Transient Abnormal Myelopoiesis and AML in Down Syndrome: an Update

Total Page:16

File Type:pdf, Size:1020Kb

Transient Abnormal Myelopoiesis and AML in Down Syndrome: an Update Curr Hematol Malig Rep DOI 10.1007/s11899-016-0338-x MYELOPROLIFERATIVE DISORDERS (C HARRISON, SECTION EDITOR) Transient Abnormal Myelopoiesis and AML in Down Syndrome: an Update Neha Bhatnagar1 & Laure Nizery2 & Oliver Tunstall3 & Paresh Vyas4 & Irene Roberts5 # The Author(s) 2016. This article is published with open access at Springerlink.com Abstract Children with constitutional trisomy 21 (Down Introduction syndrome (DS)) have a unique predisposition to develop my- eloid leukaemia of Down syndrome (ML-DS). This disorder Population studies show that children with Down syndrome is preceded by a transient neonatal preleukaemic syndrome, due to constitutional trisomy 21 have a markedly increased transient abnormal myelopoiesis (TAM). TAM and ML-DS risk of developing acute leukaemia compared with children are caused by co-operation between trisomy 21, which itself without Down syndrome [1]. Both myeloid leukaemia, known perturbs fetal haematopoiesis and acquired mutations in the as myeloid leukaemia of Down syndrome (ML-DS), and acute key haematopoietic transcription factor gene GATA1.These lymphoblastic leukaemia are increased by 150- and ∼30-fold, mutations are found in almost one third of DS neonates and respectively [1, 2]. ML-DS has a distinct natural history and are frequently clinically and haematologcially ‘silent’.While clinical and biological features (reviewed in [3, 4]). It virtually the majority of cases of TAM undergo spontaneous remission, always develops before the age of 5 years, and the acute leu- ∼10 % will progress to ML-DS by acquiring transforming kaemia is preceded by a clonal neonatal preleukaemic syn- mutations in additional oncogenes. Recent advances in the drome known as transient abnormal myelopoiesis (TAM) that unique biological, cytogenetic and molecular characteristics is unique to Down syndrome [3, 4]. of TAM and ML-DS are reviewed here. TAM is characterised by increased circulating blast cells that harbour acquired N-terminal truncating mutations in the key haematopoietic transcription factor gene GATA1 [5–10]. Keywords Transient abnormal myelopoiesis . Down Around 10–15%ofneonateswithDownsyndromehavea syndrome . Acute leukaemia . Myeloproliferative disorders diagnosis of TAM with blasts >10 % and typical clinical fea- This article is part of the Topical Collection on Myeloproliferative Disorders * Irene Roberts 1 Children’s Hospital, John Radcliffe Hospital, Oxford University [email protected] Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK Neha Bhatnagar 2 Paediatric Intensive Care Unit, Robert Debré Hospital, 48 Boulevard [email protected] Sérurier, 75019 Paris, France Laure Nizery 3 Bristol Royal Hospital for Children, Paul O’Gorman Building, Upper [email protected] Maudlin St, Bristol BS2 8B, UK Oliver Tunstall 4 Molecular Haematology Unit, Weatherall Institute of Molecular [email protected] Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK Paresh Vyas [email protected] 5 Department of Paediatrics, Children’s Hospital, University of Oxford, John Radcliffe Hospital, OX3 9DU Oxford, UK Curr Hematol Malig Rep tures that require close monitoring in the neonatal period since particular ERG and DYRK1a, as important mediators of the mortality rate may be up to 20 %. A further 10–15 % of the abnormal megakaryopoiesis, although this does not neonates with Down syndrome have one or more acquired seem to be sufficient to cause leukaemia in trisomic or GATA1 mutations in association with a low number of circu- disomic mouse models even when co-expressed with an lating blast cells (<10 %) and have clinically and N-terminally truncated GATA1 gene. Interestingly, recent haematologically silent disease (silent TAM) [11••]. In the data using a panel of iPSC lines, suggest that trisomy of majority of cases of TAM and silent TAM, the GATA1 mutant RUNX1, ETS2,andERG might be sufficient, in combina- clone goes into complete and permanent remission without the tion with mutant GATA1, to explain many of the need for chemotherapy. However, 10–20 % of neonates with haematopoietic abnormalities seen in primary human fetal TAM and silent TAM subsequently develop ML-DS in the liver and TAM cells. Despite these interesting findings, it first 5 years of life when persistent GATA1 mutant cells ac- is now clear that trisomy 21 causes genome-wide changes quire additional oncogenic mutations, most often in cohesin or in gene expression directly or indirectly affecting multiple epigenetic regulator genes [12••, 13]. This review article dis- genes on most chromosomes [23]. cusses the recent clinical and biological advances in TAM and (ii) N-terminal truncating GATA1 mutations in TAM and ML-DS and how these may impact on clinical management. ML-DS The link between acquired mutations in the GATA1 Cellular and Molecular Pathogenesis of TAM and ML-DS gene and ML-DS was first identified more than 12 years ago in John Crispino’slab[5] and rapidly followed by The cellular and molecular events involved in initiation and studies from a number of groups confirming the link with evolution of TAM and ML-DS can best be understood as a trisomy 21 as well as showing the same N-terminal trun- three-step model which requires the presence within a fetal cating mutations in TAM [6–10]. The GATA1 mutations liver-derived haematopoietic stem or progenitor cell of (i) tri- that can be detected in all cases disappear when TAM somy 21, (ii) an acquired GATA1 mutation, and (iii) at least (or ML-DS) enter remission indicating that these are ac- one additional oncogenic mutation (Fig. 1). quired events [9, 12••]. Application of highly sensitive next-generation sequencing (NGS)-based methodology (i) Perturbation of fetal haematopoiesis by trisomy 21 has recently shown that GATA1 mutations are present The initial event in trisomy 21-associated preleukaemic in all cases of TAM or ML-DS and that they are present and leukaemic conditions is the perturbation of fetal in 25–30 % of all neonates with Down syndrome [11••]. haematopoiesis by trisomy 21 itself. It is known that by This means that GATA1 mutations are necessary for the late in the first trimester of fetal life, haematopoiesis in the development of TAM/ML-DS, that they are acquired pri- liver is abnormal in fetuses with trisomy 21 and that these or to birth in fetal cells and that they occur at an aston- changes precede the acquisition of GATA1 mutations ishingly high frequency. It seems likely that acquisition [14, 15••]. Specifically, trisomy 21 causes an increase in of such mutations confers a selective growth advantage the numbers of megakaryocyte-erythroid progenitors to these cells during fetal life. The presence of multiple (MEP) and an increase in the size and characteristics of GATA1 mutant clones in up to 25 % of neonates with the immunophenotypic haematopoietic stem cell (HSC) Down syndrome is consistent with this [9, 11••]; howev- compartment [15••]. HSC and multipotent myeloid pro- er, the reason for their high frequency in Down syndrome genitors in trisomy 21 fetal liver proliferate more in vitro remains unknown (these mutations are not found in compared with normal fetal liver at the same stage of de- normal, disomic cord blood) [9, 11••]. velopment and have increased erythroid-megakaryocyte The vast majority of acquired GATA1 mutations output and gene expression [15••]. Despite the increase (∼97 %) are found in exon 2 and the remainder in exon in megakaryocytes (MK) in trisomy 21 fetal liver, 3.1 of the GATA1 gene, including insertions, deletions MK differentiation is impaired and platelet counts are re- and point mutations [10]. These mutations lead to ex- duced both in fetal blood and in neonates with Down pression of a truncated GATA1s protein [5, 6] and the syndrome suggesting that trisomy 21 itself causes type of GATA1 mutation does not predict which patients dysmegakaryopoiesis [11••, 14, 15••]. with TAM will later progress to ML-DS [10]. Since the The molecular basis for these dramatic changes in fetal GATA1 gene is on the X chromosome, haematopoietic erythro-megakaryopoiesis is not yet clear. Some, but not cells harbouring GATA1 mutations express only GATA1s all, of the features can be recapitulated either in elegant and no longer have the ability to produce the full-length animal models [16–20] or in studies in human embryonic GATA1 protein [5]. The main physiological role of stem cells (ESC) and induced pluripotent stem cells (iPSC) GATA1 is as a regulator of normal megakaryocyte and [21, 22]. Together, these studies have implicated increased erythroid differentiation [24]. How GATA1 transforms expression of various genes on chromosome 21, in trisomy 21 fetal haematopoietic cells is unclear. Forced Curr Hematol Malig Rep Fig. 1 Natural history and pathogenesis of TAM and ML-DS. Schematic of megakaryocyte-erythroid progenitors (MEP) and megakaryocytes. These representation of molecular, biological and clinical data, indicating that cells subsequently acquire N-terminal truncating GATA1 mutations resulting transient abnormal myelopoiesis (TAM) and myeloid leukaemia of Down in TAM in late fetal or early neonatal life. Although most cases of TAM syndrome (ML-DS) are initiated before birth when fetal liver spontaneously and permanently remit (∼90 %) by the age of 6 months, in haematopoietic stem and progenitor cells (HSPC)trisomicfor ∼10 % of cases, additional genetic/epigenetic events lead to further clonal chromosome 21 demonstrate perturbed haematopoiesis with an expansion expansion resulting
Recommended publications
  • Aberrant Methylation Underlies Insulin Gene Expression in Human Insulinoma
    ARTICLE https://doi.org/10.1038/s41467-020-18839-1 OPEN Aberrant methylation underlies insulin gene expression in human insulinoma Esra Karakose1,6, Huan Wang 2,6, William Inabnet1, Rajesh V. Thakker 3, Steven Libutti4, Gustavo Fernandez-Ranvier 1, Hyunsuk Suh1, Mark Stevenson 3, Yayoi Kinoshita1, Michael Donovan1, Yevgeniy Antipin1,2, Yan Li5, Xiaoxiao Liu 5, Fulai Jin 5, Peng Wang 1, Andrew Uzilov 1,2, ✉ Carmen Argmann 1, Eric E. Schadt 1,2, Andrew F. Stewart 1,7 , Donald K. Scott 1,7 & Luca Lambertini 1,6 1234567890():,; Human insulinomas are rare, benign, slowly proliferating, insulin-producing beta cell tumors that provide a molecular “recipe” or “roadmap” for pathways that control human beta cell regeneration. An earlier study revealed abnormal methylation in the imprinted p15.5-p15.4 region of chromosome 11, known to be abnormally methylated in another disorder of expanded beta cell mass and function: the focal variant of congenital hyperinsulinism. Here, we compare deep DNA methylome sequencing on 19 human insulinomas, and five sets of normal beta cells. We find a remarkably consistent, abnormal methylation pattern in insu- linomas. The findings suggest that abnormal insulin (INS) promoter methylation and altered transcription factor expression create alternative drivers of INS expression, replacing cano- nical PDX1-driven beta cell specification with a pathological, looping, distal enhancer-based form of transcriptional regulation. Finally, NFaT transcription factors, rather than the cano- nical PDX1 enhancer complex, are predicted to drive INS transactivation. 1 From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
    [Show full text]
  • Insights Into Hp1a-Chromatin Interactions
    cells Review Insights into HP1a-Chromatin Interactions Silvia Meyer-Nava , Victor E. Nieto-Caballero, Mario Zurita and Viviana Valadez-Graham * Instituto de Biotecnología, Departamento de Genética del Desarrollo y Fisiología Molecular, Universidad Nacional Autónoma de México, Cuernavaca Morelos 62210, Mexico; [email protected] (S.M.-N.); [email protected] (V.E.N.-C.); [email protected] (M.Z.) * Correspondence: [email protected]; Tel.: +527773291631 Received: 26 June 2020; Accepted: 21 July 2020; Published: 9 August 2020 Abstract: Understanding the packaging of DNA into chromatin has become a crucial aspect in the study of gene regulatory mechanisms. Heterochromatin establishment and maintenance dynamics have emerged as some of the main features involved in genome stability, cellular development, and diseases. The most extensively studied heterochromatin protein is HP1a. This protein has two main domains, namely the chromoshadow and the chromodomain, separated by a hinge region. Over the years, several works have taken on the task of identifying HP1a partners using different strategies. In this review, we focus on describing these interactions and the possible complexes and subcomplexes associated with this critical protein. Characterization of these complexes will help us to clearly understand the implications of the interactions of HP1a in heterochromatin maintenance, heterochromatin dynamics, and heterochromatin’s direct relationship to gene regulation and chromatin organization. Keywords: heterochromatin; HP1a; genome stability 1. Introduction Chromatin is a complex of DNA and associated proteins in which the genetic material is packed in the interior of the nucleus of eukaryotic cells [1]. To organize this highly compact structure, two categories of proteins are needed: histones [2] and accessory proteins, such as chromatin regulators and histone-modifying proteins.
    [Show full text]
  • Suppression of RAD21 Gene Expression Decreases Cell Growth and Enhances Cytotoxicity of Etoposide and Bleomycin in Human Breast Cancer Cells
    Molecular Cancer Therapeutics 361 Suppression of RAD21 gene expression decreases cell growth and enhances cytotoxicity of etoposide and bleomycin in human breast cancer cells Josephine M. Atienza,1 Richard B. Roth,1 Introduction 1 1 Caridad Rosette, Kevin J. Smylie, The RAD21 gene codes for a human homologue of Stefan Kammerer,1 Joachim Rehbock,2 1 1 Saccharomyces pombe Rad21 protein. The current knowledge Jonas Ekblom, and Mikhail F. Denissenko about this protein points to a role in modulation of cell 1Sequenom, Inc., San Diego, California and 2Frauena¨rzte growth and in cell defense against DNA damage, both Rosenstrasse, Munich, Germany processes being central to carcinogenesis. Several DNA repair genes including rad21 were initially identified in the fission yeast S. pombe as radiation-sensitive mutants (1, 2). Abstract Specifically, Rad21 has been implicated in homologous A genome-wide case-control association study done in our recombination–mediated double-strand break (DSB) re- laboratory has identified a single nucleotide polymorphism pair, and is unique among the radiation response genes in located in RAD21 as being significantly associated with that it also plays a role in cell cycle regulation (3, 4). Yeast breast cancer susceptibility. RAD21 is believed to function Rad21 and its mammalian homologue were subsequently in sister chromatid alignment as part of the cohesin complex identified as components of a conserved cohesin complex and also in double-strand break (DSB) repair. Following our (5, 6), which is believed to function in aligning sister initial finding, expression studies revealed a 1.25- to 2.5- chromatids during the early stages of cellular division.
    [Show full text]
  • Mutational Inactivation of STAG2 Causes Aneuploidy in Human Cancer
    REPORTS mean difference for all rubric score elements was ing becomes a more commonly supported facet 18. C. L. Townsend, E. Heit, Mem. Cognit. 39, 204 (2011). rejected. Univariate statistical tests of the observed of STEM graduate education then students’ in- 19. D. F. Feldon, M. Maher, B. Timmerman, Science 329, 282 (2010). mean differences between the teaching-and- structional training and experiences would alle- 20. B. Timmerman et al., Assess. Eval. High. Educ. 36,509 research and research-only conditions indicated viate persistent concerns that current programs (2011). significant results for the rubric score elements underprepare future STEM faculty to perform 21. No outcome differences were detected as a function of “testability of hypotheses” [mean difference = their teaching responsibilities (28, 29). the type of teaching experience (TA or GK-12) within the P sample population participating in both research and 0.272, = 0.006; CI = (.106, 0.526)] with the null teaching. hypothesis rejected in 99.3% of generated data References and Notes 22. Materials and methods are available as supporting samples (Fig. 1) and “research/experimental de- 1. W. A. Anderson et al., Science 331, 152 (2011). material on Science Online. ” P 2. J. A. Bianchini, D. J. Whitney, T. D. Breton, B. A. Hilton-Brown, 23. R. L. Johnson, J. Penny, B. Gordon, Appl. Meas. Educ. 13, sign [mean difference = 0.317, = 0.002; CI = Sci. Educ. 86, 42 (2001). (.106, 0.522)] with the null hypothesis rejected in 121 (2000). 3. C. E. Brawner, R. M. Felder, R. Allen, R. Brent, 24. R. J. A. Little, J.
    [Show full text]
  • Correction of Cognitive Deficits in Mouse Models Of
    Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological inhibitor of DYRK1A Thu Lan Nguyen, Arnaud Duchon, Antigoni Manousopoulou, Nadège Loaëc, Benoît Villiers, Guillaume Pani, Meltem Karatas, Anna E Mechling, Laura-Adela Harsan, Emmanuelle Limanton, et al. To cite this version: Thu Lan Nguyen, Arnaud Duchon, Antigoni Manousopoulou, Nadège Loaëc, Benoît Villiers, et al.. Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological in- hibitor of DYRK1A. Disease Models & Mechanisms, Cambridge Company of Biologists, 2018, 11 (9), pp.dmm035634. 10.1242/dmm.035634. hal-01862465 HAL Id: hal-01862465 https://hal-univ-rennes1.archives-ouvertes.fr/hal-01862465 Submitted on 17 Jul 2019 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. © 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm035634. doi:10.1242/dmm.035634 RESEARCH ARTICLE Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological inhibitor of DYRK1A Thu Lan Nguyen1,2,3,4,5, Arnaud Duchon1,2,3,4, Antigoni Manousopoulou6, Nadegè Loaëc5, Benoît Villiers5, Guillaume Pani1,2,3,4, Meltem Karatas7,8, Anna E. Mechling8, Laura-Adela Harsan7,8, Emmanuelle Limanton9, Jean-Pierre Bazureau9, François Carreaux9, Spiros D.
    [Show full text]
  • The Mutational Landscape of Myeloid Leukaemia in Down Syndrome
    cancers Review The Mutational Landscape of Myeloid Leukaemia in Down Syndrome Carini Picardi Morais de Castro 1, Maria Cadefau 1,2 and Sergi Cuartero 1,2,* 1 Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916 Badalona, Spain; [email protected] (C.P.M.d.C); [email protected] (M.C.) 2 Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain * Correspondence: [email protected] Simple Summary: Leukaemia occurs when specific mutations promote aberrant transcriptional and proliferation programs, which drive uncontrolled cell division and inhibit the cell’s capacity to differentiate. In this review, we summarize the most frequent genetic lesions found in myeloid leukaemia of Down syndrome, a rare paediatric leukaemia specific to individuals with trisomy 21. The evolution of this disease follows a well-defined sequence of events and represents a unique model to understand how the ordered acquisition of mutations drives malignancy. Abstract: Children with Down syndrome (DS) are particularly prone to haematopoietic disorders. Paediatric myeloid malignancies in DS occur at an unusually high frequency and generally follow a well-defined stepwise clinical evolution. First, the acquisition of mutations in the GATA1 transcription factor gives rise to a transient myeloproliferative disorder (TMD) in DS newborns. While this condition spontaneously resolves in most cases, some clones can acquire additional mutations, which trigger myeloid leukaemia of Down syndrome (ML-DS). These secondary mutations are predominantly found in chromatin and epigenetic regulators—such as cohesin, CTCF or EZH2—and Citation: de Castro, C.P.M.; Cadefau, in signalling mediators of the JAK/STAT and RAS pathways.
    [Show full text]
  • Redundant and Specific Roles of Cohesin STAG Subunits in Chromatin Looping and Transcriptional Control
    Downloaded from genome.cshlp.org on October 10, 2021 - Published by Cold Spring Harbor Laboratory Press Research Redundant and specific roles of cohesin STAG subunits in chromatin looping and transcriptional control Valentina Casa,1,6 Macarena Moronta Gines,1,6 Eduardo Gade Gusmao,2,3,6 Johan A. Slotman,4 Anne Zirkel,2 Natasa Josipovic,2,3 Edwin Oole,5 Wilfred F.J. van IJcken,1,5 Adriaan B. Houtsmuller,4 Argyris Papantonis,2,3 and Kerstin S. Wendt1 1Department of Cell Biology, Erasmus MC, 3015 GD Rotterdam, The Netherlands; 2Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany; 3Institute of Pathology, University Medical Center, Georg-August University of Göttingen, 37075 Göttingen, Germany; 4Optical Imaging Centre, Erasmus MC, 3015 GD Rotterdam, The Netherlands; 5Center for Biomics, Erasmus MC, 3015 GD Rotterdam, The Netherlands Cohesin is a ring-shaped multiprotein complex that is crucial for 3D genome organization and transcriptional regulation during differentiation and development. It also confers sister chromatid cohesion and facilitates DNA damage repair. Besides its core subunits SMC3, SMC1A, and RAD21, cohesin in somatic cells contains one of two orthologous STAG sub- units, STAG1 or STAG2. How these variable subunits affect the function of the cohesin complex is still unclear. STAG1- and STAG2-cohesin were initially proposed to organize cohesion at telomeres and centromeres, respectively. Here, we uncover redundant and specific roles of STAG1 and STAG2 in gene regulation and chromatin looping using HCT116 cells with an auxin-inducible degron (AID) tag fused to either STAG1 or STAG2. Following rapid depletion of either subunit, we perform high-resolution Hi-C, gene expression, and sequential ChIP studies to show that STAG1 and STAG2 do not co-occupy in- dividual binding sites and have distinct ways by which they affect looping and gene expression.
    [Show full text]
  • RAD21 Gene RAD21 Cohesin Complex Component
    RAD21 gene RAD21 cohesin complex component Normal Function The RAD21 gene provides instructions for making a protein that is involved in regulating the structure and organization of chromosomes during cell division. Before cells divide, they must copy all of their chromosomes. The copied DNA from each chromosome is arranged into two identical structures, called sister chromatids, which are attached to one another during the early stages of cell division. The RAD21 protein is part of a protein group called the cohesin complex that holds the sister chromatids together. Researchers believe that the RAD21 protein, as a structural component of the cohesin complex, also plays important roles in stabilizing cells' genetic information, repairing damaged DNA, and regulating the activity of certain genes that are essential for normal development. Health Conditions Related to Genetic Changes Cornelia de Lange syndrome At least eight mutations in the RAD21 gene have been identified in people with Cornelia de Lange syndrome, a developmental disorder that affects many parts of the body. Mutations in this gene appear to be an uncommon cause of this condition. Some cases of Cornelia de Lange syndrome have resulted from a deletion that removes a segment of DNA on chromosome 8 including the RAD21 gene. In these cases, the entire gene is missing from one copy of the chromosome in each cell, so cells produce a reduced amount of RAD21 protein. In other cases, the condition is caused by mutations within the RAD21 gene that impair or eliminate the function of the RAD21 protein. A defective or missing RAD21 protein likely alters the activity of the cohesin complex, impairing its ability to regulate genes that are critical for normal development.
    [Show full text]
  • Strategies and Opportunities for Small Molecule Drug Discovery to Target Neurodegenerative Diseases Andrea I
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.01.020206; this version posted April 2, 2020. The copyright holder has placed this preprint (which was not certified by peer review) in the Public Domain. It is no longer restricted by copyright. Anyone can legally share, reuse, remix, or adapt this material for any purpose without crediting the original authors. Defining the Neural Kinome: Strategies and Opportunities for Small Molecule Drug Discovery to Target Neurodegenerative Diseases Andrea I. Krahn, Carrow Wells, David H. Drewry, Lenore K. Beitel, Thomas M. Durcan, Alison D. Axtman* ABSTRACT: Kinases are highly tractable drug targets that have reached unparalleled success in fields such as cancer but whose potential has not yet been realized in neuroscience. There are currently 55 approved small molecule kinase-targeting drugs, 48 of which have an anti-cancer indication. The intrinsic complexity linked to central nervous system (CNS) drug development and a lack of validated targets has hindered progress in developing kinase inhibitors for CNS disorders when compared to other therapeutic areas such as oncology. Identification and/or characterization of new kinases as potential drug targets for neurodegenerative diseases will create opportunities for development of CNS drugs in the future. The track record of kinase inhibitors in other disease indications supports the idea that with the best targets identified small molecule kinase modulators will become impactful therapeutics for neurodegenerative diseases. KEYWORDS: kinase, neurodegeneration,
    [Show full text]
  • DYRK1A Binds to an Evolutionarily Conserved WD40-Repeat Title Protein WDR68 and Induces Its Nuclear Translocation
    DYRK1A binds to an evolutionarily conserved WD40-repeat Title protein WDR68 and induces its nuclear translocation. Author(s) Miyata, Yoshihiko; Nishida, Eisuke Citation Biochimica et biophysica acta (2011), 1813(10): 1728-1739 Issue Date 2011-10 URL http://hdl.handle.net/2433/148020 © 2011 Elsevier B.V.; This is not the published version. Please cite only the published version.; この論文は出版社版であり Right ません。引用の際には出版社版をご確認ご利用ください 。 Type Journal Article Textversion author Kyoto University *REVISED Manuscript (text UNmarked) Click here to view linked References DYRK1A binds to an evolutionarily conserved WD40-repeat protein WDR68 and induces its nuclear translocation Yoshihiko Miyata*, Eisuke Nishida Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Kyoto 606-8502, Japan * Corresponding author. Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan. Tel.: +81-75-753-4231; fax: +81-75-753-4235. 1 ABSTRACT DYRK1A is encoded in the Down’s syndrome critical region on human chromosome 21, and plays an important role in the functional and developmental regulation of many types of cells, including neuronal cells. Here we have identified WDR68, an evolutionarily conserved protein with WD40-repeat domains, as a cellular binding partner of DYRK1A. WDR68 was originally identified in petunia as AN11 that controls the pigmentation of flowers by stimulating the transcription of anthocyanin biosynthetic genes. Experiments with RNA interference showed that WDR68 was indispensable for the optimal proliferation and survival of mammalian cultured cell, and WDR68 depletion induced cell apoptosis. DYRK1A and DYRK1B, but not DYRK2, DYRK3, or DYRK4, bound to endogenous and expressed WDR68.
    [Show full text]
  • A Requirement for STAG2 in Replication Fork Progression Creates a Targetable Synthetic Lethality in Cohesin-Mutant Cancers
    ARTICLE https://doi.org/10.1038/s41467-019-09659-z OPEN A requirement for STAG2 in replication fork progression creates a targetable synthetic lethality in cohesin-mutant cancers Gourish Mondal1, Meredith Stevers1, Benjamin Goode 1, Alan Ashworth2,3 & David A. Solomon 1,2 Cohesin is a multiprotein ring that is responsible for cohesion of sister chromatids and formation of DNA loops to regulate gene expression. Genomic analyses have identified that 1234567890():,; the cohesin subunit STAG2 is frequently inactivated by mutations in cancer. However, the reason STAG2 mutations are selected during tumorigenesis and strategies for therapeutically targeting mutant cancer cells are largely unknown. Here we show that STAG2 is essential for DNA replication fork progression, whereby STAG2 inactivation in non-transformed cells leads to replication fork stalling and collapse with disruption of interaction between the cohesin ring and the replication machinery as well as failure to establish SMC3 acetylation. As a con- sequence, STAG2 mutation confers synthetic lethality with DNA double-strand break repair genes and increased sensitivity to select cytotoxic chemotherapeutic agents and PARP or ATR inhibitors. These studies identify a critical role for STAG2 in replication fork procession and elucidate a potential therapeutic strategy for cohesin-mutant cancers. 1 Department of Pathology, University of California, San Francisco, CA 94143, USA. 2 UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA. 3 Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA 94158, USA. Correspondence and requests for materials should be addressed to D.A.S. (email: [email protected]) NATURE COMMUNICATIONS | (2019) 10:1686 | https://doi.org/10.1038/s41467-019-09659-z | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-09659-z ohesin is a multi-protein complex composed of four core transcriptional dysregulation22,23.
    [Show full text]
  • Loss of Cohesin Complex Components STAG2 Or STAG3 Confers Resistance to BRAF Inhibition in Melanoma
    Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Shen, C., S. H. Kim, S. Trousil, D. T. Frederick, A. Piris, P. Yuan, L. Cai, et al. 2016. “Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma.” Nature medicine 22 (9): 1056-1061. doi:10.1038/nm.4155. http:// dx.doi.org/10.1038/nm.4155. Published Version doi:10.1038/nm.4155 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:31731818 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Nat Med Manuscript Author . Author manuscript; Manuscript Author available in PMC 2017 March 01. Published in final edited form as: Nat Med. 2016 September ; 22(9): 1056–1061. doi:10.1038/nm.4155. Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma Che-Hung Shen1, Sun Hye Kim1, Sebastian Trousil1, Dennie T. Frederick2, Adriano Piris3, Ping Yuan1, Li Cai1, Lei Gu4, Man Li1, Jung Hyun Lee1, Devarati Mitra1, David E. Fisher1,2, Ryan J. Sullivan2, Keith T. Flaherty2, and Bin Zheng1,* 1Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 2Department of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA 3Department of Dermatology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 4Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA.
    [Show full text]