Epigenetic Regulation of Neurogenesis by Citron Kinase Matthew Irg Genti University of Connecticut - Storrs, [email protected]

Total Page:16

File Type:pdf, Size:1020Kb

Epigenetic Regulation of Neurogenesis by Citron Kinase Matthew Irg Genti University of Connecticut - Storrs, Matthew.Girgenti@Gmail.Com University of Connecticut OpenCommons@UConn Doctoral Dissertations University of Connecticut Graduate School 12-10-2015 Epigenetic Regulation of Neurogenesis By Citron Kinase Matthew irG genti University of Connecticut - Storrs, [email protected] Follow this and additional works at: https://opencommons.uconn.edu/dissertations Recommended Citation Girgenti, Matthew, "Epigenetic Regulation of Neurogenesis By Citron Kinase" (2015). Doctoral Dissertations. 933. https://opencommons.uconn.edu/dissertations/933 Epigenetic Regulation Of Neurogenesis By Citron Kinase Matthew J. Girgenti, Ph.D. University of Connecticut, 2015 Patterns of neural progenitor division are controlled by a combination of asymmetries in cell division, extracellular signals, and changes in gene expression programs. In a screen for proteins that complex with citron kinase (CitK), a protein essential to cell division in developing brain, I identified the histone methyltransferase- euchromatic histone-lysine N-methyltransferse 2 (G9a). CitK is present in the nucleus and binds to positions in the genome clustered around transcription start sites of genes involved in neuronal development and differentiation. CitK and G9a co-occupy these genomic positions in S/G2-phase of the cell cycle in rat neural progenitors, and CitK functions with G9a to repress gene expression in several developmentally important genes including CDKN1a, H2afz, and Pou3f2/Brn2. The study indicates a novel function for citron kinase in gene repression, and contributes additional evidence to the hypothesis that mechanisms that coordinate gene expression states are directly linked to mechanisms that regulate cell division in the developing nervous system. Epigenetic Regulation Of Neurogenesis By Citron Kinase Matthew J. Girgenti B.S., Fairfield University, 2002 M.S., Southern Connecticut State University, 2004 A Dissertation Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy at the University of Connecticut 2015 ii Copyright by Matthew J. Girgenti 2015 iii APPROVAL PAGE Doctor of Philosophy Dissertation Epigenetic Regulation Of Neurogenesis By Citron Kinase Presented by Matthew J. Girgenti, B.S., M.S. Major Advisor:_____________________________________________________ Kent Holsinger, Ph.D. Associate Advisor:_________________________________________________ Charles Giardina, Ph.D. Associate Advisor:_________________________________________________ Stormy Chamberlain, Ph.D. Associate Advisor:_________________________________________________ James Li, Ph.D. University of Connecticut, 2015 iv This work is dedicated to my family. To my parents Jack and Eileen Girgenti for instilling in me a love of learning and to my wife Alicia Che for all of her love, encouragement, and support. v ACKNOWLEDGMENTS I would like to recognize those who aided me during the course of my dissertation work. I would like to thank my dissertation committee: Dr. Kent Holsinger, Dr. Charles Giardina, Dr. Stormy Chamberlain, and Dr. James Li for their insightful comments and hard questions. I would especially like to thank Dr. Jim Wohl, whose tireless dedication as Ombudsman and brilliant advice guided me through the toughest times in graduate school. I would also like to thank all of my undergraduates without whom this dissertation may never have been completed: Nicholas Heliotis, Amanda Collins, Christine Dougherty, Palak Shelat, Faseeha Altaf, Aditya Makol, and Shan Parikh. A special note of thanks to Dr. Aarti Tarkar for imparting with me her friendship and expertise in all things western blotting. Last but certainly not least, I would like to thank my family: my parents Jack and Eileen Girgenti for all of their support throughout my education; my brother Mark Girgenti, for all of our insightful discussions about mass spectrometry. And finally to my beautiful wife Alicia Che, whose brilliance never ceases to awe and inspire me both at home and in the lab. Matthew J. Girgenti University of Connecticut vi TABLE OF CONTENTS Page LIST OF TABLES ............................................................................................... viii LIST OF FIGURES ............................................................................................... ix CHAPTER 1. INTRODUCTION ............................................................................ 1 Gene Expression States throughout Neurogenesis .............................................. 1 Citron Kinase and Neurogenesis .......................................................................... 3 Epigenetics and Development .............................................................................. 5 Histone Methylation ............................................................................................... 7 The Histone Methyltransferase G9a ..................................................................... 8 G9a Inhibitors ...................................................................................................... 10 Dissertation Organization .................................................................................... 11 CHAPTER 2. Citron Kinase protein is nuclear in neural progenitor cells and interacts with histone methyltransferase G9a .............................. 13 Abstract ............................................................................................................... 13 Introduction ......................................................................................................... 13 Materials and Methods ........................................................................................ 15 Results ................................................................................................................ 20 CitK is present in the nucleus of neural progenitors but masked by fixation ................................................................................... 20 CitK interacts with the histone methyltransferase G9a in neural progenitors ............................................................................... 21 Discussion ........................................................................................................... 30 CHAPTER 3. Chromatin-associated Citron Kinase Regulates Gene Expression in Rat Neural Progenitor Cells .................................................... 34 Abstract ............................................................................................................... 34 Introduction ......................................................................................................... 35 Materials and Methods ........................................................................................ 37 Results ................................................................................................................ 42 Identification of CitK genomic binding sites by ChIP-seq .................................... 42 Citron kinase regulates expression of thousands of genes ................................. 44 Citron kinase is required for G9a action but no localization to target genes ....... 46 Citron kinase and G9a co-localize to target genomic loci in S-phase ................. 48 Citron kinase function requires interaction with G9a ........................................... 49 Discussion ........................................................................................................... 70 vii CHAPTER 4. Assessing Citron Kinase’s ability to phosphorylate G9a In Rat Neural Progenitor Cells ......................................................................... 73 Abstract ............................................................................................................... 73 Introduction ......................................................................................................... 74 Materials and Methods ........................................................................................ 75 Results ................................................................................................................ 79 G9a phosphorylation levels are normal in mutant neural progenitor cells .......... 79 Citron kinase phosphorylates G9a in vitro .......................................................... 80 Phosphomimetic rescue in Citron kinase deficient neural progenitors ................ 81 Citron kinase does not phosphorylate G9a in NPs or HEK293T cells ................ 83 Discussion ........................................................................................................... 87 CHAPTER 5. Summary and Conclusion ........................................................ 90 Final Remarks ..................................................................................................... 94 APPENDIX A: LIST OF ABBREVIATIONS ...................................................... 95 APPENDIX B: PRIMERS ................................................................................... 98 B.1: Exon spanning primers used for real-time PCR of regulated transcripts ....................................................................................... 98 B.2: Promoter primers used for real-time PCR of CitK target genomic loci ............................................................................... 101 APPENDIX C: CitK Genomic Occupancy in Neural Progenitors ............... 106 APPENDIX D: Mass spectrometry Phospo-Peptides .................................. 129 D.1: In vitro Kinase Assay Phospho-peptides .................................................
Recommended publications
  • 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).
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [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]
  • PDF Hosted at the Radboud Repository of the Radboud University Nijmegen
    PDF hosted at the Radboud Repository of the Radboud University Nijmegen The following full text is a publisher's version. For additional information about this publication click this link. http://hdl.handle.net/2066/48948 Please be advised that this information was generated on 2021-09-28 and may be subject to change. MOLECULAR AND CELLULAR BIOLOGY, Feb. 2005, p. 1402–1414 Vol. 25, No. 4 0270-7306/05/$08.00ϩ0 doi:10.1128/MCB.25.4.1402–1414.2005 Copyright © 2005, American Society for Microbiology. All Rights Reserved. Divergent Mitochondrial and Endoplasmic Reticulum Association of DMPK Splice Isoforms Depends on Unique Sequence Arrangements in Tail Anchors† Rene´ E. M. A. van Herpen,1 Ralph J. A. Oude Ophuis,1 Mietske Wijers,1 Miranda B. Bennink,2 Fons A. J. van de Loo,2 Jack Fransen,1 Be´ Wieringa,1* and Derick G. Wansink1 Department of Cell Biology1 and Rheumatology Research and Advanced Therapeutics,2 Nijmegen Center for Molecular Life Sciences, University Medical Center, Nijmegen, The Netherlands Received 3 September 2004/Returned for modification 23 September 2004/Accepted 10 November 2004 Myotonic dystrophy protein kinase (DMPK) is a Ser/Thr-type protein kinase with unknown function, originally identified as the product of the gene that is mutated by triplet repeat expansion in patients with myotonic dystrophy type 1 (DM1). Alternative splicing of DMPK transcripts results in multiple protein isoforms carrying distinct C termini. Here, we demonstrate by expressing individual DMPKs in various cell ؊/؊ types, including C2C12 and DMPK myoblast cells, that unique sequence arrangements in these tails control the specificity of anchoring into intracellular membranes.
    [Show full text]
  • Kinase Profiling Book
    Custom and Pre-Selected Kinase Prof iling to f it your Budget and Needs! As of July 1, 2021 19.8653 mm 128 196 12 Tyrosine Serine/Threonine Lipid Kinases Kinases Kinases Carna Biosciences, Inc. 2007 Carna Biosciences, Inc. Profiling Assays available from Carna Biosciences, Inc. As of July 1, 2021 Page Kinase Name Assay Platform Page Kinase Name Assay Platform 4 ABL(ABL1) MSA 21 EGFR[T790M/C797S/L858R] MSA 4 ABL(ABL1)[E255K] MSA 21 EGFR[T790M/L858R] MSA 4 ABL(ABL1)[T315I] MSA 21 EPHA1 MSA 4 ACK(TNK2) MSA 21 EPHA2 MSA 4 AKT1 MSA 21 EPHA3 MSA 5 AKT2 MSA 22 EPHA4 MSA 5 AKT3 MSA 22 EPHA5 MSA 5 ALK MSA 22 EPHA6 MSA 5 ALK[C1156Y] MSA 22 EPHA7 MSA 5 ALK[F1174L] MSA 22 EPHA8 MSA 6 ALK[G1202R] MSA 23 EPHB1 MSA 6 ALK[G1269A] MSA 23 EPHB2 MSA 6 ALK[L1196M] MSA 23 EPHB3 MSA 6 ALK[R1275Q] MSA 23 EPHB4 MSA 6 ALK[T1151_L1152insT] MSA 23 Erk1(MAPK3) MSA 7 EML4-ALK MSA 24 Erk2(MAPK1) MSA 7 NPM1-ALK MSA 24 Erk5(MAPK7) MSA 7 AMPKα1/β1/γ1(PRKAA1/B1/G1) MSA 24 FAK(PTK2) MSA 7 AMPKα2/β1/γ1(PRKAA2/B1/G1) MSA 24 FER MSA 7 ARG(ABL2) MSA 24 FES MSA 8 AurA(AURKA) MSA 25 FGFR1 MSA 8 AurA(AURKA)/TPX2 MSA 25 FGFR1[V561M] MSA 8 AurB(AURKB)/INCENP MSA 25 FGFR2 MSA 8 AurC(AURKC) MSA 25 FGFR2[V564I] MSA 8 AXL MSA 25 FGFR3 MSA 9 BLK MSA 26 FGFR3[K650E] MSA 9 BMX MSA 26 FGFR3[K650M] MSA 9 BRK(PTK6) MSA 26 FGFR3[V555L] MSA 9 BRSK1 MSA 26 FGFR3[V555M] MSA 9 BRSK2 MSA 26 FGFR4 MSA 10 BTK MSA 27 FGFR4[N535K] MSA 10 BTK[C481S] MSA 27 FGFR4[V550E] MSA 10 BUB1/BUB3 MSA 27 FGFR4[V550L] MSA 10 CaMK1α(CAMK1) MSA 27 FGR MSA 10 CaMK1δ(CAMK1D) MSA 27 FLT1 MSA 11 CaMK2α(CAMK2A) MSA 28
    [Show full text]
  • Testing for Parallel Genomic and Epigenomic Footprints of Adaptation to Urban Life in a Passerine Bird
    SUPPLEMENTARY INFORMATION Testing for parallel genomic and epigenomic footprints of adaptation to urban life in a passerine bird Authors: Aude E. Caizergues1*, Jeremy Le Luyer2, Arnaud Grégoire1, Marta Szulkin3, Juan-Carlos Señar4, Anne Charmantier1†, Charles Perrier5† 1 CEFE, Univ Montpellier, CNRS, Univ Paul Valéry Montpellier 3, EPHE, IRD, Montpellier, France 2 Ifremer, UMR EIO 241, Centre du Pacifique, Taravao, Tahiti, Polynésie française, France 3 Centre of New Technologies, University of Warsaw, S. Banacha 2c, 02-097 Warsaw, Poland 4 Museu de Ciències Naturals de Barcelona, Parc Ciutadella, 08003 Barcelona, Spain 5 CBGP, INRAe, CIRAD, IRD, Montpellier SupAgro, Univ. Montpellier, Montpellier, France † shared senior authorship * Corresponding author: Aude E. Caizergues, 1919 route de Mende, 34293 Montpellier cedex 5, FRANCE Email : [email protected] SUPPLEMENTARY TABLES Table S1: Redundancy analysis (RDA) performed on the genetic data including the Z chromosome. adjusted R- P-value RDA1 RDA2 RDA3 RDA4 squared % variance explained by axes Full RDA 0.018 0.001 0.024 0.021 0.021 0.019 Variables Biplot scores City – Montpellier 0.57 -0.819 0.059 -0.039 0.001 City – Warsaw 0.381 0.883 0.214 0.17 Habitat – Urban 0.001 -0.21 -0.1 0.968 -0.089 Sex – Male 0.004 0.184 0.143 -0.0358 -0.972 % variance explained by axe Partial RDA for city 0.012 0.001 0.025 0.022 Variable Biplot scores City – Montpellier 0.63 -0.776 0.001 City – Warsaw 0.356 0.934 Partial RDA for % variance explained by axe 0.004 0.001 habitat 0.022 Variable Biplot score Habitat – Urban 0.001 0.999 % variance explained by axe Partial RDA for sex 0.002 0.004 0.02 Variable Biplot score Sex – Male 0.005 -0.999 Table S2: Redundancy analysis (RDA) performed on the genetic data without Z chromosome.
    [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]
  • Lenalidomide Induces Cell Death in an MDS-Derived Cell Line with Deletion of Chromosome 5Q by Inhibition of Cytokinesis
    Leukemia (2010) 24, 748–755 & 2010 Macmillan Publishers Limited All rights reserved 0887-6924/10 $32.00 www.nature.com/leu ORIGINAL ARTICLE Lenalidomide induces cell death in an MDS-derived cell line with deletion of chromosome 5q by inhibition of cytokinesis A Matsuoka1, A Tochigi1, M Kishimoto1, T Nakahara1, T Kondo1, T Tsujioka1, T Tasaka1, Y Tohyama2 and K Tohyama1 1Department of Laboratory Medicine, Kawasaki Medical School, Okayama, Japan; and 2Division of Biochemistry, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Hyogo, Japan Myelodysplastic syndromes (MDS) are a group of hematopoietic higher-risk groups with del(5q) that are susceptible to leukemic stem cell disorders characterized by refractory cytopenias transformation.6 Several hematopoiesis-related genes and tumor and susceptibility to leukemic transformation. On a subset of suppressor genes are located at 5q locus, and SPARC,7 MDS patients with deletion of the long arm of chromosome5 8 9 10 11 (del(5q)), lenalidomide exerts hematological and cytogenetic CTNNA1, EGR1, RPS14 and CDC25C are reported as effects, but the underlying pharmacological mechanisms are the candidate genes of MDS with del(5q) or 5q- syndrome. not fully understood. In this study, we have investigated the Lenalidomide, a derivative of thalidomide, is shown to in vitro effects of lenalidomide on an MDS-derived cell line, exert plenty of biological actions including inhibition of MDS-L, which carries del(5q) and complex chromosome angiogenesis,12 suppression of proinflammatory cytokine abnormalities. We found that the growth of MDS-L cells was production such as TNF-a,13 enhancement of T- and NK-cell specifically suppressed mainly by apoptosis, and in addition, 14–16 multinucleated cells were frequently formed and finally died activation.
    [Show full text]
  • PDF Hosted at the Radboud Repository of the Radboud University Nijmegen
    PDF hosted at the Radboud Repository of the Radboud University Nijmegen The following full text is a publisher's version. For additional information about this publication click this link. http://hdl.handle.net/2066/85871 Please be advised that this information was generated on 2021-09-29 and may be subject to change. ISOFORMS IN MUSCLE AND BRAIN CELLS localization and function • ralph j.a. oude ophuis • 2011 9 789088 912344 > ISBN 978-90-8891234,-4 DMPK ISOFORMS IN MUSCLE AND BRAIN CELLS LOCALIZATION AND FUNCTION Voor het bijwonen van de openbare verdediging van het proefschrift van RALPH J.A. OUDE OPHUIS DMPK ISOFORMS IN MUSCLE AND BRAIN CELLS LOCALIZATION AND FUNCTION op vrijdag 1 april 2011 om 13:00u precies in de Aula van de Radboud Universiteit Nijmegen aan de Comeniuslaan 2 te Nijmegen Na afloop van de verdediging is er een receptie ter plaatse PARANIMFEN Susan Mulders [email protected] Rinske van de Vorstenbosch r.vandevorstenbosch(§) ncmls.ru.nl DMPK ISOFORMS IN MUSCLE AND BRAIN CELLS LOCALIZATION AND FUNCTION ISBN-13 978-90-8891234-4 ISBN-10 90-8891-234-3 Printed by Proefsohriftmaken.nl || Printyourthesis.com Published by Uitgeverij BOXPress, Oisterwijk DMPK ISOFORMS IN MUSCLE AND BRAIN CELLS LOCALIZATION AND FUNCTION Een wetenschappelijke proeve op het gebied van de Medische Wetenschappen Proefschrift ter verkrijging van de graad van doctor aan de Radboud Universiteit Nijmegen op gezag van de rector magnificus prof. mr. S.C.J.J. Kortmann, volgens besluit van het college van decanen in het openbaar te verdedigen op vrijdag 1 april 2011 om 13:00 uur precies door Raphaël Johannes Antonius Oude Ophuis geboren op 24 oktober 1978 te Sint-Oedenrode Promotor Prof.
    [Show full text]
  • Regulation of P27kip1 and P57kip2 Functions by Natural Polyphenols
    biomolecules Review Regulation of p27Kip1 and p57Kip2 Functions by Natural Polyphenols Gian Luigi Russo 1,* , Emanuela Stampone 2 , Carmen Cervellera 1 and Adriana Borriello 2,* 1 National Research Council, Institute of Food Sciences, 83100 Avellino, Italy; [email protected] 2 Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 81031 Napoli, Italy; [email protected] * Correspondence: [email protected] (G.L.R.); [email protected] (A.B.); Tel.: +39-0825-299-331 (G.L.R.) Received: 31 July 2020; Accepted: 9 September 2020; Published: 13 September 2020 Abstract: In numerous instances, the fate of a single cell not only represents its peculiar outcome but also contributes to the overall status of an organism. In turn, the cell division cycle and its control strongly influence cell destiny, playing a critical role in targeting it towards a specific phenotype. Several factors participate in the control of growth, and among them, p27Kip1 and p57Kip2, two proteins modulating various transitions of the cell cycle, appear to play key functions. In this review, the major features of p27 and p57 will be described, focusing, in particular, on their recently identified roles not directly correlated with cell cycle modulation. Then, their possible roles as molecular effectors of polyphenols’ activities will be discussed. Polyphenols represent a large family of natural bioactive molecules that have been demonstrated to exhibit promising protective activities against several human diseases. Their use has also been proposed in association with classical therapies for improving their clinical effects and for diminishing their negative side activities. The importance of p27Kip1 and p57Kip2 in polyphenols’ cellular effects will be discussed with the aim of identifying novel therapeutic strategies for the treatment of important human diseases, such as cancers, characterized by an altered control of growth.
    [Show full text]
  • Kinase-Targeted Cancer Therapies: Progress, Challenges and Future Directions Khushwant S
    Bhullar et al. Molecular Cancer (2018) 17:48 https://doi.org/10.1186/s12943-018-0804-2 REVIEW Open Access Kinase-targeted cancer therapies: progress, challenges and future directions Khushwant S. Bhullar1, Naiara Orrego Lagarón2, Eileen M. McGowan3, Indu Parmar4, Amitabh Jha5, Basil P. Hubbard1 and H. P. Vasantha Rupasinghe6,7* Abstract The human genome encodes 538 protein kinases that transfer a γ-phosphate group from ATP to serine, threonine, or tyrosine residues. Many of these kinases are associated with human cancer initiation and progression. The recent development of small-molecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy. Significantly, protein kinases are the second most targeted group of drug targets, after the G-protein- coupled receptors. Since the development of the first protein kinase inhibitor, in the early 1980s, 37 kinase inhibitors have received FDA approval for treatment of malignancies such as breast and lung cancer. Furthermore, about 150 kinase-targeted drugs are in clinical phase trials, and many kinase-specific inhibitors are in the preclinical stage of drug development. Nevertheless, many factors confound the clinical efficacy of these molecules. Specific tumor genetics, tumor microenvironment, drug resistance, and pharmacogenomics determine how useful a compound will be in the treatment of a given cancer. This review provides an overview of kinase-targeted drug discovery and development in relation to oncology and highlights the challenges and future potential for kinase-targeted cancer therapies. Keywords: Kinases, Kinase inhibition, Small-molecule drugs, Cancer, Oncology Background Recent advances in our understanding of the fundamen- Kinases are enzymes that transfer a phosphate group to a tal molecular mechanisms underlying cancer cell signaling protein while phosphatases remove a phosphate group have elucidated a crucial role for kinases in the carcino- from protein.
    [Show full text]