Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Volume 15 - Number 5 May 2011

The PDF version of the Atlas of Genetics and Cytogenetics in Oncology and Haematology is a reissue of the original articles published in collaboration with the Institute for Scientific and Technical Information (INstitut de l’Information Scientifique et Technique - INIST) of the French National Center for Scientific Research (CNRS) on its electronic publishing platform I-Revues. Online and PDF versions of the Atlas of Genetics and Cytogenetics in Oncology and Haematology are hosted by INIST-CNRS. Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Scope

The Atlas of Genetics and Cytogenetics in Oncology and Haematology is a peer reviewed on-line journal in open access, devoted to , cytogenetics, and clinical entities in cancer, and cancer-prone diseases. It presents structured review articles ("cards") on genes, leukaemias, solid tumours, cancer-prone diseases, more traditional review articles on these and also on surrounding topics ("deep insights"), case reports in hematology, and educational items in the various related topics for students in Medicine and in Sciences.

Editorial correspondance

Jean-Loup Huret Genetics, Department of Medical Information, University Hospital F-86021 Poitiers, France tel +33 5 49 44 45 46 or +33 5 49 45 47 67 [email protected] or [email protected]

Staff Mohammad Ahmad, Mélanie Arsaban, Houa Delabrousse, Marie-Christine Jacquemot-Perbal, Maureen Labarussias, Vanessa Le Berre, Anne Malo, Catherine Morel-Pair, Laurent Rassinoux, Sylvie Yau Chun Wan - Senon, Alain Zasadzinski. Philippe Dessen is the Database Director, and Alain Bernheim the Chairman of the on-line version (Gustave Roussy Institute – Villejuif – France).

The Atlas of Genetics and Cytogenetics in Oncology and Haematology (ISSN 1768-3262) is published 12 times a year by ARMGHM, a non profit organisation, and by the INstitute for Scientific and Technical Information of the French National Center for Scientific Research (INIST-CNRS) since 2008.

The Atlas is hosted by INIST-CNRS (http://www.inist.fr)

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Editor

Jean-Loup Huret (Poitiers, France) Editorial Board

Sreeparna Banerjee (Ankara, Turkey) Solid Tumours Section Alessandro Beghini (Milan, Italy) Genes Section Anne von Bergh (Rotterdam, The Netherlands) Genes / Leukaemia Sections Judith Bovée (Leiden, The Netherlands) Solid Tumours Section Vasantha Brito-Babapulle (London, UK) Leukaemia Section Charles Buys (Groningen, The Netherlands) Deep Insights Section Anne Marie Capodano (Marseille, France) Solid Tumours Section Fei Chen (Morgantown, West Virginia) Genes / Deep Insights Sections Antonio Cuneo (Ferrara, Italy) Leukaemia Section Paola Dal Cin (Boston, Massachussetts) Genes / Solid Tumours Section Louis Dallaire (Montreal, Canada) Education Section Brigitte Debuire (Villejuif, France) Deep Insights Section François Desangles (Paris, France) Leukaemia / Solid Tumours Sections Enric Domingo-Villanueva (London, UK) Solid Tumours Section Ayse Erson (Ankara, Turkey) Solid Tumours Section Richard Gatti (Los Angeles, California) Cancer-Prone Diseases / Deep Insights Sections Ad Geurts van Kessel (Nijmegen, The Netherlands) Cancer-Prone Diseases Section Oskar Haas (Vienna, Austria) Genes / Leukaemia Sections Anne Hagemeijer (Leuven, Belgium) Deep Insights Section Nyla Heerema (Colombus, Ohio) Leukaemia Section Jim Heighway (Liverpool, UK) Genes / Deep Insights Sections Sakari Knuutila (Helsinki, Finland) Deep Insights Section Lidia Larizza (Milano, Italy) Solid Tumours Section Lisa Lee-Jones (Newcastle, UK) Solid Tumours Section Edmond Ma (Hong Kong, China) Leukaemia Section Roderick McLeod (Braunschweig, Germany) Deep Insights / Education Sections Cristina Mecucci (Perugia, Italy) Genes / Leukaemia Sections Yasmin Mehraein (Homburg, Germany) Cancer-Prone Diseases Section Fredrik Mertens (Lund, Sweden) Solid Tumours Section Konstantin Miller (Hannover, Germany) Education Section Felix Mitelman (Lund, Sweden) Deep Insights Section Hossain Mossafa (Cergy Pontoise, France) Leukaemia Section Stefan Nagel (Braunschweig, Germany) Deep Insights / Education Sections Florence Pedeutour (Nice, France) Genes / Solid Tumours Sections Elizabeth Petty (Ann Harbor, Michigan) Deep Insights Section Susana Raimondi (Memphis, Tennesse) Genes / Leukaemia Section Mariano Rocchi (Bari, Italy) Genes Section Alain Sarasin (Villejuif, France) Cancer-Prone Diseases Section Albert Schinzel (Schwerzenbach, Switzerland) Education Section Clelia Storlazzi (Bari, Italy) Genes Section Sabine Strehl (Vienna, Austria) Genes / Leukaemia Sections Nancy Uhrhammer (Clermont Ferrand, France) Genes / Cancer-Prone Diseases Sections Dan Van Dyke (Rochester, Minnesota) Education Section Roberta Vanni (Montserrato, Italy) Solid Tumours Section Franck Viguié (Paris, France) Leukaemia Section José Luis Vizmanos (Pamplona, Spain) Leukaemia Section Thomas Wan (Hong Kong, China) Genes / Leukaemia Sections

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Volume 15, Number 5, May 2011

Table of contents

Gene Section

ASXL1 (additional sex combs like 1 (Drosophila)) 391 Marie-Joelle Mozziconacci, Daniel Birnbaum EWSR1 (Ewing sarcoma breakpoint region 1) 395 Jean-Loup Huret FAM57A (family with sequence similarity 57, member A) 408 Zhiao Chen, Xianghuo He FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) 410 Hideyuki Ihara, Cong-xiao Gao, Yoshitaka Ikeda, Naoyuki Taniguchi IGSF8 (immunoglobulin superfamily, member 8) 415 Yanhui H Zhang, Mekel M Richardson, Xin A Zhang MIXL1 (Mix1 homeobox-like 1 (Xenopus laevis)) 419 Aaron Raymond, Lalitha Nagarajan PEG3 (paternally expressed 3) 422 Yinhua Yu, Weiwei Feng, Zhen Lu, Robert C Bast Jr RPL10 (ribosomal L10) 425 Mohit Goel, Ranjan Tamuli SNAI1 (snail homolog 1 (Drosophila)) 428 Joerg Schwock, William R Geddie VAV3 (vav 3 guanine nucleotide exchange factor) 436 Leah Lyons, Kerry L Burnstein CAMTA1 (calmodulin binding transcription activator 1) 441 Kai-Oliver Henrich DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) 443 Maria L Arbonés, Susana de la Luna FBLN1 (fibulin 1) 450 Lorenzo Castagnoli, Elda Tagliabue, Serenella M Pupa HUS1 (HUS1 checkpoint homolog (S. pombe)) 455 Amrita Madabushi, Randall C Gunther, A-Lien Lu OTX2 (orthodenticle homeobox 2) 460 Matthew Wortham

Leukaemia Section dic(3;9)(p14;p13) PAX5/FOXP1 463 Jean-Loup Huret dic(9;18)(p13;q11) PAX5/ZNF521 465 Atlast(11;14)(q 13;q32)of Genetics in multiple myeloma and Cytogenetics Huret JL, Laï JL in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Jean-Loup Huret

Case Report Section

Chronic lymphocytic leukaemia/Small lymphocytic lymphoma (CLL/SLL) associated with translocation t(1;6)(p35;p25) as part of complex karyotype 467 Elvira D Rodrigues Pereira Velloso, Daniela Borri, Cristina Alonso Ratis, Guilherme Fleury Perin, Nelson Hamerschlak, Nydia S Bacal, Paulo A A Silveira, Alanna M P S Bezerra, Denise C Pasqualin

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

ASXL1 (additional sex combs like 1 (Drosophila)) Marie-Joelle Mozziconacci, Daniel Birnbaum Institut Paoli-Calmettes, Centre de Recherche en Cancerologie de Marseille, Departement de Biopathologie and Laboratoire d'Oncologie Moleculaire, 232 Boulevard de Sainte-Marguerite, 13009 Marseille, France (MJM), Laboratoire d'Oncologie Moleculaire, Centre de Recherche en Cancerologie de Marseille, UMR891 Inserm, Institut Paoli-Calmettes 232 Boulevard de Sainte-Marguerite 13009 Marseille, France (DB)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/ASXL1ID44553ch20q11.html DOI: 10.4267/2042/45011 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Location: 20q11.21 Local order: centromere 5' - 3' telomere. Other names: KIAA0978, MGC117280, MGC71111 HGNC (Hugo): ASXL1

Representation of ASXL1 locus. A: 20 with localisation of ASXL1; B: ASXL1 gene; C: Amino acid count; D: Protein with domains. ASXN, conserved domain at the N-terminus; ASXM, conserved domain in the middle part; NR, nuclear receptor; PHD, plant homeodomain; E: examples of mutations.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 391 ASXL1 (additional sex combs like 1 (Drosophila)) Mozziconacci MJ, Birnbaum D

DNA/RNA Homology There are 3 mammalian homologs of the Additional sex Description combs (Asx) gene of Drosophila: ASXL1, 2 The ASXL1 gene spans around 80 kb of genomic DNA (chromosome 2p24 in humans) and 3 (chromosome and is composed of 12 exons. 18q11 in humans). Transcription Mutations Alternative splicing results in multiple transcript variants. Note Acquired ASXL1 mutations are frequently frameshift Protein and nonsense. All mutations are in exon 12, mostly around the Gly-rich domain (amino acids 642-685). Description The most common somatic mutation is The longer ASXL1 transcript encodes a 1541 amino p.Gly646Trpfsx12. acid (170 kDa) protein. Mammalian ASXL are These mutations cause truncation of the ASXL1 protein characterized by an amino-terminal ASX homology downstream of the ASXH domain leading to the loss of (ASXH) region containing 2 putative nuclear receptor the C-terminal PHD domain. coregulator binding motifs (NR box), 3 other NR box Some possible single nucleotide polymorphisms have motifs and a C-terminal plant homeodomain protein- been described: protein interaction domain. Contains one Leu-Xaa-Xaa- p.Arg1224Thr, p.Thr769Ala, p.Gly1007Arg, Leu-Leu (LXXLL) motif, which may be required for an p.Thr688Met, p.Gln1074Leu, p.Arg693Gly, p.T1139K, association with nuclear receptors. p.Gly652Ser, p.Val1072Asp... All mutations have been found in myeloid diseases so Expression far. A fusion has been found in B-cell leukemia. ASXL1 is expressed all hematopoietic cell fractions in mice. Asxl1 knockout mice exhibit mild defects in Implicated in differentiation of lymphoid and myeloid progenitors, but not in multipotent progenitors and do not develop Myelodysplastic syndromes (11-21%) MDS or other hematological malignancy. Note ASXL1 is widely expressed at low level in heart, brain, Mutations p.Arg596Profsx23, p.Gly646Trpfsx12 (the skeletal muscle, placenta, pancreas, spleen, prostate, most common mutation), p.Gln1102Asp, small intestine, colon, peripheral blood, leukocytes, p.Leu1395Val, p.Ser1457Profsx18... bone marrow and fetal liver. Highly expressed in testes. Prognosis Localisation More frequent in advanced and high-risk MDSs (>40% Nucleus (probable). in RAEB2). Function Cytogenetics Normal or abnormal karyotype. - ASXL1 acts as a co-regulator of retinoic acid (RA) receptor in RA sensitive cell lines, and as a co- Chronic myelomonocytic leukemia (33- repressor of RA receptor in RA resistant cell lines. 43%) Either a coactivator or corepressor for the retinoid Note receptors retinoic acid receptor and retinoid X receptor p.His630Profsx66, p.Lys618X, p.Gly646trpfsx12, in a cell type-specific manner. p.Gln768X, p.Thr836Leufsx2, p.Ser846Glnfsx5, - ASXL1 cooperates with HP1 (heterochromatin p.Lys888Glufsx6, p.Arg1068X, p.pro1263Glnfsx17, protein 1) to modulate histone H3 demethylase LSD1 p.Leu1266Hisfsx9, p.Thr1271lysfsx10... activity, leading to a change in histone H3 methylation and RAR repression. Prognosis - ASXL1 belongs to the Enhancer of Trithorax and Associated with acute transformation. Polycomb (ETP) group in drosophila. Cytogenetics - ASXL1 is required for maintenance of both activation Normal or abnormal karyotype. More common in and silencing of Hox genes in mice and drosophila in a patients with -7/7q-. Infrequent in the presence of - context-dependent manner. 5/5q-. - ASXL1 is one of the fusion protein partners of PAX5 in B-cell precursor acute lymphoblastic leukemias. Juvenile myelomonocytic leukemia - ASXL1 may function as a tumor suppressor in (JMML) (2/49 patients 4%) myeloid malignancies by affecting stem or progenitor Note cell self renewal or differentiation. p.Arg693X, p.Ser846ValfsX21.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 392 ASXL1 (additional sex combs like 1 (Drosophila)) Mozziconacci MJ, Birnbaum D

Cytogenetics Scotto L, Narayan G, Nandula SV, Arias-Pulido H, Subramaniyam S, Schneider A, Kaufmann AM, Wright JD, Normal karyotype or monosomy 7. Pothuri B, Mansukhani M, Murty VV. Identification of copy Acute myeloid leukemia (20-30%) number gain and overexpressed genes on chromosome arm 20q by an integrative genomic approach in cervical cancer: Note potential role in progression. Genes Cancer. 30% in primary AMLs, 47% in secondary AMLs and 2008 Sep;47(9):755-65 23% in post-MDS AMLs. An Q, Wright SL, Moorman AV, Parker H, Griffiths M, Ross FM, Davies T, Harrison CJ, Strefford JC. Heterogeneous Prognosis breakpoints in patients with acute lymphoblastic leukemia and Shorter overall survival. the dic(9;20)(p11-13;q11) show recurrent involvement of genes Cytogenetics at 20q11.21. Haematologica. 2009 Aug;94(8):1164-9 Normal or abnormal karyotype, associated with trisomy Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adélaïde J, 8, inversely correlated with NPM1 mutation. Rey J, Vainchenker W, Bernard OA, Chaffanet M, Vey N, Birnbaum D, Mozziconacci MJ. Mutations of ASXL1 gene in Myeloproliferative neoplasms (8%) myeloproliferative neoplasms. Leukemia. 2009 Nov;23(11):2183-6 Note Present in all forms, including chronic myeloid Gelsi-Boyer V, Trouplin V, Adélaïde J, Bonansea J, Cervera N, Carbuccia N, Lagarde A, Prebet T, Nezri M, Sainty D, leukemia. More frequent in primary myelofibrosis. Olschwang S, Xerri L, Chaffanet M, Mozziconacci MJ, Vey N, Prognosis Birnbaum D. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic Occur in both chronic and blast-phase MPNs. leukaemia. Br J Haematol. 2009 Jun;145(6):788-800 B-cell acute lymphoblastic leukemia Abdel-Wahab O, Kilpivaara O, Patel J, Busque L, Levine RL. Cytogenetics The most commonly reported variant in ASXL1 (c.1934dupG;p.Gly646TrpfsX12) is not a somatic alteration. Dicentric chromosome dic(9;20)(p11-13;q11). Leukemia. 2010 Sep;24(9):1656-7 Hybrid/Mutated gene Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat PAX5 on 9p and ASXL1 on 20q. C, Heguy A, Bueso-Ramos C, Kantarjian H, Levine RL, Verstovsek S. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. To be noted Cancer Res. 2010 Jan 15;70(2):447-52 Note Acquaviva C, Gelsi-Boyer V, Birnbaum D. Myelodysplastic ASXL1 can be altered by small local deletion detected syndromes: lost between two states? Leukemia. 2010 only by array CGH or SNP arrays. However, ASXL1 Jan;24(1):1-5 seems to be centromeric to the main deleted region in Boultwood J, Perry J, Pellagatti A, Fernandez-Mercado M, classical 20q deletion. Fernandez-Santamaria C, Calasanz MJ, Larrayoz MJ, Garcia- Delgado M, Giagounidis A, Malcovati L, Della Porta MG, Jädersten M, Killick S, Hellström-Lindberg E, Cazzola M, References Wainscoat JS. Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute Fisher CL, Berger J, Randazzo F, Brock HW. A human myeloid leukemia. Leukemia. 2010 May;24(5):1062-5 homolog of Additional sex combs, ADDITIONAL SEX COMBS- LIKE 1, maps to chromosome 20q11. Gene. 2003 Mar Boultwood J, Perry J, Zaman R, Fernandez-Santamaria C, 13;306:115-26 Littlewood T, Kusec R, Pellagatti A, Wang L, Clark RE, Wainscoat JS. High-density single nucleotide polymorphism Katoh M, Katoh M. Identification and characterization of ASXL2 array analysis and ASXL1 gene mutation screening in chronic gene in silico. Int J Oncol. 2003 Sep;23(3):845-50 myeloid leukemia during disease progression. Leukemia. 2010 Katoh M, Katoh M. Identification and characterization of ASXL3 Jun;24(6):1139-45 gene in silico. Int J Oncol. 2004 Jun;24(6):1617-22 Carbuccia N, Trouplin V, Gelsi-Boyer V, Murati A, Rocquain J, Adélaïde J, Olschwang S, Xerri L, Vey N, Chaffanet M, Katoh M, Katoh M. Identification and characterization of human CXXC10 gene in silico. Int J Oncol. 2004 Oct;25(4):1193-9 Birnbaum D, Mozziconacci MJ. Mutual exclusion of ASXL1 and NPM1 mutations in a series of acute myeloid leukemias. Cho YS, Kim EJ, Park UH, Sin HS, Um SJ. Additional sex Leukemia. 2010 Feb;24(2):469-73 comb-like 1 (ASXL1), in cooperation with SRC-1, acts as a ligand-dependent coactivator for retinoic acid receptor. J Biol Chou WC, Huang HH, Hou HA, Chen CY, Tang JL, Yao M, Chem. 2006 Jun 30;281(26):17588-98 Tsay W, Ko BS, Wu SJ, Huang SY, Hsu SC, Chen YC, Huang YN, Chang YC, Lee FY, Liu MC, Liu CW, Tseng MH, Huang Fisher CL, Randazzo F, Humphries RK, Brock HW. CF, Tien HF. Distinct clinical and biological features of de novo Characterization of Asxl1, a murine homolog of Additional sex acute myeloid leukemia with additional sex comb-like 1 combs, and analysis of the Asx-like gene family. Gene. 2006 (ASXL1) mutations. Blood. 2010 Nov 18;116(20):4086-94 Mar 15;369:109-18 Davids MS, Steensma DP. The molecular pathogenesis of An Q, Wright SL, Konn ZJ, Matheson E, Minto L, Moorman AV, myelodysplastic syndromes. Cancer Biol Ther. 2010 Parker H, Griffiths M, Ross FM, Davies T, Hall AG, Harrison Aug;10(4):309-19 CJ, Irving JA, Strefford JC. Variable breakpoints target PAX5 in patients with dicentric chromosomes: a model for the basis of Delhommeau F, Jeziorowska D, Marzac C, Casadevall N. unbalanced translocations in cancer. Proc Natl Acad Sci U S Molecular aspects of myeloproliferative neoplasms. Int J Hematol. 2010 Mar;91(2):165-73 A. 2008 Nov 4;105(44):17050-4

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 393 ASXL1 (additional sex combs like 1 (Drosophila)) Mozziconacci MJ, Birnbaum D

Fisher CL, Lee I, Bloyer S, Bozza S, Chevalier J, Dahl A, CBL, FLT3, IDH1, IDH2, JAK2, KRAS, NPM1, NRAS, RUNX1, Bodner C, Helgason CD, Hess JL, Humphries RK, Brock HW. TET2 and WT1 genes in myelodysplastic syndromes and Additional sex combs-like 1 belongs to the enhancer of acute myeloid leukemias. BMC Cancer. 2010 Aug 2;10:401 trithorax and polycomb group and genetically interacts with Cbx2 in mice. Dev Biol. 2010 Jan 1;337(1):9-15 Sugimoto Y, Muramatsu H, Makishima H, Prince C, Jankowska AM, Yoshida N, Xu Y, Nishio N, Hama A, Yagasaki H, Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Takahashi Y, Kato K, Manabe A, Kojima S, Maciejewski JP. Bodner C, Hess JL, Humphries RK, Brock HW. Loss-of- Spectrum of molecular defects in juvenile myelomonocytic function Additional sex combs like 1 mutations disrupt leukaemia includes ASXL1 mutations. Br J Haematol. 2010 hematopoiesis but do not cause severe myelodysplasia or Jul;150(1):83-7 leukemia. Blood. 2010 Jan 7;115(1):38-46 Szpurka H, Jankowska AM, Makishima H, Bodo J, Bejanyan N, Gelsi-Boyer V, Trouplin V, Rocquain J, Adelaide J, Carbuccia Hsi ED, Sekeres MA, Maciejewski JP. Spectrum of mutations N, Esterni B, Finetti P, Murati M, Arnoulet C, Zerazhi H, Fezoui in RARS-T patients includes TET2 and ASXL1 mutations. Leuk H, Tadrist Z, Nezri M, Chaffanet M, Mozziconacci MJ, Vey N, Res. 2010 Aug;34(8):969-73 Birnbaum D.. ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic Tefferi A. Mutational analysis in BCR-ABL-negative classic leukaemia. British Journal of Haematology, 2010 Sept 29. myeloproliferative neoplasms: impact on prognosis and therapeutic choices. Leuk Lymphoma. 2010 Apr;51(4):576-82 Lee SW, Cho YS, Na JM, Park UH, Kang M, Kim EJ, Um SJ. ASXL1 represses retinoic acid receptor-mediated transcription Tefferi A. Novel mutations and their functional and clinical through associating with HP1 and LSD1. J Biol Chem. 2010 relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, Jan 1;285(1):18-29 ASXL1, CBL, IDH and IKZF1. Leukemia. 2010 Jun;24(6):1128- 38 Mittelman M, Oster HS, Hoffman M, Neumann D. The lower risk MDS patient at risk of rapid progression. Leuk Res. 2010 This article should be referenced as such: Dec;34(12):1551-5 Mozziconacci MJ, Birnbaum D. ASXL1 (additional sex combs Rocquain J, Carbuccia N, Trouplin V, Raynaud S, Murati A, like 1 (Drosophila)). Atlas Genet Cytogenet Oncol Haematol. Nezri M, Tadrist Z, Olschwang S, Vey N, Birnbaum D, Gelsi- 2011; 15(5):391-394. Boyer V, Mozziconacci MJ. Combined mutations of ASXL1,

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 394 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

EWSR1 (Ewing sarcoma breakpoint region 1) Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/EWSR1ID85.html DOI: 10.4267/2042/45012 This article is an update of : Mugneret F. EWSR1 (Ewing sarcoma region 1). Atlas Genet Cytogenet Oncol Haematol 1998;2(3):79-80

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Protein Other names: EWS Description HGNC (Hugo): EWSR1 656 amino acids for the canonical form identified in Location: 22q12.2 1993 (Plougastel et al., 1993), 68.5 kDa. From N-term to C-term: a transactivation domain (TAD) containing DNA/RNA multiple degenerate hexapeptide repeats (consensus SYGQQS) (glycine, glutamine, serine, tyrosine rich or Description SYGQ rich, where the tyrosine is mandatory): amino Spans 32.5 kb, in a centromere to telomere direction on acids 1 to 285, with a site interacting with SF1 from aa plus strand; transcript of 2654 bp from 17 exons for the 228 to 264 and an IQ domain, which binds calmodulin canonical form, with a coding sequence of 1971 nt. (aa 256-285), 3 arginine/glycine rich domains (RGG regions) (aa 300-340, 454-513 (arginine/glycine/proline Transcription rich), and aa 559-640), a RNA recognition motif (RRM According to Ensembl, there are 25 transcripts, of or RNA-binding domain (RBD): aa 361-447), and a which 16 different transcripts code for proteins. RanBP2 type Zinc finger (aa 518-549).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 395 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Expression al., 1998). It associates with heterogeneous RNA- binding proteins (hnRNPs), such as RBM38 and EWSR1 is ubiquitely expressed (Alliegro and Alliegro, RBM39 (RNA binding motif proteins 38 and 39, 20q13 1996; Andersson et al., 2008). In particular, EWSR1 is and 20q11 respectively) (Zinszner et al., 1994). required for cell survival in the central nervous system EWSR1 associates with EP300 and CREBBP. EWSR1 (Azuma et al., 2007). functions as a co-activator of CREBBP-dependent Localisation transcription factors. EWSR1-EP300/CREBBP mediates FOS activation, as well as HNF4 genes Mainly in the nucleus. It has also been found in the activation (Rossow and Janknecht, 2001; Araya et al., cytoplasm, and associated with the plasma membrane. 2003). CREBBP is a transcription co-activator which Expression of EWSR1 in the various subcellular enables the interaction between various transcription compartments is affected by the methylation state of its factors and RNA Pol II, brings enzymes to the RNA-binding domain (Belyanskaya et al., 2003). promoter, and remodels the chromatin favouring the EWSR1 is mainly found in the nucleus, and more open status (Gervasini, 2009). rarely in the cytoplasm than its two homologs FUS and EWSR1 also activates other transcription factors such TAF15; the 3 proteins participate in nucleo- as POU4F1 (or BRN3A, 13q13) (Gascoyne et al., cytoplasmic shutlling; EWSR1 localized poorly in 2004), POU5F1 (or OCT4, 6p21) (Lee et al., 2005). stress granules when cells were exposed to SF1 (splicing factor 1, 11q13, also called ZFM1) environmental stress (stress granules are cytoplasmic represses the transactivation domain of EWSR1; SF1, a particles composed of translation pre-initiation transcription activator or repressor involved with many complexes, mRNAs and RNA-binding proteins) pathways, may negatively modulate transcription of (Andersson et al., 2008), in Cajal bodies, and nucloli. target genes coordinated by EWSR1 (Zhang et al., Localization of EWSR1 in different subcellular 1998). compartments reflects a dynamic distribution during EWS functions as a docking molecule by recruiting cell cycle: predominant nuclear localization in serine-arginine (SR) splicing factors such as SRSF10 interphase cells, perichromosomal localization in (serine/arginine-rich splicing factor 10, 1p36, or TASR, prometaphase cells, and cytoplasmic localization in which represses pre-mRNA splicing) to RNA Pol II, metaphase cells, association with microtubules in coupling gene transcription to RNA splicing by binding quiescent cells (Leemann-Zakaryan et al., 2009). to hyperphosphorylated RNA Pol II through its N-term Function part domain, and SR splicing factors through its C-term RNA binding protein, single strand DNA binding. domain (Yang et al., 2000). Role in transcriptional regulation for specific genes and YBX1 (Y box binding protein 1, 1p34), a in mRNA splicing: Transcription and pre-mRNA multifunctional protein that shuttles between the slicing, a post-transcriptional activity, are closely cytoplasm (where it binds to mRNA and regulates related. mRNA translation) and the nucleus (where it regulates EWSR1 plays a role in transcription initiation: EWSR1 transcription of diverse target genes), interacts with the is able to associate with the basal transcription factor C-term domain of EWS. This interaction docks YBX1 TFIID (a multiprotein complex composed of the to RNA Pol II to participate in pre-mRNA splicing TATA-binding protein (TBP) and TBP-associated (Chansky et al., 2001). factors (TAFIIs)) and the RNA polymerase II complex. SMN1 (survival of motor neuron 1, telomeric, 5q13) EWSR1 acts as a transcriptional activator (Bertolotti et plays a major role in the pre-mRNA splicing machinery

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 396 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

(role in spliceosomal snRNP assembly in the cytoplasm mitosis, and maintains mitotic spindle integrity (Azuma and in pre-mRNA splicing in the nucleus) (Pellizzoni et et al., 2007). al., 1998). SMN1, through its Tudor region, binds the It also interacts with BARD1 (BRCA1 associated RG1 region of EWSR1 (Young et al., 2003). RING domain 1) (Spahn et al., 2002). RNU1-2 (RNA, U1 small nuclear 2, 1p36, also called EWSR1 and CCNL1 (cyclin L1, 3q25), are interacting U1C), another splicing protein, also interacts with partners of TFIP11 (tuftelin-interacting protein 11, EWSR1 (Knoop and Baker, 2000). 22q12), a protein functionally related to the EWSR1 is phosphorylated by PRKC (protein kinase C) spliceosome and involved in pre-mRNA splicing) though its IQ domain, which inhibits RNA binding of (Tannukit et al., 2008). EWSR1; CALM (calmodulin) binding to EWSR1 Homology inhibits PRKC phosphorylation (Deloulme et al., 1997). Member of the TET family of RNA binding proteins, EWSR1 interacts with POU4F2 (4q31), a gene which with FUS (TLS) and TAF15 (TAFII68). TET is for Tls, regulates differentiation of neuronal cells (Gascoyne et Ewsr1, TafII68). TET proteins contain specific al., 2004). EWSR1 interacts with LMNA (lamin A/C, structural domains not found elsewhere in other RNA 1q22, a component of the nuclear envelope which binding proteins, i.e. a N-term SYGQ rich (TAD: interacts with DNA, histones and transcriptional transactivation domain), a conserved RNA-binding repressors) is essential for pre-B lymphocyte domain (RRM: RNA recognition motif), RG rich development and, during meiosis, in XY pairing and in regions, and a Cys2-Cys2 Zinc finger which can bind meiotic recombination, as cross-overs are reduced in nucleic acids; they are also functionally related ews-/- spermatocytes. Loss of EWSR1 results in (reviews in Law et al., 2006; Tan and Manley, 2009). premature cellular senescence (Li et al., 2007). EWSR1 is required for proper localization of aurora B during Implicated in Various tumours (see details below)

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 397 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Ewing's sarcoma/Peripheral Hybrid/Mutated gene neurectodermal tumour (ES/PNET) t(11;22)(q24;q12): 5' EWSR1 - 3' FLI1; breakpoints clustered over a 2-3 kb genomic region and over a 30- Note 40 kb genomic region. Various junctions between - With: t(11;22)(q24;q12) --> FLI1 - EWSR1 (Delattre EWSR1 exon 7 or 10 with FLI1 exon 5, 6, or 8. In the et al., 1992; Bailly et al., 1994; Thomas et al., 1994; most common fusion type (type 1), EWSR1 exon 7 is records in the Mitelman Database). fused in frame to FLI1 exon 6; in type 2, EWSR1 exon - With: t(2;22)(q36;q12) --> FEV - EWSR1 (Llombart- 7 is fused in frame to FLI1 exon 5. (Obata et al., 1999; Bosch et al., 2000; Peter et al., 2001; Navarro et al., de Alava et al., 1998). 2002; Hattinger et al., 1999; Wang et al., 2007). t(21;22)(q21;q12): 5' EWSR1 - 3' ERG; the orientation - With: t(7;22)(p22;q12) --> ETV1 - EWSR1 (Jeon et of the ERG gene is from telomere to centromere, al., 1995; Peter et al., 2001; Zielenska et al., 2001; opposite to that demonstrated for EWSR1. Wang et al., 2007). Other translocations: 5' EWSR1 - 3' FEV; 5' EWSR1 - - With: t(17;22)(q21;q12) --> ETV4 - EWSR1 (Kaneko 3' ETV1; 5' EWSR1 - 3' ETV4; 5' EWSR1 - 3' et al., 1996; Urano et al., 1996; Urano et al., 1998). NFATC2; 5' EWSR1 - 3' PATZ1. - With: t(20;22)(q13;q12) --> NFATC2 - EWSR1 (Szuhai et al., 2009). Abnormal protein - With: t(21;22)(q21;q12) --> ERG - EWSR1 (Dunn et t(11;22)(q24;q12): oncogenic protein on the der(22) al., 1994; Giovannini et al., 1994; Kaneko et al., 1997; chromosome generated by fusion of the N terminal Maire et al., 2008; Minoletti et al., 1998; Sorensen et domain of EWSR1 protein (transactivation domain, e.g. al., 1994; Zoubek et al., 1994; Zucman et al., 1993b; fusion of EWSR1 amino acids 1-265) with the DNA Shing et al., 2003; Peter et al., 1996). binding domain (ETS type, amino acids 281-361) of - With: inv(22)(q12q12) --> PATZ1 - EWSR1 the human FLI1 protein, a 452 amino acids protein (e.g. (Mastrangelo et al., 2000) fusion from amino acids 260). Note: Rare cases of ES/PNET have been described t(21;22)(q21;q12): oncogenic protein on the der(22) without EWSR1 involvement, but, instead: chromosome generated by fusion of the N terminal - with: t(2;16)(q35;p11) --> FUS - FEV (Navarro et al., domain of EWSR1 protein with DNA binding domain 2002) or, of human ERG protein. - with: t(16;21)(p11;q22) --> FUS - ERG (Shing et al., Other translocations: Most of the EWSR1 partners in 2003; Berg et al., 2009). To be noted that the same ES/PNET are ETS family members (FLI1, ERG, t(16;21)(p11;q22) has been found in rare cases of acute ETV1, ETV4, FEV) and translocation results in the myeloid leukaemia. juxtaposition of the transactivation domain of EWSR1 A t(11;22)(q24;q12) has also been found in a case of with the DNA binding domain -ETS type of the cerebellar PNET (Jay et al., 1996). partner. PATZ1 is a transcription regulator with a AT hook (DNA-binding motif), a POZ domain (mediates Disease dimerisation) and Zn fingers (DNA-binding). NFATC2 Ewing's sarcoma/Peripheral neurectodermal tumour is a cytokine inducer; translocates into the nucleus to spectrum is a group of sarcomas with small blue round regulate transcription. cells with more (PNET side) or less (Ewing side) features of neuroectodermal differentiation. ES/PNET Oncogenesis entity also includes peripheral neuroepithelioma and EWSR1-FLI1 is a dominant oncogene transformed Askin tumour. These tumours display both cells by subverting normal transcriptional mesenchymal stem cell and neural crest stem cell controls/FLI1 member of ETS family. The target gene properties. It is mainly found in adolescents and young repertoire of EWSR1-FLI1 varies according to the host adults. Cytogenetics and immunochemistry are cell type. EWSR1-FLI1 induces a TP53-dependent essential for the differential diagnosis with other growth arrest in fibroblasts, supporting the importance sarcomas (review in Romeo and Dei Tos, 2010). of TP53 loss in the genesis of Ewing's tumours (Lessnick et al., 2002). EWSR1-FLI1 activates CASP3 Prognosis and promotes apoptosis in mouse embryonic fibroblasts Prognosis has dramatically improved, from less than (Sohn et al., 2010). EWSR1-FLI1 induces expression 10% of long survivors, to a 5-year disease free survival of the embryonic stem cell genes OCT4, SOX2, and of 75% for patients with a localized disease, and 25- NANOG in paediatric mesenchymal stem cells but not 30% for those with a metastatic disease (Ludwig, in adult mesenchymal stem cells. SOX2 is a target for 2008). EWSR1-FLI1 and miRNA145 and may be critical in Cytogenetics Ewing sarcoma pathogenesis (Riggi et al., 2010). The t(11;22)(q24;q12) EWSR1/FLI1 is found in 85% EWSR1-FLI1 expression in a rhabdomyosarcoma cell of cases of Ewing tumours. The t(21;22)(q21;q12) with line induced upregulation of many genes involved in EWSR1/ERG is the second in frequency, found in neural crest differentiation, and the cell phenotype was about 10% of cases. modified, resembling ewing tumour cells (Hu-

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 398 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Lieskovan et al., 2005; Riggi et al., 2008). EZH2 is a and senescence, is upregulated by EWSR1-ETS fusion target of EWSR1-FLI1. EZH2 regulates stemness and proteins), escape from apoptosis (EWSR1-FLI1 was genes involved in neuroectodermal and endothelial shown to repress IGFBP-3, preventing apoptosis) differentiation (Richter et al., 2009). EWSR1-FLI1 (Janknecht et al., 2005). induced T-cell neoplasia in committed lymphoid cells, Desmoplastic small round cell sarcoma showing that the oncogenetic potential of EWSR1-FLI1 is not restricted to pluripotent progenitors or (DSRCT) mesenchymal cells (Codrington et al., 2005). EWSR1- Note FLI1 in transgenic mouse induced limb developmental - With: t(11;22)(p13;q12) --> WT1 - EWSR1 (Ladanyi defects by disruption of the normal development of and Gerald, 1994; Gerald et al., 1995; Benjamin et al., connective tissues; homozygous deletion of p53 in 1996; records in the Mitelman Database). mice provoke sarcomas, in particular osteosarcomas, - With: t(21;22)(q21;q12) --> ERG - EWSR1 (Ordi et introduction of EWSR1-FLI1 changed the tumour al., 1998). phenotype from osteosarcomas to poorly differentiated Disease sarcomas (Lin et al., 2008). It is believed that Desmoplastic small round cell sarcoma is a small blue EW/PNET arise from mesenchymal stem cells in which round cells tumour, often intra-abdominal, with a male terminal differentiation is blocked by EWSR1-FLI1 predominance, arising in children and young adults, (Tirode et al., 2007). with a very poor prognosis. Cytogenetics and Transcriptional repressors such as NKX2-2 (Smith et immunochemistry are essential for the differential al., 2006) or NR0B1 are induced by EWSR1-FLI1. diagnosis with other sarcomas with small blue round Furthermore, EWSR1-FLI and NR0B1 physically cells. interact (Kinsey et al., 2009). The transcriptional complex of EWSR1-FLI1 includes RNA polymerase II, Cytogenetics CREB1 (cAMP responsive element binding protein 1) Most of the cases are cases of t(11;22)(p13;q12). and DHX9 (RHA, RNA helicase A) (Toretsky et al., Hybrid/Mutated gene 2006; Erkizan et al., 2009). EWSR1-Ets proteins t(11;22)(p13;q12): 5' EWSR1 - 3' WT1; breakpoints: cooperatively bind DNA with FOS-JUN (Kim et al., between EWSR1 exons 7 and 8 and between WT1 2006). EWSR1-FLI1 is involved in the spliceosome exons 7 and 8. (review in Erkizan et al., 2010). EWSR1-FLI1 chimeric t(21;22)(q21;q12): 5' EWSR1 - 3' ERG. protein, but not wild EWSR1, can oppose the change in Abnormal protein splicing pattern induced by expression of hnRNPA1 Transcription activator. (Knoop and Baker, 2001). EWSR1-FLI1 (dis)regulates many pathways (Jedlicka, Oncogenesis 2010). CD99, a transmembrane protein highly N terminal domain of EWSR1 fused to the Zn fingers expressed in Ewing sarcoma cells, has a key role in of WT1. various biological functions, including inhibition of Clear cell sarcoma of soft neuronal differentiation that may occur through the parts/malignant melanoma of soft parts RAS/RAF/MAPK pathway in Ewing's tumours (Rocchi et al., 2010). PDGFC (Zwerner and May, 2001) as well (CCSSP) as IGF1 are induced by EWSR1-FLI1, and also by Note EWSR1-ERG or FUS-ERG (Cironi et al., 2008). GLI1 - With: t(2;22)(q34;q12) --> CREB1 - EWSR1 is upregulated by EWSR1-FLI1, independently of the (Antonescu et al., 2006; Wang et al., 2009). Hedgehog pathway (Beauchamp et al., 2009; Joo et al., - With: t(12;22)(q13;q12) --> ATF1 - EWSR1 (Zucman 2009). Expression of DKK1 (which antagonizes Wnt et al., 1993a; records in the Mitelman Database). signaling) is inhibited by EWSR1-FLI1 (Navarro et al., Disease 2010), and DKK2 enhanced (Miyagawa et al., 2009). Clear cell sarcoma of tendons and aponeuroses affects TGFbR2 is inhibited by EWSR1-FLI1 (Hahm, 1999). young adults. It is characterized by slow progression, Other features were summarized in Janknecht et al., with recurrences and metastases, with only 40-50% 2005: EWSR1-FLI1 induces proliferation independent long survivors. of exogenous growth factors (EWSR1-ETS proteins upregulate PDGFC and also CCND1), evasion of Hybrid/Mutated gene growth inhibition (downregulation of TGFbR2 may 5' EWSR1 - 3' ATF1 (t(12;22)(q13;q12) cases) or, help cells escape growth surveillance), suppression of more rarely, 5' EWSR1 - 3' CREB1 (t(2;22)(q33;q12) differentiation (ID2, overexpressed in Ewing tumours, cases). is able to suppress differentiation of a variety of cells), Abnormal protein immortality (hTERT (human telomerase reverse N terminal domain of EWSR1 fused to the bZIP transcriptase)), limiting factor for telomerase activity domain of CREB1 or ATF1.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 399 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Angiomatoid fibrous histiocytoma variant round cell liposarcoma is much more (AFH) aggressive. Note Cytogenetics - With: t(2;22)(q34;q12) --> CREB1 - EWSR1 A t(12;22)(q13;q12) --> DDIT3 - EWSR1 (Antonescu et al., 2007; Shao et al., 2009; Rossi et al., (Panagopoulos et al., 1994) is found in 5% of cases, 2007). whereas a t(12;16)(q13;p11) is found in most cases. - With: t(12;22)(q13;q12) --> ATF1 - EWSR1 Hybrid/Mutated gene (Dunham et al., 2008; Hallor et al., 2005; Hallor et al., 5' EWSR1 - 3' DDIT3 (also called CHOP). The 2007; Rossi et al., 2007; Tanas et al., 2010). t(12;16)(q13;p11) with 5' FUS - 3' DDIT3 is more Note: Cases of t(12;16)(q13;p11) with 5' FUS - 3' frequent. ATF1 have also been described (Raddaoui et al., 2002; Abnormal protein Waters et al., 2000). Fuses the N-term transactivation domain of EWSR1 Disease with the bZIP domain of DDIT3. Angiomatoid fibrous histiocytoma is a rare soft-tissue Acute leukaemia tumour of low metastatic potential (local recurrence below 15% of cases, and metastases occur in less than Disease 2% of patients); it is mostly found in children and Acute lymphoblastic leukaemia (B-cell ALL), young adults. Surgical excision is the treatment of biphenotypic leukaemia choice. Cytogenetics Hybrid/Mutated gene A t(12;22)(p13;q12) was found in 2 cases (Martini et 5' EWSR1 - 3' ATF1 (t(12;22)(q13;q12) cases), or 5' al., 2002). Note: the equivalent t(12;17)(p13;q11) --> EWSR1 - 3' CREB1 (t(2;22)(q33;q12) cases). TAF15 - ZNF384 has also been found in other cases of the same series. Abnormal protein N terminal domain of EWSR1 fused to the bZIP Hybrid/Mutated gene domain of ATF1 or CREB1. 5' EWSR1 - 3' ZNF384. Extraskeletal myxoid chondrosarcoma Abnormal protein (EMCS) Fuses the N-term transactivation domain of EWSR1 with the leucine-serine rich-proline-nuclear localization Disease signal-Kruppel-type C2H2 Zinc finger domains of Extra-skeletal myxoid chondrosarcomas represent ZNF384. about 5% of chondrosarcomas. There is male Note predominance. It affects adults mainly, in the forties or EWSR1 involvement has also been described in a the fifties. The estimated 5-, 10-, and 15-year survival number of other tumours. In some instances, the rates were 90%, 70%, and 60%, respectively (Meis- diagnosis is unambiguous; in other cases, pathological Kindblom et al., 1999). diagnoses are difficult to reach, when the tumour is Cytogenetics undifferentiated or polyphenotypic. t(9;22)(q22;q12) --> NR4A3 - EWSR1 (Labelle et al., 1995; Brody et al., 1997). Rhabdomyosarcoma (RMS) Note: Cases of t(3;9)(q12;q31) --> NR4A3 - TFG Disease (Hisaoka et al., 2004), t(9;15)(q31;q21) --> NR4A3 - Rhabdomyosarcomas, the most common pediatric soft TCF12 (Sjögren et al., 2000), t(9;17)(q22;q11) --> tissue sarcomas, are tumours related to the skeletal NR4A3 - TAF15 (Sjögren et al., 1999; Attwooll et al., muscle lineage. The 2 major subtypes are alveolar 1999; records in the Mitelman Database) have also rhabdomyosarcoma (ARMS) and embryonal been reported. rhabdomyosarcoma (ERMS). Most ARMS cases are Hybrid/Mutated gene characterised by either a t(2;13)(q35;q14), resulting in a 5' EWSR1 - 3' NR4A3 (NR4A3 is also known as TEC PAX3 - FOXO1 hybrid gene, or a t(1;13)(p36;q14) or CHN). resulting in a PAX7 - FOXO1 hybrid gene (Reichek Myxoid liposarcoma/round cell and Barr, 2009). liposarcoma (MLS) However, 4 cases of RMS have been described, Disease instead, with an EWSR1 rearrangement. Myxoid liposarcoma is the most frequent type of Cytogenetics liposarcoma, found in about half of the cases. It occurs A t(4;22)(q35;q12) was found in a case of embryonal in male and female patients equally, in their thirties to rhabdomyosarcoma (Sirvent et al., 2009), and a fifties. It has a relatively favorable prognosis; the t(11;22)(q24;q12) in the other cases (Sorensen et al., 1993; Thorner et al., 1996).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 400 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Hybrid/Mutated gene Neuroblastoma 5' EWSR1 - 3' DUX4 in the t(4;22)(q35;q12) case, and Disease 5' EWSR1 - 3' FLI1 in the t(11;22) cases. Neuroblastomas are peripheral neuroblastic tumours Giant cell tumour of bone derived from cells of the sympathetic nervous system. Disease They occur mainly in infants and young children, with Locally destructive tumour, usually seen in patients a median age of 1.5 years. over 20 years of age, with good prognosis, despite of Hybrid/Mutated gene recurrences and pulmonary metastases (Forsyth and Two patients, aged 3 years and 5.5 years, were Hogendoorn, 2003). described with a 5' EWSR1 - 3' FLI1 transcript in Cytogenetics typical neuroblastomas with elevated urinary In giant-cell tumour of bone, the most frequent finding catecholamines. Prognosis had been very poor: the two is telomeric association. One study showed that a very patients relapsed during -or at the end of- treatment and minor clone with EWSR1/FLI1 translocation could be died within 2 months (Burchill et al., 1997). detected in most of the cases studied (Scotlandi et al., Olfactory neuroblastoma 2000). Disease Hybrid/Mutated gene Olfactory neuroblastoma or esthesioneuroblastoma, is a 5' EWSR1 - 3' FLI1. malignant neurectodermal tumour, from the olfactory Myoepithelioma/myoepithelial neuroepithelium that typically occurs in the superior carcinoma of soft parts nasal cavity. It is keratin negative, neuroendocrine marker positive, and S100 positive. It arises at any age, Disease often in the adult, with a 5-year survival rate above Myoepithelioma tumours of soft tissue cover a wide 50% (the 5-year overall survival for patients treated for range of tumours of various behaviour. While most are nonmetastatic olfactory neuroblastoma was recently of intermediate aggressivity, some metastase. found at 64% (Ozsahin et al., 2010)). Cytogenetics Cytogenetics t(1;22)(q23;q12) in one case (Brandal et al., 2008), t(11;22)(q24;q12), inducing a 5' EWSR1 - 3' FLI1 t(19;22)(q13;q12) in another case (Brandal et al., hybrid gene (Sorensen et al., 1996). However, recent 2009). review rejects cases with EWSR1 involvement, as Hybrid/Mutated gene being misdiagnosed cases of ES/PNET (Thompson, 5 ' EWSR1 - 3' PBX1, which fuses exons 8 from 2009). EWSR1 to exons 5 of PBX1 in the most benign case; 5' Solid pseudopapillary tumour of the EWSR1- 3' ZNF444 in the malignant case; fuses pancreas (SPTP) EWSR1 exon 8 to the very near end of ZNF444. Disease Hidradenoma of the skin Solid pseudopapillary tumour of the pancreas is a rare Disease epithelial tumour, typically occuring in young female Hidradenoma or eccrine/apocrine acrospiroma, is a patients, rarely metastasizing (Yu et al., 2010). benign adnexal tumour developing most often in adults. Cytogenetics 3 cases were found with a t(6;22)(p21;q12) and/or its One case showed a t(11;22)(q24;q12) (Maitra et al., fusion transcript equivalent (Möller et al., 2008). 2000) Abnormal protein Hybrid/Mutated gene 5' EWSR1 - 3' POU5F1. 5' EWSR1 - 3' FLI1. Mucoepidermoid carcinoma of the "Small round cell tumours", salivary glands "polyphenotypic mesenchymal Disease malignancies", and "undifferentiated Mucoepidermoid carcinoma is the most common type of malignant salivary gland tumour, often associated sarcomas" with a t(11;19)(q21;p13) translocation with expression Disease of chimeric genes 5' CRTC1 - 3' MAML2. One case of An undifferentiated sarcoma derived from pelvic bone mucoepidermoid carcinoma was found with a exhibited a t(6;22)(p21;q12) with 5' EWSR1 - 3' t(6;22)(p21;q12) (Möller et al., 2008). POU5F1. This resulted in the fusion of exons 1-6 of Abnormal protein EWSR1 and exons 2-5 and a part of exon 1 of 5' EWSR1 - 3' POU5F1. POU5F1.The patient died 6 months after diagnosis (Yamaguchi et al., 2005).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 401 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

A small round cell tumour was found to have a differentiation), or polyphenotypic sarcomas present t(2;22)(q31;q12), with 5' EWSR1 - 3' SP3 hybrid gene; with the classical t(11;22)(q24;q12) / 5' EWSR1 - 3' fuses the exon 7 of EWSR1 to exon 6 of SP3. N-term FLI1 or other variants, such as the t(2;22)(q36;q12) / 5' transactivation domain of EWSR1 fused with the Zinc EWSR1 - 3' FEV (Wang et al., 2007), the fingers of SP3. The patient died 20 months after t(11;22)(p13;q12) / 5' EWSR1 - 3' WT1 (Alaggio et al., diagnosis (Wang et al., 2007). 2007), the t(12;22)(q13;q12) / 5' EWSR1 - 3' ATF1 Other cases of spindle cell tumours, small round cell (Somers et al., 2005), or the t(21;22)(q21;q12) / 5' poorly differentiated, biphenotypic (myogenic/neural EWSR1 - 3' ERG (Tan et al., 2001). Breakpoints

Note Clustered over a 2.3 kb genomic region. References Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, malignant melanoma of soft parts. Nat Genet. 1993 Peter M, Kovar H, Joubert I, de Jong P, Rouleau G. Gene Aug;4(4):341-5 fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature. 1992 Zucman J, Melot T, Desmaze C, Ghysdael J, Plougastel B, Sep 10;359(6391):162-5 Peter M, Zucker JM, Triche TJ, Sheer D, Turc-Carel C. Combinatorial generation of variable fusion proteins in the Plougastel B, Zucman J, Peter M, Thomas G, Delattre O. Ewing family of tumours. EMBO J. 1993 Dec;12(12):4481-7 Genomic structure of the EWS gene and its relationship to EWSR1, a site of tumor-associated chromosome translocation. Bailly RA, Bosselut R, Zucman J, Cormier F, Delattre O, Genomics. 1993 Dec;18(3):609-15 Roussel M, Thomas G, Ghysdael J. DNA-binding and transcriptional activation properties of the EWS-FLI-1 fusion Sorensen PH, Liu XF, Delattre O, Rowland JM, Biggs CA, protein resulting from the t(11;22) translocation in Ewing Thomas G, Triche TJ. Reverse transcriptase PCR amplification sarcoma. Mol Cell Biol. 1994 May;14(5):3230-41 of EWS/FLI-1 fusion transcripts as a diagnostic test for peripheral primitive neuroectodermal tumors of childhood. Dunn T, Praissman L, Hagag N, Viola MV. ERG gene is Diagn Mol Pathol. 1993 Sep;2(3):147-57 translocated in an Ewing's sarcoma cell line. Cancer Genet Cytogenet. 1994 Aug;76(1):19-22 Zucman J, Delattre O, Desmaze C, Epstein AL, Stenman G, Speleman F, Fletchers CD, Aurias A, Thomas G. EWS and Giovannini M, Biegel JA, Serra M, Wang JY, Wei YH, Nycum ATF-1 gene fusion induced by t(12;22) translocation in L, Emanuel BS, Evans GA. EWS-erg and EWS-Fli1 fusion transcripts in Ewing's sarcoma and primitive neuroectodermal

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 402 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

tumors with variant translocations. J Clin Invest. 1994 related to Ewing sarcoma. Proc Natl Acad Sci U S A. 1996 Feb Aug;94(2):489-96 6;93(3):1038-43 Ladanyi M, Gerald W. Fusion of the EWS and WT1 genes in Thorner P, Squire J, Chilton-MacNeil S, Marrano P, Bayani J, the desmoplastic small round cell tumor. Cancer Res. 1994 Malkin D, Greenberg M, Lorenzana A, Zielenska M. Is the Jun 1;54(11):2837-40 EWS/FLI-1 fusion transcript specific for Ewing sarcoma and peripheral primitive neuroectodermal tumor? A report of four Panagopoulos I, Mandahl N, Ron D, Höglund M, Nilbert M, cases showing this transcript in a wider range of tumor types. Mertens F, Mitelman F, Aman P. Characterization of the CHOP Am J Pathol. 1996 Apr;148(4):1125-38 breakpoints and fusion transcripts in myxoid liposarcomas with the 12;16 translocation. Cancer Res. 1994 Dec Urano F, Umezawa A, Hong W, Kikuchi H, Hata J. A novel 15;54(24):6500-3 chimera gene between EWS and E1A-F, encoding the adenovirus E1A enhancer-binding protein, in extraosseous Sorensen PH, Lessnick SL, Lopez-Terrada D, Liu XF, Triche Ewing's sarcoma. Biochem Biophys Res Commun. 1996 Feb TJ, Denny CT. A second Ewing's sarcoma translocation, 15;219(2):608-12 t(21;22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat Genet. 1994 Feb;6(2):146-51 Brody RI, Ueda T, Hamelin A, Jhanwar SC, Bridge JA, Healey JH, Huvos AG, Gerald WL, Ladanyi M. Molecular analysis of Thomas G, Delattre O, Zucman J, Merel P, Desmaze C, Melot the fusion of EWS to an orphan nuclear receptor gene in T, Sanson M, Hoang-Xuan K, Plougastel B, Dejong P. Genetic extraskeletal myxoid chondrosarcoma. Am J Pathol. 1997 alterations in the chromosome 22q12 region associated with Mar;150(3):1049-58 development of neuroectodermal tumors. Cold Spring Harb Symp Quant Biol. 1994;59:555-64 Burchill SA, Wheeldon J, Cullinane C, Lewis IJ. EWS-FLI1 fusion transcripts identified in patients with typical Zinszner H, Albalat R, Ron D. A novel effector domain from the neuroblastoma. Eur J Cancer. 1997 Feb;33(2):239-43 RNA-binding protein TLS or EWS is required for oncogenic transformation by CHOP. Genes Dev. 1994 Nov 1;8(21):2513- Deloulme JC, Prichard L, Delattre O, Storm DR. The 26 prooncoprotein EWS binds calmodulin and is phosphorylated by protein kinase C through an IQ domain. J Biol Chem. 1997 Zoubek A, Pfleiderer C, Salzer-Kuntschik M, Amann G, Oct 24;272(43):27369-77 Windhager R, Fink FM, Koscielniak E, Delattre O, Strehl S, Ambros PF. Variability of EWS chimaeric transcripts in Ewing Desmaze C, Brizard F, Turc-Carel C, Melot T, Delattre O, tumours: a comparison of clinical and molecular data. Br J Thomas G, Aurias A. Multiple chromosomal mechanisms Cancer. 1994 Nov;70(5):908-13 generate an EWS/FLI1 or an EWS/ERG fusion gene in Ewing tumors. Cancer Genet Cytogenet. 1997 Aug;97(1):12-9 Gerald WL, Rosai J, Ladanyi M. Characterization of the genomic breakpoint and chimeric transcripts in the EWS-WT1 Kaneko Y, Kobayashi H, Handa M, Satake N, Maseki N. EWS- gene fusion of desmoplastic small round cell tumor. Proc Natl ERG fusion transcript produced by chromosomal insertion in a Acad Sci U S A. 1995 Feb 14;92(4):1028-32 Ewing sarcoma. Genes Chromosomes Cancer. 1997 Mar;18(3):228-31 Jeon IS, Davis JN, Braun BS, Sublett JE, Roussel MF, Denny CT, Shapiro DN. A variant Ewing's sarcoma translocation Bertolotti A, Melot T, Acker J, Vigneron M, Delattre O, Tora L. (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene. EWS, but not EWS-FLI-1, is associated with both TFIID and 1995 Mar 16;10(6):1229-34 RNA polymerase II: interactions between two members of the TET family, EWS and hTAFII68, and subunits of TFIID and Labelle Y, Zucman J, Stenman G, Kindblom LG, Knight J, RNA polymerase II complexes. Mol Cell Biol. 1998 Turc-Carel C, Dockhorn-Dworniczak B, Mandahl N, Desmaze Mar;18(3):1489-97 C, Peter M. Oncogenic conversion of a novel orphan nuclear receptor by chromosome translocation. Hum Mol Genet. 1995 de Alava E, Kawai A, Healey JH, Fligman I, Meyers PA, Huvos Dec;4(12):2219-26 AG, Gerald WL, Jhanwar SC, Argani P, Antonescu CR, Pardo- Mindan FJ, Ginsberg J, Womer R, Lawlor ER, Wunder J, Alliegro MC, Alliegro MA. A nuclear protein regulated during Andrulis I, Sorensen PH, Barr FG, Ladanyi M. EWS-FLI1 the transition from active to quiescent phenotype in cultured fusion transcript structure is an independent determinant of endothelial cells. Dev Biol. 1996 Mar 15;174(2):288-97 prognosis in Ewing's sarcoma. J Clin Oncol. 1998 Benjamin LE, Fredericks WJ, Barr FG, Rauscher FJ 3rd. Apr;16(4):1248-55 Fusion of the EWS1 and WT1 genes as a result of the Minoletti F, Sozzi G, Tornielli S, Pilotti S, Azzarelli A, Pierotti t(11;22)(p13;q12) translocation in desmoplastic small round MA, Radice P. A novel EWS-ERG rearrangement generating cell tumors. Med Pediatr Oncol. 1996 Nov;27(5):434-9 two hybrid mRNAs in a peripheral primitive neuroectodermal Jay V, Zielenska M, Lorenzana A, Drake J. An unusual tumour (pPNET) with a t(15;22) translocation. J Pathol. 1998 cerebellar primitive neuroectodermal tumor with t(11;22) Dec;186(4):434-7 translocation: pathological and molecular analysis. Pediatr Ordi J, de Alava E, Torné A, Mellado B, Pardo-Mindan J, Pathol Lab Med. 1996 Jan-Feb;16(1):119-28 Iglesias X, Cardesa A. Intraabdominal desmoplastic small Kaneko Y, Yoshida K, Handa M, Toyoda Y, Nishihira H, round cell tumor with EWS/ERG fusion transcript. Am J Surg Tanaka Y, Sasaki Y, Ishida S, Higashino F, Fujinaga K. Fusion Pathol. 1998 Aug;22(8):1026-32 of an ETS-family gene, EIAF, to EWS by t(17;22)(q12;q12) Pellizzoni L, Kataoka N, Charroux B, Dreyfuss G. A novel chromosome translocation in an undifferentiated sarcoma of function for SMN, the spinal muscular atrophy disease gene infancy. Genes Chromosomes Cancer. 1996 Feb;15(2):115-21 product, in pre-mRNA splicing. Cell. 1998 Nov 25;95(5):615-24 Peter M, Mugneret F, Aurias A, Thomas G, Magdelenat H, Urano F, Umezawa A, Yabe H, Hong W, Yoshida K, Fujinaga Delattre O. An EWS/ERG fusion with a truncated N-terminal K, Hata J. Molecular analysis of Ewing's sarcoma: another domain of EWS in a Ewing's tumor. Int J Cancer. 1996 Jul fusion gene, EWS-E1AF, available for diagnosis. Jpn J Cancer 29;67(3):339-42 Res. 1998 Jul;89(7):703-11 Sorensen PH, Wu JK, Berean KW, Lim JF, Donn W, Frierson Zhang D, Paley AJ, Childs G. The transcriptional repressor HF, Reynolds CP, López-Terrada D, Triche TJ. Olfactory ZFM1 interacts with and modulates the ability of EWS to neuroblastoma is a peripheral primitive neuroectodermal tumor

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 403 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

activate transcription. J Biol Chem. 1998 Jul 17;273(29):18086- Arvand A, Welford SM, Teitell MA, Denny CT. The COOH- 91 terminal domain of FLI-1 is necessary for full tumorigenesis and transcriptional modulation by EWS/FLI-1. Cancer Res. Attwooll C, Tariq M, Harris M, Coyne JD, Telford N, Varley JM. 2001 Jul 1;61(13):5311-7 Identification of a novel fusion gene involving hTAFII68 and CHN from a t(9;17)(q22;q11.2) translocation in an extraskeletal Chansky HA, Hu M, Hickstein DD, Yang L. Oncogenic myxoid chondrosarcoma. Oncogene. 1999 Dec 9;18(52):7599- TLS/ERG and EWS/Fli-1 fusion proteins inhibit RNA splicing 601 mediated by YB-1 protein. Cancer Res. 2001 May 1;61(9):3586-90 Hahm KB. Repression of the gene encoding the TGF-beta type II receptor is a major target of the EWS-FLI1 oncoprotein. Nat Knoop LL, Baker SJ. EWS/FLI alters 5'-splice site selection. J Genet. 1999 Dec;23(4):481 Biol Chem. 2001 Jun 22;276(25):22317-22 Hattinger CM, Rumpler S, Strehl S, Ambros IM, Zoubek A, Peter M, Gilbert E, Delattre O. A multiplex real-time pcr assay Pötschger U, Gadner H, Ambros PF. Prognostic impact of for the detection of gene fusions observed in solid tumors. Lab deletions at 1p36 and numerical aberrations in Ewing tumors. Invest. 2001 Jun;81(6):905-12 Genes Chromosomes Cancer. 1999 Mar;24(3):243-54 Rossow KL, Janknecht R. The Ewing's sarcoma gene product functions as a transcriptional activator. Cancer Res. 2001 Mar 15;61(6):2690-5 Meis-Kindblom JM, Bergh P, Gunterberg B, Kindblom LG. Extraskeletal myxoid chondrosarcoma: a reappraisal of its Tan SY, Burchill S, Brownhill SC, Gerrard MP, Watmore A, morphologic spectrum and prognostic factors based on 117 Wagner BE, Variend S. Small round cell tumor with cases. Am J Surg Pathol. 1999 Jun;23(6):636-50 biphenotypic differentiation and variant of t(21;22)(q22;q12). Pediatr Dev Pathol. 2001 Jul-Aug;4(4):391-6 Obata K, Hiraga H, Nojima T, Yoshida MC, Abe S. Molecular characterization of the genomic breakpoint junction in a Zielenska M, Zhang ZM, Ng K, Marrano P, Bayani J, Ramirez t(11;22) translocation in Ewing sarcoma. Genes Chromosomes OC, Sorensen P, Thorner P, Greenberg M, Squire JA. Cancer. 1999 May;25(1):6-15 Acquisition of secondary structural chromosomal changes in pediatric ewing sarcoma is a probable prognostic factor for Sjögren H, Meis-Kindblom J, Kindblom LG, Aman P, Stenman tumor response and clinical outcome. Cancer. 2001 Jun G. Fusion of the EWS-related gene TAF2N to TEC in 1;91(11):2156-64 extraskeletal myxoid chondrosarcoma. Cancer Res. 1999 Oct 15;59(20):5064-7 Zwerner JP, May WA. PDGF-C is an EWS/FLI induced transforming growth factor in Ewing family tumors. Oncogene. Knoop LL, Baker SJ. The splicing factor U1C represses 2001 Feb 1;20(5):626-33 EWS/FLI-mediated transactivation. J Biol Chem. 2000 Aug 11;275(32):24865-71 Lessnick SL, Dacwag CS, Golub TR. The Ewing's sarcoma oncoprotein EWS/FLI induces a p53-dependent growth arrest Llombart-Bosch A, Pellín A, Carda C, Noguera R, Navarro S, in primary human fibroblasts. Cancer Cell. 2002 May;1(4):393- Peydró-Olaya A. Soft tissue Ewing sarcoma--peripheral 401 primitive neuroectodermal tumor with atypical clear cell pattern shows a new type of EWS-FEV fusion transcript. Diagn Mol Martini A, La Starza R, Janssen H, Bilhou-Nabera C, Pathol. 2000 Sep;9(3):137-44 Corveleyn A, Somers R, Aventin A, Foà R, Hagemeijer A, Mecucci C, Marynen P. Recurrent rearrangement of the Maitra A, Weinberg AG, Schneider N, Patterson K. Detection Ewing's sarcoma gene, EWSR1, or its homologue, TAF15, with of t(11;22)(q24;q12) translocation and EWS-FLI-1 fusion the transcription factor CIZ/NMP4 in acute leukemia. Cancer transcript in a case of solid pseudopapillary tumor of the Res. 2002 Oct 1;62(19):5408-12 pancreas. Pediatr Dev Pathol. 2000 Nov-Dec;3(6):603-5 Navarro S, Noguera R, Pellín A, López-Guerrero JA, Roselló- Mastrangelo T, Modena P, Tornielli S, Bullrich F, Testi MA, Sastre E, Cremades A, Llombart-Bosch A. Atypical Mezzelani A, Radice P, Azzarelli A, Pilotti S, Croce CM, pleomorphic extraosseous ewing tumor/peripheral primitive Pierotti MA, Sozzi G. A novel zinc finger gene is fused to EWS neuroectodermal tumor with unusual phenotypic/genotypic in small round cell tumor. Oncogene. 2000 Aug 3;19(33):3799- profile. Diagn Mol Pathol. 2002 Mar;11(1):9-15 804 Raddaoui E, Donner LR, Panagopoulos I. Fusion of the FUS Scotlandi K, Chano T, Benini S, Serra M, Manara MC, and ATF1 genes in a large, deep-seated angiomatoid fibrous Cerisano V, Picci P, Baldini N. Identification of EWS/FLI-1 histiocytoma. Diagn Mol Pathol. 2002 Sep;11(3):157-62 transcripts in giant-cell tumor of bone. Int J Cancer. 2000 Aug 1;87(3):328-35 Spahn L, Petermann R, Siligan C, Schmid JA, Aryee DN, Kovar H. Interaction of the EWS NH2 terminus with BARD1 Sjögren H, Wedell B, Meis-Kindblom JM, Kindblom LG, links the Ewing's sarcoma gene to a common tumor Stenman G. Fusion of the NH2-terminal domain of the basic suppressor pathway. Cancer Res. 2002 Aug 15;62(16):4583-7 helix-loop-helix protein TCF12 to TEC in extraskeletal myxoid chondrosarcoma with translocation t(9;15)(q22;q21). Cancer Araya N, Hirota K, Shimamoto Y, Miyagishi M, Yoshida E, Res. 2000 Dec 15;60(24):6832-5 Ishida J, Kaneko S, Kaneko M, Nakajima T, Fukamizu A. Cooperative interaction of EWS with CREB-binding protein Waters BL, Panagopoulos I, Allen EF. Genetic characterization selectively activates hepatocyte nuclear factor 4-mediated of angiomatoid fibrous histiocytoma identifies fusion of the FUS transcription. J Biol Chem. 2003 Feb 14;278(7):5427-32 and ATF-1 genes induced by a chromosomal translocation involving bands 12q13 and 16p11. Cancer Genet Cytogenet. Belyanskaya LL, Delattre O, Gehring H. Expression and 2000 Sep;121(2):109-16 subcellular localization of Ewing sarcoma (EWS) protein is affected by the methylation process. Exp Cell Res. 2003 Aug Yang L, Chansky HA, Hickstein DD. EWS.Fli-1 fusion protein 15;288(2):374-81 interacts with hyperphosphorylated RNA polymerase II and interferes with serine-arginine protein-mediated RNA splicing. J Biol Chem. 2000 Dec 1;275(48):37612-8

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 404 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Forsyth RG, Hogendoorn PCW.. Bone: Giant cell tumour. Atlas Smith R, Owen LA, Trem DJ, Wong JS, Whangbo JS, Golub Genet Cytogenet Oncol Haematol. June 2003. URL: TR, Lessnick SL. Expression profiling of EWS/FLI identifies http://AtlasGeneticsOncology.org/Tumors/BoneGiantCellTumI NKX2.2 as a critical target gene in Ewing's sarcoma. Cancer D5150.html. Cell. 2006 May;9(5):405-16 Shing DC, McMullan DJ, Roberts P, Smith K, Chin SF, Toretsky JA, Erkizan V, Levenson A, Abaan OD, Parvin JD, Nicholson J, Tillman RM, Ramani P, Cullinane C, Coleman N. Cripe TP, Rice AM, Lee SB, Uren A. Oncoprotein EWS-FLI1 FUS/ERG gene fusions in Ewing's tumors. Cancer Res. 2003 activity is enhanced by RNA helicase A. Cancer Res. 2006 Jun Aug 1;63(15):4568-76 1;66(11):5574-81 Young PJ, Francis JW, Lince D, Coon K, Androphy EJ, Lorson Alaggio R, Rosolen A, Sartori F, Leszl A, d'Amore ES, Bisogno CL. The Ewing's sarcoma protein interacts with the Tudor G, Carli M, Cecchetto G, Coffin CM, Ninfo V. Spindle cell tumor domain of the survival motor neuron protein. Brain Res Mol with EWS-WT1 transcript and a favorable clinical course: a Brain Res. 2003 Nov 6;119(1):37-49 variant of DSCT, a variant of leiomyosarcoma, or a new entity? Report of 2 pediatric cases. Am J Surg Pathol. 2007 Gascoyne DM, Thomas GR, Latchman DS. The effects of Brn- Mar;31(3):454-9 3a on neuronal differentiation and apoptosis are differentially modulated by EWS and its oncogenic derivative EWS/Fli-1. Antonescu CR, Dal Cin P, Nafa K, Teot LA, Surti U, Fletcher Oncogene. 2004 May 6;23(21):3830-40 CD, Ladanyi M. EWSR1-CREB1 is the predominant gene fusion in angiomatoid fibrous histiocytoma. Genes Hisaoka M, Ishida T, Imamura T, Hashimoto H. TFG is a novel Chromosomes Cancer. 2007 Dec;46(12):1051-60 fusion partner of NOR1 in extraskeletal myxoid chondrosarcoma. Genes Chromosomes Cancer. 2004 Azuma M, Embree LJ, Sabaawy H, Hickstein DD. Ewing Aug;40(4):325-8 sarcoma protein ewsr1 maintains mitotic integrity and proneural cell survival in the zebrafish embryo. PLoS One. Codrington R, Pannell R, Forster A, Drynan LF, Daser A, 2007 Oct 3;2(10):e979 Lobato N, Metzler M, Rabbitts TH. The Ews-ERG fusion protein can initiate neoplasia from lineage-committed Hallor KH, Micci F, Meis-Kindblom JM, Kindblom LG, Bacchini haematopoietic cells. PLoS Biol. 2005 Aug;3(8):e242 P, Mandahl N, Mertens F, Panagopoulos I. Fusion genes in angiomatoid fibrous histiocytoma. Cancer Lett. 2007 Jun Hallor KH, Mertens F, Jin Y, Meis-Kindblom JM, Kindblom LG, 18;251(1):158-63 Behrendtz M, Kalén A, Mandahl N, Panagopoulos I. Fusion of the EWSR1 and ATF1 genes without expression of the MITF- Li H, Watford W, Li C, Parmelee A, Bryant MA, Deng C, M transcript in angiomatoid fibrous histiocytoma. Genes O'Shea J, Lee SB. Ewing sarcoma gene EWS is essential for Chromosomes Cancer. 2005 Sep;44(1):97-102 meiosis and B lymphocyte development. J Clin Invest. 2007 May;117(5):1314-23 Hu-Lieskovan S, Zhang J, Wu L, Shimada H, Schofield DE, Triche TJ. EWS-FLI1 fusion protein up-regulates critical genes Rossi S, Szuhai K, Ijszenga M, Tanke HJ, Zanatta L, Sciot R, in neural crest development and is responsible for the Fletcher CD, Dei Tos AP, Hogendoorn PC. EWSR1-CREB1 observed phenotype of Ewing's family of tumors. Cancer Res. and EWSR1-ATF1 fusion genes in angiomatoid fibrous 2005 Jun 1;65(11):4633-44 histiocytoma. Clin Cancer Res. 2007 Dec 15;13(24):7322-8 Janknecht R. EWS-ETS oncoproteins: the linchpins of Ewing Tirode F, Laud-Duval K, Prieur A, Delorme B, Charbord P, tumors. Gene. 2005 Dec 19;363:1-14 Delattre O. Mesenchymal stem cell features of Ewing tumors. Cancer Cell. 2007 May;11(5):421-9 Lee J, Rhee BK, Bae GY, Han YM, Kim J. Stimulation of Oct-4 activity by Ewing's sarcoma protein. Stem Cells. 2005 Jun- Wang L, Bhargava R, Zheng T, Wexler L, Collins MH, Roulston Jul;23(6):738-51 D, Ladanyi M. Undifferentiated small round cell sarcomas with rare EWS gene fusions: identification of a novel EWS-SP3 Somers GR, Viero S, Nathan PC, Teshima I, Pereira C, fusion and of additional cases with the EWS-ETV1 and EWS- Zielenska M. Association of the t(12;22)(q13;q12) EWS/ATF1 FEV fusions. J Mol Diagn. 2007 Sep;9(4):498-509 rearrangement with polyphenotypic round cell sarcoma of bone: a case report. Am J Surg Pathol. 2005 Dec;29(12):1673- Andersson MK, Ståhlberg A, Arvidsson Y, Olofsson A, Semb 9 H, Stenman G, Nilsson O, Aman P. The multifunctional FUS, EWS and TAF15 proto-oncoproteins show cell type-specific Yamaguchi S, Yamazaki Y, Ishikawa Y, Kawaguchi N, Mukai expression patterns and involvement in cell spreading and H, Nakamura T. EWSR1 is fused to POU5F1 in a bone tumor stress response. BMC Cell Biol. 2008 Jul 11;9:37 with translocation t(6;22)(p21;q12). Genes Chromosomes Cancer. 2005 Jun;43(2):217-22 Brandal P, Panagopoulos I, Bjerkehagen B, Gorunova L, Skjeldal S, Micci F, Heim S. Detection of a t(1;22)(q23;q12) Antonescu CR, Nafa K, Segal NH, Dal Cin P, Ladanyi M. translocation leading to an EWSR1-PBX1 fusion gene in a EWS-CREB1: a recurrent variant fusion in clear cell sarcoma-- myoepithelioma. Genes Chromosomes Cancer. 2008 association with gastrointestinal location and absence of Jul;47(7):558-64 melanocytic differentiation. Clin Cancer Res. 2006 Sep 15;12(18):5356-62 Cironi L, Riggi N, Provero P, Wolf N, Suvà ML, Suvà D, Kindler V, Stamenkovic I. IGF1 is a common target gene of Ewing's Guipaud O, Guillonneau F, Labas V, Praseuth D, Rossier J, sarcoma fusion proteins in mesenchymal progenitor cells. Lopez B, Bertrand P. An in vitro enzymatic assay coupled to PLoS One. 2008 Jul 9;3(7):e2634 proteomics analysis reveals a new DNA processing activity for Ewing sarcoma and TAF(II)68 proteins. Proteomics. 2006 Dunham C, Hussong J, Seiff M, Pfeifer J, Perry A. Primary Nov;6(22):5962-72 intracerebral angiomatoid fibrous histiocytoma: report of a case with a t(12;22)(q13;q12) causing type 1 fusion of the EWS and Kim S, Denny CT, Wisdom R. Cooperative DNA binding with ATF-1 genes. Am J Surg Pathol. 2008 Mar;32(3):478-84 AP-1 proteins is required for transformation by EWS-Ets fusion proteins. Mol Cell Biol. 2006 Apr;26(7):2467-78 Lin PP, Pandey MK, Jin F, Xiong S, Deavers M, Parant JM, Lozano G. EWS-FLI1 induces developmental abnormalities Law WJ, Cann KL, Hicks GG. TLS, EWS and TAF15: a model and accelerates sarcoma formation in a transgenic mouse for transcriptional integration of gene expression. Brief Funct model. Cancer Res. 2008 Nov 1;68(21):8968-75 Genomic Proteomic. 2006 Mar;5(1):8-14

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 405 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Ludwig JA. Ewing sarcoma: historical perspectives, current Richter GH, Plehm S, Fasan A, Rössler S, Unland R, Bennani- state-of-the-art, and opportunities for targeted therapy in the Baiti IM, Hotfilder M, Löwel D, von Luettichau I, Mossbrugger I, future. Curr Opin Oncol. 2008 Jul;20(4):412-8 Quintanilla-Martinez L, Kovar H, Staege MS, Müller-Tidow C, Burdach S. EZH2 is a mediator of EWS/FLI1 driven tumor Maire G, Brown CW, Bayani J, Pereira C, Gravel DH, Bell JC, growth and metastasis blocking endothelial and neuro- Zielenska M, Squire JA. Complex rearrangement of ectodermal differentiation. Proc Natl Acad Sci U S A. 2009 Mar chromosomes 19, 21, and 22 in Ewing sarcoma involving a 31;106(13):5324-9 novel reciprocal inversion-insertion mechanism of EWS-ERG fusion gene formation: a case analysis and literature review. Shao L, Singh V, Cooley L. Angiomatoid fibrous histiocytoma Cancer Genet Cytogenet. 2008 Mar;181(2):81-92 with t(2;22)(q33;q12.2) and EWSR1 gene rearrangement. Pediatr Dev Pathol. 2009 Mar-Apr;12(2):143-6 Möller E, Stenman G, Mandahl N, Hamberg H, Mölne L, van den Oord JJ, Brosjö O, Mertens F, Panagopoulos I. POU5F1, Sirvent N, Trassard M, Ebran N, Attias R, Pedeutour F. Fusion encoding a key regulator of stem cell pluripotency, is fused to of EWSR1 with the DUX4 facioscapulohumeral muscular EWSR1 in hidradenoma of the skin and mucoepidermoid dystrophy region resulting from t(4;22)(q35;q12) in a case of carcinoma of the salivary glands. J Pathol. 2008 embryonal rhabdomyosarcoma. Cancer Genet Cytogenet. May;215(1):78-86 2009 Nov;195(1):12-8 Riggi N, Suvà ML, Suvà D, Cironi L, Provero P, Tercier S, Szuhai K, Ijszenga M, de Jong D, Karseladze A, Tanke HJ, Joseph JM, Stehle JC, Baumer K, Kindler V, Stamenkovic I. Hogendoorn PC. The NFATc2 gene is involved in a novel EWS-FLI-1 expression triggers a Ewing's sarcoma initiation cloned translocation in a Ewing sarcoma variant that couples program in primary human mesenchymal stem cells. Cancer its function in immunology to oncology. Clin Cancer Res. 2009 Res. 2008 Apr 1;68(7):2176-85 Apr 1;15(7):2259-68 Tannukit S, Wen X, Wang H, Paine ML. TFIP11, CCNL1 and Tan AY, Manley JL. The TET family of proteins: functions and EWSR1 Protein-protein Interactions, and Their Nuclear roles in disease. J Mol Cell Biol. 2009 Dec;1(2):82-92 Localization. Int J Mol Sci. 2008;9(8):1504-1514 Thompson LD. Olfactory neuroblastoma. Head Neck Pathol. Beauchamp E, Bulut G, Abaan O, Chen K, Merchant A, Matsui 2009 Sep;3(3):252-9 W, Endo Y, Rubin JS, Toretsky J, Uren A. GLI1 is a direct transcriptional target of EWS-FLI1 oncoprotein. J Biol Chem. Wang WL, Mayordomo E, Zhang W, Hernandez VS, Tuvin D, 2009 Apr 3;284(14):9074-82 Garcia L, Lev DC, Lazar AJ, López-Terrada D. Detection and characterization of EWSR1/ATF1 and EWSR1/CREB1 Berg T, Kalsaas AH, Buechner J, Busund LT. Ewing sarcoma- chimeric transcripts in clear cell sarcoma (melanoma of soft peripheral neuroectodermal tumor of the kidney with a FUS- parts). Mod Pathol. 2009 Sep;22(9):1201-9 ERG fusion transcript. Cancer Genet Cytogenet. 2009 Oct;194(1):53-7 Erkizan HV, Uversky VN, Toretsky JA. Oncogenic partnerships: EWS-FLI1 protein interactions initiate key Brandal P, Panagopoulos I, Bjerkehagen B, Heim S. pathways of Ewing's sarcoma. Clin Cancer Res. 2010 Aug t(19;22)(q13;q12) Translocation leading to the novel fusion 15;16(16):4077-83 gene EWSR1-ZNF444 in soft tissue myoepithelial carcinoma. Genes Chromosomes Cancer. 2009 Dec;48(12):1051-6 Jedlicka P. Ewing Sarcoma, an enigmatic malignancy of likely progenitor cell origin, driven by transcription factor oncogenic Erkizan HV, Kong Y, Merchant M, Schlottmann S, Barber- fusions. Int J Clin Exp Pathol. 2010 Mar 19;3(4):338-47 Rotenberg JS, Yuan L, Abaan OD, Chou TH, Dakshanamurthy S, Brown ML, Uren A, Toretsky JA. A small molecule blocking Navarro D, Agra N, Pestaña A, Alonso J, González-Sancho oncogenic protein EWS-FLI1 interaction with RNA helicase A JM. The EWS/FLI1 oncogenic protein inhibits expression of the inhibits growth of Ewing's sarcoma. Nat Med. 2009 Wnt inhibitor DICKKOPF-1 gene and antagonizes beta- Jul;15(7):750-6 catenin/TCF-mediated transcription. Carcinogenesis. 2010 Mar;31(3):394-401 Gervasini C.. CREBBP (CREB binding protein). Atlas Genet Cytogenet Oncol Haematol. March 2009. URL: Ozsahin M, Gruber G, Olszyk O, Karakoyun-Celik O, Pehlivan http://AtlasGeneticsOncology.org/Genes/CBPID42.html B, Azria D, Roelandts M, Kaanders JH, Cengiz M, Krengli M, Matzinger O, Zouhair A. Outcome and prognostic factors in Joo J, Christensen L, Warner K, States L, Kang HG, Vo K, olfactory neuroblastoma: a rare cancer network study. Int J Lawlor ER, May WA. GLI1 is a central mediator of EWS/FLI1 Radiat Oncol Biol Phys. 2010 Nov 15;78(4):992-7 signaling in Ewing tumors. PLoS One. 2009 Oct 27;4(10):e7608 Riggi N, Suvà ML, De Vito C, Provero P, Stehle JC, Baumer K, Cironi L, Janiszewska M, Petricevic T, Suvà D, Tercier S, Kinsey M, Smith R, Iyer AK, McCabe ER, Lessnick SL. Joseph JM, Guillou L, Stamenkovic I. EWS-FLI-1 modulates EWS/FLI and its downstream target NR0B1 interact directly to miRNA145 and SOX2 expression to initiate mesenchymal modulate transcription and oncogenesis in Ewing's sarcoma. stem cell reprogramming toward Ewing sarcoma cancer stem Cancer Res. 2009 Dec 1;69(23):9047-55 cells. Genes Dev. 2010 May;24(9):916-32 Leemann-Zakaryan RP, Pahlich S, Sedda MJ, Quero L, Rocchi A, Manara MC, Sciandra M, Zambelli D, Nardi F, Grossenbacher D, Gehring H. Dynamic subcellular localization Nicoletti G, Garofalo C, Meschini S, Astolfi A, Colombo MP, of the Ewing sarcoma proto-oncoprotein and its association Lessnick SL, Picci P, Scotlandi K. CD99 inhibits neural with and stabilization of microtubules. J Mol Biol. 2009 Feb differentiation of human Ewing sarcoma cells and thereby 13;386(1):1-13 contributes to oncogenesis. J Clin Invest. 2010 Mar 1;120(3):668-80 Miyagawa Y, Okita H, Itagaki M, Toyoda M, Katagiri YU, Fujimoto J, Hata J, Umezawa A, Kiyokawa N. EWS/ETS Romeo S, Dei Tos AP. Soft tissue tumors associated with regulates the expression of the Dickkopf family in Ewing family EWSR1 translocation. Virchows Arch. 2010 Feb;456(2):219-34 tumor cells. PLoS One. 2009;4(2):e4634 Sohn EJ, Li H, Reidy K, Beers LF, Christensen BL, Lee SB. Reichek J, Barr FG.. Soft tissue tumors: Rhabdomyosarcoma. EWS/FLI1 oncogene activates caspase 3 transcription and Atlas Genet Cytogenet Oncol Haematol. January 2009. URL: triggers apoptosis in vivo. Cancer Res. 2010 Feb 1;70(3):1154- http://AtlasGeneticsOncology.org/Tumors/rhab5004.html. 63

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 406 EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Tanas MR, Rubin BP, Montgomery EA, Turner SL, Cook JR, 553 cases in Chinese literature. World J Gastroenterol. 2010 Tubbs RR, Billings SD, Goldblum JR. Utility of FISH in the Mar 14;16(10):1209-14 diagnosis of angiomatoid fibrous histiocytoma: a series of 18 cases. Mod Pathol. 2010 Jan;23(1):93-7 This article should be referenced as such: Yu PF, Hu ZH, Wang XB, Guo JM, Cheng XD, Zhang YL, Xu Huret JL. EWSR1 (Ewing sarcoma breakpoint region 1). Atlas Q. Solid pseudopapillary tumor of the pancreas: a review of Genet Cytogenet Oncol Haematol. 2011; 15(5):395-407.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 407 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

FAM57A (family with sequence similarity 57, member A) Zhiao Chen, Xianghuo He State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China (ZC, XH)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/FAM57AID40183ch17p13.html DOI: 10.4267/2042/45013 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Expression CT120 is universally expressed in different human Other names: CT120, FLJ22282 normal tissues and in various human tumor cell lines. HGNC (Hugo): FAM57A Localisation Location: 17p13.3 CT120 is a novel plasma membrane-associated gene. DNA/RNA Function CT120 may assume very essential physiological Description functions involving in amino acid transport and Gene size: 2145 bp in length, ORF 774 bp. glutathione metabolism through interaction with Full-length cDNA of CT120/FAM57A contains 2145 SLC3A2 and GGTL3B. base pairs and encodes a protein with 257 amino acids. Homology Transcription Homology comparison revealed that CT120 is highly The CT120 contains two isoforms in human: one conserved during biological evolution. isoform identified was termed CT120A; another isoform (AAH26023.1) was named CT120B, which Implicated in consists of four exons and encodes a protein with 225 amino acids (the fourth exon in CT120A is spliced). Lung cancer Prognosis Protein CT120A protein was a potential molecular target for treatment of lung cancers. CT120A was overexpressed in 15 cases of the 16 primary lung cancer specimens. Knockdown of CT120A by small hairpin RNA in the human lung adenocarcinoma cell line SPC-A-1 cells resulted in a reduced cell growth rate in vitro and decrease of the capacity for anchorage-independent growth and tumorigenicity in nude mice. The suppression of CT120A expression also sensitized cells to ultraviolet-induced apoptosis. Atlas cDNA Description expression array revealed that the expressions of - CT120: 257 aa; 29 kDa. several apoptosis- and growth-associated genes were - CT120B: 225 aa; 25 kDa. altered underlying the molecular mechanisms of these cell biological behaviors.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 408 FAM57A (family with sequence similarity 57, member A) Chen Z, He X

Oncogenesis References CT120 ectopic expression could promote cell proliferation activity of NIH3T3 cells, and two major He X, Di Y, Li J, Xie Y, Tang Y, Zhang F, Wei L, Zhang Y, Qin signaling pathways involved in cell proliferation, cell W, Huo K, Li Y, Wan D, Gu J. Molecular cloning and characterization of CT120, a novel membrane-associated gene survival and anti-apoptosis were overexpressed and involved in amino acid transport and glutathione metabolism. activated in response to CT120: one is the Biochem Biophys Res Commun. 2002 Sep 27;297(3):528-36 Raf/MEK/Erk signal cascades and the other is the He XH, Li JJ, Xie YH, Tang YT, Yao GF, Qin WX, Wan DF, Gu PI3K/Akt signal cascades, suggesting that CT120 JR. Altered gene expression profiles of NIH3T3 cells regulated might contribute, at least in part, to the constitutively by human lung cancer associated gene CT120. Cell Res. 2004 activation of Erk and Akt in human lung cancer cells. Dec;14(6):487-96 In addition, some tumor metastasis associated genes Pan DN, Li JJ, Wei L, Yao M, Wan DF, Gu JR. Inhibitory effect cathepsin B, cathepsin D, cathepsin L, MMP-2/TIMP-2 of CT120B, an alternative splice variant of CT120A, on lung were also upregulated by CT120, upon which CT120 cancer cell growth. Acta Biochim Biophys Sin (Shanghai). 2005 might be involved in tumor invasiveness and Sep;37(9):588-92 metastasis. Pan D, Wei L, Yao M, Wan D, Gu J. Down-regulation of In addition, CT120 might play an important role in CT120A by RNA interference suppresses lung cancer cells growth and sensitizes to ultraviolet-induced apoptosis. Cancer tumor progression through modulating the expression Lett. 2006 Apr 8;235(1):26-33 of some candidate "lung tumor progression" genes Li Z, Shao S, Xie S, Jiao F, Ma Y, Shi S. Silencing of CT120 by including B-Raf, Rab-2, BAX, BAG-1, YB-1 and antisense oligonucleotides could inhibit the lung cancer cells Cdc42. growth. Ir J Med Sci. 2010 Jun;179(2):217-23

This article should be referenced as such: Chen Z, He X. FAM57A (family with sequence similarity 57, member A). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):408-409.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 409 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Hideyuki Ihara, Cong-xiao Gao, Yoshitaka Ikeda, Naoyuki Taniguchi Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine, Saga, Japan (HI, YI); Department of Disease Glycomics, Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan (CXG, NT); Disease Glycomics Team, Systems Glycobiology Research Group, Chemical Biology Department, Japan (NT)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/FUT8ID40649ch14q23.html DOI: 10.4267/2042/45014 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Transcription Some splicing variants of the 5'-untranslated region Other names: MGC26465 arise in a developmental stage-specific and tissue- HGNC (Hugo): FUT8 specific manner (Martinez-Duncker et al., 2004). At Location: 14q23.3 least three different promoters appear to be functional in regulating the expression of the FUT8 gene. Three DNA/RNA transcripts with different 5'-untranslated regions have been identified. With respect to coding region, four Description variants were reported to encode polypeptides Human FUT8 gene is located on chromosome 14q23.3 containing 575, 446, 308 and 169 amino acid residues. (Yamaguchi et al., 1999). This gene encompasses The 575 residue protein is a fully active alpha1,6- approximately 333 kb and contains nine exons with fucosyltransferase, which was first of the variants to be coding regions and three 5'-untranslated exons identified. (Yamaguchi et al., 2000; Martinez-Duncker et al., 2004).

Figure 1. Genomic organization of human FUT8 gene. Exons are represented by vertical bars. Exons denoted by ATG or TAA contain start and stop codons, respectively. These exons also have a part of the noncoding region.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 410 FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

Figure 2. Protein structure of FUT8. CT and TM denote the cytoplasmic tail and the transmembrane domain, respectively. I, II and III represent the conserved motifs in alpha1,2-, alpha1,6- and protein O-fucosyltransferases.

The other variants have not yet been examined for Function enzymatic activity and biological function. The 308 FUT8 catalyzes the transfer of a fucose residue from amino acid variant is known to be expressed in the GDP-fucose to the reducing terminal GlcNAc of Asn- retina (Yamaguchi et al., 2000). linked oligosaccharide (N-glycan) via an alpha1.6- linkage (Figure 3). The resulting fucosyl residue is Protein often refered to as a core fucose. The reaction does not Description require any divalent cations or cofactors. The deletion of the FUT8 gene in mice leads to severe phenotypes FUT8 was purified and cloned as a cDNA from porcine that exhibit growth retardation, lung emphysema and brain and a human gastric cancer cell line (Uozumi et death during postnatal development (Wang et al., al., 1996, Yanagidani et al., 1997). Human FUT8 is 2005). As has been clearly shown in studies using comprised of 575 amino acids, with a calculated knockout mice, the lack of core fucosylation resulted in molecular weight of 66516. FUT8 contains no N- the biological activities of various proteins to be glycosylation sites. This enzyme belongs to the GT23 perturbed (Taniguchi et al., 2006; Takahashi et al., family of the CaZY classification. The structual 2009). Examples of this include the TGF-beta1 receptor analysis of a transmembrane domain-truncated form of (Wang et al., 2005), EGF receptor (Wang et al., 2006), FUT8 showed that the enzyme consists of a catalytic VEGF receptor-2 (Wang et al., 2009), LRP-1 (Lee et domain, an N-terminal coiled-coil domain and a C- al., 2006), E-cadherin (Osumi et al., 2009), alpha3beta1 terminal SH3 domain (Ihara et al., 2007). The catalytic integrin (Zhao et al., 2006), VCAM and alpha4beta1 domain was structurally classfied as a member of the integrin (Li et al., 2008). The binding affinity of the GT-B group of glycosylatransferases. core fucose-deleted TGF-beta receptor to TGF-beta 1 is Expression diminished in fut8-null mice, resulting in the FUT8 gene is widely expressed in human tissues downregulation of TGF-beta 1 signaling (Wang et al., (Martinez-Duncker et al., 2004). The FUT8 gene is 2005). The unusual overexpression of matrix expressed at relatively high levels in the brain, metalloproteinases such as MMP-12 and MMP-13 is placenta, lung, stomach, small intestine and jejunum, associated with the impaired receptor function, and has while pancreas, uterus, kidney and urinary bladder been proposed to cause the lung-destructive exhibit moderate expression. The FUT8 gene is weakly phenotypes. The EGF receptor in fut8 null mice is also expressed in the heart, ileum, colon and spleen. On the affected in terms of its binding affinity to EGF and other hand, the expression is not detectable in the EGF-induced phoshorylation (Wang et al., 2006). normal liver (Miyoshi et al., 1997). These studies strongly suggest that FUT8 and core fucose structures regulate the receptor function. Localisation In addition, core fucosylation was reported to be FUT8 is a typical type II membrane protein and is involved in antibodydependent cellular cytotoxicity localized in the Golgi apparatus. (ADCC) (Shields et al., 2002; Shinkawa et al., 2003). The lack of core fucose of N-glycan in the Fc region of the IgG1 molecule enhances ADCC activity up to 50- 100-fold. This discorvery promises to be useful in the development of antibody therapy in cancer treatment. Homology The sequence identities between human FUT8 and other organisms are as follows : Chimpangee (100%), Dog (97.7%), Cow (97.5%), Pig (95.6%), Rat (96.6%), Mouse (96.5%), Chicken (93.9%), Clawed frog (90.3%), Zebrafish (79.5%), Takifugu (80.2%), Tetraodon (79.8%), Sea squirt Figure 3. The reaction catalysed by FUT8. (23.2%), Fruit fly (43.7%), C. elegans (34.8%).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 411 FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

Eight cysteine residues in the catalytic domain are facilitate core fucosyltion of AFP (Noda et al., 2003). conserved among these species, except for ciona (Ihara Core fucosylation appears to serve as a sorting signal et al., 2007). for a glycoprotein to be directed to the bile, as revealed FUT8 contains three short regions that are highly by the predominant distribution of fucosylated conserved in FUT8, alpha1,2-, bacterial alpha1,6-, and glycoproteins in bile rather than serum (Nakagawa et protein O-fucosyltransferases (Oriol et al., 1999; al., 2006). In fact, the levels of alpha-antitrypsin and Takahashi et al., 2000a; Chazalet et al., 2001; alpha1-acid glycoprotein, both of which are fucosylated Martinez-Duncker et al., 2003). The structual analysis glycoproteins, are quite low in the bile of Fut8-null has shown that these regions are located adjacent to one mice. The loss of polarity in cancer cells is likely to another in the Rossmann fold of FUT8 (Ihara et al., impair the regulated sorting, thus allowing abnormal 2007). In addition, the C-terminal SH3 domain of secretion into the serum. FUT8 is structually similar to the typical SH3 domain Prognosis that is found in many proteins. AFP-L3-positive HCC patients were reported to show a Mutations poor prognosis (Yamashita et al., 1996). Ovarian cancer Note Note One frame-shit mutation and 4 substitution mutants FUT8 activity and mRNA levels are highly and have been identified to date in various SNPs of the specifically elavated in cases of ovarian serous FUT8 gene. The frame shift mutant is due to the adenocarcinoma, as compared to nomal ovary and other insertion of a T at position 2 of the codon for Val-85, types of epithelial ovarian carcinoma (Takahashi et al., resulting in 85-VLEEQLVK-92 being change to 85- 2000b). In addition, core fucosylation levels in VFRRAACter-92. The four substitution mutants are glycoproteins is also significantlly increased in cases of K101Q, L153V, E181G and T267K. These mutants are serous adenocarcinoma tissues. due to A being substituted by C at position 1 of codon 101, C to G at position 1 of codon 153, A to G at Thyroid cancer position 2 of codon 181, and C to A at posision 2 of Note codon 267, respectively. Effects of these substitutions The overexpression of FUT8 occurs in 33.3% of cases on enzymatic activity are not currently known. of papillary carcinoma of the thyroid (Ito et al., 2003), although FUT8 was not expressed in normal follicular Implicated in cells. This overexpression was also shown to be correlated with tumor size and lymph node metastasis. Hepatocellular carcinoma (HCC) These phenomena were not observed in cases of Note follicular carcinoma and anaplastic carcinoma. It is well known that the core fucosylation of alpha- Pancreatic cancer fetoprotein (AFP) is implicated in the development of HCC. AFP is a major fetal plasma protein, and its Note expression is elevated in hepatic diseases such as HCC, Haptoglobin was identified as a highly fucosylated hepatitis and liver cirrhosis (Alpert et al., 1968; glycoprotein in the serum of patients with pancreatic Ruoslahti et al., 1974). The AFP-L3 fraction was cancer (Okuyama et al., 2006). The increment of identified as the core-fucosylated isoform of AFP. The fucosylated haptoglobin was observed in pancreatic elevation in serum and liver tissue was found to be cancer rather than other diseases such as HCC, liver specific to HCC, but was not observed in other liver cirrhosis, gastric cancer and colon cancer, and also diseases (Taketa, 1990; Aoyagi, 1995; Miyoshi et al., appeared to be correlated with the clinical stage. 1999). Structural analyses using lectin blotting and mass Thus, it appears that AFP-L3 could be used as a marker spectrometry showed that core fucosylation as well as for HCC. The FUT8 gene is not expressed in the alpha1,3/4-fucosylation is increased in haptoglobin normal adult liver, but is highly expressed in HCC from the serum of such patients. In addition, it was tissue. Surprisingly, however, such an elevation was shown that interleukin 6 expressed in pancreatic cancer also observed in liver cirrhosis in spite of the absence is a possible inducing factor for increasing the of a concomitant increase in AFP-L3 levels (Noda et production of fucosylated haptoglobin in the liver al., 1998). This discrepancy can be attributed to the (Narisada et al., 2008). difference in the synthesis of GDP-fucose, a glycosyl Colorectal cancer donor substrate for fucosyltransferases, including FUT8, and by the altered intracellular sorting of Note fucosylated glycoproteins in hepatic cells. Because the The enzymatic activity and protein expression of FUT8 intracellular concentration of GDP-fucose is higher in were increased in tumor tissues of human colorectal HCC, as compared to a normal liver, chronic hepatitis carcinoma, but not in healthy tissues (Muinelo-Romay and liver cirrhosis, this increase would be expected to et al., 2008). This increment was well observed in cases

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 412 FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

of male, polypoid growth, no regional lymph node vertebrates, invertebrates, and bacteria. Glycobiology. 1999 metastasis and early clinical stage. In addition, Apr;9(4):323-34 immunohistochemical examination has demonstrated Yamaguchi Y, Fujii J, Inoue S, Uozumi N, Yanagidani S, Ikeda that FUT8 is expressed at higher levels in tumor tissues Y, Egashira M, Miyoshi O, Niikawa N, Taniguchi N. Mapping of the alpha-1,6-fucosyltransferase gene, FUT8, to human of colorectal carcinoma than in healthy and transitional chromosome 14q24.3. Cytogenet Cell Genet. 1999;84(1-2):58- tissues. 60 Cystic fibrosis Takahashi T, Ikeda Y, Miyoshi E, Yaginuma Y, Ishikawa M, Taniguchi N. alpha1,6fucosyltransferase is highly and Note specifically expressed in human ovarian serous Fucosylation is known to be increased in cystic adenocarcinomas. Int J Cancer. 2000 Dec 15;88(6):914-9 fibrosis. The alpha1,6-fucosylation of a membrane Takahashi T, Ikeda Y, Tateishi A, Yamaguchi Y, Ishikawa M, glycoprotein is elevated in cystic fibrosis fibroblast Taniguchi N. A sequence motif involved in the donor substrate (Wang et al., 1990). binding by alpha1,6-fucosyltransferase: the role of the conserved arginine residues. Glycobiology. 2000 References May;10(5):503-10 Yamaguchi Y, Ikeda Y, Takahashi T, Ihara H, Tanaka T, Sasho Alpert ME, Uriel J, de Nechaud B. Alpha-1 fetoglobulin in the C, Uozumi N, Yanagidani S, Inoue S, Fujii J, Taniguchi N. diagnosis of human hepatoma. N Engl J Med. 1968 May Genomic structure and promoter analysis of the human alpha1, 2;278(18):984-6 6-fucosyltransferase gene (FUT8). Glycobiology. 2000 Ruoslahti E, Salaspuro M, Pihko H, Andersson L, Seppälä M. Jun;10(6):637-43 Serum alpha-fetoprotein: diagnostic significance in liver Shields RL, Lai J, Keck R, O'Connell LY, Hong K, Meng YG, disease. Br Med J. 1974 Jun 8;2(5918):527-9 Weikert SH, Presta LG. Lack of fucose on human IgG1 N- Taketa K. Alpha-fetoprotein: reevaluation in hepatology. linked oligosaccharide improves binding to human Fcgamma Hepatology. 1990 Dec;12(6):1420-32 RIII and antibody-dependent cellular toxicity. J Biol Chem. 2002 Jul 26;277(30):26733-40 Wang YM, Hare TR, Won B, Stowell CP, Scanlin TF, Glick MC, Hård K, van Kuik JA, Vliegenthart JF. Additional fucosyl Ito Y, Miyauchi A, Yoshida H, Uruno T, Nakano K, Takamura residues on membrane glycoproteins but not a secreted Y, Miya A, Kobayashi K, Yokozawa T, Matsuzuka F, Taniguchi glycoprotein from cystic fibrosis fibroblasts. Clin Chim Acta. N, Matsuura N, Kuma K, Miyoshi E. Expression of alpha1,6- 1990 May;188(3):193-210 fucosyltransferase (FUT8) in papillary carcinoma of the thyroid: its linkage to biological aggressiveness and anaplastic Aoyagi Y. Carbohydrate-based measurements on alpha- transformation. Cancer Lett. 2003 Oct 28;200(2):167-72 fetoprotein in the early diagnosis of hepatocellular carcinoma. Glycoconj J. 1995 Jun;12(3):194-9 Noda K, Miyoshi E, Gu J, Gao CX, Nakahara S, Kitada T, Honke K, Suzuki K, Yoshihara H, Yoshikawa K, Kawano K, Uozumi N, Yanagidani S, Miyoshi E, Ihara Y, Sakuma T, Gao Tonetti M, Kasahara A, Hori M, Hayashi N, Taniguchi N. CX, Teshima T, Fujii S, Shiba T, Taniguchi N. Purification and Relationship between elevated FX expression and increased cDNA cloning of porcine brain GDP-L-Fuc:N-acetyl-beta-D- production of GDP-L-fucose, a common donor substrate for glucosaminide alpha1-->6fucosyltransferase. J Biol Chem. fucosylation in human hepatocellular carcinoma and hepatoma 1996 Nov 1;271(44):27810-7 cell lines. Cancer Res. 2003 Oct 1;63(19):6282-9 Yamashita F, Tanaka M, Satomura S, Tanikawa K. Prognostic Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda significance of Lens culinaris agglutinin A-reactive alpha- Y, Sakurada M, Uchida K, Anazawa H, Satoh M, Yamasaki M, fetoprotein in small hepatocellular carcinomas. Hanai N, Shitara K. The absence of fucose but not the Gastroenterology. 1996 Oct;111(4):996-1001 presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical Miyoshi E, Uozumi N, Noda K, Hayashi N, Hori M, Taniguchi role of enhancing antibody-dependent cellular cytotoxicity. J N. Expression of alpha1-6 fucosyltransferase in rat tissues and Biol Chem. 2003 Jan 31;278(5):3466-73 human cancer cell lines. Int J Cancer. 1997 Sep 17;72(6):1117-21 Martinez-Duncker I, Michalski JC, Bauvy C, Candelier JJ, Mennesson B, Codogno P, Oriol R, Mollicone R. Activity and Yanagidani S, Uozumi N, Ihara Y, Miyoshi E, Yamaguchi N, tissue distribution of splice variants of alpha6- Taniguchi N. Purification and cDNA cloning of GDP-L-Fuc:N- fucosyltransferase in human embryogenesis. Glycobiology. acetyl-beta-D-glucosaminide:alpha1-6 fucosyltransferase 2004 Jan;14(1):13-25 (alpha1-6 FucT) from human gastric cancer MKN45 cells. J Biochem. 1997 Mar;121(3):626-32 Wang X, Inoue S, Gu J, Miyoshi E, Noda K, Li W, Mizuno- Horikawa Y, Nakano M, Asahi M, Takahashi M, Uozumi N, Noda K, Miyoshi E, Uozumi N, Yanagidani S, Ikeda Y, Gao C, Ihara S, Lee SH, Ikeda Y, Yamaguchi Y, Aze Y, Tomiyama Y, Suzuki K, Yoshihara H, Yoshikawa K, Kawano K, Hayashi N, Fujii J, Suzuki K, Kondo A, Shapiro SD, Lopez-Otin C, Kuwaki Hori M, Taniguchi N. Gene expression of alpha1-6 T, Okabe M, Honke K, Taniguchi N. Dysregulation of TGF- fucosyltransferase in human hepatoma tissues: a possible beta1 receptor activation leads to abnormal lung development implication for increased fucosylation of alpha-fetoprotein. and emphysema-like phenotype in core fucose-deficient mice. Hepatology. 1998 Oct;28(4):944-52 Proc Natl Acad Sci U S A. 2005 Nov 1;102(44):15791-6 Miyoshi E, Noda K, Yamaguchi Y, Inoue S, Ikeda Y, Wang W, Lee SH, Takahashi M, Honke K, Miyoshi E, Osumi D, Ko JH, Uozumi N, Li W, Taniguchi N. The alpha1-6- Sakiyama H, Ekuni A, Wang X, Inoue S, Gu J, Kadomatsu K, fucosyltransferase gene and its biological significance. Biochim Taniguchi N. Loss of core fucosylation of low-density Biophys Acta. 1999 Dec 6;1473(1):9-20 lipoprotein receptor-related protein-1 impairs its function, Oriol R, Mollicone R, Cailleau A, Balanzino L, Breton C. leading to the upregulation of serum levels of insulin-like Divergent evolution of fucosyltransferase genes from growth factor-binding protein 3 in Fut8-/- mice. J Biochem. 2006 Mar;139(3):391-8

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 413 FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

Nakagawa T, Uozumi N, Nakano M, Mizuno-Horikawa Y, fucosyltransferase deficient mice. Glycobiology. 2008 Okuyama N, Taguchi T, Gu J, Kondo A, Taniguchi N, Miyoshi Jan;18(1):114-24 E. Fucosylation of N-glycans regulates the secretion of hepatic glycoproteins into bile ducts. J Biol Chem. 2006 Oct Muinelo-Romay L, Vázquez-Martín C, Villar-Portela S, Cuevas 6;281(40):29797-806 E, Gil-Martín E, Fernández-Briera A. Expression and enzyme activity of alpha(1,6)fucosyltransferase in human colorectal Okuyama N, Ide Y, Nakano M, Nakagawa T, Yamanaka K, cancer. Int J Cancer. 2008 Aug 1;123(3):641-6 Moriwaki K, Murata K, Ohigashi H, Yokoyama S, Eguchi H, Ishikawa O, Ito T, Kato M, Kasahara A, Kawano S, Gu J, Narisada M, Kawamoto S, Kuwamoto K, Moriwaki K, Taniguchi N, Miyoshi E. Fucosylated haptoglobin is a novel Nakagawa T, Matsumoto H, Asahi M, Koyama N, Miyoshi E. marker for pancreatic cancer: a detailed analysis of the Identification of an inducible factor secreted by pancreatic oligosaccharide structure and a possible mechanism for cancer cell lines that stimulates the production of fucosylated fucosylation. Int J Cancer. 2006 Jun 1;118(11):2803-8 haptoglobin in hepatoma cells. Biochem Biophys Res Commun. 2008 Dec 19;377(3):792-6 Taniguchi N, Miyoshi E, Gu J, Honke K, Matsumoto A. Decoding sugar functions by identifying target glycoproteins. Osumi D, Takahashi M, Miyoshi E, Yokoe S, Lee SH, Noda K, Curr Opin Struct Biol. 2006 Oct;16(5):561-6 Nakamori S, Gu J, Ikeda Y, Kuroki Y, Sengoku K, Ishikawa M, Taniguchi N. Core fucosylation of E-cadherin enhances cell- Wang X, Gu J, Ihara H, Miyoshi E, Honke K, Taniguchi N. Core cell adhesion in human colon carcinoma WiDr cells. Cancer fucosylation regulates epidermal growth factor receptor- Sci. 2009 May;100(5):888-95 mediated intracellular signaling. J Biol Chem. 2006 Feb 3;281(5):2572-7 Takahashi M, Kuroki Y, Ohtsubo K, Taniguchi N. Core fucose and bisecting GlcNAc, the direct modifiers of the N-glycan Zhao Y, Itoh S, Wang X, Isaji T, Miyoshi E, Kariya Y, Miyazaki core: their functions and target proteins. Carbohydr Res. 2009 K, Kawasaki N, Taniguchi N, Gu J. Deletion of core Aug 17;344(12):1387-90 fucosylation on alpha3beta1 integrin down-regulates its functions. J Biol Chem. 2006 Dec 15;281(50):38343-50 Wang X, Fukuda T, Li W, Gao CX, Kondo A, Matsumoto A, Miyoshi E, Taniguchi N, Gu J. Requirement of Fut8 for the Ihara H, Ikeda Y, Toma S, Wang X, Suzuki T, Gu J, Miyoshi E, expression of vascular endothelial growth factor receptor-2: a Tsukihara T, Honke K, Matsumoto A, Nakagawa A, Taniguchi new mechanism for the emphysema-like changes observed in N. Crystal structure of mammalian alpha1,6-fucosyltransferase, Fut8-deficient mice. J Biochem. 2009 May;145(5):643-51 FUT8. Glycobiology. 2007 May;17(5):455-66 This article should be referenced as such: Li W, Ishihara K, Yokota T, Nakagawa T, Koyama N, Jin J, Mizuno-Horikawa Y, Wang X, Miyoshi E, Taniguchi N, Kondo Ihara H, Gao CX, Ikeda Y, Taniguchi N. FUT8 A. Reduced alpha4beta1 integrin/VCAM-1 interactions lead to (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)). Atlas impaired pre-B cell repopulation in alpha 1,6- Genet Cytogenet Oncol Haematol. 2011; 15(5):410-414.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 414 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

IGSF8 (immunoglobulin superfamily, member 8) Yanhui H Zhang, Mekel M Richardson, Xin A Zhang Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, USA (YHZ, MMR, XAZ)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/IGSF8ID45820ch1q23.html DOI: 10.4267/2042/45015 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity DNA/RNA Other names: CD316, CD81P3, EWI-2, EWI2, KCT- Description 4, LIR-D1, PGRL Gene type: protein coding. HGNC (Hugo): IGSF8 Gene size: 7604 bp, 7 exons. Location: 1q23.2 Transcription Local order: Centromere -- PIGM - KCNJ10 - mRNA 2366 bp (length may vary for alternative LOC100287448 - KCNJ9 - CD316/IGSF8 - ATP1A2 - splicing forms). ATP1A4 - CASQ1 - PEA15 -- Telomere (NCBI Map Viewer).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 415 IGSF8 (immunoglobulin superfamily, member 8) Zhang YH, et al.

CD316/IGSF8 gene typically contains 7 exons. The green bars represent the non-coding exons while the blue bars represent the coding ones. The length of each intron is indicated above and the size of each exon is indicated below (information sourced from Ensembl (ENSG00000162729)).

There are 5 transcripts (Ensembl).

Name Transcript ID Length (bp) Protein ID Length (aa) Exon

IGSF8-001 ENST00000314485 2343 ENSP00000316664 613 7 IGSF8-002 ENST00000448417 1059 ENSP00000397464 301 4 IGSF8-003 ENST00000368086 2366 ENSP00000357065 613 7 IGSF8-201 ENST00000358475 2004 ENSP00000351261 526 8 IGSF8-004 ENST00000460351 876 No protein product - 2

There are 7 putative alternative splicing forms (AceView).

NCBI 36, mRNA Protein size IGSF8 alternative variant Exons March 2006 Aa pI size (bp) (kDa) genome (kb) aApr07 2352 7 7.36 674 71.8 8.2 bApr07 2304 8 7.60 664 70.4 8.3 cApr07 1995 4 2.92 311 32.7 8.2 dApr07 1005 4 5.58 283 30.5 6.7 eApr07-unspliced 6184 1 6.18 166 18.1 6.7 fApr07 (partial mRNA) 571 4 4.71 165 17.9 7.3 0.58 91 9.6 11.5 variant gApr07-unspliced 581 1 (partial mRNA) Appear non Appear non Appear non Appear non coding coding coding coding

Pseudogene CD316 protein is highly expressed in human brain (cortex, white matter, hippocampus and cerebellum), IGSF8-004, an alternative splicing form of astrocytes, hepatocytes and lymphoid cells (majority of IGSF8/CD316, is predicted to have no protein product B-cells, T-cells and natural killer cells, but not on (Ensembl). monocytes, polynuclear cells and platelets). CD316 is constitutively expressed on plasmacytoid dendritic cells Protein and on cord blood-derived Langerhans-like cells. Upon Description stimulation, CD316 is expressed on monocytes, monocytes derived dendritic cells and myeloid 613 amino acids, molecular weight is 65034 Da. dendritic cells. Basal isoelectric point: 8.23 (PhosphoSitePlus). Localisation Expression Plasma membrane, cell-cell contacts, microvilli. CD316 mRNA is ubiquitously expressed in human tissues, with high expression in brain, kidney, testis, Function liver and placenta, with low expression in peripheral 1. Suppresses cell movement and cell aggregation. blood cells, lung, and skeletal muscle. 2. Regulates integrin alpha3beta1- and alpha4beta1- dependent cell morphology and cell spreading.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 416 IGSF8 (immunoglobulin superfamily, member 8) Zhang YH, et al.

3. May participate in the regulation of neurite outgrowth and maintenance of the neural network in Mutations the adult brain. Note 4. Interacts with its ligand, HSPA8, and may influence Currently there is no known disease-related or the behavior of dendritic cells and control biologically significant mutation (see HGMD). adaptive immune response. 5. Links tetraspanin web to the actin cytoskeleton Implicated in through direct associations with ezrin-radixin-moesin proteins. Glioma or glioblastoma 6. Inhibits glioblastoma growth in vitro and in vivo. Note 7. EWI-2 wint inhibits hepatitis C virus entry. CD316 inhibits glioblastoma growth in vitro and in 8. May play a role in fertilization. Lack of CD316 vivo. Loss of CD316 expression correlates with a present at the cell surface of CD9-null oocytes may shorter survival time in human glioma patients contribute to the loss of ability of CD9-null oocytes to (Kolesnikova et al., 2009). fuse with sperms. CD316 typically inhibits cell migration and negatively Hepatitis and liver cancer regulates cell proliferation. It associates with Note tetraspanins CD9, CD81, and CD82 and likely Hepatitis C virus (HCV)-infected population has higher contributes to various functions of these associated risk of developing liver cancer. Ectopic expression of tetraspanins. It also regulates the functions of EWI-2wint, i.e., EWI-2 without its N-terminus, can alpha3beta1 and alpha4beta1 integrins, probably inhibit HCV entry and reduce HCV infection (Rocha- through its associated tetraspanins (Clark et al., 2001; Perugini et al., 2008). Stipp et al., 2001; Stipp et al., 2003; Zhang et al., 2003; Kolesnikova et al., 2004; Kolesnikova et al., 2009; Autoimmune diseases Sala-Valdés et al., 2006). Note It was reported that CD316 is an inducible receptor of HSPA8 on human dendritic cells, it may control the adaptive immune response through its influence on the behavior of dendritic cells. Therefore it maybe utilized in the treatment of antoimmune diseases such as rheumatoid arthritis (Kettner et al., 2007). Infertility Note CD316 plays a role in fertilization. Oocytes from CD9 null mice cannot fuse with sperm. The level of CD316 proteins on the CD9-null oocyte surface is less than 10% of that on the wild-type one. The loss of CD316 on the CD9-null oocyte surface may be responsible for the loss of fusion ability (He et al., 2009; Glazar et al., 2009). Type 2 diabetes mellitus Note CD316 is a candidate for human disorders on 1q22-

Type I transmembrane protein CD316 contains an ectodomain q23, including type 2 diabetes mellitus (Murdoch et al., that consists largely of four immunoglobulin domains, a 2003). transmembrane region, and a positively charged, 10-amino acid residue cytoplasmic tail. Glycosylation sites are found in the ectodomain and palmitoylation sites in the cytoplasmic domain. To be noted CD316 is constitutively palmitoylated and linked to actin cytoskeleton through direct association of its cytoplasmic Note domain with ezrin-radixin-moesin proteins. CD316 associates EWI-2wint, a cleavage product of EWI-2 in which the with tetraspanins such as CD9, CD81, and CD82. first Ig-domain of the 4 extracellular Ig-domains is Homology cleaved off. CD316 protein is conserved in chimpanzee, cow, References mouse, rat, and zebrafish and belongs to the EWI subfamily of Ig superfamily. Other human EWI Clark KL, Zeng Z, Langford AL, Bowen SM, Todd SC. PGRL is subfamily proteins include FPRP/CD9P-1, IGSF3, and a major CD81-associated protein on lymphocytes and CD101.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 417 IGSF8 (immunoglobulin superfamily, member 8) Zhang YH, et al.

distinguishes a new family of cell surface proteins. J Immunol. Yang XH, Kovalenko OV, Kolesnikova TV, Andzelm MM, 2001 Nov 1;167(9):5115-21 Rubinstein E, Strominger JL, Hemler ME. Contrasting effects of EWI proteins, integrins, and protein palmitoylation on cell Stipp CS, Kolesnikova TV, Hemler ME. EWI-2 is a major CD9 surface CD9 organization. J Biol Chem. 2006 May and CD81 partner and member of a novel Ig protein subfamily. 5;281(18):12976-85 J Biol Chem. 2001 Nov 2;276(44):40545-54 Kettner S, Kalthoff F, Graf P, Priller E, Kricek F, Lindley I, Charrin S, Le Naour F, Labas V, Billard M, Le Caer JP, Emile Schweighoffer T. EWI-2/CD316 is an inducible receptor of JF, Petit MA, Boucheix C, Rubinstein E. EWI-2 is a new HSPA8 on human dendritic cells. Mol Cell Biol. 2007 component of the tetraspanin web in hepatocytes and Nov;27(21):7718-26 lymphoid cells. Biochem J. 2003 Jul 15;373(Pt 2):409-21 Rocha-Perugini V, Montpellier C, Delgrange D, Wychowski C, Murdoch JN, Doudney K, Gerrelli D, Wortham N, Paternotte C, Helle F, Pillez A, Drobecq H, Le Naour F, Charrin S, Levy S, Stanier P, Copp AJ. Genomic organization and embryonic Rubinstein E, Dubuisson J, Cocquerel L. The CD81 partner expression of Igsf8, an immunoglobulin superfamily member EWI-2wint inhibits hepatitis C virus entry. PLoS One. 2008 Apr implicated in development of the nervous system and organ 2;3(4):e1866 epithelia. Mol Cell Neurosci. 2003 Jan;22(1):62-74 Charrin S, Yalaoui S, Bartosch B, Cocquerel L, Franetich JF, Stipp CS, Kolesnikova TV, Hemler ME. EWI-2 regulates Boucheix C, Mazier D, Rubinstein E, Silvie O. The Ig domain alpha3beta1 integrin-dependent cell functions on laminin-5. J protein CD9P-1 down-regulates CD81 ability to support Cell Biol. 2003 Dec 8;163(5):1167-77 Plasmodium yoelii infection. J Biol Chem. 2009 Nov Zhang XA, Lane WS, Charrin S, Rubinstein E, Liu L. 13;284(46):31572-8 EWI2/PGRL associates with the metastasis suppressor Delandre C, Penabaz TR, Passarelli AL, Chapes SK, Clem RJ. KAI1/CD82 and inhibits the migration of prostate cancer cells. Mutation of juxtamembrane cysteines in the tetraspanin CD81 Cancer Res. 2003 May 15;63(10):2665-74 affects palmitoylation and alters interaction with other proteins Kolesnikova TV, Stipp CS, Rao RM, Lane WS, Luscinskas FW, at the cell surface. Exp Cell Res. 2009 Jul 1;315(11):1953-63 Hemler ME. EWI-2 modulates lymphocyte integrin alpha4beta1 Glazar AI, Evans JP. Immunoglobulin superfamily member functions. Blood. 2004 Apr 15;103(8):3013-9 IgSF8 (EWI-2) and CD9 in fertilisation: evidence of distinct Little KD, Hemler ME, Stipp CS. Dynamic regulation of a functions for CD9 and a CD9-associated protein in mammalian GPCR-tetraspanin-G protein complex on intact cells: central sperm-egg interaction. Reprod Fertil Dev. 2009;21(2):293-303 role of CD81 in facilitating GPR56-Galpha q/11 association. He ZY, Gupta S, Myles D, Primakoff P. Loss of surface EWI-2 Mol Biol Cell. 2004 May;15(5):2375-87 on CD9 null oocytes. Mol Reprod Dev. 2009 Jul;76(7):629-36 Sala-Valdés M, Ursa A, Charrin S, Rubinstein E, Hemler ME, Kolesnikova TV, Kazarov AR, Lemieux ME, Lafleur MA, Kesari Sánchez-Madrid F, Yáñez-Mó M. EWI-2 and EWI-F link the S, Kung AL, Hemler ME. Glioblastoma inhibition by cell surface tetraspanin web to the actin cytoskeleton through their direct immunoglobulin protein EWI-2, in vitro and in vivo. Neoplasia. association with ezrin-radixin-moesin proteins. J Biol Chem. 2009 Jan;11(1):77-86, 4p following 86 2006 Jul 14;281(28):19665-75 Yamada O, Tamura K, Yagihara H, Isotani M, Washizu T, This article should be referenced as such: Bonkobara M. Neuronal expression of keratinocyte-associated Zhang YH, Richardson MM, Zhang XA. IGSF8 transmembrane protein-4, KCT-4, in mouse brain and its up- (immunoglobulin superfamily, member 8). Atlas Genet regulation by neurite outgrowth of Neuro-2a cells. Neurosci Cytogenet Oncol Haematol. 2011; 15(5):415-418. Lett. 2006 Jan 16;392(3):226-30

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 418 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

MIXL1 (Mix1 homeobox-like 1 (Xenopus laevis)) Aaron Raymond, Lalitha Nagarajan Departement of Genetics, Box 1010, MD Anderson cancer Center, 1515 Holcombe Blvd, Houston Tx 77030, USA (AR, LN)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/MIXL1ID47624ch1q42.html DOI: 10.4267/2042/45016 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity DNA/RNA Other names: MGC138179, MILD1, MIX, MIXL Description HGNC (Hugo): MIXL1 MIXL1 is 2131 bps long and consists of two exons of Location: 1q42.12 length 393 bp and 306 bp, and one intron (Guo et al., Local order: MIXL1 is flanked on its 3' end by Lin9. 2002; Sahr et al., 2002). This proximity is evolutionarily conserved. Transcription MIXL1 is transcribed to a full-length 699 bp mRNA. There are no known splice variants. Pseudogene There are no known pseudogenes for MIXL1.

MIXL1 genomic context on 1q42.12.

Genomic organisation of MIXL1.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 419 MIXL1 (Mix1 homeobox-like 1 (Xenopus laevis)) Raymond A, Nagarajan L

Representation and comparison of Human MIXL1, Mouse Mixl1, Chicken Mixl1, and Xenopus Mix.1 protein domains. The Mix family of proteins all contain evolutionarily conserved homeodomain and C-terminal acidic domains. Human, mouse and chicken also share a conserved N-terminal proline rich domain. While these three domains are highly conserved, the remainder of protein varies significantly between species.

Protein Homology MIXL1 is a member of the Mix/Bix family of Description transcription factors, of which it is the only member MIXL1 is a paired type homeobox protein which has identified in humans. It is also a member of the larger 232 amino acids, and a molecular weight of 27 kDa. grouping of paired type homeoboxes, a family of genes The protein contains three identified domains: a which share sequence similarity in the homeobox proline-rich domain, a paired-type homeobox, and a c- domain with paired box family (PAX). terminal acidic domain. While MIXL1 does have an MIXL1 shares 41% sequence similarity to its chicken expected weight of 27 kDa, it will migrate on a homolog, and 69% to its mouse homolog. Its Western Blot at 36 kDa (Guo et al., 2002). MIXL1 is homeodomain is highly conserved across species, phosphorylated in the amino-terminal region at Tyr20 sharing identity of 66% to that of Xenopus Mix.1, 79% (Guo et al., 2006). to that of chicken Mixl1, and 94% to that of mouse Expression Mixl1. MIXL1 expression is restricted to embryonic Implicated in mesendoderm precursors and adult hematopoietic stem cells and progenitors. Hodgkin's lymphoma Localisation Disease MIXL1 expression is predominantly nuclear. MIXL1 is aberrantly expressed in patient samples derived from Hodgkin's lymphoma, along with the Function following Hodgkin cell lines: L-1236, L-428, HD- MIXL1 is paired-type homeobox transcription factor, MyZ, HD-LM2, MDA-E, MDA-V, KM-H2, and Daudi and as such preferentially binds to the DNA sequence (Drakos et al., 2007). TAAT. MIXL1 homologs preferentially bind as dimers T-cell NHL lymphoma to 11 bp palindromic sequences consisting of two TAAT segments and a three nucleotide spacer (Wilson Disease et al., 1993). MIXL1 is aberrantly expressed in patient samples MIXL1 expression is required for both mesendoderm derived from High Grade T-cell non-Hodgkin's development and hematopoiesis. The MIXL1 homologs lymphoma, along with the following T-cell NHL are necessary intermediate factors to the BMP4 (bone established lines: Karpas 299, MAC2A, SR-786, and morphogenetic protein 4)-mediated mesendoderm Peer (Drakos et al., 2007; Guo et al., 2002). formation, as dominant negative mutants block this B-cell NHL lymphoma pathway (Mead et al., 1996). Development into Disease mesoderm and endoderm cell layers is dependant on MIXL1 is aberrantly expressed in patient samples the expression collaborating factors. derived from High Grade B-cell non-Hodgkin's MIXL1 expression is required for the early stages of lymphoma, along with the following B-cell NHL hematopoiesis and is normally expressed in all early established lines: SKI-DLBL, DB, DOHH1, IM-9, hematopoietic precursor types (Guo et al., 2002).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 420 MIXL1 (Mix1 homeobox-like 1 (Xenopus laevis)) Raymond A, Nagarajan L

Mino, Sp-53, Z-138, and CJ (Drakos et al., 2007; Guo et al., 2002). References Wilson D, Sheng G, Lecuit T, Dostatni N, Desplan C. Acute myeloid leukemia Cooperative dimerization of paired class homeo domains on Disease DNA. Genes Dev. 1993 Nov;7(11):2120-34 Retroviral transduction of Mixl1 into mouse bone Mead PE, Brivanlou IH, Kelley CM, Zon LI. BMP-4-responsive marrow resulted in transplantable acute myeloid regulation of dorsal-ventral patterning by the homeobox protein leukemia in all lethally irradiated recipient mice after a Mix.1. Nature. 1996 Jul 25;382(6589):357-60 latency period (Glaser et al., 2006). Guo W, Chan AP, Liang H, Wieder ED, Molldrem JJ, et al. A The following established AML cell lines aberrantly human Mix-like homeobox gene MIXL shows functional express MIXL1: U937, KG1, and ML3 (Guo et al., similarity to Xenopus Mix.1. Blood. 2002 Jul;100(1):89-95 2002). Hwang HC, Martins CP, Bronkhorst Y, Randel E, Berns A, Fero M, Clurman BE. Identification of oncogenes collaborating Chronic myeloid leukemia with p27Kip1 loss by insertional mutagenesis and high- throughput insertion site analysis. Proc Natl Acad Sci U S A. Disease 2002 Aug 20;99(17):11293-8 MIXL1 is aberrantly expressed in the K562 established cell line (Guo et al., 2002). Sahr K, Dias DC, Sanchez R, Chen D, Chen SW, Gudas LJ, Baron MH. Structure, upstream promoter region, and functional T-cell leukemia domains of a mouse and human Mix paired-like homeobox gene. Gene. 2002 May 29;291(1-2):135-47 Disease Glaser S, Metcalf D, Wu L, Hart AH, DiRago L, et al. Enforced The Mixl1 promoter in mouse was identified as a site expression of the homeobox gene Mixl1 impairs hematopoietic of viral insertion, using the Moloney murine leukemia differentiation and results in acute myeloid leukemia. Proc Natl virus, which collaborates with loss of p27 in induction Acad Sci U S A. 2006 Oct 31;103(44):16460-5 of lymphomagenesis (Hwang et al., 2002). Guo W, Nagarajan L. Amino terminal tyrosine phosphorylation MIXL1 is aberrantly expressed in the following T-cell of human MIXL1. J Mol Signal. 2006 Dec 5;1:6 leukemia established lines: Jurkat, SKW-3, and CEM Drakos E, Rassidakis GZ, Leventaki V, Guo W, Medeiros LJ, (Drakos et al., 2007; Guo et al., 2002). Nagarajan L. Differential expression of the human MIXL1 gene product in non-Hodgkin and Hodgkin lymphomas. Hum Pathol. B-cell leukemia 2007 Mar;38(3):500-7 Disease MIXL1 is aberrantly expressed in the following B-cell This article should be referenced as such: leukemia established lines: NALM6, REH-1 (Drakos et Raymond A, Nagarajan L. MIXL1 (Mix1 homeobox-like 1 al., 2007; Guo et al., 2002). (Xenopus laevis)). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):419-421.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 421 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

PEG3 (paternally expressed 3) Yinhua Yu, Weiwei Feng, Zhen Lu, Robert C Bast Jr The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Box 354, Houston, TX 77030, USA (YY, WF, ZL, JrB)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/PEG3ID41690ch19q13.html DOI: 10.4267/2042/45017 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity HGNC (Hugo): PEG3 Location: 19q13.43 Other names: DKFZp781A095, KIAA0287, PW1, ZNF904, ZSCAN24

Figure A. PEG3 DNA located on chromosome 19, starts at 62013257 and ends at 62043906 bp. Figure B. The PEG3 gene contains 7- 10 exons with 5 different transcripts. Figure C. PEG3 promoter and first exon. The transcription start site and direction are indicated with an arrow. The horizontal bars are 32 CpG sites. The dark blue box is a CpG island surrounded the first exon (Figure modified from Feng et al., 2008).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 422 PEG3 (paternally expressed 3) Yu Y, et al.

breast cancers and other gynecologic cancers (Kohda et DNA/RNA al., 2001; Dowdy et al., 2005; Feng et al., 2008). Description There are several studies revealed murine Peg3 acts as an intermediary between p53 and Bax in a cell death Human PEG3 was originally identified as an ortholog pathway activated by DNA damage in primary mouse of murine Peg3 that is the first imprinted gene detected cortical neurons, inhibiting Peg3 activity blocks p53- in mouse chromosome 7. Human PEG3 is located ~2 induced apoptosis (Johnson et al., 2002). Pw1/Peg3 Mb proximal of the 19q telomere, which is rich in interacts with a p53-inducible gene product Siah1a. Krüppel-type ZNF genes (Kim et al., 1997). Bisulfite Coexpression of Pw1/Peg3 with Siah1a induces sequencing across PEG3 revealed that all CpG apoptosis independently of p53. Inhibiting Pw1/Peg3 dinucleotides examined were differentially methylated activity blocks p53-induced apoptosis (Relaix et al., in human fetal brain, kidney, liver, and pancreas. PEG3 2000). Since human PEG3 is highly conserved with is monoallelically expressed during fetal development, murine Peg3, PEG3 may have same function, Jiang et it is a maternal-imprinted gene (Murphy et al., 2001). al. (2010) demonstrated that enforced overexpression Transcription of PEG3 mRNA during zebrafish embryogenesis The PEG3 gene contains 7-10 exons with 5 different decreased beta-catenin protein expression and inhibited transcripts. Wnt-dependent tail development. Peg3/Pw1 also inhibited Wnt signaling in human cells by binding to Pseudogene beta-catenin and promoting its degradation via a There are no known pseudogenes, but an antisense p53/Siah1-dependent, GSK3beta-independent transcript gene (APEG3), which is transcribed in the proteasomal pathway. Hypermethylation of the PEG3 opposite direction of PEG3, has been identified from promoter in primary human gliomas led to a loss of human. It is confirmed the presence of APEG3 in total imprinting and decreased PEG3 mRNA expression that RNAs from brain thalamus and testis. The functional correlated with increasing tumor grade (Jiang et al., significance of APEG3 is not known. The paternal 2010). allele-specific expression of anti-sense Peg3 The transcription factor YY1 can bind to the first intron (imprinting) was detected in mouse brain, but not in of human PEG3, and specifically to the paternal allele human yet (Choo et al., 2002). of the gene. YY1-binding sites are methylated only on the maternal chromosome. YY1-binding region may Protein function as a methylation-sensitive insulator that could influence the imprinted expression of PEG3 (Kim et al., Description 2003). PEG3 is maternal-imprinted and paternally expressed. Homology The imprinted status of PEG3 throughout development and adult life. PEG3 may be a susceptibility locus for Human PEG3 is known to have orthologs in mice and cancer and for neurobehavioral deficits (Murphy et al., cow. Human PEG3 gene sequences revealed a high 2001). level of conservation with Murine Peg3 (83% similarity), but one of the two proline-rich repeats is Expression absent from the human PEG3 (Kim et al., 1997). PEG3 is expressed at higher levels in ovary and placenta (Kim et al., 1997; Hiby et al., 2001). It was Mutations also strongly expressed in the adult brain (Kohda et al., Note 2001). Mutations have not been detected. Localisation High levels of PEG3 have localized to the layer of Implicated in villous cytotrophoblast cells in the human placenta, PEG3 expression is also detected in the ovary stroma Glioma, glioblastoma (Hiby et al., 2001). Note Function A significant decrease in PEG3 expression was more commonly observed in glioma cell lines as compared PEG3 is an imprinted gene expressed exclusively from with that in primary cultures of astrocytes. Transfection the paternal allele. The precise function of PEG3 is not of PEG3 cDNA in a glioma cell line resulted in a loss clear, but recent evidence suggests that it plays an of tumorigenicity in nude mice (Kohda et al., 2001). important role in the p53/c-myc-mediated apoptosis The epigenetic silencing of PEG3 expression in glioma pathway. cell lines depends on aberrant DNA methylation of an PEG3 is a maternally imprinted tumor suppressor gene exonic CpG island. Treatment of glioma cell lines with that is downregulated in gliomas, ovarian cancers, the DNA demethylating agent 5-aza-2'-deoxycytidine

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 423

PEG3 (paternally expressed 3) Yu Y, et al.

reversed the silencing of PEG3 biallelically (Maegawa Hiby SE, Lough M, Keverne EB, Surani MA, Loke YW, King A. et al., 2001). Re-expression of PEG3 in glioma cells Paternal monoallelic expression of PEG3 in the human placenta. Hum Mol Genet. 2001 May 1;10(10):1093-100 suppresses their proliferation. Hypermethylation of the PEG3 promoter in primary human gliomas led to a loss Kohda T, Asai A, Kuroiwa Y, Kobayashi S, Aisaka K, Nagashima G, Yoshida MC, Kondo Y, Kagiyama N, Kirino T, of imprinting and decreased PEG3 mRNA expression Kaneko-Ishino T, Ishino F. Tumour suppressor activity of that correlated with tumor grade (Jiang et al., 2010). human imprinted gene PEG3 in a glioma cell line. Genes Cells. The lower gene expression was confirmed statistically 2001 Mar;6(3):237-47 in glioblastoma (Otsuka et al., 2009). Maegawa S, Yoshioka H, Itaba N, Kubota N, Nishihara S, Endometrial cancer, cervical cancer, Shirayoshi Y, Nanba E, Oshimura M. Epigenetic silencing of PEG3 gene expression in human glioma cell lines. Mol choriocarcinomas Carcinog. 2001 May;31(1):1-9 Note Murphy SK, Wylie AA, Jirtle RL. Imprinting of PEG3, the PEG3 is silenced in all endometrial and cervical cancer human homologue of a mouse gene involved in nurturing cell lines studied. In contrast, loss of maternal behavior. Genomics. 2001 Jan 1;71(1):110-7 imprinting and relatively high PEG3 expression levels Van den Veyver IB, Norman B, Tran CQ, Bourjac J, Slim R. were detected in all four choriocarcinomas cell lines The human homologue (PEG3) of the mouse paternally expressed gene 3 (Peg3) is maternally imprinted but not studied (Dowdy et al., 2005). mutated in women with familial recurrent hydatidiform molar Breast cancer, ovarian cancer pregnancies. J Soc Gynecol Investig. 2001 Sep-Oct;8(5):305- 13 Note Johnson MD, Wu X, Aithmitti N, Morrison RS. Peg3/Pw1 is a Five of the eight ovarian cancer cell lines were found to mediator between p53 and Bax in DNA damage-induced be PEG3 negative, the remaining three express low neuronal death. J Biol Chem. 2002 Jun 21;277(25):23000-7 levels of PEG3 mRNA (Dowdy et al., 2005). PEG3 Kim J, Kollhoff A, Bergmann A, Stubbs L. Methylation-sensitive was down-regulated in 75% of ovarian cancers. PEG3 binding of transcription factor YY1 to an insulator sequence was hypermethylated in 11 of 42 ovarian cancers within the paternally expressed imprinted gene, Peg3. Hum (26%), and PEG3 expression was down-regulated in 10 Mol Genet. 2003 Feb 1;12(3):233-45 of those 11 cancers. LOH was detected in 5 of 25 Dowdy SC, Gostout BS, Shridhar V, Wu X, Smith DI, Podratz informative cases for PEG3 (20%). Re-expression of KC, Jiang SW. Biallelic methylation and silencing of paternally PEG3 markedly inhibited ovarian cancer growth. PEG3 expressed gene 3 (PEG3) in gynecologic cancer cell lines. Gynecol Oncol. 2005 Oct;99(1):126-34 expression could be restored by treatment with 5-aza- 2'-deoxycytidine and trichostatin A (Feng et al., 2008). Choo JH, Kim JD, Kim J. Imprinting of an evolutionarily conserved antisense transcript gene APeg3. Gene. 2008 Feb Hydatidiform moles 15;409(1-2):28-33 Note Feng W, Marquez RT, Lu Z, Liu J, Lu KH, Issa JP, Fishman PEG3 is not mutated in women with familial recurrent DM, Yu Y, Bast RC Jr. Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently down-regulated in hydatidiform moles, there is allele-specific methylation human ovarian cancers by loss of heterozygosity and promoter of the CpG island and expression from the paternal methylation. Cancer. 2008 Apr 1;112(7):1489-502 allele in two independent informative pedigrees (Van Otsuka S, Maegawa S, Takamura A, Kamitani H, Watanabe T, den Veyver et al., 2001). Oshimura M, Nanba E. Aberrant promoter methylation and expression of the imprinted PEG3 gene in glioma. Proc Jpn References Acad Ser B Phys Biol Sci. 2009;85(4):157-65 Kim J, Ashworth L, Branscomb E, Stubbs L. The human Jiang X, Yu Y, Yang HW, Agar NY, Frado L, Johnson MD. The homolog of a mouse-imprinted gene, Peg3, maps to a zinc imprinted gene PEG3 inhibits Wnt signaling and regulates finger gene-rich region of human chromosome 19q13.4. glioma growth. J Biol Chem. 2010 Mar 12;285(11):8472-80 Genome Res. 1997 May;7(5):532-40 This article should be referenced as such: Relaix F, Wei X, Li W, Pan J, Lin Y, Bowtell DD, Sassoon DA, Wu X. Pw1/Peg3 is a potential cell death mediator and Yu Y, Feng W, Lu Z, Bast RC Jr. PEG3 (paternally expressed cooperates with Siah1a in p53-mediated apoptosis. Proc Natl 3). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):422- Acad Sci U S A. 2000 Feb 29;97(5):2105-10 424.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 424

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

RPL10 (ribosomal protein L10) Mohit Goel, Ranjan Tamuli Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati-781 039, Assam, India (MG, RT)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/RPL10ID42148chXq28.html DOI: 10.4267/2042/45018 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

dispersed in the genome. Moreover, transcript variants Identity utilizing alternative polyA Other names: DKFZp686J1851, DXS648, DXS648E, signals exist; the variant with the longest 3' UTR FLJ23544, FLJ27072, NOV, QM overlaps the deoxyribonuclease I-like 1 gene on the HGNC (Hugo): RPL10 opposite strand. Location: Xq28 Protein DNA/RNA Description Description 214 amino acids; 24604 Da. The protein is a component of the large ribosomal (60S) subunit and DNA size 3.96 kb, mRNA size 2172 bp, 7 exons. The belongs to the L10E family of ribosomal proteins. RPL10 gene is co-transcribed with the small nucleolar Three natural variants of the RPL10 protein, RNA gene U70 that is located in its fifth intron. VAR_006922 (N202S, dbSNP rs4909 and dbSNP Multiple processed pseudogenes of the gene RPL10 are rs12012747), VAR_027795 (L206M), and VAR_027796 (H213Q) have been reported.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 425 RPL10 (ribosomal protein L10) Goel M, Tamuli R

Expression tumors, the positive areas were mostly confined to peripheral aspects of tumors and were particularly Ubiquitous. RPL10 is expressed in a wide variety strong in foci of perineural invasion. These results of embryonic and adult tissues, down-regulated during suggested that decreased RPL10 expression may be adipocyte, kidney, and heart differentiation. associated with early development of prostate cancer, Localisation but later a high level of RPL10 may facilitate Cytoplasm. progression of the tumors to a more aggressive phenotype. Function The ribosomal protein L10 (RPL10), a member of the Ovarian cancer L10E family of ribosomal proteins, is a key protein in Note assembling 60S ribosomal subunit and organizes the Both adenine (A)/guanine (G) replacement was architecture of the aminoacyl-tRNA binding site. detected at the 605th nucleotide which changes the RPL10 was originally identified as QM, a candidate for coding from serine to asparagines in 17 (58.6%) of the a Wilms' tumor suppressor; however, later studies did 29 ovarian tumors studied. The frequencies of A/A, not support the original hypothesis. In vitro studies G/G and A/G homo- or hetero-zygosity were 3.5%, have shown the interaction of RPL10 with the 37.9% and 58.6%, respectively in cancer tissues but transcript regulator the c-Jun, as well as with the proto- they were 26.1%, 52.2% and 21.7%, respectively in the oncogene c-Yes; however, these interactions yet to adjacent normal tissues, indicating a higher demonstrate in vivo. heterozygous rate in cancer (58.6% vs 21.7%, p<0.01). Homology These results suggest that high frequencies of loss of the A/G heterozygosity at the 605th nt of the RPL10 The percent identity below represents identity of gene may be associated with ovarian cancer. RPL10 over an aligned region in UniGene. - Mus musculus: 100 (percent identity) Wilms' tumor - Xenopus tropicalis: 99.5 Note - Monodelphis domestica: 99.5 RPL10 was originally isolated by subtractive - Pan troglodytes: 99.5 hybridization between a tumorigenic cell line (deleted - Xenopus laevis: 99.1 for part of 11p) and a non-tumorigenic cell line (the - Danio rerio: 97.7 tumorigenic cell line carrying an extra t(X;11) - Drosophila melanogaster: 88.9 translocation chromosome). The RPL10 mRNA level - Caenorhabditis elegans: 85.5 was found modulated between the tumorigenic and - Neurospora crassa: 84.7 nontumorigenic cell lines and suspected to be involved - Saccharomyces cerevisiae: 78.3 in the maintenance of the nontumorigenic phenotype. However, later study had shown that the RPL10 gene is Mutations X-linked and therefore not involved in suppression of tumorigenesis in Wilms' tumor. Note Two missense mutations L206M and H213Q at the C- References terminal end of RPL10 were identified in two independent families with autism, a disorder of neural Dowdy SF, Lai KM, Weissman BE, Matsui Y, Hogan BL, Stanbridge EJ. The isolation and characterization of a novel development. cDNA demonstrating an altered mRNA level in nontumorigenic Wilms' microcell hybrid cells. Nucleic Acids Res. 1991 Oct Implicated in 25;19(20):5763-9 van den Ouweland AM, Verdijk M, Mannens MM, van Oost BA. Prostatic adenocarcinoma The QM gene is X-linked and therefore not involved in Note suppression of tumorigenesis in Wilms' tumor. Hum Genet. 1992 Sep-Oct;90(1-2):144-6 RPL10 gene showed up-regulation in androgen- independent C81 passage cells, derived from the Karan D, Kelly DL, Rizzino A, Lin MF, Batra SK. Expression LNCaP cell model that recapitulates prostate cancer profile of differentially-regulated genes during progression of androgen-independent growth in human prostate cancer cells. progression. In a study using immunohistochemical Carcinogenesis. 2002 Jun;23(6):967-75 technique, human prostatic tissues showed expression Altinok G, Powell IJ, Che M, Hormont K, Sarkar FH, Sakr WA, of RPL10 protein in all normal prostate glands adjacent Grignon D, Liao DJ. Reduction of QM protein expression to prostate cancer and in various intraepithelial correlates with tumor grade in prostatic adenocarcinoma. neoplasia (PIN). However, in prostate cancer, the Prostate Cancer Prostatic Dis. 2006;9(1):77-82 staining intensity and stained areas were decreased, Klauck SM, Felder B, Kolb-Kokocinski A, Schuster C, compared to the normal glands and PIN lesions. There Chiocchetti A, Schupp I, Wellenreuther R, Schmötzer G, was an inverse correlation from normal to low-grade Poustka F, Breitenbach-Koller L, Poustka A. Mutations in the tumors and then to high-grade tumors. In high-grade ribosomal protein gene RPL10 suggest a novel modulating

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 426

RPL10 (ribosomal protein L10) Goel M, Tamuli R

disease mechanism for autism. Mol Psychiatry. 2006 C, Leboyer M, Bourgeron T. An investigation of ribosomal Dec;11(12):1073-84 protein L10 gene in autism spectrum disorders. BMC Med Genet. 2009 Jan 23;10:7 Shen XJ, Ali-Fehmi R, Weng CR, Sarkar FH, Grignon D, Liao DJ. Loss of heterozygosity and microsatellite instability at the This article should be referenced as such: Xq28 and the A/G heterozygosity of the QM gene are associated with ovarian cancer. Cancer Biol Ther. 2006 Goel M, Tamuli R. RPL10 (ribosomal protein L10). Atlas Genet May;5(5):523-8 Cytogenet Oncol Haematol. 2011; 15(5):425-427. Gong X, Delorme R, Fauchereau F, Durand CM, Chaste P, Betancur C, Goubran-Botros H, Nygren G, Anckarsäter H, Rastam M, Gillberg IC, Kopp S, Mouren-Simeoni MC, Gillberg

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 427

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

SNAI1 (snail homolog 1 (Drosophila)) Joerg Schwock, William R Geddie University of Toronto, Department of Laboratory Medicine and Pathobiology, Division of Anatomical Pathology, Toronto General Hospital, 200 Elizabeth Street, Room E11-219, M5G 2C4 Toronto, Ontario, Canada (JS, WRG)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SNAI1ID452ch20q13.html DOI: 10.4267/2042/45019 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Transcription A single transcript of 1686 bp size gives rise to a Other names: SLUGH2, SNA, SNAH, dJ710H13.1 protein of 264 aa and approximately 29.1 kDa. HGNC (Hugo): SNAI1 Pseudogene Location: 20q13.13 SNAI1P. DNA/RNA Protein Note Note Human Snail homolog 1 (SNAI1, SLUGH2, SNA, Charge 13.0, isoelectric point 8.7563, molecular weight SNAH, dJ710H13.1), homolog of the Drosophila gene 29082.97 Da (source: Uswest.Ensembl). sna, is localized on 20q13.13 (Paznekas et al., 1999; Twigg and Wilkie, 1999). Both publications describe a Description SNAI1-related pseudogene SNAI1P mapped to The N-terminal portion (aa 1-150) of the Snail protein chromosome 2q33-37. contains a SNAG (SNAI1/GFI) domain (aa 1-9) which Description includes the consensus sequence PRSFLV found in all Snail family members. This motif is highly conserved SNAI1 has 3 exons (1: 143 bp, 2: 528 bp and 3: 1015 among species and also found in several other bp size) separated by intron 1-2 (682 bp) and intron 2-3 transcription factors where it is associated with (3520 bp); spanning an approximately 6 kb region. A repressive functions. A serine-rich domain (SRD: aa CpG island has been described upstream of the coding 90-120) and a nuclear export sequence (NES: aa 139- sequence. 148) are involved in the regulation of Snail protein Two silent single nucleotide polymorphisms, a T/C stability and subcellular localization, respectively. transition at position 783 and a G/A transition at The C-terminal portion (aa 151-264) contains 3 typical position 1035, have been described (Twigg and Wilkie, (154-176, 178-202, 208-230) and one atypical (236- 1999). 259) C2H2-type zinc finger (ZF) domains.

Snail protein structure.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 428 SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

Expression characterized by a down-regulation of epithelial (cell- cell adhesion, apical-basal polarity) and up-regulation Nucleus. of mesenchymal (cell-extracellular matrix interaction, Localisation front-back polarity) features associated with respective The human SNAI1 transcript has been detected in changes in molecular composition of the cell which placenta, adult heart and lung. Lower levels were include (among others) cell adhesion molecules and reported for adult brain, liver and skeletal muscle intermediate filaments. This process, termed epithelial- (Paznekas et al., 1999). Several human fetal tissues mesenchymal transition (EMT) (Hay, 1989; Thiery, were reported to express the SNAI1 transcript, with the 2002; Peinado et al., 2007), has been classified into 3 highest levels detected in kidney (Twigg and Wilkie, types: type 1 in the context of developmental processes, 1999). type 2 in inflammation, tissue repair and fibrosis, and Reliable high protein expression is present in the type 3 in tumor invasion and metastasis (Kalluri and extravillous trophoblast of the human placenta which Weinberg, 2009). can be utilized as positive control (Rosivatz et al., To exert its function as repressor, Snail nuclear import 2006). is mediated by importins which recognize a nuclear localization signal that consists of basic residues situated in the zinc finger region (Mingot et al., 2009). Inside the nucleus Snail is required to form ternary complexes with co-repressors via the Snag domain. Different ternary complexes have been described which consist of Ajuba as mediator for the interaction with PRC2 (Herranz et al., 2008), 14-3-3 and PRMT5 (Hou et al., 2010), Sin3A for the interaction with HDAC1/HDAC2 (Peinado et al., 2004), and LSD1 for the interaction with CoREST (Lin et al., 2010). Binding to DNA occurs via E-box elements (5'-CACCTG-3') found in the promoter region of different genes including the E-cadherin gene CDH1 (Batlle et al., 2000; Cano et al., 2000). The regulation of Snail activity mainly involves the central part of the protein which contains most sites for Snail protein expression in 1st trimester human placenta. post-translational modification: serine phosphorylation Method and Antibody: Schwock et al., 2010. sites (Ser92, 96, 100, 104, 107) in the SRD as well as Function two lysine oxidation sites (Lys98 and 137), and the NES for Crm1-dependent nuclear export. Two Snail protein (SNAI1) is part of a superfamily of additional serine phosphorylation sites are found N- transcription factors composed of the SNAI and the terminal at Ser11 and C-terminal at Ser246. SCRATCH family (Nieto, 2002; Barrallo-Gimeno and Phosphorylation of Ser 96, 100, 104 and 107 by Nieto, 2009). The SNAI family contains two more GSK3beta is associated with nuclear export, members: Slug (SNAI2 (Cohen et al., 1998)) and Smuc ubiquitination by beta-TrCP1 or FBXL14 and (SNAI3 (Katoh, 2003)) on chromosomes 8 and 16, proteasomal degradation (Dominguez et al., 2003; respectively. Zhou et al., 2004; Vinas-Castells et al., 2010). Snail is The Snail gene in Drosophila (sna), first identified positively regulated by phosphorylation of Ser11, 92 during analysis of dorso-ventral patterning, is a zinc and 246 and its protein stability is increased by finger gene with repressor function required for interaction with SCP, PKA, CK2, PAK1 and LOXL2 mesoderm formation (Boulay et al., 1987; Leptin, (Peinado et al., 2005; Yang et al., 2005; Wu et al., 1991). Isolation of other Snail homologues in different 2009; MacPherson et al., 2010). species including the human indicated a high degree of Limited information is available on the factors directly conservation in coding sequence and predicted protein controlling the SNAI1 promoter. Snail up-regulation in pointing towards a conserved role in early cells has been reported as a result of diverse stimuli morphogenesis (Paznekas et al., 1999). including cytokines (Interleukin-6), growth factors Snail protein functions as E-cadherin repressor and is (TGFbeta, FGF, PDGF, EGF) and activation of their essential during early developmental stages (Cano et corresponding receptor tyrosine kinases as well as al., 2000; LaBonne and Bronner-Fraser, 2000). Snail is activation of developmental signaling pathways such as re-expressed during adult life in tissue repair as well as Wnt and Hedgehog. Notably, TGFbeta has been in neoplasia, and, in the latter, thought to contribute to described as important EMT trigger leading to HMGA2 the acquisition of a metastatic potential in tumor cells and Smad binding at the SNAI1 promoter (Thuault et (Batlle et al., 2000). The process by which Snail al., 2008). Another example is Snail expression confers increased motility in individual cells is stimulated by HGF via the MAPK pathway and Egr-1

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 429 SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

which also includes a negative feedback mechanism adjacent to tumor (Pena et al., 2009). Early studies due to Snail binding at the Egr-1 promoter (Grotegut et based on detection of the SNAI1 transcript found al., 2006). Also, a conserved 3' enhancer element has associations with lymph node metastasis (Cheng et al., been described which interacts with the SNAI1 2001; Blanco et al., 2002) and malignant effusion promoter (Palmer et al., 2007). Furthermore, Snail has (Elloul et al., 2005) in breast cancer. A mouse model been found to control its own expression by binding to reported by Moody et al. (2005) implicated Snail an E-box in its own promoter (Peiro et al., 2006). A expression with mammary cancer recurrence. Other more detailed overview over the complex signaling studies described associations between elevated SNAI1 pathways regulating Snail expression is given in transcript levels and hypoxia in ovarian cancer (Imai et several recent review publications (Peinado et al., al., 2003), downregulation of Vitamin D Receptor in 2007; De Herreros et al., 2010). colon cancer (Palmer et al., 2004; Pena et al., 2005), Consequences of Snail up-regulation not only include invasion and distant metastasis in oesophageal the repression of E-cadherin transcription, but the squamous cell carcinoma (Takeno et al., 2004), negative as well as positive control of a series of genes invasiveness (Sugimachi et al., 2003) and poor involved in a range of biological functions such as cell- prognosis (Miyoshi et al., 2005) in hepatocellular cell adhesion, cell-extracellular matrix interaction, cell carcinoma, and spindle cell phenotype in synovial polarity, cytoskeleton, cell cycle, survival, and sarcoma (Saito et al., 2004). However, it has been angiogenesis leading to a phenotypic shift towards pointed out that transcript levels may not correlate well more mesenchymal cellular characteristics (De Craene with Snail protein which is tightly regulated and subject et al., 2005; Higashikawa et al., 2008). In the context of to a short half-life previously reported as approximately tumor-associated EMT these mesenchymal-like 25 minutes (Zhou et al., 2004). Also, transcript levels characteristics have been correlated with a greater may be confounded by Snail expression in the stromal resistance to different therapeutic modalities (Kajita et tumor component if no micro-dissection is performed al., 2004; Kurrey et al., 2009), escape from attack by (Peinado et al., 2007). Immunohistochemical detection the immune system (Kudo-Saito et al., 2009), and of Snail has been documented for a range of cancers adoption of a cancer stem cell phenotype (Mani et al., including the upper gastrointestinal tract (Rosivatz et 2008; Morel et al., 2008). al., 2006; Natsugoe et al., 2007; Usami et al., 2008; The importance and the exact biological implications of Kim et al., 2009), head and neck (Yang et al., 2007; Snail expression in human tumors remain a focus of Peinado et al., 2008; Yang et al., 2008; Zidar et al., current research (Schwock et al., 2010). Some of the 2008; Schwock et al., 2010), colorectum (Roy et al., challenges in this area may be due to the transient and 2005; Franci et al., 2009), neuroendocrine tumors of dynamic nature of tumor-associated EMT. Also, it has the ileum (Fendrich et al., 2007), uterine cervix (Franci been proposed that Snail is required as initial trigger in et al., 2006), endometrium (Blechschmidt et al., 2007), EMT, whereas maintenance of the phenotype is taken ovary (Blechschmidt et al., 2008; Jin et al., 2009; over by other factors potentially leaving behind a Tuhkanen et al., 2009), prostate (Heeboll et al., 2009), mesenchymal cell devoid of Snail expression (Peinado breast (Zhou et al., 2004), bladder (Bruyere et al., et al., 2007). Another intriguing recent observation is 2009), adrenal gland (Waldmann et al., 2008), thyroid the presence of Snail in tumor-associated stroma and its gland (Hardy et al., 2007), parathyroid gland (Fendrich impact on tumor prognosis (Franci et al., 2009). et al., 2009) as well as pheochromocytoma (Waldmann Homology et al., 2009) and sarcomas (Franci et al., 2006). Differences in immunoreactivity seem to depend on the Human Snail protein is 97.3, 87.2, 87.5, 57.3 and 58.4 individual tumor entity examined as well as technical identical to SNAI1 in chimpanzee (Pan troglodytes), issues (Schwock et al., 2010). SNAI1 in dog (Canis lupus familiaris), Snai1 in mouse (Mus musculus), snai1a and snai1b in zebrafish (Danio Neoplasms of the gastro-intestinal tract rerio), respectively. Note Rosivatz et al. (2006) examined Snail expression in Implicated in adenocarcinomas of the upper gastrointestinal tract. 7.9% (27/340) of their cases were reported with Various cancers positive staining for Snail. There was no correlation Note between Snail and E-cadherin expression or Snail and Involvement of Snail as a major factor in clinicopathological parameters. Natsugoe et al. (2007) craniosynostosis was excluded (Paznekas et al., 1999; examined 194 cases with oesophageal squamous cell Twigg and Wilkie, 1999). Expression of SNAI1 at the carcinoma. 61.7% (84/194) were reported with positive transcript level has been detected in benign conditions staining. Snail expression was associated with deep such as tissue fibrosis (Sato et al., 2003; Yanez-Mo et invasion, increased lymph node metastasis, and al., 2003; Jayachandran et al., 2009), a range of advanced stage. No correlation was found between malignant neoplasms (Cheng et al., 2001; Rosivatz et Snail and E-cadherin expression. Usami et al. (2008) al., 2002; Takeno et al., 2004), and in normal tissue reported a cohort of 72 cases of oesophageal squamous

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 430

SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

cell carcinoma for which 38% (27/72) were considered tumor cells in excess of 10% was rare, but associated positive. Elevated Snail expression was found at the with poor outcome by univariate analysis. invasion front, and was associated with lymphatic and Neoplasms of the genitourinary tract venous vessel invasion, lymph node metastasis and tumor stage. Furthermore, a recent study by Kim et al. Note (2009) reported Snail positivity in 42.9% (245/571) of 87 primary endometrioid-type adenocarcinomas of the gastric carcinomas where it was associated with endometrium and 26 unrelated metastases were invasion and lymph node metastasis. Snail staining was examined in a study by Blechschmidt et al. (2007). an independent indicator of prognosis by multivariate Among the primary tumors and the metastases a analysis in this study. Fendrich et al., 2007 examined proportion of 28.7% and 53.8% were reported with Snail expression in neuroendocrine tumors of the positive Snail staining, respectively. Snail ileum. 59% (22/37) of the primary tumors and 6 of 7 immunoreactivity in metastases was found to correlate liver metastases were reported with immunoreactivity with higher grade and reduced E-cadherin expression. for Snail. 53% (16/30) of the neuroendocrine tumors A subsequent study by Blechschmidt et al. (2008) on displayed positivity for Snail as well as Sonic 48 primary ovarian neoplasms and 50 metastases found Hedgehog. Roy et al. (2005) found a proportion of 78% Snail immunoreactivity in 37.5% and 52%, (46/59) cases with positive staining in their study on respectively. A borderline significant difference in colorectal cancers as well as a trend towards increased overall survival with Snail expression in metastases presence of Snail in tumors with distant metastasis. A was noted. There was no correlation between Snail and more recent study on colorectal cancer by Franci et al. E-cadherin expression in this study. A similar study by (2009) reported a similarly high proportion of 79% Jin et al. (2009) examined 41 serous adenocarcinomas (128/162) of cases with Snail immunoreactivity. of the ovary with 14 matched metastases, 12 serous Interestingly, in this study a correlation between borderline tumors, 5 cystadenomas and 4 normal stromal Snail expression and decreased survival was ovarian controls. There was a range of Snail found. immunoreactivity with increased nuclear positivity noted in the carcinoma group. Tuhkanen et al. (2009) Neoplasms of the head and neck compared 74 ovarian carcinomas with 24 borderline Note tumors, 21 benign ovarian neoplasms and 14 normal Yang et al. (2007) reported a proportion of 37.4% controls. Increased nuclear staining was noted with (n=147) of primary head and neck squamous cell increasing malignancy both in the epithelial as well as carcinomas with positive immunoreactivity for Snail. the stromal compartment. 23% (17/74) of the ovarian Snail expression was associated with lymph node carcinomas were reported to show focal Snail metastasis, and co-expression with Nijmegen breakage positivity. No association with clinicopathological syndrome 1 (NBS1) indicated short metastasis-free factors was seen in this study. Heeboll et al., 2009 period and overall survival. Another study by Yang et examined Snail in 327 prostate cancer specimens, 15 al. (2008) reported a positive correlation between Snail specimens with high-grade prostatic intraepithelial and reduced metastasis-free and overall survival. neoplasia (PIN), 30 specimens from patients with Peinado et al. (2008) examined a large cohort of benign prostatic hyperplasia and 30 benign prostate laryngeal squamous cell carcinomas for which 16% tissue controls. Approximately 50% of the prostate (40/251) were reported Snail positive including 3% cancers were found to have high Snail (8/251) high-positive. They found a correlation immunoreactivity compared to only 7% of the high- between Snail and LOXL2 expression, but no grade PIN specimens. Snail expression in this study association between Snail and disease-free or overall was associated with Gleason score, but not with survival. Zidar et al. (2008) reported their findings on progression or prognosis. Bruyere et al., 2009 studied two cohorts of head and neck squamous cell Snail expression in transitional cell carcinoma of the carcinomas specifically distinguishing between spindle bladder using a microarray of 87 cases. Strong Snail cell carcinomas and those of moderately differentiated positivity was found in 43.7% and weak positivity in phenotype. 19/30 of the spindle cell, but only 4/30 the remainder of the cases. Snail immunoreactivity in cases in the moderately differentiated group were found this study was prognostic for tumor recurrence by uni- to display positive immunoreactivity. There was no and multivariate analysis. correlation between Snail and E-cadherin expression. Breast cancer Schwock et al. (2010) examined a cohort of 46 cases of oral squamous cell carcinoma including corresponding Note metastases. Nuclear Snail-positivity equal or in excess Zhou et al. (2004) reported a study on Snail expression of a 5% threshold was observed in 10 tumors and 5 in breast cancer which found positive staining in 56% metastases which corresponded to 12 cases. Individual (72/129) of their cases; 17 with low and 55 with high Snail-positive tumor cells below this threshold, Snail immunoreactivity. Snail correlated with GSK- however, were present more frequently and found in 3beta inhibition and E-cadherin downregulation, and primary tumors of 30 patients. Snail expression in clinically with metastasis in this study.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 431 SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

Endocrine neoplasms References Note Boulay JL, Dennefeld C, Alberga A. The Drosophila Waldmann et al. (2008) reported their findings with developmental gene snail encodes a protein with nucleic acid Snail expression in adrenocortical carcinomas obtained binding fingers. Nature. 1987 Nov 26-Dec 2;330(6146):395-8 in a study including 26 primary tumors as well as two Hay ED. Theory for epithelial-mesenchymal transformation lymph node and one liver metastases. 65% (17/26) based on the "fixed cortex" cell motility model. Cell Motil primary tumors showed staining for Snail with strong Cytoskeleton. 1989;14(4):455-7 positivity found at the invasion front of 7 tumors and in Leptin M. twist and snail as positive and negative regulators 2 of 3 metastases. Snail positivity was associated with during Drosophila mesoderm development. Genes Dev. 1991 advanced stage, decreased survival and higher risk for Sep;5(9):1568-76 distant metastases. The same group reported a study on Cohen ME, Yin M, Paznekas WA, Schertzer M, Wood S, Jabs Snail in pheochromocytomas including 44 primary EW. Human SLUG gene organization, expression, and tumors, 3 lymph node and 2 peritoneal metastases chromosome map location on 8q. Genomics. 1998 Aug (Waldmann et al., 2009). Snail positivity was reported 1;51(3):468-71 for 28% (13/47) cases, and positive staining was Paznekas WA, Okajima K, Schertzer M, Wood S, Jabs EW. associated with malignant behaviour. Hardy et al. Genomic organization, expression, and chromosome location of the human SNAIL gene (SNAI1) and a related processed (2007) published their results focused on thyroid pseudogene (SNAI1P). Genomics. 1999 Nov 15;62(1):42-9 neoplasms. 18/31 follicular and 28/32 papillary thyroid cancers as well as all of 4 lymph node metastases of Twigg SR, Wilkie AO. Characterisation of the human snail (SNAI1) gene and exclusion as a major disease gene in papillary thyroid cancer were found to stain positive for craniosynostosis. Hum Genet. 1999 Oct;105(4):320-6 Snail whereas normal thyroid tissue was negative. Snail Batlle E, Sancho E, Francí C, Domínguez D, Monfar M, staining was reported to be restricted to the invasive Baulida J, García De Herreros A. The transcription factor snail front and associated with a concomitant reduction in E- is a repressor of E-cadherin gene expression in epithelial cadherin reactivity. The authors of this study also tumour cells. Nat Cell Biol. 2000 Feb;2(2):84-9 included their findings from a Combi-TA mouse model Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, showing development of papillary thyroid carcinomas. del Barrio MG, Portillo F, Nieto MA. The transcription factor Fendrich et al. (2009) recently published results of a snail controls epithelial-mesenchymal transitions by repressing study focused on parathyroid neoplasms including 9 E-cadherin expression. Nat Cell Biol. 2000 Feb;2(2):76-83 cases of parathyroid carcinoma, 25 adenomas and 25 LaBonne C, Bronner-Fraser M. Snail-related transcriptional cases of hyperplasia. Snail staining was positive in all repressors are required in Xenopus for both the induction of cases of hyperplasia and 22/25 adenomas. In the neural crest and its subsequent migration. Dev Biol. 2000 May 1;221(1):195-205 carcinomas a change in staining pattern towards the invasion front was noted. Cheng CW, Wu PE, Yu JC, Huang CS, Yue CT, Wu CW, Shen CY. Mechanisms of inactivation of E-cadherin in breast Mesenchymal neoplasms carcinoma: modification of the two-hit hypothesis of tumor suppressor gene. Oncogene. 2001 Jun 28;20(29):3814-23 Note Franci et al. (2006) studied Snail in a series of different Blanco MJ, Moreno-Bueno G, Sarrio D, Locascio A, Cano A, Palacios J, Nieto MA. Correlation of Snail expression with neoplasm which included sarcomas and infantile histological grade and lymph node status in breast carcinomas. fibromatosis as well as epithelial neoplasms (squamous Oncogene. 2002 May 9;21(20):3241-6 cell carcinoma of the uterine cervix and Nieto MA. The snail superfamily of zinc-finger transcription adenocarcinoma of the colon). High Snail expression factors. Nat Rev Mol Cell Biol. 2002 Mar;3(3):155-66 was present in fibrosarcomas and other sarcomas. Snail Rosivatz E, Becker I, Specht K, Fricke E, Luber B, Busch R, expression in neoplasms of epithelial origin was Höfler H, Becker KF. Differential expression of the epithelial- restricted to the tumor-stroma interface. mesenchymal transition regulators snail, SIP1, and twist in gastric cancer. Am J Pathol. 2002 Nov;161(5):1881-91 To be noted Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002 Jun;2(6):442-54 Note Mouse model. Domínguez D, Montserrat-Sentís B, Virgós-Soler A, Guaita S, Grueso J, Porta M, Puig I, Baulida J, Francí C, García de A CombitTA conditional mouse model of Snail Herreros A. Phosphorylation regulates the subcellular location expression has been described without morphological and activity of the snail transcriptional repressor. Mol Cell Biol. alterations, but associated with the development of both 2003 Jul;23(14):5078-89 epithelial and mesenchymal tumors (leukemias) (Perez- Imai T, Horiuchi A, Wang C, Oka K, Ohira S, Nikaido T, Mancera et al., 2005). Notably, suppression of the Snail Konishi I. Hypoxia attenuates the expression of E-cadherin via transgene did not rescue the malignant phenotype, up-regulation of SNAIL in ovarian carcinoma cells. Am J Pathol. 2003 Oct;163(4):1437-47 indicating that the alterations induced by Snail were irreversible.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 432

SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

Katoh M, Katoh M. Identification and characterization of human Peinado H, Del Carmen Iglesias-de la Cruz M, Olmeda D, SNAIL3 (SNAI3) gene in silico. Int J Mol Med. 2003 Csiszar K, Fong KS, Vega S, Nieto MA, Cano A, Portillo F. A Mar;11(3):383-8 molecular role for lysyl oxidase-like 2 enzyme in snail regulation and tumor progression. EMBO J. 2005 Oct Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A. 5;24(19):3446-58 Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral Peña C, García JM, Silva J, García V, Rodríguez R, Alonso I, ureteral obstruction. J Clin Invest. 2003 Nov;112(10):1486-94 Millán I, Salas C, de Herreros AG, Muñoz A, Bonilla F. E- cadherin and vitamin D receptor regulation by SNAIL and Sugimachi K, Tanaka S, Kameyama T, Taguchi K, Aishima S, ZEB1 in colon cancer: clinicopathological correlations. Hum Shimada M, Sugimachi K, Tsuneyoshi M. Transcriptional Mol Genet. 2005 Nov 15;14(22):3361-70 repressor snail and progression of human hepatocellular carcinoma. Clin Cancer Res. 2003 Jul;9(7):2657-64 Pérez-Mancera PA, Pérez-Caro M, González-Herrero I, Flores T, Orfao A, de Herreros AG, Gutiérrez-Adán A, Pintado B, Yáñez-Mó M, Lara-Pezzi E, Selgas R, Ramírez-Huesca M, Sagrera A, Sánchez-Martín M, Sánchez-García I. Cancer Domínguez-Jiménez C, Jiménez-Heffernan JA, Aguilera A, development induced by graded expression of Snail in mice. Sánchez-Tomero JA, Bajo MA, Alvarez V, Castro MA, del Hum Mol Genet. 2005 Nov 15;14(22):3449-61 Peso G, Cirujeda A, Gamallo C, Sánchez-Madrid F, López- Cabrera M. Peritoneal dialysis and epithelial-to-mesenchymal Roy HK, Smyrk TC, Koetsier J, Victor TA, Wali RK. The transition of mesothelial cells. N Engl J Med. 2003 Jan transcriptional repressor SNAIL is overexpressed in human 30;348(5):403-13 colon cancer. Dig Dis Sci. 2005 Jan;50(1):42-6 Kajita M, McClinic KN, Wade PA. Aberrant expression of the Yang Z, Rayala S, Nguyen D, Vadlamudi RK, Chen S, Kumar transcription factors snail and slug alters the response to R. Pak1 phosphorylation of snail, a master regulator of genotoxic stress. Mol Cell Biol. 2004 Sep;24(17):7559-66 epithelial-to-mesenchyme transition, modulates snail's subcellular localization and functions. Cancer Res. 2005 Apr Pálmer HG, Larriba MJ, García JM, Ordóñez-Morán P, Peña 15;65(8):3179-84 C, Peiró S, Puig I, Rodríguez R, de la Fuente R, Bernad A, Pollán M, Bonilla F, Gamallo C, de Herreros AG, Muñoz A. The Francí C, Takkunen M, Dave N, Alameda F, Gómez S, transcription factor SNAIL represses vitamin D receptor Rodríguez R, Escrivà M, Montserrat-Sentís B, Baró T, Garrido expression and responsiveness in human colon cancer. Nat M, Bonilla F, Virtanen I, García de Herreros A. Expression of Med. 2004 Sep;10(9):917-9 Snail protein in tumor-stroma interface. Oncogene. 2006 Aug 24;25(37):5134-44 Peinado H, Ballestar E, Esteller M, Cano A. Snail mediates E- cadherin repression by the recruitment of the Sin3A/histone Grotegut S, von Schweinitz D, Christofori G, Lehembre F. deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol. 2004 Hepatocyte growth factor induces cell scattering through Jan;24(1):306-19 MAPK/Egr-1-mediated upregulation of Snail. EMBO J. 2006 Aug 9;25(15):3534-45 Saito T, Oda Y, Kawaguchi K, Sugimachi K, Yamamoto H, Tateishi N, Tanaka K, Matsuda S, Iwamoto Y, Ladanyi M, Peiró S, Escrivà M, Puig I, Barberà MJ, Dave N, Herranz N, Tsuneyoshi M. E-cadherin mutation and Snail overexpression Larriba MJ, Takkunen M, Francí C, Muñoz A, Virtanen I, as alternative mechanisms of E-cadherin inactivation in Baulida J, García de Herreros A. Snail1 transcriptional synovial sarcoma. Oncogene. 2004 Nov 11;23(53):8629-38 repressor binds to its own promoter and controls its expression. Nucleic Acids Res. 2006;34(7):2077-84 Takeno S, Noguchi T, Fumoto S, Kimura Y, Shibata T, Kawahara K. E-cadherin expression in patients with Rosivatz E, Becker KF, Kremmer E, Schott C, Blechschmidt K, esophageal squamous cell carcinoma: promoter Höfler H, Sarbia M. Expression and nuclear localization of hypermethylation, Snail overexpression, and clinicopathologic Snail, an E-cadherin repressor, in adenocarcinomas of the implications. Am J Clin Pathol. 2004 Jul;122(1):78-84 upper gastrointestinal tract. Virchows Arch. 2006 Mar;448(3):277-87 Zhou BP, Deng J, Xia W, Xu J, Li YM, Gunduz M, Hung MC. Dual regulation of Snail by GSK-3beta-mediated Blechschmidt K, Kremmer E, Hollweck R, Mylonas I, Höfler H, phosphorylation in control of epithelial-mesenchymal transition. Kremer M, Becker KF. The E-cadherin repressor snail plays a Nat Cell Biol. 2004 Oct;6(10):931-40 role in tumor progression of endometrioid adenocarcinomas. Diagn Mol Pathol. 2007 Dec;16(4):222-8 De Craene B, Gilbert B, Stove C, Bruyneel E, van Roy F, Berx G. The transcription factor snail induces tumor cell invasion Fendrich V, Waldmann J, Esni F, Ramaswamy A, Mullendore through modulation of the epithelial cell differentiation program. M, Buchholz M, Maitra A, Feldmann G. Snail and Sonic Cancer Res. 2005 Jul 15;65(14):6237-44 Hedgehog activation in neuroendocrine tumors of the ileum. Endocr Relat Cancer. 2007 Sep;14(3):865-74 Elloul S, Elstrand MB, Nesland JM, Tropé CG, Kvalheim G, Goldberg I, Reich R, Davidson B. Snail, Slug, and Smad- Hardy RG, Vicente-Dueñas C, González-Herrero I, Anderson interacting protein 1 as novel parameters of disease C, Flores T, Hughes S, Tselepis C, Ross JA, Sánchez-García aggressiveness in metastatic ovarian and breast carcinoma. I. Snail family transcription factors are implicated in thyroid Cancer. 2005 Apr 15;103(8):1631-43 carcinogenesis. Am J Pathol. 2007 Sep;171(3):1037-46 Miyoshi A, Kitajima Y, Kido S, Shimonishi T, Matsuyama S, Natsugoe S, Uchikado Y, Okumura H, Matsumoto M, Kitahara K, Miyazaki K. Snail accelerates cancer invasion by Setoyama T, Tamotsu K, Kita Y, Sakamoto A, Owaki T, upregulating MMP expression and is associated with poor Ishigami S, Aikou T. Snail plays a key role in E-cadherin- prognosis of hepatocellular carcinoma. Br J Cancer. 2005 Jan preserved esophageal squamous cell carcinoma. Oncol Rep. 31;92(2):252-8 2007 Mar;17(3):517-23 Moody SE, Perez D, Pan TC, Sarkisian CJ, Portocarrero CP, Palmer MB, Majumder P, Green MR, Wade PA, Boss JM. A 3' Sterner CJ, Notorfrancesco KL, Cardiff RD, Chodosh LA. The enhancer controls snail expression in melanoma cells. Cancer transcriptional repressor Snail promotes mammary tumor Res. 2007 Jul 1;67(13):6113-20 recurrence. Cancer Cell. 2005 Sep;8(3):197-209

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 433 SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in benign and malignant parathyroid neoplasia. Eur J Endocrinol. tumour progression: an alliance against the epithelial 2009 Apr;160(4):695-703 phenotype? Nat Rev Cancer. 2007 Jun;7(6):415-28 Francí C, Gallén M, Alameda F, Baró T, Iglesias M, Virtanen I, Yang MH, Chang SY, Chiou SH, Liu CJ, Chi CW, Chen PM, García de Herreros A. Snail1 protein in the stroma as a new Teng SC, Wu KJ. Overexpression of NBS1 induces epithelial- putative prognosis marker for colon tumours. PLoS One. mesenchymal transition and co-expression of NBS1 and Snail 2009;4(5):e5595 predicts metastasis of head and neck cancer. Oncogene. 2007 Mar 1;26(10):1459-67 Heebøll S, Borre M, Ottosen PD, Dyrskjøt L, Orntoft TF, Tørring N. Snail1 is over-expressed in prostate cancer. APMIS. Blechschmidt K, Sassen S, Schmalfeldt B, Schuster T, Höfler 2009 Mar;117(3):196-204 H, Becker KF. The E-cadherin repressor Snail is associated with lower overall survival of ovarian cancer patients. Br J Jayachandran A, Königshoff M, Yu H, Rupniewska E, Hecker Cancer. 2008 Jan 29;98(2):489-95 M, Klepetko W, Seeger W, Eickelberg O. SNAI transcription factors mediate epithelial-mesenchymal transition in lung Herranz N, Pasini D, Díaz VM, Francí C, Gutierrez A, Dave N, fibrosis. Thorax. 2009 Dec;64(12):1053-61 Escrivà M, Hernandez-Muñoz I, Di Croce L, Helin K, García de Herreros A, Peiró S. Polycomb complex 2 is required for E- Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal cadherin repression by the Snail1 transcription factor. Mol Cell transition. J Clin Invest. 2009 Jun;119(6):1420-8 Biol. 2008 Aug;28(15):4772-81 Kim MA, Lee HS, Lee HE, Kim JH, Yang HK, Kim WH. Higashikawa K, Yoneda S, Taki M, Shigeishi H, Ono S, Prognostic importance of epithelial-mesenchymal transition- Tobiume K, Kamata N. Gene expression profiling to identify related protein expression in gastric carcinoma. genes associated with high-invasiveness in human squamous Histopathology. 2009 Mar;54(4):442-51 cell carcinoma with epithelial-to-mesenchymal transition. Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. Cancer Cancer Lett. 2008 Jun 18;264(2):256-64 metastasis is accelerated through immunosuppression during Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Snail-induced EMT of cancer cells. Cancer Cell. 2009 Mar Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, 3;15(3):195-206 Polyak K, Brisken C, Yang J, Weinberg RA. The epithelial- Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar mesenchymal transition generates cells with properties of stem PD, Doiphode RY, Bapat SA. Snail and slug mediate cells. Cell. 2008 May 16;133(4):704-15 radioresistance and chemoresistance by antagonizing p53- Morel AP, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux mediated apoptosis and acquiring a stem-like phenotype in A. Generation of breast cancer stem cells through epithelial- ovarian cancer cells. Stem Cells. 2009 Sep;27(9):2059-68 mesenchymal transition. PLoS One. 2008 Aug 6;3(8):e2888 Mingot JM, Vega S, Maestro B, Sanz JM, Nieto MA. Peinado H, Moreno-Bueno G, Hardisson D, Pérez-Gómez E, Characterization of Snail nuclear import pathways as Santos V, Mendiola M, de Diego JI, Nistal M, Quintanilla M, representatives of C2H2 zinc finger transcription factors. J Cell Portillo F, Cano A. Lysyl oxidase-like 2 as a new poor Sci. 2009 May 1;122(Pt 9):1452-60 prognosis marker of squamous cell carcinomas. Cancer Res. Peña C, García JM, Larriba MJ, Barderas R, Gómez I, Herrera 2008 Jun 15;68(12):4541-50 M, García V, Silva J, Domínguez G, Rodríguez R, Cuevas J, Thuault S, Tan EJ, Peinado H, Cano A, Heldin CH, Moustakas de Herreros AG, Casal JI, Muñoz A, Bonilla F. SNAI1 A. HMGA2 and Smads co-regulate SNAIL1 expression during expression in colon cancer related with CDH1 and VDR induction of epithelial-to-mesenchymal transition. J Biol Chem. downregulation in normal adjacent tissue. Oncogene. 2009 2008 Nov 28;283(48):33437-46 Dec 10;28(49):4375-85 Usami Y, Satake S, Nakayama F, Matsumoto M, Ohnuma K, Tuhkanen H, Soini Y, Kosma VM, Anttila M, Sironen R, Komori T, Semba S, Ito A, Yokozaki H. Snail-associated Hämäläinen K, Kukkonen L, Virtanen I, Mannermaa A. Nuclear epithelial-mesenchymal transition promotes oesophageal expression of Snail1 in borderline and malignant epithelial squamous cell carcinoma motility and progression. J Pathol. ovarian tumours is associated with tumour progression. BMC 2008 Jul;215(3):330-9 Cancer. 2009 Aug 20;9:289 Waldmann J, Feldmann G, Slater EP, Langer P, Buchholz M, Waldmann J, Slater EP, Langer P, Buchholz M, Ramaswamy Ramaswamy A, Saeger W, Rothmund M, Fendrich V. A, Walz MK, Schmid KW, Feldmann G, Bartsch DK, Fendrich Expression of the zinc-finger transcription factor Snail in V. Expression of the transcription factor snail and its target adrenocortical carcinoma is associated with decreased gene twist are associated with malignancy in survival. Br J Cancer. 2008 Dec 2;99(11):1900-7 pheochromocytomas. Ann Surg Oncol. 2009 Jul;16(7):1997- 2005 Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, Teng SC, Wu KJ. Direct regulation of TWIST by HIF-1alpha Wu Y, Evers BM, Zhou BP. Small C-terminal domain promotes metastasis. Nat Cell Biol. 2008 Mar;10(3):295-305 phosphatase enhances snail activity through dephosphorylation. J Biol Chem. 2009 Jan 2;284(1):640-8 Zidar N, Gale N, Kojc N, Volavsek M, Cardesa A, Alos L, Höfler H, Blechschmidt K, Becker KF. Cadherin-catenin Bruyere F, Namdarian B, Corcoran NM, Pedersen J, Ockrim J, complex and transcription factor Snail-1 in spindle cell Voelzke BB, Mete U, Costello AJ, Hovens CM. Snail carcinoma of the head and neck. Virchows Arch. 2008 expression is an independent predictor of tumor recurrence in Sep;453(3):267-74 superficial bladder cancers. Urol Oncol. 2010 Nov- Dec;28(6):591-6 Barrallo-Gimeno A, Nieto MA. Evolutionary history of the Snail/Scratch superfamily. Trends Genet. 2009 Jun;25(6):248- de Herreros AG, Peiró S, Nassour M, Savagner P. Snail family 52 regulation and epithelial mesenchymal transitions in breast cancer progression. J Mammary Gland Biol Neoplasia. 2010 Fendrich V, Waldmann J, Feldmann G, Schlosser K, König A, Jun;15(2):135-47 Ramaswamy A, Bartsch DK, Karakas E. Unique expression pattern of the EMT markers Snail, Twist and E-cadherin in

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 434

SNAI1 (snail homolog 1 (Drosophila)) Schwock J, Geddie WR

Hou Z, Peng H, White DE, Wang P, Lieberman PM, cAMP-activated kinase protein kinase A. Mol Biol Cell. 2010 Halazonetis T, Rauscher FJ 3rd. 14-3-3 binding sites in the Jan 15;21(2):244-53 snail protein are essential for snail-mediated transcriptional repression and epithelial-mesenchymal differentiation. Cancer Schwock J, Bradley G, Ho JC, Perez-Ordonez B, Hedley DW, Res. 2010 Jun 1;70(11):4385-93 Irish JC, Geddie WR. SNAI1 expression and the mesenchymal phenotype: an immunohistochemical study performed on 46 Jin H, Yu Y, Zhang T, Zhou X, Zhou J, Jia L, Wu Y, Zhou BP, cases of oral squamous cell carcinoma. BMC Clin Pathol. 2010 Feng Y. Snail is critical for tumor growth and metastasis of Feb 5;10:1 ovarian carcinoma. Int J Cancer. 2010 May 1;126(9):2102-11 Viñas-Castells R, Beltran M, Valls G, Gómez I, García JM, Lin Y, Wu Y, Li J, Dong C, Ye X, Chi YI, Evers BM, Zhou BP. Montserrat-Sentís B, Baulida J, Bonilla F, de Herreros AG, The SNAG domain of Snail1 functions as a molecular hook for Díaz VM. The hypoxia-controlled FBXL14 ubiquitin ligase recruiting lysine-specific demethylase 1. EMBO J. 2010 Jun targets SNAIL1 for proteasome degradation. J Biol Chem. 2;29(11):1803-16 2010 Feb 5;285(6):3794-805

MacPherson MR, Molina P, Souchelnytskyi S, Wernstedt C, This article should be referenced as such: Martin-Pérez J, Portillo F, Cano A. Phosphorylation of serine 11 and serine 92 as new positive regulators of human Snail1 Schwock J, Geddie WR. SNAI1 (snail homolog 1 (Drosophila)). function: potential involvement of casein kinase-2 and the Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):428-435.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 435 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

VAV3 (vav 3 guanine nucleotide exchange factor) Leah Lyons, Kerry L Burnstein Nova Southeastern University, College of Medical Sciences, Department of Physiology, Florida, USA (LL), University of Miami, Miller School of Medicine, Department of Molecular and Cellular Pharmacology, Miami, Florida, USA (KLB)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/VAV3ID42782ch1p13.html DOI: 10.4267/2042/45020 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

be derived from alternate promoter usage (Vav3.1). Identity The alpha isoform is the canonical sequence and is Other names: FLJ40431 derived from the full 27 exons. Isoform beta differs in HGNC (Hugo): VAV3 the N terminus from the alpha isoform as follows: The residues 1-107 in the alpha isoform, Location: 1p13.3 MEPWKQCAQW...DLFDVRDFGK, are replaced by Local order: VAV3 maps to the minus strand of MQLPDCPCRAHLP in the beta isoform. The beta chromosome 1. isoform is produced from a unique exon 1 spliced to exons 4-27 (Maier et al., 2005). Additionally, a DNA/RNA transcript variant encoding only the C terminal SH3 SH2 SH3 domains has been identified and is known as Description Vav3.1. This variant is derived from a unique exon 18 The VAV3 gene is comprised of 27 exons spanning and exons 19-27 (Maier et al., 2005) and is thought to 393.7 kb on chromosome 1p13.3. It is located on the be produced either by alternative splicing or through reverse strand 108113782 bp from pter -108507545 alternate promoter usage. The Vav3 mRNA consists of basepairs from pter. a 54 5 prime UTR and a 2171 basepair 3 Transcription prime UTR (Trenkle et al., 2000). The promoter region of Vav3 contains predicted binding sites for the There are two known isoforms produced by alternative following transcription factors: STAT3, c-MYB, splicing and a third transcript thought to

Figure 1. Upper figure shows gene organization for the alpha (canonical) isoform (ID NM 006113.4) and isoform 2 (ID NM 001079874.1) which corresponds to the 287 amino acid Vav3.1 transcript variant (described below). Lower panel illustrates neighboring genes. Figures adapted from NCBI Gene database.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 436 VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

LMO2, GATA-1, GCNF-2, E47, GCNF-1, PAX-5, acidic domain, a DBL homology domain which confers POU2F1, and FOXO1A (information obtained from GEF function. The DBL homology domain is data deposited in Genecard database through use of comprised of residues 192-371, followed by a SABiosciences' text mining application and the UCSC pleckstrin homology domain, spanning residues 400- genome browser). It is worth mentioning that the gene 502 a cysteine rich domain (also termed a zinc finger locus is complex and could potentially produce up to domain) comprising residues 513-562 and two SH3 13 different isoforms resulting from alternative splicing domains flanking a single SH2 domain. The SH3-SH2- and alternate promoter usage (Thierry-Mieg and SH3 cassette comprises the C terminal portion of Vav3 Thierry-Mieg, 2006). and extends from the N terminal SH3 domain (residues Vav3 can be modified posttranslationally by 592-660), to the C terminal SH3 domain (residues 788- phosphorylation. Phosphorylation site prediction 847) and includes the intervening SH2 domain identifies phosphorylation sites at T131, S134, Y141, (residues 672-766) (Trenkle et al., 2000). Residing Y173, S511, T606 and Y797. Sites residing in the N within the N terminal SH3 domain is a proline rich terminal region have been shown to regulate activation region which may be involved in facilitating of Vav3 GEF function (Movilla and Bustelo, 1999). In intramolecular interactions between the C terminal the unphosphorylated state, the GEF domain is regions (our unpublished observations). prevented from physical association with Rho proteins by the Vav3 N terminal domains. These domains (calponin homology and acidic domains) form an autoinhibitory loop via intramolecular interactions. Vav3 is recruited via its SH2 domain to phosphotyrosine residues on interacting proteins, including activated growth factor receptors. Once bound to active growth factor receptors, or other molecules containing intrinsic tyrosine kinase activity, Vav3 becomes tyrosine phosphorylated (Movilla and Bustelo, 1999; Bustelo, 2002; Zugaza et al., 2002). Tyrosine phosphorylation of Vav3 results in a conformational change that relieves the autoinhibition, thus activating the GEF function by allowing access of Figure 3. Schematic showing inactive (top panel) and active Rho proteins to the GEF domain (Movilla and Bustelo, (bottom panel) conformations of Vav3. Movement of the N terminal regions to allow RhoGTPase access to the DH domain 1999; Yu et al., 2010). Tyrosine 173 in particular is a is regulated by phosphorylation events. critical residue in this process (Llorca et al., 2005; Yu et al., 2010). Consistent with an autoinhibitory role of Expression the N terminal regions, removal of both the calponin Vav3 is broadly expressed but with highest levels in homology and the acidic domains results in constitutive cells of hematopoietic lineages (Trenkle et al., 2000). activation of Vav3 GEF function (Movilla and Bustelo, Localisation 1999; Zeng et al., 2000; Zugaza et al., 2002). Vav3 is located predominantly in the cytoplasm, and is Protein often recruited to the membrane upon activation of the various cell surface receptors that are coupled to Vav3 phosphorylation (Zeng et al., 2000). Function

Figure 2. Functional domains of Vav3 proteins and their relative Vav3 functions as a guanine nucleotide exchange factor positions. Abbreviations are as follows: CH: calponin homology, mediating activation of Rho GTPases by stabilization AD: acidic domain, DH: DBL homology, PH: pleckstrin of the nucleotide free state of Rho proteins. homology, CRD: cysteine rich domain, SH3: Src homology 3 Specifically, Vav3 has been shown to act as a GEF for and SH2: Src homology 2. RhoA, RhoG and RAC1 (Movilla and Bustelo, 1999; Description Zugaza et al., 2002). Vav3 couples the activation of The VAV3 gene encodes a 847 amino acid mature growth factor type receptors such as IGFR, EGFR, protein. The mature protein has a molecular mass of PDGFR, insulin receptor and ROS receptor (Zeng et approximately 98 kDa and functions as a guanine al., 2000) to downstream signaling molecules including nucleotide exchange factor (GEF) for members of the but not limited to Jun kinase, NFKappa B, MAPK and Rho family of small GTPases (Movilla and Stat pathways (Moores et al., 2000; Sachdev et al., Bustelo,1999; Trenkle et al., 2000). Vav3 is structurally 2002). More recently, Vav3 activation by Eph complex consisting of multiple functional domains. Receptors has been demonstrated (Fang et al., 2008) These domains consist sequentially of a single calponin and a large number of studies have shown the homology domain encompassing residues 1-119, an activation of Vav3 upon integrin signaling (Gakidis et

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 437 VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

al., 2004; Faccio et al., 2005; Pearce et al., 2007; presence of tumor cells. Additionally Vav2 and Vav3 Sindrilaru et al., 2009). were found to be necessary and sufficient for Eph A Vav3 is implicated in B cell induced antigen receptor-mediated angiogenesis both in vitro and in presentation to T cells (Malhotra et al., 2009) and vivo (Hunter et al., 2006). mediates both B and T cell signaling events and Homology alteration of macrophage morphology (Sindrilaru et al., 2009). Additionally, protein interactions with the C Vav3 is conserved among vertebrates including dog, terminal SH3 SH2, SH3 cassette have revealed roles in cow, mouse, rat, chicken and zebrafish, and has been scaffolding through adaptor like actions (Bustelo, 2001; shown to be present and conserved in Drosophila Yabana and Shibuya, 2002). melanogaster (Movilla and Bustelo, 1999; Couceiro et Additional functions of Vav3 in distinct tissues are al., 2005). Vav3 displays over 50% amino acid identity listed below. with other members of the Vav family of GEFS, Vav1 Nervous system: NGF-induced neurite outgrowth in and Vav2 which have a similar arrangement of PC12 cells requires Vav3-mediated activation of Rac. functional domains and regulation (Trenkle et al., This process involves P13K activation which occurs 2000). upstream of Vav3 (Aoki et al., 2005). Vav3 is also important for neuronal migration during development Mutations (Khodosevich et al., 2009). Additionally, Vav3 Note knockout mice show defects in Purkinje cell dendrite None described. SNP analysis has revealed several branching, granule cell migration and survival. genetic polymorphisms, the implications of which Functionally the animals show deficiencies in motor remain unclear. The single nucleotide polymorphisms coordination and gaiting consistent with a role for Vav3 resulting in differing amino acid sequence are as in neuronal guidance, cerebellar development and follows: residue 139, D to N, residue 298, T to S, function (Quevedo et al., 2010). residue 616 P to S, and residue Q to H. There are Skeletal system: Studies in osteoclasts support a role multiple SNPS residing in both the 3'UTR and 5'UTR for Vav3 in mediating proper bone deposition. regions. The implications of these are not known. Specifically, Vav3 deficient osteoclasts exhibit abnormalities in actin cytoskeletal rearrangements, cell spreading, and resorptive activities. Consistent with the Implicated in actions of Vav3 on integrin signaling, the osteoclast Prostate cancer defects were found to be due to impaired integrin engagement. Further, Vav3 deficient mice have Note increased bone density and are refractory to PTH- Vav3 mRNA and protein are up-regulated during mediated bone resorption (Faccio et al., 2005). progression of human prostate cancer cells to androgen Cardiovascular system: An important role for Vav3 in independence in cell culture and in vivo experimental maintaining proper cardiovascular homeostasis was studies (Lyons and Burnstein, 2006; Lyons et al., suggested by experiments performed in Vav3 null 2008). Further, the importance of this upregulation to mice. These mice exhibited many symptoms of the disease process has been elucidated by more recent cardiovascular dysfunction including tachycardia, studies showing that Vav3 mRNA is up-regulated in hypertension and cardiovascular remodeling. prostate cancer tumor specimens obtained from men Consistent with these symptoms, the mice also undergoing androgen deprivation therapy compared to exhibited a high degree of sympathetic tone including levels in primary tumors (Holzbeierlein et al., 2004; elevated circulating levels of catecholamines and renin- Best et al., 2005; data deposited in public databases). angiotensin-aldosterone hyperactivity, resulting in Vav3 protein is overexpressed (relative to benign progressive loss of both cardiovascular and renal tissue) in almost one-third of prostate cancer tumor homeostasis (Sauzeau et al., 2006). specimens (Dong et al., 2006). Vascular smooth muscle: Vav3 is both necessary and Additionally, Vav3 mRNA is up-regulated in androgen sufficient for rat vascular smooth muscle cell independent tumors in the Nkx3.1; Pten mouse model proliferation. These effects occur through a Rac-1 of prostate cancer (Banach-Petrosky et al., 2007; dependent mechanism, involving the effector Pak 1 Ouyang et al., 2008) and targeted expression of a (Toumaniantz et al., 2010). constitutively active form of Vav3 in prostate Platelets: Consistent with a role for Vav3 in mediating epithelium of transgenic mice leads to overactivity of integrin-based responses, Vav3 and Vav1 together are the androgen receptor signaling axis and required for collagen exposure-mediated PLC adenocarcinoma (Liu et al., 2008). Mechanistic studies activation in platelets. This signaling pathway occurs show that Vav3 stimulates ligand independent through the major platelet integrin alphaIIbbetaIII androgen receptor activation by a GEF-dependent (Pearce et al., 2004). mechanism that requires the Rho GTPase, Rac 1 in Angiogenesis: Mice deficient in both Vav3 and Vav2 prostate cancer cells (Lyons et al., 2008). Additionally, show reduced endothelial migration in response to the Vav3 enhances androgen receptor transcriptional

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 438 VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

activity in the presence of low concentrations of Type 1 diabetes mellitus androgen through a GEF independent pathway that Note requires the Vav3 PH domain (Lyons and Burnstein, Alteration in Vav3 expression may be an etiological 2006). factor in the development of beta islet cell destruction Breast cancer characteristic of type 1 diabetes (Fraser et al., 2010). Note Lee et al. reported that 81% of human breast cancer References specimens exhibited higher levels of Vav3 compared to Shen L, Qui D, Fang J. [Correlation between hypomethylation benign tissue (Lee et al., 2008). In addition, Vav3 of c-myc and c-N-ras oncogenes and pathological changes in enhances the transcriptional activity of the estrogen human hepatocellular carcinoma]. Zhonghua Zhong Liu Za Zhi. receptor in a GEF dependent manner (Lee et al., 2008). 1997 May;19(3):173-6 Movilla N, Bustelo XR. Biological and regulatory properties of Gastric cancer Vav-3, a new member of the Vav family of oncoproteins. Mol Note Cell Biol. 1999 Nov;19(11):7870-85 Downregulation of RUNX3, a member of the runt Moores SL, Selfors LM, Fredericks J, Breit T, Fujikawa K, Alt domain-containing family of transcription factors that FW, Brugge JS, Swat W. Vav family proteins couple to diverse has tumor suppressive actions, has been implicated in cell surface receptors. Mol Cell Biol. 2000 Sep;20(17):6364-73 promoting human gastric carcinogenesis. Silencing of Trenkle T, McClelland M, Adlkofer K, Welsh J. Major transcript RUNX3 expression via methylation was found in 75% variants of VAV3, a new member of the VAV family of guanine of primary tumors and 100% of gastric metastasis. nucleotide exchange factors. Gene. 2000 Mar 7;245(1):139-49 Stable reexpression of RUNX3 strongly inhibited Zeng L, Sachdev P, Yan L, Chan JL, Trenkle T, McClelland M, peritoneal metastases. Further analysis suggested that Welsh J, Wang LH. Vav3 mediates receptor protein tyrosine kinase signaling, regulates GTPase activity, modulates cell Runx3 expression resulted in the downregulation of a morphology, and induces cell transformation. Mol Cell Biol. number of genes including Vav3 thereby providing a 2000 Dec;20(24):9212-24 potential line between Vav3 expression and gastric Bustelo XR. Vav proteins, adaptors and cell signaling. malignancy (Sakakura et al., 2005). Oncogene. 2001 Oct 1;20(44):6372-81 Hepatocellular carcinoma Sachdev P, Zeng L, Wang LH. Distinct role of Note phosphatidylinositol 3-kinase and Rho family GTPases in Vav3-induced cell transformation, cell motility, and Vav3.1 was down regulated in HepG2 cells in response morphological changes. J Biol Chem. 2002 May to treatment with the hepatocellular carcinoma 17;277(20):17638-48 chemotherapeutic triterpenoid agent astragoloside. Yabana N, Shibuya M. Adaptor protein APS binds the NH2- Downregulation of Vav3.1 was highly correlated with a terminal autoinhibitory domain of guanine nucleotide exchange decrease in malignant transformation, suggesting a role factor Vav3 and augments its activity. Oncogene. 2002 Oct for Vav3.1 in the antitumor actions of astragoloside 31;21(50):7720-9 (Shen et al., 1997). Zugaza JL, López-Lago MA, Caloca MJ, Dosil M, Movilla N, Bustelo XR. Structural determinants for the biological activity of Glioblastoma Vav proteins. J Biol Chem. 2002 Nov 22;277(47):45377-92 Note Gakidis MA, Cullere X, Olson T, Wilsbacher JL, Zhang B, Vav3 is upregulated in glioblastoma as compared to Moores SL, Ley K, Swat W, Mayadas T, Brugge JS. Vav GEFs nonneoplastic or lower grade gliomas. Down regulation are required for beta2 integrin-dependent functions of neutrophils. J Cell Biol. 2004 Jul 19;166(2):273-82 of Vav3 by siRNA reduced glioblastoma invasion and migration. Further upregulation of Vav3 was shown to Holzbeierlein J, Lal P, LaTulippe E, Smith A, Satagopan J, be an indicator of poor patient survival (Salhia et al., Zhang L, Ryan C, Smith S, Scher H, Scardino P, Reuter V, Gerald WL. Gene expression analysis of human prostate 2008). carcinoma during hormonal therapy identifies androgen- responsive genes and mechanisms of therapy resistance. Am Tumor growth and angiogenesis J Pathol. 2004 Jan;164(1):217-27 Note Pearce AC, Senis YA, Billadeau DD, Turner M, Watson SP, A role for Vav3 in promoting tumor growth and Vigorito E. Vav1 and vav3 have critical but redundant roles in angiogensis has been revealed through studies using mediating platelet activation by collagen. J Biol Chem. 2004 mice deficient in both Vav2 and Vav3 (Brantley- Dec 24;279(52):53955-62 Sieders et al., 2009). Vav2, Vav3 knockout mice Aoki K, Nakamura T, Fujikawa K, Matsuda M. Local transplanted with B16 melanoma or Lewis lung phosphatidylinositol 3,4,5-trisphosphate accumulation recruits carcinoma cells showed decreases in tumor growth, Vav2 and Vav3 to activate Rac1/Cdc42 and initiate neurite outgrowth in nerve growth factor-stimulated PC12 cells. Mol tumor survival and neovascularization of tumors as Biol Cell. 2005 May;16(5):2207-17 compared to wild type control mice. The reduction in Best CJ, Gillespie JW, Yi Y, Chandramouli GV, Perlmutter MA, vascularization and tumor growth was found to be Gathright Y, Erickson HS, Georgevich L, Tangrea MA, Duray secondary to a reduction in endothelial cell migration PH, González S, Velasco A, Linehan WM, Matusik RJ, Price (Brantley-Sieders et al., 2009). DK, Figg WD, Emmert-Buck MR, Chuaqui RF. Molecular

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 439 VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

alterations in primary prostate cancer after androgen ablation involved in human breast cancer. BMC Cancer. 2008 Jun therapy. Clin Cancer Res. 2005 Oct 1;11(19 Pt 1):6823-34 2;8:158 Couceiro JR, Martín-Bermudo MD, Bustelo XR. Phylogenetic Liu Y, Mo JQ, Hu Q, Boivin G, Levin L, Lu S, Yang D, Dong Z, conservation of the regulatory and functional properties of the Lu S. Targeted overexpression of vav3 oncogene in prostatic Vav oncoprotein family. Exp Cell Res. 2005 Aug epithelium induces nonbacterial prostatitis and prostate cancer. 15;308(2):364-80 Cancer Res. 2008 Aug 1;68(15):6396-406 Faccio R, Teitelbaum SL, Fujikawa K, Chappel J, Zallone A, Lyons LS, Rao S, Balkan W, Faysal J, Maiorino CA, Burnstein Tybulewicz VL, Ross FP, Swat W. Vav3 regulates osteoclast KL. Ligand-independent activation of androgen receptors by function and bone mass. Nat Med. 2005 Mar;11(3):284-90 Rho GTPase signaling in prostate cancer. Mol Endocrinol. 2008 Mar;22(3):597-608 Llorca O, Arias-Palomo E, Zugaza JL, Bustelo XR. Global conformational rearrangements during the activation of the Ouyang X, Jessen WJ, Al-Ahmadie H, Serio AM, Lin Y, Shih GDP/GTP exchange factor Vav3. EMBO J. 2005 Apr WJ, Reuter VE, Scardino PT, Shen MM, Aronow BJ, Vickers 6;24(7):1330-40 AJ, Gerald WL, Abate-Shen C. Activator protein-1 transcription factors are associated with progression and recurrence of Maier LM, Smyth DJ, Vella A, Payne F, Cooper JD, Pask R, prostate cancer. Cancer Res. 2008 Apr 1;68(7):2132-44 Lowe C, Hulme J, Smink LJ, Fraser H, Moule C, Hunter KM, Chamberlain G, Walker N, Nutland S, Undlien DE, Rønningen Salhia B, Tran NL, Chan A, Wolf A, Nakada M, Rutka F, Ennis KS, Guja C, Ionescu-Tîrgoviste C, Savage DA, Strachan DP, M, McDonough WS, Berens ME, Symons M, Rutka JT. The Peterson LB, Todd JA, Wicker LS, Twells RC. Construction guanine nucleotide exchange factors trio, Ect2, and Vav3 and analysis of tag single nucleotide polymorphism maps for mediate the invasive behavior of glioblastoma. Am J Pathol. six human-mouse orthologous candidate genes in type 1 2008 Dec;173(6):1828-38 diabetes. BMC Genet. 2005 Feb 18;6:9 Brantley-Sieders DM, Zhuang G, Vaught D, Freeman T, Sakakura C, Hasegawa K, Miyagawa K, Nakashima S, Hwang Y, Hicks D, Chen J. Host deficiency in Vav2/3 guanine Yoshikawa T, Kin S, Nakase Y, Yazumi S, Yamagishi H, nucleotide exchange factors impairs tumor growth, survival, Okanoue T, Chiba T, Hagiwara A. Possible involvement of and angiogenesis in vivo. Mol Cancer Res. 2009 May;7(5):615- RUNX3 silencing in the peritoneal metastases of gastric 23 cancers. Clin Cancer Res. 2005 Sep 15;11(18):6479-88 Khodosevich K, Seeburg PH, Monyer H. Major signaling Dong Z, Liu Y, Lu S, Wang A, Lee K, Wang LH, Revelo M, Lu pathways in migrating neuroblasts. Front Mol Neurosci. S. Vav3 oncogene is overexpressed and regulates cell growth 2009;2:7 and androgen receptor activity in human prostate cancer. Mol Endocrinol. 2006 Oct;20(10):2315-25 Malhotra S, Kovats S, Zhang W, Coggeshall KM. B cell antigen receptor endocytosis and antigen presentation to T cells Hunter SG, Zhuang G, Brantley-Sieders D, Swat W, Cowan require Vav and dynamin. J Biol Chem. 2009 Sep CW, Chen J. Essential role of Vav family guanine nucleotide 4;284(36):24088-97 exchange factors in EphA receptor-mediated angiogenesis. Mol Cell Biol. 2006 Jul;26(13):4830-42 Sindrilaru A, Peters T, Schymeinsky J, Oreshkova T, Wang H, Gompf A, Mannella F, Wlaschek M, Sunderkötter C, Rudolph Lyons LS, Burnstein KL. Vav3, a Rho GTPase guanine KL, Walzog B, Bustelo XR, Fischer KD, Scharffetter-Kochanek nucleotide exchange factor, increases during progression to K. Wound healing defect of Vav3-/- mice due to impaired androgen independence in prostate cancer cells and {beta}2-integrin-dependent macrophage phagocytosis of potentiates androgen receptor transcriptional activity. Mol apoptotic neutrophils. Blood. 2009 May 21;113(21):5266-76 Endocrinol. 2006 May;20(5):1061-72 Fraser HI, Dendrou CA, Healy B, Rainbow DB, Howlett S, Sauzeau V, Sevilla MA, Rivas-Elena JV, de Alava E, Montero Smink LJ, Gregory S, Steward CA, Todd JA, Peterson LB, MJ, López-Novoa JM, Bustelo XR. Vav3 proto-oncogene Wicker LS. Nonobese diabetic congenic strain analysis of deficiency leads to sympathetic hyperactivity and autoimmune diabetes reveals genetic complexity of the Idd18 cardiovascular dysfunction. Nat Med. 2006 Jul;12(7):841-5 locus and identifies Vav3 as a candidate gene. J Immunol. 2010 May 1;184(9):5075-84 Thierry-Mieg D, Thierry-Mieg J. AceView: a comprehensive cDNA-supported gene and transcripts annotation. Genome Quevedo C, Sauzeau V, Menacho-Márquez M, Castro-Castro Biol. 2006;7 Suppl 1:S12.1-14 A, Bustelo XR. Vav3-deficient mice exhibit a transient delay in cerebellar development. Mol Biol Cell. 2010 Mar;21(6):1125-39 Banach-Petrosky W, Jessen WJ, Ouyang X, Gao H, Rao J, Quinn J, Aronow BJ, Abate-Shen C. Prolonged exposure to Toumaniantz G, Ferland-McCollough D, Cario-Toumaniantz C, reduced levels of androgen accelerates prostate cancer Pacaud P, Loirand G. The Rho protein exchange factor Vav3 progression in Nkx3.1; Pten mutant mice. Cancer Res. 2007 regulates vascular smooth muscle cell proliferation and Oct 1;67(19):9089-96 migration. Cardiovasc Res. 2010 Apr 1;86(1):131-40 Pearce AC, McCarty OJ, Calaminus SD, Vigorito E, Turner M, Yu B, Martins IR, Li P, Amarasinghe GK, Umetani J, Watson SP. Vav family proteins are required for optimal Fernandez-Zapico ME, Billadeau DD, Machius M, Tomchick regulation of PLCgamma2 by integrin alphaIIbbeta3. Biochem DR, Rosen MK. Structural and energetic mechanisms of J. 2007 Feb 1;401(3):753-61 cooperative autoinhibition and activation of Vav1. Cell. 2010 Jan 22;140(2):246-56 Fang WB, Brantley-Sieders DM, Hwang Y, Ham AJ, Chen J. Identification and functional analysis of phosphorylated This article should be referenced as such: tyrosine residues within EphA2 receptor tyrosine kinase. J Biol Chem. 2008 Jun 6;283(23):16017-26 Lyons L, Burnstein KL. VAV3 (vav 3 guanine nucleotide exchange factor). Atlas Genet Cytogenet Oncol Haematol. Lee K, Liu Y, Mo JQ, Zhang J, Dong Z, Lu S. Vav3 oncogene 2011; 15(5):436-440. activates estrogen receptor and its overexpression may be

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 440 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

CAMTA1 (calmodulin binding transcription activator 1) Kai-Oliver Henrich Division of Tumor Genetics B030, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (KOH)

Published in Atlas Database: September 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/CAMTA1ID908ch1p36.html DOI: 10.4267/2042/45021 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Expression Although expression of CAMTA1 is found in various Other names: KIAA0833 organs, highest levels are seen in neuronal tissues HGNC (Hugo): CAMTA1 (Nagase et al., 1998). In the brain, CAMTA1 levels are Location: 1p36.31 highest in the temporal cortex, entorhinal cortex and in the cerebellum (Huentelman et al., 2007). DNA/RNA Localisation Description Nucleus. The CAMTA1 gene (Henrich and Westermann, 2008) Function comprises 23 exons (all coding) spanning 984.38 kb of Largely unknown. CAMTA1 has been shown to act as genomic DNA. a transcription activator in a reporter expression system Transcription (Bouché et al., 2002). Further data on the functional role are scarce. The homolog CAMTA2 is a co- 8442 bp mRNA. Start codon at 208 bp. Stop codon at activator of the transcription factor Nkx2-5. This 5227 bp. function is inhibited by binding of class II histone Protein deacetylases (Song et al., 2006). Homology Description Members of the CAMTA family are conserved in 1673 amino acids; the protein's primary structure various eukaryotes including ciliates, plants, contains a nuclear localization signal, two DNA- nematodes, insects, birds and mammals (Finkler et al., binding domains (CG-1 and TIG), calmodulin binding 2007). In human, two homologous CAMTAs are found, motifs (IQ motifs) and ankyrin domains that may CAMTA1 and CAMTA2. mediate protein-protein interactions.

Genomic organization of CAMTA1. Arrows illustrate genomic distance. Exons are represented by vertical blue lines or bars.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 441 CAMTA1 (calmodulin binding transcription activator 1) Henrich KO

Schematic representation of the CAMTA1 protein. Protein domains are indicated by boxes. NLS, nuclear localization signal.

multicellular organisms. J Biol Chem. 2002 Jun Implicated in 14;277(24):21851-61 Neuroblastoma Barbashina V, Salazar P, Holland EC, Rosenblum MK, Ladanyi M. Allelic losses at 1p36 and 19q13 in gliomas: correlation with Note histologic classification, definition of a 150-kb minimal deleted The 1p36 smallest region of overlapping deletion in region on 1p36, and evaluation of CAMTA1 as a candidate tumor suppressor gene. Clin Cancer Res. 2005 Feb neuroblastomas spans only 261 kb and pinpoints the 1;11(3):1119-28 CAMTA1 gene (Henrich et al., 2006). In the absence of somatic mutations (Henrich et al., 2007), low Henrich KO, Fischer M, Mertens D, Benner A, et al. Reduced expression of CAMTA1 correlates with adverse outcome in CAMTA1 mRNA expression is significantly associated neuroblastoma patients. Clin Cancer Res. 2006 Jan with markers of unfavorable tumor biology and poor 1;12(1):131-8 neuroblastoma outcome. In multivariate survival Kim MY, Yim SH, Kwon MS, Kim TM, Shin SH, Kang HM, Lee analysis, this prognostic value is independent of C, Chung YJ. Recurrent genomic alterations with impact on established risk markers, including 1p deletion survival in colorectal cancer identified by genome-wide array (Henrich et al., 2006). comparative genomic hybridization. Gastroenterology. 2006 Dec;131(6):1913-24 Glioma Song K, Backs J, McAnally J, Qi X, Gerard RD, Richardson JA, Note Hill JA, Bassel-Duby R, Olson EN. The transcriptional CAMTA1 is homozygously deleted in a subset of coactivator CAMTA2 stimulates cardiac growth by opposing class II histone deacetylases. Cell. 2006 May 5;125(3):453-66 gliomas (Ichimura et al., 2008) and is the only gene mapping to the smallest region of overlapping Finkler A, Ashery-Padan R, Fromm H. CAMTAs: calmodulin- binding transcription activators from plants to human. FEBS heterozygous deletion in this entity (Barbashina et al., Lett. 2007 Aug 21;581(21):3893-8 2005). Henrich KO, Claas A, Praml C, Benner A, Mollenhauer J, Colon cancer Poustka A, Schwab M, Westermann F. Allelic variants of CAMTA1 and FLJ10737 within a commonly deleted region at Note 1p36 in neuroblastoma. Eur J Cancer. 2007 Feb;43(3):607-16 In colorectal cancer, loss of a 2 Mb region encompassing CAMTA1 had the strongest impact on Huentelman MJ, Papassotiropoulos A, Craig DW, Hoerndli FJ, Pearson JV, Huynh KD, Corneveaux J, Hänggi J, Mondadori survival among all copy number alterations identified CR, Buchmann A, Reiman EM, Henke K, de Quervain DJ, by genome-wide copy number analysis (Kim et al., Stephan DA. Calmodulin-binding transcription activator 1 2006). As in neuroblastoma, low expression of (CAMTA1) alleles predispose human episodic memory CAMTA1 mRNA is an independent predictor of poor performance. Hum Mol Genet. 2007 Jun 15;16(12):1469-77 outcome in colorectal cancer (Kim et al., 2006). Henrich KO and Westermann F.. CAMTA1. Encyclopedia of Cancer 2008; M. Schwab, Ed., Berlin, New York, Heidelberg, References Springer. (REVIEW) Ichimura K, Vogazianou AP, Liu L, Pearson DM, Bäcklund LM, Nagase T, Ishikawa K, Suyama M, Kikuno R, Hirosawa M, Plant K, Baird K, Langford CF, Gregory SG, Collins VP. 1p36 Miyajima N, Tanaka A, Kotani H, Nomura N, Ohara O. is a preferential target of chromosome 1 deletions in astrocytic Prediction of the coding sequences of unidentified human tumours and homozygously deleted in a subset of genes. XII. The complete sequences of 100 new cDNA clones glioblastomas. Oncogene. 2008 Mar 27;27(14):2097-108 from brain which code for large proteins in vitro. DNA Res. 1998 Dec 31;5(6):355-64 This article should be referenced as such: Bouché N, Scharlat A, Snedden W, Bouchez D, Fromm H. A Henrich KO. CAMTA1 (calmodulin binding transcription novel family of calmodulin-binding transcription activators in activator 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):441-442.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 442 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

DYRK1A (dual-specificity tyrosine-(Y)- phosphorylation regulated kinase 1A) Maria L Arbonés, Susana de la Luna Center for Genomic Regulation (CRG) and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain (MLA), Institucio Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain (SdlL)

Published in Atlas Database: September 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/DYRK1AID43234ch21q22.html DOI: 10.4267/2042/45022 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

- DSCR8 (Down syndrome critical region gene 8; Identity miscRNA). Location 21q22.2. Other names: DYRK, DYRK1, HP86, MNB, MNBH Note HGNC (Hugo): DYRK1A The chromosomal region depicted in the above figure is Location: 21q22.13 part of the "critical region" for Down syndrome, commonly named Down syndrome critical region Local order: (DSCR). This region, which encompasses 5.4 Mb of Genes surrounding DYRK1A and their chromosome chromosome 21 and contains around 30 genes, has locations ordered from centromere to telomere been defined in phenotype-genotype correlation studies (according to NCBI Map Viewer): of individuals with partial trisomy 21 (Delabar et al., - TTC3 (tetratricopeptide repeat domain 3). Location 1993). Similar correlation studies of patients with 21q22.13. partial monosomy 21 have defined a common region - DSCR9 (Down syndrome critical region gene 9; within DSCR that when deleted causes microcephaly miscRNA, non-protein coding). Location 21q22.13. and developmental delay. This region expands 1.2 Mb - DSCR3 (Down syndrome critical region gene 3). and contains 10 genes including DYRK1A (Matsumoto Location 21q22.13. et al., 1997). The identification of two unrelated - DYRK1A. Location 21q22.13. patients with microcephaly carrying a DYRK1A - LOC100289229 (hypothetical gene). Location truncated mutation in hemizygosis (see bellow; Moller 21q22.13. et al., 2007) indicates that haploinsufficiency of - KCNJ6 (potassium inwardly-rectifying channel, DYRK1A is most likely the cause of the common subfamily J, member 6). Location: 21q22.1; 21q22.1. features presented by these two patients and by patients - DSCR4 (Down syndrome critical region gene 4). with partial monosomy 21. Location 21q22.2.

Map of the Chromosomal region in 21q22.13 - 21q22.2 where DYRK1A is located. Genes (in green) and predicted gene (in black) surrounding DYRK1A are depicted according to the chromosomal position in NCBI Map Viewer (version released in August 2009). Direction of the arrowhead indicates the orientation of the gene.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 443 DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) Arbonés ML, de la Luna S

Schematic representation of the exon-intron organization of human DYRK1A gene. The size of introns and exons is in kb. Exons (boxes) are numbered and drawn to scale: in orange, coding exons; in blue, alternative first exons. The position of major transcription start sites (TSSs) according to Maenz et al. (2008) is indicated with blue arrows. Alternative splicing events are shown with dotted lines and numbered from a-e.

Western blot with antibodies raised against the C- DNA/RNA terminal, the protein appears as three bands around 90 Description kDa. The DYRK1A gene contains at least 12 exons spanning Expression approximately 149.7 kb of genomic DNA. At the RNA level, DYRK1A is expressed as an Transcription approximately 6 kb transcript in many fetal and adult tissues including brain, heart, lung and skeletal muscle Several transcripts have been identified as result of (Guimerá et al., 1996; Shindoh et al., 1996; Song et al., alternative splicing (Wang et al., 1998; Guimerá et al., 1996). At the protein level, human DYRK1A 1999; Maenz et al., 2008). The use of two alternative expression has been mainly studied in the central first exons, controlled by different promoter sequences, nervous system, where it is detected in cortex, does not affect the open reading frame (alternative hippocampus, amygdala, thalamus and substantia nigra splicing events a and b). These two promoters differ in (Wegiel et al., 2004). The number of DYRK1A their strength and regulation by the transcription factor immunopositive neurons increases with maturation of E2F1 (Maenz et al., 2008). Exclusion of exon 2 would human brain and an increase in the number of give rise to a N-terminal truncated protein (alternative DYRK1A-positive astrocytes in aged people has also splicing event c). The use of an alternative acceptor site been observed (Weigel et al., 2004). Work done in within exon 4 (alternative splicing events d and e) mice has led to the proposal that Dyrk1a is expressed in generates two protein variants that differ by the sequential phases of central nervous system inclusion/exclusion of 9 amino acids in the N-terminal development; i) scattered expression in individual pre- region (Kentrup et al., 1996; Guimerá et al., 1999). No neurogenic progenitors; ii) throughout the cell cycle in functional differences have been associated to any of neurogenic progenitors; ii) down-regulated in post- these variants. Further splice variants, affecting the C- mitotic neurons as they migrate radially; and iv) terminal region, were identified by PCR cloning sustained expression in late differentiating neurons (Guimerá et al., 1999), although the existence of the (Hammerle et al., 2008). Based on these observations, protein isoforms encoded by these transcripts has not DYRK1A has been suggested to be critical for the yet been confirmed. coupling of the sequential events required for proper Pseudogene neuronal development. No pseudogene reported. The analysis of the human DYRK1A promoter found two promoter regions that respond differentially to the Protein cell cycle-related transcription factor E2F1 (Maenz et al., 2008). Binding of the AP4-geminin complex to the Description human DYRK1A promoter has been shown by The DYRK1A gene encodes two main protein isoforms chromatin immunoprecipitation assays; moreover, the of 763 and 754 amino acids. DYRK1A is a protein DYRK1A promoter is downregulated by AP4 kinase that belongs to the DYRK family of dual- overexpression in gene reporter assays, suggesting that specificity protein kinases (CMGC group: DYRK the AP4-geminin repressor complex could be family: DYRK subfamily). The kinase domain is responsible of the DYRK1A downregulation in non- located centrally in the primary structure of the protein. neuronal cells (Kim et al., 2004). In mice, Dyrk1a is DYRK1A shares with the other DYRKs a conserved transcriptionally induced by the receptor activator of motif N-terminal to the kinase domain known as nuclear factor kappa-B ligand (RANKL) cytokine DYRK homology (DH)-box. It also harbors a through the activity of the nuclear factor of activated T- functional, bipartite nuclear localization signal (NLS) cells (NFAT) transcription factors (Lee et al., 2009). N-terminal to the DH-box, a second NLS between Localisation subdomains X and XI within the kinase domain, a C- When overexpressed in mammalian cells, DYRK1A terminal PEST motif, and a polyhistidine tract that acts protein mainly localizes in the nucleus and shows a as a nuclear speckle targeting signal. When analyzed by punctuated staining that it is

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 444 DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) Arbonés ML, de la Luna S

Schematic representation of DYRK1A protein. NLS: nuclear localization signal. DH: DYRK-homology box. PEST: Pest motif. His: Polyhistidine stretch. S/T: Serine and threonine-rich region. The line shows the alternatively spliced segment of 9 amino acids. compatible with its accumulation in nuclear speckles potentiates nerve growth factor (NGF)-mediated (or splicing factor compartment) (Becker et al., 1998; neuronal differentiation in PC12 cells through an Alvarez et al., 2003). Two nuclear localization signals enhancement of Ras/MAPK signalling (Kelly and contribute to DYRK1A nuclear translocation and the Ramahni, 2005) or exit from the cell cycle in neuronal histidine stretch is responsible for the accumulation of progenitors (Park et al., 2010; Yabut et al., 2010). the protein in nuclear speckles (Alvarez et al., 2003). Furthermore, an increase in DYRK1A gene dosage Endogenous DYRK1A is detected in both the nucleus alters the levels of neuron-restrictive silencer factor and the cytosol of human neurons, but only in the (NRSF or REST), a key regulator of neuronal cytosol of astrocytes (Wegiel et al., 2004). Within the differentiation (Canzonetta et al., 2008). In the adult cytosol, DYRK1A accumulates in the cell bodies, brain, DYRK1A regulates epidermal growth factor dendrites and synapses (Wegiel et al., 2004). Similar (EGF) signalling in stem cell daughters and reduced behaviour has been described for mouse and chicken levels of DYRK1A compromise stem cell longevity Dyrk1a (Marti et al., 2003; Hammerle et al., 2003; (Ferron et al., 2010). These evidences together with Laguna et al., 2008). These findings also correlate with DYRK1A overexpression in Down syndrome the observed nuclear-cytosolic partitioning of Dyrk1a individuals (Dowjat et al., 2007) and the phenotype of in mouse brain by biochemical fractionation (Aranda et mouse models of overexpression (Smith et al., 1997; al., 2008). Altafaj et al., 2001; Ahn et al., 2006), have led to the In certain pathological situations like the proposal of DYRK1A as a main contributor to Down neurodegeneration associated to sporadic Alzheimer's syndrome neurological alterations (reviewed in Park et disease and Down syndrome, DYRK1A is detected in al., 2009a). Finally, there are increasing evidences of a neurofibrillary tangles, in granules in granulovacuolar link of DYRK1A with neurodegeneration, given that it degeneration and in corpora amylacea (Wegiel et al., phosphorylates several proteins related to this cellular 2008). process including tau, alpha-synuclein, septin-4, Function presenilin or amyloid beta precursor protein (APP) and accumulates in amyloid lesions (reviewed in Park et al., DYRK1A is a dual-specificity protein kinase that 2009b). autophosphorylates on tyrosine serine and threonine Additionally, the participation of DYRK1A in different residues, but phosphorylate substrates only on serine or cellular processes and signal transduction pathways has threonine residues. Autophosphorylation of Tyr been inferred from the activity of its interactors and 312/321 (754/763 variants) in the activation loop is substrates. Functional activities include apoptosis, required for full catalytic activity (Himpel et al., 2001; exerting a protective role through phosphorylation of Lochhed et al., 2005). A full list of the residues caspase-9 or the deactelylase sirtuin-1 (SIRT1) (Seifert phosphorylated in DYRK1A can be found at et al., 2008; Laguna et al., 2008; Guo et al., 2010); PhoSphositePlus. A consensus phosphorylation endocytosis, through the interaction and sequence has been proposed for DYRK1A (RPXS/TP) phosphorylation of the GTPase dynamin-1, the (Himpel et al., 2000), although some phosphorylation phosphatase synaptojanin-1 or the scaffold protein sites have been found that do not fit within the amphiphysin-1 (Murakami et al., 2009; and references consensus such as in the case of LTAT(434)P in therein); cytoskeletal-related processes, through SF3B1/SAP155 or RPAS(640)V in glycogen synthase phosphorylation of tau or microtubule-associated (Skurat et al., 2004; de Graaf et al., 2006). protein 1B (MAP1B) (Woods et al., 2001; Scales et al., The phenotypic analysis of a loss-of-function mouse 2009); receptor tyrosine kinase-dependent signalling, model has provided information about DYRK1A through the interaction with the kinase B-Raf (NGF) or functional roles. Null Dyrk1a embryos present a severe the inhibitor Sprouty-2 (FGF) (Kelly and Rahmani, developmental delay and die around embryonic day 2005; Aranda et al., 2008). 10.5, and the analysis of the heterozygous animals has A relevant group of DYRK1A substrates are chromatin indicated that DYRK1A plays a role in brain regulators and transcription factors suggesting a role development (Fotaki et al., 2002). In this context, for DYRK1A in the regulation of gene expression DYRK1A has been shown to participate in programs. When assayed in gene reporter assays, neuritogenesis (Benavides-Piccione et al., 2005; DYRK1A works as an activator of cAMP responsive Gockler et al., 2009; Lepagnol-Bestel et al., 2009; element binding protein 1 Scales et al., 2009), and DYRK1A overexpression

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 445 DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) Arbonés ML, de la Luna S

Schematic representation of DYRK1A gene indicating the location of the breakpoints in chromosome 21 (vertical arrows) of the reported patients with a de novo balanced translocation.

(CREB1) (Yang et al., 2001), Gli1 (Mao et al., 2002), forkhead box protein O1 (FOXO1/FKHR) (von Groote- Implicated in Bidlingmaier et al., 2003) or androgen receptor- Various types of cancer and disease interacting protein 4 (ARIP4/RAD54L2) (Sitz et al., 2004) and as an inhibitor of Notch-dependent Disease transcription (Fernandez-Martinez et al., 2009). There are evidences suggesting a role of DYRK1A in DYRK1A acts as negative regulator of NFAT several human diseases. All the evidences, with the transcription factors in distinct cellular environments exception of those reported in Moller et al. (2008) by inducing their translocation to the cytosol (Arron et regarding microcephaly, are based on in vitro al., 2006; Kuhn et al., 2009; Lee et al., 2009). molecular and biochemical studies, on RNA expression DYRK1A cooperation with glycogen synthase kinase 3 studies and on studies in model organisms, mainly in (GSK3) promotes the degradation of cryptochrome 2 the mouse. The list of human diseases includes Down (CRY2), thus contributing to the internal cellular clock syndrome (reviewed in Hammerle et al., 2003; Park et (Kurabayashi et al., 2010). Another DYRK1A target is al., 2009a), early onset microcephaly (Moller et al., the tumour protein p53, either by being a direct 2008), Alzheimer and Huntington diseases (reviewed in DYRK1A substrate or indirectly through the Park et al., 2009b), several cancer types (Baek et al., phosphorylation by DYRK1A of the p53 deactelylase 2009; de Wit et al., 2002) and cardiac hypertrophy SIRT1 (Guo et al., 2010; Park et al., 2010). Finally, (Khun at al., 2009; Raaf et al., 2009). DYRK1A has been proposed as a regulator of splicing Oncogenesis based on DYRK1A localization in nuclear speckles Experiments performed in the Ts65Dn trisomic mouse (Alvarez et al., 2003) and on having several splicing model for Down syndrome have shown that factors such SF3b1 or ASF as substrates (de Graaf et overexpression of DYRK1A contributes to the al., 2006; Shi et al., 2008). In fact, DYRK1A attenuation of the calcineurin pathway induced by the phosphorylation of the alternative splicing factor ASF increased dosage of RCAN1 (also named DSCR1), a prevents ASF-mediated inclusion of the alternatively chromosome 21 gene that like DYRK1A is triplicated spliced exon 10 in tau mRNA (Shi et al., 2008). in this model (Baek et al., 2009). The reduction in Homology calcineurin signalling leads to a significant reduction of angiogenesis and tumour growth. Because the Mouse and human DYRK1A show a high degree of incidence of many cancer types is significantly reduced homology at the amino acid level (99%). The in people with Down syndrome (complete trisomy of Drosophila DYRK1A orthologue is the minibrain gene chromosome 21), the results obtained in mice suggest (Tejedor et al., 1995). Within the DYRK family of that small differences in the amount of DYRK1A kinases, the closest paralogous is DYRK1B. kinase could, through the calcineurin-dependent regulation of angiogenesis, modify the growth of some Mutations type of tumours also in humans (Pussegoda et al., Note 2010). By contrary, phosphorylation of caspase-9 by A truncated mutation in DYRK1A gene has been DYRK1A may have a negative role in cancer because a identified in two unrelated patients that present prenatal reduction of caspase-9 apoptotic activity protects mitotic cells from apoptosis and promotes cell survival onset of microcephaly, intrauterine growth retardation, during tumorigenesis (Allan and Clarke, 2007). developmental delay, severe feeding problems and DYRK1A has been shown to interact with two viral febrile seizures. In both patients, truncation of oncoproteins, adenovirus E1A and human papilloma DYRK1A results from a de novo balanced translocation involving chromosome 21, virus (HPV) E7 (Zhang et al., 2001; Liang et al., 2008; t(9;21)(p12;q22) in one patient and t(2;21)(q22;q22) in Komorek et al., 2010), which could be suggestive of the involvement of DYRK1A in oncogenic the other (Moller et al., 2008). Location of the transformation. breakpoints within DYRK1A is depicted in the diagram above.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 446 DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) Arbonés ML, de la Luna S

Melanoma Song WJ, Sternberg LR, Kasten-Sportes C, Keuren ML, Chung SH, Slack AC, Miller DE, Glover TW, Chiang PW, Lou L, Kurnit Note DM.. Isolation of human and murine homologues of the DYRK1A mRNA levels in a melanoma cell line with Drosophila minibrain gene: human homologue maps to 21q22.2 in the Down syndrome "critical region". Genomics. high metastatic potential (Mel57) are lower than in a 1996 Dec 15;38(3):331-9. melanoma cell line (1F6) with poor metastatic potential. DYRK1A mRNA levels are down-regulated Matsumoto N, Ohashi H, Tsukahara M, Kim KC, Soeda E, Niikawa N. Possible narrowed assignment of the loci of in vivo during melanocytic tumour progression, and in monosomy 21-associated microcephaly and intrauterine tumour samples from lung, oesophagus, colon, growth retardation to a 1.2-Mb segment at 21q22.2. Am J Hum pancreas and testis when compared to normal samples Genet. 1997 Apr;60(4):997-9 from the same tissues (de Wit et al., 2002). Smith DJ, Stevens ME, Sudanagunta SP, Bronson RT, Makhinson M, Watabe AM, O'Dell TJ, Fung J, Weier HU, Cervical cancer Cheng JF, Rubin EM. Functional screening of 2 Mb of human Note chromosome 21q22.2 in transgenic mice implicates minibrain in learning defects associated with Down syndrome. Nat HPV type 16 (HPV16) is a tumorigenic virus that Genet. 1997 May;16(1):28-36 causes the development of cervical cancer. DYRK1A is present in HPV16 immortalized keratinocytes but not Becker W, Weber Y, Wetzel K, Eirmbter K, Tejedor FJ, Joost HG. Sequence characteristics, subcellular localization, and in primary keratynocytes; moreover, malignant cervical substrate specificity of DYRK-related kinases, a novel family of lesions contain more DYRK1A than normal tissue dual specificity protein kinases. J Biol Chem. 1998 Oct (Chang et al., 2007). Biochemical data lead to the 2;273(40):25893-902 suggestion that the increased expression of DYRK1A Wang J, Kudoh J, Shintani A, Minoshima S, Shimizu N. in immortalized keratinocytes and in cervical tissues Identification of two novel 5' noncoding exons in human acts as an antiapoptotic factor in the FKHR-dependent MNB/DYRK gene and alternatively spliced transcripts. pathway leading to tumour development (Chang et al., Biochem Biophys Res Commun. 1998 Sep 29;250(3):704-10 2007). Additionally, DYRK1A interacts and Guimera J, Casas C, Estivill X, Pritchard M. Human minibrain phosphorylates the HPV16 protein E7 leading to its homologue (MNBH/DYRK1): characterization, alternative splicing, differential tissue expression, and overexpression in stabilization and to an increase in its capacity for Down syndrome. Genomics. 1999 May 1;57(3):407-18 forming clones in a colony-formation assay (Liang et al., 2008). Himpel S, Tegge W, Frank R, Leder S, Joost HG, Becker W. Specificity determinants of substrate recognition by the protein Pancreatic endocrine neoplasms kinase DYRK1A. J Biol Chem. 2000 Jan 28;275(4):2431-8 Note Altafaj X, Dierssen M, Baamonde C, Martí E, Visa J, Guimerà J, Oset M, González JR, Flórez J, Fillat C, Estivill X. Microarray hybridization data showed up-regulation of Neurodevelopmental delay, motor abnormalities and cognitive DYRK1A in metastatic pancreatic endocrine deficits in transgenic mice overexpressing Dyrk1A (minibrain), neoplasms when compared with nonmetastatic a murine model of Down's syndrome. Hum Mol Genet. 2001 pancreatic endocrine neoplasms (Hansel et al., 2004). Sep 1;10(18):1915-23 Himpel S, Panzer P, Eirmbter K, Czajkowska H, Sayed M, References Packman LC, Blundell T, Kentrup H, Grötzinger J, Joost HG, Becker W. Identification of the autophosphorylation sites and Delabar JM, Theophile D, Rahmani Z, Chettouh Z, Blouin JL, characterization of their effects in the protein kinase DYRK1A. Prieur M, Noel B, Sinet PM. Molecular mapping of twenty-four Biochem J. 2001 Nov 1;359(Pt 3):497-505 features of Down syndrome on chromosome 21. Eur J Hum Genet. 1993;1(2):114-24 Woods YL, Cohen P, Becker W, Jakes R, Goedert M, Wang X, Proud CG.. The kinase DYRK phosphorylates protein- Tejedor F, Zhu XR, Kaltenbach E, Ackermann A, Baumann A, synthesis initiation factor eIF2Bepsilon at Ser539 and the Canal I, Heisenberg M, Fischbach KF, Pongs O. minibrain: a microtubule-associated protein tau at Thr212: potential role for new protein kinase family involved in postembryonic DYRK as a glycogen synthase kinase 3-priming kinase. neurogenesis in Drosophila. Neuron. 1995 Feb;14(2):287-301 Biochem J. 2001 May 1;355(Pt 3):609-15. Guimerá J, Casas C, Pucharcòs C, Solans A, Domènech A, Yang EJ, Ahn YS, Chung KC. Protein kinase Dyrk1 activates Planas AM, Ashley J, Lovett M, Estivill X, Pritchard MA. A cAMP response element-binding protein during neuronal human homologue of Drosophila minibrain (MNB) is expressed differentiation in hippocampal progenitor cells. J Biol Chem. in the neuronal regions affected in Down syndrome and maps 2001 Oct 26;276(43):39819-24 to the critical region. Hum Mol Genet. 1996 Sep;5(9):1305-10 Zhang Z, Smith MM, Mymryk JS. Interaction of the E1A Kentrup H, Becker W, Heukelbach J, Wilmes A, Schürmann A, oncoprotein with Yak1p, a novel regulator of yeast Huppertz C, Kainulainen H, Joost HG. Dyrk, a dual specificity pseudohyphal differentiation, and related mammalian kinases. protein kinase with unique structural features whose activity is Mol Biol Cell. 2001 Mar;12(3):699-710 dependent on tyrosine residues between subdomains VII and VIII. J Biol Chem. 1996 Feb 16;271(7):3488-95 de Wit NJ, Burtscher HJ, Weidle UH, Ruiter DJ, van Muijen GN. Differentially expressed genes identified in human Shindoh N, Kudoh J, Maeda H, Yamaki A, Minoshima S, melanoma cell lines with different metastatic behaviour using Shimizu Y, Shimizu N. Cloning of a human homolog of the high density oligonucleotide arrays. Melanoma Res. 2002 Drosophila minibrain/rat Dyrk gene from "the Down syndrome Feb;12(1):57-69 critical region" of chromosome 21. Biochem Biophys Res Commun. 1996 Aug 5;225(1):92-9 Fotaki V, Dierssen M, Alcántara S, Martínez S, Martí E, Casas C, Visa J, Soriano E, Estivill X, Arbonés ML. Dyrk1A

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 447 DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) Arbonés ML, de la Luna S

haploinsufficiency affects viability and causes developmental de Graaf K, Czajkowska H, Rottmann S, Packman LC, delay and abnormal brain morphology in mice. Mol Cell Biol. Lilischkis R, Lüscher B, Becker W. The protein kinase 2002 Sep;22(18):6636-47 DYRK1A phosphorylates the splicing factor SF3b1/SAP155 at Thr434, a novel in vivo phosphorylation site. BMC Biochem. Mao J, Maye P, Kogerman P, Tejedor FJ, Toftgard R, Xie W, 2006 Mar 2;7:7 Wu G, Wu D. Regulation of Gli1 transcriptional activity in the nucleus by Dyrk1. J Biol Chem. 2002 Sep 20;277(38):35156- Kim MY, Jeong BC, Lee JH, Kee HJ, Kook H, Kim NS, Kim 61 YH, Kim JK, Ahn KY, Kim KK. A repressor complex, AP4 transcription factor and geminin, negatively regulates Alvarez M, Estivill X, de la Luna S. DYRK1A accumulates in expression of target genes in nonneuronal cells. Proc Natl splicing speckles through a novel targeting signal and induces Acad Sci U S A. 2006 Aug 29;103(35):13074-9 speckle disassembly. J Cell Sci. 2003 Aug 1;116(Pt 15):3099- 107 Allan LA, Clarke PR. Phosphorylation of caspase-9 by CDK1/cyclin B1 protects mitotic cells against apoptosis. Mol Hämmerle B, Carnicero A, Elizalde C, Ceron J, Martínez S, Cell. 2007 Apr 27;26(2):301-10 Tejedor FJ. Expression patterns and subcellular localization of the Down syndrome candidate protein MNB/DYRK1A suggest Chang HS, Lin CH, Yang CH, Yen MS, Lai CR, Chen YR, a role in late neuronal differentiation. Eur J Neurosci. 2003 Liang YJ, Yu WC. Increased expression of Dyrk1a in HPV16 Jun;17(11):2277-86 immortalized keratinocytes enable evasion of apoptosis. Int J Cancer. 2007 Jun 1;120(11):2377-85 Martí E, Altafaj X, Dierssen M, de la Luna S, Fotaki V, Alvarez M, Pérez-Riba M, Ferrer I, Estivill X. Dyrk1A expression Dowjat WK, Adayev T, Kuchna I, Nowicki K, Palminiello S, pattern supports specific roles of this kinase in the adult central Hwang YW, Wegiel J. Trisomy-driven overexpression of nervous system. Brain Res. 2003 Feb 28;964(2):250-63 DYRK1A kinase in the brain of subjects with Down syndrome. Neurosci Lett. 2007 Feb 8;413(1):77-81 von Groote-Bidlingmaier F, Schmoll D, Orth HM, Joost HG, Becker W, Barthel A. DYRK1 is a co-activator of FKHR Aranda S, Alvarez M, Turró S, Laguna A, de la Luna S. (FOXO1a)-dependent glucose-6-phosphatase gene Sprouty2-mediated inhibition of fibroblast growth factor expression. Biochem Biophys Res Commun. 2003 Jan signaling is modulated by the protein kinase DYRK1A. Mol Cell 17;300(3):764-9 Biol. 2008 Oct;28(19):5899-911 Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo CJ, Canzonetta C, Mulligan C, Deutsch S, Ruf S, O'Doherty A, Lyle Maitra A. Met proto-oncogene and insulin-like growth factor R, Borel C, Lin-Marq N, Delom F, Groet J, Schnappauf F, De binding protein 3 overexpression correlates with metastatic Vita S, Averill S, Priestley JV, Martin JE, Shipley J, Denyer G, ability in well-differentiated pancreatic endocrine neoplasms. Epstein CJ, Fillat C, Estivill X, Tybulewicz VL, Fisher EM, Clin Cancer Res. 2004 Sep 15;10(18 Pt 1):6152-8 Antonarakis SE, Nizetic D. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating pluripotency and Sitz JH, Tigges M, Baumgärtel K, Khaspekov LG, Lutz B. embryonic stem cell fate in Down syndrome. Am J Hum Genet. Dyrk1A potentiates steroid hormone-induced transcription via 2008 Sep;83(3):388-400 the chromatin remodeling factor Arip4. Mol Cell Biol. 2004 Jul;24(13):5821-34 Hämmerle B, Elizalde C, Tejedor FJ. The spatio-temporal and subcellular expression of the candidate Down syndrome gene Skurat AV, Dietrich AD. Phosphorylation of Ser640 in muscle Mnb/Dyrk1A in the developing mouse brain suggests distinct glycogen synthase by DYRK family protein kinases. J Biol sequential roles in neuronal development. Eur J Neurosci. Chem. 2004 Jan 23;279(4):2490-8 2008 Mar;27(5):1061-74 Wegiel J, Kuchna I, Nowicki K, Frackowiak J, Dowjat K, Laguna A, Aranda S, Barallobre MJ, Barhoum R, Fernández E, Silverman WP, Reisberg B, DeLeon M, Wisniewski T, Adayev Fotaki V, Delabar JM, de la Luna S, de la Villa P, Arbonés ML. T, Chen-Hwang MC, Hwang YW. Cell type- and brain The protein kinase DYRK1A regulates caspase-9-mediated structure-specific patterns of distribution of minibrain kinase in apoptosis during retina development. Dev Cell. 2008 human brain. Brain Res. 2004 Jun 4;1010(1-2):69-80 Dec;15(6):841-53 Benavides-Piccione R, Dierssen M, Ballesteros-Yáñez I, Liang YJ, Chang HS, Wang CY, Yu WC. DYRK1A stabilizes Martínez de Lagrán M, Arbonés ML, Fotaki V, DeFelipe J, HPV16E7 oncoprotein through phosphorylation of the Elston GN. Alterations in the phenotype of neocortical threonine 5 and threonine 7 residues. Int J Biochem Cell Biol. pyramidal cells in the Dyrk1A+/- mouse. Neurobiol Dis. 2005 2008;40(11):2431-41 Oct;20(1):115-22 Maenz B, Hekerman P, Vela EM, Galceran J, Becker W. Kelly PA, Rahmani Z. DYRK1A enhances the mitogen- Characterization of the human DYRK1A promoter and its activated protein kinase cascade in PC12 cells by forming a regulation by the transcription factor E2F1. BMC Mol Biol. 2008 complex with Ras, B-Raf, and MEK1. Mol Biol Cell. 2005 Mar 26;9:30 Aug;16(8):3562-73 Møller RS, Kübart S, Hoeltzenbein M, Heye B, Vogel I, Hansen Lochhead PA, Sibbet G, Morrice N, Cleghon V. Activation-loop CP, Menzel C, Ullmann R, Tommerup N, Ropers HH, Tümer Z, autophosphorylation is mediated by a novel transitional Kalscheuer VM. Truncation of the Down syndrome candidate intermediate form of DYRKs. Cell. 2005 Jun 17;121(6):925-36 gene DYRK1A in two unrelated patients with microcephaly. Am Ahn KJ, Jeong HK, Choi HS, Ryoo SR, Kim YJ, Goo JS, Choi J Hum Genet. 2008 May;82(5):1165-70 SY, Han JS, Ha I, Song WJ. DYRK1A BAC transgenic mice Seifert A, Allan LA, Clarke PR. DYRK1A phosphorylates show altered synaptic plasticity with learning and memory caspase 9 at an inhibitory site and is potently inhibited in defects. Neurobiol Dis. 2006 Jun;22(3):463-72 human cells by harmine. FEBS J. 2008 Dec;275(24):6268-80 Arron JR, Winslow MM, Polleri A, Chang CP, Wu H, Gao X, Shi J, Zhang T, Zhou C, Chohan MO, Gu X, Wegiel J, Zhou J, Neilson JR, Chen L, Heit JJ, Kim SK, Yamasaki N, Miyakawa Hwang YW, Iqbal K, Grundke-Iqbal I, Gong CX, Liu F. T, Francke U, Graef IA, Crabtree GR. NFAT dysregulation by Increased dosage of Dyrk1A alters alternative splicing factor increased dosage of DSCR1 and DYRK1A on chromosome (ASF)-regulated alternative splicing of tau in Down syndrome. 21. Nature. 2006 Jun 1;441(7093):595-600 J Biol Chem. 2008 Oct 17;283(42):28660-9

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 448 DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) Arbonés ML, de la Luna S

Wegiel J, Dowjat K, Kaczmarski W, Kuchna I, Nowicki K, Park J, Song WJ, Chung KC. Function and regulation of Frackowiak J, Mazur Kolecka B, Wegiel J, Silverman WP, Dyrk1A: towards understanding Down syndrome. Cell Mol Life Reisberg B, Deleon M, Wisniewski T, Gong CX, Liu F, Adayev Sci. 2009 Oct;66(20):3235-40 T, Chen-Hwang MC, Hwang YW. The role of overexpressed DYRK1A protein in the early onset of neurofibrillary Scales TM, Lin S, Kraus M, Goold RG, Gordon-Weeks PR. degeneration in Down syndrome. Acta Neuropathol. 2008 Nonprimed and DYRK1A-primed GSK3 beta-phosphorylation Oct;116(4):391-407 sites on MAP1B regulate microtubule dynamics in growing axons. J Cell Sci. 2009 Jul 15;122(Pt 14):2424-35 Baek KH, Zaslavsky A, Lynch RC, Britt C, Okada Y, Siarey RJ, Lensch MW, Park IH, Yoon SS, Minami T, Korenberg JR, Ferron SR, Pozo N, Laguna A, Aranda S, Porlan E, Moreno M, Folkman J, Daley GQ, Aird WC, Galdzicki Z, Ryeom S. Down's Fillat C, de la Luna S, Sánchez P, Arbonés ML, Fariñas I. syndrome suppression of tumour growth and the role of the Regulated segregation of kinase Dyrk1A during asymmetric calcineurin inhibitor DSCR1. Nature. 2009 Jun neural stem cell division is critical for EGFR-mediated biased 25;459(7250):1126-30 signaling. Cell Stem Cell. 2010 Sep 3;7(3):367-79 Fernandez-Martinez J, Vela EM, Tora-Ponsioen M, Ocaña OH, Guo X, Williams JG, Schug TT, Li X. DYRK1A and DYRK3 Nieto MA, Galceran J. Attenuation of Notch signalling by the promote cell survival through phosphorylation and activation of Down-syndrome-associated kinase DYRK1A. J Cell Sci. 2009 SIRT1. J Biol Chem. 2010 Apr 23;285(17):13223-32 May 15;122(Pt 10):1574-83 Komorek J, Kuppuswamy M, Subramanian T, Vijayalingam S, Göckler N, Jofre G, Papadopoulos C, Soppa U, Tejedor FJ, Lomonosova E, Zhao LJ, Mymryk JS, Schmitt K, Chinnadurai Becker W. Harmine specifically inhibits protein kinase DYRK1A G. Adenovirus type 5 E1A and E6 proteins of low-risk and interferes with neurite formation. FEBS J. 2009 cutaneous beta-human papillomaviruses suppress cell Nov;276(21):6324-37 transformation through interaction with FOXK1/K2 transcription factors. J Virol. 2010 Mar;84(6):2719-31 Kuhn C, Frank D, Will R, Jaschinski C, Frauen R, Katus HA, Frey N. DYRK1A is a novel negative regulator of Kurabayashi N, Hirota T, Sakai M, Sanada K, Fukada Y. cardiomyocyte hypertrophy. J Biol Chem. 2009 Jun DYRK1A and glycogen synthase kinase 3beta, a dual-kinase 19;284(25):17320-7 mechanism directing proteasomal degradation of CRY2 for circadian timekeeping. Mol Cell Biol. 2010 Apr;30(7):1757-68 Lee Y, Ha J, Kim HJ, Kim YS, Chang EJ, Song WJ, Kim HH. Negative feedback Inhibition of NFATc1 by DYRK1A regulates Park J, Oh Y, Yoo L, Jung MS, Song WJ, Lee SH, Seo H, bone homeostasis. J Biol Chem. 2009 Nov 27;284(48):33343- Chung KC. Dyrk1A phosphorylates p53 and inhibits 51 proliferation of embryonic neuronal cells. J Biol Chem. 2010 Oct 8;285(41):31895-906 Lepagnol-Bestel AM, Zvara A, Maussion G, Quignon F, Ngimbous B, Ramoz N, Imbeaud S, Loe-Mie Y, Benihoud K, Pussegoda KA. Down's syndrome patients are less likely to Agier N, Salin PA, Cardona A, Khung-Savatovsky S, Kallunki develop cancer. Clin Genet. 2010 Jul;78(1):35-7 P, Delabar JM, Puskas LG, Delacroix H, Aggerbeck L, Raaf L, Noll C, Cherifi M, Benazzoug Y, Delabar JM, Janel N. Delezoide AL, Delattre O, Gorwood P, Moalic JM, Simonneau Hyperhomocysteinemia-induced Dyrk1a downregulation M. DYRK1A interacts with the REST/NRSF-SWI/SNF results in cardiomyocyte hypertrophy in rats. Int J Cardiol. 2010 chromatin remodelling complex to deregulate gene clusters Nov 19;145(2):306-7 involved in the neuronal phenotypic traits of Down syndrome. Hum Mol Genet. 2009 Apr 15;18(8):1405-14 Yabut O, Domogauer J, D'Arcangelo G. Dyrk1A overexpression inhibits proliferation and induces premature Murakami N, Bolton D, Hwang YW. Dyrk1A binds to multiple neuronal differentiation of neural progenitor cells. J Neurosci. endocytic proteins required for formation of clathrin-coated 2010 Mar 17;30(11):4004-14 vesicles. Biochemistry. 2009 Oct 6;48(39):9297-305 Park J, Oh Y, Chung KC. Two key genes closely implicated This article should be referenced as such: with the neuropathological characteristics in Down syndrome: Arbonés ML, de la Luna S. DYRK1A (dual-specificity tyrosine- DYRK1A and RCAN1. BMB Rep. 2009 Jan 31;42(1):6-15 (Y)-phosphorylation regulated kinase 1A). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):443-449.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 449 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

FBLN1 (fibulin 1) Lorenzo Castagnoli, Elda Tagliabue, Serenella M Pupa Fondazione IRCCS Istituto Nazionale Dei Tumori - Molecular Targeting Unit, Dept of Experimental Oncology and Molecular Medicine, Via Amadeo 42, Milan, Italy (LC, ET, SMP)

Published in Atlas Database: September 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/FBLN1ID44462ch22q13.html DOI: 10.4267/2042/45023 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

exon length: 818, minimun exon length: 50. Number of Identity SNPs 1038. Four splice variants have been identified Other names: FBLN, FIBL1 which differ in the 3' end and encode different isoforms HGNC (Hugo): FBLN1 (A, B, C and D) (Pan et al., 1999). Variant D: This variant is considered the canonic Location: 22q13.31 transcript form: 2947 bp; Local order: Orientation: plus strand. - Including exons 18, 19 and 20; Note: Fibulin-1 is an extracellular matrix (ECM) and - Lacking exons 15 and 16. blood glycoprotein. It is a member of the fibulin Variant B: 2530 bp; glycoprotein family which includes 6 proteins thought - Including exon 17; to function as bridges in the organization of ECM - Lacking exons, 16, 18, 19 and 20. supramolecular structures (Argraves et al., 1990; Timpl Variant A: 2350bp; et al., 2003). - Including exon 15; - Lackings exons 16 to 20. DNA/RNA Variant C: 2313bp; - Including exon 16; Description - Lacking exons 15 and 17 to 20. Sequence length: 97, 71 kb, 20 exons, maximum

Transcription factor.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 450 FBLN1 (fibulin 1) Castagnoli L, et al.

Protein Description

Position Length Description 36-76 41 Anaphylatoxin-like 1 77-111 35 Anaphylatoxin-like 2 112-144 33 Anaphylatoxin-like 3 176-215 40 EGF-like 1 216-261 46 EGF-like 2; calcium-binding 262-307 46 EGF-like 3; calcium-binding 308-355 48 EGF-like 4; calcium-binding 356-398 43 EGF-like 5; calcium-binding 399-440 42 EGF-like 6; calcium-binding 441-480 40 EGF-like 7; calcium-binding 481-524 44 EGF-like 8; calcium-binding 525-578 54 EGF-like 9; calcium-binding Glycosylation 98; 535; 539 36↔61; 37↔68; 50↔69; 78↔109; 91↔110; 112↔136; 113↔143; 126↔144; 180↔190; 186↔199; 201↔214; 220↔233; 227↔242; 248↔260; 266↔279; 273↔288; 294↔306; Disulfide 312↔325; 319↔334; 341↔354; 360↔373; 367↔382; 384↔397; 403↔415; 411↔424; bond 426↔439; 445↔454; 450↔463; 465↔479; 485↔498; 494↔507; 509↔523; 529↔542; 536↔551; 556↔577

Amino acid modifications (UNIPROT)

Fibulin-1 homomultimerizes and interacts with various 1 also interacts with amyloid beta A4 (APP) (Ohsawa ECM components such as fibronectin (FN) (Balbona et et al., 2001), insulin-like growth factor-binding protein al., 1992), laminin subunits alpha-1 and apha-2 9 (NOV) (Perbal et al., 1999), fibrinogen (FGB) (Tran (LAMA1 and LAMA2), nidogen (NID), Aggrecan core et al., 1995), and human papillomavirus (HPV) type 16, protein (ACAN), versican core protein (CSPG2) and 18, 31 proteins (Du et al., 2002). type IV collagen proteins (Timpl et al., 2003). Fibulin-

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 451 FBLN1 (fibulin 1) Castagnoli L, et al.

Localisation Implicated in Secreted, extracellular space, extracellular matrix. Various cancers Function Note Fibulin-1 is incorporated into FN-containing matrix Several studies have reported that fibulin-1 is fibers. It plays a role in cell adhesion and migration overexpressed in various human neoplasias and it is along protein fibers within the ECM and is important implicated in processes such as invasion, motility, and for certain developmental processes. Fibulin-1 in vivo tumor growth (Qing et al., 1997; Twal et al., contributes to the supramolecular organization of ECM 2001; Du et al., 2002; Moll et al., 2002; Greene et al., architecture, in particular to that of the basement 2003). Fibulin-1 inhibits in vitro adhesion and motility membrane. It is implicated in cellular transformation of various carcinoma cell lines (Twal et al., 2001). and tumor invasion, and can behave both as an oncosuppressor and oncogene depending on tissue Breast cancer environment. It also plays a role in hemostasis and Prognosis thrombosis owing to its ability to bind fibrinogen and Fibulin-1 was found aberrantly expressed in ~35% of incorporate into clots and plays a significant role in 528 human primary breast cancers. Its expression is modulating the neurotrophic activities of APP, associated with improved survival in patients with particularly soluble APP (Timpl et al., 2003). lymphoid infiltrate at the tumor site (Pupa et al., 2004), Homology suggesting a possible involvement in triggering a Paralogs: latent-transforming growth factor beta- protective antitumor immune response. Fibulin-1 binding protein 1 (LTBP1), LTBP2, LTBP3, LTBP4, induces specific B- and T-cell-mediated responses in fibrillin-1 (FBN1), FBN2, FBN3, FBLN5, fibulin-2 breast cancer patients (Forti et al., 2002; Pupa et al., (FBLN2), epidermal growth factor-like protein 6 2004). Its overexpression can serve as a tool for early (EGFL6), nephronectin (NPNT), EGF-containing detection of breast cancer (Pupa et al., 2002) and acts to fibulin-like extracellular matrix protein 2 (EFEMP2), promote breast cancer cell survival during doxorubicin Von Willebrand factor C (VWCE). treatment (Pupa et al., 2007). In a series of 36 primary breast carcinomas, the expression of mature fibulin-1 Mutations polypeptide (100 kDa) did not correlate with estrogen receptor-alpha (ERalpha) or progesterone receptor (PR) Note levels, whereas a positive correlation was found - Location: exon 19 (acceptor splice site) between fibulin-1 processing (50 kDa fragment) and - Substitution: G-A ERalpha and PR protein levels (Greene et al., 2003). - Phenotype: Bernard-Soulier syndrome

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 452 FBLN1 (fibulin 1) Castagnoli L, et al.

Ovarian cancer Placenta dysplasia Disease Disease The molecular mechanisms involved in ovarian Placental dysplasia is a rare human placental disorder carcinogenesis remain unclear, but growing evidence in which the placenta is enlarged and contains cystic indicates that estrogens promote progression of ovarian villi and dilated vasculature. A significant correlation cancer and increase expression levels of some secreted was observed between fibulin-1 gene overexpression proteins. Differential overexpression of ERalpha versus and murine placental overgrowth (Singh et al., 2006), ERbeta has been demonstrated during ovarian suggesting that the gene and its product have a carcinogenesis (Clinton et al., 1996; Moll et al., 2002), functional role in placenta development. suggesting that estrogen-induced proteins, including Morphogenesis of neural crest-derived fibulin-1, may act as ovarian tumor-promoting agents. In ovarian tissues and cancer cell lines, fibulin-1C and - structures 1D are the predominant forms, whereas fibulin-1A and Disease -1B are weakly expressed. An increased fibulin-1C:-1D A significant negative correlation between fibulin-1 mRNA ratio in ovarian cancer cells as compared to that gene expression and some morphogenic anomalies of in normal ovaries has been observed, suggesting that neural crest-derived structures in mice has been the C variant is the main one involved in ovarian reported (Cooley et al., 2008). Such fibulin-1-deficient carcinogenesis. Fibulin-1C overexpression might mice exhibit cardiac ventricular wall thinning and provide a clue in understanding the putative role of ventricular septal defects, with double outlet right estrogens in ERalpha-promoted ovarian tumor ventricle or overriding aorta, as well as aortic arch progression (Moll et al., 2002). arteries anomalies, hypoplasia of the thymus and Synpolydactyly thyroid, underdeveloped skull bones, malformations of cranial nerves and hemorrhagic blood vessels in the Disease head and neck. The spectrum of malformations is Synpolydactyly is a rare genetic disorder characterized consistent with a role for fibulin-1 in neural crest cell- by malformations in the hands and feet, with dependent development of these tissues. abnormalities including increased finger and toe numbers and fusion of digits into a single digit. Acute aortic dissection Molecular analysis of the reciprocal chromosomal Disease translocation t(12;22)(p11.2;q13.3) cosegregating with Acute aortic dissection (AAD) is a tear in the wall of a complex type of synpolydactyly indicated the aorta that causes blood to flow between the layers involvement of an alternatively spliced exon of the of the wall of the aorta and force the layers apart. AAD fibulin-1 gene (FBLN1 located in 22q13.3). is a life-threatening condition with high mortality and a Investigation of the possible functional involvement of mostly unclear pathophysiological mechanism. the fibulin-1 protein in the observed phenotype showed Downregulation of fibulin-1 noted in AAD compared that fibulin-1 is expressed in the ECM in association to control samples might determine a weakening of with the digits in the developing limb (Debeer et al., ECM in the aorta and/or interfere with the transmission 2002). Thus, t(12;22) might result in haplo- of cellular signals causing AAD (Mohamed et al., insufficiency of the fibulin-1D variant, leading to the 2009). observed limb malformations. Atherosclerotic lesions Bernard-Soulier syndrome Disease Disease Fibulin-1 deposits were found in association with Bernard-Soulier syndrome is an autosomic-dominant fibrinogen in atherosclerotic lesions and in regions disease that causes alterations of the containing fresh thrombi. By contrast, fibulin-1 was not megakaryocyte/platelet lineage and is characterized by detected in sclerotic regions and low levels were bleeding tendency, giant blood platelets and low associated with smooth muscle cells within and outside platelet counts. An unexpected mutation in the splice lesions (Argraves et al., 2009). acceptor site of fibulin-1 exon 19 was found in affected Thrombosis individuals of the Israeli Fechtner family. In all affected individuals from eight families, expression of Disease fibulin-1 variant D was inhibited by putative antisense Thrombosis, the formation of a blood clot (thrombus) RNA (Lanza, 2006), raising the possibility that these inside a blood vessel, obstructs blood flow through the autosomal-dominant giant platelet syndromes are circulatory system. Analyses of blood plasma revealed associated with aberrant antisense gene regulation of an interaction between fibulin-1 and fibrinogen (Tran et fibulin-1. al., 1995), pointing to potential new roles for fibulin-1 in hemostasis as well as thrombosis.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 453 FBLN1 (fibulin 1) Castagnoli L, et al.

fibronectin-regulated cell adhesion and motility. J Cell Sci. Breakpoints 2001 Dec;114(Pt 24):4587-98 Note Debeer P, Schoenmakers EF, Twal WO, Argraves WS, De Smet L, Fryns JP, Van De Ven WJ. The fibulin-1 gene (FBLN1) Description: Translocation t(12;22)(p11.2;q13.3). is disrupted in a t(12;22) associated with a complex type of Phenotype: Synpolydactyly. synpolydactyly. J Med Genet. 2002 Feb;39(2):98-104 Analysis of a Belgian family with a complex type of Du M, Fan X, Hong E, Chen JJ. Interaction of oncogenic synpolydactyly (SPD2) and a t(12;22)(p11.2;q13.3) papillomavirus E6 proteins with fibulin-1. Biochem Biophys Res translocation identified the breakpoints located in the Commun. 2002 Aug 30;296(4):962-9 intron between the last 2 exons of the fibulin-1D splice Forti S, Scanlan MJ, Invernizzi A, Castiglioni F, Pupa S, variant isoform (exons 19, 20) and the 5' UTR of the Agresti R, Fontanelli R, Morelli D, Old LJ, Pupa SM, Ménard S. C12ORF2 gene. Fibroblasts derived from the patients Identification of breast cancer-restricted antigens by antibody displayed decreased levels of ECM-related fibulin-1D screening of SKBR3 cDNA library using a preselected patient's secreted into the culture medium, whereas levels of the serum. Breast Cancer Res Treat. 2002 Jun;73(3):245-56 fibulin-1C variant were normal. The findings are Moll F, Katsaros D, Lazennec G, Hellio N, Roger P, Giacalone consistent with the notion that the PL, Chalbos D, Maudelonde T, Rochefort H, Pujol P. Estrogen induction and overexpression of fibulin-1C mRNA in ovarian t(12;22)(p11.2;q13.3) translocation results in haplo- cancer cells. Oncogene. 2002 Feb 7;21(7):1097-107 insufficiency of the fibulin-1D variant, leading to the observed limb malformations. The authors noted that Pupa SM, Forti S, Balsari A, Ménard S. Humoral immune response for early diagnosis of breast carcinoma. Ann Oncol. the entire fibulin-1 gene is deleted in the chromosome 2002 Mar;13(3):483 22q13.3 deletion syndrome (Debeer et al., 2002). Greene LM, Twal WO, Duffy MJ, McDermott EW, Hill AD, O'Higgins NJ, McCann AH, Dervan PA, Argraves WS, References Gallagher WM. Elevated expression and altered processing of fibulin-1 protein in human breast cancer. Br J Cancer. 2003 Argraves WS, Tran H, Burgess WH, Dickerson K. Fibulin is an Mar 24;88(6):871-8 extracellular matrix and plasma glycoprotein with repeated domain structure. J Cell Biol. 1990 Dec;111(6 Pt 2):3155-64 Timpl R, Sasaki T, Kostka G, Chu ML. Fibulins: a versatile family of extracellular matrix proteins. Nat Rev Mol Cell Biol. Balbona K, Tran H, Godyna S, Ingham KC, Strickland DK, 2003 Jun;4(6):479-89 Argraves WS. Fibulin binds to itself and to the carboxyl- terminal heparin-binding region of fibronectin. J Biol Chem. Pupa SM, Argraves WS, Forti S, Casalini P, Berno V, Agresti 1992 Oct 5;267(28):20120-5 R, Aiello P, Invernizzi A, Baldassari P, Twal WO, Mortarini R, Anichini A, Ménard S. Immunological and pathobiological roles Tran H, Tanaka A, Litvinovich SV, Medved LV, Haudenschild of fibulin-1 in breast cancer. Oncogene. 2004 Mar CC, Argraves WS. The interaction of fibulin-1 with fibrinogen. A 18;23(12):2153-60 potential role in hemostasis and thrombosis. J Biol Chem. 1995 Aug 18;270(33):19458-64 Lanza F. Bernard-Soulier syndrome (hemorrhagiparous thrombocytic dystrophy). Orphanet J Rare Dis. 2006 Nov Clinton GM, Rougeot C, Derancourt J, Roger P, Defrenne A, 16;1:46 Godyna S, Argraves WS, Rochefort H. Estrogens increase the expression of fibulin-1, an extracellular matrix protein secreted Singh U, Sun T, Larsson T, Elliott RW, Kostka G, Fundele RH. by human ovarian cancer cells. Proc Natl Acad Sci U S A. Expression and functional analysis of fibulin-1 (Fbln1) during 1996 Jan 9;93(1):316-20 normal and abnormal placental development of the mouse. Placenta. 2006 Sep-Oct;27(9-10):1014-21 Qing J, Maher VM, Tran H, Argraves WS, Dunstan RW, McCormick JJ. Suppression of anchorage-independent growth Pupa SM, Giuffré S, Castiglioni F, Bertola L, Cantú M, et al. and matrigel invasion and delayed tumor formation by elevated Regulation of breast cancer response to chemotherapy by expression of fibulin-1D in human fibrosarcoma-derived cell fibulin-1. Cancer Res. 2007 May 1;67(9):4271-7 lines. Oncogene. 1997 Oct;15(18):2159-68 Cooley MA, Kern CB, Fresco VM, Wessels A, Thompson RP, Pan TC, Kostka G, Zhang RZ, Timpl R, Chu ML. Complete McQuinn TC, Twal WO, Mjaatvedt CH, Drake CJ, Argraves exon-intron organization of the mouse fibulin-1 gene and its WS. Fibulin-1 is required for morphogenesis of neural crest- comparison with the human fibulin-1 gene. FEBS Lett. 1999 derived structures. Dev Biol. 2008 Jul 15;319(2):336-45 Feb 5;444(1):38-42 Argraves WS, Tanaka A, Smith EP, Twal WO, Argraves KM, Perbal B, Martinerie C, Sainson R, Werner M, He B, Roizman Fan D, Haudenschild CC. Fibulin-1 and fibrinogen in human B. The C-terminal domain of the regulatory protein NOVH is atherosclerotic lesions. Histochem Cell Biol. 2009 sufficient to promote interaction with fibulin 1C: a clue for a role Nov;132(5):559-65 of NOVH in cell-adhesion signaling. Proc Natl Acad Sci U S A. 1999 Feb 2;96(3):869-74 Mohamed SA, Sievers HH, Hanke T, Richardt D, et al. Pathway analysis of differentially expressed genes in patients Ohsawa I, Takamura C, Kohsaka S. Fibulin-1 binds the amino- with acute aortic dissection. Biomark Insights. 2009 May terminal head of beta-amyloid precursor protein and modulates 6;4:81-90 its physiological function. J Neurochem. 2001 Mar;76(5):1411- 20 This article should be referenced as such: Twal WO, Czirok A, Hegedus B, Knaak C, Chintalapudi MR, Castagnoli L, Tagliabue E, Pupa SM. FBLN1 (fibulin 1). Atlas Okagawa H, Sugi Y, Argraves WS. Fibulin-1 suppression of Genet Cytogenet Oncol Haematol. 2011; 15(5):450-454.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 454 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

HUS1 (HUS1 checkpoint homolog (S. pombe)) Amrita Madabushi, Randall C Gunther, A-Lien Lu Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, 108 North Greene Street, Baltimore, Maryland 21201, USA (AM, RCG, ALL)

Published in Atlas Database: September 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/HUS1ID40899ch7p12.html DOI: 10.4267/2042/45024 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Protein HGNC (Hugo): HUS1 Description Location: 7p12.3 Amino acids: 280. Molecular weight: 31.7 kDa. Hus1 Note: NCBI accession: NM_004507.2; NP_004498.1. is a component of the 9-1-1 cell cycle checkpoint complex that plays a critical role in sensing DNA DNA/RNA damage and maintaining genomic stability. Description Expression Found in all tissues. 15464 bp; 8 exons. Transcription Localisation Nucleus and cytoplasm. In discrete nuclear foci upon The transcribed mRNA has 2143 bp and the coding DNA damage. region is 840 bp, encodes a 280 amino acids, 31691 Da protein.

Pseudogene None.

Figure 1. HUS1 gene adapted from NCBI database Homo sapiens chromosome 7, GrCh37 primary reference assembly with kilobases from the telomere of p-arm on bottom. The exons 1-8 are represented by boxes with transcribed and untranscribed sequences in pink and orange, respectively. The exon numbers are labeled on top. The grey arrowhead symbolizes the direction of transcription and the arrows show the ATG and the stop codons, respectively.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 455 HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

Function (MYH) has a strong preference to bind Hus1 (Chang and Lu, 2005; Shi et al., 2006). Hus1 along with and Rad1 forms the 9-1-1 complex Recent structural and functional analyses indicate that (Hang and Lieberman, 2000; St Onge et al., 1999; the Hus1 binding region of MYH adopts a stabilized Volkmer and Karnitz, 1999). Hus1 and Rad1 can also conformation projecting away from the catalytic form a stable 1:1 complex (Doré et al., 2009; Sohn and domain to form a docking scaffold for Hus1 and binds Cho, 2009; Xu et al., 2009). The 9-1-complex is loaded to Hus1 through electrostatic interaction (Luncsford et onto the DNA by a specific clamp loader (Rad17- al., 2010). RFC2-RFC3-RFC4-RFC5) in response to many The 9-1-1 complex is required to activate two different genotoxic stresses (alkylation, oxidation, checkpoint sensors-ATM (ataxia telangiectasia [AT] ultraviolet light radiation and ionizing radiation), and mutated protein) and ATR (ATM- and Rad3-related replication inhibitors (Bermudez et al., 2003; Ellison protein), which are phosphoinositol phosphate 3 and Stillman, 2003). The Rad1-Rad9 interface may be kinase-related kinases (PIKKs) (Zhou and Elledge, opened to encircle DNA (Doré et al., 2009; Sohn and 2000). The DNA-bound 9-1-1 complex facilitates Cho, 2009; Xu et al., 2009). ATM- or ATR-mediated phosphorylation of more than 700 proteins including Chk1, Chk2, p53, and BRCA1 (Zhou and Elledge, 2000). Hus1 facilitated phosphorylation of Chk1 kinase is required for the ATR-dependent checkpoint; and regulates S-phase progression, G2/M arrest, and replication fork stabilization (Sancar et al., 2004; Sancar et al., 2004). However, Hus1 is not required for Chk2 phosphorylation in response to certain genotoxins (Weiss et al., 2003). Besides acting as a DNA damage sensor, the 9-1-1 complex plays an integral role in several DNA repair pathways including base excision repair (BER), mismatch repair (MMR), and nucleotide excision repair (NER) (see figure 2) (Helt et al., 2005). In the BER pathway, the 9-1-1 complex facilitates and interacts with several DNA glycosylases including MYH (Chang and Lu, 2005; Shi et al., 2006; Chang and Lu, 2005; Shi et al., 2006), 8-oxoG glycosylase (OGG1) (Park et al., 2009), NEIL1 (Guan et al., 2007a), and thymine DNA glycosylase (TDG) (Guan et Figure 2. hHus1 and hRad1 associate with each other prior to al., 2007b). The 9-1-1 complex also interacts with and forming the 9-1-1 complex with Rad9. In response to DNA stimulates other BER enzymes including APE1 damage and replication block, the 9-1-1 complex is loaded into (Gembka et al., 2007), POLbeta (Toueille et al., 2004), DNA by Rad17/RFC clamp loader and acts as checkpoint FEN1 (Friedrich-Heineken et al., 2005; Wang et al., sensor to activate ATM (ataxia telangiectasia [AT] mutated protein) or ATR (ATM- and Rad3-related protein) protein 2004a), RPA (Wu et al., 2005), and DNA ligase 1 kinase. The 9-1-1 complex interacts with and stimulates many (Smirnova et al., 2005; Wang et al., 2006a). Thus, the DNA repair enzymes involved in base excision repair (BER), 9-1-1 complex may provide a platform for the assembly nucleotide excision repair (NER), and mismatch repair (MMR). and function of the BER machinery (Balakrishnan et The damage recognition repair enzymes may serve as adaptors to signal DNA damage responses including enhanced DNA al., 2009; Lu et al., 2006). The 9-1-1 complex enhances repair, cell cycle arrest, and apoptosis. mismatch repair via direct interaction with mismatch The structure of the 9-1-1 complex (Doré et al., 2009; recognition proteins (MSH2/MSH3, MSH2/MSH6, and Sohn and Cho, 2009; Xu et al., 2009) is similar to the MLH1/PMS2) (Bai et al., 2010; He et al., 2008). Hus1 sliding clamp proliferating cell nuclear antigen protein interacts with MSH2/MSH3 and MSH2/MSH6, but not (PCNA) (Gulbis et al., 1996; Krishna et al., 1994). with MLH1/PMS2 (Bai et al., 2010; He et al., 2008). Hus1 interacts with Rad9 and Rad1 through its N In the NER pathway, interactions between terminal domain and C terminal domain, respectively. Saccharomyces cerevisiae Rad14 (hXPA homolog) and The structure and surface charge distribution of the the checkpoint proteins ScDdc1 (hRad9 homolog) and interdomain connecting loop (IDCL) of Hus1 differs ScMec3 (hHus1 homolog) have been demonstrated from those of other two subunits (Doré et al., 2009; (Giannattasio et al., 2004). Inactivation of NER by Sohn and Cho, 2009; Xu et al., 2009). The IDCL of knock down of XPA and XPC resulted in a decrease of Hus1 contains an N-terminal alpha helix and positive G1 phase cells that displayed Rad9 foci in response to charge cluster. These differences among Hus1, Rad9, UV light (Warmerdam et al., 2009). UV light-induced and Rad1 may contribute to different binding affinities Rad9 foci also colocalized with TopBP1 and gamma- to their partner proteins. For example, MutY homolog H2AX (Warmerdam et al., 2009).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 456 HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

Hus1 interacts with histone deacetylase HDAC1 (Cai et cells and impaired tissue regeneration (Yazinski et al., al., 2000). A novel pathway has been proposed that 2009). HDAC1 is involved in G(2)/M checkpoint control Oncogenesis through the interaction with the 9-1-1 complex. Single nucleotide polymorphism (SNP) analysis Jab1 physically associates with the 9-1-1 complex, supports the potential role of Hus1 in sporadic breast causes the translocation of the 9-1-1 complex from the cancer (Vega et al., 2009). SNPs in hHUS1 at nucleus to the cytoplasm, and mediates the rapid chromosome 7 base pair positions 47778020 (C) and degradation of the 9-1-1 complex (Huang et al., 2007). 47789957 (G) are statistically associated with breast Homology cancer development (Vega et al., 2009). hHus1 homologues from many eukaryotes are highly Ovarian tumors conserved. The following diagram shows the sequence Oncogenesis alignment of human Hus1 with Hus1 of fission yeast Hus1 expression levels correlate significantly with the Schizosaccharomyces pombe. The N-terminal domain clinicopathologic factors of bad prognosis of ovarian of Hus1 is structurally similar to the C-terminal tumors (de la Torre et al., 2008). domain. The structure of Hus1 (Doré et al., 2009; Sohn and Cho, 2009; Xu et al., 2009) is similar to those of Cancer therapy Rad9, Rad1, and PCNA (Gulbis et al., 1996; Krishna et Note al., 1994). The status of Hus1 can influence response to cancer therapy. Down-regulation of hHus1 by antisense RNA Mutations enhances the sensitivity of human lung carcinoma cells to cisplatin (a DNA cross-linker), presumably by Note enhancing apoptosis (Kinzel et al., 2002). Complete inactivation of the mouse Hus1 results in chromosomal instability, genotoxin hypersensitivity, Genomic stability and embryonic lethality. Note Inactivation of mouse or human Hus1 results in Implicated in impaired DNA damage signaling and severe spontaneous chromosomal instability (Weiss et al., Breast cancer 2002; Weiss et al., 2000; Kinzel et al., 2002). Note Drug sensitivity Hus1 inactivation in the mammary epithelium resulted in genome damage that induced apoptosis and led to Note depletion of Hus1-null cells from the mammary gland. Cells lacking Hus1 are hypersensitive to certain Dual inactivation of Hus1 and p53 in the mouse genotoxins including camptothecin (CPT), hydroxyurea mammary gland results in accumulation of damaged (HU), ultraviolet (UV), and ionizing radiation (IR) (Weiss et al., 2000; Wang et al., 2004b; Wang et al., 2006b).

Figure 3. Sequence alignment of the human Hus1 (hHus1) and S. pombe (SpHus1) with secondary structure motifs shown corresponding to the hHus1 based on hHus1 crystal structure (Xu et al., 2009). The arrows and coils represent the beta-sheets and alpha-helix, respectively. Hus1 contains 18 beta sheets and 5 alpha helices. Its N terminal and C terminal domains are connected by an interdomain connecting loop (IDCL, residues 134-155). The yellow shaded regions are the identical residues between hHus1 and SpHus1.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 457 HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

Loss of Hus1 sensitizes the cells to etoposide-induced Weiss RS, Matsuoka S, Elledge SJ, Leder P. Hus1 acts apoptosis by inducing Bim and Puma expressions and upstream of chk1 in a mammalian DNA damage response pathway. Curr Biol. 2002 Jan 8;12(1):73-7 releasing Rad9 into the cytosol (Levitt et al., 2005; Levitt et al., 2007; Meyerkord et al., 2008). The role of Bermudez VP, Lindsey-Boltz LA, Cesare AJ, Maniwa Y, Griffith JD, Hurwitz J, Sancar A. Loading of the human 9-1-1 Hus1 affecting the sensitivity of cells to IR-induced checkpoint complex onto DNA by the checkpoint clamp loader killing is independent of nonhomologous end-joining hRad17-replication factor C complex in vitro. Proc Natl Acad (NHEJ) but might be linked to homologous Sci U S A. 2003 Feb 18;100(4):1633-8 recombination repair (HRR) (Wang et al., 2006b). Ellison V, Stillman B. Biochemical characterization of DNA Development damage checkpoint complexes: clamp loader and clamp complexes with specificity for 5' recessed DNA. PLoS Biol. Note 2003 Nov;1(2):E33 Targeted deletion of mouse Hus1 results in embryonic Weiss RS, Leder P, Vaziri C. Critical role for mouse Hus1 in an lethality (Weiss et al., 2000). S-phase DNA damage cell cycle checkpoint. Mol Cell Biol. 2003 Feb;23(3):791-803 Telomere maintenance Giannattasio M, Lazzaro F, Longhese MP, Plevani P, Muzi- Note Falconi M. Physical and functional interactions between Severe telomere shortening has been observed in both nucleotide excision repair and DNA damage checkpoint. Hus1-deficient mouse embryonic fibroblasts and EMBO J. 2004 Jan 28;23(2):429-38 thymocytes from conditional Hus1-knockout mice Sancar A, Lindsey-Boltz LA, Unsal-Kaçmaz K, Linn S. (Francia et al., 2006). Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem. 2004;73:39-85 HIV infection (AIDS) Toueille M, El-Andaloussi N, Frouin I, Freire R, Funk D, Note Shevelev I, Friedrich-Heineken E, Villani G, Hottiger MO, Hus1 is required for Human Immunodeficiency Virus Hübscher U. The human Rad9/Rad1/Hus1 damage sensor clamp interacts with DNA polymerase beta and increases its type 1 Vpr-mediated G2 cell cycle arrest (Zimmerman DNA substrate utilisation efficiency: implications for DNA et al., 2004). repair. Nucleic Acids Res. 2004;32(11):3316-24 Wang W, Brandt P, Rossi ML, Lindsey-Boltz L, Podust V, References Fanning E, Sancar A, Bambara RA. The human Rad9-Rad1- Hus1 checkpoint complex stimulates flap endonuclease 1. Krishna TS, Kong XP, Gary S, Burgers PM, Kuriyan J. Crystal Proc Natl Acad Sci U S A. 2004 Nov 30;101(48):16762-7 structure of the eukaryotic DNA polymerase processivity factor PCNA. Cell. 1994 Dec 30;79(7):1233-43 Wang X, Guan J, Hu B, Weiss RS, Iliakis G, Wang Y. Involvement of Hus1 in the chain elongation step of DNA Gulbis JM, Kelman Z, Hurwitz J, O'Donnell M, Kuriyan J. replication after exposure to camptothecin or ionizing radiation. Structure of the C-terminal region of p21(WAF1/CIP1) Nucleic Acids Res. 2004;32(2):767-75 complexed with human PCNA. Cell. 1996 Oct 18;87(2):297- 306 Zimmerman ES, Chen J, Andersen JL, Ardon O, Dehart JL, Blackett J, Choudhary SK, Camerini D, Nghiem P, Planelles V. St Onge RP, Udell CM, Casselman R, Davey S. The human Human immunodeficiency virus type 1 Vpr-mediated G2 arrest G2 checkpoint control protein hRAD9 is a nuclear requires Rad17 and Hus1 and induces nuclear BRCA1 and phosphoprotein that forms complexes with hRAD1 and hHUS1. gamma-H2AX focus formation. Mol Cell Biol. 2004 Mol Biol Cell. 1999 Jun;10(6):1985-95 Nov;24(21):9286-94 Volkmer E, Karnitz LM. Human homologs of Chang DY, Lu AL. Interaction of checkpoint proteins Schizosaccharomyces pombe rad1, hus1, and rad9 form a Hus1/Rad1/Rad9 with DNA base excision repair enzyme MutY DNA damage-responsive protein complex. J Biol Chem. 1999 homolog in fission yeast, Schizosaccharomyces pombe. J Biol Jan 8;274(2):567-70 Chem. 2005 Jan 7;280(1):408-17 Cai RL, Yan-Neale Y, Cueto MA, Xu H, Cohen D. HDAC1, a Friedrich-Heineken E, Toueille M, Tännler B, Bürki C, Ferrari histone deacetylase, forms a complex with Hus1 and Rad9, E, Hottiger MO, Hübscher U. The two DNA clamps two G2/M checkpoint Rad proteins. J Biol Chem. 2000 Sep Rad9/Rad1/Hus1 complex and proliferating cell nuclear 8;275(36):27909-16 antigen differentially regulate flap endonuclease 1 activity. J Hang H, Lieberman HB. Physical interactions among human Mol Biol. 2005 Nov 11;353(5):980-9 checkpoint control proteins HUS1p, RAD1p, and RAD9p, and Helt CE, Wang W, Keng PC, Bambara RA. Evidence that DNA implications for the regulation of cell cycle progression. damage detection machinery participates in DNA repair. Cell Genomics. 2000 Apr 1;65(1):24-33 Cycle. 2005 Apr;4(4):529-32 Weiss RS, Enoch T, Leder P. Inactivation of mouse Hus1 Levitt PS, Liu H, Manning C, Weiss RS. Conditional results in genomic instability and impaired responses to inactivation of the mouse Hus1 cell cycle checkpoint gene. genotoxic stress. Genes Dev. 2000 Aug 1;14(15):1886-98 Genomics. 2005 Aug;86(2):212-24 Zhou BB, Elledge SJ. The DNA damage response: putting Smirnova E, Toueille M, Markkanen E, Hübscher U. The checkpoints in perspective. Nature. 2000 Nov human checkpoint sensor and alternative DNA clamp Rad9- 23;408(6811):433-9 Rad1-Hus1 modulates the activity of DNA ligase I, a Kinzel B, Hall J, Natt F, Weiler J, Cohen D. Downregulation of component of the long-patch base excision repair machinery. Hus1 by antisense oligonucleotides enhances the sensitivity of Biochem J. 2005 Jul 1;389(Pt 1):13-7 human lung carcinoma cells to cisplatin. Cancer. 2002 Mar 15;94(6):1808-14

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 458 HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

Wu X, Shell SM, Zou Y. Interaction and colocalization of Meyerkord CL, Takahashi Y, Araya R, Takada N, Weiss RS, Rad9/Rad1/Hus1 checkpoint complex with replication protein A Wang HG. Loss of Hus1 sensitizes cells to etoposide-induced in human cells. Oncogene. 2005 Jul 7;24(29):4728-35 apoptosis by regulating BH3-only proteins. Oncogene. 2008 Dec 11;27(58):7248-59 Francia S, Weiss RS, Hande MP, Freire R, d'Adda di Fagagna F. Telomere and telomerase modulation by the mammalian Balakrishnan L, Brandt PD, Lindsey-Boltz LA, Sancar A, Rad9/Rad1/Hus1 DNA-damage-checkpoint complex. Curr Biol. Bambara RA. Long patch base excision repair proceeds via 2006 Aug 8;16(15):1551-8 coordinated stimulation of the multienzyme DNA repair complex. J Biol Chem. 2009 May 29;284(22):15158-72 Lu AL, Bai H, Shi G, Chang DY. MutY and MutY homologs (MYH) in genome maintenance. Front Biosci. 2006 Sep Doré AS, Kilkenny ML, Rzechorzek NJ, Pearl LH. Crystal 1;11:3062-80 structure of the rad9-rad1-hus1 DNA damage checkpoint complex--implications for clamp loading and regulation. Mol Shi G, Chang DY, Cheng CC, Guan X, Venclovas C, Lu AL. Cell. 2009 Jun 26;34(6):735-45 Physical and functional interactions between MutY glycosylase homologue (MYH) and checkpoint proteins Rad9-Rad1-Hus1. Park MJ, Park JH, Hahm SH, Ko SI, Lee YR, Chung JH, Sohn Biochem J. 2006 Nov 15;400(1):53-62 SY, Cho Y, Kang LW, Han YS. Repair activities of human 8- oxoguanine DNA glycosylase are stimulated by the interaction Wang W, Lindsey-Boltz LA, Sancar A, Bambara RA. with human checkpoint sensor Rad9-Rad1-Hus1 complex. Mechanism of stimulation of human DNA ligase I by the Rad9- DNA Repair (Amst). 2009 Oct 2;8(10):1190-200 rad1-Hus1 checkpoint complex. J Biol Chem. 2006 Jul 28;281(30):20865-72 Sohn SY, Cho Y. Crystal structure of the human rad9-hus1- rad1 clamp. J Mol Biol. 2009 Jul 17;390(3):490-502 Wang X, Hu B, Weiss RS, Wang Y. The effect of Hus1 on ionizing radiation sensitivity is associated with homologous Vega A, Salas A, Milne RL, Carracedo B, Ribas G, Ruibal A, recombination repair but is independent of nonhomologous de León AC, González-Hernández A, Benítez J, Carracedo A. end-joining. Oncogene. 2006 Mar 23;25(13):1980-3 Evaluating new candidate SNPs as low penetrance risk factors in sporadic breast cancer: a two-stage Spanish case-control Gembka A, Toueille M, Smirnova E, Poltz R, Ferrari E, Villani study. Gynecol Oncol. 2009 Jan;112(1):210-4 G, Hübscher U. The checkpoint clamp, Rad9-Rad1-Hus1 complex, preferentially stimulates the activity of Warmerdam DO, Freire R, Kanaar R, Smits VA. Cell cycle- apurinic/apyrimidinic endonuclease 1 and DNA polymerase dependent processing of DNA lesions controls localization of beta in long patch base excision repair. Nucleic Acids Res. Rad9 to sites of genotoxic stress. Cell Cycle. 2009 Jun 2007;35(8):2596-608 1;8(11):1765-74 Guan X, Bai H, Shi G, Theriot CA, Hazra TK, Mitra S, Lu AL. Xu M, Bai L, Gong Y, Xie W, Hang H, Jiang T. Structure and The human checkpoint sensor Rad9-Rad1-Hus1 interacts with functional implications of the human rad9-hus1-rad1 cell cycle and stimulates NEIL1 glycosylase. Nucleic Acids Res. checkpoint complex. J Biol Chem. 2009 Jul 31;284(31):20457- 2007;35(8):2463-72 61 Guan X, Madabushi A, Chang DY, Fitzgerald ME, Shi G, Yazinski SA, Westcott PM, Ong K, Pinkas J, Peters RM, Weiss Drohat AC, Lu AL. The human checkpoint sensor Rad9-Rad1- RS. Dual inactivation of Hus1 and p53 in the mouse mammary Hus1 interacts with and stimulates DNA repair enzyme TDG gland results in accumulation of damaged cells and impaired glycosylase. Nucleic Acids Res. 2007;35(18):6207-18 tissue regeneration. Proc Natl Acad Sci U S A. 2009 Dec 15;106(50):21282-7 Huang J, Yuan H, Lu C, Liu X, Cao X, Wan M. Jab1 mediates protein degradation of the Rad9-Rad1-Hus1 checkpoint Bai H, Madabushi A, Guan X, Lu AL. Interaction between complex. J Mol Biol. 2007 Aug 10;371(2):514-27 human mismatch repair recognition proteins and checkpoint sensor Rad9-Rad1-Hus1. DNA Repair (Amst). 2010 May Levitt PS, Zhu M, Cassano A, Yazinski SA, Liu H, Darfler J, 4;9(5):478-87 Peters RM, Weiss RS. Genome maintenance defects in cultured cells and mice following partial inactivation of the Luncsford PJ, Chang DY, Shi G, Bernstein J, Madabushi A, essential cell cycle checkpoint gene Hus1. Mol Cell Biol. 2007 Patterson DN, Lu AL, Toth EA. A structural hinge in eukaryotic Mar;27(6):2189-201 MutY homologues mediates catalytic activity and Rad9-Rad1- Hus1 checkpoint complex interactions. J Mol Biol. 2010 Oct de la Torre J, Gil-Moreno A, García A, Rojo F, Xercavins J, 29;403(3):351-70 Salido E, Freire R. Expression of DNA damage checkpoint protein Hus1 in epithelial ovarian tumors correlates with This article should be referenced as such: prognostic markers. Int J Gynecol Pathol. 2008 Jan;27(1):24- 32 Madabushi A, Gunther RC, Lu AL. HUS1 (HUS1 checkpoint homolog (S. pombe)). Atlas Genet Cytogenet Oncol Haematol. He W, Zhao Y, Zhang C, An L, Hu Z, Liu Y, Han L, Bi L, Xie Z, 2011; 15(5):455-459. Xue P, Yang F, Hang H. Rad9 plays an important role in DNA mismatch repair through physical interaction with MLH1. Nucleic Acids Res. 2008 Nov;36(20):6406-17

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 459 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

OTX2 (orthodenticle homeobox 2) Matthew Wortham Department of Pathology, Duke University Medical Center, DUMC-3156, 199B-MSRB, Research Drive, Durham, NC 27710, USA (MW)

Published in Atlas Database: September 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/OTX2ID46429ch14q22.html DOI: 10.4267/2042/45025 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Protein Other names: MCOPS5, MGC45000 Note HGNC (Hugo): OTX2 Isoforms a and b share the same coding exons, therefore both isoforms encode full-length (289 amino Location: 14q22.3 acid) Otx2 protein. Local order: Chr14:57267427-57277184 (isoform a), Description Chr14:57267427-57277097 (isoform b). 289 amino acids, see diagram for domain organization. From plus strand: C14orf101, OTX2, EXOC5, Expression MUDENG. Rostral neural tube (mid-late gestation; Larsen et al., DNA/RNA 2010), hippocampus, cerebellar rhombic lip, choroid plexus (Larsen et al., 2010), retinal pigment epithelium Description (Glubrecht et al., 2009; Larsen et al., 2009). Characterized in rodents: epiblast (Fossat et al., 2006), Total gene sequence: 9757 bp. anterior neural ectoderm and anterior visceral Transcription endoderm (Fossat et al., 2006), external granular layer From minus strand. Isoform a: 5 exons, 4 introns; of the postnatal cerebellum (Frantz et al., 1994), isoform b: 3 exons, 2 introns. posterior lobes of the adult cerebellum (Fossat et al., Isoform a: Full-length unspliced transcript: 9757 bp; 2006). spliced transcript: 2209 bp; Localisation Isoform b: Full-length unspliced transcript: 9670 bp; Predominately nuclear but in some cell types can be spliced transcript: 2082 bp. retained in the cytoplasm (Baas et al., 2000) as well as Pseudogene transferred from cell to cell (Sugiyama et al., 2008). OTX2P1 located at 9q21.

Otx2 protein domains. Domains were defined based on sequence conservation and, when possible, functional analysis as described in Chau et al., 2000 and Chatelain et al., 2006. Conserved OTX family domain identified in the CDD database (Marchler-Bauer et al., 2009). Domain abbreviations and boundaries are as follows: HD: Paired-class homeobox domain, spans amino acids (aa) 37-97; NRS: nuclear retention signal, spans aa 117-146; grey box: WSP domain, spans aa 150-159; OTX: OTX family domain, spans aa 178-243; TA: transactivation domain, comprised of two separate transactivation motifs spanning aa 255-267 and aa 273-285; b: basic regions (aa 36- 42, aa 89-94, and aa 107-114); Q: polyglutamine repeat (aa 95-101).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 460 OTX2 (orthodenticle homeobox 2) Wortham M

Function activity is transcriptional activation of MYC (Adamson et al., 2010). Homeobox transcription factor, binds the DNA sequence TAATCC (Chatelain et al., 2006). OTX2 Retinoblastoma plays a critical role in anteroposterior patterning of the Cytogenetics embryo (Matsuo et al., 1995), anterior neuroectoderm Secondary events cooperating with loss of Rb gene formation (Acampora et al., 1995), neuronal function have remained elusive. However, genomewide differentiation in various CNS compartments (Vernay copy number analysis has revealed recurrent regions of et al., 2005; Omodei et al., 2008), and experience- gain or loss at the megabase resolution, and induced plasticity (Sugiyama et al., 2009). chromosome 14 aberrations have indeed been described Homology (Zielinski et al., 2005). Shares and general domain Oncogenesis organization with OTX family members Otx1 and Crx. Considering the restricted expression pattern of OTX2 mRNA in adult tissues (Boon et al., 2002) and the well- Mutations established oncogenic function of Otx2 in medulloblastomas (Adamson et al., 2010), expression Germinal of Otx2 in retinoblastoma may indicate a role for this Dominant-inherited OTX2 mutations exhibiting gene in retinoblastoma pathogenesis (Glubrecht et al., variable penetrance have been associated with 2009). Interestingly, Otx2 is expressed in the most developmental defects of the eye (Ragge et al., 2005; undifferentiated compartments of retinoblastomas Wyatt et al., 2008; see the "Implicated in" section (Glubrecht et al., 2009). Although, Otx2 is expressed below for further discussion) and pituitary (Diaczok et broadly among retinoblastoma samples, its potential al., 2008) as well as recurrent seizure disorders (Ragge role as an oncogene in this tumor type has not been et al., 2005). None associated with hereditary tumor experimentally assessed; the possibility that Otx2 is predisposition syndromes. solely a cell lineage marker maintained in transformed retinal progenitor cells has yet to be excluded based on Somatic functional studies. None detected in medulloblastoma. Coloboma Implicated in Note Developmental defects of the eye. Pediatric CNS cancer (medullo- Disease blastoma) Coloboma, defined as a fissure in the ocular tissue Prognosis (Onwochei et al., 2000). These result from incomplete 5-year survival rates average 50-60%; predictors of closure of the fetal fissure (an invagination of the optic poor outcome include young age (younger than 3 years stalk and optic vesicle), whose function is to provide a old) and presence of metastases. OTX2 copy number scaffold for the formation of the optic cup and for the gain has been associated with shorter survival vessels responsible for retinal vascularization. (Adamson et al., 2010). Colobomata are predominately developmental defects that present at birth. Various genes, including OTX2, Cytogenetics have been implicated in hereditary syndromes Various broad and focal copy number changes have predisposing to coloboma (Omwochei et al., 2000; been identified in medulloblastoma (reviewed in: Wyatt et al., 2008), and sporadic cases have implicated Northcott et al., 2010), whereas OTX2 is the most teratogens, though evidence implicating particular common target of focal copy number gain in the agents is generally anecdotal (Omwochei et al., 2000). medulloblastoma genome (Adamson et al., 2010). Cytogenetics Oncogenesis Germline OTX2 mutations have been identified in Otx2 is overexpressed in the majority (~74%) of patients with bilateral eye defects including colobomata medulloblastomas (Adamson et al., 2010). A subset of and anophthalmia (Wyatt et al., 2008). these tumors (~21%) harbor copy number gains of the OTX2 genomic locus; the mechanism of Otx2 Anophthalmia and microphthalmia overexpression in the remaining tumors remains (absent or small eyes, respectively) unidentified. Otx2 is distinctly overexpressed in Shh- Note independent medulloblastomas (i.e. tumor subtypes not Developmental defects of the eye. expressing gene signatures of Shh pathway activation; Adamson et al., 2010). Otx2 has been implicated in Disease medulloblastoma tumor progression and is required for Microphthalmia is clinically defined as an eye with an tumor maintenance. One mechanism of Otx2 oncogenic axial diameter measuring at least two standard

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 461 OTX2 (orthodenticle homeobox 2) Wortham M

deviations below the mean for the corresponding age comparative genomic hybridization. Genes Chromosomes group (Omwochei et al., 2000), whereas anophthalmia Cancer. 2005 Jul;43(3):294-301 is diagnosed when no clinically apparent eye structure Chatelain G, Fossat N, Brun G, Lamonerie T. Molecular is present. Those affected generally harbor bilateral dissection reveals decreased activity and not dominant negative effect in human OTX2 mutants. J Mol Med. 2006 malformations. Like coloboma, some forms of Jul;84(7):604-15 anophthalmia/microphthalmia are clearly inheritable, while for other cases environmental factors have been Fossat N, Chatelain G, Brun G, Lamonerie T. Temporal and spatial delineation of mouse Otx2 functions by conditional self- implicated but not definitively so (Verma et al., 2007). knockout. EMBO Rep. 2006 Aug;7(8):824-30 Anophthalmia/microphthalmia can present as secondary malformations following colobomata. Verma AS, Fitzpatrick DR. Anophthalmia and microphthalmia. Orphanet J Rare Dis. 2007 Nov 26;2:47 Cytogenetics Diaczok D, Romero C, Zunich J, Marshall I, Radovick S. A Various genes have been implicated, including SOX2 novel dominant negative mutation of OTX2 associated with (autosomal dominant inheritance), OTX2 (autosomal combined pituitary hormone deficiency. J Clin Endocrinol dominant), CHX10 (autosomal recessive), and RAX Metab. 2008 Nov;93(11):4351-9 (autosomal recessive; Verma et al., 2007; Wyatt et al., Omodei D, Acampora D, Mancuso P, Prakash N, Di 2008). Giovannantonio LG, Wurst W, Simeone A. Anterior-posterior graded response to Otx2 controls proliferation and differentiation of dopaminergic progenitors in the ventral References mesencephalon. Development. 2008 Oct;135(20):3459-70 Frantz GD, Weimann JM, Levin ME, McConnell SK. Otx1 and Sugiyama S, Di Nardo AA, Aizawa S, Matsuo I, Volovitch M, Otx2 define layers and regions in developing cerebral cortex Prochiantz A, Hensch TK. Experience-dependent transfer of and cerebellum. J Neurosci. 1994 Oct;14(10):5725-40 Otx2 homeoprotein into the visual cortex activates postnatal plasticity. Cell. 2008 Aug 8;134(3):508-20 Acampora D, Mazan S, Lallemand Y, Avantaggiato V, Maury M, Simeone A, Brûlet P. Forebrain and midbrain regions are Wyatt A, Bakrania P, Bunyan DJ, Osborne RJ, Crolla JA, et al. deleted in Otx2-/- mutants due to a defective anterior Novel heterozygous OTX2 mutations and whole gene deletions neuroectoderm specification during gastrulation. Development. in anophthalmia, microphthalmia and coloboma. Hum Mutat. 1995 Oct;121(10):3279-90 2008 Nov;29(11):E278-83 Matsuo I, Kuratani S, Kimura C, Takeda N, Aizawa S. Mouse Glubrecht DD, Kim JH, Russell L, Bamforth JS, Godbout R. Otx2 functions in the formation and patterning of rostral head. Differential CRX and OTX2 expression in human retina and Genes Dev. 1995 Nov 1;9(21):2646-58 retinoblastoma. J Neurochem. 2009 Oct;111(1):250-63 Baas D, Bumsted KM, Martinez JA, Vaccarino FM, Wikler KC, Larsen KB, Lutterodt M, Rath MF, Møller M. Expression of the Barnstable CJ. The subcellular localization of Otx2 is cell-type homeobox genes PAX6, OTX2, and OTX1 in the early human specific and developmentally regulated in the mouse retina. fetal retina. Int J Dev Neurosci. 2009 Aug;27(5):485-92 Brain Res Mol Brain Res. 2000 May 31;78(1-2):26-37 Marchler-Bauer A, Anderson JB, Chitsaz F, Derbyshire MK, et Chau KY, Chen S, Zack DJ, Ono SJ. Functional domains of al. CDD: specific functional annotation with the Conserved the cone-rod homeobox (CRX) transcription factor. J Biol Domain Database. Nucleic Acids Res. 2009 Jan;37(Database Chem. 2000 Nov 24;275(47):37264-70 issue):D205-10 Onwochei BC, Simon JW, Bateman JB, Couture KC, Mir E. Sugiyama S, Prochiantz A, Hensch TK. From brain formation Ocular colobomata. Surv Ophthalmol. 2000 Nov- to plasticity: insights on Otx2 homeoprotein. Dev Growth Differ. Dec;45(3):175-94 2009 Apr;51(3):369-77 Boon K, Osorio EC, Greenhut SF, Schaefer CF, Shoemaker J, Adamson DC, Shi Q, Wortham M, Northcott PA, Di C, Duncan Polyak K, Morin PJ, Buetow KH, Strausberg RL, De Souza SJ, CG, Li J, McLendon RE, Bigner DD, Taylor MD, Yan H. OTX2 Riggins GJ. An anatomy of normal and malignant gene is critical for the maintenance and progression of Shh- expression. Proc Natl Acad Sci U S A. 2002 Aug independent medulloblastomas. Cancer Res. 2010 Jan 20;99(17):11287-92 1;70(1):181-91 Ragge NK, Brown AG, Poloschek CM, Lorenz B, et al. Larsen KB, Lutterodt MC, Møllgård K, Møller M. Expression of Heterozygous mutations of OTX2 cause severe ocular the homeobox genes OTX2 and OTX1 in the early developing malformations. Am J Hum Genet. 2005 Jun;76(6):1008-22 human brain. J Histochem Cytochem. 2010 Jul;58(7):669-78 Vernay B, Koch M, Vaccarino F, Briscoe J, Simeone A, Northcott PA, Rutka JT, Taylor MD. Genomics of Kageyama R, Ang SL. Otx2 regulates subtype specification medulloblastoma: from Giemsa-banding to next-generation and neurogenesis in the midbrain. J Neurosci. 2005 May sequencing in 20 years. Neurosurg Focus. 2010 Jan;28(1):E6 11;25(19):4856-67 This article should be referenced as such: Zielinski B, Gratias S, Toedt G, Mendrzyk F, Stange DE, Radlwimmer B, Lohmann DR, Lichter P. Detection of Wortham M. OTX2 (orthodenticle homeobox 2). Atlas Genet chromosomal imbalances in retinoblastoma by matrix-based Cytogenet Oncol Haematol. 2011; 15(5):460-462.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 462 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Leukaemia Section Short Communication dic(3;9)(p14;p13) PAX5/FOXP1 Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/dic0309p14p13ID1553.html DOI: 10.4267/2042/45026 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Entry of common lymphoid progenitors into the B cell Identity lineage depends on E2A, EBF1, and PAX5; activates Note B-cell specific genes and repress genes involved in See also the paper on dic(9;20)(p11-13;q11). other lineage commitments. Activates the surface cell receptor CD19 and repress FLT3. Pax5 physically Clinics and pathology interacts with the RAG1/RAG2 complex, and removes the inhibitory signal of the lysine-9-methylated histone Disease H3, and induces V-to-DJ rearrangements. Genes Acute lymphoblastic leukaemia (ALL). repressed by PAX5 expression in early B cells are restored in their function in mature B cells and plasma Phenotype/cell stem origin cells, and PAX5 repressed (Fuxa et al., 2004; Johnson B-cell precursor ALL. et al., 2004; Zhang et al., 2006; Cobaleda et al., 2007). Epidemiology FOXP1 One case to date (Mullighan et al., 2007). Location Prognosis 3p14 No data. Protein Transcriptional repressor. Involved in cardiomyocyte Genes involved and proteins proliferation, motor neuron migration, B-lymphocyte development, and the generation of quiescent naive T PAX5 cells (Shi et al., 2008; Feng et al., 2010; Rao et al., 2010; Zhang et al., 2010; Hisaoka et al., 2010). Location 9p13.2 Result of the chromosomal Protein Lineage-specific transcription factor; recognizes the anomaly concensus recognition sequence GNCCANTGAAGCGTGAC, where N is any Hybrid gene nucleotide. Involved in B-cell differentiation. Description Fusion of PAX5 exon 6 to FOXP1 exon 7.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 463 dic(3;9)(p14;p13) PAX5/FOXP1 Huret JL

Fusion protein

Description Shi C, Sakuma M, Mooroka T, Liscoe A, Gao H, Croce KJ, Sharma A, Kaplan D, Greaves DR, Wang Y, Simon DI. Down- The predicted fusion protein contains the DNA binding regulation of the forkhead transcription factor Foxp1 is required paired domain of PAX5 and the DNA-binding and for monocyte differentiation and macrophage function. Blood. transcriptional regulator domain of FOXP1. 877 amino 2008 Dec 1;112(12):4699-711 acids. Feng X, Ippolito GC, Tian L, Wiehagen K, Oh S, Sambandam A, Willen J, Bunte RM, Maika SD, Harriss JV, Caton AJ, References Bhandoola A, Tucker PW, Hu H. Foxp1 is an essential transcriptional regulator for the generation of quiescent naive T Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, cells during thymocyte development. Blood. 2010 Jan Busslinger M. Pax5 induces V-to-DJ rearrangements and locus 21;115(3):510-8 contraction of the immunoglobulin heavy-chain gene. Genes Dev. 2004 Feb 15;18(4):411-22 Hisaoka T, Nakamura Y, Senba E, Morikawa Y. The forkhead transcription factors, Foxp1 and Foxp2, identify different Johnson K, Pflugh DL, Yu D, Hesslein DG, Lin KI, Bothwell AL, subpopulations of projection neurons in the mouse cerebral Thomas-Tikhonenko A, Schatz DG, Calame K. B cell-specific cortex. Neuroscience. 2010 Mar 17;166(2):551-63 loss of histone 3 lysine 9 methylation in the V(H) locus depends on Pax5. Nat Immunol. 2004 Aug;5(8):853-61 Rao DS, O'Connell RM, Chaudhuri AA, Garcia-Flores Y, Geiger TL, Baltimore D. MicroRNA-34a perturbs B lymphocyte Zhang Z, Espinoza CR, Yu Z, Stephan R, He T, Williams GS, development by repressing the forkhead box transcription Burrows PD, Hagman J, Feeney AJ, Cooper MD. Transcription factor Foxp1. Immunity. 2010 Jul 23;33(1):48-59 factor Pax5 (BSAP) transactivates the RAG-mediated V(H)-to- DJ(H) rearrangement of immunoglobulin genes. Nat Immunol. Zhang Y, Li S, Yuan L, Tian Y, Weidenfeld J, Yang J, Liu F, 2006 Jun;7(6):616-24 Chokas AL, Morrisey EE. Foxp1 coordinates cardiomyocyte proliferation through both cell-autonomous and Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: the nonautonomous mechanisms. Genes Dev. 2010 Aug guardian of B cell identity and function. Nat Immunol. 2007 15;24(16):1746-57 May;8(5):463-70 This article should be referenced as such: Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, Girtman K, Mathew S, Ma J, Pounds SB, Su X, Huret JL. dic(3;9)(p14;p13) PAX5/FOXP1. Atlas Genet Pui CH, Relling MV, Evans WE, Shurtleff SA, Downing JR. Cytogenet Oncol Haematol. 2011; 15(5):463-464. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature. 2007 Apr 12;446(7137):758- 64

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 464 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Leukaemia Section Short Communication dic(9;18)(p13;q11) PAX5/ZNF521 Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/dic0918p13q11ID1556.html DOI: 10.4267/2042/45027 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

lineage depends on E2A, EBF1, and PAX5; activates Identity B-cell specific genes and repress genes involved in Note other lineage commitments. Activates the surface cell See also the paper on dic(9;20)(p11-13;q11). receptor CD19 and repress FLT3. Pax5 physically interacts with the RAG1/RAG2 complex, and removes Clinics and pathology the inhibitory signal of the lysine-9-methylated histone H3, and induces V-to-DJ rearrangements. Genes Disease repressed by PAX5 expression in early B cells are Acute lymphoblastic leukaemia (ALL). restored in their function in mature B cells and plasma cells, and PAX5 repressed (Fuxa et al., 2004; Johnson Phenotype/cell stem origin et al., 2004; Zhang et al., 2006; Cobaleda et al., 2007). B-cell precursor ALL. ZNF521 Epidemiology Location One case to date (Mullighan et al., 2007). 18q11 Prognosis Protein No data. Transcription factor. Involved in B-cell lineage commitment, in the differentiation of neural Genes involved and proteins progenitors; Inhibits EBF1. Binds Runx2 and represses its transcriptional activity. Regulates osteoblast PAX5 differentiation and bone formation (Bond et al., 2008; Lobo et al., 2008; Wu et al., 2009; Hesse et al., 2010). Location 9p13.2 Result of the chromosomal Protein Lineage-specific transcription factor; recognizes the anomaly concensus recognition sequence GNCCANTGAAGCGTGAC, where N is any Hybrid gene nucleotide. Involved in B-cell differentiation. Entry of Description common lymphoid progenitors into the B cell Fusion of PAX5 exon 7 to ZNF521 exon 4.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 465 dic(9;18)(p13;q11) PAX5/ZNF521 Huret JL

Fusion protein

Description Pui CH, Relling MV, Evans WE, Shurtleff SA, Downing JR. Genome-wide analysis of genetic alterations in acute The predicted fusion protein contains the DNA binding lymphoblastic leukaemia. Nature. 2007 Apr 12;446(7137):758- paired domain of PAX5 and the DNA-binding and 64 transcriptional regulator domain of ZNF521. 1541 Bond HM, Mesuraca M, Amodio N, Mega T, Agosti V, Fanello amino acids. D, Pelaggi D, Bullinger L, Grieco M, Moore MA, Venuta S, Morrone G. Early hematopoietic zinc finger protein-zinc finger References protein 521: a candidate regulator of diverse immature cells. Int J Biochem Cell Biol. 2008;40(5):848-54 Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, Busslinger M. Pax5 induces V-to-DJ rearrangements and locus Lobo MK, Yeh C, Yang XW. Pivotal role of early B-cell factor 1 contraction of the immunoglobulin heavy-chain gene. Genes in development of striatonigral medium spiny neurons in the Dev. 2004 Feb 15;18(4):411-22 matrix compartment. J Neurosci Res. 2008 Aug 1;86(10):2134- 46 Johnson K, Pflugh DL, Yu D, Hesslein DG, Lin KI, Bothwell AL, Thomas-Tikhonenko A, Schatz DG, Calame K. B cell-specific Wu M, Hesse E, Morvan F, Zhang JP, Correa D, Rowe GC, loss of histone 3 lysine 9 methylation in the V(H) locus Kiviranta R, Neff L, Philbrick WM, Horne WC, Baron R. Zfp521 depends on Pax5. Nat Immunol. 2004 Aug;5(8):853-61 antagonizes Runx2, delays osteoblast differentiation in vitro, and promotes bone formation in vivo. Bone. 2009 Zhang Z, Espinoza CR, Yu Z, Stephan R, He T, Williams GS, Apr;44(4):528-36 Burrows PD, Hagman J, Feeney AJ, Cooper MD. Transcription factor Pax5 (BSAP) transactivates the RAG-mediated V(H)-to- Hesse E, Kiviranta R, Wu M, Saito H, Yamana K, Correa D, DJ(H) rearrangement of immunoglobulin genes. Nat Immunol. Atfi A, Baron R. Zinc finger protein 521, a new player in bone 2006 Jun;7(6):616-24 formation. Ann N Y Acad Sci. 2010 Mar;1192:32-7 Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: the This article should be referenced as such: guardian of B cell identity and function. Nat Immunol. 2007 May;8(5):463-70 Huret JL. dic(9;18)(p13;q11) PAX5/ZNF521. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):465-466. Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, Girtman K, Mathew S, Ma J, Pounds SB, Su X,

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 466 Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Case Report Section Paper co-edited with the European LeukemiaNet

Chronic lymphocytic leukaemia/Small lymphocytic lymphoma (CLL/SLL) associated with translocation t(1;6)(p35;p25) as part of complex karyotype Elvira D Rodrigues Pereira Velloso, Daniela Borri, Cristina Alonso Ratis, Guilherme Fleury Perin, Nelson Hamerschlak, Nydia S Bacal, Paulo A A Silveira, Alanna M P S Bezerra, Denise C Pasqualin Clinical Laboratory of Hospital Israelita Albert Einstein, Sao Paulo, Brazil (EDRPV, DB, CAR, NSB, PAAS); Pathology Department of Hospital Israelita Albert Einstein, Sao Paulo, Brazil (AMPSB, DCP); Hematology Department of Hospital Israelita Albert Einstein, Sao Paulo, Brazil (GFP, NH)

Published in Atlas Database: August 2010 Online updated version : http://AtlasGeneticsOncology.org/Reports/t0106VellosoID100047.html DOI: 10.4267/2042/45028 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Clinics Cyto-Pathology Age and sex Classification 64 years old male patient. Immunophenotype Previous history Small lymphocytic lymphoma/chronic lymphocytic No preleukemia, previous malignancy Chronic leukemia (SLL/CLL) lymphocytic leukaemia/Small lymphocytic lymphoma Rearranged Ig Tcr (CLL/SLL) diagnosed 19 months earlier, in first relapse after 4 cycles of RFC (Fludarabine, cyclophosphamide Not done and Rituximab), no inborn condition of note. Pathology Organomegaly Lymph node biopsy showed SLL/CLL, CD20, CD23, No hepatomegaly, no splenomegaly, enlarged lymph CD5, CD43 and BCL2 positive; CD10 and Cyclin D1 nodes (diffuse lymphadenopathy), no central nervous negative and Ki-67 positive in 25% of neoplastic cells. system involvement Electron microscopy Not done Blood Diagnosis WBC : 1.5 X 109/l Chronic lymphocytic leukaemia/Small lymphocytic HB : 10.3g/dl lymphoma (CLL/SLL) with high Ki-67 index. Platelets : 52 X 109/l Bone marrow : Aspirate and immunophenotype study: Survival 69.8% lymphoid cells, CD19+, CD5++, CD11c+, Date of diagnosis: 03-2010 CD23++, cyIgM+, cylambda+. Treatment: Bendamustine +Rituximab (2 cycles).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 467 Chronic lymphocytic leukaemia/Small lymphocytic lymphoma (CLL/SLL) Rodrigues Pereira Velloso ED, et al. associated with translocation t(1;6)(p35;p25) as part of complex karyotype

Complete remission : no Results: Treatment related death : + (using ISCN): 44,XY, t(1;6)(p35;p25), der(4)(q21), -9, add(17)(p13), -21, +mar[6]/46,XY[14] Status : Dead; Last follow up 07-2010. Survival: 4 months Karyotype at Relapse: not done Other molecular cytogenetics technics: Karyotype Fish using deletion probe XLP53 (MetaSystems) confirms 17p13/P53 locus deletion nuc Sample: bone marrow cells. ish(D17Z1x2,p53x1)[76/100] Culture time: 72 hours with TPA (o-tetradecanoyl phorbol-13-acetate). Other Molecular Studies Banding: G Technics: not done

Partial karyotypes, G- bands, showing the t(1;6)(p35;q21), del(4)(q21), and add(17)(p13).

Fish interphase study using probes D17Z1 (green) and p53 (orange) showing 2 green signals and 1 orange signal, confirming p53 deletion.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 468 Chronic lymphocytic leukaemia/Small lymphocytic lymphoma (CLL/SLL) Rodrigues Pereira Velloso ED, et al. associated with translocation t(1;6)(p35;p25) as part of complex karyotype

Ki-67 immunostaining in lymph node at time of t(1;6) detection.Comments

Until now, 16 patients with CLL and t(1;6)(p35;p25) Rodrigues Pereira Velloso ED, Ratis C, Brasil SAB, Guerra JC, were described. 8 of these patients were described by Bacal N, Pitangueira CPMLM.. Translocation t(1;6)(p35;p25) in B-cell lymphoproliferative disorder with evolution to Diffuse Michaux et al. in 2005, showing that this rearrangement Large B-cell Lymphoma. Atlas Genet Cytogenet Oncol was associated with bad prognosis: unmutated B-CLL Haematol. July 2007. URL : and evolution to diffuse large B-cell Lymphoma http://AtlasGeneticsOncology.org/Reports/0106RodriguesID10 (DLBCL). Our group also described in 2007 a case of 0030.html atypical CLL with evolution to aggressive B-cell Van Den Neste E, Robin V, Francart J, Hagemeijer A, Stul M, Lymphoma. In the case reported herein, clinical and Vandenberghe P, Delannoy A, Sonet A, Deneys V, Costantini S, Ferrant A, Robert A, Michaux L. Chromosomal pathological evolution was associated with the translocations independently predict treatment failure, detection of t(1;6) as part of complex karyotype treatment-free survival and overall survival in B-cell chronic including deletion of p53. Although no transformation lymphocytic leukemia patients treated with cladribine. to DLBCL was seen, increase in proliferation rate in Leukemia. 2007 Aug;21(8):1715-22 lymph node biopsy (Ki-67 increase from less than10 to This article should be referenced as such: 25%) was detected, associated with bad prognosis and short survival. Rodrigues Pereira Velloso ED, Borri D, Alonso Ratis C, Fleury Perin G, Hamerschlak N, Bacal NS, Silveira PAA, Bezerra AMPS, Pasqualin DC. Chronic lymphocytic leukaemia/Small References lymphocytic lymphoma (CLL/SLL) associated with translocation t(1;6)(p35;p25) as part of complex karyotype. Michaux L, Wlodarska I, Rack K, Stul M, Criel A, Maerevoet M, Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):467-469. Marichal S, Demuynck H, Mineur P, Kargar Samani K, Van Hoof A, Ferrant A, Marynen P, Hagemeijer A. Translocation t(1;6)(p35.3;p25.2): a new recurrent aberration in "unmutated" B-CLL. Leukemia. 2005 Jan;19(1):77-82

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 469 Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Instructions to Authors Manuscripts submitted to the Atlas must be submitted solely to the Atlas. Iconography is most welcome: there is no space restriction. The Atlas publishes "cards", "deep insights", "case reports", and "educational items". Cards are structured review articles. Detailed instructions for these structured reviews can be found at: http://AtlasGeneticsOncology.org/Forms/Gene_Form.html for reviews on genes, http://AtlasGeneticsOncology.org/Forms/Leukaemia_Form.html for reviews on leukaemias, http://AtlasGeneticsOncology.org/Forms/SolidTumour_Form.html for reviews on solid tumours, http://AtlasGeneticsOncology.org/Forms/CancerProne_Form.html for reviews on cancer-prone diseases. According to the length of the paper, cards are divided, into "reviews" (texts exceeding 2000 words), "mini reviews" (between), and "short communications" (texts below 400 words). The latter category may not be accepted for indexing by bibliographic databases. Deep Insights are written as traditional papers, made of paragraphs with headings, at the author's convenience. No length restriction. Case Reports in haematological malignancies are dedicated to recurrent -but rare- chromosomes abnormalities in leukaemias/lymphomas. Cases of interest shall be: 1- recurrent (i.e. the chromosome anomaly has already been described in at least 1 case), 2- rare (previously described in less than 20 cases), 3- with well documented clinics and laboratory findings, and 4- with iconography of chromosomes. It is mandatory to use the specific "Submission form for Case reports": see http://AtlasGeneticsOncology.org/Reports/Case_Report_Submission.html. Educational Items must be didactic, give full information and be accompanied with iconography. Translations into French, German, Italian, and Spanish are welcome.

Subscription: The Atlas is FREE!

Corporate patronage, sponsorship and advertising Enquiries should be addressed to [email protected].

Rules, Copyright Notice and Disclaimer Conflicts of Interest: Authors must state explicitly whether potential conflicts do or do not exist. Reviewers must disclose to editors any conflicts of interest that could bias their opinions of the manuscript. The editor and the editorial board members must disclose any potential conflict. Privacy and Confidentiality – Iconography: Patients have a right to privacy. Identifying details should be omitted. If complete anonymity is difficult to achieve, informed consent should be obtained. Property: As "cards" are to evolve with further improvements and updates from various contributors, the property of the cards belongs to the editor, and modifications will be made without authorization from the previous contributor (who may, nonetheless, be asked for refereeing); contributors are listed in an edit history manner. Authors keep the rights to use further the content of their papers published in the Atlas, provided that the source is cited. Copyright: The information in the Atlas of Genetics and Cytogenetics in Oncology and Haematology is issued for general distribution. All rights are reserved. The information presented is protected under international conventions and under national laws on copyright and neighbouring rights. Commercial use is totally forbidden. Information extracted from the Atlas may be reviewed, reproduced or translated for research or private study but not for sale or for use in conjunction with commercial purposes. Any use of information from the Atlas should be accompanied by an acknowledgment of the Atlas as the source, citing the uniform resource locator (URL) of the article and/or the article reference, according to the Vancouver convention. Reference to any specific commercial products, process, or service by trade name, trademark, manufacturer, or otherwise, does not necessarily constitute or imply its endorsement, recommendation, or favouring. The views and opinions of contributors and authors expressed herein do not necessarily state or reflect those of the Atlas editorial staff or of the web site holder, and shall not be used for advertising or product endorsement purposes. The Atlas does not make any warranty, express or implied, including the warranties of merchantability and fitness for a particular purpose, or assumes any legal liability or responsibility for the accuracy, completeness, or usefulness of any information, and shall not be liable whatsoever for any damages incurred as a result of its use. In particular, information presented in the Atlas is only for research purpose, and shall not be used for diagnosis or treatment purposes. No responsibility is assumed for any injury and/or damage to persons or property for any use or operation of any methods products, instructions or ideas contained in the material herein. See also: "Uniform Requirements for Manuscripts Submitted to Biomedical Journals: Writing and Editing for Biomedical Publication - Updated October 2004": http://www.icmje.org.

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

© ATLAS - ISSN 1768-3262

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 471