RASSF1A Interacts with and Activates the Mitotic Kinase Aurora-A

Total Page:16

File Type:pdf, Size:1020Kb

RASSF1A Interacts with and Activates the Mitotic Kinase Aurora-A Oncogene (2008) 27, 6175–6186 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $32.00 www.nature.com/onc ORIGINAL ARTICLE RASSF1A interacts with and activates the mitotic kinase Aurora-A L Liu1, C Guo1, R Dammann2, S Tommasi1 and GP Pfeifer1 1Division of Biology, Beckman Research Institute, City of Hope Cancer Center, Duarte, CA, USA and 2Institute of Genetics, University of Giessen, Giessen, Germany The RAS association domain family 1A (RASSF1A) gene tumorigenesis and carcinogen-induced tumorigenesis is located at chromosome 3p21.3 within a specific area of (Tommasi et al., 2005; van der Weyden et al., 2005), common heterozygous and homozygous deletions. RASS- supporting the notion that RASSF1A is a bona fide F1A frequently undergoes promoter methylation-asso- tumor suppressor. However, it is not fully understood ciated inactivation in human cancers. Rassf1aÀ/À mice how RASSF1A is involved in tumor suppression. are prone to both spontaneous and carcinogen-induced The biochemical function of the RASSF1A protein is tumorigenesis, supporting the notion that RASSF1A is a largely unknown. The homology of RASSF1A with the tumor suppressor. However, it is not fully understood how mammalian Ras effector novel Ras effector (NORE)1 RASSF1A is involved in tumor suppression pathways. suggests that the RASSF1A gene product may function Here we show that overexpression of RASSF1A inhibits in signal transduction pathways involving Ras-like centrosome separation. RASSF1A interacts with Aurora-A, proteins. However, recent data indicate that RASSF1A a mitotic kinase. Surprisingly, knockdown of RASS- itself binds to RAS only weakly and that binding to F1A by siRNA led to reduced activation of Aurora-A, RAS may require heterodimerization of RASSF1A and whereas overexpression of RASSF1A resulted in in- NORE1 (Ortiz-Vega et al., 2002). In addition, there is creased activation of Aurora-A, suggesting that RASS- evidence for an association of both NORE1 and F1A is involved in Aurora-A activation. Like other RASSF1A with the pro-apoptotic kinase MST1 and Aurora-A activators, RASSF1A was also a substrate of that this interaction is involved in apoptosis induced by Aurora-A in vitro. The failure of recombinant RASSF1A activated Ras (Khokhlatchev et al., 2002). to activate recombinant Aurora-A indicates that RASS- Recently, several groups, including ours, have re- F1A may not activate Aurora-A directly and suggests that ported that RASSF1A is a microtubule- and centro- RASSF1A may function as a scaffold to bring together some-associated protein, which regulates mitotic Aurora-A and its activator(s). Inhibition of centrosome progression (Liu et al., 2003, 2005c; Dallol et al., 2004; separation by RASSF1A overexpression is most likely a Rong et al., 2004; Song et al., 2004; Vos et al., 2004). We consequence of hyperstabilization of microtubules by this have shown that overexpressed RASSF1A co-localized protein. with microtubules in interphase cells and with spindles Oncogene (2008) 27, 6175–6186; doi:10.1038/onc.2008.220; and centrosomes during mitosis (Liu et al., 2003). published online 21 July 2008 Forced expression of RASSF1A induced mitotic arrest with aberrant mitotic cells (Liu et al., 2003). Similar Keywords: RASSF1A; Aurora-A; centrosome; mitosis results been have been obtained by several other groups (Dallol et al., 2004; Rong et al., 2004; Song et al., 2004; Vos et al., 2004). Song et al. (2004) have reported that RASSF1A regulated mitosis by inhibiting the anaphase- Introduction promoting complex (APC) through binding to CDC20 and induces G2–M arrest at pro-metaphase. The The RAS association domain family 1A (RASSF1A) function of RASSF1A was independent of the protein gene is located at chromosome 3p21.3 within a specific early mitotic inhibitor 1, and therefore Song et al. (2004) area of common heterozygous and homozygous dele- proposed that RASSF1A acted in early prometaphase tions (Dammann et al., 2000; Lerman and Minna, 2000). to prevent the degradation of mitotic cyclins and to The RASSF1A gene is one of the most frequently delay mitotic progression. However, our laboratory has methylation-silenced genes described in human cancer been unable to confirm an interaction between RASS- to date (Agathanggelou et al., 2005; Dammann et al., F1A and CDC20 (Liu et al., 2007). RASSF1A may 2005). Rassf1aÀ/À mice are prone to both spontaneous function as a tumor suppressor through promoting apoptosis and/or by controlling mitotic cell division (Agathanggelou et al., 2005; Dammann et al., 2005). Correspondence:Dr GP Pfeifer, Division of Biology, Beckman However, its exact mechanism of action remains to be Research Institute, City of Hope Cancer Center, City of Hope, 1500 determined. East Duarte Road, Duarte, CA 91010, USA. E-mail:[email protected] Aurora-A belongs to a family of highly conserved Received 14 April 2008; revised 4 June 2008; accepted 10 June 2008; serine/threonine kinases, which includes Aurora-A, -B published online 21 July 2008 and -C in mammals. Aurora kinases combined with RASSF1A interacts with and activates Aurora-A LLiuet al 6176 cyclin-dependent kinase 1 (or p34cdc2), Polo-like kinases 2005; Weaver and Cleveland, 2006). Being a putative and NIMA-related kinases are four families of known oncogene, Aurora-A has attracted a great deal of mitotic kinases that regulate mitotic progression. All attention as a potential antitumor target (Keen and three members of the Aurora kinase family share a Taylor, 2004; Mortlock et al., 2005; Naruganahalli conserved C-terminal catalytic domain and have been et al., 2006). found to have multiple mitotic functions (Andrews Here we demonstrate that endogenous RASSF1A et al., 2003; Carmena and Earnshaw, 2003). Among interacts with and activates Aurora-A. them, Aurora-A is the most intensively studied one. Originally Aurora-A was identified in a screen for Drosophila melanogaster mutants that have abnormal spindle-pole phenotypes (Glover et al., 1995). Aurora-A Results is located on centrosomes from late S/early G to the 2 RASSF1A regulates centrosome separation time when the cells exit mitosis (Carmena and Earn- shaw, 2003). Aurora-A is important in centrosome RASSF1A is a microtubule- and centrosome-associated maturation and separation as well as in spindle assembly protein in interphase cells and is found at the spindle et al (Andrews et al., 2003; Carmena and Earnshaw, 2003; poles and midzone region in mitotic cells (Liu ., et al et al Marumoto et al., 2005). Moreover, activity of Aurora-A 2003, 2005a, c; Dallol ., 2004; Rong ., 2004; Vos et al et al is required for timely mitotic entry (Marumoto et al., ., 2004; van der Weyden ., 2005). Endogenous 2002, 2003; Hirota et al., 2003; Hachet et al., 2007; RASSF1A is primarily a centrosomal protein in et al Portier et al., 2007). Both upregulation and down- interphase cells (Guo ., 2007) and it colocalizes regulation of Aurora-A results in cytokinesis failure with mitotic spindles in mitosis (Figure 1a). Using (Meraldi et al., 2002; Anand et al., 2003; Marumoto EGFP-tagged deletion fragments of RASSF1A, we et al., 2003; Zhang et al., 2004), indicating that the determined that the centrosome and spindle pole timing of Aurora-A activity is important for proper localization of RASSF1A requires a minimal domain cytokinesis. fragment from amino acid D120 to G193 (Figure 1b). Expression and localization of Aurora-A is cell-cycle This domain is distinct from the RAS association regulated. Its protein level peaks in the middle of mitosis domain present in RASSF1A (R194–S288) and only and is diminished in late mitosis/early G by the Cdh1- partially overlaps with the minimum microtubule- 1 et al activated APC/C (APC/cyclosome) (Honda et al., 2000; binding domain (D120-S288) (Liu ., 2003). Castro et al., 2002; Taguchi et al., 2002). The activity of Previously, we found that overexpression of RASS- Aurora-A is also controlled by phosphorylation. Phos- F1A resulted in over 50% of mitoses with monopolar et al phorylation of Thr288 of human Aurora-A is essential spindles (Liu ., 2003). Generally, monopolar for its kinase activity (Walter et al., 2000; Littlepage spindles may result from defects in centrosome duplica- et al., 2002). Several activators of Aurora-A have been tion or from failure of centrosome separation. When identified, which include TPX2, Ajuba, HEF1, Bora and these cells were stained with an anti-g-tubulin antibody, PAK1 (Kufer et al., 2002; Hirota et al., 2003; Crane we observed that there were two closely spaced g-tubulin et al., 2004; Satinover et al., 2004; Pugacheva and spots in 96% of the monopolar spindle containing cells, Golemis, 2005; Zhao et al., 2005; Hutterer et al., 2006). which indicates that the centrosomes have duplicated Activation of Aurora-A by TPX2 is required for but were not separated far enough to organize a bipolar bipolar-spindle assembly (Trieselmann et al., 2003; Tsai spindle (Figure 2). et al., 2003), whereas activation of Aurora-A by Ajuba is Microtubules are important in centrosome separation et al et al et al essential for mitotic entry (Hirota et al., 2003). It is (Waters ., 1993; Gaglio ., 1996; Uzbekov ., tempting to propose that Aurora-A activators may at 2002). Therefore, we speculated that inhibition of least partially control the temporal and spatial distribu- microtubule dynamics by RASSF1A may at least in tion of Aurora-A activity along the cell division cycle. part account for its effect on centrosome separation. Aurora-A is frequently overexpressed in various Indeed, the monopolar spindles were hyperstabilized human tumors (Bischoff et al., 1998; Zhou et al., and resistant to nocodazole-induced depolymerization 1998). Overexpression of Aurora-A was sufficient to (Figure 2). transform NIH3T3 and Rat1 cells (Bischoff et al., 1998; Zhou et al., 1998). However, the same regimen did not RASSF1A interacts with Aurora-A transform primary murine embryonic fibroblasts We asked if there are other mechanisms by which (MEFs), suggesting that upregulation of Aurora-A itself RASSF1A may regulate centrosome separation.
Recommended publications
  • Aurora Kinase a in Gastrointestinal Cancers: Time to Target Ahmed Katsha1, Abbes Belkhiri1, Laura Goff3 and Wael El-Rifai1,2,4*
    Katsha et al. Molecular Cancer (2015) 14:106 DOI 10.1186/s12943-015-0375-4 REVIEW Open Access Aurora kinase A in gastrointestinal cancers: time to target Ahmed Katsha1, Abbes Belkhiri1, Laura Goff3 and Wael El-Rifai1,2,4* Abstract Gastrointestinal (GI) cancers are a major cause of cancer-related deaths. During the last two decades, several studies have shown amplification and overexpression of Aurora kinase A (AURKA) in several GI malignancies. These studies demonstrated that AURKA not only plays a role in regulating cell cycle and mitosis, but also regulates a number of key oncogenic signaling pathways. Although AURKA inhibitors have moved to phase III clinical trials in lymphomas, there has been slower progress in GI cancers and solid tumors. Ongoing clinical trials testing AURKA inhibitors as a single agent or in combination with conventional chemotherapies are expected to provide important clinical information for targeting AURKA in GI cancers. It is, therefore, imperative to consider investigations of molecular determinants of response and resistance to this class of inhibitors. This will improve evaluation of the efficacy of these drugs and establish biomarker based strategies for enrollment into clinical trials, which hold the future direction for personalized cancer therapy. In this review, we will discuss the available data on AURKA in GI cancers. We will also summarize the major AURKA inhibitors that have been developed and tested in pre-clinical and clinical settings. Keywords: Aurora kinases, Therapy, AURKA inhibitors, MNL8237, Alisertib, Gastrointestinal, Cancer, Signaling pathways Introduction stage [9-11]. Furthermore, AURKA is critical for Mitotic kinases are the main proteins that coordinate ac- bipolar-spindle assembly where it interacts with Ran- curate mitotic processing [1].
    [Show full text]
  • Therapeutic Implications for Ovarian Cancer Emerging from the Tumor Cancer Genome Atlas
    Review Article Therapeutic implications for ovarian cancer emerging from the Tumor Cancer Genome Atlas Claire Verschraegen, Karen Lounsbury, Alan Howe, Marc Greenblatt University of Vermont Cancer Center, Burlington, VT 05405, USA Correspondence to: Claire Verschraegen, MD. University of Vermont Cancer Center, 89 Beaumont Avenue, Given E 214, Burlington, VT 05405, USA. Email: [email protected] Abstract: With increasing insights into the molecular landscape of ovarian cancer, subtypes are emerging that might require differential targeted therapies. While the combination of a platinum and a taxane remains the standard of care, newer therapies, specifically targeted to molecular anomalies, are rapidly being tested in various cancers. A major effort to better understand ovarian cancer occurred through the Cancer Atlas Project. The Catalogue of Somatic Mutations in Cancer (COSMIC) is a database that collates mutation data and associated information extracted from the primary literature. The information from the Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC), which systematically analyzed hundreds of ovarian cancer, are included in COSMIC, sometimes with discordant results. In this manuscript, the published data (mainly from TCGA) on somatic high grade papillary serous ovarian cancer (HGSOC) mutations has been used as the basis to propose a more granular approach to ovarian cancer treatment, already a reality for tumors such as lung and breast cancers. TP53 mutations are the most common molecular anomaly in HGSOC, and lead to genomic instability, perhaps through the FOXM1 node. Normalizing P53 has been a therapeutic challenge, and is being extensively studied. BRCAness is found an about 50% of HGSOC and can be targeted with poly (ADP-ribose) polymerase (PARP) inhibitors, such as olaparib, recently approved for ovarian cancer treatment.
    [Show full text]
  • HIPK2 and Extrachromosomal Histone H2B Are Separately Recruited by Aurora-B for Cytokinesis
    Oncogene https://doi.org/10.1038/s41388-018-0191-6 ARTICLE HIPK2 and extrachromosomal histone H2B are separately recruited by Aurora-B for cytokinesis 1 1 2 3 2 Laura Monteonofrio ● Davide Valente ● Manuela Ferrara ● Serena Camerini ● Roberta Miscione ● 3 1,2 1 Marco Crescenzi ● Cinzia Rinaldo ● Silvia Soddu Received: 14 November 2017 / Revised: 24 January 2018 / Accepted: 5 February 2018 © The Author(s) 2018. This article is published with open access Abstract Cytokinesis, the final phase of cell division, is necessary to form two distinct daughter cells with correct distribution of genomic and cytoplasmic materials. Its failure provokes genetically unstable states, such as tetraploidization and polyploidization, which can contribute to tumorigenesis. Aurora-B kinase controls multiple cytokinetic events, from chromosome condensation to abscission when the midbody is severed. We have previously shown that HIPK2, a kinase involved in DNA damage response and development, localizes at the midbody and contributes to abscission by phosphorylating extrachromosomal histone H2B at Ser14. Of relevance, HIPK2-defective cells do not phosphorylate H2B 1234567890();,: and do not successfully complete cytokinesis leading to accumulation of binucleated cells, chromosomal instability, and increased tumorigenicity. However, how HIPK2 and H2B are recruited to the midbody during cytokinesis is still unknown. Here, we show that regardless of their direct (H2B) and indirect (HIPK2) binding of chromosomal DNA, both H2B and HIPK2 localize at the midbody independently of nucleic acids. Instead, by using mitotic kinase-specific inhibitors in a spatio-temporal regulated manner, we found that Aurora-B kinase activity is required to recruit both HIPK2 and H2B to the midbody.
    [Show full text]
  • Cell Division- Ch 5
    Cell Division- Mitosis and Meiosis When do cells divide? Cell size . One of most important factors affecting size of the cell is size of cell membrane . Cell must remain relatively small to survive (why?) – Cell membrane has to be big enough to take in nutrients and eliminate wastes – As cells get bigger, the volume increases faster than the surface area – Small cells have a larger surface area to volume ratio than larger cells to help with nutrient intake and waste elimination . When a cell reaches its max size, the nucleus starts cell division: called MITOSIS or MEIOSIS Mitosis . General Information – Occurs in somatic (body) cells ONLY!! – Nickname: called “normal” cell division – Produces somatic cells only . Background Info – Starts with somatic cell in DIPLOID (2n) state . Cell contains homologous chromosomes- chromosomes that control the same traits but not necessarily in the same way . 1 set from mom and 1 set from dad – Ends in diploid (2n) state as SOMATIC cells – Goes through one set of divisions – Start with 1 cell and end with 2 cells Mitosis (cont.) . Accounts for three essential life processes – Growth . Result of cell producing new cells . Develop specialized shapes/functions in a process called differentiation . Rate of cell division controlled by GH (Growth Hormone) which is produced in the pituitary gland . Ex. Nerve cell, intestinal cell, etc. – Repair . Cell regenerates at the site of injury . Ex. Skin (replaced every 28 days), blood vessels, bone Mitosis (cont.) – Reproduction . Asexual – Offspring produced by only one parent – Produce offspring that are genetically identical – MITOSIS – Ex. Bacteria, fungi, certain plants and animals .
    [Show full text]
  • The Kinase Activity of Aurora B Is Required for Kinetochore-Microtubule Interactions During Mitosis
    Current Biology, Vol. 12, 894–899, June 4, 2002, 2002 Elsevier Science Ltd. All rights reserved. PII S0960-9822(02)00848-5 The Kinase Activity of Aurora B Is Required for Kinetochore-Microtubule Interactions during Mitosis Maki Murata-Hori and Yu-li Wang1 chromosomal congression, we monitored the move- Department of Physiology ment of individual centromeres in cells expressing a high University of Massachusetts Medical School level of aurora B(K-R)-GFP or control cells expressing a Worcester, Massachusetts 01605 similar level of wild-type aurora B-GFP (Figure 1B). Both aurora B-GFP and aurora B(K-R)-GFP were localized at centromeres and spindle poles [6] and to a less extent Summary along chromosomal arms (see Figures 3A and 3D). In control cells, the centromeres of neighboring chro- As a component of the “chromosomal passenger pro- mosomes moved independently of each other at an av- Ϯ ␮ Ϯ ϭ tein complex,” the aurora B kinase is associated with erage rate of 1.8 1.2 m/min (mean SD, n 22), centromeres during prometaphase and with midzone with frequent changes in direction (Figure 1B, left). They ف microtubules during anaphase and is required for both eventually accumulated at the metaphase plate 20 min mitosis and cytokinesis [1–6]. Ablation of aurora B after nuclear envelope breakdown (19/19, Figures 1Aa– causes defects in both prometaphase chromosomal 1Ad; Supplementary Movie 1 available with this article congression and the spindle checkpoint [4–6]; how- online). In contrast, in cells expressing aurora B(K-R)- ever, the mechanisms underlying these defects are GFP, centromeres of neighboring chromosomes moved Ϯ ␮ unclear.
    [Show full text]
  • Ran Controls Microtubule Asters and Nuclear Assembly 2455 Chromatin Rounded up (Fig
    Journal of Cell Science 112, 2453-2461 (1999) 2453 Printed in Great Britain © The Company of Biologists Limited 1999 JCS0524 Ran-GTP stabilises microtubule asters and inhibits nuclear assembly in Xenopus egg extracts Chuanmao Zhang1,2, Mike Hughes1 and Paul R. Clarke1,* 1Biomedical Research Centre, University of Dundee, Level 5, Ninewells Hospital and Medical School, Dundee DD1 9SY, Scotland, UK 2Department of Cell Biology and Genetics, College of Life Sciences, Peking University, Beijing 100871, China *Author for correspondence (e-mail: [email protected]) Accepted 25 May; published on WWW 24 June 1999 SUMMARY Ran is an abundant GTPase of the Ras superfamily that is nucleus and blocks chromatin decondensation. In contrast, highly conserved in eukaryotes. In interphase cells, Ran is Ran GDP does not stabilise microtubules or inhibit nuclear mainly nuclear and thought to be predominantly GTP- assembly. RanT24N and RanBP1, which oppose the bound, but it is also present in the cytoplasm, probably generation of Ran-GTP by RCC1, arrest nuclear growth GDP-bound. This asymmetric distribution plays an after disappearance of the aster. Ran associates with important role in directing nucleocytoplasmic transport. microtubule asters in egg extracts and with mitotic spindles Ran has also been implicated in cell cycle control, including in somatic Xenopus cells, suggesting that it may affect the transition from mitosis to interphase when the microtubule stability directly. These results show that Ran compartmentalisation of the nucleus is established. Here, has a novel function in the control of microtubule stability we have examined the role of Ran in this transition using that is clearly distinct from nucleocytoplasmic transport.
    [Show full text]
  • The CHEK2 Variant C.349A>G Is Associated with Prostate Cancer
    cancers Article The CHEK2 Variant C.349A>G Is Associated with Prostate Cancer Risk and Carriers Share a Common Ancestor Andreia Brandão 1 , Paula Paulo 1, Sofia Maia 1, Manuela Pinheiro 1, Ana Peixoto 2, Marta Cardoso 1, Maria P. Silva 1, Catarina Santos 2, Rosalind A. Eeles 3,4 , Zsofia Kote-Jarai 3, 5,6 7, 8,9 10 Kenneth Muir , UKGPCS Collaborators y, Johanna Schleutker , Ying Wang , Nora Pashayan 11,12 , Jyotsna Batra 13,14, APCB BioResource 13,14, Henrik Grönberg 15, David E. Neal 16,17,18, Børge G. Nordestgaard 19,20 , Catherine M. Tangen 21, Melissa C. Southey 22,23,24, Alicja Wolk 25,26, Demetrius Albanes 27, Christopher A. Haiman 28, Ruth C. Travis 29, Janet L. Stanford 30,31, Lorelei A. Mucci 32, Catharine M. L. West 33, Sune F. Nielsen 19,20, Adam S. Kibel 34, Olivier Cussenot 35,36, Sonja I. Berndt 27, Stella Koutros 27, Karina Dalsgaard Sørensen 37,38 , Cezary Cybulski 39 , Eli Marie Grindedal 40, Jong Y. Park 41 , Sue A. Ingles 42, Christiane Maier 43, Robert J. Hamilton 44,45, Barry S. Rosenstein 46,47, 48,49,50 7, Ana Vega , The IMPACT Study Steering Committee and Collaborators z, Manolis Kogevinas 51,52,53,54, Fredrik Wiklund 15 , Kathryn L. Penney 55, Hermann Brenner 56,57,58 , Esther M. John 59, Radka Kaneva 60, Christopher J. Logothetis 61, Susan L. Neuhausen 62, Kim De Ruyck 63, Azad Razack 64, Lisa F. Newcomb 30,65, Canary PASS Investigators 30,65, Davor Lessel 66 , Nawaid Usmani 67,68, Frank Claessens 69, Manuela Gago-Dominguez 70,71 , Paul A.
    [Show full text]
  • Mtor REGULATES AURORA a VIA ENHANCING PROTEIN STABILITY
    mTOR REGULATES AURORA A VIA ENHANCING PROTEIN STABILITY Li Fan Submitted to the faculty of the University Graduate School in partial fulfillment of the requirements for the degree Doctor of Philosophy in the Department of Biochemistry and Molecular Biology, Indiana University December 2013 Accepted by the Graduate Faculty, of Indiana University, in partial fulfillment of the requirements for the degree of Doctor of Philosophy. Lawrence A. Quilliam, Ph.D., Chair Doctoral Committee Simon J. Atkinson, Ph.D. Mark G. Goebl, Ph.D. October 22, 2013 Maureen A. Harrington, Ph.D. Ronald C. Wek, Ph.D. ii © 2013 Li Fan iii DEDICATION I dedicate this thesis to my family: to my parents, Xiu Zhu Fan and Shu Qin Yang, who have been loving, supporting, and encouraging me from the beginning of my life; to my husband Fei Huang, who provided unconditional support and encouragement through these years; to my son, David Yan Huang, who has made my life highly enjoyable and meaningful. iv ACKNOWLEDGMENTS I sincerely thank my mentor Dr. Lawrence Quilliam for his guidance, motivation, support, and encouragement during my dissertation work. His passion for science and the scientific and organizational skills I have learned from Dr. Quilliam made it possible for me to achieve this accomplishment. Many thanks to Drs. Ron Wek, Mark Goebl, Maureen Harrington, and Simon Atkinson for serving on my committee and providing constructive suggestions and technical advice during my Ph.D. program. I have had a pleasurable experience working with all the people in our laboratory. Thanks Drs. Justin Babcock and Sirisha Asuri, and Mr.
    [Show full text]
  • The Emerging Role of Ncrnas and RNA-Binding Proteins in Mitotic Apparatus Formation
    non-coding RNA Review The Emerging Role of ncRNAs and RNA-Binding Proteins in Mitotic Apparatus Formation Kei K. Ito, Koki Watanabe and Daiju Kitagawa * Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan; [email protected] (K.K.I.); [email protected] (K.W.) * Correspondence: [email protected] Received: 11 November 2019; Accepted: 13 March 2020; Published: 20 March 2020 Abstract: Mounting experimental evidence shows that non-coding RNAs (ncRNAs) serve a wide variety of biological functions. Recent studies suggest that a part of ncRNAs are critically important for supporting the structure of subcellular architectures. Here, we summarize the current literature demonstrating the role of ncRNAs and RNA-binding proteins in regulating the assembly of mitotic apparatus, especially focusing on centrosomes, kinetochores, and mitotic spindles. Keywords: ncRNA; centrosome; kinetochore; mitotic spindle 1. Introduction Non-coding RNAs (ncRNAs) are defined as a class of RNA molecules that are transcribed from genomic DNA, but not translated into proteins. They are mainly classified into the following two categories according to their length—small RNA (<200 nt) and long non-coding RNA (lncRNA) (>200 nt). Small RNAs include traditional RNA molecules, such as transfer RNA (tRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), PIWI-interacting RNA (piRNA), and micro RNA (miRNA), and they have been studied extensively [1]. Research on lncRNA is behind that on small RNA despite that recent transcriptome analysis has revealed that more than 120,000 lncRNAs are generated from the human genome [2–4].
    [Show full text]
  • Kinetochores Accelerate Centrosome Separation to Ensure Faithful Chromosome Segregation
    906 Research Article Kinetochores accelerate centrosome separation to ensure faithful chromosome segregation Nunu Mchedlishvili1,*, Samuel Wieser1,2,*, Rene´ Holtackers1, Julien Mouysset1, Mukta Belwal1, Ana C. Amaro1 and Patrick Meraldi1,` 1Institute of Biochemistry, ETH Zurich, Schafmattstrasse 18, 8093 Zu¨rich, Switzerland 2Wellcome Trust/Cancer Research Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK *These authors contributed equally to this work `Author for correspondence ([email protected]) Accepted 28 August 2011 Journal of Cell Science 125, 906–918 ß 2012. Published by The Company of Biologists Ltd doi: 10.1242/jcs.091967 Summary At the onset of mitosis, cells need to break down their nuclear envelope, form a bipolar spindle and attach the chromosomes to microtubules via kinetochores. Previous studies have shown that spindle bipolarization can occur either before or after nuclear envelope breakdown. In the latter case, early kinetochore–microtubule attachments generate pushing forces that accelerate centrosome separation. However, until now, the physiological relevance of this prometaphase kinetochore pushing force was unknown. We investigated the depletion phenotype of the kinetochore protein CENP-L, which we find to be essential for the stability of kinetochore microtubules, for a homogenous poleward microtubule flux rate and for the kinetochore pushing force. Loss of this force in prometaphase not only delays centrosome separation by 5–6 minutes, it also causes massive chromosome alignment and segregation defects due to the formation of syntelic and merotelic kinetochore–microtubule attachments. By contrast, CENP-L depletion has no impact on mitotic progression in cells that have already separated their centrosomes at nuclear envelope breakdown.
    [Show full text]
  • The Aurora B Kinase Activity Is Required for the Maintenance of the Differentiated State of Murine Myoblasts
    Cell Death and Differentiation (2009) 16, 321–330 & 2009 Macmillan Publishers Limited All rights reserved 1350-9047/09 $32.00 www.nature.com/cdd The Aurora B kinase activity is required for the maintenance of the differentiated state of murine myoblasts G Amabile1,2, AM D’Alise1, M Iovino1, P Jones3, S Santaguida4, A Musacchio4, S Taylor5 and R Cortese*,1 Reversine is a synthetic molecule capable of inducing dedifferentiation of C2C12, a murine myoblast cell line, into multipotent progenitor cells, which can be redirected to differentiate in nonmuscle cell types under appropriate conditions. Reversine is also a potent inhibitor of Aurora B, a protein kinase required for mitotic chromosome segregation, spindle checkpoint function, cytokinesis and histone H3 phosphorylation, raising the possibility that the dedifferentiation capability of reversine is mediated through the inhibition of Aurora B. Indeed, here we show that several other well-characterized Aurora B inhibitors are capable of dedifferentiating C2C12 myoblasts. Significantly, expressing drug-resistant Aurora B mutants, which are insensitive to reversine block the dedifferentiation process, indicating that Aurora B kinase activity is required to maintain the differentiated state. We show that the inhibition of the spindle checkpoint or cytokinesis per se is not sufficient for dedifferentiation. Rather, our data support a model whereby changes in histone H3 phosphorylation result in chromatin remodeling, which in turn restores the multipotent state. Cell Death and Differentiation (2009) 16, 321–330; doi:10.1038/cdd.2008.156; published online 31 October 2008 Lineage-restricted cells can be reprogramed to a state Evidence is emerging that the role of Aurora B is not of pluripotency by several different manipulations, including restricted to mitosis and cell division.
    [Show full text]
  • Discovery of BPR1K871, a Quinazoline Based
    www.impactjournals.com/oncotarget/ Oncotarget, 2016, Vol. 7, (No. 52), pp: 86239-86256 Research Paper Discovery of BPR1K871, a quinazoline based, multi-kinase inhibitor for the treatment of AML and solid tumors: Rational design, synthesis, in vitro and in vivo evaluation Yung Chang Hsu1,*, Mohane Selvaraj Coumar2,*, Wen-Chieh Wang1,*, Hui-Yi Shiao1,*, Yi-Yu Ke1, Wen-Hsing Lin1, Ching-Chuan Kuo1, Chun-Wei Chang1, Fu-Ming Kuo1, Pei-Yi Chen1, Sing-Yi Wang1, An-Siou Li1, Chun-Hwa Chen1, Po-Chu Kuo1, Ching- Ping Chen1, Ming-Hsine Wu1, Chen-Lung Huang1, Kuei-Jung Yen1, Yun-I Chang1, John T.-A. Hsu1, Chiung-Tong Chen1, Teng-Kuang Yeh1, Jen-Shin Song1, Chuan Shih1, Hsing-Pang Hsieh1,3 1Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC 2Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, India 3Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan, ROC *These authors contributed equally to this work Correspondence to: Hsing-Pang Hsieh, email: [email protected] Keywords: acute myeloid leukemia, aurora kinase, FLT3, quinazoline, multi-kinase inhibitor Received: July 25, 2016 Accepted: November 07, 2016 Published: November 15, 2016 ABSTRACT The design and synthesis of a quinazoline-based, multi-kinase inhibitor for the treatment of acute myeloid leukemia (AML) and other malignancies is reported. Based on the previously reported furanopyrimidine 3, quinazoline core containing lead 4 was synthesized and found to impart dual FLT3/AURKA inhibition (IC50 = 127/5 nM), as well as improved physicochemical properties. A detailed structure-activity relationship study of the lead 4 allowed FLT3 and AURKA inhibition to be finely tuned, resulting in AURKA selective (5 and 7; 100-fold selective over FLT3), FLT3 selective (13; 30-fold selective over AURKA) and dual FLT3/AURKA selective (BPR1K871; IC50 = 19/22 nM) agents.
    [Show full text]