Greatwall Is Essential to Prevent Mitotic Collapse After Nuclear Envelope Breakdown in Mammals

Total Page:16

File Type:pdf, Size:1020Kb

Greatwall Is Essential to Prevent Mitotic Collapse After Nuclear Envelope Breakdown in Mammals Greatwall is essential to prevent mitotic collapse after nuclear envelope breakdown in mammals Mónica Álvarez-Fernándeza, Ruth Sánchez-Martíneza,1, Belén Sanz-Castilloa, Pei Pei Gana, María Sanz-Floresa, Marianna Trakalaa, Miguel Ruiz-Torresa, Thierry Lorcab, Anna Castrob, and Marcos Malumbresa,2 aCell Division and Cancer Group, Spanish National Cancer Research Centre, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain; and bUniversités Montpellier 2 et 1, Centre de Recherches de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5237, 34293 Montpellier, France Edited* by Tim Hunt, Cancer Research UK, South Mimms, United Kingdom, and approved September 19, 2013 (received for review June 6, 2013) Greatwall is a protein kinase involved in the inhibition of protein cyclin B–Cdk1. The lack of Greatwall activity results in defects in phosphatase 2 (PP2A)-B55 complexes to maintain the mitotic state. chromosome condensation after NEB, and these defects can be Although its biochemical activity has been deeply characterized in rescued by concomitant ablation of B55 proteins. Our results Xenopus, its specific relevance during the progression of mitosis imply that cells are subjected to a mitotic stress resulting from is not fully understood. By using a conditional knockout of the NEB, a moment in which nuclear chromatin becomes exposed to mouse ortholog, Mastl, we show here that mammalian Greatwall cytoplasmic phosphatases. We therefore propose that Greatwall is essential for mouse embryonic development and cell cycle pro- shuttles to the cytoplasm before NEB and prevents mitotic col- gression. Yet, Greatwall-null cells enter into mitosis with normal lapse by inhibiting the PP2A–B55-dependent dephosphorylation kinetics. However, these cells display mitotic collapse after nuclear of Cdk substrates. envelope breakdown (NEB) characterized by defective chromo- some condensation and prometaphase arrest. Intriguingly, Great- Results wall is exported from the nucleus to the cytoplasm in a CRM1- Genetic Ablation of Mastl in the Mouse. Mastl(+/−) mice (Fig. 1 A dependent manner before NEB. This export occurs after the nuclear and B) develop normally and are fertile. However, no viable importofcyclinB–Cdk1 complexes, requires the kinase activity homozygous mutants were found after intercrosses between of Greatwall, and is mediated by Cdk-, but not Polo-like kinase these mice, suggesting embryonic lethality in the absence of CELL BIOLOGY 1-dependent phosphorylation. The mitotic collapse observed in Greatwall. Mastl(−/−) embryos developed normally in vitro Greatwall-deficient cells is partially rescued after concomitant de- during a few days and generate normal blastocysts (Fig. S1). pletion of B55 regulatory subunits, which are mostly cytoplasmic However, these mutant embryos were not detected in the utero before NEB. These data suggest that Greatwall is an essential pro- by embryonic day (E) 10.5, suggesting lethality at periimplanta- tein in mammals required to prevent mitotic collapse after NEB. tional stages (Fig. 1C). Because the germ-line null mutation in Greatwall resulted in cell cycle regulation | cell division | mitotic kinases | mitotic phosphatases | early embryonic lethality, we intercrossed Mastl(+/lox) mice nuclear export harboring an additional allele that expresses the Cre-estrogen receptor (ERT2) recombinase in the ubiquitous RNA polymerase reatwall was originally identified in Drosophila as a modu- II locus (13). We treated pregnant females at E12.5 with ta- Glator of Polo activity and a protein required for DNA con- moxifen to induce Cre activity, and embryos were analyzed at densation and normal progression through mitosis (1–4). E14.5. Genetic ablation of Greatwall resulted in increased mitotic fi Mastl Δ Δ Biochemical assays in Xenopus extracts have demonstrated that gures in the neuroepithelium of ( / ) embryos compared Mastl + Δ D fi Greatwall is able to inhibit PP2A–B55 phosphatase complexes by with ( / ) littermates (Fig. 1 ). These mitotic gures dis- phosphorylating the cAMP-regulated phosphoprotein Arpp19 played an accumulation of prometaphases/metaphases (PM) in and α-endosulfine, thus participating in the maintenance of the the absence of Greatwall compared with more abundant ana- D mitotic state (5–8). The control of PP2A through the Greatwall- phases/telophases (AT) in control sections (Fig. 1 ), thus sug- dependent phosphorylation of Arpp19/Ensa proteins has also gesting that Greatwall is essential for proper mitotic progression been supported by genetic studies in Drosophila (9, 10). The in vivo in mammals. mammalian ortholog of Greatwall, also known as microtubule- associated serine/threonine kinase-like protein (Mastl), also Significance participates in the maintenance of the mitotic state by inhibiting PP2A phosphatases (11, 12). Inhibition of Greatwall is required Nuclear envelope breakdown (NEB) leads to the exposure of for the activation of PP2A–B55α,δ complexes during mitotic exit nuclear structures to cytoplasmic activities. Greatwall is a ki- (13), thus suggesting the relevance of this pathway in maintaining nase able to inhibit PP2A phosphatases that counteract Cdk- the mitotic state (4). How Greatwall activity and function is dependent phosphorylation required for mitosis. Here we show regulated is not well established. Several evidences support a role that Greatwall, an essential protein in mammals, is exported to for cyclin-dependent kinase (Cdk)-dependent phosphorylation in the cytoplasm in a Cdk-dependent manner before NEB, thus the activation of Greatwall, and several phosphorylation sites for protecting mitotic phosphates from phosphatase activity. multiple kinases have been mapped (14, 15). In addition, although Greatwall is mostly nuclear in interphase (3, 11), the cellular and Author contributions: M.M. designed research; M.A.-F., R.S.-M., B.S.-C., P.P.G., M.S.-F., molecular basis of the control of its dynamic intracellular M.T., and M.R.-T. performed research; M.T., T.L., and A.C. contributed new reagents/analytic fi tools; M.A.-F., R.S.-M., B.S.-C., P.P.G., M.S.-F., M.T., M.R.-T., and M.M. analyzed data; and traf cking and its activity remains largely unknown. M.A.-F. and M.M. wrote the paper. We show in this work that the murine Greatwall ortholog, fl encoded by the Mastl gene, is essential for mouse development The authors declare no con ict of interest. and cell cycle progression. Greatwall-null cultures, however, *This Direct Submission article had a prearranged editor. 1Present address: Madrid Institute for Advanced Studies (IMDEA)-Food, 28049 Madrid, Spain. display normal kinetics during the G2/M transition, suggesting that this protein is not required for mitotic entry. Greatwall is 2To whom correspondence should be addressed. E-mail: [email protected]. exported to the cytoplasm before nuclear envelope breakdown This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. (NEB) but, interestingly, this export follows nuclear import of 1073/pnas.1310745110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1310745110 PNAS Early Edition | 1of6 Downloaded by guest on October 2, 2021 A B serum, cells were monitored by using time-lapse microscopy Mastl(+) Wild-type during 2 d (Fig. 2A). No differences were observed in the kinetics 1234 56789 101112 Mastl(+/+) (–/–) (+/–) of mitotic entry (determined by cell rounding and chromosome F1 F2 R1 Mastl(+) Mastl(loxfrt) Hypomorphic? B C 34frt-neo-frt condensation) in these cultures (Fig. 2 and ). In addition, no Flp Mastl(–) differences were detected in the accumulation of MPM2-positive Mastl(lox) Conditional 34 F1/R1 cells by immunofluorescence (Fig. 2D) or in the accumulation of Mastl(–) Cre Mastl(+) Null E Mastl( ) cyclin B1 or phospho-histone H3 (Fig. 2 ). However, Greatwall- F2/R1 3 loxP frt C Developmental stage Mastl(+/+) Mastl(+/–) Mastl(–/–) Total Live births 22 (27%) 60 (73%) 0 (0%) 82 Serum addition/ Embryos E13.5-18.5 15 (37%) 23 (56%) 0 (0%) 38 A Mastl(lox/lox) nucleofection H2B-mRFP Start video Embryos E10.5-11.5 14 (21%) 35 (51%) 0 (0%) 49 Serum deprivation (72h) 5h 48h Embryos E1.5 + 4 days i.v.c 5 (25%) 8 (40%) 7 (35%) 20 Adeno infection (48h) t=0 60h 84h post infection D pH3 ** ** E GFP Cre GFP Cre B ) 120 min 100 Greatwall (lox/lox) (20-min ( frames) 80 Actin Mastl Mastl 60 Mastl(lox/lox) 4 ) Mastl( ) 40 360 min ) 3 ( (60-min Percentage of cells Percentage ( 20 2 frames) Mastl Mastl 0 I MIM PM AT PM AT 1 PDL (rel to day 0) Mastl( ) ( )( )( ) 0 0246 C D 100 Mastl 25 3 * Days (lox/lox) Mastl(lox/lox) DAPI -tub pH3 merged Mastl( ) Mastl n.s. F 80 20 ( ) 2 60 15 (lox/lox) n.s. 40 10 Mastl 1 n.s. control) (rel. to 20 MPM2+ cells (%) 5 MPM2 levels per cell MPM2 levels per Cumulative mitotic index ) Cumulative mitotic entry (%) 0 0 0 ( 0 10 20 30 h 0 20 24 36 h Mastl (lox/lox) ( ) Time Time Mastl E Mastl(lox/lox) Mastl( ) F Mastl G Normal division Binucleated (lox/lox)( ) No segregation Death in mitosis 02024 36020 24 36 h Fig. 1. Greatwall mutant mice and cells. (A) The exon 4 of the murine gene Greatwall 100 fl Greatwall encoding Greatwall, Mastl, was anked by loxP sites (green triangles) and 75 fi P-Cdk a selection cassette [Mastl(loxfrt) allele]. Site-speci c recombination by Flp CyclinB1 substrates results in the Mastl(lox) allele, which expresses normal levels of Greatwall. 50 pH3 Cre-mediated recombination results in the germ-line [Mastl(–)] or conditionally 25 CyclinB1 of Percentage cells induced [Mastl(Δ)] null alleles. (B) Representative PCR products showing the -actin Securin 0 indicated alleles after amplification using oligonucleotides indicated in A. (lox/lox) ( )((lox/lox) ) (C) The number (and percentage) of mice or embryos with the indicated + siMad2 *** *** genotype is shown, indicating that Mastl(−/−) embryos develop normally H 600 Mastl(lox/lox) Mastl( until blastocyte stage. (D) Immunohistochemical analysis of E14.5 embryos siLuc siMad2 siLuc siMad2 fi showing an increase in mitotic (M) gures (pH3, phosphohistone H3 signal, 400 0 28000 28 28 82 h brown) versus interphasic (I) cells in the subventricular zone in Mastl(Δ/Δ) vs.
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Transcriptomic Analysis of Native Versus Cultured Human and Mouse Dorsal Root Ganglia Focused on Pharmacological Targets Short
    bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. Transcriptomic analysis of native versus cultured human and mouse dorsal root ganglia focused on pharmacological targets Short title: Comparative transcriptomics of acutely dissected versus cultured DRGs Andi Wangzhou1, Lisa A. McIlvried2, Candler Paige1, Paulino Barragan-Iglesias1, Carolyn A. Guzman1, Gregory Dussor1, Pradipta R. Ray1,#, Robert W. Gereau IV2, # and Theodore J. Price1, # 1The University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson, TX, 75080, USA 2Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine # corresponding authors [email protected], [email protected] and [email protected] Funding: NIH grants T32DA007261 (LM); NS065926 and NS102161 (TJP); NS106953 and NS042595 (RWG). The authors declare no conflicts of interest Author Contributions Conceived of the Project: PRR, RWG IV and TJP Performed Experiments: AW, LAM, CP, PB-I Supervised Experiments: GD, RWG IV, TJP Analyzed Data: AW, LAM, CP, CAG, PRR Supervised Bioinformatics Analysis: PRR Drew Figures: AW, PRR Wrote and Edited Manuscript: AW, LAM, CP, GD, PRR, RWG IV, TJP All authors approved the final version of the manuscript. 1 bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Role of MASTL in Mammals: Molecular Functions and Physiological Relevance
    Departamento de Biología Molecular Facultad de Ciencias Universidad Autónoma de Madrid Role of MASTL in mammals: Molecular functions and physiological relevance Belén Sanz Castillo Degree in Biotechnology Thesis Directors: Dr. Marcos Malumbres Dra. Mónica Álvarez Fernández Centro Nacional de Investigaciones Oncológicas (CNIO) Madrid, 2017 Marcos Malumbres Martínez, Jefe del Grupo de División Celular y Cáncer del Centro Nacional de Investigaciones Oncológicas (CNIO) y, Mónica Álvarez Fernández, Investigadora del Grupo de División Celular y Cáncer del CNIO. Certifican: que Belén Sanz Castillo ha realizado bajo su dirección el trabajo de Tesis Doctoral titulado: ”Role of MASTL in mammals: Molecular functions and physiological relevance” en el Centro Nacional de Investigaciones Oncológicas y tutelada en el departamento de Biología Molecular de la Universidad Autónoma de Madrid. Revisado el presente trabajo, considera que reúne todas las condiciones requeridas por la legislación vigente y la originalidad y calidad científica para su presentación y defensa con el fin de optar al grado de Doctor. Y para que conste donde proceda, firmamos el presente certificado. Dr. Marcos Malumbres Martínez Dra. Mónica Álvarez Fernández 3 A mi familia “Tu llegada allí es tu destino. Mas no apresures nunca el viaje. Mejor que dure muchos años y atracar, viejo ya, en la isla, enriquecido de cuanto ganaste en el camino” Constantino Cavafis 5 Acknowledgements Esta tesis hubiera sido completamente imposible sin la ayuda y apoyo de muchas personas. En primer lugar Marcos, muchas gracias por haberme acogido en tu laboratorio, aún recuerdo cuando llegué cargada de ilusión y ahora pienso que no podría haber escogido un sitio mejor. Gracias por tu entusiasmo, pasión y fuerza que nos transmites a cada uno de los que trabajamos contigo.
    [Show full text]
  • Kinase-Independent Functions of MASTL in Cancer: a New Perspective on MASTL Targeting
    cells Perspective Kinase-Independent Functions of MASTL in Cancer: A New Perspective on MASTL Targeting 1, 1, 1, James Ronald William Conway y , Elisa Närvä y, Maria Emilia Taskinen y and Johanna Ivaska 1,2,* 1 Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; james.conway@utu.fi (J.R.W.C.); elisa.narva@utu.fi (E.N.); maria.taskinen@utu.fi (M.E.T.) 2 Department of Biochemistry, University of Turku, 20520 Turku, Finland * Correspondence: Johanna.ivaska@utu.fi These authors contributed equally to this work. y Received: 1 June 2020; Accepted: 1 July 2020; Published: 6 July 2020 Abstract: Microtubule-associated serine/threonine kinase-like (MASTL; Greatwall) is a well-characterized kinase, whose catalytic role has been extensively studied in relation to cell-cycle acceleration. Importantly, MASTL has been implicated to play a substantial role in cancer progression and subsequent studies have shown that MASTL is a significant regulator of the cellular actomyosin cytoskeleton. Several kinases have non-catalytic properties, which are essential or even sufficient for their functions. Likewise, MASTL functions have been attributed both to kinase-dependent phosphorylation of downstream substrates, but also to kinase-independent regulation of the actomyosin contractile machinery. In this review, we aimed to highlight the catalytic and non-catalytic roles of MASTL in proliferation, migration, and invasion. Further, we discussed the implications of this dual role for therapeutic design. Keywords: MASTL; actin; contractility; cell cycle; cancer; therapeutic targeting; kinase inhibitor 1. Introduction The serine/threonine kinase MASTL (microtubule-associated serine/threonine kinase-like; Greatwall) was first described in Drosophila melanogaster for the role it played in supporting cell-cycle progression [1].
    [Show full text]
  • MASTL Promotes Cell Contractility and Motility Through Kinase-Independent Signaling
    ARTICLE MASTL promotes cell contractility and motility through kinase-independent signaling Maria Emilia Taskinen1*, Elisa Narv¨ a¨1*, James R.W. Conway1, Laura Soto Hinojosa2,SergioLilla3,AnjaMai1, Nicola De Franceschi1, Laura L. Elo1, Robert Grosse2,SaraZanivan3,4, Jim C. Norman3,4, and Johanna Ivaska1,5 Microtubule-associated serine/threonine-protein kinase-like (MASTL) is a mitosis-accelerating kinase with emerging roles in cancer progression. However, possible cell cycle–independent mechanisms behind its oncogenicity remain ambiguous. Here, we identify MASTL as an activator of cell contractility and MRTF-A/SRF (myocardin-related transcription factor A/serum Downloaded from https://rupress.org/jcb/article-pdf/219/6/e201906204/1042522/jcb_201906204.pdf by guest on 05 May 2020 response factor) signaling. Depletion of MASTL increased cell spreading while reducing contractile actin stress fibers in normal and breast cancer cells and strongly impairing breast cancer cell motility and invasion. Transcriptome and proteome profiling revealed MASTL-regulated genes implicated in cell movement and actomyosin contraction, including Rho guanine nucleotide exchange factor 2 (GEF-H1, ARHGEF2) and MRTF-A target genes tropomyosin 4.2 (TPM4), vinculin (VCL), and nonmuscle myosin IIB (NM-2B, MYH10). Mechanistically, MASTL associated with MRTF-A and increased its nuclear retention and transcriptional activity. Importantly, MASTL kinase activity was not required for regulation of cell spreading or MRTF-A/SRF transcriptional activity. Taken together, we
    [Show full text]
  • Comprehensive Assessment of Indian Variations in the Druggable Kinome Landscape Highlights Distinct Insights at the Sequence, Structure and Pharmacogenomic Stratum
    SUPPLEMENTARY MATERIAL Comprehensive assessment of Indian variations in the druggable kinome landscape highlights distinct insights at the sequence, structure and pharmacogenomic stratum Gayatri Panda1‡, Neha Mishra1‡, Disha Sharma2,3, Rahul C. Bhoyar3, Abhinav Jain2,3, Mohamed Imran2,3, Vigneshwar Senthilvel2,3, Mohit Kumar Divakar2,3, Anushree Mishra3, Priyanka Banerjee4, Sridhar Sivasubbu2,3, Vinod Scaria2,3, Arjun Ray1* 1 Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla, India. 2 Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India. 3 CSIR-Institute of Genomics and Integrative Biology, Mathura Road, Delhi-110020, India. 4 Institute for Physiology, Charite-University of Medicine, Berlin, 10115 Berlin, Germany. ‡These authors contributed equally to this work. * [email protected] TABLE OF CONTENTS Name Title Supplemental_Figure_S1 Fauchere and Pliska hydrophobicity scale for variations in structure data Supplemental_Figure_S2 Phenotypic drug-drug correlogram Supplemental_Table_S1 545 kinase coding genes used in the study Supplemental_Table_S2 Classes and count of kinase coding genes Supplemental_Table_S3 Allele frequency Indian v/s other populations from 1000 genome data(1000g2015). Supplemental_Table_S4 IndiGen Structure Data- consisting of 12 genes and their 22 variants Supplemental_Table_S5 Genes, PDB ids, mutations in IndiGen data and associated drugs (FDA approved) Supplemental_Table_S6 Data used for docking and binding pocket similarity analysis Supplemental_Table_S7
    [Show full text]
  • Knockdown of Microtubule Associated Serine/Threonine Kinase Like
    Current Medical Science 41(1):108-117,2021 108 DOI https://doi.org/10.1007/s11596-021-2325-2Current Medical Science 41(1):2021 Knockdown of Microtubule Associated Serine/threonine Kinase Like Expression Inhibits Gastric Cancer Cell Growth and Induces Apoptosis by Activation of ERK1/2 and Inactivation of NF-κB Signaling* Cai-xia AN1†, Shou-pin XIE2†, Hai-long LI3, Yong-hua HU3, Rong NIU4, Lin-jie ZHANG5, Yan JIANG5, Qiang LI6, Yong-ning Zhou1# 1Division of Gastroenterology and Hepatology, The First Hospital of Lanzhou University, Lanzhou 730000, China 2Department of Neurology, The First People’s Hospital of Lanzhou City, Lanzhou 730050, China 3Department of Internal Mddicine, The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China 4Department of External Chest, Gansu Provincial Cancer Hospital, Lanzhou 730030, China 5Division of Pediatric Emergency, Gansu Provincial Maternal and Child Health Hospital, Lanzhou 730050, China 6Division of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou 730000, China The Author(s) 2021 Summary: Microtubule-associated serine/threonine kinase (MASTL) functions to regulate chromosome condensation and mitotic progression. Therefore, aberrant MASTL expression is commonly implicated in various human cancers. This study analyzed MASTL expression in gastric cancer vs. adjacent normal tissue for elucidating the association with clinicopathological data from patients. This work was then extended to investigate the effects of MASTL knockdown on tumor cells in vitro. The level of MASTL expression in gastric cancer tissue was assessed from the UALCAN, GEPIA, and Oncomine online databases. Lentivirus carrying MASTL or negative control shRNA was infected into gastric cancer cells. RT-qPCR, Western blotting, cell viability, cell counting, flow cytometric apoptosis and cell cycle, and colony formation assays were performed.
    [Show full text]
  • Thrombocytopenia-Associated Mutations in Ser/Thr Kinase MASTL
    Thrombocytopenia-associated mutations in Ser/Thr kinase MASTL deregulate actin cytoskeleton dynamics in platelets by Begoña Hurtado et al. SUPPLEMENTARY MATERIAL List of Supplementary Figures Figure S1. Hematopoietic precursors and megakaryocytes maturation in Mastl mutant mice. Figure S2. Sialylation and apoptosis profile of Mastl mutant platelets. Figure S3. Mastl E166D activity results in increased phosphorylation levels. Figure S4. Phospho-proteomic analysis in Mastl mutant platelets after thrombin activation. Figure S5. Signaling pathways differentially phosphorylated in Mastl mutant platelets. Figure S6. Changes in the phosphorylation status of signaling molecules in Mastl-mutant platelets. Figure S7. Full scan of blots showed in the manuscript. List of Supplementary Tables Table S1. Full data from phospho-proteomic studies. 1 Table S2. KEGG pathways enriched (FDR>0.05) in hyperphosphorylated proteins in resting Mastl(ED/ED) platelets (log2FC ED/WT>0.75), considering as statistical background the mouse platelet proteome. Table S3. KEGG pathways enriched (FDR>0.05) in hypophosphorylated proteins in resting Mastl(/) platelets (log2FC/WT<0.75), considering as statistical background the mouse platelet proteome. Table S4. KEGG pathways enriched (FDR>0.01) in hyperphosphorylated proteins in Mastl(ED/ED) platelets 3 minutes after stimulation with thrombin (log2FC ED/WT>0.75). Table S5. KEGG pathways enriched (FDR>0.01) in hyperphosphorylated proteins in both resting and 3-min-activated Mastl(ED/ED) platelets (log2FC ED/WT>0.5), considering as statistical background the mouse platelet proteome. Table S6. KEGG pathways enriched (FDR>0.05) in hyperphosphorylated proteins in Mastl(ED/ED) platelets 15 minutes after stimulation with thrombin (log2FC ED/WT>1.0), considering as statistical background the mouse platelet proteome.
    [Show full text]
  • MASTL Overexpression Promotes Chromosome Instability and Metastasis in Breast Cancer
    Oncogene (2018) 37:4518–4533 https://doi.org/10.1038/s41388-018-0295-z ARTICLE MASTL overexpression promotes chromosome instability and metastasis in breast cancer 1,2 1 3 1,4 1 Samuel Rogers ● Rachael A. McCloy ● Benjamin L. Parker ● David Gallego-Ortega ● Andrew M. K. Law ● 1 1 4 5,6,7 8,9 1,10 Venessa T. Chin ● James R. W. Conway ● Dirk Fey ● Ewan K. A. Millar ● Sandra O’Toole ● Niantao Deng ● 1,10 11 2 1,10 1,10 Alexander Swarbrick ● Paul D. Chastain ● Anthony J. Cesare ● Paul Timpson ● C. Elizabeth Caldon ● 1,10,12 3,13 1,10,14 15 David R. Croucher ● David E. James ● D. Neil Watkins ● Andrew Burgess Received: 20 October 2017 / Revised: 27 February 2018 / Accepted: 8 April 2018 / Published online: 10 May 2018 © The Author(s) 2018. This article is published with open access Abstract MASTL kinase is essential for correct progression through mitosis, with loss of MASTL causing chromosome segregation errors, mitotic collapse and failure of cytokinesis. However, in cancer MASTL is most commonly amplified and overexpressed. This correlates with increased chromosome instability in breast cancer and poor patient survival in breast, ovarian and lung cancer. Global phosphoproteomic analysis of immortalised breast MCF10A cells engineered to 1234567890();,: 1234567890();,: overexpressed MASTL revealed disruption to desmosomes, actin cytoskeleton, PI3K/AKT/mTOR and p38 stress kinase signalling pathways. Notably, these pathways were also disrupted in patient samples that overexpress MASTL. In MCF10A cells, these alterations corresponded with a loss of contact inhibition and partial epithelial–mesenchymal transition, which disrupted migration and allowed cells to proliferate uncontrollably in 3D culture.
    [Show full text]
  • Human Kinases Info Page
    Human Kinase Open Reading Frame Collecon Description: The Center for Cancer Systems Biology (Dana Farber Cancer Institute)- Broad Institute of Harvard and MIT Human Kinase ORF collection from Addgene consists of 559 distinct human kinases and kinase-related protein ORFs in pDONR-223 Gateway® Entry vectors. All clones are clonal isolates and have been end-read sequenced to confirm identity. Kinase ORFs were assembled from a number of sources; 56% were isolated as single cloned isolates from the ORFeome 5.1 collection (horfdb.dfci.harvard.edu); 31% were cloned from normal human tissue RNA (Ambion) by reverse transcription and subsequent PCR amplification adding Gateway® sequences; 11% were cloned into Entry vectors from templates provided by the Harvard Institute of Proteomics (HIP); 2% additional kinases were cloned into Entry vectors from templates obtained from collaborating laboratories. All ORFs are open (stop codons removed) except for 5 (MST1R, PTK7, JAK3, AXL, TIE1) which are closed (have stop codons). Detailed information can be found at: www.addgene.org/human_kinases Handling and Storage: Store glycerol stocks at -80oC and minimize freeze-thaw cycles. To access a plasmid, keep the plate on dry ice to prevent thawing. Using a sterile pipette tip, puncture the seal above an individual well and spread a portion of the glycerol stock onto an agar plate. To patch the hole, use sterile tape or a portion of a fresh aluminum seal. Note: These plasmid constructs are being distributed to non-profit institutions for the purpose of basic
    [Show full text]
  • MASTL Datasheet
    Catalog # Aliquot Size M72-10G-05 5 µg M72-10G-10 10 µg MASTL, Active Full-length recombinant human protein expressed in Sf9 cells Catalog # M72-10G Lot # Q2393-5 Product Description Specific Activity Full-length recombinant human MASTL was expressed by baculovirus in Sf9 insect cells using an N-terminal GST tag. 120,000 The gene accession number for MASTL is NM_001172303. 90,000 Gene Aliases 60,000 GREATWALL; GW; GWL; hGWL; RP11-85G18.2; THC2 30,000 (cpm) Activity Formulation 0 0 50 100 150 200 Recombinant protein stored in 50mM Tris-HCl, pH 7.5, Protein (ng) 150mM NaCl, 10mM glutathione, 0.1mM EDTA, 0.25mM DTT, 0.1mM PMSF, 25% glycerol. The specific activity of MASTL was determined to be 18 nmol /min/mg as per activity assay protocol. Storage and Stability Store product at –70oC. For optimal storage, aliquot target Purity into smaller quantities after centrifugation and store at recommended temperature. For most favorable performance, avoid repeated handling and multiple freeze/thaw cycles. The purity of MASTL protein was determined to be ~70% by Scientific Background densitometry, MASTL approx. MW 150kDa. Microtubule associated serine/threonine kinase-like (MASTL) belongs to the MAST family, AGC Ser/Thr protein kinase group. MASTL promotes mitotic progression and cell cycle reentry after DNA damage. Loss of MASTL leads to defects in chromosome condensation, separation, and other aspects of mitotic progression, which is primarily mediated by inhibiting PP2A/B55δ. Overexpression of MASTL, Active MASTL is correlated with cancer progression, poor patient Full-length recombinant human protein expressed in Sf9 cells survival, and tumor recurrence in various cancers.
    [Show full text]
  • 39UTR Shortening Identifies High-Risk Cancers with Targeted Dysregulation
    OPEN 39UTR shortening identifies high-risk SUBJECT AREAS: cancers with targeted dysregulation of GENE REGULATORY NETWORKS the ceRNA network REGULATORY NETWORKS Li Li1*, Duolin Wang2*, Mengzhu Xue1*, Xianqiang Mi1, Yanchun Liang2 & Peng Wang1,3 Received 1 2 15 April 2014 Key Laboratory of Systems Biology, Shanghai Advanced Research Institute, Chinese Academy of Sciences, College of Computer Science and Technology, Jilin University, 3School of Life Science and Technology, ShanghaiTech University. Accepted 3 June 2014 Competing endogenous RNA (ceRNA) interactions form a multilayered network that regulates gene Published expression in various biological pathways. Recent studies have demonstrated novel roles of ceRNA 23 June 2014 interactions in tumorigenesis, but the dynamics of the ceRNA network in cancer remain unexplored. Here, we examine ceRNA network dynamics in prostate cancer from the perspective of alternative cleavage and polyadenylation (APA) and reveal the principles of such changes. Analysis of exon array data revealed that both shortened and lengthened 39UTRs are abundant. Consensus clustering with APA data stratified Correspondence and cancers into groups with differing risks of biochemical relapse and revealed that a ceRNA subnetwork requests for materials enriched with cancer genes was specifically dysregulated in high-risk cancers. The novel connection between should be addressed to 39UTR shortening and ceRNA network dysregulation was supported by the unusually high number of P.W. (wangpeng@ microRNA response elements (MREs) shared by the dysregulated ceRNA interactions and the significantly sari.ac.cn) altered 39UTRs. The dysregulation followed a fundamental principle in that ceRNA interactions connecting genes that show opposite trends in expression change are preferentially dysregulated. This targeted dysregulation is responsible for the majority of the observed expression changes in genes with significant * These authors ceRNA dysregulation and represents a novel mechanism underlying aberrant oncogenic expression.
    [Show full text]