstrasse 50, 19117, Freiburg, Germany.
| | - Heike Richly
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Annette Frost
- Department of Medical Oncology, Tumour Biology Center, Breisacherstrasse 117, 79106, Freiburg, Germany.,Department of Hematology and Oncology, University Hospital, Breisacherstr. 117, 79106, Freiburg, Germany
| | - Dirk Scharr
- Department of Medical Oncology, Tumour Biology Center, Breisacherstrasse 117, 79106, Freiburg, Germany
| | - Bahar Nokay
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Ralph Graeser
- ProQinase GmbH, Breisacherstrasse 117, 79106, Freiburg, Germany.,Boehringer Ingelheim Pharma GmbH & Co. KG., Birkendorfer Strasse 65, 88397, Biberach an der Riss, Germany
| | - Chooi Lee
- Boehringer Ingelheim Ltd., Ellesfield Avenue, Bracknell, Berkshire, RG12 8YS, UK
| | - James Hilbert
- Boehringer Ingelheim Pharmaceuticals, Inc, 900 Ridgebury Road, Ridgefield, CT, 06877, USA.,Applied Biomath LLC, Wincester, MA, USA
| | - Rainer-George Goeldner
- Boehringer Ingelheim Pharma GmbH & Co. KG., Birkendorfer Strasse 65, 88397, Biberach an der Riss, Germany
| | - Oliver Fietz
- Boehringer Ingelheim Pharma GmbH & Co. KG., Birkendorfer Strasse 65, 88397, Biberach an der Riss, Germany
| | - Max E Scheulen
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
58
|
Kotak S, Afshar K, Busso C, Gönczy P. Aurora A kinase regulates proper spindle positioning in C. elegans and in human cells. J Cell Sci 2016; 129:3015-25. [PMID: 27335426 DOI: 10.1242/jcs.184416] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 06/16/2016] [Indexed: 01/04/2023] Open
Abstract
Accurate spindle positioning is essential for error-free cell division. The one-cell Caenorhabditis elegans embryo has proven instrumental for dissecting mechanisms governing spindle positioning. Despite important progress, how the cortical forces that act on astral microtubules to properly position the spindle are modulated is incompletely understood. Here, we report that the PP6 phosphatase PPH-6 and its associated subunit SAPS-1, which positively regulate pulling forces acting on spindle poles, associate with the Aurora A kinase AIR-1 in C. elegans embryos. We show that acute inactivation of AIR-1 during mitosis results in excess pulling forces on astral microtubules. Furthermore, we uncover that AIR-1 acts downstream of PPH-6-SAPS-1 in modulating spindle positioning, and that PPH-6-SAPS-1 negatively regulates AIR-1 localization at the cell cortex. Moreover, we show that Aurora A and the PP6 phosphatase subunit PPP6C are also necessary for spindle positioning in human cells. There, Aurora A is needed for the cortical localization of NuMA and dynein during mitosis. Overall, our work demonstrates that Aurora A kinases and PP6 phosphatases have an ancient function in modulating spindle positioning, thus contributing to faithful cell division.
Collapse
Affiliation(s)
- Sachin Kotak
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne CH-1015, Switzerland
| | - Katayon Afshar
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne CH-1015, Switzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne CH-1015, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne CH-1015, Switzerland
| |
Collapse
|
59
|
Leishmania donovani Aurora kinase: A promising therapeutic target against visceral leishmaniasis. Biochim Biophys Acta Gen Subj 2016; 1860:1973-88. [PMID: 27288586 DOI: 10.1016/j.bbagen.2016.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 06/02/2016] [Accepted: 06/06/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Aurora kinases are key mitotic kinases executing multiple aspects of eukaryotic cell-division. The apicomplexan homologs being essential for survival, suggest that the Leishmania homolog, annotated LdAIRK, may be equally important. METHODS Bioinformatics, stage-specific immunofluorescence microscopy, immunoblotting, RT-PCR, molecular docking, in-vitro kinase assay, anti-leishmanial activity assays, flow cytometry, fluorescence microscopy. RESULTS Ldairk expression is seen to vary as the cell-cycle progresses from G1 through S and finally G2M and cytokinesis. Kinetic studies demonstrate their enzymatic activity exhibiting a Km and Vmax of 6.12μM and 82.9pmoles·min(-1)mg(-1) respectively against ATP using recombinant Leishmania donovani H3, its physiological substrate. Due to the failure of LdAIRK-/+ knock-out parasites to survive, we adopted a chemical knock-down approach. Based on the conservation of key active site residues, three mammalian Aurora kinase inhibitors were investigated to evaluate their potential as inhibitors of LdAIRK activity. Interestingly, the cell-cycle progressed unhindered, despite treatment with GSK-1070916 or Barasertib, inhibitors with greater potencies for the ATP-binding pocket compared to Hesperadin, which at nanomolar concentrations, severely compromised viability at IC50s 105.9 and 36.4nM for promastigotes and amastigotes, respectively. Cell-cycle and morphological studies implicated their role in both mitosis and cytokinesis. CONCLUSION We identified an Aurora kinase homolog in L. donovani implicated in cell-cycle progression, whose inhibition led to aberrant changes in cell-cycle progression and reduced viability. GENERAL SIGNIFICANCE Human homologs being actively pursued drug targets and the observations with LdAIRK in both promastigotes and amastigotes suggest their potential as therapeutic-targets. Importantly, our results encourage the exploration of other proteins identified herein as potential novel drug targets.
Collapse
|
60
|
Sarvagalla S, Hsieh HP, Coumar MS. Therapeutic polymeric nanoparticles and the methods of making and using thereof: a patent evaluation of WO2015036792. Expert Opin Ther Pat 2016; 26:751-5. [PMID: 27167102 DOI: 10.1080/13543776.2016.1188919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Evaluation of the patent application WO2015036792 claiming therapeutic polymeric nanoparticles loaded with AZD1152-hqpa (aurora kinase inhibitor), and methods of making and using same for the treatment of cancer, is described. The claimed polymeric nano-formulations containing hydrophobic acid significantly improved the pharmacokinetic profiles (slow/sustained drug release profile) of the drug AZD1152-hqpa, as compared to the control agent (AZD1152). Drug efficacy and tolerability were also improved, and toxicity decreased in in vivo animal experiments, resulting in a better therapeutic index for the nano-formulation. Hence, the nano-formulated AZD1152-hqpa could be tested in the clinic at a dose level similar to, or higher than, that used for AZD1152, with lower incidence of toxicity.
Collapse
Affiliation(s)
- Sailu Sarvagalla
- a Centre for Bioinformatics, School of Life Sciences , Pondicherry University , Kalapet , Puducherry , India
| | - Hsing Pang Hsieh
- b Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Zhunan , Taiwan , ROC
| | - Mohane Selvaraj Coumar
- a Centre for Bioinformatics, School of Life Sciences , Pondicherry University , Kalapet , Puducherry , India
| |
Collapse
|
61
|
Rao SR, Flores-Rodriguez N, Page SL, Wong C, Robinson PJ, Chircop M. The Clathrin-dependent Spindle Proteome. Mol Cell Proteomics 2016; 15:2537-53. [PMID: 27174698 DOI: 10.1074/mcp.m115.054809] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Indexed: 01/07/2023] Open
Abstract
The mitotic spindle is required for chromosome congression and subsequent equal segregation of sister chromatids. These processes involve a complex network of signaling molecules located at the spindle. The endocytic protein, clathrin, has a "moonlighting" role during mitosis, whereby it stabilizes the mitotic spindle. The signaling pathways that clathrin participates in to achieve mitotic spindle stability are unknown. Here, we assessed the mitotic spindle proteome and phosphoproteome in clathrin-depleted cells using quantitative MS/MS (data are available via ProteomeXchange with identifier PXD001603). We report a spindle proteome that consists of 3046 proteins and a spindle phosphoproteome consisting of 5157 phosphosites in 1641 phosphoproteins. Of these, 2908 (95.4%) proteins and 1636 (99.7%) phosphoproteins are known or predicted spindle-associated proteins. Clathrin-depletion from spindles resulted in dysregulation of 121 proteins and perturbed signaling to 47 phosphosites. The majority of these proteins increased in mitotic spindle abundance and six of these were validated by immunofluorescence microscopy. Functional pathway analysis confirmed the reported role of clathrin in mitotic spindle stabilization for chromosome alignment and highlighted possible new mechanisms of clathrin action. The data also revealed a novel second mitotic role for clathrin in bipolar spindle formation.
Collapse
Affiliation(s)
- Sushma R Rao
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Neftali Flores-Rodriguez
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Scott L Page
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Chin Wong
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Phillip J Robinson
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| | - Megan Chircop
- From the ‡Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, Australia
| |
Collapse
|
62
|
Blas-Rus N, Bustos-Morán E, Pérez de Castro I, de Cárcer G, Borroto A, Camafeita E, Jorge I, Vázquez J, Alarcón B, Malumbres M, Martín-Cófreces NB, Sánchez-Madrid F. Aurora A drives early signalling and vesicle dynamics during T-cell activation. Nat Commun 2016; 7:11389. [PMID: 27091106 PMCID: PMC4838898 DOI: 10.1038/ncomms11389] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 03/21/2016] [Indexed: 01/09/2023] Open
Abstract
Aurora A is a serine/threonine kinase that contributes to the progression of mitosis by inducing microtubule nucleation. Here we have identified an unexpected role for Aurora A kinase in antigen-driven T-cell activation. We find that Aurora A is phosphorylated at the immunological synapse (IS) during TCR-driven cell contact. Inhibition of Aurora A with pharmacological agents or genetic deletion in human or mouse T cells severely disrupts the dynamics of microtubules and CD3ζ-bearing vesicles at the IS. The absence of Aurora A activity also impairs the activation of early signalling molecules downstream of the TCR and the expression of IL-2, CD25 and CD69. Aurora A inhibition causes delocalized clustering of Lck at the IS and decreases phosphorylation levels of tyrosine kinase Lck, thus indicating Aurora A is required for maintaining Lck active. These findings implicate Aurora A in the propagation of the TCR activation signal. Aurora A is a protein kinase that contributes to the progression of mitosis by stimulating microtubule nucleation. Here the authors show that Aurora A also functions during T cell activation by maintaining TCR signaling through Lck activation.
Collapse
Affiliation(s)
- Noelia Blas-Rus
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, C/ Diego de León 62, Madrid 28006, Spain
| | - Eugenio Bustos-Morán
- Cell-cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Ignacio Pérez de Castro
- Cell Division and Cancer Group, Centro Nacional de Investigaciones Oncológicas (CNIO), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Guillermo de Cárcer
- Cell Division and Cancer Group, Centro Nacional de Investigaciones Oncológicas (CNIO), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Aldo Borroto
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/ Nicolás cabrera 1, Madrid 28049, Spain
| | - Emilio Camafeita
- Laboratory of Cardiovascular Proteomics, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Inmaculada Jorge
- Laboratory of Cardiovascular Proteomics, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/ Nicolás cabrera 1, Madrid 28049, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Centro Nacional de Investigaciones Oncológicas (CNIO), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Noa B Martín-Cófreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, C/ Diego de León 62, Madrid 28006, Spain.,Cell-cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, C/ Diego de León 62, Madrid 28006, Spain.,Cell-cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), C/ Melchor Fdz Almagro 3, Madrid 28029, Spain
| |
Collapse
|
63
|
Berry L, Chen CT, Reininger L, Carvalho TG, El Hajj H, Morlon-Guyot J, Bordat Y, Lebrun M, Gubbels MJ, Doerig C, Daher W. The conserved apicomplexan Aurora kinase TgArk3 is involved in endodyogeny, duplication rate and parasite virulence. Cell Microbiol 2016; 18:1106-1120. [PMID: 26833682 DOI: 10.1111/cmi.12571] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 12/25/2022]
Abstract
Aurora kinases are eukaryotic serine/threonine protein kinases that regulate key events associated with chromatin condensation, centrosome and spindle function and cytokinesis. Elucidating the roles of Aurora kinases in apicomplexan parasites is crucial to understand the cell cycle control during Plasmodium schizogony or Toxoplasma endodyogeny. Here, we report on the localization of two previously uncharacterized Toxoplasma Aurora-related kinases (Ark2 and Ark3) in tachyzoites and of the uncharacterized Ark3 orthologue in Plasmodium falciparum erythrocytic stages. In Toxoplasma gondii, we show that TgArk2 and TgArk3 concentrate at specific sub-cellular structures linked to parasite division: the mitotic spindle and intranuclear mitotic structures (TgArk2), and the outer core of the centrosome and the budding daughter cells cytoskeleton (TgArk3). By tagging the endogenous PfArk3 gene with the green fluorescent protein in live parasites, we show that PfArk3 protein expression peaks late in schizogony and localizes at the periphery of budding schizonts. Disruption of the TgArk2 gene reveals no essential function for tachyzoite propagation in vitro, which is surprising giving that the P. falciparum and P. berghei orthologues are essential for erythrocyte schizogony. In contrast, knock-down of TgArk3 protein results in pronounced defects in parasite division and a major growth deficiency. TgArk3-depleted parasites display several defects, such as reduced parasite growth rate, delayed egress and parasite duplication, defect in rosette formation, reduced parasite size and invasion efficiency and lack of virulence in mice. Our study provides new insights into cell cycle control in Toxoplasma and malaria parasites and highlights Aurora kinase 3 as potential drug target.
Collapse
Affiliation(s)
- Laurence Berry
- Dynamique des Interactions Membranaires Normales et Pathologiques, UMR5235 CNRS, Université Montpellier, Montpellier, France
| | - Chun-Ti Chen
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Luc Reininger
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, INSERM U1016, Institut Cochin, Paris, France
| | - Teresa G Carvalho
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria, Australia, 3800
| | - Hiba El Hajj
- Department of Internal Medicine and Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Juliette Morlon-Guyot
- Dynamique des Interactions Membranaires Normales et Pathologiques, UMR5235 CNRS, Université Montpellier, Montpellier, France
| | - Yann Bordat
- Dynamique des Interactions Membranaires Normales et Pathologiques, UMR5235 CNRS, Université Montpellier, Montpellier, France
| | - Maryse Lebrun
- Dynamique des Interactions Membranaires Normales et Pathologiques, UMR5235 CNRS, Université Montpellier, Montpellier, France
| | - Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Christian Doerig
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria, Australia, 3800
| | - Wassim Daher
- Dynamique des Interactions Membranaires Normales et Pathologiques, UMR5235 CNRS, Université Montpellier, Montpellier, France
| |
Collapse
|
64
|
Dos Santos EO, Carneiro-Lobo TC, Aoki MN, Levantini E, Bassères DS. Aurora kinase targeting in lung cancer reduces KRAS-induced transformation. Mol Cancer 2016; 15:12. [PMID: 26842935 PMCID: PMC4739397 DOI: 10.1186/s12943-016-0494-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 01/20/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Activating mutations in KRAS are prevalent in lung cancer and have been causally linked to the oncogenic process. However, therapies targeted to oncogenic RAS have been ineffective to date and identification of KRAS targets that impinge on the oncogenic phenotype is warranted. Based on published studies showing that mitotic kinases Aurora A (AURKA) and B (AURKB) cooperate with oncogenic RAS to promote malignant transformation and that AURKA phosphorylates RAS effector pathway components, the aim of this study was to investigate whether AURKA and AURKB are KRAS targets in lung cancer and whether targeting these kinases might be therapeutically beneficial. METHODS In order to determine whether oncogenic KRAS induces Aurora kinase expression, we used qPCR and western blotting in three different lung cell-based models of gain- or loss-of-function of KRAS. In order to determine the functional role of these kinases in KRAS-induced transformation, we generated KRAS-positive A549 and H358 cells with stable and inducible shRNA-mediated knockdown of AURKA or AURKB and evaluated transformation in vitro and tumor growth in vivo. In order to validate AURKA and/or AURKB as therapeutically relevant KRAS targets in lung cancer, we treated A549 and H358 cells, as well as two different lung cell based models of gain-of-function of KRAS with a dual Aurora kinase inhibitor and performed functional in vitro assays. RESULTS We determined that KRAS positively regulates AURKA and AURKB expression. Furthermore, in KRAS-positive H358 and A549 cell lines, inducible knockdown of AURKA or AURKB, as well as treatment with a dual AURKA/AURKB inhibitor, decreased growth, viability, proliferation, transformation, and induced apoptosis in vitro. In addition, inducible shRNA-mediated knockdown of AURKA in A549 cells decreased tumor growth in vivo. More importantly, dual pharmacological inhibiton of AURKA and AURKB reduced growth, viability, transformation, and induced apoptosis in vitro in an oncogenic KRAS-dependent manner, indicating that Aurora kinase inhibition therapy can specifically target KRAS-transformed cells. CONCLUSIONS Our results support our hypothesis that Aurora kinases are important KRAS targets in lung cancer and suggest Aurora kinase inhibition as a novel approach for KRAS-induced lung cancer therapy.
Collapse
Affiliation(s)
| | | | - Mateus Nobrega Aoki
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, Brazil.
| | - Elena Levantini
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy.
| | - Daniela Sanchez Bassères
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
65
|
Lindon C, Grant R, Min M. Ubiquitin-Mediated Degradation of Aurora Kinases. Front Oncol 2016; 5:307. [PMID: 26835416 PMCID: PMC4716142 DOI: 10.3389/fonc.2015.00307] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/25/2015] [Indexed: 11/18/2022] Open
Abstract
The Aurora kinases are essential regulators of mitosis in eukaryotes. In somatic cell divisions of higher eukaryotes, the paralogs Aurora kinase A (AurA) and Aurora kinase B (AurB) play non-overlapping roles that depend on their distinct spatiotemporal activities. These mitotic roles of Aurora kinases depend on their interactions with different partners that direct them to different mitotic destinations and different substrates: AurB is a component of the chromosome passenger complex that orchestrates the tasks of chromosome segregation and cytokinesis, while AurA has many known binding partners and mitotic roles, including a well-characterized interaction with TPX2 that mediates its role in mitotic spindle assembly. Beyond the spatial control conferred by different binding partners, Aurora kinases are subject to temporal control of their activation and inactivation. Ubiquitin-mediated proteolysis is a critical route to irreversible inactivation of these kinases, which must occur for ordered transition from mitosis back to interphase. Both AurA and AurB undergo targeted proteolysis after anaphase onset as substrates of the anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase, even while they continue to regulate steps during mitotic exit. Temporal control of Aurora kinase destruction ensures that AurB remains active at the midbody during cytokinesis long after AurA activity has been largely eliminated from the cell. Differential destruction of Aurora kinases is achieved despite the fact that they are targeted at the same time and by the same ubiquitin ligase, making these substrates an interesting case study for investigating molecular determinants of ubiquitin-mediated proteolysis in higher eukaryotes. The prevalence of Aurora overexpression in cancers and their potential as therapeutic targets add importance to the task of understanding the molecular determinants of Aurora kinase stability. Here, we review what is known about ubiquitin-mediated targeting of these critical mitotic regulators and discuss the different factors that contribute to proteolytic control of Aurora kinase activity in the cell.
Collapse
Affiliation(s)
- Catherine Lindon
- Department of Pharmacology, University of Cambridge , Cambridge , UK
| | - Rhys Grant
- Department of Pharmacology, University of Cambridge , Cambridge , UK
| | - Mingwei Min
- Department of Cell Biology, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
66
|
Erythroblast enucleation is a dynein-dependent process. Exp Hematol 2015; 44:247-56.e12. [PMID: 26724640 DOI: 10.1016/j.exphem.2015.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/14/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023]
Abstract
Mammalian erythroblasts undergo enucleation through a process thought to be similar to cytokinesis. Microtubule-organizing centers (MTOCs) mediate organization of the mitotic spindle apparatus that separates the chromosomes during mitosis and are known to be crucial for proper cytokinesis. However, the role of MTOCs in erythroblast enucleation remains unknown. We therefore investigated the effect of various MTOC inhibitors on cytokinesis and enucleation using human colony-forming units-erythroid (CFU-Es) and mature erythroblasts generated from purified CD34(+) cells. We found that erythro-9-[3-(2-hydroxynonyl)]adenine (EHNA), a dynein inhibitor, and monastrol, a kinesin Eg5 inhibitor, as well as various inhibitors of MTOC regulators, including ON-01910 (Plk-1), MLN8237 (aurora A), hesperadin (aurora B), and LY294002 (PI3K), all inhibited CFU-E cytokinesis. Among these inhibitors, however, only EHNA blocked enucleation. Moreover, terminally differentiated erythroblasts expressed only dynein; little or none of the other tested proteins was detected. Over the course of the terminal differentiation of human erythroblasts, the fraction of cells with nuclei at the cell center declined, whereas the fraction of polarized cells, with nuclei shifted to a position near the plasma membrane, increased. Dynein inhibition impaired nuclear polarization, thereby blocking enucleation. These data indicate that dynein plays an essential role not only in cytokinesis but also in enucleation. We therefore conclude that human erythroblast enucleation is a process largely independent of MTOCs, but dependent on dynein.
Collapse
|
67
|
Reboutier D, Benaud C, Prigent C. Aurora A's Functions During Mitotic Exit: The Guess Who Game. Front Oncol 2015; 5:290. [PMID: 26734572 PMCID: PMC4685928 DOI: 10.3389/fonc.2015.00290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/07/2015] [Indexed: 11/24/2022] Open
Abstract
Until recently, the knowledge of Aurora A kinase functions during mitosis was limited to pre-metaphase events, particularly centrosome maturation, G2/M transition, and mitotic spindle assembly. However, an involvement of Aurora A in post-metaphase events was also suspected, but not clearly demonstrated due to the technical difficulty to perform the appropriate experiments. Recent developments of both an analog-specific version of Aurora A and small molecule inhibitors have led to the first demonstration that Aurora A is required for the early steps of cytokinesis. As in pre-metaphase, Aurora A plays diverse functions during anaphase, essentially participating in astral microtubules dynamics and central spindle assembly and functioning. The present review describes the experimental systems used to decipher new functions of Aurora A during late mitosis and situate these functions into the context of cytokinesis mechanisms.
Collapse
Affiliation(s)
- David Reboutier
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Christelle Benaud
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Claude Prigent
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| |
Collapse
|
68
|
de Souza EE, Hehnly H, Perez AM, Meirelles GV, Smetana JHC, Doxsey S, Kobarg J. Human Nek7-interactor RGS2 is required for mitotic spindle organization. Cell Cycle 2015; 14:656-67. [PMID: 25664600 DOI: 10.4161/15384101.2014.994988] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The mitotic spindle apparatus is composed of microtubule (MT) networks attached to kinetochores organized from 2 centrosomes (a.k.a. spindle poles). In addition to this central spindle apparatus, astral MTs assemble at the mitotic spindle pole and attach to the cell cortex to ensure appropriate spindle orientation. We propose that cell cycle-related kinase, Nek7, and its novel interacting protein RGS2, are involved in mitosis regulation and spindle formation. We found that RGS2 localizes to the mitotic spindle in a Nek7-dependent manner, and along with Nek7 contributes to spindle morphology and mitotic spindle pole integrity. RGS2-depletion leads to a mitotic-delay and severe defects in the chromosomes alignment and congression. Importantly, RGS2 or Nek7 depletion or even overexpression of wild-type or kinase-dead Nek7, reduced γ-tubulin from the mitotic spindle poles. In addition to causing a mitotic delay, RGS2 depletion induced mitotic spindle misorientation coinciding with astral MT-reduction. We propose that these phenotypes directly contribute to a failure in mitotic spindle alignment to the substratum. In conclusion, we suggest a molecular mechanism whereupon Nek7 and RGS2 may act cooperatively to ensure proper mitotic spindle organization.
Collapse
Key Words
- CREST, calcium-responsive transactivator
- EB1, end-binding protein 1
- GAP, GTPase-activating protein
- MT, microtubule
- Nek, NIMA-related kinase
- Nek7
- PCM, centrosomal pericentriolar material
- PD, pull-down
- PPI, protein-protein interaction
- RGS, regulators of G protein signaling
- RGS2
- WB, Western blotting
- cell division
- mitotic spindle
- mitotic spindle orientation
- shRNA, short-interfering RNA
Collapse
Affiliation(s)
- Edmarcia Elisa de Souza
- a Laboratório Nacional de Biociências-LNBio ; Centro Nacional de Pesquisa em Energia e Materiais-CNPEM ; Campinas , SP Brasil
| | | | | | | | | | | | | |
Collapse
|
69
|
Premkumar DR, Jane EP, Pollack IF. Cucurbitacin-I inhibits Aurora kinase A, Aurora kinase B and survivin, induces defects in cell cycle progression and promotes ABT-737-induced cell death in a caspase-independent manner in malignant human glioma cells. Cancer Biol Ther 2015; 16:233-43. [PMID: 25482928 DOI: 10.4161/15384047.2014.987548] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Because STAT signaling is commonly activated in malignant gliomas as a result of constitutive EGFR activation, strategies for inhibiting the EGFR/JAK/STAT cascade are of significant interest. We, therefore, treated a panel of established glioma cell lines, including EGFR overexpressors, and primary cultures derived from patients diagnosed with glioblastoma with the JAK/STAT inhibitor cucurbitacin-I. Treatment with cucurbitacin-I depleted p-STAT3, p-STAT5, p-JAK1 and p-JAK2 levels, inhibited cell proliferation, and induced G2/M accumulation, DNA endoreduplication, and multipolar mitotic spindles. Longer exposure to cucurbitacin-I significantly reduced the number of viable cells and this decrease in viability was associated with cell death, as confirmed by an increase in the subG1 fraction. Our data also demonstrated that cucurbitacin-I strikingly downregulated Aurora kinase A, Aurora kinase B and survivin. We then searched for agents that exhibited a synergistic effect on cell death in combination with cucurbitacin-I. We found that cotreatment with cucurbitacin-I significantly increased Bcl(-)2/Bcl(-)xL family member antagonist ABT-737-induced cell death regardless of EGFR/PTEN/p53 status of malignant human glioma cell lines. Although >50% of the cucurbitacin-I plus ABT-737 treated cells were annexin V and propidium iodide positive, PARP cleavage or caspase activation was not observed. Pretreatment of z-VAD-fmk, a pan caspase inhibitor did not inhibit cell death, suggesting a caspase-independent mechanism of cell death. Genetic inhibition of Aurora kinase A or Aurora kinase B or survivin by RNA interference also sensitized glioma cells to ABT-737, suggesting a link between STAT activation and Aurora kinases in malignant gliomas.
Collapse
Key Words
- Aurora kinases
- BSA, bovine serum albumin
- DMSO, dimethyl sulfoxide
- EGFR, epidermal growth factor receptor
- FITC, fluorescein isothiocyanate
- Glioma
- MTS, 3-[4, 5-dimethylthiazol- 2yl]-5-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]-2H, tetrazolium
- NF-кB, nuclear factor кB
- PAGE, polyacrylamide gel electrophoresis
- PBS, phosphate-buffered saline
- PDGFR, platelet derived growth factor receptor
- PI, propidium iodide
- PI3K, Phosphatidylinositol 3-Kinase
- TBS, Tris-buffered saline
- TRAIL, tumor necrosis factor–related apoptosis inducing ligand
- caspase-independent cell death
- cell cycle arrest
Collapse
Affiliation(s)
- Daniel R Premkumar
- a Department of Neurosurgery ; University of Pittsburgh School of Medicine ; Pittsburgh , PA USA
| | | | | |
Collapse
|
70
|
Abstract
A universal feature of mitosis is that all chromosomes become aligned at the spindle equator--the halfway point between the two spindle poles--prior to anaphase onset. This migratory event is called congression, and is powered by centromere-bound protein machines called kinetochores. This Commentary aims to document recent advances concerning the two kinetochore-based force-generating mechanisms that drive mitotic chromosome congression in vertebrate cells: depolymerisation-coupled pulling (DCP) and lateral sliding. We aim to explore how kinetochores can 'read-out' their spatial position within the spindle, and adjust these force-generating mechanisms to ensure chromosomes reach, and then remain, at the equator. Finally, we will describe the 'life history' of a chromosome, and provide a working model for how individual mechanisms are integrated to ensure efficient and successful congression.
Collapse
Affiliation(s)
- Philip Auckland
- Mechanochemical Cell Biology Building, Division of Biomedical Cell Biology, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Andrew D McAinsh
- Mechanochemical Cell Biology Building, Division of Biomedical Cell Biology, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
71
|
Thoma G, Veenstra S, Strang R, Blanz J, Vangrevelinghe E, Berghausen J, Lee CC, Zerwes HG. Orally bioavailable Syk inhibitors with activity in a rat PK/PD model. Bioorg Med Chem Lett 2015; 25:4642-7. [PMID: 26320624 DOI: 10.1016/j.bmcl.2015.08.037] [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] [Received: 08/05/2015] [Revised: 08/12/2015] [Accepted: 08/13/2015] [Indexed: 12/15/2022]
Abstract
Design and optimization of benzo- and pyrido-thiazoles/isothiazoles are reported leading to the discovery of the potent, orally bioavailable Syk inhibitor 5, which was found to be active in a rat PK/PD model. Compound 5 showed acceptable overall kinase selectivity. However, in addition to Syk it also inhibited Aurora kinase in enzymatic and cellular settings leading to findings in the micronucleus assay. As a consequence, compound 5 was not further pursued.
Collapse
Affiliation(s)
- Gebhard Thoma
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland.
| | - Siem Veenstra
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Ross Strang
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Joachim Blanz
- Analytical Sciences & Imaging, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Eric Vangrevelinghe
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Jörg Berghausen
- Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Christian C Lee
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Hans-Günter Zerwes
- Autoimmunity, Transplantation and Inflammation Research, Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| |
Collapse
|
72
|
Yang KT, Tang CJC, Tang TK. Possible Role of Aurora-C in Meiosis. Front Oncol 2015; 5:178. [PMID: 26322271 PMCID: PMC4534787 DOI: 10.3389/fonc.2015.00178] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/20/2015] [Indexed: 01/02/2023] Open
Abstract
The meiotic generation of haploid gametes with equal contents of genetic material is important for sexual reproduction in mammals. Errors in the transmission of chromosomes during meiosis may lead to aneuploidy, which is the leading cause of miscarriage and congenital birth defects in humans. The Aurora kinases, which include Aurora-A, Aurora-B, and Aurora-C, are highly conserved serine–threonine kinases that play essential roles in centrosome function, chromosome segregation, and cytokinesis during mitosis and meiosis. While Aurora-A and Aurora-B have been extensively studied in mitosis, the role of Aurora-C in meiosis is only now starting to be revealed. For example, the perturbation of Aurora-C kinase activity by microinjection of Aurora-C-kinase-dead mutant mRNAs into mouse oocytes induced multiple defects, including chromosome misalignment, abnormal kinetochore–microtubule attachment, premature chromosome segregation, and failure of cytokinesis during meiotic division. However, the analysis of such defects is complicated by the possibility that Aurora-B may be present in mammalian germ cells. Interestingly, a homozygous mutation of Aurora-C in humans leads to the production of large-headed polyploid spermatozoa and causes male infertility, but homozygous females are fertile. Mouse studies regarding the roles of Aurora-B and Aurora-C in female meiotic divisions have yielded inconsistent results, and it has proven difficult to explain why homozygous human females have no significant clinical phenotype. In this review, we will discuss the controversial status of Aurora-B in oocytes and the possible role of Aurora-C during meiotic division.
Collapse
Affiliation(s)
- Kuo-Tai Yang
- Department of Animal Science and Technology, National Taiwan University , Taipei , Taiwan
| | - Chieh-Ju C Tang
- Institute of Biomedical Sciences, Academia Sinica , Taipei , Taiwan
| | - Tang K Tang
- Institute of Biomedical Sciences, Academia Sinica , Taipei , Taiwan
| |
Collapse
|
73
|
Carvalhal S, Ribeiro SA, Arocena M, Kasciukovic T, Temme A, Koehler K, Huebner A, Griffis ER. The nucleoporin ALADIN regulates Aurora A localization to ensure robust mitotic spindle formation. Mol Biol Cell 2015; 26:3424-38. [PMID: 26246606 PMCID: PMC4591688 DOI: 10.1091/mbc.e15-02-0113] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022] Open
Abstract
The nucleoporin ALADIN, which is mutated in patients with triple A syndrome, is necessary for proper spindle formation. Without ALADIN, active Aurora A moves away from centrosomes. The relocalization of active Aurora A leads to a redistribution of specific spindle assembly factors that make spindles less stable and slows their formation. The formation of the mitotic spindle is a complex process that requires massive cellular reorganization. Regulation by mitotic kinases controls this entire process. One of these mitotic controllers is Aurora A kinase, which is itself highly regulated. In this study, we show that the nuclear pore protein ALADIN is a novel spatial regulator of Aurora A. Without ALADIN, Aurora A spreads from centrosomes onto spindle microtubules, which affects the distribution of a subset of microtubule regulators and slows spindle assembly and chromosome alignment. ALADIN interacts with inactive Aurora A and is recruited to the spindle pole after Aurora A inhibition. Of interest, mutations in ALADIN cause triple A syndrome. We find that some of the mitotic phenotypes that we observe after ALADIN depletion also occur in cells from triple A syndrome patients, which raises the possibility that mitotic errors may underlie part of the etiology of this syndrome.
Collapse
Affiliation(s)
- Sara Carvalhal
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom
| | - Susana Abreu Ribeiro
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543 Wellcome Trust Centre for Cell Biology, Institute of Cell and Molecular Biology, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Miguel Arocena
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom
| | - Taciana Kasciukovic
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom
| | - Achim Temme
- Department of Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Katrin Koehler
- Department of Paediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Angela Huebner
- Department of Paediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Eric R Griffis
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543
| |
Collapse
|
74
|
Lavogina D, Kisand K, Raidaru G, Uri A. Fluorescent photoaffinity probes for mitotic protein kinase Aurora A. Bioorg Med Chem Lett 2015; 25:3290-4. [DOI: 10.1016/j.bmcl.2015.05.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 12/30/2022]
|
75
|
Platani M, Trinkle-Mulcahy L, Porter M, Jeyaprakash AA, Earnshaw WC. Mio depletion links mTOR regulation to Aurora A and Plk1 activation at mitotic centrosomes. J Cell Biol 2015; 210:45-62. [PMID: 26124292 PMCID: PMC4494011 DOI: 10.1083/jcb.201410001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Coordination of cell growth and proliferation in response to nutrient supply is mediated by mammalian target of rapamycin (mTOR) signaling. In this study, we report that Mio, a highly conserved member of the SEACAT/GATOR2 complex necessary for the activation of mTORC1 kinase, plays a critical role in mitotic spindle formation and subsequent chromosome segregation by regulating the proper concentration of active key mitotic kinases Plk1 and Aurora A at centrosomes and spindle poles. Mio-depleted cells showed reduced activation of Plk1 and Aurora A kinase at spindle poles and an impaired localization of MCAK and HURP, two key regulators of mitotic spindle formation and known substrates of Aurora A kinase, resulting in spindle assembly and cytokinesis defects. Our results indicate that a major function of Mio in mitosis is to regulate the activation/deactivation of Plk1 and Aurora A, possibly by linking them to mTOR signaling in a pathway to promote faithful mitotic progression.
Collapse
Affiliation(s)
- Melpomeni Platani
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Laura Trinkle-Mulcahy
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H8M5, Canada Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H8M5, Canada
| | - Michael Porter
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - A Arockia Jeyaprakash
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - William C Earnshaw
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| |
Collapse
|
76
|
Li S, Deng Z, Fu J, Xu C, Xin G, Wu Z, Luo J, Wang G, Zhang S, Zhang B, Zou F, Jiang Q, Zhang C. Spatial Compartmentalization Specializes the Function of Aurora A and Aurora B. J Biol Chem 2015; 290:17546-58. [PMID: 25987563 DOI: 10.1074/jbc.m115.652453] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Indexed: 12/20/2022] Open
Abstract
Aurora kinase A and B share great similarity in sequences, structures, and phosphorylation motif, yet they show different localizations and play distinct crucial roles. The factors that determine such differences are largely unknown. Here we targeted Aurora A to the localization of Aurora B and found that Aurora A phosphorylates the substrate of Aurora B and substitutes its function in spindle checkpoint. In return, the centrosome targeting of Aurora B substitutes the function of Aurora A in the mitotic entry. Expressing the chimera proteins of the Auroras with exchanged N termini in cells indicates that the divergent N termini are also important for their spatiotemporal localizations and functions. Collectively, we demonstrate that functional divergence of Aurora kinases is determined by spatial compartmentalization, and their divergent N termini also contribute to their spatial and functional differentiation.
Collapse
Affiliation(s)
- Si Li
- From the Ministry of Education Key Laboratory of Bio-resources and Eco-environment, College of Life Sciences, Sichuan University, Chengdu, Sichuan 610064 and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Zhaoxuan Deng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jingyan Fu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Caiyue Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guangwei Xin
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Zhige Wu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jia Luo
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Gang Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Shuli Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Boyan Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Fangdong Zou
- From the Ministry of Education Key Laboratory of Bio-resources and Eco-environment, College of Life Sciences, Sichuan University, Chengdu, Sichuan 610064 and
| | - Qing Jiang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Chuanmao Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
77
|
Shoji W, Suenaga Y, Kaneko Y, Islam SMR, Alagu J, Yokoi S, Nio M, Nakagawara A. NCYM promotes calpain-mediated Myc-nick production in human MYCN-amplified neuroblastoma cells. Biochem Biophys Res Commun 2015; 461:501-6. [PMID: 25896758 DOI: 10.1016/j.bbrc.2015.04.050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 12/15/2022]
Abstract
NCYM is a cis-antisense gene of MYCN and is amplified in human neuroblastomas. High NCYM expression is associated with poor prognoses, and the NCYM protein stabilizes MYCN to promote proliferation of neuroblastoma cells. However, the molecular mechanisms of NCYM in the regulation of cell survival have remained poorly characterized. Here we show that NCYM promotes cleavage of MYCN to produce the anti-apoptotic protein, Myc-nick, both in vitro and in vivo. NCYM and Myc-nick were induced at G2/M phase, and NCYM knockdown induced apoptotic cell death accompanied by Myc-nick downregulation. These results reveal a novel function of NCYM as a regulator of Myc-nick production in human neuroblastomas.
Collapse
Affiliation(s)
- Wataru Shoji
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan; Department of Pediatric Surgery, Graduate School of Medicine, Tohoku University, Sendai 980-8574, Japan
| | - Yusuke Suenaga
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan; Cancer Genome Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan.
| | - Yoshiki Kaneko
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan
| | - S M Rafiqul Islam
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan
| | - Jennifer Alagu
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan
| | - Sana Yokoi
- Cancer Genome Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan
| | - Masaki Nio
- Department of Pediatric Surgery, Graduate School of Medicine, Tohoku University, Sendai 980-8574, Japan
| | - Akira Nakagawara
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan.
| |
Collapse
|
78
|
Gottardo M, Callaini G, Riparbelli MG. Aurora A inhibition by MNL8054 promotes centriole elongation during Drosophila male meiosis. Cell Cycle 2015; 14:2844-52. [PMID: 25785740 DOI: 10.1080/15384101.2015.1026488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Aurora A kinase plays an important role in several aspects of cell division, including centrosome maturation and separation, a crucial step for the correct organization of the bipolar spindle. Although it has long been showed that this kinase accumulates at the centrosome throughout mitosis its precise contribution to centriole biogenesis and structure has until now not been reported. It is not surprising that so little is known, due to the small size of somatic centrioles, where only dramatic structural changes may be identified by careful electron microscopy analysis. Conversely, centrioles of Drosophila primary spermatocytes increase tenfold in length during the first prophase, thus making any change easily detectable. Therefore, we examined the consequence of the pharmacological inhibition of Aurora A by MLN8054 on centriole biogenesis during early Drosophila gametogenesis. Here, we show that depletion of this kinase results in longer centrioles, mainly during transition from prophase to prometaphase of the first meiosis. We also found abnormal ciliogenesis characterized by irregularly growing axonemal doublets. Our results represent the first documentation of a potential requirement of Aurora A in centriole integrity and elongation.
Collapse
Affiliation(s)
- Marco Gottardo
- a Department of Life Sciences ; University of Siena ; Siena , Italy
| | | | | |
Collapse
|
79
|
Telford BJ, Chen A, Beetham H, Frick J, Brew TP, Gould CM, Single A, Godwin T, Simpson KJ, Guilford P. Synthetic Lethal Screens Identify Vulnerabilities in GPCR Signaling and Cytoskeletal Organization in E-Cadherin–Deficient Cells. Mol Cancer Ther 2015; 14:1213-23. [DOI: 10.1158/1535-7163.mct-14-1092] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 03/06/2015] [Indexed: 11/16/2022]
|
80
|
Suvorova ES, Francia M, Striepen B, White MW. A novel bipartite centrosome coordinates the apicomplexan cell cycle. PLoS Biol 2015; 13:e1002093. [PMID: 25734885 PMCID: PMC4348508 DOI: 10.1371/journal.pbio.1002093] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/30/2015] [Indexed: 12/11/2022] Open
Abstract
Apicomplexan parasites can change fundamental features of cell division during their life cycles, suspending cytokinesis when needed and changing proliferative scale in different hosts and tissues. The structural and molecular basis for this remarkable cell cycle flexibility is not fully understood, although the centrosome serves a key role in determining when and how much replication will occur. Here we describe the discovery of multiple replicating core complexes with distinct protein composition and function in the centrosome of Toxoplasma gondii. An outer core complex distal from the nucleus contains the TgCentrin1/TgSfi1 protein pair, along with the cartwheel protein TgSas-6 and a novel Aurora-related kinase, while an inner core closely aligned with the unique spindle pole (centrocone) holds distant orthologs of the CEP250/C-Nap protein family. This outer/inner spatial relationship of centrosome cores is maintained throughout the cell cycle. When in metaphase, the duplicated cores align to opposite sides of the kinetochores in a linear array. As parasites transition into S phase, the cores sequentially duplicate, outer core first and inner core second, ensuring that each daughter parasite inherits one copy of each type of centrosome core. A key serine/threonine kinase distantly related to the MAPK family is localized to the centrosome, where it restricts core duplication to once per cycle and ensures the proper formation of new daughter parasites. Genetic analysis of the outer core in a temperature-sensitive mutant demonstrated this core functions primarily in cytokinesis. An inhibition of ts-TgSfi1 function at high temperature caused the loss of outer cores and a severe block to budding, while at the same time the inner core amplified along with the unique spindle pole, indicating the inner core and spindle pole are independent and co-regulated. The discovery of a novel bipartite organization in the parasite centrosome that segregates the functions of karyokinesis and cytokinesis provides an explanation for how cell cycle flexibility is achieved in apicomplexan life cycles. The apicomplexan parasite Toxoplasma gondii has a unique centrosome with two specialized compartments, potentially explaining the remarkable flexibility in life cycle that these organisms can show in diverse host cells. Apicomplexan parasites infect many different hosts and tissues, causing numerous human diseases, including malaria. These important pathogens have a peculiar cell cycle in which chromosomes sometimes amplify to remarkable levels, followed by concerted cell division—providing an unusual proliferative capacity. This capacity for proliferation, combined with an ability to change the scale of replication when needed, are hallmarks of the cell cycles of these parasites. Yet the molecular mechanism responsible for these peculiar cell cycles remains one of the unsolved mysteries of Apicomplexa biology. Here we show that the centrosome—an organelle that orchestrates several aspects of the cell cycle—of the apicomplexan parasite Toxoplasma gondii contains specialized structures that coordinate parasite cell division. Our findings demonstrate that a two-part centrosomal architecture, comprising an inner and an outer core with distinct protein compositions, segregates the processes of mitosis from the assembly of new daughter parasites. The modular organization of the centrosome offers an explanation for how cell division can be suspended while the parasites amplify their genome to the biotic scale required for their life cycles. It is unknown whether these distinct centrosome core complexes evolved independently in Apicompexa. Another possibility is that the foundations for these mechanisms were present in the original eukaryote, which could explain how the distinct extranuclear centrosome of animal cells and the novel yeast spindle pole body of the nuclear envelope may have evolved from a common ancestor.
Collapse
Affiliation(s)
- Elena S. Suvorova
- Departments of Molecular Medicine & Global Health and the Florida Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida, United States of America
| | - Maria Francia
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Boris Striepen
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Michael W. White
- Departments of Molecular Medicine & Global Health and the Florida Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
81
|
Yount AL, Zong H, Walczak CE. Regulatory mechanisms that control mitotic kinesins. Exp Cell Res 2015; 334:70-7. [PMID: 25576382 DOI: 10.1016/j.yexcr.2014.12.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 12/26/2014] [Indexed: 11/18/2022]
Abstract
During mitosis, the mitotic spindle is assembled to align chromosomes at the spindle equator in metaphase, and to separate the genetic material equally to daughter cells in anaphase. The spindle itself is a macromolecular machine composed of an array of dynamic microtubules and associated proteins that coordinate the diverse events of mitosis. Among the microtubule associated proteins are a plethora of molecular motor proteins that couple the energy of ATP hydrolysis to force production. These motors, including members of the kinesin superfamily, must function at the right time and in the right place to insure the fidelity of mitosis. Misregulation of mitotic motors in disease states, such as cancer, underlies their potential utility as targets for antitumor drug development and highlights the importance of understanding the molecular mechanisms for regulating their function. Here, we focus on recent progress about regulatory mechanisms that control the proper function of mitotic kinesins and highlight new findings that lay the path for future studies.
Collapse
Affiliation(s)
- Amber L Yount
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, United States
| | - Hailing Zong
- Department of Biology, Indiana University, Bloomington, IN 47405, United States
| | - Claire E Walczak
- Medical Sciences, Indiana University, Myers Hall 262, 915 East 3rd Street, Bloomington, IN 47405, United States.
| |
Collapse
|
82
|
Kalatova B, Jesenska R, Hlinka D, Dudas M. Tripolar mitosis in human cells and embryos: occurrence, pathophysiology and medical implications. Acta Histochem 2015; 117:111-25. [PMID: 25554607 DOI: 10.1016/j.acthis.2014.11.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 01/08/2023]
Abstract
Tripolar mitosis is a specific case of cell division driven by typical molecular mechanisms of mitosis, but resulting in three daughter cells instead of the usual count of two. Other variants of multipolar mitosis show even more mitotic poles and are relatively rare. In nature, this phenomenon was frequently observed or suspected in multiple common cancers, infected cells, the placenta, and in early human embryos with impaired pregnancy-yielding potential. Artificial causes include radiation and various toxins. Here we combine several pieces of the most recent evidence for the existence of different types of multipolar mitosis in preimplantation embryos together with a detailed review of the literature. The related molecular and cellular mechanisms are discussed, including the regulation of centriole duplication, mitotic spindle biology, centromere functions, cell cycle checkpoints, mitotic autocorrection mechanisms, and the related complicating factors in healthy and affected cells, including post-mitotic cell-cell fusion often associated with multipolar cell division. Clinical relevance for oncology and embryo selection in assisted reproduction is also briefly discussed in this context.
Collapse
|
83
|
Guo L, Xiao Y, Wang Y. Application of adenosine triphosphate affinity probe and scheduled multiple-reaction monitoring analysis for profiling global kinome in human cells in response to arsenite treatment. Anal Chem 2014; 86:10700-7. [PMID: 25301106 PMCID: PMC4222629 DOI: 10.1021/ac502592d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Phosphorylation of cellular components
catalyzed by kinases plays
important roles in cell signaling and proliferation. Quantitative
assessment of perturbation in global kinome may provide crucial knowledge
for elucidating the mechanisms underlying the cytotoxic effects of
environmental toxicants. Here, we utilized an adenosine triphosphate
(ATP) affinity probe coupled with stable isotope labeling by amino
acids in cell culture (SILAC) to assess quantitatively the arsenite-induced
alteration of global kinome in human cells. We constructed a SILAC-compatible
kinome library for scheduled multiple-reaction monitoring (MRM) analysis
and adopted on-the-fly recalibration of retention time shift, which
provided better throughput of the analytical method and enabled the
simultaneous quantification of the expression of ∼300 kinases
in two LC-MRM runs. With this improved analytical method, we conducted
an in-depth quantitative analysis of the perturbation of kinome of
GM00637 human skin fibroblast cells induced by arsenite exposure.
Several kinases involved in cell cycle progression, including cyclin-dependent
kinases (CDK1 and CDK4) and Aurora kinases A, B, and C, were found
to be hyperactivated, and the altered expression of CDK1 was further
validated by Western analysis. In addition, treatment with a CDK inhibitor,
flavopiridol, partially restored the arsenite-induced growth inhibition
of human skin fibroblast cells. Thus, sodium arsenite may confer its
cytotoxic effect partly through the aberrant activation of CDKs and
the resultant perturbation of cell cycle progression. Together, we
developed a high-throughput, SILAC-compatible, and MRM-based kinome
profiling method and demonstrated that the method is powerful in deciphering
the molecular modes of action of a widespread environmental toxicant.
The method should be generally applicable for uncovering the cellular
pathways triggered by other extracellular stimuli.
Collapse
Affiliation(s)
- Lei Guo
- Environmental Toxicology Graduate Program and ‡Department of Chemistry, University of California , Riverside, California 92521-0403, United States
| | | | | |
Collapse
|
84
|
Jayanthan A, Ruan Y, Truong TH, Narendran A. Aurora kinases as druggable targets in pediatric leukemia: heterogeneity in target modulation activities and cytotoxicity by diverse novel therapeutic agents. PLoS One 2014; 9:e102741. [PMID: 25048812 PMCID: PMC4105567 DOI: 10.1371/journal.pone.0102741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/23/2014] [Indexed: 11/19/2022] Open
Abstract
Leukemia is the most common pediatric malignancy, constituting more than 30% of all childhood cancers. Although cure rates have improved greatly, approximately one in five children relapse and poor survival rates post relapse remain a challenge. Given this, more effective and innovative therapeutic strategies are needed in order to improve prognosis. Aurora kinases, a family of serine/threonine kinases essential for the regulation of several mitotic processes, have been identified as potential targets for cancer therapeutics. Elevated expression of Aurora kinases has been demonstrated in several malignancies and is associated with aberrant mitotic activity, aneuploidy and alterations in chromosomal structure and genome instability. Based on this rationale, a number of small molecule inhibitors have been formulated and advanced to human studies in the recent past. A comparative analysis of these agents in cytotoxicity and target modulation analyses against a panel of leukemia cells provides novel insights into the unique mechanisms and codependent activity pathways involved in targeting Aurora kinases, constituting a distinctive preclinical experimental framework to identify appropriate agents and combinations in future clinical studies.
Collapse
Affiliation(s)
- Aarthi Jayanthan
- Pediatric Oncology Experimental Therapeutics Investigators Consortium (POETIC) Laboratory for Pre-Clinical and Drug Discovery Studies, University of Calgary, Calgary, Alberta, Canada
| | - Yibing Ruan
- Pediatric Oncology Experimental Therapeutics Investigators Consortium (POETIC) Laboratory for Pre-Clinical and Drug Discovery Studies, University of Calgary, Calgary, Alberta, Canada
| | - Tony H. Truong
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Aru Narendran
- Pediatric Oncology Experimental Therapeutics Investigators Consortium (POETIC) Laboratory for Pre-Clinical and Drug Discovery Studies, University of Calgary, Calgary, Alberta, Canada
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|
85
|
Newnham L, Jordan PW, Carballo JA, Newcombe S, Hoffmann E. Ipl1/Aurora kinase suppresses S-CDK-driven spindle formation during prophase I to ensure chromosome integrity during meiosis. PLoS One 2013; 8:e83982. [PMID: 24386320 PMCID: PMC3873974 DOI: 10.1371/journal.pone.0083982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 10/29/2013] [Indexed: 11/18/2022] Open
Abstract
Cells coordinate spindle formation with DNA repair and morphological modifications to chromosomes prior to their segregation to prevent cell division with damaged chromosomes. Here we uncover a novel and unexpected role for Aurora kinase in preventing the formation of spindles by Clb5-CDK (S-CDK) during meiotic prophase I and when the DDR is active in budding yeast. This is critical since S-CDK is essential for replication during premeiotic S-phase as well as double-strand break induction that facilitates meiotic recombination and, ultimately, chromosome segregation. Furthermore, we find that depletion of Cdc5 polo kinase activity delays spindle formation in DDR-arrested cells and that ectopic expression of Cdc5 in prophase I enhances spindle formation, when Ipl1 is depleted. Our findings establish a new paradigm for Aurora kinase function in both negative and positive regulation of spindle dynamics.
Collapse
Affiliation(s)
- Louise Newnham
- MRC Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
| | - Philip W. Jordan
- MRC Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
| | - Jesus A. Carballo
- MRC Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
| | - Sonya Newcombe
- MRC Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
| | - Eva Hoffmann
- MRC Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
- * E-mail:
| |
Collapse
|
86
|
Kim S, Meyer R, Chuong H, Dawson DS. Dual mechanisms prevent premature chromosome segregation during meiosis. Genes Dev 2013; 27:2139-46. [PMID: 24115770 PMCID: PMC3850097 DOI: 10.1101/gad.227454.113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In meiosis I, homologous chromosomes pair and then attach to the spindle so that the homologs can be pulled apart at anaphase I. The segregation of homologs before pairing would be catastrophic. We describe two mechanisms that prevent this. First, in early meiosis, Ipl1, the budding yeast homolog of the mammalian Aurora B kinase, triggers shedding of a kinetochore protein, preventing microtubule attachment. Second, Ipl1 localizes to the spindle pole bodies (SPBs), where it blocks spindle assembly. These processes are reversed upon expression of Ndt80. Previous studies have shown that Ndt80 is expressed when homologs have successfully partnered, and this triggers a rise in the levels of cyclin-dependent kinase (CDK). We found that CDK phosphorylates Ipl1, delocalizing it from SPBs, triggering spindle assembly. At the same time, kinetochores reassemble. Thus, dual mechanisms controlled by Ipl1 and Ntd80 coordinate chromosome and spindle behaviors to prevent the attachment of unpartnered chromosomes to the meiotic spindle.
Collapse
Affiliation(s)
- Seoyoung Kim
- Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | | | | | | |
Collapse
|
87
|
EBNA3C-mediated regulation of aurora kinase B contributes to Epstein-Barr virus-induced B-cell proliferation through modulation of the activities of the retinoblastoma protein and apoptotic caspases. J Virol 2013; 87:12121-38. [PMID: 23986604 DOI: 10.1128/jvi.02379-13] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Epstein-Barr virus (EBV) is an oncogenic gammaherpesvirus that is implicated in several human malignancies, including Burkitt's lymphoma (BL), posttransplant lymphoproliferative disease (PTLD), nasopharyngeal carcinoma (NPC), and AIDS-associated lymphomas. Epstein-Barr nuclear antigen 3C (EBNA3C), one of the essential EBV latent antigens, can induce mammalian cell cycle progression through its interaction with cell cycle regulators. Aurora kinase B (AK-B) is important for cell division, and deregulation of AK-B is associated with aneuploidy, incomplete mitotic exit, and cell death. Our present study shows that EBNA3C contributes to upregulation of AK-B transcript levels by enhancing the activity of its promoter. Further, EBNA3C also increased the stability of the AK-B protein, and the presence of EBNA3C leads to reduced ubiquitination of AK-B. Importantly, EBNA3C in association with wild-type AK-B but not with its kinase-dead mutant led to enhanced cell proliferation, and AK-B knockdown can induce nuclear blebbing and cell death. This phenomenon was rescued in the presence of EBNA3C. Knockdown of AK-B resulted in activation of caspase 3 and caspase 9, along with poly(ADP-ribose) polymerase 1 (PARP1) cleavage, which is known to be an important contributor to apoptotic signaling. Importantly, EBNA3C failed to stabilize the kinase-dead mutant of AK-B compared to wild-type AK-B, which suggests a role for the kinase domain in AK-B stabilization and downstream phosphorylation of the cell cycle regulator retinoblastoma protein (Rb). This study demonstrates the functional relevance of AK-B kinase activity in EBNA3C-regulated B-cell proliferation and apoptosis.
Collapse
|