Open Full Page

Total Page:16

File Type:pdf, Size:1020Kb

Open Full Page Published OnlineFirst June 29, 2015; DOI: 10.1158/1541-7786.MCR-15-0125 Oncogenes and Tumor Suppressors Molecular Cancer Research Structure, Dynamics, and Functionality of Tankyrase Inhibitor-Induced Degradasomes Tor Espen Thorvaldsen1,2, Nina Marie Pedersen1,2, Eva M. Wenzel1,2, Sebastian W. Schultz1,2, Andreas Brech1,2, Knut Liestøl1,3, Jo Waaler4, Stefan Krauss4, and Harald Stenmark1,2 Abstract Tankyrase (TNKS) enzymes, due to their poly(ADP-ribose) poly- revealed that the induced degradasomes in SW480 cells are mem- merase activity, have emerged as potential targets in experimental brane-free structures that consist of a filamentous assembly of high cancer therapy. However, the functional consequences of TNKS electron densities and discrete subdomains of various destruction inhibition remain incompletely resolved because of the binding complex components. Fluorescence recovery after photobleaching promiscuity of TNKS. One of the hallmarks of small-molecule experiments further demonstrated that b-catenin–mCherry was TNKS inhibitors (TNKSi) is the stabilization of AXIN, which plays rapidly turned over in the G007-LK-induced degradasomes, where- a pivotal role in the WNT/b-catenin signaling pathway. The present as GFP-TNKS1 remained stable. In conclusion, TNKS inhibition study focused on the known ability of TNKSi to induce cytoplasmic attenuates WNT/b-catenin signaling by promoting dynamic assem- puncta (degradasomes) consisting of components of the signal- blies of functional active destruction complexes into a TNKS-con- limiting WNT/b-catenin destruction complex. Using the colorectal taining scaffold even in the presence of an APC truncation. cancer cell line SW480 stably transfected with GFP-TNKS1, it was demonstrated that a TNKS-specific inhibitor (G007-LK) induces Implications: This study demonstrates that b-catenin is rapidly highly dynamic and mobile degradasomes that contain phosphor- turned over in highly dynamic assemblies of WNT destruction ylated b-catenin, ubiquitin, and b-TrCP. Likewise, G007-LK was complexes (degradasomes) upon tankyrase inhibition and pro- found to induce similar degradasomes in other colorectal cancer vides a direct mechanistic link between degradasome formation cell lines expressing wild-type or truncated versions of the degrada- and reduced WNT signaling in colorectal cancer cells. Mol Cancer some component APC. Super-resolution and electron microscopy Res; 13(11); 1487–501. Ó2015 AACR. Introduction regarded as the core complex components (5). In the WNT-off state, transcriptionally active b-catenin (ABC) levels are kept low The WNT/b-catenin signaling pathway plays a pivotal role in by CK1a/GSK3-mediated N-terminal phosphorylation of b-cate- fundamental biologic processes, including cell proliferation, cell nin and subsequent degradation by the ubiquitin-proteasome polarity, energy metabolism, and cell fate determination during system. Upon WNT activation, ABC escapes N-terminal phos- embryonic development and adult tissue homeostasis (1, 2). phorylation and proteasomal degradation, translocates to the Consequently, mutations in this pathway are linked to a broad nucleus, and initiates transcription of WNT/b-catenin-responsive range of human diseases, including cancer (3). The WNT/b-cate- genes by complexing predominantly with the TCF/LEF family of nin destruction complex regulates protein turnover of b-catenin, transcription factors (6). The mechanisms by which destruction the key mediator of canonical WNT signaling output (4). The complex activity is inhibited in the WNT-on state are currently structural proteins adenomatous polyposis coli (APC) and axis debated (7, 8). inhibition protein 1 and 2 (AXIN1/2), and the kinases casein The poly-ADP-ribosyltransferases tankyrase 1 (TNKS1) and kinase 1a (CK1a) and glycogen synthase kinase 3 (GSK3) are tankyrase 2 (TNKS2) modify acceptor proteins by transferring ADP-ribose moieties (poly-ADP-ribosylation) to amino acid side chains. Modified proteins are subsequently poly-ubiqui- 1 Centre for Cancer Biomedicine, Faculty of Medicine, Oslo University tinated and predominantly turned over by proteasomal degra- Hospital, Montebello, Oslo, Norway. 2Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Mon- dation (9). TNKS1/2 are involved in a wide range of cellular tebello, Oslo, Norway. 3Department of Informatics, University of Oslo, functions (10) and were recently shown to positively regulate 4 Oslo, Norway. Department of Microbiology, Unit for Cell Signaling, the WNT/b-catenin pathway through poly-ADP-ribosylation of Oslo University Hospital, Forskningsparken, Oslo, Norway. AXIN (11), the rate-limiting factor for destruction complex Note: Supplementary data for this article are available at Molecular Cancer stability and function (12). Implication of TNKS1/2 as drug- Research Online (http://mcr.aacrjournals.org/). gable targets in the WNT/b-catenin signaling pathway has Corresponding Author: Harald Stenmark, Institute for Cancer Research, The generated profound research on developing novel small-mol- Norwegian Radium Hospital, Montebello, N-0379 Oslo, Norway. Phone: 47- ecule inhibitors (9, 13, 14). Inhibition of the catalytic activity 22781818; Fax: 47-22781845; E-mail: [email protected] of TNKS1/2 reduces WNT/b-catenin signaling in both APC doi: 10.1158/1541-7786.MCR-15-0125 wild-type cells (e.g., in HEK293) and colorectal cancer cells Ó2015 American Association for Cancer Research. harboring APC truncations (e.g., in SW480; refs. 11, 15, 16). www.aacrjournals.org 1487 Downloaded from mcr.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst June 29, 2015; DOI: 10.1158/1541-7786.MCR-15-0125 Thorvaldsen et al. Interestingly, immunofluorescence imaging of colorectal cancer Cell-based assays cells has revealed cytoplasmic puncta formation of destruction SW480, COLO320, and LS174T cell lines were purchased from complex components upon TNKSi treatment (15–17). Based the ATCC. Upon receipt, cells were frozen, and individual aliquots on the presence of phosphorylated b-catenin (phospho-b-cate- were taken into cell culture, typically for analysis within 15 nin [PBC]) in puncta, they have been designated functional passages. Cells were grown in RPMI (SW480 and COLO320) or degradasomes (18) that promote phosphorylation and subse- DMEM F12 (LS174T) medium supplemented with 10% (SW480 quent degradation of b-catenin. However, extensive structural and COLO320) or 15% (LS174T) FBS and 1% penicillin/strep- and functional studies on these central inhibitor-induced com- tomycin. Testing for mycoplasma contamination was performed plexes are lacking. every sixth week. For inhibition of TNKS activity, cells were treated G007-LK is a highly selective TNKSi that attenuates WNT- with 0.5 mmol/L G007-LK for 24 hours, unless specified otherwise. induced cell growth both in vivo and in vitro (19, 20). In the DMSO was used as a control. For inhibition of proteasomal present study, we used endogenous proteins and stably activity, cells were treated with 10 mmol/L MG132 for 1 hour, expressed fluorescent fusion proteins to investigate the molec- either alone or in combination with G007-LK. To generate a stable ular effects of G007-LK in APC-truncated SW480 cells. Key SW480 cell line expressing GFP-TNKS1, third-generation lenti- experiments were also reproduced in other colorectal cancer viral transduction was used as previously described (25). Detailed cell lines expressing wild-type or truncated versions of APC. We cloning procedures can be requested from the authors. Cells were examined the composition and structure of inhibitor-induced sorted by FACS and frozen. Individual aliquots were grown in cell degradasomes in SW480 cells by combining confocal, super- culture as described for SW480 cells. resolution, and electron microscopy. Moreover, dynamic prop- erties and functionality of the protein complexes were eluci- Western blot analysis dated by Western blotting, live-cell imaging, and quantitative Cells were rinsed in PBS and lysed in Laemmli lysis buffer [65.8 image analysis. Here, we show that TNKS inhibition by G007- mmol/L Tris-HCl, pH 6.8, 2.1% SDS, 26.3% (w/v) glycerol, LK induces highly mobile and structurally dynamic degrada- 0.01% bromophenol blue, dithiothreitol (DTT)]. Equal amounts somes. Importantly, there is a rapid turnover of b-catenin in the of whole cell lysate were separated by SDS-PAGE (Bio-Rad Lab- degradasomes as shown by photobleaching experiments. Fur- oratories) and blotted with polyvinylidene difluoride membranes thermore, high resolution microscopy enabled us for the first (Millipore). Immunodetection was performed with IRDye-con- time to reveal structural details of these complexes beyond the jugated secondary antibodies (LI-COR Biosciences). The Odyssey resolution limits of confocal microscopy. Our data give novel Imager system (LI-COR Biosciences) was used to scan all blots. insight into the mechanisms of TNKS inhibition and attenua- Protein bands were quantified using the Odyssey software. tion of WNT/b-catenin signaling in colorectal cancer cells and provide a direct mechanistic link between degradasome for- Confocal fluorescence microscopy mation and b-catenin degradation. Cells were grown on coverslips, fixed in paraformaldehyd, and further processed for antibody staining as previously described (26). Fluorescence signals were investigated with Zeiss LSM 710/ Materials and Methods 780 microscopes (Carl Zeiss MicroImaging GmbH) using stan-
Recommended publications
  • 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]
  • Dynamics of the Linc Complex
    DYNAMICS OF THE LINC COMPLEX Loic Gazquez University of Manchester School of Biological Sciences 2017 A thesis submitted to the University of Manchester for the degree of Doctor of Philosophy in the Faculty of Biology, Medicine and Health TABLE OF CONTENTS Table of Contents .................................................................................................................................... 2 Abstract.................................................................................................................................................... 4 Declaration .............................................................................................................................................. 5 Copyright statement ................................................................................................................................ 5 Acknowledgements ................................................................................................................................. 6 I. Introduction .................................................................................................................................. 7 Nuclear Envelope and the LINC complex ................................................................................... 7 I. 1.1. The first LINC component: the SUN ................................................................................ 8 I. 1.2. The second LINC component: the KASH ........................................................................ 8 I. 1.1. Structure
    [Show full text]
  • RET Gene Fusions in Malignancies of the Thyroid and Other Tissues
    G C A T T A C G G C A T genes Review RET Gene Fusions in Malignancies of the Thyroid and Other Tissues Massimo Santoro 1,*, Marialuisa Moccia 1, Giorgia Federico 1 and Francesca Carlomagno 1,2 1 Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; [email protected] (M.M.); [email protected] (G.F.); [email protected] (F.C.) 2 Institute of Endocrinology and Experimental Oncology of the CNR, 80131 Naples, Italy * Correspondence: [email protected] Received: 10 March 2020; Accepted: 12 April 2020; Published: 15 April 2020 Abstract: Following the identification of the BCR-ABL1 (Breakpoint Cluster Region-ABelson murine Leukemia) fusion in chronic myelogenous leukemia, gene fusions generating chimeric oncoproteins have been recognized as common genomic structural variations in human malignancies. This is, in particular, a frequent mechanism in the oncogenic conversion of protein kinases. Gene fusion was the first mechanism identified for the oncogenic activation of the receptor tyrosine kinase RET (REarranged during Transfection), initially discovered in papillary thyroid carcinoma (PTC). More recently, the advent of highly sensitive massive parallel (next generation sequencing, NGS) sequencing of tumor DNA or cell-free (cfDNA) circulating tumor DNA, allowed for the detection of RET fusions in many other solid and hematopoietic malignancies. This review summarizes the role of RET fusions in the pathogenesis of human cancer. Keywords: kinase; tyrosine kinase inhibitor; targeted therapy; thyroid cancer 1. The RET Receptor RET (REarranged during Transfection) was initially isolated as a rearranged oncoprotein upon the transfection of a human lymphoma DNA [1].
    [Show full text]
  • Rat Anti-TERF1 Monoclonal Antibody, Clone 683D (CABT-RM172) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Rat Anti-TERF1 monoclonal antibody, clone 683D (CABT-RM172) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Specificity Specifically detects murine Telomeric repeat-binding factor 1 (TRF1). Target TERF1 Immunogen His-tagged full-length recombinant mouse Telomeric repeat-binding factor 1 (TRF1). Isotype IgG1, κ Source/Host Rat Species Reactivity Mouse Clone 683D Purification Protein G purified Conjugate unconjugated Applications FC, ICC, IF, WB Molecular Weight ~51 kDa observed; 48.22 kDa calculated. Uncharacterized bands may be observed in some lysate(s). Format Liquid Size 100 μg, 25 μg Buffer 0.1 M Tris-Glycine (pH 7.4), 150 mM NaCl Preservative 0.05% sodium azide Storage Stable for 1 year at 2-8°C from date of receipt. Warnings Unless otherwise stated in our catalog or other company documentation accompanying the product(s), our products are intended for research use only and are not to be used for any other purpose, which includes but is not limited to, unauthorized commercial uses, in vitro diagnostic uses, ex vivo or in vivo therapeutic uses or any type of consumption or application to humans or animals. 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved BACKGROUND Introduction Telomeric repeat-binding factor 1 is encoded by the Terf1 gene in murine species. TRF1 is a component of the shelterin complex that is involved in the regulation of telomere length and protection. It binds to telomeric DNA as a homodimer and protects telomeres.
    [Show full text]
  • Nuclear Envelope Laminopathies: Evidence for Developmentally Inappropriate Nuclear Envelope-Chromatin Associations
    Nuclear Envelope Laminopathies: Evidence for Developmentally Inappropriate Nuclear Envelope-Chromatin Associations by Jelena Perovanovic M.S. in Molecular Biology and Physiology, September 2009, University of Belgrade M.Phil. in Molecular Medicine, August 2013, The George Washington University A Dissertation submitted to The Faculty of The Columbian College of Arts and Sciences of The George Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy August 31, 2015 Dissertation directed by Eric P. Hoffman Professor of Integrative Systems Biology The Columbian College of Arts and Sciences of The George Washington University certifies that Jelena Perovanovic has passed the Final Examination for the degree of Doctor of Philosophy as of May 5, 2015. This is the final and approved form of the dissertation. Nuclear Envelope Laminopathies: Evidence for Developmentally Inappropriate Nuclear Envelope-Chromatin Associations Jelena Perovanovic Dissertation Research Committee: Eric P. Hoffman, Professor of Integrative Systems Biology, Dissertation Director Anamaris Colberg-Poley, Professor of Integrative Systems Biology, Committee Member Robert J. Freishtat, Associate Professor of Pediatrics, Committee Member Vittorio Sartorelli, Senior Investigator, National Institutes of Health, Committee Member ii © Copyright 2015 by Jelena Perovanovic All rights reserved iii Acknowledgments I am deeply indebted to countless individuals for their support and encouragement during the past five years of graduate studies. First and foremost, I would like to express my gratitude to my mentor, Dr. Eric P. Hoffman, for his unwavering support and guidance, and keen attention to my professional development. This Dissertation would not have been possible without the critical input he provided and the engaging environment he created.
    [Show full text]
  • Whole Proteome Analysis of Human Tankyrase Knockout Cells Reveals Targets of Tankyrase- Mediated Degradation
    ARTICLE DOI: 10.1038/s41467-017-02363-w OPEN Whole proteome analysis of human tankyrase knockout cells reveals targets of tankyrase- mediated degradation Amit Bhardwaj1, Yanling Yang2, Beatrix Ueberheide2 & Susan Smith1 Tankyrase 1 and 2 are poly(ADP-ribose) polymerases that function in pathways critical to cancer cell growth. Tankyrase-mediated PARylation marks protein targets for proteasomal 1234567890 degradation. Here, we generate human knockout cell lines to examine cell function and interrogate the proteome. We show that either tankyrase 1 or 2 is sufficient to maintain telomere length, but both are required to resolve telomere cohesion and maintain mitotic spindle integrity. Quantitative analysis of the proteome of tankyrase double knockout cells using isobaric tandem mass tags reveals targets of degradation, including antagonists of the Wnt/β-catenin signaling pathway (NKD1, NKD2, and HectD1) and three (Notch 1, 2, and 3) of the four Notch receptors. We show that tankyrases are required for Notch2 to exit the plasma membrane and enter the nucleus to activate transcription. Considering that Notch signaling is commonly activated in cancer, tankyrase inhibitors may have therapeutic potential in targeting this pathway. 1 Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY 10016, USA. 2 Proteomics Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA. Correspondence and requests for materials should be addressed to S.S. (email: [email protected]) NATURE COMMUNICATIONS | 8: 2214 | DOI: 10.1038/s41467-017-02363-w | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02363-w ankyrases function in cellular pathways that are critical to function in human cells will provide insights into the clinical cancer cell growth including telomere cohesion and length utility of tankyrase inhibitors.
    [Show full text]
  • Table S1. Identified Proteins with Exclusive Expression in Cerebellum of Rats of Control, 10Mg F/L and 50Mg F/L Groups
    Table S1. Identified proteins with exclusive expression in cerebellum of rats of control, 10mg F/L and 50mg F/L groups. Accession PLGS Protein Name Group IDa Score Q3TXS7 26S proteasome non-ATPase regulatory subunit 1 435 Control Q9CQX8 28S ribosomal protein S36_ mitochondrial 197 Control P52760 2-iminobutanoate/2-iminopropanoate deaminase 315 Control Q60597 2-oxoglutarate dehydrogenase_ mitochondrial 67 Control P24815 3 beta-hydroxysteroid dehydrogenase/Delta 5-->4-isomerase type 1 84 Control Q99L13 3-hydroxyisobutyrate dehydrogenase_ mitochondrial 114 Control P61922 4-aminobutyrate aminotransferase_ mitochondrial 470 Control P10852 4F2 cell-surface antigen heavy chain 220 Control Q8K010 5-oxoprolinase 197 Control P47955 60S acidic ribosomal protein P1 190 Control P70266 6-phosphofructo-2-kinase/fructose-2_6-bisphosphatase 1 113 Control Q8QZT1 Acetyl-CoA acetyltransferase_ mitochondrial 402 Control Q9R0Y5 Adenylate kinase isoenzyme 1 623 Control Q80TS3 Adhesion G protein-coupled receptor L3 59 Control B7ZCC9 Adhesion G-protein coupled receptor G4 139 Control Q6P5E6 ADP-ribosylation factor-binding protein GGA2 45 Control E9Q394 A-kinase anchor protein 13 60 Control Q80Y20 Alkylated DNA repair protein alkB homolog 8 111 Control P07758 Alpha-1-antitrypsin 1-1 78 Control P22599 Alpha-1-antitrypsin 1-2 78 Control Q00896 Alpha-1-antitrypsin 1-3 78 Control Q00897 Alpha-1-antitrypsin 1-4 78 Control P57780 Alpha-actinin-4 58 Control Q9QYC0 Alpha-adducin 270 Control Q9DB05 Alpha-soluble NSF attachment protein 156 Control Q6PAM1 Alpha-taxilin 161
    [Show full text]
  • Major Differences Between Human Atopic Dermatitis and Murine Models As Determined by Global Transcriptomic Profiling
    Downloaded from orbit.dtu.dk on: Jul 09, 2018 Major differences between human atopic dermatitis and murine models as determined by global transcriptomic profiling Ewald, David Adrian; Noda, Shinji; Oliva, Margeaux; Litman, Thomas; Nakajima, Saeko; Li, Xuan; Xu, Hui; Scheipers, Peter; Svitacheva, Naila; Labuda, Tord; Krueger, James G.; Suárez-Fariñas, Mayte; Kabashima, Kenji; Guttman-Yassky, Emma Published in: Journal of Allergy and Clinical Immunology Link to article, DOI: 10.1016/j.jaci.2016.08.029 Publication date: 2017 Document Version Peer reviewed version Link back to DTU Orbit Citation (APA): Ewald, D. A., Noda, S., Oliva, M., Litman, T., Nakajima, S., Li, X., ... Guttman-Yassky, E. (2017). Major differences between human atopic dermatitis and murine models as determined by global transcriptomic profiling. Journal of Allergy and Clinical Immunology, 139(2), 562-571. DOI: 10.1016/j.jaci.2016.08.029 General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.
    [Show full text]
  • Whole Exome Sequencing in Families at High Risk for Hodgkin Lymphoma: Identification of a Predisposing Mutation in the KDR Gene
    Hodgkin Lymphoma SUPPLEMENTARY APPENDIX Whole exome sequencing in families at high risk for Hodgkin lymphoma: identification of a predisposing mutation in the KDR gene Melissa Rotunno, 1 Mary L. McMaster, 1 Joseph Boland, 2 Sara Bass, 2 Xijun Zhang, 2 Laurie Burdett, 2 Belynda Hicks, 2 Sarangan Ravichandran, 3 Brian T. Luke, 3 Meredith Yeager, 2 Laura Fontaine, 4 Paula L. Hyland, 1 Alisa M. Goldstein, 1 NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, Stephen J. Chanock, 5 Neil E. Caporaso, 1 Margaret A. Tucker, 6 and Lynn R. Goldin 1 1Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 2Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 3Ad - vanced Biomedical Computing Center, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Frederick, MD; 4Westat, Inc., Rockville MD; 5Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; and 6Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA ©2016 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2015.135475 Received: August 19, 2015. Accepted: January 7, 2016. Pre-published: June 13, 2016. Correspondence: [email protected] Supplemental Author Information: NCI DCEG Cancer Sequencing Working Group: Mark H. Greene, Allan Hildesheim, Nan Hu, Maria Theresa Landi, Jennifer Loud, Phuong Mai, Lisa Mirabello, Lindsay Morton, Dilys Parry, Anand Pathak, Douglas R. Stewart, Philip R. Taylor, Geoffrey S. Tobias, Xiaohong R. Yang, Guoqin Yu NCI DCEG Cancer Genomics Research Laboratory: Salma Chowdhury, Michael Cullen, Casey Dagnall, Herbert Higson, Amy A.
    [Show full text]
  • Mayer-Rokitansky-Küster- Hauser Syndrome
    Morcel et al. Orphanet Journal of Rare Diseases 2011, 6:9 http://www.ojrd.com/content/6/1/9 RESEARCH Open Access Utero-vaginal aplasia (Mayer-Rokitansky-Küster- Hauser syndrome) associated with deletions in known DiGeorge or DiGeorge-like loci Karine Morcel1,2*†, Tanguy Watrin1†, Laurent Pasquier1,3, Lucie Rochard1, Cédric Le Caignec4,5, Christèle Dubourg1,6, Philippe Loget7, Bernard-Jean Paniel8, Sylvie Odent1,3, Véronique David1,6, Isabelle Pellerin1, Claude Bendavid1,6 and Daniel Guerrier1 Abstract Background: Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is characterized by congenital aplasia of the uterus and the upper part of the vagina in women showing normal development of secondary sexual characteristics and a normal 46, XX karyotype. The uterovaginal aplasia is either isolated (type I) or more frequently associated with other malformations (type II or Müllerian Renal Cervico-thoracic Somite (MURCS) association), some of which belong to the malformation spectrum of DiGeorge phenotype (DGS). Its etiology remains poorly understood. Thus the phenotypic manifestations of MRKH and DGS overlap suggesting a possible genetic link. This would potentially have clinical consequences. Methods: We searched DiGeorge critical chromosomal regions for chromosomal anomalies in a cohort of 57 subjects with uterovaginal aplasia (55 women and 2 aborted fetuses). For this candidate locus approach, we used a multiplex ligation-dependent probe amplification (MLPA) assay based on a kit designed for investigation of the chromosomal regions known to be involved in DGS. The deletions detected were validated by Duplex PCR/liquid chromatography (DP/LC) and/or array-CGH analysis. Results: We found deletions in four probands within the four chromosomal loci 4q34-qter, 8p23.1, 10p14 and 22q11.2 implicated in almost all cases of DGS syndrome.
    [Show full text]
  • APC Mutations As a Potential Biomarker for Sensitivity To
    Published OnlineFirst February 8, 2017; DOI: 10.1158/1535-7163.MCT-16-0578 Companion Diagnostics and Cancer Biomarkers Molecular Cancer Therapeutics APC Mutations as a Potential Biomarker for Sensitivity to Tankyrase Inhibitors in Colorectal Cancer Noritaka Tanaka1,2, Tetsuo Mashima1, Anna Mizutani1, Ayana Sato1,3, Aki Aoyama3,4, Bo Gong3,4, Haruka Yoshida1, Yukiko Muramatsu1, Kento Nakata1,5, Masaaki Matsuura6, Ryohei Katayama4, Satoshi Nagayama7, Naoya Fujita3,4,5, Yoshikazu Sugimoto2, and Hiroyuki Seimiya1,3,5 Abstract In most colorectal cancers, Wnt/b-catenin signaling is acti- "short" truncated APCs lacking all seven b-catenin-binding vated by loss-of-function mutations in the adenomatous polyposis 20-amino acid repeats (20-AARs). In contrast, the drug-resistant coli (APC) gene and plays a critical role in tumorigenesis. cells possessed "long" APC retaining two or more 20-AARs. Knock- Tankyrases poly(ADP-ribosyl)ate and destabilize Axins, a neg- down of the long APCs with two 20-AARs increased b-catenin, ative regulator of b-catenin, and upregulate b-catenin signaling. Tcf/LEF transcriptional activity and its target gene AXIN2 expres- Tankyrase inhibitors downregulate b-catenin and are expected sion. Under these conditions, tankyrase inhibitors were able to to be promising therapeutics for colorectal cancer. However, downregulate b-catenin in the resistant cells. These results indicate colorectal cancer cells are not always sensitive to tankyrase that the long APCs are hypomorphic mutants, whereas they exert inhibitors, and predictive biomarkers for the drug sensitivity a dominant-negative effect on Axin-dependent b-catenin degra- remain elusive. Here we demonstrate that the short-form APC dation caused by tankyrase inhibitors.
    [Show full text]
  • Crystal Structure of a Tankyrase-Axin Complex and Its Implications for Axin Turnover and Tankyrase Substrate Recruitment
    Crystal structure of a Tankyrase-Axin complex and its implications for Axin turnover and Tankyrase substrate recruitment Seamus Morronea,1, Zhihong Chenga,1, Randall T. Moonb, Feng Congc, and Wenqing Xua,2 aDepartment of Biological Structure, University of Washington School of Medicine, Seattle, WA 98195; bDepartment of Pharmacology, Howard Hughes Medical Institute, and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98195; and cNovartis Institutes for Biomedical Research, Cambridge, MA 02139 Edited by* Stephen C. Harrison, Children's Hospital, Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, and approved December8, 2011 (received for review October 9, 2011) Axin is a tumor suppressor and a key negative regulator of the Ubiquitination of Axin, which leads to its subsequent turnover, Wnt/β-catenin signaling pathway. Axin turnover is controlled by its requires its poly-ADP-ribosylation (PARylation) (11). PARylation poly-ADP-ribosylation catalyzed by tankyrase (TNKS), which re- of proteins is catalyzed by a family of poly-ADP-ribose poly- quires the direct interaction of Axin with TNKS. This interaction merases (PARPs), with 18 known members in humans, which re- is thus an attractive drug target for treating cancers, brain injuries, gulate many aspects of biology including genomic stability, cell and other diseases where β-catenin is involved. Here we report the cycle, and energy metabolism (15, 16). Axin PARylation is speci- crystal structure of a mouse TNKS1 fragment containing ankyrin- fically catalyzed by tankyrases 1 and 2 (a.k.a. TNKS1/PARP5a repeat clusters 2 and 3 (ARC2-3) in a complex with the TNKS-bind- and TNKS2/PARP5b, respectively).
    [Show full text]