VolumeVolume 15 1 -- NumberNumber 51 May -May Sept 2011ember 1997

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

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

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)) 393 Marie-Joelle Mozziconacci, Daniel Birnbaum EWSR1 (Ewing sarcoma breakpoint region 1) 397 Jean-Loup Huret FAM57A (family with sequence similarity 57, member A) 410 Zhiao Chen, Xianghuo He FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) 412 Hideyuki Ihara, Cong-xiao Gao, Yoshitaka Ikeda, Naoyuki Taniguchi IGSF8 (immunoglobulin superfamily, member 8) 417 Yanhui H Zhang, Mekel M Richardson, Xin A Zhang MIXL1 (Mix1 homeobox-like 1 (Xenopus laevis)) 421 Aaron Raymond, Lalitha Nagarajan PEG3 (paternally expressed 3) 424 Yinhua Yu, Weiwei Feng, Zhen Lu, Robert C Bast Jr RPL10 (ribosomal L10) 427 Mohit Goel, Ranjan Tamuli SNAI1 (snail homolog 1 (Drosophila)) 430 Joerg Schwock, William R Geddie VAV3 (vav 3 guanine nucleotide exchange factor) 438 Leah Lyons, Kerry L Burnstein CAMTA1 (calmodulin binding transcription activator 1) 444 Kai-Oliver Henrich DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) 446 Maria L Arbonés, Susana de la Luna FBLN1 (fibulin 1) 453 Lorenzo Castagnoli, Elda Tagliabue, Serenella M Pupa HUS1 (HUS1 checkpoint homolog (S. pombe)) 458 Amrita Madabushi, Randall C Gunther, A-Lien Lu OTX2 (orthodenticle homeobox 2) 463 Matthew Wortham

Leukaemia Section dic(3;9)(p14;p13) PAX5/FOXP1 466 Jean-Loup Huret dic(9;18)(p13;q11) PAX5/ZNF521 468 Jean-Loup Huret

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

Case Report Section

Chronic lymphocytic leukaemia/Small lymphocytic lymphoma (CLL/SLL) associated with translocation t(1;6)(p35;p25) as part of complex karyotype 470 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

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI ASXL1ID44553ch20q11.txt

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

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) 393

ASXL1 (additional sex combs like 1 (Drosophila)) Mozziconacci MJ, Birnbaum D

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 394

ASXL1 (additional sex combs like 1 (Drosophila)) Mozziconacci MJ, Birnbaum D

Juvenile myelomonocytic leukemia An Q, Wright SL, Konn ZJ, Matheson E, Minto L, Moorman AV, Parker H, Griffiths M, Ross FM, Davies T, Hall AG, (JMML) (2/49 patients 4%) Harrison CJ, Irving JA, Strefford JC. Variable breakpoints target PAX5 in patients with dicentric : a Note model for the basis of unbalanced translocations in cancer. p.Arg693X, p.Ser846ValfsX21. Proc Natl Acad Sci U S A. 2008 Nov 4;105(44):17050-4 Cytogenetics Scotto L, Narayan G, Nandula SV, Arias-Pulido H, Normal karyotype or monosomy 7. Subramaniyam S, Schneider A, Kaufmann AM, Wright JD, 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 Note cancer: potential role in progression. Genes Chromosomes 30% in primary AMLs, 47% in secondary AMLs Cancer. 2008 Sep;47(9):755-65 and 23% in post-MDS AMLs. An Q, Wright SL, Moorman AV, Parker H, Griffiths M, Ross Prognosis FM, Davies T, Harrison CJ, Strefford JC. Heterogeneous Shorter overall survival. breakpoints in patients with acute lymphoblastic leukemia and the dic(9;20)(p11-13;q11) show recurrent involvement Cytogenetics of genes at 20q11.21. Haematologica. 2009 Normal or abnormal karyotype, associated with Aug;94(8):1164-9 trisomy 8, inversely correlated with NPM1 Carbuccia N, Murati A, Trouplin V, Brecqueville M, mutation. Adélaïde J, Rey J, Vainchenker W, Bernard OA, Chaffanet M, Vey N, Birnbaum D, Mozziconacci MJ. Mutations of Myeloproliferative neoplasms (8%) ASXL1 gene in 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, leukemia. More frequent in primary myelofibrosis. Sainty D, Olschwang S, Xerri L, Chaffanet M, Prognosis Mozziconacci MJ, Vey N, Birnbaum D. Mutations of polycomb-associated gene ASXL1 in myelodysplastic Occur in both chronic and blast-phase MPNs. syndromes and chronic myelomonocytic leukaemia. Br J B-cell acute lymphoblastic leukemia Haematol. 2009 Jun;145(6):788-800 Cytogenetics Abdel-Wahab O, Kilpivaara O, Patel J, Busque L, Levine RL. The most commonly reported variant in ASXL1 Dicentric chromosome dic(9;20)(p11-13;q11). (c.1934dupG;p.Gly646TrpfsX12) is not a somatic Hybrid/Mutated gene alteration. Leukemia. 2010 Sep;24(9):1656-7 PAX5 on 9p and ASXL1 on 20q. Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C, Heguy A, Bueso-Ramos C, Kantarjian H, Levine RL, Verstovsek S. Genetic analysis of transforming events To be noted that convert chronic myeloproliferative neoplasms to Note leukemias. Cancer Res. 2010 Jan 15;70(2):447-52 ASXL1 can be altered by small local deletion Acquaviva C, Gelsi-Boyer V, Birnbaum D. Myelodysplastic detected only by array CGH or SNP arrays. syndromes: lost between two states? Leukemia. 2010 Jan;24(1):1-5 However, ASXL1 seems to be centromeric to the main deleted region in classical 20q deletion. Boultwood J, Perry J, Pellagatti A, Fernandez-Mercado M, Fernandez-Santamaria C, Calasanz MJ, Larrayoz MJ, Garcia-Delgado M, Giagounidis A, Malcovati L, Della Porta References MG, Jädersten M, Killick S, Hellström-Lindberg E, Cazzola M, Wainscoat JS. Frequent mutation of the polycomb- Fisher CL, Berger J, Randazzo F, Brock HW. A human associated gene ASXL1 in the myelodysplastic syndromes homolog of Additional sex combs, ADDITIONAL SEX and in acute myeloid leukemia. Leukemia. 2010 COMBS-LIKE 1, maps to chromosome 20q11. Gene. 2003 May;24(5):1062-5 Mar 13;306:115-26 Boultwood J, Perry J, Zaman R, Fernandez-Santamaria C, Katoh M, Katoh M. Identification and characterization of Littlewood T, Kusec R, Pellagatti A, Wang L, Clark RE, ASXL2 gene in silico. Int J Oncol. 2003 Sep;23(3):845-50 Wainscoat JS. High-density single nucleotide Katoh M, Katoh M. Identification and characterization of polymorphism array analysis and ASXL1 gene mutation ASXL3 gene in silico. Int J Oncol. 2004 Jun;24(6):1617-22 screening in chronic myeloid leukemia during disease progression. Leukemia. 2010 Jun;24(6):1139-45 Katoh M, Katoh M. Identification and characterization of human CXXC10 gene in silico. Int J Oncol. 2004 Carbuccia N, Trouplin V, Gelsi-Boyer V, Murati A, Oct;25(4):1193-9 Rocquain J, Adélaïde J, Olschwang S, Xerri L, Vey N, Chaffanet M, Birnbaum D, Mozziconacci MJ. Mutual Cho YS, Kim EJ, Park UH, Sin HS, Um SJ. Additional sex exclusion of ASXL1 and NPM1 mutations in a series of comb-like 1 (ASXL1), in cooperation with SRC-1, acts as a acute myeloid leukemias. Leukemia. 2010 Feb;24(2):469- ligand-dependent coactivator for retinoic acid receptor. J 73 Biol Chem. 2006 Jun 30;281(26):17588-98 Chou WC, Huang HH, Hou HA, Chen CY, Tang JL, Yao M, Fisher CL, Randazzo F, Humphries RK, Brock HW. Tsay W, Ko BS, Wu SJ, Huang SY, Hsu SC, Chen YC, Characterization of Asxl1, a murine homolog of Additional Huang YN, Chang YC, Lee FY, Liu MC, Liu CW, Tseng sex combs, and analysis of the Asx-like gene family. Gene. MH, Huang CF, Tien HF. Distinct clinical and biological 2006 Mar 15;369:109-18 features of de novo acute myeloid leukemia with additional

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 395

ASXL1 (additional sex combs like 1 (Drosophila)) Mozziconacci MJ, Birnbaum D

sex comb-like 1 (ASXL1) mutations. Blood. 2010 Nov Rocquain J, Carbuccia N, Trouplin V, Raynaud S, Murati 18;116(20):4086-94 A, Nezri M, Tadrist Z, Olschwang S, Vey N, Birnbaum D, Gelsi-Boyer V, Mozziconacci MJ. Combined mutations of Davids MS, Steensma DP. The molecular pathogenesis of ASXL1, CBL, FLT3, IDH1, IDH2, JAK2, KRAS, NPM1, myelodysplastic syndromes. Cancer Biol Ther. 2010 NRAS, RUNX1, TET2 and WT1 genes in myelodysplastic Aug;10(4):309-19 syndromes and acute myeloid leukemias. BMC Cancer. Delhommeau F, Jeziorowska D, Marzac C, Casadevall N. 2010 Aug 2;10:401 Molecular aspects of myeloproliferative neoplasms. Int J Sugimoto Y, Muramatsu H, Makishima H, Prince C, Hematol. 2010 Mar;91(2):165-73 Jankowska AM, Yoshida N, Xu Y, Nishio N, Hama A, Fisher CL, Lee I, Bloyer S, Bozza S, Chevalier J, Dahl A, Yagasaki H, Takahashi Y, Kato K, Manabe A, Kojima S, Bodner C, Helgason CD, Hess JL, Humphries RK, Brock Maciejewski JP. Spectrum of molecular defects in juvenile HW. Additional sex combs-like 1 belongs to the enhancer myelomonocytic leukaemia includes ASXL1 mutations. Br of trithorax and polycomb group and genetically interacts J Haematol. 2010 Jul;150(1):83-7 with Cbx2 in mice. Dev Biol. 2010 Jan 1;337(1):9-15 Szpurka H, Jankowska AM, Makishima H, Bodo J, Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Bejanyan N, Hsi ED, Sekeres MA, Maciejewski JP. Bodner C, Hess JL, Humphries RK, Brock HW. Loss-of- Spectrum of mutations in RARS-T patients includes TET2 function Additional sex combs like 1 mutations disrupt and ASXL1 mutations. Leuk Res. 2010 Aug;34(8):969-73 hematopoiesis but do not cause severe myelodysplasia or Tefferi A. Mutational analysis in BCR-ABL-negative classic leukemia. Blood. 2010 Jan 7;115(1):38-46 myeloproliferative neoplasms: impact on prognosis and Gelsi-Boyer V, Trouplin V, Rocquain J, Adelaide J, therapeutic choices. Leuk Lymphoma. 2010 Apr;51(4):576- Carbuccia N, Esterni B, Finetti P, Murati M, Arnoulet C, 82 Zerazhi H, Fezoui H, Tadrist Z, Nezri M, Chaffanet M, Tefferi A. Novel mutations and their functional and clinical Mozziconacci MJ, Vey N, Birnbaum D.. ASXL1 mutation is relevance in myeloproliferative neoplasms: JAK2, MPL, associated with poor prognosis and acute transformation in TET2, ASXL1, CBL, IDH and IKZF1. Leukemia. 2010 chronic myelomonocytic leukaemia. British Journal of Jun;24(6):1128-38 Haematology, 2010 Sept 29. Lee SW, Cho YS, Na JM, Park UH, Kang M, Kim EJ, Um This article should be referenced as such: SJ. ASXL1 represses retinoic acid receptor-mediated Mozziconacci MJ, Birnbaum D. ASXL1 (additional sex transcription through associating with HP1 and LSD1. J combs like 1 (Drosophila)). Atlas Genet Cytogenet Oncol Biol Chem. 2010 Jan 1;285(1):18-29 Haematol. 2011; 15(5):393-396. Mittelman M, Oster HS, Hoffman M, Neumann D. The lower risk MDS patient at risk of rapid progression. Leuk Res. 2010 Dec;34(12):1551-5

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 396

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI EWSR1ID85.txt

This article is an update of : Mugneret F. EWSR1 (Ewing sarcoma region 1). Atlas Genet Cytogenet Oncol Haematol 1998;3(2)

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) DNA/RNA containing multiple degenerate hexapeptide repeats (consensus SYGQQS) (glycine, glutamine, serine, Description tyrosine rich or SYGQ rich, where the tyrosine is Spans 32.5 kb, in a centromere to telomere mandatory): amino acids 1 to 285, with a site direction on plus strand; transcript of 2654 bp from interacting with SF1 from aa 228 to 264 and an IQ 17 exons for the canonical form, with a coding domain, which binds calmodulin (aa 256-285), 3 sequence of 1971 nt. arginine/glycine rich domains (RGG regions) (aa 300-340, 454-513 (arginine/glycine/proline rich), Transcription 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) 397

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 398

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

interaction docks YBX1 to RNA Pol II to premature cellular senescence (Li et al., 2007). participate in pre-mRNA splicing (Chansky et al., EWSR1 is required for proper localization of aurora 2001). B during mitosis, and maintains mitotic spindle SMN1 (survival of motor neuron 1, telomeric, integrity (Azuma et al., 2007). 5q13) plays a major role in the pre-mRNA splicing It also interacts with BARD1 (BRCA1 associated machinery (role in spliceosomal snRNP assembly RING domain 1) (Spahn et al., 2002). in the cytoplasm and in pre-mRNA splicing in the EWSR1 and CCNL1 (cyclin L1, 3q25), are nucleus) (Pellizzoni et al., 1998). SMN1, through interacting partners of TFIP11 (tuftelin-interacting its Tudor region, binds the RG1 region of EWSR1 protein 11, 22q12), a protein functionally related to (Young et al., 2003). the spliceosome and involved in pre-mRNA RNU1-2 (RNA, U1 small nuclear 2, 1p36, also splicing) (Tannukit et al., 2008). called U1C), another splicing protein, also interacts Homology with EWSR1 (Knoop and Baker, 2000). EWSR1 is phosphorylated by PRKC (protein Member of the TET family of RNA binding kinase C) though its IQ domain, which inhibits proteins, with FUS (TLS) and TAF15 (TAFII68). RNA binding of EWSR1; CALM (calmodulin) TET is for Tls, Ewsr1, TafII68). TET proteins binding to EWSR1 inhibits PRKC phosphorylation contain specific structural domains not found (Deloulme et al., 1997). elsewhere in other RNA binding proteins, i.e. a N- EWSR1 interacts with POU4F2 (4q31), a gene term SYGQ rich (TAD: transactivation domain), a which regulates differentiation of neuronal cells conserved RNA-binding domain (RRM: RNA (Gascoyne et al., 2004). EWSR1 interacts with recognition motif), RG rich regions, and a Cys2- LMNA (lamin A/C, 1q22, a component of the Cys2 Zinc finger which can bind nucleic acids; they nuclear envelope which interacts with DNA, are also functionally related (reviews in Law et al., histones and transcriptional repressors) is essential 2006; Tan and Manley, 2009). for pre-B lymphocyte development and, during meiosis, in XY pairing and in meiotic Implicated in recombination, as cross-overs are reduced in ews-/- spermatocytes. Loss of EWSR1 results in Various tumours (see details below)

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 399

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Ewing's sarcoma/Peripheral t(21;22)(q21;q12) with EWSR1/ERG is the second neurectodermal tumour (ES/PNET) in frequency, found in about 10% of cases. Note Hybrid/Mutated gene - With: t(11;22)(q24;q12) --> FLI1 - EWSR1 t(11;22)(q24;q12): 5' EWSR1 - 3' FLI1; breakpoints (Delattre et al., 1992; Bailly et al., 1994; Thomas et clustered over a 2-3 kb genomic region and over a al., 1994; records in the Mitelman Database). 30-40 kb genomic region. Various junctions - With: t(2;22)(q36;q12) --> FEV - EWSR1 between EWSR1 exon 7 or 10 with FLI1 exon 5, 6, (Llombart-Bosch et al., 2000; Peter et al., 2001; or 8. In the most common fusion type (type 1), Navarro et al., 2002; Hattinger et al., 1999; Wang et EWSR1 exon 7 is fused in frame to FLI1 exon 6; in al., 2007). type 2, EWSR1 exon 7 is fused in frame to FLI1 - With: t(7;22)(p22;q12) --> ETV1 - EWSR1 (Jeon exon 5. (Obata et al., 1999; de Alava et al., 1998). et al., 1995; Peter et al., 2001; Zielenska et al., t(21;22)(q21;q12): 5' EWSR1 - 3' ERG; the 2001; Wang et al., 2007). orientation of the ERG gene is from telomere to - With: t(17;22)(q21;q12) --> ETV4 - EWSR1 centromere, opposite to that demonstrated for (Kaneko et al., 1996; Urano et al., 1996; Urano et EWSR1. al., 1998). Other translocations: 5' EWSR1 - 3' FEV; 5' - With: t(20;22)(q13;q12) --> NFATC2 - EWSR1 EWSR1 - 3' ETV1; 5' EWSR1 - 3' ETV4; 5' (Szuhai et al., 2009). EWSR1 - 3' NFATC2; 5' EWSR1 - 3' PATZ1. - With: t(21;22)(q21;q12) --> ERG - EWSR1 Abnormal protein (Dunn et al., 1994; Giovannini et al., 1994; Kaneko t(11;22)(q24;q12): oncogenic protein on the der(22) et al., 1997; Maire et al., 2008; Minoletti et al., chromosome generated by fusion of the N terminal 1998; Sorensen et al., 1994; Zoubek et al., 1994; domain of EWSR1 protein (transactivation domain, Zucman et al., 1993b; Shing et al., 2003; Peter et e.g. fusion of EWSR1 amino acids 1-265) with the al., 1996). DNA binding domain (ETS type, amino acids 281- - With: inv(22)(q12q12) --> PATZ1 - EWSR1 361) of the human FLI1 protein, a 452 amino acids (Mastrangelo et al., 2000) protein (e.g. 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 domain of EWSR1 protein with DNA binding al., 2002) or, domain of human ERG protein. - with: t(16;21)(p11;q22) --> FUS - ERG (Shing et Other translocations: Most of the EWSR1 partners al., 2003; Berg et al., 2009). To be noted that the in ES/PNET are ETS family members (FLI1, ERG, same t(16;21)(p11;q22) has been found in rare ETV1, ETV4, FEV) and translocation results in the cases of acute myeloid leukaemia. juxtaposition of the transactivation domain of A t(11;22)(q24;q12) has also been found in a case EWSR1 with the DNA binding domain -ETS type of cerebellar PNET (Jay et al., 1996). of the partner. PATZ1 is a transcription regulator Disease with a AT hook (DNA-binding motif), a POZ Ewing's sarcoma/Peripheral neurectodermal tumour domain (mediates dimerisation) and Zn fingers spectrum is a group of sarcomas with small blue (DNA-binding). NFATC2 is a cytokine inducer; round cells with more (PNET side) or less (Ewing translocates into the nucleus to regulate side) features of neuroectodermal differentiation. transcription. ES/PNET entity also includes peripheral Oncogenesis neuroepithelioma and Askin tumour. These tumours EWSR1-FLI1 is a dominant oncogene transformed display both mesenchymal stem cell and neural cells by subverting normal transcriptional crest stem cell properties. It is mainly found in controls/FLI1 member of ETS family. The target adolescents and young adults. Cytogenetics and gene repertoire of EWSR1-FLI1 varies according to immunochemistry are essential for the differential the host cell type. EWSR1-FLI1 induces a TP53- diagnosis with other sarcomas (review in Romeo dependent growth arrest in fibroblasts, supporting and Dei Tos, 2010). the importance of TP53 loss in the genesis of Prognosis Ewing's tumours (Lessnick et al., 2002). EWSR1- Prognosis has dramatically improved, from less FLI1 activates CASP3 and promotes apoptosis in than 10% of long survivors, to a 5-year disease free mouse embryonic fibroblasts (Sohn et al., 2010). survival of 75% for patients with a localized EWSR1-FLI1 induces expression of the embryonic disease, and 25-30% for those with a metastatic stem cell genes OCT4, SOX2, and NANOG in disease (Ludwig, 2008). paediatric mesenchymal stem cells but not in adult mesenchymal stem cells. SOX2 is a target for Cytogenetics EWSR1-FLI1 and miRNA145 and may be critical The t(11;22)(q24;q12) EWSR1/FLI1 is found in in Ewing sarcoma pathogenesis (Riggi et al., 2010). 85% of cases of Ewing tumours. The EWSR1-FLI1 expression in a rhabdomyosarcoma

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 400

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 401

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 402

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

However, 4 cases of RMS have been described, Mucoepidermoid carcinoma of the instead, with an EWSR1 rearrangement. salivary glands Cytogenetics Disease A t(4;22)(q35;q12) was found in a case of Mucoepidermoid carcinoma is the most common embryonal rhabdomyosarcoma (Sirvent et al., type of malignant salivary gland tumour, often 2009), and a t(11;22)(q24;q12) in the other cases associated with a t(11;19)(q21;p13) translocation (Sorensen et al., 1993; Thorner et al., 1996). with expression of chimeric genes 5' CRTC1 - 3' Hybrid/Mutated gene MAML2. One case of mucoepidermoid carcinoma 5' EWSR1 - 3' DUX4 in the t(4;22)(q35;q12) case, was found with a t(6;22)(p21;q12) (Möller et al., and 5' EWSR1 - 3' FLI1 in the t(11;22) cases. 2008). Giant cell tumour of bone Abnormal protein Disease 5' EWSR1 - 3' POU5F1. Locally destructive tumour, usually seen in patients Neuroblastoma over 20 years of age, with good prognosis, despite Disease of recurrences and pulmonary metastases (Forsyth Neuroblastomas are peripheral neuroblastic and Hogendoorn, 2003). tumours derived from cells of the sympathetic Cytogenetics nervous system. They occur mainly in infants and In giant-cell tumour of bone, the most frequent young children, with a median age of 1.5 years. finding is telomeric association. One study showed Hybrid/Mutated gene that a very minor clone with EWSR1/FLI1 Two patients, aged 3 years and 5.5 years, were translocation could be detected in most of the cases described with a 5' EWSR1 - 3' FLI1 transcript in studied (Scotlandi et al., 2000). typical neuroblastomas with elevated urinary Hybrid/Mutated gene catecholamines. Prognosis had been very poor: the 5' EWSR1 - 3' FLI1. two patients relapsed during -or at the end of- Myoepithelioma/myoepithelial treatment and died within 2 months (Burchill et al., 1997). carcinoma of soft parts Disease Olfactory neuroblastoma Myoepithelioma tumours of soft tissue cover a wide Disease range of tumours of various behaviour. While most Olfactory neuroblastoma or esthesioneuroblastoma, are of intermediate aggressivity, some metastase. is a malignant neurectodermal tumour, from the olfactory neuroepithelium that typically occurs in Cytogenetics the superior nasal cavity. It is keratin negative, t(1;22)(q23;q12) in one case (Brandal et al., 2008), neuroendocrine marker positive, and S100 positive. t(19;22)(q13;q12) in another case (Brandal et al., It arises at any age, often in the adult, with a 5-year 2009). survival rate above 50% (the 5-year overall survival Hybrid/Mutated gene for patients treated for nonmetastatic olfactory 5 ' EWSR1 - 3' PBX1, which fuses exons 8 from neuroblastoma was recently found at 64% (Ozsahin EWSR1 to exons 5 of PBX1 in the most benign et al., 2010)). case; 5' EWSR1- 3' ZNF444 in the malignant case; Cytogenetics fuses EWSR1 exon 8 to the very near end of t(11;22)(q24;q12), inducing a 5' EWSR1 - 3' FLI1 ZNF444. hybrid gene (Sorensen et al., 1996). However, Hidradenoma of the skin recent review rejects cases with EWSR1 Disease involvement, as being misdiagnosed cases of Hidradenoma or eccrine/apocrine acrospiroma, is a ES/PNET (Thompson, 2009). benign adnexal tumour developing most often in Solid pseudopapillary tumour of the adults. 3 cases were found with a t(6;22)(p21;q12) pancreas (SPTP) and/or its fusion transcript equivalent (Möller et al., 2008). Disease Solid pseudopapillary tumour of the pancreas is a Abnormal protein rare epithelial tumour, typically occuring in young 5' EWSR1 - 3' POU5F1. female patients, rarely metastasizing (Yu et al., 2010).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 403

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Cytogenetics A small round cell tumour was found to have a One case showed a t(11;22)(q24;q12) (Maitra et al., t(2;22)(q31;q12), with 5' EWSR1 - 3' SP3 hybrid 2000) gene; fuses the exon 7 of EWSR1 to exon 6 of SP3. Hybrid/Mutated gene N-term transactivation domain of EWSR1 fused 5' EWSR1 - 3' FLI1. with the Zinc fingers of SP3. The patient died 20 months after diagnosis (Wang et al., 2007). "Small round cell tumours", Other cases of spindle cell tumours, small round "polyphenotypic mesenchymal cell poorly differentiated, biphenotypic malignancies", and "undifferentiated (myogenic/neural differentiation), or sarcomas" polyphenotypic sarcomas present with the classical t(11;22)(q24;q12) / 5' EWSR1 - 3' FLI1 or other Disease variants, such as the t(2;22)(q36;q12) / 5' EWSR1 - An undifferentiated sarcoma derived from pelvic 3' FEV (Wang et al., 2007), the t(11;22)(p13;q12) / bone exhibited a t(6;22)(p21;q12) with 5' EWSR1 - 5' EWSR1 - 3' WT1 (Alaggio et al., 2007), the 3' POU5F1. This resulted in the fusion of exons 1-6 t(12;22)(q13;q12) / 5' EWSR1 - 3' ATF1 (Somers et of EWSR1 and exons 2-5 and a part of exon 1 of al., 2005), or the t(21;22)(q21;q12) / 5' EWSR1 - 3' POU5F1.The patient died 6 months after diagnosis ERG (Tan et al., 2001). (Yamaguchi et al., 2005).

Breakpoints

Note Clustered over a 2.3 kb genomic region. References Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, Zucman J, Delattre O, Desmaze C, Epstein AL, Stenman Peter M, Kovar H, Joubert I, de Jong P, Rouleau G. Gene G, Speleman F, Fletchers CD, Aurias A, Thomas G. EWS fusion with an ETS DNA-binding domain caused by and ATF-1 gene fusion induced by t(12;22) translocation in chromosome translocation in human tumours. Nature. malignant melanoma of soft parts. Nat Genet. 1993 1992 Sep 10;359(6391):162-5 Aug;4(4):341-5 Plougastel B, Zucman J, Peter M, Thomas G, Delattre O. Zucman J, Melot T, Desmaze C, Ghysdael J, Plougastel B, Genomic structure of the EWS gene and its relationship to Peter M, Zucker JM, Triche TJ, Sheer D, Turc-Carel C. EWSR1, a site of tumor-associated chromosome Combinatorial generation of variable fusion proteins in the translocation. Genomics. 1993 Dec;18(3):609-15 Ewing family of tumours. EMBO J. 1993 Dec;12(12):4481- 7 Sorensen PH, Liu XF, Delattre O, Rowland JM, Biggs CA, Thomas G, Triche TJ. Reverse transcriptase PCR Bailly RA, Bosselut R, Zucman J, Cormier F, Delattre O, amplification of EWS/FLI-1 fusion transcripts as a Roussel M, Thomas G, Ghysdael J. DNA-binding and diagnostic test for peripheral primitive neuroectodermal transcriptional activation properties of the EWS-FLI-1 tumors of childhood. Diagn Mol Pathol. 1993 Sep;2(3):147- fusion protein resulting from the t(11;22) translocation in 57 Ewing sarcoma. Mol Cell Biol. 1994 May;14(5):3230-41

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 404

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Dunn T, Praissman L, Hagag N, Viola MV. ERG gene is undifferentiated sarcoma of infancy. Genes Chromosomes translocated in an Ewing's sarcoma cell line. Cancer Genet Cancer. 1996 Feb;15(2):115-21 Cytogenet. 1994 Aug;76(1):19-22 Peter M, Mugneret F, Aurias A, Thomas G, Magdelenat H, Giovannini M, Biegel JA, Serra M, Wang JY, Wei YH, Delattre O. An EWS/ERG fusion with a truncated N- Nycum L, Emanuel BS, Evans GA. EWS-erg and EWS- terminal domain of EWS in a Ewing's tumor. Int J Cancer. Fli1 fusion transcripts in Ewing's sarcoma and primitive 1996 Jul 29;67(3):339-42 neuroectodermal tumors with variant translocations. J Clin Invest. 1994 Aug;94(2):489-96 Sorensen PH, Wu JK, Berean KW, Lim JF, Donn W, Frierson HF, Reynolds CP, López-Terrada D, Triche TJ. Ladanyi M, Gerald W. Fusion of the EWS and WT1 genes Olfactory neuroblastoma is a peripheral primitive in the desmoplastic small round cell tumor. Cancer Res. neuroectodermal tumor related to Ewing sarcoma. Proc 1994 Jun 1;54(11):2837-40 Natl Acad Sci U S A. 1996 Feb 6;93(3):1038-43 Panagopoulos I, Mandahl N, Ron D, Höglund M, Nilbert M, Thorner P, Squire J, Chilton-MacNeil S, Marrano P, Bayani Mertens F, Mitelman F, Aman P. Characterization of the J, Malkin D, Greenberg M, Lorenzana A, Zielenska M. Is CHOP breakpoints and fusion transcripts in myxoid the EWS/FLI-1 fusion transcript specific for Ewing sarcoma liposarcomas with the 12;16 translocation. Cancer Res. and peripheral primitive neuroectodermal tumor? A report 1994 Dec 15;54(24):6500-3 of four cases showing this transcript in a wider range of tumor types. Am J Pathol. 1996 Apr;148(4):1125-38 Sorensen PH, Lessnick SL, Lopez-Terrada D, Liu XF, Triche TJ, Denny CT. A second Ewing's sarcoma Urano F, Umezawa A, Hong W, Kikuchi H, Hata J. A novel translocation, t(21;22), fuses the EWS gene to another chimera gene between EWS and E1A-F, encoding the ETS-family transcription factor, ERG. Nat Genet. 1994 adenovirus E1A enhancer-binding protein, in extraosseous Feb;6(2):146-51 Ewing's sarcoma. Biochem Biophys Res Commun. 1996 Feb 15;219(2):608-12 Thomas G, Delattre O, Zucman J, Merel P, Desmaze C, Melot T, Sanson M, Hoang-Xuan K, Plougastel B, Dejong Brody RI, Ueda T, Hamelin A, Jhanwar SC, Bridge JA, P. Genetic alterations in the chromosome 22q12 region Healey JH, Huvos AG, Gerald WL, Ladanyi M. Molecular associated with development of neuroectodermal tumors. analysis of the fusion of EWS to an orphan nuclear Cold Spring Harb Symp Quant Biol. 1994;59:555-64 receptor gene in extraskeletal myxoid chondrosarcoma. Am J Pathol. 1997 Mar;150(3):1049-58 Zinszner H, Albalat R, Ron D. A novel effector domain from the RNA-binding protein TLS or EWS is required for Burchill SA, Wheeldon J, Cullinane C, Lewis IJ. EWS-FLI1 oncogenic transformation by CHOP. Genes Dev. 1994 Nov fusion transcripts identified in patients with typical 1;8(21):2513-26 neuroblastoma. Eur J Cancer. 1997 Feb;33(2):239-43 Zoubek A, Pfleiderer C, Salzer-Kuntschik M, Amann G, Deloulme JC, Prichard L, Delattre O, Storm DR. The Windhager R, Fink FM, Koscielniak E, Delattre O, Strehl S, prooncoprotein EWS binds calmodulin and is Ambros PF. Variability of EWS chimaeric transcripts in phosphorylated by protein kinase C through an IQ domain. Ewing tumours: a comparison of clinical and molecular J Biol Chem. 1997 Oct 24;272(43):27369-77 data. Br J Cancer. 1994 Nov;70(5):908-13 Desmaze C, Brizard F, Turc-Carel C, Melot T, Delattre O, Gerald WL, Rosai J, Ladanyi M. Characterization of the Thomas G, Aurias A. Multiple chromosomal mechanisms genomic breakpoint and chimeric transcripts in the EWS- generate an EWS/FLI1 or an EWS/ERG fusion gene in WT1 gene fusion of desmoplastic small round cell tumor. Ewing tumors. Cancer Genet Cytogenet. 1997 Proc Natl Acad Sci U S A. 1995 Feb 14;92(4):1028-32 Aug;97(1):12-9 Jeon IS, Davis JN, Braun BS, Sublett JE, Roussel MF, Kaneko Y, Kobayashi H, Handa M, Satake N, Maseki N. Denny CT, Shapiro DN. A variant Ewing's sarcoma EWS-ERG fusion transcript produced by chromosomal translocation (7;22) fuses the EWS gene to the ETS gene insertion in a Ewing sarcoma. Genes Chromosomes ETV1. Oncogene. 1995 Mar 16;10(6):1229-34 Cancer. 1997 Mar;18(3):228-31 Labelle Y, Zucman J, Stenman G, Kindblom LG, Knight J, Bertolotti A, Melot T, Acker J, Vigneron M, Delattre O, Tora Turc-Carel C, Dockhorn-Dworniczak B, Mandahl N, L. EWS, but not EWS-FLI-1, is associated with both TFIID Desmaze C, Peter M. Oncogenic conversion of a novel and RNA polymerase II: interactions between two orphan nuclear receptor by chromosome translocation. members of the TET family, EWS and hTAFII68, and Hum Mol Genet. 1995 Dec;4(12):2219-26 subunits of TFIID and RNA polymerase II complexes. Mol Cell Biol. 1998 Mar;18(3):1489-97 Alliegro MC, Alliegro MA. A nuclear protein regulated during the transition from active to quiescent phenotype in de Alava E, Kawai A, Healey JH, Fligman I, Meyers PA, cultured endothelial cells. Dev Biol. 1996 Mar Huvos AG, Gerald WL, Jhanwar SC, Argani P, Antonescu 15;174(2):288-97 CR, Pardo-Mindan FJ, Ginsberg J, Womer R, Lawlor ER, Wunder J, Andrulis I, Sorensen PH, Barr FG, Ladanyi M. Benjamin LE, Fredericks WJ, Barr FG, Rauscher FJ 3rd. EWS-FLI1 fusion transcript structure is an independent Fusion of the EWS1 and WT1 genes as a result of the determinant of prognosis in Ewing's sarcoma. J Clin Oncol. t(11;22)(p13;q12) translocation in desmoplastic small 1998 Apr;16(4):1248-55 round cell tumors. Med Pediatr Oncol. 1996 Nov;27(5):434-9 Minoletti F, Sozzi G, Tornielli S, Pilotti S, Azzarelli A, Pierotti MA, Radice P. A novel EWS-ERG rearrangement Jay V, Zielenska M, Lorenzana A, Drake J. An unusual generating two hybrid mRNAs in a peripheral primitive cerebellar primitive neuroectodermal tumor with t(11;22) neuroectodermal tumour (pPNET) with a t(15;22) translocation: pathological and molecular analysis. Pediatr translocation. J Pathol. 1998 Dec;186(4):434-7 Pathol Lab Med. 1996 Jan-Feb;16(1):119-28 Ordi J, de Alava E, Torné A, Mellado B, Pardo-Mindan J, Kaneko Y, Yoshida K, Handa M, Toyoda Y, Nishihira H, Iglesias X, Cardesa A. Intraabdominal desmoplastic small Tanaka Y, Sasaki Y, Ishida S, Higashino F, Fujinaga K. round cell tumor with EWS/ERG fusion transcript. Am J Fusion of an ETS-family gene, EIAF, to EWS by Surg Pathol. 1998 Aug;22(8):1026-32 t(17;22)(q12;q12) chromosome translocation in an

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 405

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Pellizzoni L, Kataoka N, Charroux B, Dreyfuss G. A novel identifies fusion of the FUS and ATF-1 genes induced by a function for SMN, the spinal muscular atrophy disease chromosomal translocation involving bands 12q13 and gene product, in pre-mRNA splicing. Cell. 1998 Nov 16p11. Cancer Genet Cytogenet. 2000 Sep;121(2):109-16 25;95(5):615-24 Yang L, Chansky HA, Hickstein DD. EWS.Fli-1 fusion Urano F, Umezawa A, Yabe H, Hong W, Yoshida K, protein interacts with hyperphosphorylated RNA Fujinaga K, Hata J. Molecular analysis of Ewing's polymerase II and interferes with serine-arginine protein- sarcoma: another fusion gene, EWS-E1AF, available for mediated RNA splicing. J Biol Chem. 2000 Dec diagnosis. Jpn J Cancer Res. 1998 Jul;89(7):703-11 1;275(48):37612-8 Zhang D, Paley AJ, Childs G. The transcriptional repressor Arvand A, Welford SM, Teitell MA, Denny CT. The COOH- ZFM1 interacts with and modulates the ability of EWS to terminal domain of FLI-1 is necessary for full activate transcription. J Biol Chem. 1998 Jul tumorigenesis and transcriptional modulation by EWS/FLI- 17;273(29):18086-91 1. Cancer Res. 2001 Jul 1;61(13):5311-7 Attwooll C, Tariq M, Harris M, Coyne JD, Telford N, Varley Chansky HA, Hu M, Hickstein DD, Yang L. Oncogenic JM. Identification of a novel fusion gene involving hTAFII68 TLS/ERG and EWS/Fli-1 fusion proteins inhibit RNA and CHN from a t(9;17)(q22;q11.2) translocation in an splicing mediated by YB-1 protein. Cancer Res. 2001 May extraskeletal myxoid chondrosarcoma. Oncogene. 1999 1;61(9):3586-90 Dec 9;18(52):7599-601 Knoop LL, Baker SJ. EWS/FLI alters 5'-splice site Hahm KB. Repression of the gene encoding the TGF-beta selection. J Biol Chem. 2001 Jun 22;276(25):22317-22 type II receptor is a major target of the EWS-FLI1 oncoprotein. Nat Genet. 1999 Dec;23(4):481 Peter M, Gilbert E, Delattre O. A multiplex real-time pcr assay for the detection of gene fusions observed in solid Hattinger CM, Rumpler S, Strehl S, Ambros IM, Zoubek A, tumors. Lab Invest. 2001 Jun;81(6):905-12 Pötschger U, Gadner H, Ambros PF. Prognostic impact of deletions at 1p36 and numerical aberrations in Ewing Rossow KL, Janknecht R. The Ewing's sarcoma gene tumors. Genes Chromosomes Cancer. 1999 product functions as a transcriptional activator. Cancer Mar;24(3):243-54 Res. 2001 Mar 15;61(6):2690-5 Meis-Kindblom JM, Bergh P, Gunterberg B, Kindblom LG. Tan SY, Burchill S, Brownhill SC, Gerrard MP, Watmore A, Extraskeletal myxoid chondrosarcoma: a reappraisal of its Wagner BE, Variend S. Small round cell tumor with morphologic spectrum and prognostic factors based on biphenotypic differentiation and variant of 117 cases. Am J Surg Pathol. 1999 Jun;23(6):636-50 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 Zielenska M, Zhang ZM, Ng K, Marrano P, Bayani J, junction in a t(11;22) translocation in Ewing sarcoma. Ramirez OC, Sorensen P, Thorner P, Greenberg M, Genes Chromosomes Cancer. 1999 May;25(1):6-15 Squire JA. Acquisition of secondary structural chromosomal changes in pediatric ewing sarcoma is a Sjögren H, Meis-Kindblom J, Kindblom LG, Aman P, probable prognostic factor for tumor response and clinical Stenman G. Fusion of the EWS-related gene TAF2N to outcome. Cancer. 2001 Jun 1;91(11):2156-64 TEC in 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. Knoop LL, Baker SJ. The splicing factor U1C represses Oncogene. 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 Llombart-Bosch A, Pellín A, Carda C, Noguera R, Navarro growth arrest in primary human fibroblasts. Cancer Cell. S, Peydró-Olaya A. Soft tissue Ewing sarcoma--peripheral 2002 May;1(4):393-401 primitive neuroectodermal tumor with atypical clear cell pattern shows a new type of EWS-FEV fusion transcript. Martini A, La Starza R, Janssen H, Bilhou-Nabera C, Diagn Mol 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. Ewing's sarcoma gene, EWSR1, or its homologue, TAF15, Detection of t(11;22)(q24;q12) translocation and EWS-FLI- with the transcription factor CIZ/NMP4 in acute leukemia. 1 fusion transcript in a case of solid pseudopapillary tumor Cancer Res. 2002 Oct 1;62(19):5408-12 of the pancreas. Pediatr Dev Pathol. 2000 Nov- Dec;3(6):603-5 Navarro S, Noguera R, Pellín A, López-Guerrero JA, Roselló-Sastre E, Cremades A, Llombart-Bosch A. Mastrangelo T, Modena P, Tornielli S, Bullrich F, Testi MA, Atypical pleomorphic extraosseous ewing tumor/peripheral Mezzelani A, Radice P, Azzarelli A, Pilotti S, Croce CM, primitive neuroectodermal tumor with unusual Pierotti MA, Sozzi G. A novel zinc finger gene is fused to phenotypic/genotypic profile. Diagn Mol Pathol. 2002 EWS in small round cell tumor. Oncogene. 2000 Aug Mar;11(1):9-15 3;19(33):3799-804 Raddaoui E, Donner LR, Panagopoulos I. Fusion of the Scotlandi K, Chano T, Benini S, Serra M, Manara MC, FUS and ATF1 genes in a large, deep-seated angiomatoid Cerisano V, Picci P, Baldini N. Identification of EWS/FLI-1 fibrous histiocytoma. Diagn Mol Pathol. 2002 transcripts in giant-cell tumor of bone. Int J Cancer. 2000 Sep;11(3):157-62 Aug 1;87(3):328-35 Spahn L, Petermann R, Siligan C, Schmid JA, Aryee DN, Sjögren H, Wedell B, Meis-Kindblom JM, Kindblom LG, Kovar H. Interaction of the EWS NH2 terminus with Stenman G. Fusion of the NH2-terminal domain of the BARD1 links the Ewing's sarcoma gene to a common basic helix-loop-helix protein TCF12 to TEC in tumor suppressor pathway. Cancer Res. 2002 Aug extraskeletal myxoid chondrosarcoma with translocation 15;62(16):4583-7 t(9;15)(q22;q21). Cancer Res. 2000 Dec 15;60(24):6832-5 Araya N, Hirota K, Shimamoto Y, Miyagishi M, Yoshida E, Waters BL, Panagopoulos I, Allen EF. Genetic Ishida J, Kaneko S, Kaneko M, Nakajima T, Fukamizu A. characterization of angiomatoid fibrous histiocytoma Cooperative interaction of EWS with CREB-binding protein

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 406

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

selectively activates hepatocyte nuclear factor 4-mediated Kim S, Denny CT, Wisdom R. Cooperative DNA binding transcription. J Biol Chem. 2003 Feb 14;278(7):5427-32 with AP-1 proteins is required for transformation by EWS- Ets fusion proteins. Mol Cell Biol. 2006 Apr;26(7):2467-78 Belyanskaya LL, Delattre O, Gehring H. Expression and subcellular localization of Ewing sarcoma (EWS) protein is Law WJ, Cann KL, Hicks GG. TLS, EWS and TAF15: a affected by the methylation process. Exp Cell Res. 2003 model for transcriptional integration of gene expression. Aug 15;288(2):374-81 Brief Funct Genomic Proteomic. 2006 Mar;5(1):8-14 Forsyth RG, Hogendoorn PCW.. Bone: Giant cell tumour. Smith R, Owen LA, Trem DJ, Wong JS, Whangbo JS, Atlas Genet Cytogenet Oncol Haematol. June 2003. URL: Golub TR, Lessnick SL. Expression profiling of EWS/FLI http://AtlasGeneticsOncology.org/Tumors/BoneGiantCellT identifies NKX2.2 as a critical target gene in Ewing's umID5150.html. sarcoma. Cancer 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 Nicholson J, Tillman RM, Ramani P, Cullinane C, Coleman JD, Cripe TP, Rice AM, Lee SB, Uren A. Oncoprotein N. FUS/ERG gene fusions in Ewing's tumors. Cancer Res. EWS-FLI1 activity is enhanced by RNA helicase A. Cancer 2003 Aug 1;63(15):4568-76 Res. 2006 Jun 1;66(11):5574-81 Young PJ, Francis JW, Lince D, Coon K, Androphy EJ, Alaggio R, Rosolen A, Sartori F, Leszl A, d'Amore ES, Lorson CL. The Ewing's sarcoma protein interacts with the Bisogno G, Carli M, Cecchetto G, Coffin CM, Ninfo V. Tudor domain of the survival motor neuron protein. Brain Spindle cell tumor with EWS-WT1 transcript and a Res Mol Brain Res. 2003 Nov 6;119(1):37-49 favorable clinical course: a variant of DSCT, a variant of leiomyosarcoma, or a new entity? Report of 2 pediatric Gascoyne DM, Thomas GR, Latchman DS. The effects of cases. Am J Surg Pathol. 2007 Mar;31(3):454-9 Brn-3a on neuronal differentiation and apoptosis are differentially modulated by EWS and its oncogenic Antonescu CR, Dal Cin P, Nafa K, Teot LA, Surti U, derivative EWS/Fli-1. Oncogene. 2004 May 6;23(21):3830- Fletcher CD, Ladanyi M. EWSR1-CREB1 is the 40 predominant gene fusion in angiomatoid fibrous histiocytoma. Genes Chromosomes Cancer. 2007 Hisaoka M, Ishida T, Imamura T, Hashimoto H. TFG is a Dec;46(12):1051-60 novel 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, haematopoietic cells. PLoS Biol. 2005 Aug;3(8):e242 Bacchini P, Mandahl N, Mertens F, Panagopoulos I. Fusion genes in angiomatoid fibrous histiocytoma. Cancer Hallor KH, Mertens F, Jin Y, Meis-Kindblom JM, Kindblom Lett. 2007 Jun 18;251(1):158-63 LG, Behrendtz M, Kalén A, Mandahl N, Panagopoulos I. Fusion of the EWSR1 and ATF1 genes without expression Li H, Watford W, Li C, Parmelee A, Bryant MA, Deng C, of the MITF-M transcript in angiomatoid fibrous O'Shea J, Lee SB. Ewing sarcoma gene EWS is essential histiocytoma. Genes Chromosomes Cancer. 2005 for meiosis and B lymphocyte development. J Clin Invest. Sep;44(1):97-102 2007 May;117(5):1314-23 Hu-Lieskovan S, Zhang J, Wu L, Shimada H, Schofield Rossi S, Szuhai K, Ijszenga M, Tanke HJ, Zanatta L, Sciot DE, Triche TJ. EWS-FLI1 fusion protein up-regulates R, Fletcher CD, Dei Tos AP, Hogendoorn PC. EWSR1- critical genes in neural crest development and is CREB1 and EWSR1-ATF1 fusion genes in angiomatoid responsible for the observed phenotype of Ewing's family fibrous histiocytoma. Clin Cancer Res. 2007 Dec of tumors. Cancer Res. 2005 Jun 1;65(11):4633-44 15;13(24):7322-8 Janknecht R. EWS-ETS oncoproteins: the linchpins of Tirode F, Laud-Duval K, Prieur A, Delorme B, Charbord P, Ewing 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. Wang L, Bhargava R, Zheng T, Wexler L, Collins MH, 2005 Jun-Jul;23(6):738-51 Roulston D, Ladanyi M. Undifferentiated small round cell sarcomas with rare EWS gene fusions: identification of a Somers GR, Viero S, Nathan PC, Teshima I, Pereira C, novel EWS-SP3 fusion and of additional cases with the Zielenska M. Association of the t(12;22)(q13;q12) EWS-ETV1 and EWS-FEV fusions. J Mol Diagn. 2007 EWS/ATF1 rearrangement with polyphenotypic round cell Sep;9(4):498-509 sarcoma of bone: a case report. Am J Surg Pathol. 2005 Dec;29(12):1673-9 Andersson MK, Ståhlberg A, Arvidsson Y, Olofsson A, Semb H, Stenman G, Nilsson O, Aman P. The Yamaguchi S, Yamazaki Y, Ishikawa Y, Kawaguchi N, multifunctional FUS, EWS and TAF15 proto-oncoproteins Mukai H, Nakamura T. EWSR1 is fused to POU5F1 in a show cell type-specific expression patterns and bone tumor with translocation t(6;22)(p21;q12). Genes involvement in cell spreading and stress response. BMC Chromosomes Cancer. 2005 Jun;43(2):217-22 Cell Biol. 2008 Jul 11;9:37 Antonescu CR, Nafa K, Segal NH, Dal Cin P, Ladanyi M. Brandal P, Panagopoulos I, Bjerkehagen B, Gorunova L, EWS-CREB1: a recurrent variant fusion in clear cell Skjeldal S, Micci F, Heim S. Detection of a sarcoma--association with gastrointestinal location and t(1;22)(q23;q12) translocation leading to an EWSR1-PBX1 absence of melanocytic differentiation. Clin Cancer Res. fusion gene in a myoepithelioma. Genes Chromosomes 2006 Sep 15;12(18):5356-62 Cancer. 2008 Jul;47(7):558-64 Guipaud O, Guillonneau F, Labas V, Praseuth D, Rossier Cironi L, Riggi N, Provero P, Wolf N, Suvà ML, Suvà D, J, Lopez B, Bertrand P. An in vitro enzymatic assay Kindler V, Stamenkovic I. IGF1 is a common target gene of coupled to proteomics analysis reveals a new DNA Ewing's sarcoma fusion proteins in mesenchymal processing activity for Ewing sarcoma and TAF(II)68 progenitor cells. PLoS One. 2008 Jul 9;3(7):e2634 proteins. Proteomics. 2006 Nov;6(22):5962-72

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 407

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

Dunham C, Hussong J, Seiff M, Pfeifer J, Perry A. Primary localization of the Ewing sarcoma proto-oncoprotein and intracerebral angiomatoid fibrous histiocytoma: report of a its association with and stabilization of microtubules. J Mol case with a t(12;22)(q13;q12) causing type 1 fusion of the Biol. 2009 Feb 13;386(1):1-13 EWS and ATF-1 genes. Am J Surg Pathol. 2008 Mar;32(3):478-84 Miyagawa Y, Okita H, Itagaki M, Toyoda M, Katagiri YU, Fujimoto J, Hata J, Umezawa A, Kiyokawa N. EWS/ETS Lin PP, Pandey MK, Jin F, Xiong S, Deavers M, Parant regulates the expression of the Dickkopf family in Ewing JM, Lozano G. EWS-FLI1 induces developmental family tumor cells. PLoS One. 2009;4(2):e4634 abnormalities and accelerates sarcoma formation in a transgenic mouse model. Cancer Res. 2008 Nov Reichek J, Barr FG.. Soft tissue tumors: 1;68(21):8968-75 Rhabdomyosarcoma. Atlas Genet Cytogenet Oncol Haematol. January 2009. URL: Ludwig JA. Ewing sarcoma: historical perspectives, current http://AtlasGeneticsOncology.org/Tumors/rhab5004.html. state-of-the-art, and opportunities for targeted therapy in the future. Curr Opin Oncol. 2008 Jul;20(4):412-8 Richter GH, Plehm S, Fasan A, Rössler S, Unland R, Bennani-Baiti IM, Hotfilder M, Löwel D, von Luettichau I, Maire G, Brown CW, Bayani J, Pereira C, Gravel DH, Bell Mossbrugger I, Quintanilla-Martinez L, Kovar H, Staege JC, Zielenska M, Squire JA. Complex rearrangement of MS, Müller-Tidow C, Burdach S. EZH2 is a mediator of chromosomes 19, 21, and 22 in Ewing sarcoma involving a EWS/FLI1 driven tumor growth and metastasis blocking novel reciprocal inversion-insertion mechanism of EWS- endothelial and neuro-ectodermal differentiation. Proc Natl ERG fusion gene formation: a case analysis and literature Acad Sci U S A. 2009 Mar 31;106(13):5324-9 review. Cancer Genet Cytogenet. 2008 Mar;181(2):81-92 Shao L, Singh V, Cooley L. Angiomatoid fibrous Möller E, Stenman G, Mandahl N, Hamberg H, Mölne L, histiocytoma with t(2;22)(q33;q12.2) and EWSR1 gene van den Oord JJ, Brosjö O, Mertens F, Panagopoulos I. rearrangement. Pediatr Dev Pathol. 2009 Mar- POU5F1, encoding a key regulator of stem cell Apr;12(2):143-6 pluripotency, is fused to EWSR1 in hidradenoma of the skin and mucoepidermoid carcinoma of the salivary Sirvent N, Trassard M, Ebran N, Attias R, Pedeutour F. glands. J Pathol. 2008 May;215(1):78-86 Fusion of EWSR1 with the DUX4 facioscapulohumeral muscular dystrophy region resulting from t(4;22)(q35;q12) Riggi N, Suvà ML, Suvà D, Cironi L, Provero P, Tercier S, in a case of embryonal rhabdomyosarcoma. Cancer Genet Joseph JM, Stehle JC, Baumer K, Kindler V, Stamenkovic Cytogenet. 2009 Nov;195(1):12-8 I. EWS-FLI-1 expression triggers a Ewing's sarcoma initiation program in primary human mesenchymal stem Szuhai K, Ijszenga M, de Jong D, Karseladze A, Tanke HJ, cells. Cancer Res. 2008 Apr 1;68(7):2176-85 Hogendoorn PC. The NFATc2 gene is involved in a novel cloned translocation in a Ewing sarcoma variant that Tannukit S, Wen X, Wang H, Paine ML. TFIP11, CCNL1 couples its function in immunology to oncology. Clin and EWSR1 Protein-protein Interactions, and Their Cancer Res. 2009 Apr 1;15(7):2259-68 Nuclear Localization. Int J Mol Sci. 2008;9(8):1504-1514 Tan AY, Manley JL. The TET family of proteins: functions Beauchamp E, Bulut G, Abaan O, Chen K, Merchant A, and roles in disease. J Mol Cell Biol. 2009 Dec;1(2):82-92 Matsui W, Endo Y, Rubin JS, Toretsky J, Uren A. GLI1 is a direct transcriptional target of EWS-FLI1 oncoprotein. J Thompson LD. Olfactory neuroblastoma. Head Neck Biol Chem. 2009 Apr 3;284(14):9074-82 Pathol. 2009 Sep;3(3):252-9 Berg T, Kalsaas AH, Buechner J, Busund LT. Ewing Wang WL, Mayordomo E, Zhang W, Hernandez VS, Tuvin sarcoma-peripheral neuroectodermal tumor of the kidney D, Garcia L, Lev DC, Lazar AJ, López-Terrada D. with a FUS-ERG fusion transcript. Cancer Genet Detection and characterization of EWSR1/ATF1 and Cytogenet. 2009 Oct;194(1):53-7 EWSR1/CREB1 chimeric transcripts in clear cell sarcoma (melanoma of soft parts). Mod Pathol. 2009 Brandal P, Panagopoulos I, Bjerkehagen B, Heim S. Sep;22(9):1201-9 t(19;22)(q13;q12) Translocation leading to the novel fusion gene EWSR1-ZNF444 in soft tissue myoepithelial Erkizan HV, Uversky VN, Toretsky JA. Oncogenic carcinoma. Genes Chromosomes Cancer. 2009 partnerships: EWS-FLI1 protein interactions initiate key Dec;48(12):1051-6 pathways of Ewing's sarcoma. Clin Cancer Res. 2010 Aug 15;16(16):4077-83 Erkizan HV, Kong Y, Merchant M, Schlottmann S, Barber- Rotenberg JS, Yuan L, Abaan OD, Chou TH, Jedlicka P. Ewing Sarcoma, an enigmatic malignancy of Dakshanamurthy S, Brown ML, Uren A, Toretsky JA. A likely progenitor cell origin, driven by transcription factor small molecule blocking oncogenic protein EWS-FLI1 oncogenic fusions. Int J Clin Exp Pathol. 2010 Mar interaction with RNA helicase A inhibits growth of Ewing's 19;3(4):338-47 sarcoma. Nat Med. 2009 Jul;15(7):750-6 Navarro D, Agra N, Pestaña A, Alonso J, González- Gervasini C.. CREBBP (CREB binding protein). Atlas Sancho JM. The EWS/FLI1 oncogenic protein inhibits Genet Cytogenet Oncol Haematol. March 2009. URL: expression of the Wnt inhibitor DICKKOPF-1 gene and http://AtlasGeneticsOncology.org/Genes/CBPID42.html antagonizes beta-catenin/TCF-mediated transcription. Carcinogenesis. 2010 Mar;31(3):394-401 Joo J, Christensen L, Warner K, States L, Kang HG, Vo K, Lawlor ER, May WA. GLI1 is a central mediator of Ozsahin M, Gruber G, Olszyk O, Karakoyun-Celik O, EWS/FLI1 signaling in Ewing tumors. PLoS One. 2009 Oct Pehlivan B, Azria D, Roelandts M, Kaanders JH, Cengiz 27;4(10):e7608 M, Krengli M, Matzinger O, Zouhair A. Outcome and prognostic factors in olfactory neuroblastoma: a rare Kinsey M, Smith R, Iyer AK, McCabe ER, Lessnick SL. cancer network study. Int J Radiat Oncol Biol Phys. 2010 EWS/FLI and its downstream target NR0B1 interact Nov 15;78(4):992-7 directly to modulate transcription and oncogenesis in Ewing's sarcoma. Cancer Res. 2009 Dec 1;69(23):9047- Riggi N, Suvà ML, De Vito C, Provero P, Stehle JC, 55 Baumer K, Cironi L, Janiszewska M, Petricevic T, Suvà D, Tercier S, Joseph JM, Guillou L, Stamenkovic I. EWS-FLI- Leemann-Zakaryan RP, Pahlich S, Sedda MJ, Quero L, 1 modulates miRNA145 and SOX2 expression to initiate Grossenbacher D, Gehring H. Dynamic subcellular mesenchymal stem cell reprogramming toward Ewing

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 408

EWSR1 (Ewing sarcoma breakpoint region 1) Huret JL

sarcoma cancer stem cells. Genes Dev. 2010 Tanas MR, Rubin BP, Montgomery EA, Turner SL, Cook May;24(9):916-32 JR, Tubbs RR, Billings SD, Goldblum JR. Utility of FISH in the diagnosis of angiomatoid fibrous histiocytoma: a series Rocchi A, Manara MC, Sciandra M, Zambelli D, Nardi F, of 18 cases. Mod Pathol. 2010 Jan;23(1):93-7 Nicoletti G, Garofalo C, Meschini S, Astolfi A, Colombo MP, Lessnick SL, Picci P, Scotlandi K. CD99 inhibits Yu PF, Hu ZH, Wang XB, Guo JM, Cheng XD, Zhang YL, neural differentiation of human Ewing sarcoma cells and Xu Q. Solid pseudopapillary tumor of the pancreas: a thereby contributes to oncogenesis. J Clin Invest. 2010 review of 553 cases in Chinese literature. World J Mar 1;120(3):668-80 Gastroenterol. 2010 Mar 14;16(10):1209-14

Romeo S, Dei Tos AP. Soft tissue tumors associated with This article should be referenced as such: EWSR1 translocation. Virchows Arch. 2010 Feb;456(2):219-34 Huret JL. EWSR1 (Ewing sarcoma breakpoint region 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):397- Sohn EJ, Li H, Reidy K, Beers LF, Christensen BL, Lee 409. SB. EWS/FLI1 oncogene activates caspase 3 transcription and triggers apoptosis in vivo. Cancer Res. 2010 Feb 1;70(3):1154-63

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 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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI FAM57AID40183ch17p13.txt

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 Other names: CT120, FLJ22282 CT120 is universally expressed in different human normal tissues and in various human tumor cell HGNC (Hugo): FAM57A lines. Location: 17p13.3 Localisation DNA/RNA CT120 is a novel plasma membrane-associated gene. Description Function Gene size: 2145 bp in length, ORF 774 bp. CT120 may assume very essential physiological Full-length cDNA of CT120/FAM57A contains functions involving in amino acid transport and 2145 base pairs and encodes a protein with 257 glutathione metabolism through interaction with amino acids. SLC3A2 and GGTL3B. Transcription Homology The CT120 contains two isoforms in human: one Homology comparison revealed that CT120 is isoform identified was termed CT120A; another highly conserved during biological evolution. isoform (AAH26023.1) was named CT120B, which consists of four exons and encodes a protein with Implicated in 225 amino acids (the fourth exon in CT120A is spliced). Lung cancer Protein Prognosis 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. Description The suppression of CT120A expression also sensitized cells to ultraviolet-induced apoptosis. - CT120: 257 aa; 29 kDa. Atlas cDNA expression array revealed that the - CT120B: 225 aa; 25 kDa.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 410

FAM57A (family with sequence similarity 57, member A) Chen Z, He X

expressions of several apoptosis- and growth- associated genes were altered underlying the References molecular mechanisms of these cell biological He X, Di Y, Li J, Xie Y, Tang Y, Zhang F, Wei L, Zhang Y, behaviors. Qin W, Huo K, Li Y, Wan D, Gu J. Molecular cloning and characterization of CT120, a novel membrane-associated Oncogenesis gene involved in amino acid transport and glutathione CT120 ectopic expression could promote cell metabolism. Biochem Biophys Res Commun. 2002 Sep proliferation activity of NIH3T3 cells, and two 27;297(3):528-36 major signaling pathways involved in cell He XH, Li JJ, Xie YH, Tang YT, Yao GF, Qin WX, Wan DF, proliferation, cell survival and anti-apoptosis were Gu JR. Altered gene expression profiles of NIH3T3 cells regulated by human lung cancer associated gene CT120. overexpressed and activated in response to CT120: Cell Res. 2004 Dec;14(6):487-96 one is the Raf/MEK/Erk signal cascades and the other is the PI3K/Akt signal cascades, suggesting Pan DN, Li JJ, Wei L, Yao M, Wan DF, Gu JR. Inhibitory effect of CT120B, an alternative splice variant of CT120A, that CT120 might contribute, at least in part, to the on lung cancer cell growth. Acta Biochim Biophys Sin constitutively activation of Erk and Akt in human (Shanghai). 2005 Sep;37(9):588-92 lung cancer cells. Pan D, Wei L, Yao M, Wan D, Gu J. Down-regulation of In addition, some tumor metastasis associated genes CT120A by RNA interference suppresses lung cancer cells cathepsin B, cathepsin D, cathepsin L, MMP- growth and sensitizes to ultraviolet-induced apoptosis. 2/TIMP-2 were also upregulated by CT120, upon Cancer Lett. 2006 Apr 8;235(1):26-33 which CT120 might be involved in tumor Li Z, Shao S, Xie S, Jiao F, Ma Y, Shi S. Silencing of invasiveness and metastasis. CT120 by antisense oligonucleotides could inhibit the lung In addition, CT120 might play an important role in cancer cells growth. Ir J Med Sci. 2010 Jun;179(2):217-23 tumor progression through modulating the This article should be referenced as such: expression of some candidate "lung tumor progression" genes including B-Raf, Rab-2, BAX, Chen Z, He X. FAM57A (family with sequence similarity 57, member A). Atlas Genet Cytogenet Oncol Haematol. BAG-1, YB-1 and Cdc42. 2011; 15(5):410-411.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 411

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI FUT8ID40649ch14q23.txt

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 Other names: MGC26465 Some splicing variants of the 5'-untranslated region 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 DNA/RNA gene. Three transcripts with different 5'- untranslated regions have been identified. With Description respect to coding region, four variants were Human FUT8 gene is located on chromosome reported to encode polypeptides containing 575, 14q23.3 (Yamaguchi et al., 1999). This gene 446, 308 and 169 amino acid residues. The 575 encompasses approximately 333 kb and contains residue protein is a fully active alpha1,6- nine exons with coding regions and three 5'- fucosyltransferase, which was first of the variants to untranslated exons (Yamaguchi et al., 2000; be identified. 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) 412

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 amino acid variant is known to be expressed in the FUT8 catalyzes the transfer of a fucose residue retina (Yamaguchi et al., 2000). from GDP-fucose to the reducing terminal GlcNAc of Asn-linked oligosaccharide (N-glycan) via an Protein alpha1.6-linkage (Figure 3). The resulting fucosyl residue is often refered to as a core fucose. The Description reaction does not require any divalent cations or FUT8 was purified and cloned as a cDNA from cofactors. The deletion of the FUT8 gene in mice porcine brain and a human gastric cancer cell line leads to severe phenotypes that exhibit growth (Uozumi et al., 1996, Yanagidani et al., 1997). retardation, lung emphysema and death during Human FUT8 is comprised of 575 amino acids, postnatal development (Wang et al., 2005). As has with a calculated molecular weight of 66516. FUT8 been clearly shown in studies using knockout mice, contains no N-glycosylation sites. This enzyme the lack of core fucosylation resulted in the belongs to the GT23 family of the CaZY biological activities of various proteins to be classification. The structual analysis of a perturbed (Taniguchi et al., 2006; Takahashi et al., transmembrane domain-truncated form of FUT8 2009). Examples of this include the TGF-beta1 showed that the enzyme consists of a catalytic receptor (Wang et al., 2005), EGF receptor (Wang domain, an N-terminal coiled-coil domain and a C- et al., 2006), VEGF receptor-2 (Wang et al., 2009), terminal SH3 domain (Ihara et al., 2007). The LRP-1 (Lee et al., 2006), E-cadherin (Osumi et al., catalytic domain was structurally classfied as a 2009), alpha3beta1 integrin (Zhao et al., 2006), member of the GT-B group of VCAM and alpha4beta1 integrin (Li et al., 2008). glycosylatransferases. The binding affinity of the core fucose-deleted TGF-beta receptor to TGF-beta 1 is diminished in Expression fut8-null mice, resulting in the downregulation of FUT8 gene is widely expressed in human tissues TGF-beta 1 signaling (Wang et al., 2005). The (Martinez-Duncker et al., 2004). The FUT8 gene is unusual overexpression of matrix expressed at relatively high levels in the brain, metalloproteinases such as MMP-12 and MMP-13 placenta, lung, stomach, small intestine and is associated with the impaired receptor function, jejunum, while pancreas, uterus, kidney and urinary and has been proposed to cause the lung-destructive bladder exhibit moderate expression. The FUT8 phenotypes. The EGF receptor in fut8 null mice is gene is weakly expressed in the heart, ileum, colon also affected in terms of its binding affinity to EGF and spleen. On the other hand, the expression is not and EGF-induced phoshorylation (Wang et al., detectable in the normal liver (Miyoshi et al., 2006). These studies strongly suggest that FUT8 1997). 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%), Figure 3. The reaction catalysed by FUT8.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 413

FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

Takifugu (80.2%), Tetraodon (79.8%), Sea squirt fucosylated glycoproteins in hepatic cells. Because (23.2%), Fruit fly (43.7%), C. elegans (34.8%). the intracellular concentration of GDP-fucose is Eight cysteine residues in the catalytic domain are higher in HCC, as compared to a normal liver, conserved among these species, except for ciona chronic hepatitis and liver cirrhosis, this increase (Ihara et al., 2007). would be expected to facilitate core fucosyltion of FUT8 contains three short regions that are highly AFP (Noda et al., 2003). Core fucosylation appears conserved in FUT8, alpha1,2-, bacterial alpha1,6-, to serve as a sorting signal for a glycoprotein to be and protein O-fucosyltransferases (Oriol et al., directed to the bile, as revealed by the predominant 1999; Takahashi et al., 2000a; Chazalet et al., 2001; distribution of fucosylated glycoproteins in bile Martinez-Duncker et al., 2003). The structual rather than serum (Nakagawa et al., 2006). In fact, analysis has shown that these regions are located the levels of alpha-antitrypsin and alpha1-acid adjacent to one another in the Rossmann fold of glycoprotein, both of which are fucosylated FUT8 (Ihara et al., 2007). In addition, the C- glycoproteins, are quite low in the bile of Fut8-null terminal SH3 domain of FUT8 is structually similar mice. The loss of polarity in cancer cells is likely to to the typical SH3 domain that is found in many impair the regulated sorting, thus allowing proteins. abnormal secretion into the serum. Prognosis Mutations AFP-L3-positive HCC patients were reported to Note show a poor prognosis (Yamashita et al., 1996). One frame-shit mutation and 4 substitution mutants Ovarian cancer have been identified to date in various SNPs of the Note FUT8 gene. The frame shift mutant is due to the FUT8 activity and mRNA levels are highly and insertion of a T at position 2 of the codon for Val- specifically elavated in cases of ovarian serous 85, resulting in 85-VLEEQLVK-92 being change to adenocarcinoma, as compared to nomal ovary and 85-VFRRAACter-92. The four substitution mutants other types of epithelial ovarian carcinoma are K101Q, L153V, E181G and T267K. These (Takahashi et al., 2000b). In addition, core mutants are due to A being substituted by C at fucosylation levels in glycoproteins is also position 1 of codon 101, C to G at position 1 of significantlly increased in cases of serous codon 153, A to G at position 2 of codon 181, and adenocarcinoma tissues. C to A at posision 2 of codon 267, respectively. Effects of these substitutions on enzymatic activity Thyroid cancer are not currently known. Note The overexpression of FUT8 occurs in 33.3% of Implicated in cases of papillary carcinoma of the thyroid (Ito et al., 2003), although FUT8 was not expressed in Hepatocellular carcinoma (HCC) normal follicular cells. This overexpression was Note also shown to be correlated with tumor size and It is well known that the core fucosylation of alpha- lymph node metastasis. These phenomena were not fetoprotein (AFP) is implicated in the development observed in cases of follicular carcinoma and of HCC. AFP is a major fetal plasma protein, and anaplastic carcinoma. its expression is elevated in hepatic diseases such as Pancreatic cancer HCC, hepatitis and liver cirrhosis (Alpert et al., 1968; Ruoslahti et al., 1974). The AFP-L3 fraction Note was identified as the core-fucosylated isoform of Haptoglobin was identified as a highly fucosylated AFP. The elevation in serum and liver tissue was glycoprotein in the serum of patients with found to be specific to HCC, but was not observed pancreatic cancer (Okuyama et al., 2006). The in other liver diseases (Taketa, 1990; Aoyagi, 1995; increment of fucosylated haptoglobin was observed Miyoshi et al., 1999). in pancreatic cancer rather than other diseases such Thus, it appears that AFP-L3 could be used as a as HCC, liver cirrhosis, gastric cancer and colon marker for HCC. The FUT8 gene is not expressed cancer, and also appeared to be correlated with the in the normal adult liver, but is highly expressed in clinical stage. Structural analyses using lectin HCC tissue. Surprisingly, however, such an blotting and mass spectrometry showed that core elevation was also observed in liver cirrhosis in fucosylation as well as alpha1,3/4-fucosylation is spite of the absence of a concomitant increase in increased in haptoglobin from the serum of such AFP-L3 levels (Noda et al., 1998). This patients. In addition, it was shown that interleukin 6 discrepancy can be attributed to the difference in expressed in pancreatic cancer is a possible the synthesis of GDP-fucose, a glycosyl donor inducing factor for increasing the production of substrate for fucosyltransferases, including FUT8, fucosylated haptoglobin in the liver (Narisada et al., and by the altered intracellular sorting of 2008).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 414

FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

Colorectal cancer Miyoshi E, Noda K, Yamaguchi Y, Inoue S, Ikeda Y, Wang W, Ko JH, Uozumi N, Li W, Taniguchi N. The alpha1-6- Note fucosyltransferase gene and its biological significance. The enzymatic activity and protein expression of Biochim Biophys Acta. 1999 Dec 6;1473(1):9-20 FUT8 were increased in tumor tissues of human Oriol R, Mollicone R, Cailleau A, Balanzino L, Breton C. colorectal carcinoma, but not in healthy tissues Divergent evolution of fucosyltransferase genes from (Muinelo-Romay et al., 2008). This increment was vertebrates, invertebrates, and bacteria. Glycobiology. 1999 Apr;9(4):323-34 well observed in cases of male, polypoid growth, no regional lymph node metastasis and early clinical Yamaguchi Y, Fujii J, Inoue S, Uozumi N, Yanagidani S, Ikeda Y, Egashira M, Miyoshi O, Niikawa N, Taniguchi N. stage. In addition, immunohistochemical Mapping of the alpha-1,6-fucosyltransferase gene, FUT8, examination has demonstrated that FUT8 is to human chromosome 14q24.3. Cytogenet Cell Genet. expressed at higher levels in tumor tissues of 1999;84(1-2):58-60 colorectal carcinoma than in healthy and Takahashi T, Ikeda Y, Miyoshi E, Yaginuma Y, Ishikawa transitional tissues. M, Taniguchi N. alpha1,6fucosyltransferase is highly and specifically expressed in human ovarian serous Cystic fibrosis adenocarcinomas. Int J Cancer. 2000 Dec 15;88(6):914-9 Note Takahashi T, Ikeda Y, Tateishi A, Yamaguchi Y, Ishikawa Fucosylation is known to be increased in cystic M, Taniguchi N. A sequence motif involved in the donor fibrosis. The alpha1,6-fucosylation of a membrane substrate binding by alpha1,6-fucosyltransferase: the role of the conserved arginine residues. Glycobiology. 2000 glycoprotein is elevated in cystic fibrosis fibroblast May;10(5):503-10 (Wang et al., 1990). Yamaguchi Y, Ikeda Y, Takahashi T, Ihara H, Tanaka T, Sasho C, Uozumi N, Yanagidani S, Inoue S, Fujii J, References Taniguchi N. Genomic structure and promoter analysis of the human alpha1, 6-fucosyltransferase gene (FUT8). Alpert ME, Uriel J, de Nechaud B. Alpha-1 fetoglobulin in Glycobiology. 2000 Jun;10(6):637-43 the diagnosis of human hepatoma. N Engl J Med. 1968 May 2;278(18):984-6 Shields RL, Lai J, Keck R, O'Connell LY, Hong K, Meng YG, Weikert SH, Presta LG. Lack of fucose on human Ruoslahti E, Salaspuro M, Pihko H, Andersson L, Seppälä IgG1 N-linked oligosaccharide improves binding to human M. Serum alpha-fetoprotein: diagnostic significance in liver Fcgamma RIII and antibody-dependent cellular toxicity. J disease. Br Med J. 1974 Jun 8;2(5918):527-9 Biol Chem. 2002 Jul 26;277(30):26733-40 Taketa K. Alpha-fetoprotein: reevaluation in hepatology. Ito Y, Miyauchi A, Yoshida H, Uruno T, Nakano K, Hepatology. 1990 Dec;12(6):1420-32 Takamura Y, Miya A, Kobayashi K, Yokozawa T, Wang YM, Hare TR, Won B, Stowell CP, Scanlin TF, Glick Matsuzuka F, Taniguchi N, Matsuura N, Kuma K, Miyoshi MC, Hård K, van Kuik JA, Vliegenthart JF. Additional E. Expression of alpha1,6-fucosyltransferase (FUT8) in fucosyl residues on membrane glycoproteins but not a papillary carcinoma of the thyroid: its linkage to biological secreted glycoprotein from cystic fibrosis fibroblasts. Clin aggressiveness and anaplastic transformation. Cancer Chim Acta. 1990 May;188(3):193-210 Lett. 2003 Oct 28;200(2):167-72 Aoyagi Y. Carbohydrate-based measurements on alpha- Noda K, Miyoshi E, Gu J, Gao CX, Nakahara S, Kitada T, fetoprotein in the early diagnosis of hepatocellular Honke K, Suzuki K, Yoshihara H, Yoshikawa K, Kawano K, carcinoma. Glycoconj J. 1995 Jun;12(3):194-9 Tonetti M, Kasahara A, Hori M, Hayashi N, Taniguchi N. Relationship between elevated FX expression and Uozumi N, Yanagidani S, Miyoshi E, Ihara Y, Sakuma T, increased production of GDP-L-fucose, a common donor Gao CX, Teshima T, Fujii S, Shiba T, Taniguchi N. substrate for fucosylation in human hepatocellular Purification and cDNA cloning of porcine brain GDP-L- carcinoma and hepatoma cell lines. Cancer Res. 2003 Oct Fuc:N-acetyl-beta-D-glucosaminide alpha1-- 1;63(19):6282-9 >6fucosyltransferase. J Biol Chem. 1996 Nov 1;271(44):27810-7 Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda Y, Sakurada M, Uchida K, Anazawa H, Satoh M, Yamashita F, Tanaka M, Satomura S, Tanikawa K. Yamasaki M, Hanai N, Shitara K. The absence of fucose Prognostic significance of Lens culinaris agglutinin A- but not the presence of galactose or bisecting N- reactive alpha-fetoprotein in small hepatocellular acetylglucosamine of human IgG1 complex-type carcinomas. Gastroenterology. 1996 Oct;111(4):996-1001 oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J Biol Chem. Miyoshi E, Uozumi N, Noda K, Hayashi N, Hori M, 2003 Jan 31;278(5):3466-73 Taniguchi N. Expression of alpha1-6 fucosyltransferase in rat tissues and human cancer cell lines. Int J Cancer. 1997 Martinez-Duncker I, Michalski JC, Bauvy C, Candelier JJ, Sep 17;72(6):1117-21 Mennesson B, Codogno P, Oriol R, Mollicone R. Activity and tissue distribution of splice variants of alpha6- Yanagidani S, Uozumi N, Ihara Y, Miyoshi E, Yamaguchi fucosyltransferase in human embryogenesis. Glycobiology. N, Taniguchi N. Purification and cDNA cloning of GDP-L- 2004 Jan;14(1):13-25 Fuc:N-acetyl-beta-D-glucosaminide:alpha1-6 fucosyltransferase (alpha1-6 FucT) from human gastric Wang X, Inoue S, Gu J, Miyoshi E, Noda K, Li W, Mizuno- cancer MKN45 cells. J Biochem. 1997 Mar;121(3):626-32 Horikawa Y, Nakano M, Asahi M, Takahashi M, Uozumi N, Ihara S, Lee SH, Ikeda Y, Yamaguchi Y, Aze Y, Tomiyama Noda K, Miyoshi E, Uozumi N, Yanagidani S, Ikeda Y, Gao Y, Fujii J, Suzuki K, Kondo A, Shapiro SD, Lopez-Otin C, C, Suzuki K, Yoshihara H, Yoshikawa K, Kawano K, Kuwaki T, Okabe M, Honke K, Taniguchi N. Dysregulation Hayashi N, Hori M, Taniguchi N. Gene expression of of TGF-beta1 receptor activation leads to abnormal lung alpha1-6 fucosyltransferase in human hepatoma tissues: a development and emphysema-like phenotype in core possible implication for increased fucosylation of alpha- fetoprotein. Hepatology. 1998 Oct;28(4):944-52

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 415

FUT8 (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)) Ihara H, et al.

fucose-deficient mice. Proc Natl Acad Sci U S A. 2005 Nov Li W, Ishihara K, Yokota T, Nakagawa T, Koyama N, Jin J, 1;102(44):15791-6 Mizuno-Horikawa Y, Wang X, Miyoshi E, Taniguchi N, Kondo A. Reduced alpha4beta1 integrin/VCAM-1 Lee SH, Takahashi M, Honke K, Miyoshi E, Osumi D, interactions lead to impaired pre-B cell repopulation in Sakiyama H, Ekuni A, Wang X, Inoue S, Gu J, Kadomatsu alpha 1,6-fucosyltransferase deficient mice. Glycobiology. K, Taniguchi N. Loss of core fucosylation of low-density 2008 Jan;18(1):114-24 lipoprotein receptor-related protein-1 impairs its function, leading to the upregulation of serum levels of insulin-like Muinelo-Romay L, Vázquez-Martín C, Villar-Portela S, growth factor-binding protein 3 in Fut8-/- mice. J Biochem. Cuevas E, Gil-Martín E, Fernández-Briera A. Expression 2006 Mar;139(3):391-8 and enzyme activity of alpha(1,6)fucosyltransferase in human colorectal cancer. Int J Cancer. 2008 Aug Nakagawa T, Uozumi N, Nakano M, Mizuno-Horikawa Y, 1;123(3):641-6 Okuyama N, Taguchi T, Gu J, Kondo A, Taniguchi N, Miyoshi E. Fucosylation of N-glycans regulates the Narisada M, Kawamoto S, Kuwamoto K, Moriwaki K, secretion of hepatic glycoproteins into bile ducts. J Biol Nakagawa T, Matsumoto H, Asahi M, Koyama N, Miyoshi Chem. 2006 Oct 6;281(40):29797-806 E. Identification of an inducible factor secreted by pancreatic cancer cell lines that stimulates the production Okuyama N, Ide Y, Nakano M, Nakagawa T, Yamanaka K, of fucosylated haptoglobin in hepatoma cells. Biochem Moriwaki K, Murata K, Ohigashi H, Yokoyama S, Eguchi H, Biophys Res Commun. 2008 Dec 19;377(3):792-6 Ishikawa O, Ito T, Kato M, Kasahara A, Kawano S, Gu J, Taniguchi N, Miyoshi E. Fucosylated haptoglobin is a novel Osumi D, Takahashi M, Miyoshi E, Yokoe S, Lee SH, marker for pancreatic cancer: a detailed analysis of the Noda K, Nakamori S, Gu J, Ikeda Y, Kuroki Y, Sengoku K, oligosaccharide structure and a possible mechanism for Ishikawa M, Taniguchi N. Core fucosylation of E-cadherin fucosylation. Int J Cancer. 2006 Jun 1;118(11):2803-8 enhances cell-cell adhesion in human colon carcinoma WiDr cells. Cancer Sci. 2009 May;100(5):888-95 Taniguchi N, Miyoshi E, Gu J, Honke K, Matsumoto A. Decoding sugar functions by identifying target Takahashi M, Kuroki Y, Ohtsubo K, Taniguchi N. Core glycoproteins. Curr Opin Struct Biol. 2006 Oct;16(5):561-6 fucose and bisecting GlcNAc, the direct modifiers of the N- glycan core: their functions and target proteins. Carbohydr Wang X, Gu J, Ihara H, Miyoshi E, Honke K, Taniguchi N. Res. 2009 Aug 17;344(12):1387-90 Core fucosylation regulates epidermal growth factor receptor-mediated intracellular signaling. J Biol Chem. Wang X, Fukuda T, Li W, Gao CX, Kondo A, Matsumoto A, 2006 Feb 3;281(5):2572-7 Miyoshi E, Taniguchi N, Gu J. Requirement of Fut8 for the expression of vascular endothelial growth factor receptor- Zhao Y, Itoh S, Wang X, Isaji T, Miyoshi E, Kariya Y, 2: a new mechanism for the emphysema-like changes Miyazaki K, Kawasaki N, Taniguchi N, Gu J. Deletion of observed in Fut8-deficient mice. J Biochem. 2009 core fucosylation on alpha3beta1 integrin down-regulates May;145(5):643-51 its functions. J Biol Chem. 2006 Dec 15;281(50):38343-50 Ihara H, Ikeda Y, Toma S, Wang X, Suzuki T, Gu J, This article should be referenced as such: Miyoshi E, Tsukihara T, Honke K, Matsumoto A, Ihara H, Gao CX, Ikeda Y, Taniguchi N. FUT8 Nakagawa A, Taniguchi N. Crystal structure of mammalian (fucosyltransferase 8 (alpha (1,6) fucosyltransferase)). alpha1,6-fucosyltransferase, FUT8. Glycobiology. 2007 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):412- May;17(5):455-66 416.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 416

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI IGSF8ID45820ch1q23.txt

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, Description KCT-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 - splicing forms). ATP1A2 - ATP1A4 - CASQ1 - PEA15 -- Telomere (NCBI Map Viewer).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 417

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 cells (majority of B-cells, T-cells and natural killer cells, but not on monocytes, polynuclear cells and IGSF8-004, an alternative splicing form of platelets). CD316 is constitutively expressed on IGSF8/CD316, is predicted to have no protein plasmacytoid dendritic cells and on cord blood- product (Ensembl). derived Langerhans-like cells. Upon stimulation, CD316 is expressed on monocytes, monocytes Protein derived dendritic cells and myeloid dendritic cells. Description Localisation 613 amino acids, molecular weight is 65034 Da. Plasma membrane, cell-cell contacts, microvilli. Basal isoelectric point: 8.23 (PhosphoSitePlus). Function Expression 1. Suppresses cell movement and cell aggregation. CD316 mRNA is ubiquitously expressed in human 2. Regulates integrin alpha3beta1- and alpha4beta1- tissues, with high expression in brain, kidney, testis, dependent cell morphology and cell spreading. liver and placenta, with low expression in 3. May participate in the regulation of neurite peripheral blood cells, lung, and skeletal muscle. outgrowth and maintenance of the neural network CD316 protein is highly expressed in human brain in the adult brain. (cortex, white matter, hippocampus and 4. Interacts with its ligand, HSPA8, and may cerebellum), astrocytes, hepatocytes and lymphoid influence the behavior of dendritic cells and control

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 418

IGSF8 (immunoglobulin superfamily, member 8) Zhang YH, et al.

adaptive immune response. 5. Links tetraspanin web to the actin cytoskeleton Implicated in through direct associations with ezrin-radixin- Glioma or glioblastoma moesin proteins. 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 (Kolesnikova et al., 2009). to fuse with sperms. Hepatitis and liver cancer CD316 typically inhibits cell migration and Note negatively regulates cell proliferation. It associates Hepatitis C virus (HCV)-infected population has with tetraspanins CD9, CD81, and CD82 and likely higher risk of developing liver cancer. Ectopic contributes to various functions of these associated expression of EWI-2wint, i.e., EWI-2 without its N- tetraspanins. It also regulates the functions of terminus, can inhibit HCV entry and reduce HCV alpha3beta1 and alpha4beta1 integrins, probably infection (Rocha-Perugini et al., 2008). through its associated tetraspanins (Clark et al., 2001; Stipp et al., 2001; Stipp et al., 2003; Zhang et Autoimmune diseases al., 2003; Kolesnikova et al., 2004; Kolesnikova et Note al., 2009; Sala-Valdés et al., 2006). 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 q23, including type 2 diabetes mellitus (Murdoch et ectodomain that consists largely of four immunoglobulin al., 2003). domains, a transmembrane region, and a positively charged, 10-amino acid residue cytoplasmic tail. Glycosylation sites are found in the ectodomain and To be noted palmitoylation sites in the cytoplasmic domain. CD316 is constitutively palmitoylated and linked to actin cytoskeleton Note through direct association of its cytoplasmic domain with EWI-2wint, a cleavage product of EWI-2 in which ezrin-radixin-moesin proteins. CD316 associates with the first Ig-domain of the 4 extracellular Ig-domains tetraspanins such as CD9, CD81, and CD82. is cleaved off. Homology CD316 protein is conserved in chimpanzee, cow, References mouse, rat, and zebrafish and belongs to the EWI Clark KL, Zeng Z, Langford AL, Bowen SM, Todd SC. subfamily of Ig superfamily. Other human EWI PGRL is a major CD81-associated protein on lymphocytes subfamily proteins include FPRP/CD9P-1, IGSF3, and distinguishes a new family of cell surface proteins. J and CD101. Immunol. 2001 Nov 1;167(9):5115-21 Stipp CS, Kolesnikova TV, Hemler ME. EWI-2 is a major Mutations CD9 and CD81 partner and member of a novel Ig protein subfamily. J Biol Chem. 2001 Nov 2;276(44):40545-54 Note Charrin S, Le Naour F, Labas V, Billard M, Le Caer JP, Currently there is no known disease-related or Emile JF, Petit MA, Boucheix C, Rubinstein E. EWI-2 is a biologically significant mutation (see HGMD).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 419

IGSF8 (immunoglobulin superfamily, member 8) Zhang YH, et al.

new component of the tetraspanin web in hepatocytes and Kettner S, Kalthoff F, Graf P, Priller E, Kricek F, Lindley I, lymphoid cells. Biochem J. 2003 Jul 15;373(Pt 2):409-21 Schweighoffer T. EWI-2/CD316 is an inducible receptor of HSPA8 on human dendritic cells. Mol Cell Biol. 2007 Murdoch JN, Doudney K, Gerrelli D, Wortham N, Nov;27(21):7718-26 Paternotte C, Stanier P, Copp AJ. Genomic organization and embryonic expression of Igsf8, an immunoglobulin Rocha-Perugini V, Montpellier C, Delgrange D, Wychowski superfamily member implicated in development of the C, Helle F, Pillez A, Drobecq H, Le Naour F, Charrin S, nervous system and organ epithelia. Mol Cell Neurosci. Levy S, Rubinstein E, Dubuisson J, Cocquerel L. The 2003 Jan;22(1):62-74 CD81 partner EWI-2wint inhibits hepatitis C virus entry. PLoS One. 2008 Apr 2;3(4):e1866 Stipp CS, Kolesnikova TV, Hemler ME. EWI-2 regulates alpha3beta1 integrin-dependent cell functions on laminin- Charrin S, Yalaoui S, Bartosch B, Cocquerel L, Franetich 5. J Cell Biol. 2003 Dec 8;163(5):1167-77 JF, Boucheix C, Mazier D, Rubinstein E, Silvie O. The Ig domain protein CD9P-1 down-regulates CD81 ability to Zhang XA, Lane WS, Charrin S, Rubinstein E, Liu L. support Plasmodium yoelii infection. J Biol Chem. 2009 EWI2/PGRL associates with the metastasis suppressor Nov 13;284(46):31572-8 KAI1/CD82 and inhibits the migration of prostate cancer cells. Cancer Res. 2003 May 15;63(10):2665-74 Delandre C, Penabaz TR, Passarelli AL, Chapes SK, Clem RJ. Mutation of juxtamembrane cysteines in the Kolesnikova TV, Stipp CS, Rao RM, Lane WS, Luscinskas tetraspanin CD81 affects palmitoylation and alters FW, Hemler ME. EWI-2 modulates lymphocyte integrin interaction with other proteins at the cell surface. Exp Cell alpha4beta1 functions. Blood. 2004 Apr 15;103(8):3013-9 Res. 2009 Jul 1;315(11):1953-63 Little KD, Hemler ME, Stipp CS. Dynamic regulation of a Glazar AI, Evans JP. Immunoglobulin superfamily member GPCR-tetraspanin-G protein complex on intact cells: IgSF8 (EWI-2) and CD9 in fertilisation: evidence of distinct central role of CD81 in facilitating GPR56-Galpha q/11 functions for CD9 and a CD9-associated protein in association. Mol Biol Cell. 2004 May;15(5):2375-87 mammalian sperm-egg interaction. Reprod Fertil Dev. Sala-Valdés M, Ursa A, Charrin S, Rubinstein E, Hemler 2009;21(2):293-303 ME, Sánchez-Madrid F, Yáñez-Mó M. EWI-2 and EWI-F He ZY, Gupta S, Myles D, Primakoff P. Loss of surface link the tetraspanin web to the actin cytoskeleton through EWI-2 on CD9 null oocytes. Mol Reprod Dev. 2009 their direct association with ezrin-radixin-moesin proteins. Jul;76(7):629-36 J Biol Chem. 2006 Jul 14;281(28):19665-75 Kolesnikova TV, Kazarov AR, Lemieux ME, Lafleur MA, Yamada O, Tamura K, Yagihara H, Isotani M, Washizu T, Kesari S, Kung AL, Hemler ME. Glioblastoma inhibition by Bonkobara M. Neuronal expression of keratinocyte- cell surface immunoglobulin protein EWI-2, in vitro and in associated transmembrane protein-4, KCT-4, in mouse vivo. Neoplasia. 2009 Jan;11(1):77-86, 4p following 86 brain and its up-regulation by neurite outgrowth of Neuro- 2a cells. Neurosci Lett. 2006 Jan 16;392(3):226-30 This article should be referenced as such: Yang XH, Kovalenko OV, Kolesnikova TV, Andzelm MM, Zhang YH, Richardson MM, Zhang XA. IGSF8 Rubinstein E, Strominger JL, Hemler ME. Contrasting (immunoglobulin superfamily, member 8). Atlas Genet effects of EWI proteins, integrins, and protein Cytogenet Oncol Haematol. 2011; 15(5):417-420. palmitoylation on cell surface CD9 organization. J Biol Chem. 2006 May 5;281(18):12976-85

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 420

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI MIXL1ID47624ch1q42.txt

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 Location: 1q42.12 of length 393 bp and 306 bp, and one intron (Guo et Local order: MIXL1 is flanked on its 3' end by al., 2002; Sahr et al., 2002). Lin9. 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) 421

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 422

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 Disease on 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- marrow resulted in transplantable acute myeloid responsive regulation of dorsal-ventral patterning by the leukemia in all lethally irradiated recipient mice homeobox protein Mix.1. Nature. 1996 Jul 25;382(6589):357-60 after a latency period (Glaser et al., 2006). The following established AML cell lines Guo W, Chan AP, Liang H, Wieder ED, Molldrem JJ, et al. aberrantly express MIXL1: U937, KG1, and ML3 A human Mix-like homeobox gene MIXL shows functional similarity to Xenopus Mix.1. Blood. 2002 Jul;100(1):89-95 (Guo et al., 2002). Hwang HC, Martins CP, Bronkhorst Y, Randel E, Berns A, Chronic myeloid leukemia Fero M, Clurman BE. Identification of oncogenes collaborating with p27Kip1 loss by insertional mutagenesis Disease and high-throughput insertion site analysis. Proc Natl Acad MIXL1 is aberrantly expressed in the K562 Sci U S A. 2002 Aug 20;99(17):11293-8 established cell line (Guo et al., 2002). Sahr K, Dias DC, Sanchez R, Chen D, Chen SW, Gudas T-cell leukemia LJ, Baron MH. Structure, upstream promoter region, and functional domains of a mouse and human Mix paired-like Disease homeobox gene. Gene. 2002 May 29;291(1-2):135-47 The Mixl1 promoter in mouse was identified as a Glaser S, Metcalf D, Wu L, Hart AH, DiRago L, et al. site of viral insertion, using the Moloney murine Enforced expression of the homeobox gene Mixl1 impairs leukemia virus, which collaborates with loss of p27 hematopoietic differentiation and results in acute myeloid in induction of lymphomagenesis (Hwang et al., leukemia. Proc Natl Acad Sci U S A. 2006 Oct 2002). 31;103(44):16460-5 MIXL1 is aberrantly expressed in the following T- Guo W, Nagarajan L. Amino terminal tyrosine cell leukemia established lines: Jurkat, SKW-3, and phosphorylation of human MIXL1. J Mol Signal. 2006 Dec 5;1:6 CEM (Drakos et al., 2007; Guo et al., 2002). Drakos E, Rassidakis GZ, Leventaki V, Guo W, Medeiros B-cell leukemia LJ, Nagarajan L. Differential expression of the human Disease MIXL1 gene product in non-Hodgkin and Hodgkin lymphomas. Hum Pathol. 2007 Mar;38(3):500-7 MIXL1 is aberrantly expressed in the following B- cell leukemia established lines: NALM6, REH-1 This article should be referenced as such: (Drakos et al., 2007; Guo et al., 2002). Raymond A, Nagarajan L. MIXL1 (Mix1 homeobox-like 1 (Xenopus laevis)). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):421-423.

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

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI PEG3ID41690ch19q13.txt

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) 424

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

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 425

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

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

Relaix F, Wei X, Li W, Pan J, Lin Y, Bowtell DD, Sassoon This article should be referenced as such: DA, Wu X. Pw1/Peg3 is a potential cell death mediator and cooperates with Siah1a in p53-mediated apoptosis. Proc Yu Y, Feng W, Lu Z, Bast RC Jr. PEG3 (paternally Natl Acad Sci U S A. 2000 Feb 29;97(5):2105-10 expressed 3). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):424-426.

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

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI RPL10ID42148chXq28.txt

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

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

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

RPL10 (ribosomal protein L10) Goel M, Tamuli R

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 428

RPL10 (ribosomal protein L10) Goel M, Tamuli R

modulating disease mechanism for autism. Mol Psychiatry. Rastam M, Gillberg IC, Kopp S, Mouren-Simeoni MC, 2006 Dec;11(12):1073-84 Gillberg C, Leboyer M, Bourgeron T. An investigation of ribosomal protein L10 gene in autism spectrum disorders. Shen XJ, Ali-Fehmi R, Weng CR, Sarkar FH, Grignon D, BMC Med Genet. 2009 Jan 23;10:7 Liao DJ. Loss of heterozygosity and microsatellite instability at the Xq28 and the A/G heterozygosity of the This article should be referenced as such: QM gene are associated with ovarian cancer. Cancer Biol Ther. 2006 May;5(5):523-8 Goel M, Tamuli R. RPL10 (ribosomal protein L10). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):427-429. Gong X, Delorme R, Fauchereau F, Durand CM, Chaste P, Betancur C, Goubran-Botros H, Nygren G, Anckarsäter H,

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 429

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SNAI1ID452ch20q13.txt

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

Snail protein structure.

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

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

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 431

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

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

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

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

and clinicopathological parameters. Natsugoe et al. correlation between Snail and E-cadherin (2007) examined 194 cases with oesophageal expression. Schwock et al. (2010) examined a squamous cell carcinoma. 61.7% (84/194) were cohort of 46 cases of oral squamous cell carcinoma reported with positive staining. Snail expression including corresponding metastases. Nuclear Snail- was associated with deep invasion, increased lymph positivity equal or in excess of a 5% threshold was node metastasis, and advanced stage. No correlation observed in 10 tumors and 5 metastases which was found between Snail and E-cadherin corresponded to 12 cases. Individual Snail-positive expression. Usami et al. (2008) reported a cohort of tumor cells below this threshold, however, were 72 cases of oesophageal squamous cell carcinoma present more frequently and found in primary for which 38% (27/72) were considered positive. tumors of 30 patients. Snail expression in tumor Elevated Snail expression was found at the invasion cells in excess of 10% was rare, but associated with front, and was associated with lymphatic and poor outcome by univariate analysis. venous vessel invasion, lymph node metastasis and Neoplasms of the genitourinary tract tumor stage. Furthermore, a recent study by Kim et al. (2009) reported Snail positivity in 42.9% Note (245/571) of gastric carcinomas where it was 87 primary endometrioid-type adenocarcinomas of associated with invasion and lymph node the endometrium and 26 unrelated metastases were metastasis. Snail staining was an independent examined in a study by Blechschmidt et al. (2007). indicator of prognosis by multivariate analysis in Among the primary tumors and the metastases a this study. Fendrich et al., 2007 examined Snail proportion of 28.7% and 53.8% were reported with expression in neuroendocrine tumors of the ileum. positive Snail staining, respectively. Snail 59% (22/37) of the primary tumors and 6 of 7 liver immunoreactivity in metastases was found to metastases were reported with immunoreactivity for correlate with higher grade and reduced E-cadherin Snail. 53% (16/30) of the neuroendocrine tumors expression. A subsequent study by Blechschmidt et displayed positivity for Snail as well as Sonic al. (2008) on 48 primary ovarian neoplasms and 50 Hedgehog. Roy et al. (2005) found a proportion of metastases found Snail immunoreactivity in 37.5% 78% (46/59) cases with positive staining in their and 52%, respectively. A borderline significant study on colorectal cancers as well as a trend difference in overall survival with Snail expression towards increased presence of Snail in tumors with in metastases was noted. There was no correlation distant metastasis. A more recent study on between Snail and E-cadherin expression in this colorectal cancer by Franci et al. (2009) reported a study. A similar study by Jin et al. (2009) examined similarly high proportion of 79% (128/162) of cases 41 serous adenocarcinomas of the ovary with 14 with Snail immunoreactivity. Interestingly, in this matched metastases, 12 serous borderline tumors, 5 study a correlation between stromal Snail cystadenomas and 4 normal ovarian controls. There expression and decreased survival was found. was a range of Snail immunoreactivity with increased nuclear positivity noted in the carcinoma Neoplasms of the head and neck group. Tuhkanen et al. (2009) compared 74 ovarian Note carcinomas with 24 borderline tumors, 21 benign Yang et al. (2007) reported a proportion of 37.4% ovarian neoplasms and 14 normal controls. (n=147) of primary head and neck squamous cell Increased nuclear staining was noted with carcinomas with positive immunoreactivity for increasing malignancy both in the epithelial as well Snail. Snail expression was associated with lymph as the stromal compartment. 23% (17/74) of the node metastasis, and co-expression with Nijmegen ovarian carcinomas were reported to show focal breakage syndrome 1 (NBS1) indicated short Snail positivity. No association with metastasis-free period and overall survival. Another clinicopathological factors was seen in this study. study by Yang et al. (2008) reported a positive Heeboll et al., 2009 examined Snail in 327 prostate correlation between Snail and reduced metastasis- cancer specimens, 15 specimens with high-grade free and overall survival. Peinado et al. (2008) prostatic intraepithelial neoplasia (PIN), 30 examined a large cohort of laryngeal squamous cell specimens from patients with benign prostatic carcinomas for which 16% (40/251) were reported hyperplasia and 30 benign prostate tissue controls. Snail positive including 3% (8/251) high-positive. Approximately 50% of the prostate cancers were They found a correlation between Snail and LOXL2 found to have high Snail immunoreactivity expression, but no association between Snail and compared to only 7% of the high-grade PIN disease-free or overall survival. Zidar et al. (2008) specimens. Snail expression in this study was reported their findings on two cohorts of head and associated with Gleason score, but not with neck squamous cell carcinomas specifically progression or prognosis. Bruyere et al., 2009 distinguishing between spindle cell carcinomas and studied Snail expression in transitional cell those of moderately differentiated phenotype. 19/30 carcinoma of the bladder using a microarray of 87 of the spindle cell, but only 4/30 cases in the cases. Strong Snail positivity was found in 43.7% moderately differentiated group were found to and weak positivity in the remainder of the cases. display positive immunoreactivity. There was no Snail immunoreactivity in this study was prognostic

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 433

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

for tumor recurrence by uni- and multivariate analysis. To be noted Breast cancer Note Mouse model. Note A CombitTA conditional mouse model of Snail Zhou et al. (2004) reported a study on Snail expression has been described without expression in breast cancer which found positive morphological alterations, but associated with the staining in 56% (72/129) of their cases; 17 with low development of both epithelial and mesenchymal and 55 with high Snail immunoreactivity. Snail tumors (leukemias) (Perez-Mancera et al., 2005). correlated with GSK-3beta inhibition and E- Notably, suppression of the Snail transgene did not cadherin downregulation, and clinically with rescue the malignant phenotype, indicating that the metastasis in this study. alterations induced by Snail were irreversible. Endocrine neoplasms Note References Waldmann et al. (2008) reported their findings with Boulay JL, Dennefeld C, Alberga A. The Drosophila Snail expression in adrenocortical carcinomas developmental gene snail encodes a protein with nucleic obtained in a study including 26 primary tumors as acid binding fingers. Nature. 1987 Nov 26-Dec 2;330(6146):395-8 well as two lymph node and one liver metastases. 65% (17/26) primary tumors showed staining for Hay ED. Theory for epithelial-mesenchymal transformation Snail with strong positivity found at the invasion based on the "fixed cortex" cell motility model. Cell Motil Cytoskeleton. 1989;14(4):455-7 front of 7 tumors and in 2 of 3 metastases. Snail positivity was associated with advanced stage, Leptin M. twist and snail as positive and negative regulators during Drosophila mesoderm development. decreased survival and higher risk for distant Genes Dev. 1991 Sep;5(9):1568-76 metastases. The same group reported a study on Snail in pheochromocytomas including 44 primary Cohen ME, Yin M, Paznekas WA, Schertzer M, Wood S, Jabs EW. Human SLUG gene organization, expression, tumors, 3 lymph node and 2 peritoneal metastases and chromosome map location on 8q. Genomics. 1998 (Waldmann et al., 2009). Snail positivity was Aug 1;51(3):468-71 reported for 28% (13/47) cases, and positive Paznekas WA, Okajima K, Schertzer M, Wood S, Jabs staining was associated with malignant behaviour. EW. Genomic organization, expression, and chromosome Hardy et al. (2007) published their results focused location of the human SNAIL gene (SNAI1) and a related on thyroid neoplasms. 18/31 follicular and 28/32 processed pseudogene (SNAI1P). Genomics. 1999 Nov papillary thyroid cancers as well as all of 4 lymph 15;62(1):42-9 node metastases of papillary thyroid cancer were Twigg SR, Wilkie AO. Characterisation of the human snail found to stain positive for Snail whereas normal (SNAI1) gene and exclusion as a major disease gene in craniosynostosis. Hum Genet. 1999 Oct;105(4):320-6 thyroid tissue was negative. Snail staining was reported to be restricted to the invasive front and Batlle E, Sancho E, Francí C, Domínguez D, Monfar M, associated with a concomitant reduction in E- Baulida J, García De Herreros A. The transcription factor snail is a repressor of E-cadherin gene expression in cadherin reactivity. The authors of this study also epithelial tumour cells. Nat Cell Biol. 2000 Feb;2(2):84-9 included their findings from a Combi-TA mouse Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco model showing development of papillary thyroid MJ, del Barrio MG, Portillo F, Nieto MA. The transcription carcinomas. Fendrich et al. (2009) recently factor snail controls epithelial-mesenchymal transitions by published results of a study focused on parathyroid repressing E-cadherin expression. Nat Cell Biol. 2000 neoplasms including 9 cases of parathyroid Feb;2(2):76-83 carcinoma, 25 adenomas and 25 cases of LaBonne C, Bronner-Fraser M. Snail-related transcriptional hyperplasia. Snail staining was positive in all cases repressors are required in Xenopus for both the induction of hyperplasia and 22/25 adenomas. In carcinomas of the neural crest and its subsequent migration. Dev Biol. 2000 May 1;221(1):195-205 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 Mesenchymal neoplasms breast carcinoma: modification of the two-hit hypothesis of tumor suppressor gene. Oncogene. 2001 Jun Note 28;20(29):3814-23 Franci et al. (2006) studied Snail in a series of Blanco MJ, Moreno-Bueno G, Sarrio D, Locascio A, Cano different neoplasm which included sarcomas and A, Palacios J, Nieto MA. Correlation of Snail expression infantile fibromatosis as well as epithelial with histological grade and lymph node status in breast neoplasms (squamous cell carcinoma of the uterine carcinomas. Oncogene. 2002 May 9;21(20):3241-6 cervix and adenocarcinoma of the colon). High Nieto MA. The snail superfamily of zinc-finger transcription Snail expression was present in fibrosarcomas and factors. Nat Rev Mol Cell Biol. 2002 Mar;3(3):155-66 other sarcomas. Snail expression in neoplasms of Rosivatz E, Becker I, Specht K, Fricke E, Luber B, Busch epithelial origin was restricted to the tumor-stroma R, Höfler H, Becker KF. Differential expression of the interface. epithelial-mesenchymal transition regulators snail, SIP1,

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

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

and twist in gastric cancer. Am J Pathol. 2002 differentiation program. Cancer Res. 2005 Jul Nov;161(5):1881-91 15;65(14):6237-44 Thiery JP. Epithelial-mesenchymal transitions in tumour Elloul S, Elstrand MB, Nesland JM, Tropé CG, Kvalheim progression. Nat Rev Cancer. 2002 Jun;2(6):442-54 G, Goldberg I, Reich R, Davidson B. Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease Domínguez D, Montserrat-Sentís B, Virgós-Soler A, Guaita aggressiveness in metastatic ovarian and breast S, Grueso J, Porta M, Puig I, Baulida J, Francí C, García carcinoma. Cancer. 2005 Apr 15;103(8):1631-43 de Herreros A. Phosphorylation regulates the subcellular location and activity of the snail transcriptional repressor. Miyoshi A, Kitajima Y, Kido S, Shimonishi T, Matsuyama Mol Cell Biol. 2003 Jul;23(14):5078-89 S, Kitahara K, Miyazaki K. Snail accelerates cancer invasion by upregulating MMP expression and is Imai T, Horiuchi A, Wang C, Oka K, Ohira S, Nikaido T, associated with poor prognosis of hepatocellular Konishi I. Hypoxia attenuates the expression of E-cadherin carcinoma. Br J Cancer. 2005 Jan 31;92(2):252-8 via up-regulation of SNAIL in ovarian carcinoma cells. Am J Pathol. 2003 Oct;163(4):1437-47 Moody SE, Perez D, Pan TC, Sarkisian CJ, Portocarrero CP, Sterner CJ, Notorfrancesco KL, Cardiff RD, Chodosh Katoh M, Katoh M. Identification and characterization of LA. The transcriptional repressor Snail promotes human SNAIL3 (SNAI3) gene in silico. Int J Mol Med. 2003 mammary tumor recurrence. Cancer Cell. 2005 Mar;11(3):383-8 Sep;8(3):197-209 Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A. Peinado H, Del Carmen Iglesias-de la Cruz M, Olmeda D, Targeted disruption of TGF-beta1/Smad3 signaling Csiszar K, Fong KS, Vega S, Nieto MA, Cano A, Portillo F. protects against renal tubulointerstitial fibrosis induced by A molecular role for lysyl oxidase-like 2 enzyme in snail unilateral ureteral obstruction. J Clin Invest. 2003 regulation and tumor progression. EMBO J. 2005 Oct Nov;112(10):1486-94 5;24(19):3446-58 Sugimachi K, Tanaka S, Kameyama T, Taguchi K, Aishima Peña C, García JM, Silva J, García V, Rodríguez R, S, Shimada M, Sugimachi K, Tsuneyoshi M. Alonso I, Millán I, Salas C, de Herreros AG, Muñoz A, Transcriptional repressor snail and progression of human Bonilla F. E-cadherin and vitamin D receptor regulation by hepatocellular carcinoma. Clin Cancer Res. 2003 SNAIL and ZEB1 in colon cancer: clinicopathological Jul;9(7):2657-64 correlations. Hum Mol Genet. 2005 Nov 15;14(22):3361-70 Yáñez-Mó M, Lara-Pezzi E, Selgas R, Ramírez-Huesca M, Pérez-Mancera PA, Pérez-Caro M, González-Herrero I, Domínguez-Jiménez C, Jiménez-Heffernan JA, Aguilera A, Flores T, Orfao A, de Herreros AG, Gutiérrez-Adán A, Sánchez-Tomero JA, Bajo MA, Alvarez V, Castro MA, del Pintado B, Sagrera A, Sánchez-Martín M, Sánchez-García Peso G, Cirujeda A, Gamallo C, Sánchez-Madrid F, I. Cancer development induced by graded expression of López-Cabrera M. Peritoneal dialysis and epithelial-to- Snail in mice. Hum Mol Genet. 2005 Nov 15;14(22):3449- mesenchymal transition of mesothelial cells. N Engl J Med. 61 2003 Jan 30;348(5):403-13 Roy HK, Smyrk TC, Koetsier J, Victor TA, Wali RK. The Kajita M, McClinic KN, Wade PA. Aberrant expression of transcriptional repressor SNAIL is overexpressed in human the transcription factors snail and slug alters the response colon cancer. Dig Dis Sci. 2005 Jan;50(1):42-6 to genotoxic stress. Mol Cell Biol. 2004 Sep;24(17):7559- 66 Yang Z, Rayala S, Nguyen D, Vadlamudi RK, Chen S, Kumar R. Pak1 phosphorylation of snail, a master Pálmer HG, Larriba MJ, García JM, Ordóñez-Morán P, regulator of epithelial-to-mesenchyme transition, Peña C, Peiró S, Puig I, Rodríguez R, de la Fuente R, modulates snail's subcellular localization and functions. Bernad A, Pollán M, Bonilla F, Gamallo C, de Herreros Cancer Res. 2005 Apr 15;65(8):3179-84 AG, Muñoz A. The transcription factor SNAIL represses vitamin D receptor expression and responsiveness in Francí C, Takkunen M, Dave N, Alameda F, Gómez S, human colon cancer. Nat Med. 2004 Sep;10(9):917-9 Rodríguez R, Escrivà M, Montserrat-Sentís B, Baró T, Garrido M, Bonilla F, Virtanen I, García de Herreros A. Peinado H, Ballestar E, Esteller M, Cano A. Snail mediates Expression of Snail protein in tumor-stroma interface. E-cadherin repression by the recruitment of the Oncogene. 2006 Aug 24;25(37):5134-44 Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol. 2004 Jan;24(1):306-19 Grotegut S, von Schweinitz D, Christofori G, Lehembre F. Hepatocyte growth factor induces cell scattering through Saito T, Oda Y, Kawaguchi K, Sugimachi K, Yamamoto H, MAPK/Egr-1-mediated upregulation of Snail. EMBO J. Tateishi N, Tanaka K, Matsuda S, Iwamoto Y, Ladanyi M, 2006 Aug 9;25(15):3534-45 Tsuneyoshi M. E-cadherin mutation and Snail overexpression as alternative mechanisms of E-cadherin Peiró S, Escrivà M, Puig I, Barberà MJ, Dave N, Herranz inactivation in synovial sarcoma. Oncogene. 2004 Nov N, Larriba MJ, Takkunen M, Francí C, Muñoz A, Virtanen I, 11;23(53):8629-38 Baulida J, García de Herreros A. Snail1 transcriptional repressor binds to its own promoter and controls its Takeno S, Noguchi T, Fumoto S, Kimura Y, Shibata T, expression. Nucleic Acids Res. 2006;34(7):2077-84 Kawahara K. E-cadherin expression in patients with esophageal squamous cell carcinoma: promoter Rosivatz E, Becker KF, Kremmer E, Schott C, hypermethylation, Snail overexpression, and Blechschmidt K, Höfler H, Sarbia M. Expression and clinicopathologic implications. Am J Clin Pathol. 2004 nuclear localization of Snail, an E-cadherin repressor, in Jul;122(1):78-84 adenocarcinomas of the 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 phosphorylation in control of epithelial-mesenchymal H, Kremer M, Becker KF. The E-cadherin repressor snail transition. Nat Cell Biol. 2004 Oct;6(10):931-40 plays a role in tumor progression of endometrioid adenocarcinomas. Diagn Mol Pathol. 2007 Dec;16(4):222- De Craene B, Gilbert B, Stove C, Bruyneel E, van Roy F, 8 Berx G. The transcription factor snail induces tumor cell invasion through modulation of the epithelial cell

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 435

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

Fendrich V, Waldmann J, Esni F, Ramaswamy A, Expression of the zinc-finger transcription factor Snail in Mullendore M, Buchholz M, Maitra A, Feldmann G. Snail adrenocortical carcinoma is associated with decreased and Sonic Hedgehog activation in neuroendocrine tumors survival. Br J Cancer. 2008 Dec 2;99(11):1900-7 of the ileum. Endocr Relat Cancer. 2007 Sep;14(3):865-74 Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu Hardy RG, Vicente-Dueñas C, González-Herrero I, CJ, Teng SC, Wu KJ. Direct regulation of TWIST by HIF- Anderson C, Flores T, Hughes S, Tselepis C, Ross JA, 1alpha promotes metastasis. Nat Cell Biol. 2008 Sánchez-García I. Snail family transcription factors are Mar;10(3):295-305 implicated in thyroid carcinogenesis. Am J Pathol. 2007 Sep;171(3):1037-46 Zidar N, Gale N, Kojc N, Volavsek M, Cardesa A, Alos L, Höfler H, Blechschmidt K, Becker KF. Cadherin-catenin Natsugoe S, Uchikado Y, Okumura H, Matsumoto M, complex and transcription factor Snail-1 in spindle cell Setoyama T, Tamotsu K, Kita Y, Sakamoto A, Owaki T, carcinoma of the head and neck. Virchows Arch. 2008 Ishigami S, Aikou T. Snail plays a key role in E-cadherin- Sep;453(3):267-74 preserved esophageal squamous cell carcinoma. Oncol Rep. 2007 Mar;17(3):517-23 Barrallo-Gimeno A, Nieto MA. Evolutionary history of the Snail/Scratch superfamily. Trends Genet. 2009 Palmer MB, Majumder P, Green MR, Wade PA, Boss JM. Jun;25(6):248-52 A 3' enhancer controls snail expression in melanoma cells. Cancer Res. 2007 Jul 1;67(13):6113-20 Fendrich V, Waldmann J, Feldmann G, Schlosser K, König A, Ramaswamy A, Bartsch DK, Karakas E. Unique Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH expression pattern of the EMT markers Snail, Twist and E- factors in tumour progression: an alliance against the cadherin in benign and malignant parathyroid neoplasia. epithelial phenotype? Nat Rev Cancer. 2007 Jun;7(6):415- Eur J Endocrinol. 2009 Apr;160(4):695-703 28 Francí C, Gallén M, Alameda F, Baró T, Iglesias M, Yang MH, Chang SY, Chiou SH, Liu CJ, Chi CW, Chen Virtanen I, García de Herreros A. Snail1 protein in the PM, Teng SC, Wu KJ. Overexpression of NBS1 induces stroma as a new putative prognosis marker for colon epithelial-mesenchymal transition and co-expression of tumours. PLoS One. 2009;4(5):e5595 NBS1 and Snail 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. Blechschmidt K, Sassen S, Schmalfeldt B, Schuster T, APMIS. 2009 Mar;117(3):196-204 Höfler H, Becker KF. The E-cadherin repressor Snail is associated with lower overall survival of ovarian cancer Jayachandran A, Königshoff M, Yu H, Rupniewska E, patients. Br J Cancer. 2008 Jan 29;98(2):489-95 Hecker M, Klepetko W, Seeger W, Eickelberg O. SNAI transcription factors mediate epithelial-mesenchymal Herranz N, Pasini D, Díaz VM, Francí C, Gutierrez A, Dave transition in lung fibrosis. Thorax. 2009 Dec;64(12):1053- N, Escrivà M, Hernandez-Muñoz I, Di Croce L, Helin K, 61 García de Herreros A, Peiró S. Polycomb complex 2 is required for E-cadherin repression by the Snail1 Kalluri R, Weinberg RA. The basics of epithelial- transcription factor. Mol Cell Biol. 2008 Aug;28(15):4772- mesenchymal transition. J Clin Invest. 2009 81 Jun;119(6):1420-8 Higashikawa K, Yoneda S, Taki M, Shigeishi H, Ono S, Kim MA, Lee HS, Lee HE, Kim JH, Yang HK, Kim WH. Tobiume K, Kamata N. Gene expression profiling to Prognostic importance of epithelial-mesenchymal identify genes associated with high-invasiveness in human transition-related protein expression in gastric carcinoma. squamous cell carcinoma with epithelial-to-mesenchymal Histopathology. 2009 Mar;54(4):442-51 transition. Cancer Lett. 2008 Jun 18;264(2):256-64 Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou Cancer metastasis is accelerated through AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, immunosuppression during Snail-induced EMT of cancer Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA. cells. Cancer Cell. 2009 Mar 3;15(3):195-206 The epithelial-mesenchymal transition generates cells with Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, properties of stem cells. Cell. 2008 May 16;133(4):704-15 Chaskar PD, Doiphode RY, Bapat SA. Snail and slug Morel AP, Lièvre M, Thomas C, Hinkal G, Ansieau S, mediate radioresistance and chemoresistance by Puisieux A. Generation of breast cancer stem cells through antagonizing p53-mediated apoptosis and acquiring a epithelial-mesenchymal transition. PLoS One. 2008 Aug stem-like phenotype in ovarian cancer cells. Stem Cells. 6;3(8):e2888 2009 Sep;27(9):2059-68 Peinado H, Moreno-Bueno G, Hardisson D, Pérez-Gómez Mingot JM, Vega S, Maestro B, Sanz JM, Nieto MA. E, Santos V, Mendiola M, de Diego JI, Nistal M, Quintanilla Characterization of Snail nuclear import pathways as M, Portillo F, Cano A. Lysyl oxidase-like 2 as a new poor representatives of C2H2 zinc finger transcription factors. J prognosis marker of squamous cell carcinomas. Cancer Cell Sci. 2009 May 1;122(Pt 9):1452-60 Res. 2008 Jun 15;68(12):4541-50 Peña C, García JM, Larriba MJ, Barderas R, Gómez I, Thuault S, Tan EJ, Peinado H, Cano A, Heldin CH, Herrera M, García V, Silva J, Domínguez G, Rodríguez R, Moustakas A. HMGA2 and Smads co-regulate SNAIL1 Cuevas J, de Herreros AG, Casal JI, Muñoz A, Bonilla F. expression during induction of epithelial-to-mesenchymal SNAI1 expression in colon cancer related with CDH1 and transition. J Biol Chem. 2008 Nov 28;283(48):33437-46 VDR downregulation in normal adjacent tissue. Oncogene. 2009 Dec 10;28(49):4375-85 Usami Y, Satake S, Nakayama F, Matsumoto M, Ohnuma K, Komori T, Semba S, Ito A, Yokozaki H. Snail-associated Tuhkanen H, Soini Y, Kosma VM, Anttila M, Sironen R, epithelial-mesenchymal transition promotes oesophageal Hämäläinen K, Kukkonen L, Virtanen I, Mannermaa A. squamous cell carcinoma motility and progression. J Nuclear expression of Snail1 in borderline and malignant Pathol. 2008 Jul;215(3):330-9 epithelial ovarian tumours is associated with tumour progression. BMC Cancer. 2009 Aug 20;9:289 Waldmann J, Feldmann G, Slater EP, Langer P, Buchholz M, Ramaswamy A, Saeger W, Rothmund M, Fendrich V. Waldmann J, Slater EP, Langer P, Buchholz M, Ramaswamy A, Walz MK, Schmid KW, Feldmann G,

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 436

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

Bartsch DK, Fendrich V. Expression of the transcription Lin Y, Wu Y, Li J, Dong C, Ye X, Chi YI, Evers BM, Zhou factor snail and its target gene twist are associated with BP. The SNAG domain of Snail1 functions as a molecular malignancy in pheochromocytomas. Ann Surg Oncol. 2009 hook for recruiting lysine-specific demethylase 1. EMBO J. Jul;16(7):1997-2005 2010 Jun 2;29(11):1803-16 Wu Y, Evers BM, Zhou BP. Small C-terminal domain MacPherson MR, Molina P, Souchelnytskyi S, Wernstedt phosphatase enhances snail activity through C, Martin-Pérez J, Portillo F, Cano A. Phosphorylation of dephosphorylation. J Biol Chem. 2009 Jan 2;284(1):640-8 serine 11 and serine 92 as new positive regulators of human Snail1 function: potential involvement of casein Bruyere F, Namdarian B, Corcoran NM, Pedersen J, kinase-2 and the cAMP-activated kinase protein kinase A. Ockrim J, Voelzke BB, Mete U, Costello AJ, Hovens CM. Mol Biol Cell. 2010 Jan 15;21(2):244-53 Snail expression is an independent predictor of tumor recurrence in superficial bladder cancers. Urol Oncol. 2010 Schwock J, Bradley G, Ho JC, Perez-Ordonez B, Hedley Nov-Dec;28(6):591-6 DW, Irish JC, Geddie WR. SNAI1 expression and the mesenchymal phenotype: an immunohistochemical study de Herreros AG, Peiró S, Nassour M, Savagner P. Snail performed on 46 cases of oral squamous cell carcinoma. family regulation and epithelial mesenchymal transitions in BMC Clin Pathol. 2010 Feb 5;10:1 breast cancer progression. J Mammary Gland Biol Neoplasia. 2010 Jun;15(2):135-47 Viñas-Castells R, Beltran M, Valls G, Gómez I, García JM, Montserrat-Sentís B, Baulida J, Bonilla F, de Herreros AG, Hou Z, Peng H, White DE, Wang P, Lieberman PM, Díaz VM. The hypoxia-controlled FBXL14 ubiquitin ligase Halazonetis T, Rauscher FJ 3rd. 14-3-3 binding sites in the targets SNAIL1 for proteasome degradation. J Biol Chem. snail protein are essential for snail-mediated transcriptional 2010 Feb 5;285(6):3794-805 repression and epithelial-mesenchymal differentiation. Cancer Res. 2010 Jun 1;70(11):4385-93 This article should be referenced as such: Jin H, Yu Y, Zhang T, Zhou X, Zhou J, Jia L, Wu Y, Zhou Schwock J, Geddie WR. SNAI1 (snail homolog 1 BP, Feng Y. Snail is critical for tumor growth and (Drosophila)). Atlas Genet Cytogenet Oncol Haematol. metastasis of ovarian carcinoma. Int J Cancer. 2010 May 2011; 15(5):430-437. 1;126(9):2102-11

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 437

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI VAV3ID42782ch1p13.txt

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

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) 438

VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

LMO2, GATA-1, GCNF-2, E47, GCNF-1, PAX-5, sequentially of a single calponin homology domain POU2F1, and FOXO1A (information obtained from encompassing residues 1-119, an acidic domain, a data deposited in Genecard database through use of DBL homology domain which confers GEF SABiosciences' text mining application and the function. The DBL homology domain is comprised UCSC genome browser). It is worth mentioning of residues 192-371, followed by a pleckstrin that the gene locus is complex and could potentially homology domain, spanning residues 400-502 a produce up to 13 different isoforms resulting from cysteine rich domain (also termed a zinc finger alternative splicing and alternate promoter usage domain) comprising residues 513-562 and two SH3 (Thierry-Mieg and Thierry-Mieg, 2006). domains flanking a single SH2 domain. The SH3- Vav3 can be modified posttranslationally by SH2-SH3 cassette comprises the C terminal portion phosphorylation. Phosphorylation site prediction of Vav3 and extends from the N terminal SH3 identifies phosphorylation sites at T131, S134, domain (residues 592-660), to the C terminal SH3 Y141, Y173, S511, T606 and Y797. Sites residing domain (residues 788-847) and includes the in the N terminal region have been shown to intervening SH2 domain (residues 672-766) regulate activation of Vav3 GEF function (Movilla (Trenkle et al., 2000). Residing within the N and Bustelo, 1999). In the unphosphorylated state, terminal SH3 domain is a proline rich region which the GEF domain is prevented from physical may be involved in facilitating intramolecular association with Rho proteins by the Vav3 N interactions between the C terminal regions (our terminal domains. These domains (calponin unpublished observations). 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 Rho proteins to the GEF domain Figure 3. Schematic showing inactive (top panel) and (Movilla and Bustelo, 1999; Yu et al., 2010). active (bottom panel) conformations of Vav3. Movement of Tyrosine 173 in particular is a critical residue in the N terminal regions to allow RhoGTPase access to the DH domain is regulated by phosphorylation events. this process (Llorca et al., 2005; Yu et al., 2010). Consistent with an autoinhibitory role of the N Expression terminal regions, removal of both the calponin Vav3 is broadly expressed but with highest levels in homology and the acidic domains results in cells of hematopoietic lineages (Trenkle et al., constitutive activation of Vav3 GEF function 2000). (Movilla and Bustelo, 1999; Zeng et al., 2000; Zugaza et al., 2002). Localisation Vav3 is located predominantly in the cytoplasm, Protein and is often recruited to the membrane upon activation of the various cell surface receptors that are coupled to Vav3 phosphorylation (Zeng et al., 2000).

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 439

VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 440

VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 441

VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

roles in mediating platelet activation by collagen. J Biol Pearce AC, McCarty OJ, Calaminus SD, Vigorito E, Turner Chem. 2004 Dec 24;279(52):53955-62 M, Watson SP. Vav family proteins are required for optimal regulation of PLCgamma2 by integrin alphaIIbbeta3. Aoki K, Nakamura T, Fujikawa K, Matsuda M. Local Biochem J. 2007 Feb 1;401(3):753-61 phosphatidylinositol 3,4,5-trisphosphate accumulation recruits Vav2 and Vav3 to activate Rac1/Cdc42 and initiate Fang WB, Brantley-Sieders DM, Hwang Y, Ham AJ, Chen neurite outgrowth in nerve growth factor-stimulated PC12 J. Identification and functional analysis of phosphorylated cells. Mol Biol Cell. 2005 May;16(5):2207-17 tyrosine residues within EphA2 receptor tyrosine kinase. J Biol Chem. 2008 Jun 6;283(23):16017-26 Best CJ, Gillespie JW, Yi Y, Chandramouli GV, Perlmutter MA, Gathright Y, Erickson HS, Georgevich L, Tangrea MA, Lee K, Liu Y, Mo JQ, Zhang J, Dong Z, Lu S. Vav3 Duray PH, González S, Velasco A, Linehan WM, Matusik oncogene activates estrogen receptor and its RJ, Price DK, Figg WD, Emmert-Buck MR, Chuaqui RF. overexpression may be involved in human breast cancer. Molecular alterations in primary prostate cancer after BMC Cancer. 2008 Jun 2;8:158 androgen ablation therapy. Clin Cancer Res. 2005 Oct 1;11(19 Pt 1):6823-34 Liu Y, Mo JQ, Hu Q, Boivin G, Levin L, Lu S, Yang D, Dong Z, Lu S. Targeted overexpression of vav3 oncogene Couceiro JR, Martín-Bermudo MD, Bustelo XR. in prostatic epithelium induces nonbacterial prostatitis and Phylogenetic conservation of the regulatory and functional prostate cancer. Cancer Res. 2008 Aug 1;68(15):6396-406 properties of the Vav oncoprotein family. Exp Cell Res. 2005 Aug 15;308(2):364-80 Lyons LS, Rao S, Balkan W, Faysal J, Maiorino CA, Burnstein KL. Ligand-independent activation of androgen Faccio R, Teitelbaum SL, Fujikawa K, Chappel J, Zallone receptors by Rho GTPase signaling in prostate cancer. A, Tybulewicz VL, Ross FP, Swat W. Vav3 regulates Mol Endocrinol. 2008 Mar;22(3):597-608 osteoclast function and bone mass. Nat Med. 2005 Mar;11(3):284-90 Ouyang X, Jessen WJ, Al-Ahmadie H, Serio AM, Lin Y, Shih WJ, Reuter VE, Scardino PT, Shen MM, Aronow BJ, Llorca O, Arias-Palomo E, Zugaza JL, Bustelo XR. Global Vickers AJ, Gerald WL, Abate-Shen C. Activator protein-1 conformational rearrangements during the activation of the transcription factors are associated with progression and GDP/GTP exchange factor Vav3. EMBO J. 2005 Apr recurrence of prostate cancer. Cancer Res. 2008 Apr 6;24(7):1330-40 1;68(7):2132-44 Maier LM, Smyth DJ, Vella A, Payne F, Cooper JD, Pask Salhia B, Tran NL, Chan A, Wolf A, Nakada M, Rutka F, R, Lowe C, Hulme J, Smink LJ, Fraser H, Moule C, Hunter Ennis M, McDonough WS, Berens ME, Symons M, Rutka KM, Chamberlain G, Walker N, Nutland S, Undlien DE, JT. The guanine nucleotide exchange factors trio, Ect2, Rønningen KS, Guja C, Ionescu-Tîrgoviste C, Savage DA, and Vav3 mediate the invasive behavior of glioblastoma. Strachan DP, Peterson LB, Todd JA, Wicker LS, Twells Am J Pathol. 2008 Dec;173(6):1828-38 RC. Construction and analysis of tag single nucleotide polymorphism maps for six human-mouse orthologous Brantley-Sieders DM, Zhuang G, Vaught D, Freeman T, candidate genes in type 1 diabetes. BMC Genet. 2005 Feb Hwang Y, Hicks D, Chen J. Host deficiency in Vav2/3 18;6:9 guanine nucleotide exchange factors impairs tumor growth, survival, and angiogenesis in vivo. Mol Cancer Sakakura C, Hasegawa K, Miyagawa K, Nakashima S, Res. 2009 May;7(5):615-23 Yoshikawa T, Kin S, Nakase Y, Yazumi S, Yamagishi H, Okanoue T, Chiba T, Hagiwara A. Possible involvement of Khodosevich K, Seeburg PH, Monyer H. Major signaling RUNX3 silencing in the peritoneal metastases of gastric pathways in migrating neuroblasts. Front Mol Neurosci. cancers. Clin Cancer Res. 2005 Sep 15;11(18):6479-88 2009;2:7 Dong Z, Liu Y, Lu S, Wang A, Lee K, Wang LH, Revelo M, Malhotra S, Kovats S, Zhang W, Coggeshall KM. B cell Lu S. Vav3 oncogene is overexpressed and regulates cell antigen receptor endocytosis and antigen presentation to T growth and androgen receptor activity in human prostate cells require Vav and dynamin. J Biol Chem. 2009 Sep cancer. Mol Endocrinol. 2006 Oct;20(10):2315-25 4;284(36):24088-97 Hunter SG, Zhuang G, Brantley-Sieders D, Swat W, Sindrilaru A, Peters T, Schymeinsky J, Oreshkova T, Cowan CW, Chen J. Essential role of Vav family guanine Wang H, Gompf A, Mannella F, Wlaschek M, Sunderkötter nucleotide exchange factors in EphA receptor-mediated C, Rudolph KL, Walzog B, Bustelo XR, Fischer KD, angiogenesis. Mol Cell Biol. 2006 Jul;26(13):4830-42 Scharffetter-Kochanek K. Wound healing defect of Vav3-/- mice due to impaired {beta}2-integrin-dependent Lyons LS, Burnstein KL. Vav3, a Rho GTPase guanine macrophage phagocytosis of apoptotic neutrophils. Blood. nucleotide exchange factor, increases during progression 2009 May 21;113(21):5266-76 to androgen independence in prostate cancer cells and potentiates androgen receptor transcriptional activity. Mol Fraser HI, Dendrou CA, Healy B, Rainbow DB, Howlett S, Endocrinol. 2006 May;20(5):1061-72 Smink LJ, Gregory S, Steward CA, Todd JA, Peterson LB, Wicker LS. Nonobese diabetic congenic strain analysis of Sauzeau V, Sevilla MA, Rivas-Elena JV, de Alava E, autoimmune diabetes reveals genetic complexity of the Montero MJ, López-Novoa JM, Bustelo XR. Vav3 proto- Idd18 locus and identifies Vav3 as a candidate gene. J oncogene deficiency leads to sympathetic hyperactivity Immunol. 2010 May 1;184(9):5075-84 and cardiovascular dysfunction. Nat Med. 2006 Jul;12(7):841-5 Quevedo C, Sauzeau V, Menacho-Márquez M, Castro- Castro A, Bustelo XR. Vav3-deficient mice exhibit a Thierry-Mieg D, Thierry-Mieg J. AceView: a transient delay in cerebellar development. Mol Biol Cell. comprehensive cDNA-supported gene and transcripts 2010 Mar;21(6):1125-39 annotation. Genome Biol. 2006;7 Suppl 1:S12.1-14 Toumaniantz G, Ferland-McCollough D, Cario- Banach-Petrosky W, Jessen WJ, Ouyang X, Gao H, Rao Toumaniantz C, Pacaud P, Loirand G. The Rho protein J, Quinn J, Aronow BJ, Abate-Shen C. Prolonged exchange factor Vav3 regulates vascular smooth muscle exposure to reduced levels of androgen accelerates cell proliferation and migration. Cardiovasc Res. 2010 Apr prostate cancer progression in Nkx3.1; Pten mutant mice. 1;86(1):131-40 Cancer Res. 2007 Oct 1;67(19):9089-96

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 442

VAV3 (vav 3 guanine nucleotide exchange factor) Lyons L, Burnstein KL

Yu B, Martins IR, Li P, Amarasinghe GK, Umetani J, This article should be referenced as such: Fernandez-Zapico ME, Billadeau DD, Machius M, Tomchick DR, Rosen MK. Structural and energetic Lyons L, Burnstein KL. VAV3 (vav 3 guanine nucleotide mechanisms of cooperative autoinhibition and activation of exchange factor). Atlas Genet Cytogenet Oncol Haematol. Vav1. Cell. 2010 Jan 22;140(2):246-56 2011; 15(5):438-443.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 443

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI CAMTA1ID908ch1p36.txt

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

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) 444

CAMTA1 (calmodulin binding transcription activator 1) Henrich KO

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

Barbashina V, Salazar P, Holland EC, Rosenblum MK, Implicated in Ladanyi M. Allelic losses at 1p36 and 19q13 in gliomas: correlation with histologic classification, definition of a 150- Neuroblastoma kb minimal deleted region on 1p36, and evaluation of CAMTA1 as a candidate tumor suppressor gene. Clin Note Cancer Res. 2005 Feb 1;11(3):1119-28 The 1p36 smallest region of overlapping deletion in Henrich KO, Fischer M, Mertens D, Benner A, et al. neuroblastomas spans only 261 kb and pinpoints Reduced expression of CAMTA1 correlates with adverse the CAMTA1 gene (Henrich et al., 2006). In the outcome in neuroblastoma patients. Clin Cancer Res. absence of somatic mutations (Henrich et al., 2007), 2006 Jan 1;12(1):131-8 low CAMTA1 mRNA expression is significantly Kim MY, Yim SH, Kwon MS, Kim TM, Shin SH, Kang HM, associated with markers of unfavorable tumor Lee C, Chung YJ. Recurrent genomic alterations with biology and poor neuroblastoma outcome. In impact on survival in colorectal cancer identified by multivariate survival analysis, this prognostic value genome-wide array comparative genomic hybridization. Gastroenterology. 2006 Dec;131(6):1913-24 is independent of established risk markers, including 1p deletion (Henrich et al., 2006). Song K, Backs J, McAnally J, Qi X, Gerard RD, Richardson JA, Hill JA, Bassel-Duby R, Olson EN. The Glioma transcriptional coactivator CAMTA2 stimulates cardiac growth by opposing class II histone deacetylases. Cell. Note 2006 May 5;125(3):453-66 CAMTA1 is homozygously deleted in a subset of Finkler A, Ashery-Padan R, Fromm H. CAMTAs: gliomas (Ichimura et al., 2008) and is the only gene calmodulin-binding transcription activators from plants to mapping to the smallest region of overlapping human. FEBS Lett. 2007 Aug 21;581(21):3893-8 heterozygous deletion in this entity (Barbashina et Henrich KO, Claas A, Praml C, Benner A, Mollenhauer J, al., 2005). Poustka A, Schwab M, Westermann F. Allelic variants of CAMTA1 and FLJ10737 within a commonly deleted region Colon cancer at 1p36 in neuroblastoma. Eur J Cancer. 2007 Note Feb;43(3):607-16 In colorectal cancer, loss of a 2 Mb region Huentelman MJ, Papassotiropoulos A, Craig DW, Hoerndli encompassing CAMTA1 had the strongest impact FJ, Pearson JV, Huynh KD, Corneveaux J, Hänggi J, on survival among all copy number alterations Mondadori CR, Buchmann A, Reiman EM, Henke K, de Quervain DJ, Stephan DA. Calmodulin-binding identified by genome-wide copy number analysis transcription activator 1 (CAMTA1) alleles predispose (Kim et al., 2006). As in neuroblastoma, low human episodic memory performance. Hum Mol Genet. expression of CAMTA1 mRNA is an independent 2007 Jun 15;16(12):1469-77 predictor of poor outcome in colorectal cancer (Kim Henrich KO and Westermann F.. CAMTA1. Encyclopedia et al., 2006). of Cancer 2008; M. Schwab, Ed., Berlin, New York, Heidelberg, Springer. (REVIEW) References Ichimura K, Vogazianou AP, Liu L, Pearson DM, Bäcklund LM, Plant K, Baird K, Langford CF, Gregory SG, Collins Nagase T, Ishikawa K, Suyama M, Kikuno R, Hirosawa M, VP. 1p36 is a preferential target of chromosome 1 Miyajima N, Tanaka A, Kotani H, Nomura N, Ohara O. deletions in astrocytic tumours and homozygously deleted Prediction of the coding sequences of unidentified human in a subset of glioblastomas. Oncogene. 2008 Mar genes. XII. The complete sequences of 100 new cDNA 27;27(14):2097-108 clones 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. Henrich KO. CAMTA1 (calmodulin binding transcription A novel family of calmodulin-binding transcription activator 1). Atlas Genet Cytogenet Oncol Haematol. 2011; activators in multicellular organisms. J Biol Chem. 2002 15(5):444-445. Jun 14;277(24):21851-61

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 445

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI DYRK1AID43234ch21q22.txt

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, Note MNBH The chromosomal region depicted in the above HGNC (Hugo): DYRK1A figure is part of the "critical region" for Down Location: 21q22.13 syndrome, commonly named Down syndrome critical region (DSCR). This region, which Local order: encompasses 5.4 Mb of chromosome 21 and Genes surrounding DYRK1A and their contains around 30 genes, has been defined in chromosome locations ordered from centromere to phenotype-genotype correlation studies of telomere (according to NCBI Map Viewer): individuals with partial trisomy 21 (Delabar et al., - TTC3 (tetratricopeptide repeat domain 3). 1993). Similar correlation studies of patients with Location 21q22.13. partial monosomy 21 have defined a common - DSCR9 (Down syndrome critical region gene 9; region within DSCR that when deleted causes miscRNA, non-protein coding). Location 21q22.13. microcephaly and developmental delay. This region - DSCR3 (Down syndrome critical region gene 3). expands 1.2 Mb and contains 10 genes including Location 21q22.13. DYRK1A (Matsumoto et al., 1997). The - DYRK1A. Location 21q22.13. identification of two unrelated patients with - LOC100289229 (hypothetical gene). Location microcephaly carrying a DYRK1A truncated 21q22.13. mutation in hemizygosis (see bellow; Moller et al., - KCNJ6 (potassium inwardly-rectifying channel, 2007) indicates that haploinsufficiency of subfamily J, member 6). Location: 21q22.1; DYRK1A is most likely the cause of the common 21q22.1. features presented by these two patients and by - DSCR4 (Down syndrome critical region gene 4). patients 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) 446

DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation Arbonés ML, de la Luna S regulated kinase 1A)

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 447

DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation Arbonés ML, de la Luna S regulated kinase 1A)

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 448

DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation Arbonés ML, de la Luna S regulated kinase 1A)

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 449

DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation Arbonés ML, de la Luna S regulated kinase 1A)

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 450

DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation Arbonés ML, de la Luna S regulated kinase 1A)

de Wit NJ, Burtscher HJ, Weidle UH, Ruiter DJ, van Muijen Ahn KJ, Jeong HK, Choi HS, Ryoo SR, Kim YJ, Goo JS, GN. Differentially expressed genes identified in human Choi SY, Han JS, Ha I, Song WJ. DYRK1A BAC melanoma cell lines with different metastatic behaviour transgenic mice show altered synaptic plasticity with using high density oligonucleotide arrays. Melanoma Res. learning and memory defects. Neurobiol Dis. 2006 2002 Feb;12(1):57-69 Jun;22(3):463-72 Fotaki V, Dierssen M, Alcántara S, Martínez S, Martí E, Arron JR, Winslow MM, Polleri A, Chang CP, Wu H, Gao Casas C, Visa J, Soriano E, Estivill X, Arbonés ML. X, Neilson JR, Chen L, Heit JJ, Kim SK, Yamasaki N, Dyrk1A haploinsufficiency affects viability and causes Miyakawa T, Francke U, Graef IA, Crabtree GR. NFAT developmental delay and abnormal brain morphology in dysregulation by increased dosage of DSCR1 and mice. Mol Cell Biol. 2002 Sep;22(18):6636-47 DYRK1A on chromosome 21. Nature. 2006 Jun 1;441(7093):595-600 Mao J, Maye P, Kogerman P, Tejedor FJ, Toftgard R, Xie W, Wu G, Wu D. Regulation of Gli1 transcriptional activity de Graaf K, Czajkowska H, Rottmann S, Packman LC, in the nucleus by Dyrk1. J Biol Chem. 2002 Sep Lilischkis R, Lüscher B, Becker W. The protein kinase 20;277(38):35156-61 DYRK1A phosphorylates the splicing factor SF3b1/SAP155 at Thr434, a novel in vivo phosphorylation Alvarez M, Estivill X, de la Luna S. DYRK1A accumulates site. BMC Biochem. 2006 Mar 2;7:7 in splicing speckles through a novel targeting signal and induces speckle disassembly. J Cell Sci. 2003 Aug Kim MY, Jeong BC, Lee JH, Kee HJ, Kook H, Kim NS, Kim 1;116(Pt 15):3099-107 YH, Kim JK, Ahn KY, Kim KK. A repressor complex, AP4 transcription factor and geminin, negatively regulates Hämmerle B, Carnicero A, Elizalde C, Ceron J, Martínez expression of target genes in nonneuronal cells. Proc Natl S, Tejedor FJ. Expression patterns and subcellular Acad Sci U S A. 2006 Aug 29;103(35):13074-9 localization of the Down syndrome candidate protein MNB/DYRK1A suggest a role in late neuronal Allan LA, Clarke PR. Phosphorylation of caspase-9 by differentiation. Eur J Neurosci. 2003 Jun;17(11):2277-86 CDK1/cyclin B1 protects mitotic cells against apoptosis. Mol Cell. 2007 Apr 27;26(2):301-10 Martí E, Altafaj X, Dierssen M, de la Luna S, Fotaki V, Alvarez M, Pérez-Riba M, Ferrer I, Estivill X. Dyrk1A Chang HS, Lin CH, Yang CH, Yen MS, Lai CR, Chen YR, expression pattern supports specific roles of this kinase in Liang YJ, Yu WC. Increased expression of Dyrk1a in the adult central nervous system. Brain Res. 2003 Feb HPV16 immortalized keratinocytes enable evasion of 28;964(2):250-63 apoptosis. Int J Cancer. 2007 Jun 1;120(11):2377-85 von Groote-Bidlingmaier F, Schmoll D, Orth HM, Joost HG, Dowjat WK, Adayev T, Kuchna I, Nowicki K, Palminiello S, Becker W, Barthel A. DYRK1 is a co-activator of FKHR Hwang YW, Wegiel J. Trisomy-driven overexpression of (FOXO1a)-dependent glucose-6-phosphatase gene DYRK1A kinase in the brain of subjects with Down expression. Biochem Biophys Res Commun. 2003 Jan syndrome. Neurosci Lett. 2007 Feb 8;413(1):77-81 17;300(3):764-9 Aranda S, Alvarez M, Turró S, Laguna A, de la Luna S. Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo Sprouty2-mediated inhibition of fibroblast growth factor CJ, Maitra A. Met proto-oncogene and insulin-like growth signaling is modulated by the protein kinase DYRK1A. Mol factor binding protein 3 overexpression correlates with Cell Biol. 2008 Oct;28(19):5899-911 metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clin Cancer Res. 2004 Sep 15;10(18 Pt Canzonetta C, Mulligan C, Deutsch S, Ruf S, O'Doherty A, 1):6152-8 Lyle R, Borel C, Lin-Marq N, Delom F, Groet J, Schnappauf F, De Vita S, Averill S, Priestley JV, Martin JE, Sitz JH, Tigges M, Baumgärtel K, Khaspekov LG, Lutz B. Shipley J, Denyer G, Epstein CJ, Fillat C, Estivill X, Dyrk1A potentiates steroid hormone-induced transcription Tybulewicz VL, Fisher EM, Antonarakis SE, Nizetic D. via the chromatin remodeling factor Arip4. Mol Cell Biol. DYRK1A-dosage imbalance perturbs NRSF/REST levels, 2004 Jul;24(13):5821-34 deregulating pluripotency and embryonic stem cell fate in Down syndrome. Am J Hum Genet. 2008 Sep;83(3):388- Skurat AV, Dietrich AD. Phosphorylation of Ser640 in 400 muscle glycogen synthase by DYRK family protein kinases. J Biol Chem. 2004 Jan 23;279(4):2490-8 Hämmerle B, Elizalde C, Tejedor FJ. The spatio-temporal and subcellular expression of the candidate Down Wegiel J, Kuchna I, Nowicki K, Frackowiak J, Dowjat K, syndrome gene Mnb/Dyrk1A in the developing mouse Silverman WP, Reisberg B, DeLeon M, Wisniewski T, brain suggests distinct sequential roles in neuronal Adayev T, Chen-Hwang MC, Hwang YW. Cell type- and development. Eur J Neurosci. 2008 Mar;27(5):1061-74 brain structure-specific patterns of distribution of minibrain kinase in human brain. Brain Res. 2004 Jun 4;1010(1- Laguna A, Aranda S, Barallobre MJ, Barhoum R, 2):69-80 Fernández E, Fotaki V, Delabar JM, de la Luna S, de la Villa P, Arbonés ML. The protein kinase DYRK1A Benavides-Piccione R, Dierssen M, Ballesteros-Yáñez I, regulates caspase-9-mediated apoptosis during retina Martínez de Lagrán M, Arbonés ML, Fotaki V, DeFelipe J, development. Dev Cell. 2008 Dec;15(6):841-53 Elston GN. Alterations in the phenotype of neocortical pyramidal cells in the Dyrk1A+/- mouse. Neurobiol Dis. Liang YJ, Chang HS, Wang CY, Yu WC. DYRK1A 2005 Oct;20(1):115-22 stabilizes HPV16E7 oncoprotein through phosphorylation of the threonine 5 and threonine 7 residues. Int J Biochem Kelly PA, Rahmani Z. DYRK1A enhances the mitogen- Cell Biol. 2008;40(11):2431-41 activated protein kinase cascade in PC12 cells by forming a complex with Ras, B-Raf, and MEK1. Mol Biol Cell. 2005 Maenz B, Hekerman P, Vela EM, Galceran J, Becker W. Aug;16(8):3562-73 Characterization of the human DYRK1A promoter and its regulation by the transcription factor E2F1. BMC Mol Biol. Lochhead PA, Sibbet G, Morrice N, Cleghon V. Activation- 2008 Mar 26;9:30 loop autophosphorylation is mediated by a novel transitional intermediate form of DYRKs. Cell. 2005 Jun Møller RS, Kübart S, Hoeltzenbein M, Heye B, Vogel I, 17;121(6):925-36 Hansen CP, Menzel C, Ullmann R, Tommerup N, Ropers HH, Tümer Z, Kalscheuer VM. Truncation of the Down syndrome candidate gene DYRK1A in two unrelated

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 451

DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation Arbonés ML, de la Luna S regulated kinase 1A)

patients with microcephaly. Am J Hum Genet. 2008 Park J, Oh Y, Chung KC. Two key genes closely May;82(5):1165-70 implicated with the neuropathological characteristics in Down syndrome: DYRK1A and RCAN1. BMB Rep. 2009 Seifert A, Allan LA, Clarke PR. DYRK1A phosphorylates Jan 31;42(1):6-15 caspase 9 at an inhibitory site and is potently inhibited in human cells by harmine. FEBS J. 2008 Dec;275(24):6268- Park J, Song WJ, Chung KC. Function and regulation of 80 Dyrk1A: towards understanding Down syndrome. Cell Mol Life Sci. 2009 Oct;66(20):3235-40 Shi J, Zhang T, Zhou C, Chohan MO, Gu X, Wegiel J, Zhou J, Hwang YW, Iqbal K, Grundke-Iqbal I, Gong CX, Scales TM, Lin S, Kraus M, Goold RG, Gordon-Weeks PR. Liu F. Increased dosage of Dyrk1A alters alternative Nonprimed and DYRK1A-primed GSK3 beta- splicing factor (ASF)-regulated alternative splicing of tau in phosphorylation sites on MAP1B regulate microtubule Down syndrome. J Biol Chem. 2008 Oct dynamics in growing axons. J Cell Sci. 2009 Jul 15;122(Pt 17;283(42):28660-9 14):2424-35 Wegiel J, Dowjat K, Kaczmarski W, Kuchna I, Nowicki K, Ferron SR, Pozo N, Laguna A, Aranda S, Porlan E, Frackowiak J, Mazur Kolecka B, Wegiel J, Silverman WP, Moreno M, Fillat C, de la Luna S, Sánchez P, Arbonés ML, Reisberg B, Deleon M, Wisniewski T, Gong CX, Liu F, Fariñas I. Regulated segregation of kinase Dyrk1A during Adayev T, Chen-Hwang MC, Hwang YW. The role of asymmetric neural stem cell division is critical for EGFR- overexpressed DYRK1A protein in the early onset of mediated biased signaling. Cell Stem Cell. 2010 Sep neurofibrillary degeneration in Down syndrome. Acta 3;7(3):367-79 Neuropathol. 2008 Oct;116(4):391-407 Guo X, Williams JG, Schug TT, Li X. DYRK1A and DYRK3 Baek KH, Zaslavsky A, Lynch RC, Britt C, Okada Y, Siarey promote cell survival through phosphorylation and RJ, Lensch MW, Park IH, Yoon SS, Minami T, Korenberg activation of SIRT1. J Biol Chem. 2010 Apr JR, Folkman J, Daley GQ, Aird WC, Galdzicki Z, Ryeom S. 23;285(17):13223-32 Down's syndrome suppression of tumour growth and the role of the calcineurin inhibitor DSCR1. Nature. 2009 Jun Komorek J, Kuppuswamy M, Subramanian T, Vijayalingam 25;459(7250):1126-30 S, Lomonosova E, Zhao LJ, Mymryk JS, Schmitt K, Chinnadurai G. Adenovirus type 5 E1A and E6 proteins of Fernandez-Martinez J, Vela EM, Tora-Ponsioen M, Ocaña low-risk cutaneous beta-human papillomaviruses suppress OH, Nieto MA, Galceran J. Attenuation of Notch signalling cell transformation through interaction with FOXK1/K2 by the Down-syndrome-associated kinase DYRK1A. J Cell transcription factors. J Virol. 2010 Mar;84(6):2719-31 Sci. 2009 May 15;122(Pt 10):1574-83 Kurabayashi N, Hirota T, Sakai M, Sanada K, Fukada Y. Göckler N, Jofre G, Papadopoulos C, Soppa U, Tejedor DYRK1A and glycogen synthase kinase 3beta, a dual- FJ, Becker W. Harmine specifically inhibits protein kinase kinase mechanism directing proteasomal degradation of DYRK1A and interferes with neurite formation. FEBS J. CRY2 for circadian timekeeping. Mol Cell Biol. 2010 2009 Nov;276(21):6324-37 Apr;30(7):1757-68 Kuhn C, Frank D, Will R, Jaschinski C, Frauen R, Katus Park J, Oh Y, Yoo L, Jung MS, Song WJ, Lee SH, Seo H, HA, Frey N. DYRK1A is a novel negative regulator of Chung KC. Dyrk1A phosphorylates p53 and inhibits cardiomyocyte hypertrophy. J Biol Chem. 2009 Jun proliferation of embryonic neuronal cells. J Biol Chem. 19;284(25):17320-7 2010 Oct 8;285(41):31895-906 Lee Y, Ha J, Kim HJ, Kim YS, Chang EJ, Song WJ, Kim Pussegoda KA. Down's syndrome patients are less likely HH. Negative feedback Inhibition of NFATc1 by DYRK1A to develop cancer. Clin Genet. 2010 Jul;78(1):35-7 regulates bone homeostasis. J Biol Chem. 2009 Nov 27;284(48):33343-51 Raaf L, Noll C, Cherifi M, Benazzoug Y, Delabar JM, Janel N. Hyperhomocysteinemia-induced Dyrk1a downregulation Lepagnol-Bestel AM, Zvara A, Maussion G, Quignon F, results in cardiomyocyte hypertrophy in rats. Int J Cardiol. Ngimbous B, Ramoz N, Imbeaud S, Loe-Mie Y, Benihoud 2010 Nov 19;145(2):306-7 K, Agier N, Salin PA, Cardona A, Khung-Savatovsky S, Kallunki P, Delabar JM, Puskas LG, Delacroix H, Yabut O, Domogauer J, D'Arcangelo G. Dyrk1A Aggerbeck L, Delezoide AL, Delattre O, Gorwood P, overexpression inhibits proliferation and induces Moalic JM, Simonneau M. DYRK1A interacts with the premature neuronal differentiation of neural progenitor REST/NRSF-SWI/SNF chromatin remodelling complex to cells. J Neurosci. 2010 Mar 17;30(11):4004-14 deregulate gene clusters involved in the neuronal phenotypic traits of Down syndrome. Hum Mol Genet. This article should be referenced as such: 2009 Apr 15;18(8):1405-14 Arbonés ML, de la Luna S. DYRK1A (dual-specificity Murakami N, Bolton D, Hwang YW. Dyrk1A binds to tyrosine-(Y)-phosphorylation regulated kinase 1A). Atlas multiple endocytic proteins required for formation of Genet Cytogenet Oncol Haematol. 2011; 15(5):446-452. clathrin-coated vesicles. Biochemistry. 2009 Oct 6;48(39):9297-305

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 452

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI FBLN1ID44462ch22q13.txt

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

Transcription factor.

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 453

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 454

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 455

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 456

FBLN1 (fibulin 1) Castagnoli L, et al.

suppression of fibronectin-regulated cell adhesion and Breakpoints motility. J Cell Sci. 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 Description: Translocation t(12;22)(p11.2;q13.3). (FBLN1) is disrupted in a t(12;22) associated with a Phenotype: Synpolydactyly. complex type of synpolydactyly. J Med Genet. 2002 Analysis of a Belgian family with a complex type Feb;39(2):98-104 of synpolydactyly (SPD2) and a Du M, Fan X, Hong E, Chen JJ. Interaction of oncogenic t(12;22)(p11.2;q13.3) translocation identified the papillomavirus E6 proteins with fibulin-1. Biochem Biophys breakpoints located in the intron between the last 2 Res Commun. 2002 Aug 30;296(4):962-9 exons of the fibulin-1D splice variant isoform Forti S, Scanlan MJ, Invernizzi A, Castiglioni F, Pupa S, (exons 19, 20) and the 5' UTR of the C12ORF2 Agresti R, Fontanelli R, Morelli D, Old LJ, Pupa SM, gene. Fibroblasts derived from the patients Ménard S. Identification of breast cancer-restricted displayed decreased levels of ECM-related fibulin- antigens by antibody screening of SKBR3 cDNA library using a preselected patient's serum. Breast Cancer Res 1D secreted into the culture medium, whereas Treat. 2002 Jun;73(3):245-56 levels of the fibulin-1C variant were normal. The Moll F, Katsaros D, Lazennec G, Hellio N, Roger P, findings are consistent with the notion that the Giacalone PL, Chalbos D, Maudelonde T, Rochefort H, t(12;22)(p11.2;q13.3) translocation results in haplo- Pujol P. Estrogen induction and overexpression of fibulin- insufficiency of the fibulin-1D variant, leading to 1C mRNA in ovarian cancer cells. Oncogene. 2002 Feb the observed limb malformations. The authors 7;21(7):1097-107 noted that the entire fibulin-1 gene is deleted in the Pupa SM, Forti S, Balsari A, Ménard S. Humoral immune chromosome 22q13.3 deletion syndrome (Debeer et response for early diagnosis of breast carcinoma. Ann al., 2002). Oncol. 2002 Mar;13(3):483 Greene LM, Twal WO, Duffy MJ, McDermott EW, Hill AD, References O'Higgins NJ, McCann AH, Dervan PA, Argraves WS, Gallagher WM. Elevated expression and altered Argraves WS, Tran H, Burgess WH, Dickerson K. Fibulin is processing of fibulin-1 protein in human breast cancer. Br J an extracellular matrix and plasma glycoprotein with Cancer. 2003 Mar 24;88(6):871-8 repeated domain structure. J Cell Biol. 1990 Dec;111(6 Pt Timpl R, Sasaki T, Kostka G, Chu ML. Fibulins: a versatile 2):3155-64 family of extracellular matrix proteins. Nat Rev Mol Cell Balbona K, Tran H, Godyna S, Ingham KC, Strickland DK, Biol. 2003 Jun;4(6):479-89 Argraves WS. Fibulin binds to itself and to the carboxyl- Pupa SM, Argraves WS, Forti S, Casalini P, Berno V, terminal heparin-binding region of fibronectin. J Biol Chem. Agresti R, Aiello P, Invernizzi A, Baldassari P, Twal WO, 1992 Oct 5;267(28):20120-5 Mortarini R, Anichini A, Ménard S. Immunological and Tran H, Tanaka A, Litvinovich SV, Medved LV, pathobiological roles of fibulin-1 in breast cancer. Haudenschild CC, Argraves WS. The interaction of fibulin- Oncogene. 2004 Mar 18;23(12):2153-60 1 with fibrinogen. A potential role in hemostasis and Lanza F. Bernard-Soulier syndrome (hemorrhagiparous thrombosis. J Biol Chem. 1995 Aug 18;270(33):19458-64 thrombocytic dystrophy). Orphanet J Rare Dis. 2006 Nov Clinton GM, Rougeot C, Derancourt J, Roger P, Defrenne 16;1:46 A, Godyna S, Argraves WS, Rochefort H. Estrogens Singh U, Sun T, Larsson T, Elliott RW, Kostka G, Fundele increase the expression of fibulin-1, an extracellular matrix RH. Expression and functional analysis of fibulin-1 (Fbln1) protein secreted by human ovarian cancer cells. Proc Natl during normal and abnormal placental development of the Acad Sci U S A. 1996 Jan 9;93(1):316-20 mouse. Placenta. 2006 Sep-Oct;27(9-10):1014-21 Qing J, Maher VM, Tran H, Argraves WS, Dunstan RW, Pupa SM, Giuffré S, Castiglioni F, Bertola L, Cantú M, et McCormick JJ. Suppression of anchorage-independent al. Regulation of breast cancer response to chemotherapy growth and matrigel invasion and delayed tumor formation by fibulin-1. Cancer Res. 2007 May 1;67(9):4271-7 by elevated expression of fibulin-1D in human fibrosarcoma-derived cell lines. Oncogene. 1997 Cooley MA, Kern CB, Fresco VM, Wessels A, Thompson Oct;15(18):2159-68 RP, McQuinn TC, Twal WO, Mjaatvedt CH, Drake CJ, Argraves WS. Fibulin-1 is required for morphogenesis of Pan TC, Kostka G, Zhang RZ, Timpl R, Chu ML. Complete neural crest-derived structures. Dev Biol. 2008 Jul exon-intron organization of the mouse fibulin-1 gene and 15;319(2):336-45 its comparison with the human fibulin-1 gene. FEBS Lett. 1999 Feb 5;444(1):38-42 Argraves WS, Tanaka A, Smith EP, Twal WO, Argraves KM, Fan D, Haudenschild CC. Fibulin-1 and fibrinogen in Perbal B, Martinerie C, Sainson R, Werner M, He B, human atherosclerotic lesions. Histochem Cell Biol. 2009 Roizman B. The C-terminal domain of the regulatory Nov;132(5):559-65 protein NOVH is sufficient to promote interaction with fibulin 1C: a clue for a role of NOVH in cell-adhesion Mohamed SA, Sievers HH, Hanke T, Richardt D, et al. signaling. Proc Natl Acad Sci U S A. 1999 Feb Pathway analysis of differentially expressed genes in 2;96(3):869-74 patients with acute aortic dissection. Biomark Insights. 2009 May 6;4:81-90 Ohsawa I, Takamura C, Kohsaka S. Fibulin-1 binds the amino-terminal head of beta-amyloid precursor protein and This article should be referenced as such: modulates its physiological function. J Neurochem. 2001 Mar;76(5):1411-20 Castagnoli L, Tagliabue E, Pupa SM. FBLN1 (fibulin 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5):453- Twal WO, Czirok A, Hegedus B, Knaak C, Chintalapudi 457. MR, Okagawa H, Sugi Y, Argraves WS. Fibulin-1

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 457

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI HUS1ID40899ch7p12.txt

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. Note: NCBI accession: NM_004507.2; Hus1 is a component of the 9-1-1 cell cycle NP_004498.1. checkpoint complex that plays a critical role in sensing DNA damage and maintaining genomic DNA/RNA stability. Description Expression Found in all tissues. 15464 bp; 8 exons. Transcription Localisation Nucleus and cytoplasm. In discrete nuclear foci The transcribed mRNA has 2143 bp and the coding upon 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) 458

HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 459

HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

foci also colocalized with TopBP1 and gamma- accumulation of damaged cells and impaired tissue H2AX (Warmerdam et al., 2009). regeneration (Yazinski et al., 2009). Hus1 interacts with histone deacetylase HDAC1 Oncogenesis (Cai et al., 2000). A novel pathway has been Single nucleotide polymorphism (SNP) analysis proposed that HDAC1 is involved in G(2)/M supports the potential role of Hus1 in sporadic checkpoint control through the interaction with the breast cancer (Vega et al., 2009). SNPs in hHUS1 9-1-1 complex. at chromosome 7 base pair positions 47778020 (C) Jab1 physically associates with the 9-1-1 complex, and 47789957 (G) are statistically associated with causes the translocation of the 9-1-1 complex from breast cancer development (Vega et al., 2009). the nucleus to the cytoplasm, and mediates the rapid degradation of the 9-1-1 complex (Huang et Ovarian tumors al., 2007). Oncogenesis Homology Hus1 expression levels correlate significantly with the clinicopathologic factors of bad prognosis of hHus1 homologues from many eukaryotes are ovarian tumors (de la Torre et al., 2008). highly conserved. The following diagram shows the sequence alignment of human Hus1 with Hus1 of Cancer therapy fission yeast Schizosaccharomyces pombe. The N- Note terminal domain of Hus1 is structurally similar to The status of Hus1 can influence response to cancer the C-terminal domain. The structure of Hus1 (Doré therapy. Down-regulation of hHus1 by antisense et al., 2009; Sohn and Cho, 2009; Xu et al., 2009) is RNA enhances the sensitivity of human lung similar to those of Rad9, Rad1, and PCNA (Gulbis carcinoma cells to cisplatin (a DNA cross-linker), et al., 1996; Krishna et al., 1994). presumably by enhancing apoptosis (Kinzel et al., Mutations 2002). Genomic stability Note Note Complete inactivation of the mouse Hus1 results in Inactivation of mouse or human Hus1 results in chromosomal instability, genotoxin impaired DNA damage signaling and severe hypersensitivity, and embryonic lethality. spontaneous chromosomal instability (Weiss et al., Implicated in 2002; Weiss et al., 2000; Kinzel et al., 2002). Drug sensitivity Breast cancer Note Note Cells lacking Hus1 are hypersensitive to certain Hus1 inactivation in the mammary epithelium genotoxins including camptothecin (CPT), resulted in genome damage that induced apoptosis hydroxyurea (HU), ultraviolet (UV), and ionizing and led to depletion of Hus1-null cells from the radiation (IR) (Weiss et al., 2000; Wang et al., mammary gland. Dual inactivation of Hus1 and p53 2004b; Wang et al., 2006b). in the mouse mammary gland results in

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) 460

HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 461

HUS1 (HUS1 checkpoint homolog (S. pombe)) Madabushi A, et al.

protein A in human cells. Oncogene. 2005 Jul Meyerkord CL, Takahashi Y, Araya R, Takada N, Weiss 7;24(29):4728-35 RS, Wang HG. Loss of Hus1 sensitizes cells to etoposide- induced apoptosis by regulating BH3-only proteins. Francia S, Weiss RS, Hande MP, Freire R, d'Adda di Oncogene. 2008 Dec 11;27(58):7248-59 Fagagna F. Telomere and telomerase modulation by the mammalian Rad9/Rad1/Hus1 DNA-damage-checkpoint Balakrishnan L, Brandt PD, Lindsey-Boltz LA, Sancar A, complex. Curr Biol. 2006 Aug 8;16(15):1551-8 Bambara RA. Long patch base excision repair proceeds via coordinated stimulation of the multienzyme DNA repair Lu AL, Bai H, Shi G, Chang DY. MutY and MutY homologs complex. J Biol Chem. 2009 May 29;284(22):15158-72 (MYH) in genome maintenance. Front Biosci. 2006 Sep 1;11:3062-80 Doré AS, Kilkenny ML, Rzechorzek NJ, Pearl LH. Crystal structure of the rad9-rad1-hus1 DNA damage checkpoint Shi G, Chang DY, Cheng CC, Guan X, Venclovas C, Lu complex--implications for clamp loading and regulation. AL. Physical and functional interactions between MutY Mol Cell. 2009 Jun 26;34(6):735-45 glycosylase homologue (MYH) and checkpoint proteins Rad9-Rad1-Hus1. Biochem J. 2006 Nov 15;400(1):53-62 Park MJ, Park JH, Hahm SH, Ko SI, Lee YR, Chung JH, Sohn SY, Cho Y, Kang LW, Han YS. Repair activities of Wang W, Lindsey-Boltz LA, Sancar A, Bambara RA. human 8-oxoguanine DNA glycosylase are stimulated by Mechanism of stimulation of human DNA ligase I by the the interaction with human checkpoint sensor Rad9-Rad1- Rad9-rad1-Hus1 checkpoint complex. J Biol Chem. 2006 Hus1 complex. DNA Repair (Amst). 2009 Oct Jul 28;281(30):20865-72 2;8(10):1190-200 Wang X, Hu B, Weiss RS, Wang Y. The effect of Hus1 on Sohn SY, Cho Y. Crystal structure of the human rad9- ionizing radiation sensitivity is associated with homologous hus1-rad1 clamp. J Mol Biol. 2009 Jul 17;390(3):490-502 recombination repair but is independent of nonhomologous end-joining. Oncogene. 2006 Mar 23;25(13):1980-3 Vega A, Salas A, Milne RL, Carracedo B, Ribas G, Ruibal A, de León AC, González-Hernández A, Benítez J, Gembka A, Toueille M, Smirnova E, Poltz R, Ferrari E, Carracedo A. Evaluating new candidate SNPs as low Villani G, Hübscher U. The checkpoint clamp, Rad9-Rad1- penetrance risk factors in sporadic breast cancer: a two- Hus1 complex, preferentially stimulates the activity of stage Spanish case-control study. Gynecol Oncol. 2009 apurinic/apyrimidinic endonuclease 1 and DNA Jan;112(1):210-4 polymerase beta in long patch base excision repair. Nucleic Acids Res. 2007;35(8):2596-608 Warmerdam DO, Freire R, Kanaar R, Smits VA. Cell cycle- dependent processing of DNA lesions controls localization Guan X, Bai H, Shi G, Theriot CA, Hazra TK, Mitra S, Lu of Rad9 to sites of genotoxic stress. Cell Cycle. 2009 Jun AL. The human checkpoint sensor Rad9-Rad1-Hus1 1;8(11):1765-74 interacts with and stimulates NEIL1 glycosylase. Nucleic Acids Res. 2007;35(8):2463-72 Xu M, Bai L, Gong Y, Xie W, Hang H, Jiang T. Structure and functional implications of the human rad9-hus1-rad1 Guan X, Madabushi A, Chang DY, Fitzgerald ME, Shi G, cell cycle checkpoint complex. J Biol Chem. 2009 Jul Drohat AC, Lu AL. The human checkpoint sensor Rad9- 31;284(31):20457-61 Rad1-Hus1 interacts with and stimulates DNA repair enzyme TDG glycosylase. Nucleic Acids Res. Yazinski SA, Westcott PM, Ong K, Pinkas J, Peters RM, 2007;35(18):6207-18 Weiss RS. Dual inactivation of Hus1 and p53 in the mouse mammary gland results in accumulation of damaged cells Huang J, Yuan H, Lu C, Liu X, Cao X, Wan M. Jab1 and impaired tissue regeneration. Proc Natl Acad Sci U S mediates protein degradation of the Rad9-Rad1-Hus1 A. 2009 Dec 15;106(50):21282-7 checkpoint complex. J Mol Biol. 2007 Aug 10;371(2):514- 27 Bai H, Madabushi A, Guan X, Lu AL. Interaction between human mismatch repair recognition proteins and Levitt PS, Zhu M, Cassano A, Yazinski SA, Liu H, Darfler checkpoint sensor Rad9-Rad1-Hus1. DNA Repair (Amst). J, Peters RM, Weiss RS. Genome maintenance defects in 2010 May 4;9(5):478-87 cultured cells and mice following partial inactivation of the essential cell cycle checkpoint gene Hus1. Mol Cell Biol. Luncsford PJ, Chang DY, Shi G, Bernstein J, Madabushi 2007 Mar;27(6):2189-201 A, Patterson DN, Lu AL, Toth EA. A structural hinge in eukaryotic MutY homologues mediates catalytic activity de la Torre J, Gil-Moreno A, García A, Rojo F, Xercavins J, and Rad9-Rad1-Hus1 checkpoint complex interactions. J Salido E, Freire R. Expression of DNA damage checkpoint Mol Biol. 2010 Oct 29;403(3):351-70 protein Hus1 in epithelial ovarian tumors correlates with prognostic markers. Int J Gynecol Pathol. 2008 This article should be referenced as such: Jan;27(1):24-32 Madabushi A, Gunther RC, Lu AL. HUS1 (HUS1 He W, Zhao Y, Zhang C, An L, Hu Z, Liu Y, Han L, Bi L, checkpoint homolog (S. pombe)). Atlas Genet Cytogenet Xie Z, Xue P, Yang F, Hang H. Rad9 plays an important Oncol Haematol. 2011; 15(5):458-462. 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) 462

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI OTX2ID46429ch14q22.txt

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

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) 463

OTX2 (orthodenticle homeobox 2) Wortham M

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 464

OTX2 (orthodenticle homeobox 2) Wortham M

Disease based comparative genomic hybridization. Genes Microphthalmia is clinically defined as an eye with Chromosomes Cancer. 2005 Jul;43(3):294-301 an axial diameter measuring at least two standard Chatelain G, Fossat N, Brun G, Lamonerie T. Molecular deviations below the mean for the corresponding dissection reveals decreased activity and not dominant negative effect in human OTX2 mutants. J Mol Med. 2006 age group (Omwochei et al., 2000), whereas Jul;84(7):604-15 anophthalmia is diagnosed when no clinically apparent eye structure is present. Those affected Fossat N, Chatelain G, Brun G, Lamonerie T. Temporal and spatial delineation of mouse Otx2 functions by generally harbor bilateral malformations. Like conditional self-knockout. EMBO Rep. 2006 Aug;7(8):824- coloboma, some forms of 30 anophthalmia/microphthalmia are clearly Verma AS, Fitzpatrick DR. Anophthalmia and inheritable, while for other cases environmental microphthalmia. Orphanet J Rare Dis. 2007 Nov 26;2:47 factors have been implicated but not definitively so Diaczok D, Romero C, Zunich J, Marshall I, Radovick S. A (Verma et al., 2007). novel dominant negative mutation of OTX2 associated with Anophthalmia/microphthalmia can present as combined pituitary hormone deficiency. J Clin Endocrinol secondary malformations following colobomata. Metab. 2008 Nov;93(11):4351-9 Cytogenetics Omodei D, Acampora D, Mancuso P, Prakash N, Di Various genes have been implicated, including Giovannantonio LG, Wurst W, Simeone A. Anterior- posterior graded response to Otx2 controls proliferation SOX2 (autosomal dominant inheritance), OTX2 and differentiation of dopaminergic progenitors in the (autosomal dominant), CHX10 (autosomal ventral mesencephalon. Development. 2008 recessive), and RAX (autosomal recessive; Verma Oct;135(20):3459-70 et al., 2007; Wyatt et al., 2008). Sugiyama S, Di Nardo AA, Aizawa S, Matsuo I, Volovitch M, Prochiantz A, Hensch TK. Experience-dependent References transfer of Otx2 homeoprotein into the visual cortex activates postnatal plasticity. Cell. 2008 Aug 8;134(3):508- Frantz GD, Weimann JM, Levin ME, McConnell SK. Otx1 20 and Otx2 define layers and regions in developing cerebral Wyatt A, Bakrania P, Bunyan DJ, Osborne RJ, Crolla JA, cortex and cerebellum. J Neurosci. 1994 Oct;14(10):5725- et al. Novel heterozygous OTX2 mutations and whole gene 40 deletions in anophthalmia, microphthalmia and coloboma. Acampora D, Mazan S, Lallemand Y, Avantaggiato V, Hum Mutat. 2008 Nov;29(11):E278-83 Maury M, Simeone A, Brûlet P. Forebrain and midbrain Glubrecht DD, Kim JH, Russell L, Bamforth JS, Godbout regions are deleted in Otx2-/- mutants due to a defective R. Differential CRX and OTX2 expression in human retina anterior neuroectoderm specification during gastrulation. and retinoblastoma. J Neurochem. 2009 Oct;111(1):250- Development. 1995 Oct;121(10):3279-90 63 Matsuo I, Kuratani S, Kimura C, Takeda N, Aizawa S. Larsen KB, Lutterodt M, Rath MF, Møller M. Expression of Mouse Otx2 functions in the formation and patterning of the homeobox genes PAX6, OTX2, and OTX1 in the early rostral head. Genes Dev. 1995 Nov 1;9(21):2646-58 human fetal retina. Int J Dev Neurosci. 2009 Baas D, Bumsted KM, Martinez JA, Vaccarino FM, Wikler Aug;27(5):485-92 KC, Barnstable CJ. The subcellular localization of Otx2 is Marchler-Bauer A, Anderson JB, Chitsaz F, Derbyshire cell-type specific and developmentally regulated in the MK, et al. CDD: specific functional annotation with the mouse retina. Brain Res Mol Brain Res. 2000 May Conserved Domain Database. Nucleic Acids Res. 2009 31;78(1-2):26-37 Jan;37(Database issue):D205-10 Chau KY, Chen S, Zack DJ, Ono SJ. Functional domains Sugiyama S, Prochiantz A, Hensch TK. From brain of the cone-rod homeobox (CRX) transcription factor. J formation to plasticity: insights on Otx2 homeoprotein. Dev Biol Chem. 2000 Nov 24;275(47):37264-70 Growth Differ. 2009 Apr;51(3):369-77 Onwochei BC, Simon JW, Bateman JB, Couture KC, Mir Adamson DC, Shi Q, Wortham M, Northcott PA, Di C, E. Ocular colobomata. Surv Ophthalmol. 2000 Nov- Duncan CG, Li J, McLendon RE, Bigner DD, Taylor MD, Dec;45(3):175-94 Yan H. OTX2 is critical for the maintenance and Boon K, Osorio EC, Greenhut SF, Schaefer CF, progression of Shh-independent medulloblastomas. Shoemaker J, Polyak K, Morin PJ, Buetow KH, Strausberg Cancer Res. 2010 Jan 1;70(1):181-91 RL, De Souza SJ, Riggins GJ. An anatomy of normal and Larsen KB, Lutterodt MC, Møllgård K, Møller M. malignant gene expression. Proc Natl Acad Sci U S A. Expression of the homeobox genes OTX2 and OTX1 in the 2002 Aug 20;99(17):11287-92 early developing human brain. J Histochem Cytochem. Ragge NK, Brown AG, Poloschek CM, Lorenz B, et al. 2010 Jul;58(7):669-78 Heterozygous mutations of OTX2 cause severe ocular Northcott PA, Rutka JT, Taylor MD. Genomics of malformations. Am J Hum Genet. 2005 Jun;76(6):1008-22 medulloblastoma: from Giemsa-banding to next-generation Vernay B, Koch M, Vaccarino F, Briscoe J, Simeone A, sequencing in 20 years. Neurosurg Focus. 2010 Kageyama R, Ang SL. Otx2 regulates subtype Jan;28(1):E6 specification and neurogenesis in the midbrain. J Neurosci. 2005 May 11;25(19):4856-67 This article should be referenced as such: Zielinski B, Gratias S, Toedt G, Mendrzyk F, Stange DE, Wortham M. OTX2 (orthodenticle homeobox 2). Atlas Radlwimmer B, Lohmann DR, Lichter P. Detection of Genet Cytogenet Oncol Haematol. 2011; 15(5):463-465. chromosomal imbalances in retinoblastoma by matrix-

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 465

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI dic0309p14p13ID1553.txt

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 Identity cell lineage depends on E2A, EBF1, and PAX5; Note activates B-cell specific genes and repress genes See also the paper on dic(9;20)(p11-13;q11). involved in other lineage commitments. Activates the surface cell receptor CD19 and repress FLT3. Clinics and pathology Pax5 physically interacts with the RAG1/RAG2 complex, and removes the inhibitory signal of the Disease lysine-9-methylated histone H3, and induces V-to- Acute lymphoblastic leukaemia (ALL). DJ rearrangements. Genes repressed by PAX5 expression in early B cells are restored in their Phenotype/cell stem origin function in mature B cells and plasma cells, and B-cell precursor ALL. PAX5 repressed (Fuxa et al., 2004; Johnson et al., Epidemiology 2004; Zhang et al., 2006; Cobaleda et al., 2007). One case to date (Mullighan et al., 2007). FOXP1 Prognosis Location 3p14 No data. Protein Genes involved and Transcriptional repressor. Involved in cardiomyocyte proliferation, motor neuron proteins migration, B-lymphocyte development, and the generation of quiescent naive T cells (Shi et al., PAX5 2008; Feng et al., 2010; Rao et al., 2010; Zhang et Location al., 2010; Hisaoka et al., 2010). 9p13.2 Protein Result of the chromosomal 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) 466 dic(3;9)(p14;p13) PAX5/FOXP1 Huret JL

Fusion protein

Shi C, Sakuma M, Mooroka T, Liscoe A, Gao H, Croce KJ, Description Sharma A, Kaplan D, Greaves DR, Wang Y, Simon DI. The predicted fusion protein contains the DNA Down-regulation of the forkhead transcription factor Foxp1 binding paired domain of PAX5 and the DNA- is required for monocyte differentiation and macrophage binding and transcriptional regulator domain of function. Blood. 2008 Dec 1;112(12):4699-711 FOXP1. 877 amino acids. Feng X, Ippolito GC, Tian L, Wiehagen K, Oh S, Sambandam A, Willen J, Bunte RM, Maika SD, Harriss JV, Caton AJ, Bhandoola A, Tucker PW, Hu H. Foxp1 is an References essential transcriptional regulator for the generation of Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, quiescent naive T cells during thymocyte development. Busslinger M. Pax5 induces V-to-DJ rearrangements and Blood. 2010 Jan 21;115(3):510-8 locus contraction of the immunoglobulin heavy-chain gene. Hisaoka T, Nakamura Y, Senba E, Morikawa Y. The Genes Dev. 2004 Feb 15;18(4):411-22 forkhead transcription factors, Foxp1 and Foxp2, identify Johnson K, Pflugh DL, Yu D, Hesslein DG, Lin KI, Bothwell different subpopulations of projection neurons in the AL, Thomas-Tikhonenko A, Schatz DG, Calame K. B cell- mouse cerebral cortex. Neuroscience. 2010 Mar specific loss of histone 3 lysine 9 methylation in the V(H) 17;166(2):551-63 locus depends on Pax5. Nat Immunol. 2004 Aug;5(8):853- Rao DS, O'Connell RM, Chaudhuri AA, Garcia-Flores Y, 61 Geiger TL, Baltimore D. MicroRNA-34a perturbs B Zhang Z, Espinoza CR, Yu Z, Stephan R, He T, Williams lymphocyte development by repressing the forkhead box GS, Burrows PD, Hagman J, Feeney AJ, Cooper MD. transcription factor Foxp1. Immunity. 2010 Jul 23;33(1):48- Transcription factor Pax5 (BSAP) transactivates the RAG- 59 mediated V(H)-to-DJ(H) rearrangement of immunoglobulin Zhang Y, Li S, Yuan L, Tian Y, Weidenfeld J, Yang J, Liu genes. Nat Immunol. 2006 Jun;7(6):616-24 F, Chokas AL, Morrisey EE. Foxp1 coordinates Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: cardiomyocyte proliferation through both cell-autonomous the guardian of B cell identity and function. Nat Immunol. and nonautonomous mechanisms. Genes Dev. 2010 Aug 2007 May;8(5):463-70 15;24(16):1746-57 Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan- This article should be referenced as such: Smith E, Dalton JD, Girtman K, Mathew S, Ma J, Pounds SB, Su X, Pui CH, Relling MV, Evans WE, Shurtleff SA, Huret JL. dic(3;9)(p14;p13) PAX5/FOXP1. Atlas Genet Downing JR. Genome-wide analysis of genetic alterations Cytogenet Oncol Haematol. 2011; 15(5):466-467. in acute lymphoblastic leukaemia. Nature. 2007 Apr 12;446(7137):758-64

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 467

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI dic0918p13q11ID1556.txt

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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 468 dic(9;18)(p13;q11) PAX5/ZNF521 Huret JL

Fusion protein

SB, Su X, Pui CH, Relling MV, Evans WE, Shurtleff SA, Description Downing JR. Genome-wide analysis of genetic alterations The predicted fusion protein contains the DNA in acute lymphoblastic leukaemia. Nature. 2007 Apr binding paired domain of PAX5 and the DNA- 12;446(7137):758-64 binding and transcriptional regulator domain of Bond HM, Mesuraca M, Amodio N, Mega T, Agosti V, ZNF521. 1541 amino acids. Fanello D, Pelaggi D, Bullinger L, Grieco M, Moore MA, Venuta S, Morrone G. Early hematopoietic zinc finger protein-zinc finger protein 521: a candidate regulator of References diverse immature cells. Int J Biochem Cell Biol. Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, 2008;40(5):848-54 Busslinger M. Pax5 induces V-to-DJ rearrangements and Lobo MK, Yeh C, Yang XW. Pivotal role of early B-cell locus contraction of the immunoglobulin heavy-chain gene. factor 1 in development of striatonigral medium spiny Genes Dev. 2004 Feb 15;18(4):411-22 neurons in the matrix compartment. J Neurosci Res. 2008 Johnson K, Pflugh DL, Yu D, Hesslein DG, Lin KI, Bothwell Aug 1;86(10):2134-46 AL, Thomas-Tikhonenko A, Schatz DG, Calame K. B cell- Wu M, Hesse E, Morvan F, Zhang JP, Correa D, Rowe specific loss of histone 3 lysine 9 methylation in the V(H) GC, Kiviranta R, Neff L, Philbrick WM, Horne WC, Baron locus depends on Pax5. Nat Immunol. 2004 Aug;5(8):853- R. Zfp521 antagonizes Runx2, delays osteoblast 61 differentiation in vitro, and promotes bone formation in Zhang Z, Espinoza CR, Yu Z, Stephan R, He T, Williams vivo. Bone. 2009 Apr;44(4):528-36 GS, Burrows PD, Hagman J, Feeney AJ, Cooper MD. Hesse E, Kiviranta R, Wu M, Saito H, Yamana K, Correa Transcription factor Pax5 (BSAP) transactivates the RAG- D, Atfi A, Baron R. Zinc finger protein 521, a new player in mediated V(H)-to-DJ(H) rearrangement of immunoglobulin bone formation. Ann N Y Acad Sci. 2010 Mar;1192:32-7 genes. Nat Immunol. 2006 Jun;7(6):616-24 Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: This article should be referenced as such: the guardian of B cell identity and function. Nat Immunol. Huret JL. dic(9;18)(p13;q11) PAX5/ZNF521. Atlas Genet 2007 May;8(5):463-70 Cytogenet Oncol Haematol. 2011; 15(5):468-469. Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan- Smith E, Dalton JD, Girtman K, Mathew S, Ma J, Pounds

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 469

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 Printable original version : http://documents.irevues.inist.fr/bitstream/DOI t0106VellosoID100047.txt

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 Rearranged Ig Tcr lymphoma (CLL/SLL) diagnosed 19 months earlier, in first relapse after 4 cycles of RFC Not done (Fludarabine, cyclophosphamide and Rituximab), Pathology no inborn condition of note. Lymph node biopsy showed SLL/CLL, CD20, Organomegaly CD23, CD5, CD43 and BCL2 positive; CD10 and No hepatomegaly, no splenomegaly, enlarged Cyclin D1 negative and Ki-67 positive in 25% of lymph nodes (diffuse lymphadenopathy), neoplastic cells. no central nervous 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 Survival study: 69.8% lymphoid cells, CD19+, CD5++, Date of diagnosis: 03-2010 CD11c+, CD23++, cyIgM+, cylambda+. Treatment: Bendamustine +Rituximab (2 cycles).

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 470

Chronic lymphocytic leukaemia/Small lymphocytic lymphoma Rodrigues Pereira Velloso ED, et al. (CLL/SLL) 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.

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

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

Chronic lymphocytic leukaemia/Small lymphocytic lymphoma Rodrigues Pereira Velloso ED, et al. (CLL/SLL) associated with translocation t(1;6)(p35;p25) as part of complex karyotype

aberration in "unmutated" B-CLL. Leukemia. 2005 Comments Jan;19(1):77-82 Until now, 16 patients with CLL and Rodrigues Pereira Velloso ED, Ratis C, Brasil SAB, Guerra t(1;6)(p35;p25) were described. 8 of these patients JC, Bacal N, Pitangueira CPMLM.. Translocation t(1;6)(p35;p25) in B-cell lymphoproliferative disorder with were described by Michaux et al. in 2005, showing evolution to Diffuse Large B-cell Lymphoma. Atlas Genet that this rearrangement was associated with bad Cytogenet Oncol Haematol. July 2007. URL : prognosis: unmutated B-CLL and evolution to http://AtlasGeneticsOncology.org/Reports/0106RodriguesI diffuse large B-cell Lymphoma (DLBCL). Our D100030.html group also described in 2007 a case of atypical CLL Van Den Neste E, Robin V, Francart J, Hagemeijer A, Stul with evolution to aggressive B-cell Lymphoma. In M, Vandenberghe P, Delannoy A, Sonet A, Deneys V, Costantini S, Ferrant A, Robert A, Michaux L. the case reported herein, clinical and pathological Chromosomal translocations independently predict evolution was associated with the detection of treatment failure, treatment-free survival and overall t(1;6) as part of complex karyotype including survival in B-cell chronic lymphocytic leukemia patients deletion of p53. Although no transformation to treated with cladribine. Leukemia. 2007 Aug;21(8):1715-22 DLBCL was seen, increase in proliferation rate in This article should be referenced as such: lymph node biopsy (Ki-67 increase from less than10 to 25%) was detected, associated with bad Rodrigues Pereira Velloso ED, Borri D, Alonso Ratis C, Fleury Perin G, Hamerschlak N, Bacal NS, Silveira PAA, prognosis and short survival. Bezerra AMPS, Pasqualin DC. Chronic lymphocytic leukaemia/Small lymphocytic lymphoma (CLL/SLL) References associated with translocation t(1;6)(p35;p25) as part of complex karyotype. Atlas Genet Cytogenet Oncol Michaux L, Wlodarska I, Rack K, Stul M, Criel A, Haematol. 2011; 15(5):470-472. Maerevoet M, 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

Atlas Genet Cytogenet Oncol Haematol. 2011; 15(5) 472

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