95

Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

  • Upload
    vuthuan

  • View
    232

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics
Page 2: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

The PDF version of the Atlas of Genetics and Cytogenetics in Oncology and Haematology is a reissue of the original articles published in collaboration with the

Institute for Scientific and Technical Information (INstitut de l’Information Scientifique et Technique - INIST) of the French National Center for Scientific Research

(CNRS) on its electronic publishing platform I-Revues.

Online and PDF versions of the Atlas of Genetics and Cytogenetics in Oncology and Haematology are hosted by INIST-CNRS.

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Scope

The Atlas of Genetics and Cytogenetics in Oncology and Haematology is a peer reviewed on-line journal in open

access, devoted to genes, 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

Page 3: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

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

Page 4: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Volume 14, Number 7, July 2010

Table of contents

Gene Section

APLNR (apelin receptor) 627 Yves Audigier

CD9 (CD9 molecule) 630 Laure Humbert, Mario Chevrette

CITED4 (Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 4) 633 Miguel Torres-Martin, Juan Antonio Rey

ENO1 (Enolase 1, (alpha)) 635 Bogusz Trojanowicz, Cuong Hoang-Vu, Carsten Sekulla

LIMK1 (LIM domain kinase 1) 641 Ratna Chakrabarti

PAX6 (paired box 6) 645 Yi-Hong Zhou

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) 652 Luke B Hesson, Farida Latif

SEMA3B (sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3B) 662 Munmi Bhattacharyya, Ranjan Tamuli

TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15) 665 Gui-Li Yang, Jian-Wei Qi, Zhi-Song Zhang, Lu-Yuan Li

BAP1 (BRCA1 associated protein-1 (ubiquitin carboxy-terminal hydrolase)) 670 Frédéric Guénard, Francine Durocher

CDA (Cytidine Deaminase) 673 Yoshiro Saito

CKS1B (CDC28 protein kinase regulatory subunit 1B) 676 Yongyou Zhang

COL16A1 (collagen, type XVI, alpha 1) 679 Susanne Grässel, Sabine Ratzinger

COPS2 (COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)) 688 Susanne Jennek, Florian Kraft, Aria Baniahmad

Leukaemia Section

t(3;6)(q27;p21) 692 Jean-Loup Huret

t(3;6)(q27;p21) PIM1/BCL6 694 Jean-Loup Huret

Page 5: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

t(11;14)(q13;q32) in multiple myeloma Huret JL, Laï JL

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(3;6)(q27;p21) SFRS3/BCL6 695 Jean-Loup Huret

t(8;20)(p11;q13) 696 Marie-Joëlle Mozziconacci, Christine Pérot

Solid Tumour Section

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma 698 DunFa Peng, Wael El-Rifai

Head and neck: Retinoblastoma 704 Hayyam Kiratli, Berçin Tarlan

Case Report Section

t(1;21)(p32;q22) as a non-random abnormality in AML M4 710 Lena Reindl, Claudia Haferlach

t(3;7)(q26;q21) as a secondary abnormality in MDS RAEB-2 712 Lena Reindl, Claudia Haferlach

Page 6: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Page 7: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 627

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

APLNR (apelin receptor) Yves Audigier

Unite INSERM U-858, I2MR, equipe 13, CHU Rangueil, Bat. L3, BP 84225, 1 avenue Jean-Poulhes, 31432-

Toulouse Cedex 4, France (YA)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/APLNRID44364ch11q12.html DOI: 10.4267/2042/44792

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: AGTRL1; APJ; APJR; FLJ90771;

HG11; MGC45246

HGNC (Hugo): APLNR

Location: 11q12.1

DNA/RNA

Description

1 exon.

Transcription

3.8 kb mRNA; 1140 bp open reading frame.

Protein

Description

380 amino acids.

Expression

Blood vessels, hypothalamus, heart, stomach, colon,

endocrine pancreas, bone, skeletic muscle, spleen.

Localisation

Plasma membrane.

Function

The apelin receptor APJ belongs to the family of G

protein-coupled receptors (O'Dowd et al., 1993; Devic

et al., 1996; Devic et al., 1999; Scott et al., 2007) and is

coupled to a Gi/o protein (Masri et al., 2006). Its

activation leads to the regulation of various

intracellular effectors with the following consequences:

adenylylcyclase inhibition (Masri et al., 2006; Habata

et al., 1999), increase of intracellular calcium (Choe et

al., 2000) and activation of extracellular signal-

regulated kinases (ERKs), PI-3K, Akt or p70S6 kinase

(S6K1) (Masri et al., 2006; Masri et al., 2004 ).

Expression of apelin receptors by the endothelial cell

(Devic et al., 1996; Devic et al., 1999) is associated

with two effects :

1) NO release leading to vessel vasodilatation

(Tatemoto et al., 2001) and peripheral hypotension

(Lee et al., 2000);

2) cell proliferation and migration (Masri et al., 2004;

Kasai et al., 2004) linked to angiogenesis (Cox et al.,

2006).

Page 8: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

APLNR (apelin receptor) Audigier Y

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 628

The expression in the central nervous system is high in

hypothalamus (De Mota et al., 2000; O'Carroll et al.,

2000) where receptor activation leads to the decrease of

vasopressin release (De Mota et al., 2004). Activation

of apelin receptors expressed by cardiomyocytes results

in a strong inotropic effect (Szokodi et al., 2002). In

stomach, apelin receptors are expressed by

enterochromaffin-like cells where their stimulation

decreases gastrin-induced acid secretion (Lambrecht et

al., 2006). Apelin receptors may regulate epithelial

proliferation in the colon (Han et al., 2007). Expression

of apelin receptors both in endocrine pancreas and

skeletic muscle contributes to the regulation of insulin

plasma levels and glucose uptake (Sorhede Winzell et

al., 2005; Dray et al., 2008). Apelin signalling also

increases osteoblast proliferation (Xie et al., 2007) and

cytokine expression by T lymphocytes (Habata et al.,

1999).

Mutations

Note

No mutation has been presently described.

Implicated in

Malignant glioma

Note

On a quantitative point of view, APLNR gene

expression is highly upregulated in microvascular

proliferations of malignant gliomas (Kalin et al., 2007).

Various diseases

Note

On a qualitative point of view, two single nucleotide

polymorphisms (SNP) have been reported. A functional

SNP in an Sp1-binding site of APLNR gene is

associated with susceptibility to brain infarction (Hata

et al., 2007). The 212A variant of the APJ receptor

gene is associated with slower heart failure progression

in idiopathic dilated cardiomyopathy (Sarzani et al.,

2007).

References O'Dowd BF, Heiber M, Chan A, Heng HH, Tsui LC, Kennedy JL, Shi X, Petronis A, George SR, Nguyen T. A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene. 1993 Dec 22;136(1-2):355-60

Devic E, Paquereau L, Vernier P, Knibiehler B, Audigier Y. Expression of a new G protein-coupled receptor X-msr is associated with an endothelial lineage in Xenopus laevis. Mech Dev. 1996 Oct;59(2):129-40

Devic E, Rizzoti K, Bodin S, Knibiehler B, Audigier Y. Amino acid sequence and embryonic expression of msr/apj, the mouse homolog of Xenopus X-msr and human APJ. Mech Dev. 1999 Jun;84(1-2):199-203

Habata Y, Fujii R, Hosoya M, Fukusumi S, Kawamata Y, Hinuma S, Kitada C, Nishizawa N, Murosaki S, Kurokawa T, Onda H, Tatemoto K, Fujino M. Apelin, the natural ligand of the

orphan receptor APJ, is abundantly secreted in the colostrum. Biochim Biophys Acta. 1999 Oct 13;1452(1):25-35

Choe W, Albright A, Sulcove J, Jaffer S, Hesselgesser J, Lavi E, Crino P, Kolson DL. Functional expression of the seven-transmembrane HIV-1 co-receptor APJ in neural cells. J Neurovirol. 2000 May;6 Suppl 1:S61-9

De Mota N, Lenkei Z, Llorens-Cortès C. Cloning, pharmacological characterization and brain distribution of the rat apelin receptor. Neuroendocrinology. 2000 Dec;72(6):400-7

Lee DK, Cheng R, Nguyen T, Fan T, Kariyawasam AP, Liu Y, Osmond DH, George SR, O'Dowd BF. Characterization of apelin, the ligand for the APJ receptor. J Neurochem. 2000 Jan;74(1):34-41

O'Carroll AM, Selby TL, Palkovits M, Lolait SJ. Distribution of mRNA encoding B78/apj, the rat homologue of the human APJ receptor, and its endogenous ligand apelin in brain and peripheral tissues. Biochim Biophys Acta. 2000 Jun 21;1492(1):72-80

Tatemoto K, Takayama K, Zou MX, Kumaki I, Zhang W, Kumano K, Fujimiya M. The novel peptide apelin lowers blood pressure via a nitric oxide-dependent mechanism. Regul Pept. 2001 Jun 15;99(2-3):87-92

Szokodi I, Tavi P, Földes G, Voutilainen-Myllylä S, Ilves M, Tokola H, Pikkarainen S, Piuhola J, Rysä J, Tóth M, Ruskoaho H. Apelin, the novel endogenous ligand of the orphan receptor APJ, regulates cardiac contractility. Circ Res. 2002 Sep 6;91(5):434-40

De Mota N, Reaux-Le Goazigo A, El Messari S, Chartrel N, Roesch D, Dujardin C, Kordon C, Vaudry H, Moos F, Llorens-Cortes C. Apelin, a potent diuretic neuropeptide counteracting vasopressin actions through inhibition of vasopressin neuron activity and vasopressin release. Proc Natl Acad Sci U S A. 2004 Jul 13;101(28):10464-9

Kasai A, Shintani N, Oda M, Kakuda M, Hashimoto H, Matsuda T, Hinuma S, Baba A. Apelin is a novel angiogenic factor in retinal endothelial cells. Biochem Biophys Res Commun. 2004 Dec 10;325(2):395-400

Masri B, Morin N, Cornu M, Knibiehler B, Audigier Y. Apelin (65-77) activates p70 S6 kinase and is mitogenic for umbilical endothelial cells. FASEB J. 2004 Dec;18(15):1909-11

Sörhede Winzell M, Magnusson C, Ahrén B. The apj receptor is expressed in pancreatic islets and its ligand, apelin, inhibits insulin secretion in mice. Regul Pept. 2005 Nov;131(1-3):12-7

Cox CM, D'Agostino SL, Miller MK, Heimark RL, Krieg PA. Apelin, the ligand for the endothelial G-protein-coupled receptor, APJ, is a potent angiogenic factor required for normal vascular development of the frog embryo. Dev Biol. 2006 Aug 1;296(1):177-89

Lambrecht NW, Yakubov I, Zer C, Sachs G. Transcriptomes of purified gastric ECL and parietal cells: identification of a novel pathway regulating acid secretion. Physiol Genomics. 2006 Mar 13;25(1):153-65

Masri B, Morin N, Pedebernade L, Knibiehler B, Audigier Y. The apelin receptor is coupled to Gi1 or Gi2 protein and is differentially desensitized by apelin fragments. J Biol Chem. 2006 Jul 7;281(27):18317-26

Xie H, Tang SY, Cui RR, Huang J, Ren XH, Yuan LQ, Lu Y, Yang M, Zhou HD, Wu XP, Luo XH, Liao EY. Apelin and its receptor are expressed in human osteoblasts. Regul Pept. 2006 May 15;134(2-3):118-25

Han S, Wang G, Qiu S, de la Motte C, Wang HQ, Gomez G, Englander EW, Greeley GH Jr. Increased colonic apelin production in rodents with experimental colitis and in humans with IBD. Regul Pept. 2007 Aug 16;142(3):131-7

Page 9: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

APLNR (apelin receptor) Audigier Y

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 629

Hata J, Matsuda K, Ninomiya T, Yonemoto K, Matsushita T, Ohnishi Y, Saito S, Kitazono T, Ibayashi S, Iida M, Kiyohara Y, Nakamura Y, Kubo M. Functional SNP in an Sp1-binding site of AGTRL1 gene is associated with susceptibility to brain infarction. Hum Mol Genet. 2007 Mar 15;16(6):630-9

Kälin RE, Kretz MP, Meyer AM, Kispert A, Heppner FL, Brändli AW. Paracrine and autocrine mechanisms of apelin signaling govern embryonic and tumor angiogenesis. Dev Biol. 2007 May 15;305(2):599-614

Sarzani R, Forleo C, Pietrucci F, Capestro A, Soura E, Guida P, Sorrentino S, Iacoviello M, Romito R, Dessì-Fulgheri P, Pitzalis M, Rappelli A. The 212A variant of the APJ receptor gene for the endogenous inotrope apelin is associated with slower heart failure progression in idiopathic dilated cardiomyopathy. J Card Fail. 2007 Sep;13(7):521-9

Scott IC, Masri B, D'Amico LA, Jin SW, Jungblut B, Wehman AM, Baier H, Audigier Y, Stainier DY. The g protein-coupled receptor agtrl1b regulates early development of myocardial progenitors. Dev Cell. 2007 Mar;12(3):403-13

Dray C, Knauf C, Daviaud D, Waget A, Boucher J, Buléon M, Cani PD, Attané C, Guigné C, Carpéné C, Burcelin R, Castan-Laurell I, Valet P. Apelin stimulates glucose utilization in normal and obese insulin-resistant mice. Cell Metab. 2008 Nov;8(5):437-45

This article should be referenced as such:

Audigier Y. APLNR (apelin receptor). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):627-629.

Page 10: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 630

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

CD9 (CD9 molecule) Laure Humbert, Mario Chevrette

The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada

(LH, MC)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/CD9ID995ch12p13.html DOI: 10.4267/2042/44793

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: 5H9; BA2; P24; GIG2; MIC3; MRP-1;

BTCC-1; DRAP-27; TSPAN29

HGNC (Hugo): CD9

Location: 12p13.31

Local order: The CD9 gene is located between the

VWF and the ATP5J2P5 genes.

DNA/RNA

Description

The gene spans 38 kb of DNA, including a 10 kb intron

separating the first two exons. CD9 encodes 8 exons,

ranging from 63 to 109 base pairs. The coding

sequence is highly conserved between species. The

promoter contains neither TATA nor CAAT boxes, but

does contain several consensus sequences for the

binding of transcription factors (GATA, ETS, E2F, NF-

kB, AP2) as well as three putative Sp1 binding sites.

Transcription

The CD9 transcribed RNA has 1246 bases, of which

684 bases (from 112 (Met) to 795 (Val)) encode the

protein.

Pseudogene

None.

Protein

Description

CD9 is a member of the transmembrane 4 superfamily,

also called the tetraspanin family. As other

tetraspanins, CD9 is a cell-surface protein containing

four hydrophobic transmembrane domains (indicated in

green) and two extracellular domains (illustrated in

violet). CD9 consists of 228 amino acids and weighs

24-27 kDa. CD9 contains four small and highly

conserved hydrophobic transmembrane domains (24-27

amino acids); a small N-terminal (11 amino acids) and

a C-terminal cytoplasmic (7 amino acids) tails, and a

very small intracellular domain (4 amino acids). The

remaining part of the protein is composed of two

extracellular domains (also called loops; a small one of

20 amino acids and a large one of 83 amino acids).

Two disulfide bonds, generated by four well-conserved

cysteine residues (C), stabilize the large extracellular

domain. CD9 also contains a tetraspanin signature

(amino acids 65-89) and a CCG motif (amino acids 152

to 154), but lacks

Genomic organisation of the CD9 gene on chromosome 12.

Page 11: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CD9 (CD9 molecule) Humbert L, Chevrette M

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 631

Structure of the CD9 protein.

other motifs found on other tetraspanins (DW, PxSc3,

Gc4).

Expression

CD9 is expressed by a variety of hematopoietic and

epithelial cells. It is transiently expressed during

development of spinal motoneurons and other fetal

nervous system sites, as well as in hematopoietic

development. CD9 is glycosylated (the glycosylation

site is in the first extracellular loop unlike most

glycosylated tetraspanins where the site is located in

the second extracellular loop) and acylated. CD9 is also

phosphorylated on tyrosine following B-cell activation.

CD9 is up-regulated on activated B and T lymphocytes.

Localisation

In normal cells, CD9 localizes mainly in the

membranes while in cancer cells the protein may also

be detected throughout the cytoplasm.

Function

CD9 can interact or form complexes with many other

proteins, including other tetraspanins, integrins, EWI

molecules, TGF-a, diphtheria toxin receptor, receptor

tyrosine kinase, pregnancy specific glycoproteins, and

proteins of the immune system such as MHC class II

molecules and members of the Ig superfamily.

Moreover, probably because of its localization in the

cell membrane, CD9 is involved in platelet activation

and aggregation, as well as in cell adhesion, spreading,

cell motility and tumor metastasis. CD9 also regulates

paranodal junction formation, and is required for

gamete fusion. Furthermore, CD9 promotes muscle cell

fusion and supports myotube maintenance.

Homology

Although there are variations in the amino acid

sequence in the extracellular loops, the CD9 protein

sequence is very well conserved between species (90%

between human, mice and rat). CD9 share also some

homologies with other tetraspanins, particularly in the

transmembrane domains.

Mutations

Note

Although no genomic CD9 mutation has been reported,

in prostate cancer, there is mention of cDNA mutation

compatible with an RNA editing mechanism. So far,

CD9 has never been implicated in gene fusion that

could result in a modified protein.

Implicated in

Various cancers

Note

Decreased expression of the CD9 protein has been

associated with many types of cancer.

Disease

- Expressed in 90% of non-T cell acute lymphoblastic

leukemia cells and in 50% of chronic lymphocytic

leukemia and acute myeloblastic leukemia.

- Expression inversely correlated with metastatic

potential of melanoma.

- Expression suppresses motility and metastasis of

carcinoma cells.

- Reduction of expression correlated with poor

prognosis in breast, lung and colon carcinomas.

Page 12: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CD9 (CD9 molecule) Humbert L, Chevrette M

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 632

References Boucheix C, Nguyen-van-Cong, Perrot JY, Foubert C, Gross MS, Weil D, Laisney V, Rosenfeld C, Frezal J. Assignment to chromosome 12 of the gene coding for the human cell surface antigen CD9(p24) using the monoclonal antibody ALB6. Ann Genet. 1985;28(1):19-24

Rendu F, Boucheix C, Lebret M, Bourdeau N, Benoit P, Maclouf J, Soria C, Levy-Toledano S. Mechanisms of the mAb ALB6(CD9) induced human platelet activation: comparison with thrombin. Biochem Biophys Res Commun. 1987 Aug 14;146(3):1397-404

Seehafer JG, Tang SC, Slupsky JR, Shaw AR. The functional glycoprotein CD9 is variably acylated: localization of the variably acylated region to a membrane-associated peptide containing the binding site for the agonistic monoclonal antibody 50H.19. Biochim Biophys Acta. 1988 Dec 2;957(3):399-410

Boucheix C, Benoit P, Frachet P, Billard M, Worthington RE, Gagnon J, Uzan G. Molecular cloning of the CD9 antigen. A new family of cell surface proteins. J Biol Chem. 1991 Jan 5;266(1):117-22

Ikeyama S, Koyama M, Yamaoko M, Sasada R, Miyake M. Suppression of cell motility and metastasis by transfection with human motility-related protein (MRP-1/CD9) DNA. J Exp Med. 1993 May 1;177(5):1231-7

Rubinstein E, Benoit P, Billard M, Plaisance S, Prenant M, Uzan G, Boucheix C. Organization of the human CD9 gene. Genomics. 1993 Apr;16(1):132-8

Si Z, Hersey P. Expression of the neuroglandular antigen and analogues in melanoma. CD9 expression appears inversely related to metastatic potential of melanoma. Int J Cancer. 1993 Apr 22;54(1):37-43

Tole S, Patterson PH. Distribution of CD9 in the developing and mature rat nervous system. Dev Dyn. 1993 Jun;197(2):94-106

Higashiyama M, Taki T, Ieki Y, Adachi M, Huang CL, Koh T, Kodama K, Doi O, Miyake M. Reduced motility related protein-1 (MRP-1/CD9) gene expression as a factor of poor prognosis in non-small cell lung cancer. Cancer Res. 1995 Dec 15;55(24):6040-4

Shaw AR, Domanska A, Mak A, Gilchrist A, Dobler K, Visser L, Poppema S, Fliegel L, Letarte M, Willett BJ. Ectopic expression of human and feline CD9 in a human B cell line confers beta 1 integrin-dependent motility on fibronectin and laminin substrates and enhanced tyrosine phosphorylation. J Biol Chem. 1995 Oct 13;270(41):24092-9

Le Naour F, Prenant M, Francastel C, Rubinstein E, Uzan G, Boucheix C. Transcriptional regulation of the human CD9

gene: characterization of the 5'-flanking region. Oncogene. 1996 Aug 1;13(3):481-6

Miyake M, Nakano K, Itoi SI, Koh T, Taki T. Motility-related protein-1 (MRP-1/CD9) reduction as a factor of poor prognosis in breast cancer. Cancer Res. 1996 Mar 15;56(6):1244-9

Cajot JF, Sordat I, Silvestre T, Sordat B. Differential display cloning identifies motility-related protein (MRP1/CD9) as highly expressed in primary compared to metastatic human colon carcinoma cells. Cancer Res. 1997 Jul 1;57(13):2593-7

Maecker HT, Todd SC, Levy S. The tetraspanin superfamily: molecular facilitators. FASEB J. 1997 May;11(6):428-42

Horváth G, Serru V, Clay D, Billard M, Boucheix C, Rubinstein E. CD19 is linked to the integrin-associated tetraspans CD9, CD81, and CD82. J Biol Chem. 1998 Nov 13;273(46):30537-43

Cook GA, Wilkinson DA, Crossno JT Jr, Raghow R, Jennings LK. The tetraspanin CD9 influences the adhesion, spreading, and pericellular fibronectin matrix assembly of Chinese hamster ovary cells on human plasma fibronectin. Exp Cell Res. 1999 Sep 15;251(2):356-71

Tachibana I, Hemler ME. Role of transmembrane 4 superfamily (TM4SF) proteins CD9 and CD81 in muscle cell fusion and myotube maintenance. J Cell Biol. 1999 Aug 23;146(4):893-904

Miyado K, Yamada G, Yamada S, Hasuwa H, Nakamura Y, Ryu F, Suzuki K, Kosai K, Inoue K, Ogura A, Okabe M, Mekada E. Requirement of CD9 on the egg plasma membrane for fertilization. Science. 2000 Jan 14;287(5451):321-4

Seigneuret M, Delaguillaumie A, Lagaudrière-Gesbert C, Conjeaud H. Structure of the tetraspanin main extracellular domain. A partially conserved fold with a structurally variable domain insertion. J Biol Chem. 2001 Oct 26;276(43):40055-64

Ishibashi T, Ding L, Ikenaka K, Inoue Y, Miyado K, Mekada E, Baba H. Tetraspanin protein CD9 is a novel paranodal component regulating paranodal junctional formation. J Neurosci. 2004 Jan 7;24(1):96-102

Kovalenko OV, Metcalf DG, DeGrado WF, Hemler ME. Structural organization and interactions of transmembrane domains in tetraspanin proteins. BMC Struct Biol. 2005 Jun 28;5:11

Wang JC, Bégin LR, Bérubé NG, Chevalier S, Aprikian AG, Gourdeau H, Chevrette M. Down-regulation of CD9 expression during prostate carcinoma progression is associated with CD9 mRNA modifications. Clin Cancer Res. 2007 Apr 15;13(8):2354-61

This article should be referenced as such:

Humbert L, Chevrette M. CD9 (CD9 molecule). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):630-632.

Page 13: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 633

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

CITED4 (Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 4) Miguel Torres-Martin, Juan Antonio Rey

Unidad de Investigacion del Hospital Universitario La Paz, Madrid, Spain (MTM, JAR)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/CITED4ID44535ch1p34.html DOI: 10.4267/2042/44794

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: MRG2; MRG-2

HGNC (Hugo): CITED4

Location: 1p34.2

DNA/RNA

Description

DNA sequence is located at chromosome 1p.

Transcription

Transcription consists of a single exon without

alternative splicing. mRNA: NM_133467.

Protein

Note

CITED4 protein is 184 amino acid long with a

molecular weight of 18569 Da.

NP_597724.

Description

CITED4 has a characteristic CITED domain motif

conserved in all CITED peptides located at the

carboxyl-terminal domain that binds with p300/CBP.

Expression

In all tissues with special intensity in heart, liver,

pancreas and skeletal muscle.

Localisation

CITED4 has nuclear and cytoplasmatic location. In

most cells it has a nuclear localization, but in others it

was localized in nucleus and cytoplasm.

Function

Binds CBP and tumor suppressor protein EP300 by

carboxy terminus domain (residues 138-184).

Therefore it may be implicated in gene transcription.

As other genes of the family, CITED4 physically

interacts with transcription factor AP-2.

A. Position of CITED4 in the chromosome 1. B. Flanking genes of CITED4.

Page 14: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CITED4 (Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 4) Torres-Martin M, Rey JA

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 634

Coding and flanking regions of CITED4.

Fox et al. (2002) showed that CITED4 blocks the

binding of hypoxia-inducible factor 1alpha to p300 in

their experiments made in vitro and inhibits hypoxia-

inducible factor-1alpha transactivation and hypoxia-

mediated reporter gene activation. That is the reason

why they concluded that CITED4 might be an inhibitor

of hypoxia-inducible factor 1alpha.

Homology

CITED4 has 2 paralogues (CITED1 and CITED2) in

humans. All of them belong to CITED family, found

only in jawed vertebrates to date (Braganca et al.,

2002).

Mutations

Note

No mutations has been reported yet, but a total of 16

polymorphisms with unknown consequences has been

founded by Tews et al. (2007) and Torres-Martin et al.

(2008).

Implicated in

Oligodendroglioma

Note

CITED4 promoter is methylated in

oligodendrogliomas, especially in those with 1p/19q

deletions. This hypermethylation is responsible of

lower levels of CITED4 mRNA expression, suggesting

a way by which CITED4 is almost silenced by both

hypermethylation and chromosomal deletion (Tews et

al., 2007).

Prognosis

CITED4 hypermethylation in oligodendroglioma

patients is similar to prognosis associated to 1p/19q

deletions. Thus, CITED4 hypermethylation might be an

alternative or even a confirmation of 1p/19q testing.

Breast cancer

Note

Cytoplasmatic translocation and loss of nuclear

expression has been associated with breast cancer by

Fox et al. (2002). This loss may allow p300/CBP to

interact with hypoxia-inducible factor 1a and

oncogenes to enhance their transcriptional activity

leading to an aggressive tumor phenotype (Fox et al.,

2004).

Prognosis

CITED4 is located in the nucleus in normal tissue, but

in breast tumors is present both nuclear and

cytoplasmatic location. This characteristic might be

used as prognosis factor of this kind of tumors.

References Bragança J, Swingler T, Marques FI, Jones T, Eloranta JJ, Hurst HC, Shioda T, Bhattacharya S. Human CREB-binding protein/p300-interacting transactivator with ED-rich tail (CITED) 4, a new member of the CITED family, functions as a co-activator for transcription factor AP-2. J Biol Chem. 2002 Mar 8;277(10):8559-65

Yahata T, Takedatsu H, Dunwoodie SL, Bragança J, Swingler T, Withington SL, Hur J, Coser KR, Isselbacher KJ, Bhattacharya S, Shioda T. Cloning of mouse Cited4, a member of the CITED family p300/CBP-binding transcriptional coactivators: induced expression in mammary epithelial cells. Genomics. 2002 Dec;80(6):601-13

Fox SB, Bragança J, Turley H, Campo L, Han C, Gatter KC, Bhattacharya S, Harris AL. CITED4 inhibits hypoxia-activated transcription in cancer cells, and its cytoplasmic location in breast cancer is associated with elevated expression of tumor cell hypoxia-inducible factor 1alpha. Cancer Res. 2004 Sep 1;64(17):6075-81

Tews B, Roerig P, Hartmann C, Hahn M, Felsberg J, Blaschke B, Sabel M, Kunitz A, Toedt G, Neben K, Benner A, von Deimling A, Reifenberger G, Lichter P. Hypermethylation and transcriptional downregulation of the CITED4 gene at 1p34.2 in oligodendroglial tumours with allelic losses on 1p and 19q. Oncogene. 2007 Jul 26;26(34):5010-6

Torres-Martín M, Franco-Hernandez C, Martinez-Glez V, de Campos JM, Isla A, Casartelli C, Rey JA. Mutational analysis of the CITED4 gene in glioblastomas. Cancer Genet Cytogenet. 2008 Sep;185(2):114-6

This article should be referenced as such:

Torres-Martin M, Rey JA. CITED4 (Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 4). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):633-634.

Page 15: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 635

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

ENO1 (Enolase 1, (alpha)) Bogusz Trojanowicz, Cuong Hoang-Vu, Carsten Sekulla

AG Experimentelle and Chirurgische Onkologie, Universitatsklinik und Poliklinik fur Allgemein-, Viszeral-

und Gefasschirurgie, Martin-Luther Universitat, Magdeburger Strasse 18, 06097 Halle/S, Germany (BT,

CHV, CS); AG Experimentelle and Chirurgische Onkologie, Universitatsklinik und Poliklinik fur

Kinderchirurgie, Martin-Luther Universitat, Magdeburger Strasse 18, 06097 Halle/S, Germany (BT)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/ENO1ID40453ch1p36.html DOI: 10.4267/2042/44795

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: EC 4.2.1.11; ENO1L1; MBP-1;

MBPB1; MPB-1; MPB1; NNE; PPH; tau-crystallin

HGNC (Hugo): ENO1

Location: 1p36.23

DNA/RNA

Note

Alpha-Enolase (ENO1, alpha enolase, non-neuronal

enolase) is one of the three enolase enzymes, expressed

in a wide variety of tissues. The other two enolase

genes, ENO2 and ENO3, encode gamma (neuron-

specific) and beta (muscle-specific) enolase,

respectively. The active enolase enzymes exist as

homodimers of non-covalently bound subunits. Each

alpha, beta or gamma subunit is encoded by separate

genes. The genomic organisation of ENO1 gene is

identical with that of human gamma-enolase gene. All

the coding exons have exactly the same length and

introns occur at analogous positions.

Description

The ENO1 gene consists of 12 exons distributed over

17718 bp of genomic DNA. Single alpha-enolase

transcript contains two translation initiation positions

and encodes two structurally and functionally distinct

proteins, alpha-enolase enzyme and MYC promotor-

binding protein (MBP-1).

Transcription

Transcription start sites of ENO1 gene are

heterogeneous and spread over 38-bp region located at

116 bp upstream from the initiation codon ATG. These

multiple start sites of transcription in ENO1 gene are

consistent with lack of a canonical TATA box, usually

found at the position 19-27 bp upstream of the cap

sites. It is worth to notice that promoter of ENO1 gene

contains two perfect Myc-Max binding motifs

CACGTC. Other regulatory sites found in the 5'-

flanking region of ENO1 gene include AP1

(T[T/G]AGTCA), AP2 (CCCCAGGC), AP3

(GGGTGTGGAAAG), AP4 (CAGCTGTGG), AP5

(CTGTGGAATG), ATF/CREB ([T/G][A/T]CGTCA),

C2 (CATGTG),

Structure of ENO1 mRNA ; note that nucleotides number (nt), exon positions (1-12) and two translation initiation sites (ATG) are labelled.

Page 16: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

ENO1 (Enolase 1, (alpha)) Trojanowicz B, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 636

Structure of ENO1/MBP-1 protein; N and C termini, and amino acid (aa) positions are labelled.

CTF/NF1 (TGGCTNNNAGCCAA), E2AE-C beta

(TGGGAATT), E2F (TTTCGCGC), E4TF1

(GGAAGTG), EF-C (GTTGCNNGGCAAC),

MLTF/USF (GGTCACGTGGCC), Ig octamer

(ATTTGCAT), PEA2 (GACCGCA), SP1 (GGGCGG),

CACCC (may function as CAAT boxes) and viral core

(GTGG[A/T][A/T][A/T]G).

Pseudogene

A pseudogene has been identified that is located on the

other arm of the same chromosome (provided by

RefSeq).

Protein

Description

Alpha-Enolase (ENO1), like two other isoenzymes

(gamma-ENO2 and beta-ENO3), is made up of two

identical (homodimer), non-covalently bound alpha

alpha subunits; alpha-Enolase is resolved during 1D-

PAGE as a protein with molecular weight of about 48

kDa (434 amino acids). It was demonstrated that in

brain and neurons, specific enolases may exist as

heterodimers, such as alpha alpha, alpha beta, beta beta,

alpha gamma and gamma gamma. The proportions of

isoenzymes alpha alpha, alpha beta and beta beta

change in heart and muscle during embryonic

development. In both mentioned tissues, isoform alpha

was found predominantly in fetus. In adult heart this

subunit is replaced by types alpha beta and beta beta,

and in muscle by type beta beta. In human adult brain

tissues, apha-type and gamma-type enolase subunits are

present at similar concentrations.

Two identical subunits of alpha-Enolase facilitate each

other in an antiparallel fashion. Each subunit is made

up of two distinct domains: N-terminal domain,

consisting of three beta-sheets and four alpha-helices

(beta3 alpha4 topology), and larger C-terminal domain

with eightfold alpha beta barrel structure with beta beta

alpha alpha(beta alpha)6 topology. This domain

contains two beta-sheets at the beginning, followed by

two alpha-helices and ends with a barrel made up of

alternating beta-sheets and alpha-helices (beta - sheets

are surrounded by the alpha - helices). The N-terminal

of one subunit contacts the C-terminal of the second in

such way, that glutamic acid at position 20 (Glu20)

forms an ionic pair with arginine at position 414

(Arg414).

Alternatively translated product of ENO1 gene, called

MBP-1 (MYC promotor-binding protein), is expressed

as a 37 kDa (338 amino acids) protein and does not

posses the enolase enzyme activity.

Expression

Apha-Enolase is widely expressed in variety of tissues

including liver, brain, kidney, spleen, adipose as well as

thyroid. In comparison with gamma-type subunit found

only in neurons, type alpha subunit was also detected in

astrocytes, ependymal cells, capillary endothelial cells,

Schwann cells and arachnoidal endothelial cells.

Localisation

Alpha-Enolase is most abundantly found in cytoplasm

and also on the cell surface. MBP-1 is localised in the

nucleus.

Function

Enolase enzymes (2-phospho-D-glycerate hydrolases)

catalyse the dehydration of 2-phospho-D-glycerate

(PGA) to phosphoenolopyruvate (PEP) in Emden

Mayerhoff-Parnas glycolytic pathway (catabolic

direction). In anabolic pathway (reverse reaction)

during gluconeogenesis, the same enzyme catalyses

hydration of PEP to PGA (hence it is called

phosphopyruvate hydratase). Metal ions are cofactors

impairing the increase of enolase activity; hence it is

also called metal-activated metalloenzyme. Magnesium

is a natural cofactor causing the highest activity. The

relative activation strength profile of metal ions

involved in the enzyme activity appears in the

following rank of order Mg2+

> Zn2+

> Mn2+

> Fe(II)2+

> Cd2+

> Co2+

, Ni2+

, Sm3+

, Tb3+

and most other divalent

metal ions. In reaction catalyzed by enolases, the alpha-

proton from a carbon adjacent to a carboxylate group of

Page 17: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

ENO1 (Enolase 1, (alpha)) Trojanowicz B, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 637

Reaction catalyzed by Enolase.

PGA, is abstracted, and PGA is conversed to enolate

anion intermediate. This intermediate is further

processed in a variety of chemical reactions, including

racemization, cycloisomerization and beta-elimination

of either water or ammonia.

The smaller product of ENO1 gene, MBP-1, is known

as c-myc binding protein and negative regulator of its

expression. C-myc is a DNA-binding phosphoprotein

and a key regulator of cell behaviour. Many of c-myc

targeting pathways are deregulated in cancer cells and

contribute to its enhanced expression. There are four c-

myc promoters, designated as P0, P1, P2 and

P3,although in normal and cancer cells most mRNAs

initiate at the P2 promoter. MBP-1 binds in a region

+123 to +153 relative to the c-myc P2 promoter and

probably by preventing the formation of a transcription

initiation complex, decrease c-myc promoter activity.

Hence MBP-1 is considered as tumor suppressor.

ENO1 protein was also found as a structural component

of the eye lenses and was designated as tau-crystalin.

ENO1 enzyme and tau-crystalin are the products of the

same gene. Tau-crystalins are the major components of

vertebrate lens. These proteins are mainly found in

monomeric form with a low enzymatic activity, while

the active ENO1 enzyme exists as a dimer. Irrespective

of ENO1 enzyme activity, its significant presence in

eye lens (23% of the total protein of the lens) clearly

indicate ENO1 structural role in lens and cataracts.

In hypoxic conditions elevated ENO1 levels may

provide protection to the cells by increasing anaerobic

metabolism.

Homology

Currently, amino acid sequences of more than 50

enolase enzymes are known. The five residues that

participate in catalytic activity of this enzyme are

highly conserved throughout evolution. Studies in vitro

revealed that mutant enolase enzymes that differs at

either positions Glu168, Glu211, Lys345, Lys396 or

His159, demonstrated dramatically decreased activity

level. An integral and conserved part of enolases are

two Mg2+

ions that participate in conformational

changes of the active site of enolase and enable binding

of a substrate or its analogues.

Mutations

Note

The ENO1 gene maps to a region of chromosome 1

(1p35-p36) reported to be often deleted in several

human malignancies, including neuroblastoma,

melanoma, pheochromocytoma, breast, liver and colon

cancer. However screening of neuroblastomas at

different stages, failed to detect any mutations in ENO1

gene.

Amplification of ENO1 gene, as well as PAX7 (region

1pter-p33) was found to be a common phenomenon in

squamous cell lung carcinoma.

Implicated in

Non-small cell lung cancer (NSCLC)

Note

Higher expression of ENO1 was demonstrated in

NSCLC tissues as compared with normal lung tissues.

Detection and expression level of ENO1 in primary

tumors were the key factors contributing to overall

patient's survival rates. Relatively higher ENO1 levels

in tumors correlated with poorer survival outcomes and

tumor recurrence.

Other report suggests that ENO1 down-regulation in

patients with NSCLC, predicts more aggressive

biological behaviour. The patients whose tumors

showed decreased ENO1 production had significantly

poorer overall survival when compared with those

without ENO1 reduction.

Also in proteomic studies, ENO1 was one of the

secreted proteins demonstrated to be overexpressed by

NSCLC cell line A549 as compared to controls.

Studies in vitro performed on NSCLC cell line H1299,

revealed that MBP-1 overexpression correlated with

decreased cell proliferation as compared with

corresponding controls. Investigations in vivo

demonstrated tumor suppressive properties of MBP-1.

In mice with induced tumors (injection of H1299)

administration of adenovirus MBP-1 construct

significantly reduced tumor growth and prolonged

animal survival rates.

Page 18: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

ENO1 (Enolase 1, (alpha)) Trojanowicz B, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 638

Small cell lung cancer

Note

There is some evidence concerning the role of anti-

alpha-enolase antibodies in cancer associated

retinopathy with SCLC. In serum obtained from patient

with a sudden loss of vision, the only detectable

antibodies were those against a 35-kDa anti-retinal

protein. Surgical treatment performed after 1 week and

1 month, led to changes in the antibody response from

antibodies against p35kDa to alpha-enolase after tumor

resection. SCLC may express high levels of alpha-

enolase and anti-alpha-enolase antibodies are typically

detected after diagnosis of cancer.

Thyroid carcinoma

Note

In thyroid oncocytomas, which represent a subgroup of

follicular thyroid carcinoma (FTC),the up-regulation of

ENO1, GPI (glucose phosphate isomerase) and

GAPDH (glyceraldehydes-3-phosphate dehydrogenase)

was identified as metabolic signature of thyroid

carcinoma.

Important role of ENO1 in progression of thyroid

carcinoma was also demonstrated for cell lines

established from FTC. Pre-treatment of these cell lines

with retinoic acid (RA) used in therapy and

chemoprevention of solid cancers, led to decrease in

ENO1 and MBP-1 expression, accompanied by

reduced invasiveness of the thyroid carcinoma cells.

Similar effects were also observed after silencing of

common the MYC promoter-binding domain found in

ENO1 and MBP-1. Both, RA-mediated and siRNA

induced reduction of ENO1/MBP-1 resulted in down-

regulation of c-Myc oncoprotein. It seems that in FTC

the bi-functional role of ENO1 gene products is

diminished and ENO1 posses the enzymatic activity

only. It is worth to notice that ENO1 promoter contains

two MYC binding sites (CACGTG). C-Myc over-

expression and interaction with these sites may result in

increased ENO1 expression and/or energy production.

In well differentiated medullary thyroid carcinomas

MTC the relatively high amount of alpha beta and

gamma gamma enolase isoenzymes was observed,

indicating presumed neuroectodermal origin of these

tumors. In highly undifferentiated and anaplastic

MTCs, the majority of enzyme was represented as

alpha alpha-enolase while alpha gamma-enolase was

only weakly detectable.

Hepatocellular carcinoma (HCC)

Note

In proteomic studies performed on HCC cell lines and

tissues, ENO1 was identified as a protein that showed

stronger expression in tumor tissues when comparing to

nontumorous samples. Additionally, expression of

ENO1 increased with tumor dedifferentiation status.

Significantly higher ENO1 expression was found in

poorly differentiated HCC than in well differentiated

HCC. Moreover, expression of ENO1 positively

correlated with tumor size and venous invasion. Also

reduction of ENO1 by specific siRNAs decreased the

proliferation rates of HCC cell lines and prolonged the

G2/M phase of the cell cycle.

Investigations of MBP-1 revealed its significant

reduction in cirrhosis and even more diminished

expression in HCC. This reduction was surprisingly

accompanied by decrease in c-myc expression.

Breast carcinoma

Note

Increased expression of ENO1 was found in HER-

2/neu positive breast tumors and cell lines when

compared with corresponding controls. HER-2/neu is

the receptor tyrosine kinase found to be overexpressed

in up to 30% of breast cancers and is associated with

increased metastasis rate and poor prognosis.

Introduction of MBP-1 gene into human breast

carcinoma cells MDA-MB-231 and MCF-7 reduced

their ability to penetrate basement membrane matrix

and suppressed tumor formation in athymic nude mice.

It is worth to notice that MCF-7 cell line is estrogen

receptor positive and estrogen dependent for

tumorigenicity.

It was demonstrated that translation of ENO1 mRNA in

MCF-7 cell line is glucose concentration-dependent.

Low glucose concentrations increased the level of

MBP-1 protein accompanied by reduced proliferation

rates. The levels of ENO1 mRNA remained unaffected.

This suggests that effects induced by low glucose

concentrations are mediated by preferential translation

of MBP-1 (using the down-stream ATG codon). In

contrast, physiologic or high glucose concentrations

correlated with reduced levels of MBP-1 protein and

markedly induced growth of the cells. Interestingly the

low glucose group exhibited a dramatic increase in c-

Myc expression, not observed in physiologic or high

glucose conditions. As demonstrated for follicular

thyroid carcinoma cells, also in this case c-Myc might

directly transactivate ENO1 promoter, resulting in an

increase in glucose uptake and elevated proliferation

rates.

Prostate cancer

Note

Investigations performed on human prostate cancer

cells PC3, revealed that tumor suppressive function of

MBP-1 is diminished. Reduction of endogenous MBP-

1 by employing specific siRNAs resulted in decreased

proliferation rates accompanied by inhibition of cyclin

A1 and cyclin B1 expression. Additionally, the cell size

increased after depletion of MBP-1. Introduction of

exogenous MBP-1 restored cyclins expression, leading

to dose-dependent increase in cyclin A1 and B1 levels.

Brain tumors

Note

Generally, the increased levels and activity of alpha

alpha-enolase correlate with brain tumorigenicity. In

Page 19: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

ENO1 (Enolase 1, (alpha)) Trojanowicz B, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 639

astrocytomas with different degrees of malignancy,

oligodendrogliomas, meningiomas and ependymomas,

alpha alpha-enolase was more abundant than in normal

brain tissues. Among astrocytic tumors, glioblastomas

revealed the highest proportion of alpha alpha-enolase

as compared with control tissues.

Introduction of full length, exogenous ENO1 sequence

into 1p-deleted or other neuroblastoma cell lines, led to

reduction of cell growth. This suggests that in this cell

lines ENO1 is preferentially translated as MBP-1 and

probably does not posses the enolase enzyme activity.

Multiple myeloma (plasma cell myeloma, kahler's disease)

Note

It was demonstrated, that interleukin 6 (IL-6) is

implicated in the in vivo proliferation of malignant

plasma cells in multiple myeloma. Studies in vitro

revealed that myeloma cell line U266 treated with IL-6,

responded with increased levels of MBP-1 and XBP-1

(X-box binding protein).

Acute lung inflammation (pneumonia)

Note

Increased ENO1 cell-surface expression on peripheral

blood monocytes (PBMs) and strong ENO1 production

in mononuclear cells in the alveolar space were

demonstrated for pneumonia patients when compared

with healthy volunteers. Elevated cell-surface

expression of ENO1 on PBMs and on human leukemic

monocyte lymphoma cell line U937, led to increased

plasmin generation, enhanced monocyte migration

through epithelial monolayers and promoted matrix

degradation.

Vasculitis

Note

In sera from patients with clinically proven vaculitis,

anti-neutrophil cytoplasmic antibodies (ANCA) reacted

with proteins present in the granules of human

neutrophils. 37.3 % of these sera contained the

antibodies raised against 48kDa protein, identified

further as cytoplasmic alpha-enolase. Antibodies

directed against enolase protein, recognised only alpha

isoform and were detected in sera giving ANCA

staining pattern.

Disease

Vasculitis (inflammatory destruction of blood vessels).

Nephritis

Note

In two independent studies antibodies raised against

alpha-enolase were detected in 10/41 and 9/33 sera of

patients with clinically proved SLE, respectively. 80%

of patients from the first report and 66.7% from the

second one, suffered from active nephritis.

Disease

Nephritis (renal disease) caused by systemic lupus

erythematosus (SLE, chronic autoimmune connective

tissue disease that can affect any part of the body).

Ulcerative colitis

Note

Alpha-enolase antibodies were found in about 10% of

ulcerative colitis patients.

Crohn's disease

Note

Alpha-enolase antibodies were found in about 18% of

patients with Crohn's disease.

Disease

Crohn's disease (autoimmune, inflammatory disease of

the intestines that may affect any part of the

gastrointestinal tract).

Primary biliary cirrhosis and autoimmune hepatitis

Note

Alpha-enolase antibodies were present in 28.6% of

patients with primary biliary cirrhosis and in 31.6%

with autoimmune hepatitis. Normal subjects revealed

significantly lower levels of alpha-enolase antibodies

when compared with both diseases. Note that

antibodies against beta and gamma enolases were not

found in any serum sample analysed.

References Oskam R, Rijksen G, Lips CJ, Staal GE. Enolase isozymes in differentiated and undifferentiated medullary thyroid carcinomas. Cancer. 1985 Jan 15;55(2):394-9

Wistow GJ, Lietman T, Williams LA, Stapel SO, de Jong WW, Horwitz J, Piatigorsky J. Tau-crystallin/alpha-enolase: one gene encodes both an enzyme and a lens structural protein. J Cell Biol. 1988 Dec;107(6 Pt 2):2729-36

Giallongo A, Oliva D, Calì L, Barba G, Barbieri G, Feo S. Structure of the human gene for alpha-enolase. Eur J Biochem. 1990 Jul 5;190(3):567-73

Moodie FD, Leaker B, Cambridge G, Totty NF, Segal AW. Alpha-enolase: a novel cytosolic autoantigen in ANCA positive vasculitis. Kidney Int. 1993 Mar;43(3):675-81

Aaronson RM, Graven KK, Tucci M, McDonald RJ, Farber HW. Non-neuronal enolase is an endothelial hypoxic stress protein. J Biol Chem. 1995 Nov 17;270(46):27752-7

Ray RB, Steele R, Seftor E, Hendrix M. Human breast carcinoma cells transfected with the gene encoding a c-myc promoter-binding protein (MBP-1) inhibits tumors in nude mice. Cancer Res. 1995 Sep 1;55(17):3747-51

Joseph J, Cruz-Sánchez FF, Carreras J. Enolase activity and isoenzyme distribution in human brain regions and tumors. J Neurochem. 1996 Jun;66(6):2484-90

Weith A, Brodeur GM, Bruns GA, Matise TC, Mischke D, Nizetic D, Seldin MF, van Roy N, Vance J. Report of the second international workshop on human chromosome 1 mapping 1995. Cytogenet Cell Genet. 1996;72(2-3):114-44

Page 20: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

ENO1 (Enolase 1, (alpha)) Trojanowicz B, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 640

Akisawa N, Maeda T, Iwasaki S, Onishi S. Identification of an autoantibody against alpha-enolase in primary biliary cirrhosis. J Hepatol. 1997 Apr;26(4):845-51

Merkulova T, Lucas M, Jabet C, Lamandé N, Rouzeau JD, Gros F, Lazar M, Keller A. Biochemical characterization of the mouse muscle-specific enolase: developmental changes in electrophoretic variants and selective binding to other proteins. Biochem J. 1997 May 1;323 ( Pt 3):791-800

Wen XY, Stewart AK, Sooknanan RR, Henderson G, Hawley TS, Reimold AM, Glimcher LH, Baumann H, Malek LT, Hawley RG. Identification of c-myc promoter-binding protein and X-box binding protein 1 as interleukin-6 target genes in human multiple myeloma cells. Int J Oncol. 1999 Jul;15(1):173-8

Feo S, Arcuri D, Piddini E, Passantino R, Giallongo A. ENO1 gene product binds to the c-myc promoter and acts as a transcriptional repressor: relationship with Myc promoter-binding protein 1 (MBP-1). FEBS Lett. 2000 May 4;473(1):47-52

Pratesi F, Moscato S, Sabbatini A, Chimenti D, Bombardieri S, Migliorini P. Autoantibodies specific for alpha-enolase in systemic autoimmune disorders. J Rheumatol. 2000 Jan;27(1):109-15

Rácz A, Brass N, Höfer M, Sybrecht GW, Remberger K, Meese EU. Gene amplification at chromosome 1pter-p33 including the genes PAX7 and ENO1 in squamous cell lung carcinoma. Int J Oncol. 2000 Jul;17(1):67-73

Subramanian A, Miller DM. Structural analysis of alpha-enolase. Mapping the functional domains involved in down-regulation of the c-myc protooncogene. J Biol Chem. 2000 Feb 25;275(8):5958-65

Fan X, Solomon H, Schwarz K, Kew MC, Ray RB, Di Bisceglie AM. Expression of c-myc promoter binding protein (MBP-1), a novel eukaryotic repressor gene, in cirrhosis and human hepatocellular carcinoma. Dig Dis Sci. 2001 Mar;46(3):563-6

Pancholi V. Multifunctional alpha-enolase: its role in diseases. Cell Mol Life Sci. 2001 Jun;58(7):902-20

Chang YS, Wu W, Walsh G, Hong WK, Mao L. Enolase-alpha is frequently down-regulated in non-small cell lung cancer and predicts aggressive biological behavior. Clin Cancer Res. 2003 Sep 1;9(10 Pt 1):3641-4

Baris O, Savagner F, Nasser V, Loriod B, Granjeaud S, Guyetant S, Franc B, Rodien P, Rohmer V, Bertucci F, Birnbaum D, Malthièry Y, Reynier P, Houlgatte R. Transcriptional profiling reveals coordinated up-regulation of oxidative metabolism genes in thyroid oncocytic tumors. J Clin Endocrinol Metab. 2004 Feb;89(2):994-1005

Dot C, Guigay J, Adamus G. Anti-alpha-enolase antibodies in cancer-associated retinopathy with small cell carcinoma of the lung. Am J Ophthalmol. 2005 Apr;139(4):746-7

Ejeskär K, Krona C, Carén H, Zaibak F, Li L, Martinsson T, Ioannou PA. Introduction of in vitro transcribed ENO1 mRNA into neuroblastoma cells induces cell death. BMC Cancer. 2005 Dec 16;5:161

Takashima M, Kuramitsu Y, Yokoyama Y, Iizuka N, Fujimoto M, Nishisaka T, Okita K, Oka M, Nakamura K. Overexpression of alpha enolase in hepatitis C virus-related hepatocellular carcinoma: association with tumor progression as determined by proteomic analysis. Proteomics. 2005 Apr;5(6):1686-92

Zhang D, Tai LK, Wong LL, Chiu LL, Sethi SK, Koay ES. Proteomic study reveals that proteins involved in metabolic and detoxification pathways are highly expressed in HER-2/neu-positive breast cancer. Mol Cell Proteomics. 2005 Nov;4(11):1686-96

Chang GC, Liu KJ, Hsieh CL, Hu TS, Charoenfuprasert S, Liu HK, Luh KT, Hsu LH, Wu CW, Ting CC, Chen CY, Chen KC, Yang TY, Chou TY, Wang WH, Whang-Peng J, Shih NY. Identification of alpha-enolase as an autoantigen in lung cancer: its overexpression is associated with clinical outcomes. Clin Cancer Res. 2006 Oct 1;12(19):5746-54

Ghosh AK, Steele R, Ray RB. Knockdown of MBP-1 in human prostate cancer cells delays cell cycle progression. J Biol Chem. 2006 Aug 18;281(33):23652-7

Ghosh AK, Steele R, Ryerse J, Ray RB. Tumor-suppressive effects of MBP-1 in non-small cell lung cancer cells. Cancer Res. 2006 Dec 15;66(24):11907-12

Huang LJ, Chen SX, Luo WJ, Jiang HH, Zhang PF, Yi H. Proteomic analysis of secreted proteins of non-small cell lung cancer. Ai Zheng. 2006 Nov;25(11):1361-7

Yoon SY, Kim JM, Oh JH, Jeon YJ, Lee DS, Kim JH, Choi JY, Ahn BM, Kim S, Yoo HS, Kim YS, Kim NS. Gene expression profiling of human HBV- and/or HCV-associated hepatocellular carcinoma cells using expressed sequence tags. Int J Oncol. 2006 Aug;29(2):315-27

Sedoris KC, Thomas SD, Miller DM. c-myc promoter binding protein regulates the cellular response to an altered glucose concentration. Biochemistry. 2007 Jul 24;46(29):8659-68

Hamaguchi T, Iizuka N, Tsunedomi R, Hamamoto Y, Miyamoto T, Iida M, Tokuhisa Y, Sakamoto K, Takashima M, Tamesa T, Oka M. Glycolysis module activated by hypoxia-inducible factor 1alpha is related to the aggressive phenotype of hepatocellular carcinoma. Int J Oncol. 2008 Oct;33(4):725-31

Trojanowicz B, Winkler A, Hammje K, Chen Z, Sekulla C, Glanz D, Schmutzler C, Mentrup B, Hombach-Klonisch S, Klonisch T, Finke R, Köhrle J, Dralle H, Hoang-Vu C. Retinoic acid-mediated down-regulation of ENO1/MBP-1 gene products caused decreased invasiveness of the follicular thyroid carcinoma cell lines. J Mol Endocrinol. 2009 Mar;42(3):249-60

Wygrecka M, Marsh LM, Morty RE, Henneke I, Guenther A, Lohmeyer J, Markart P, Preissner KT. Enolase-1 promotes plasminogen-mediated recruitment of monocytes to the acutely inflamed lung. Blood. 2009 May 28;113(22):5588-98

This article should be referenced as such:

Trojanowicz B, Hoang-Vu C, Sekulla C. ENO1 (Enolase 1, (alpha)). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):635-640.

Page 21: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 641

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

LIMK1 (LIM domain kinase 1) Ratna Chakrabarti

Department of Molecular biology and Microbiology, Burnett School of Biomedical Sciences, University of

Central Florida, 12722 Research Parkway, Orlando, Florida 32826, USA (RC)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/LIMK1ID41159ch7q11.html DOI: 10.4267/2042/44796

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: EC 2.7.11.1; LIMK; LIMK-1

HGNC (Hugo): LIMK1

Location: 7q11.23

Local order: ELN, LIMK1, EIF4H, LAT2.

LIMK1 gene is located at chromosome 7 on the long arm

(q11.23).

DNA/RNA

Description

The gene starts at 73136092 bp from pter and ends at

73174790 bp from pter. Its size is 38699 bases and its

orientation lie in the plus strand. The 5' promoter

region (1.5 kb) contains putative sites for

transcriptional regulation including Sp1, MZF1, AP1

and NF-E2. No consensus TATA box is evident.

Transcription

The transcript contains 16 exons spanning a length of

3.332 kb. The mRNA contains a short 5'UTR but a

long 3' end UTR. Two LIM domains, LIM1 and LIM2

are encoded by the exons 2-4. A single PDZ domain is

encoded by the exons 2-6. A C-terminal domain is

encoded by exons 8-16.

Pseudogene

None identified.

Protein

Description

The LIMK1 protein is composed of 647 amino acids. It

belongs to a unique family of LIM domain containing

dual specificity (serine threonine and tyrosine) protein

kinase. LIMK1 also has a PDZ domain in the middle of

the gene and a kinase domain at the C-terminal end. A

stretch of basic amino acids resembling nuclear

localisation signal is present in the kinase domain and

two nuclear export signal sequences containing

hydrophobic residues are present in the PDZ domain.

A) LIMK1 gene consists of 16 exons. Exons 2-4 encode two LIM domains in tandem. Exons 4-6 encode a PDZ domain and exons 8-16 encode a serine/threonine kinase domain. B) A splice variant of LIMK1 (dLIMK1), which lacks the kinase domain. In this variant, intron 7 was extended to 61 additional bases at the 5' end of exon 8, which caused a frameshift in the LIMK1 ORF and resulted in premature termination after 12 missense mutations in dLIMK1.

Page 22: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

LIMK1 (LIM domain kinase 1) Chakrabarti R

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 642

LIMK1 consists of specific domains, nuclear localisation signal (NLS) and nuclear exit signals (NES).

Expression

LIMK1 exhibits tissue specific expression. It is

predominantly expressed in brain but to a moderate

extent in the heart and skeletal muscle. The least

amount of LIMK1 expression was noted in the liver.

LIMK1 is also expressed in lesser amounts in various

human epithelial cell lines and haematopoetic cell lines.

Localisation

LIMK1 is primarily localized in the cytoplasm but also

transported to the nucleus. In the cytoplasm, LIMK1 is

colocalized with microtubules, and actin at the focal

adhesion, stress fiber and at the lamellipodia. In the

mitotic cells, LIMK1 is localized to the centrosomes

until early telophase and to the cleavage furrow during

late telophase.

Function

LIMK1 regulates organization of actin cytoskeleton

through inactivating phosphorylation of the actin

depolymerizing family (ADF) protein cofilin. LIMK1

phosphorylates cofilin at Serine, which inhibits actin

depolymerization and results in accumulation of F

actin. LIMK1 also regulates microtubule stability and

assembly through phosphorylation of p25/TPPP

(tubulin polymerization protein), which destabilizes

microtubules. Activated LIMK1 associates with

gamma-tubulin at the centrosome during mitotic

phases. LIMK1 is a multifunctional protein and is

involved in regulation of cell motility, cell cycle,

cytokinesis and cellular morphology. LIMK1 also

regulates neurite growth, synaptic stability, growth

cone motility, axon formation through modulation of

Golgi dynamics and neuronal differentiation.

Homology

LIMK1 has 50% identity overall and 70% identity in

the kinase domain with another family member

LIMK2. Although both proteins phosphorylate cofilin

and regulate actin cytosketon reorganization current

studies showed that LIMK2 has also different cellular

function.

Mutations

Germinal

Hemizygous deletion of LIMK1 along with Elastin

gene in a 1.5 MB deletion has been noted in patients

with Williams-Beuren syndrome. Patients with

Williams Syndrome exhibit impaired visuospatial

constructive cognition possibly because of loss of

LIMK1 gene.

SNP: A single nucleotide polymorphism of LIMK1 in a

haplotype spanning Elastin gene has been linked to

susceptibility of intracranial aneurysm (IA).

Implicated in

Prostate cancer

Disease

Prostate cancer is the most prevalent malignancy

second to lung cancer in men in the western world.

Although slow growing, a subpopulation of prostate

cancer patients develops highly invasive metastatic

disease that is nonresponsive to anti-androgen therapy

and is usually fatal.

Prognosis

The gold standard for diagnosis of prostate cancer are

the Gleason scores and the serum PSA level. PSA level

is also used for prognostic purposes. LIMK1 expression

may have prognostic value for identification of

metastatic progression as overexpression of LIMK1 has

been noted in metastatic prostate cancer cells.

Cytogenetics

Through cytogenetics method such as CGH and FISH

analysis chromosomal gain in 7q11.2 region or entire

chromosome 7 including 7q11.23 locus has been

reported in some prostate cancer cases.

Oncogenesis

LIMK1 is overexpressed in prostate cancer cells and

tissues compared to benign prostatic hyperplasia.

Because LIMK1 plays an important role in mitosis,

microtubule dynamics and cytokinesis altered

expession of LIMK1 may cause mitotic defects.

Aberrant expression of LIMK1 is also involved in

induction of invasion in prostate cancer cells.

Breast cancer

Disease

Breast cancer is one of the major cancers affecting

women in the western world after skin cancer and

second leading cause of cancer death in women. About

20% of breast cancers are familial and about 10% of

breast cancer is because of inheritence of a mutated

gene. Although the cure rate has been increased

because of the improved diagnostic approaches and

Page 23: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

LIMK1 (LIM domain kinase 1) Chakrabarti R

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 643

early detection, the metastatic disease actually has been

increased since 1990.

Prognosis

Overexpression of Her2/neu oncogene product is

considered to be associated with worse prognosis.

LIMK1 expression may have a prognostic value for

metastatic breast cancer as overexpression of LIMK1

has been noted in metastatic breast cancer cells.

Cytogenetics

CGH analysis indicated a gain in chromosome 7 in

majority of the infiltrating ductal carcinoma cases.

Some of the chromosomal gains include the region

encompassing Elastin and LIMK1 loci.

Oncogenesis

Overexpression of LIMK1 has been shown to increase

invasion and metastasis in animals. LIMK1 also

involved in regulation of EGFR turnover through

endocytic pathway in invasive breast cancer cells,

which may have implication in development of an

agressive disease.

Melanoma

Disease

Malignant melanoma is an agressive type of skin

cancer, which often metastasize leading to death. The

progression of melanoma is unpredictable and

sometimes show refractoriness to available

chemotherapy.

Cytogenetics

Chromosomal analysis using tiling array and CGH

showed a gain in chromosome 7 in melanoma cells.

Increased expression of LIMK1 in melanoma cells

(Skmel 28) harboring a break at 7q11.2 has also been

reported.

Williams-beuren syndrome (WBS)

Disease

WBS is a genetic disorder with autosomal dominant

inheritence. WBS is caused by microdeletion at

7q11.23 region with a phenotype of connective tissue

abnormalities, growth and psychomotor retardation,

muscular hypotonia, loss of visuospatial cognition and

behavioural abnormalities.

Prognosis

The presence of supravalvular aortic stenosis,

pulmonary stenosis, developmental retardation and

characteristic facial features in children between 18 to

30 months.

Cytogenetics

Chromosome analyses showed a deletion at the LIMK1

locus at 7q11.23 caused by a distal recombination event

at the common telomeric breakpoint.

Alzheimer disease (AD)

Disease

Dystrophic neurites are found to be associated with

Alzheimer's pathology. Altered structures of axons and

dendrites, deposition of amyloid plaques leading to

neurofibrillary tangle formation in AD pathology are

responsible for dementia and cognitive disorder in

Alzheimer's patients.

Prognosis

Deposition of fibrillar amyloid beta in the brain is one

of the events towards developing Alzheimer Disease.

LIMK1 has been shown to be involved in amyloid

beta-induced neuronal degeneration.

Immunofluorescence analysis showed an increased

number of phosphorylated LIMK1 positive neurons in

the areas of brain with AD pathology. Inhibition of

cofilin phosphorylation prevented neuronal

degeneration, which supports the involvement of

LIMK1 in AD.

Intracranial Aneurysm

Disease

Intracranial aneurysm is the localized dilation of the

blood vessel which could be fatal upon rupture causing

hemorrhage in the subarachnoid space. It occurs more

frequently in adults than children and in women than

men. Risk factors include family history of aneurysm

and inherited disorders including polycystic kidney

disease.

Cytogenetics

Genome wide linkage studies indicated a significant

association between SNP in LIMK1 promoter sequence

at 7q11.2 locus and incidence of IA in Japanese and

Korean patients. The SNP in the promoter sequence of

LIMK1 [C(-187)T] introduced an additional

transcription factor (AP2) binding site, which leads to a

reduced transcription of LIMK1 mRNA.

References Mizuno K, Okano I, Ohashi K, Nunoue K, Kuma K, Miyata T, Nakamura T. Identification of a human cDNA encoding a novel protein kinase with two repeats of the LIM/double zinc finger motif. Oncogene. 1994 Jun;9(6):1605-12

Tassabehji M, Metcalfe K, Fergusson WD, Carette MJ, Dore JK, Donnai D, Read AP, Pröschel C, Gutowski NJ, Mao X, Sheer D. LIM-kinase deleted in Williams syndrome. Nat Genet. 1996 Jul;13(3):272-3

Higuchi O, Amano T, Yang N, Mizuno K. Inhibition of activated Ras-induced neuronal differentiation of PC12 cells by the LIM domain of LIM-kinase 1. Oncogene. 1997 Apr 17;14(15):1819-25

Jenkins RB, Qian J, Lee HK, Huang H, Hirasawa K, Bostwick DG, Proffitt J, Wilber K, Lieber MM, Liu W, Smith DI. A molecular cytogenetic analysis of 7q31 in prostate cancer. Cancer Res. 1998 Feb 15;58(4):759-66

Edwards DC, Gill GN. Structural features of LIM kinase that control effects on the actin cytoskeleton. J Biol Chem. 1999 Apr 16;274(16):11352-61

Alers JC, Rochat J, Krijtenburg PJ, Hop WC, Kranse R, Rosenberg C, Tanke HJ, Schröder FH, van Dekken H. Identification of genetic markers for prostatic cancer progression. Lab Invest. 2000 Jun;80(6):931-42

Page 24: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

LIMK1 (LIM domain kinase 1) Chakrabarti R

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 644

Davila M, Frost AR, Grizzle WE, Chakrabarti R. LIM kinase 1 is essential for the invasive growth of prostate epithelial cells: implications in prostate cancer. J Biol Chem. 2003 Sep 19;278(38):36868-75

Endo M, Ohashi K, Sasaki Y, Goshima Y, Niwa R, Uemura T, Mizuno K. Control of growth cone motility and morphology by LIM kinase and Slingshot via phosphorylation and dephosphorylation of cofilin. J Neurosci. 2003 Apr 1;23(7):2527-37

Yoshioka K, Foletta V, Bernard O, Itoh K. A role for LIM kinase in cancer invasion. Proc Natl Acad Sci U S A. 2003 Jun 10;100(12):7247-52

Rosso S, Bollati F, Bisbal M, Peretti D, Sumi T, Nakamura T, Quiroga S, Ferreira A, Cáceres A. LIMK1 regulates Golgi dynamics, traffic of Golgi-derived vesicles, and process extension in primary cultured neurons. Mol Biol Cell. 2004 Jul;15(7):3433-49

Eaton BA, Davis GW. LIM Kinase1 controls synaptic stability downstream of the type II BMP receptor. Neuron. 2005 Sep 1;47(5):695-708

Gorovoy M, Niu J, Bernard O, Profirovic J, Minshall R, Neamu R, Voyno-Yasenetskaya T. LIM kinase 1 coordinates microtubule stability and actin polymerization in human endothelial cells. J Biol Chem. 2005 Jul 15;280(28):26533-42

Okamoto I, Pirker C, Bilban M, Berger W, Losert D, Marosi C, Haas OA, Wolff K, Pehamberger H. Seven novel and stable translocations associated with oncogenic gene expression in malignant melanoma. Neoplasia. 2005 Apr;7(4):303-11

Akagawa H, Tajima A, Sakamoto Y, Krischek B, Yoneyama T, Kasuya H, Onda H, Hori T, Kubota M, Machida T, Saeki N, Hata A, Hashiguchi K, Kimura E, Kim CJ, Yang TK, Lee JY,

Kimm K, Inoue I. A haplotype spanning two genes, ELN and LIMK1, decreases their transcripts and confers susceptibility to intracranial aneurysms. Hum Mol Genet. 2006 May 15;15(10):1722-34

Bagheri-Yarmand R, Mazumdar A, Sahin AA, Kumar R. LIM kinase 1 increases tumor metastasis of human breast cancer cells via regulation of the urokinase-type plasminogen activator system. Int J Cancer. 2006 Jun 1;118(11):2703-10

Heredia L, Helguera P, de Olmos S, Kedikian G, Solá Vigo F, LaFerla F, Staufenbiel M, de Olmos J, Busciglio J, Cáceres A, Lorenzo A. Phosphorylation of actin-depolymerizing factor/cofilin by LIM-kinase mediates amyloid beta-induced degeneration: a potential mechanism of neuronal dystrophy in Alzheimer's disease. J Neurosci. 2006 Jun 14;26(24):6533-42

Nishimura Y, Yoshioka K, Bernard O, Bereczky B, Itoh K. A role of LIM kinase 1/cofilin pathway in regulating endocytic trafficking of EGF receptor in human breast cancer cells. Histochem Cell Biol. 2006 Nov;126(5):627-38

Chakrabarti R, Jones JL, Oelschlager DK, Tapia T, Tousson A, Grizzle WE. Phosphorylated LIM kinases colocalize with gamma-tubulin in centrosomes during early stages of mitosis. Cell Cycle. 2007 Dec 1;6(23):2944-52

Jönsson G, Dahl C, Staaf J, Sandberg T, Bendahl PO, Ringnér M, Guldberg P, Borg A. Genomic profiling of malignant melanoma using tiling-resolution arrayCGH. Oncogene. 2007 Jul 12;26(32):4738-48

This article should be referenced as such:

Chakrabarti R. LIMK1 (LIM domain kinase 1). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):641-644.

Page 25: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 645

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

PAX6 (paired box 6) Yi-Hong Zhou

Department of Neurological Surgery, Department of Biological Chemistry (joint), University of California,

Irvine, Med Sci I, Room C214, Irvine, CA 92697, USA (YHZ)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/PAX6ID211ch11p13.html DOI: 10.4267/2042/44797

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: AN; AN2; D11S812E; MGC17209;

MGDA; Oculorhombin; WAGR

HGNC (Hugo): PAX6

Location: 11p13

DNA/RNA

Description

The PAX6 coding region extends over a genomic

interval of 16-17 kb and comprise 10 (isoform a) and

11 exons (isoform b).

Transcription

Three transcripts have been identified, originating from

alternative promoter usage (variant 3) or alternative

splicing (variant 2, additional in-frame coding 42 bp

exon downstream of exon 5 of variant 1); transcription

is from centromere to telomere.

Protein

Description

PAX6 belongs to the paired box family of transcription

factors, contains two DNA binding domains, a paired

box (PD) and a paired-type homeodomain (HD), and a

carboxyl-terminal transactivation domain rich of

proline, serine, and threonine (PST).

Expression

PAX6, predominately in form of PAX6a, is expressed

in the developing sensory organs (including eye, nasal

and olfactory tissues), central nervous system

(including forebrain, hindbrain, and spinal cord), and

endocrine system (including anterior pituitary gland

and pancreas) in human and rodent (Walther and

Gruss., 1991; Stoykova and Gruss., 1994; Davis and

Reed., 1996; Terzic and Saraga-Babic., 1999; Pinson et

al., 2005). PAX6 expression is sustained into adulthood

in certain areas of the brain, including, hippocampal

dentate gyrus (Maekawa et al., 2005; Nacher et al.,

2005), ependymal layer and the subventricular zone of

the lateral ventricle (Hack et al., 2005; Kohwi et al.,

2005), radial glia-like cells (Gubert et al., 2009), and in

mature endocrine cells in pancreas (St-Onge et al.,

1997). PAX6 transcription is regulated by two

promoters, P0 and P1, which are remarkably conserved

in evolution in both of their nucleotide sequence

arrangement and functional control of special and

temporal expression of PAX6 in development (Xu and

Saunders, 1997; Okladnova et al., 1998a; Williams et

al., 1998; Xu and

Page 26: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

PAX6 (paired box 6) Zhou YH

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 646

There are two isoforms of PAX6, PAX6a and PAX6b with additional 14 extra amino acids in the paired box DNA binding domain. PAX6a, 423 amino acids, ~47 kDa; PAX6b, 436 amino acids, ~49 kDa.

Saunders, 1998; Kammandel et al., 1999; Plaza et al.,

1999a; Xu et al., 1999; Tyas et al., 2006), involving

multiple transcription factors, such as POU factor Brn-

3B, TFCP2, SP1, the basic helix-loop-helix

transcription factor NeuroD/BETA2, CCCTC binding

factor CTCF, PPARgamma (Plaza et al., 1999b; Zheng

et al., 2001; Schinner et al., 2002; Marsich et al., 2003;

Li et al., 2006; Wu et al., 2006). PAX6 expression is

also regulated by a long range downstream enhancer

(Kleinjan et al., 2006) and is under autoregulation

(Grocott et al., 2007) and post modification by HIPK2

and protein phosphatase 1 (Kim et al., 2006; Yan et al.,

2007). A promoter-associated polymorphic repeat was

found to modulate PAX6 expression in human brain

(Okladnova et al., 1998b).

Localisation

Nuclear.

Function

Loss of Pax6 function in rodent mutant and knock-out

model revealed that Pax6 is a key regulator of a

multitude of developmental processes of sensory

system, including eye, nasal and olfactory (Hill et al.,

1991; Grindley et al., 1995; Quinn et al., 1996; van

Raamsdonk and Tilghman, 2000; Singh et al., 2002;

van Heyningen and Williamson, 2002; Collinson et al.,

2003; Davis et al., 2003; Brill et al., 2008), CNS

(Matsuo et al., 1993; Schmahl et al., 1993; Stoykova et

al., 1996; Grindley et al., 1997; Osumi et al., 1997;

Mastick et al., 1997; Warren and Price, 1997; Gotz et

al., 1998; Sun et al., 1998; Engelkamp et al., 1999;

Kawano et al., 1999; Pratt et al., 2000; Stoykova et al.,

2000; Estivill-Torrus et al., 2002; Pratt et al., 2002;

Talamillo et al., 2003; Quinn et al., 2007), pituitary

(Bentley et al., 1999; Kioussi et al., 1999) and pancreas

(Sander et al., 1997; St-Onge et al., 1997; Dohrmann et

al., 2000; Zhang et al., 2003). Pax6 function in

development of fundamental sensory processes and

central nervous system, particularly of the

photoreceptive organ, are remarkably conserved in

evolution (Halder et al., 1995; Gehring et al., 2005).

PAX6 funciton in development were found to be under

control of Shh, notch and EGFR signaling (Ericson et

al., 1997; Kumar and Moses, 2001; Onuma et al., 2002;

Li and Lu, 2005), essential for neural stem cell

proliferation, multipotency, and neurogenesis in many

regions of the central nervous system (Warren et al.,

1999; Bishop et al., 2000; Toresson et al., 2000;

Marquardt et al., 2001; Yamasaki et al., 2001; Yun et

al., 2001; Estivill-Torrus et al., 2002; Heins et al.,

2002; Simpson and Price, 2002; Tyas et al., 2003;

Collinson et al., 2004; Haubst et al., 2004; Nomura and

Osumi, 2004; Schuurmans et al., 2004; Maekawa et al.,

2005; Bel-Vialar et al., 2007; Duparc et al., 2007;

Quinn et al., 2007; Canto-Soler et al., 2008; Oron-

Karni et al., 2008; Osumi et al., 2008), and appears to

control the balance between neural stem cell self-

renewal and neurogenesis under a dose-dependent

manner (Sansom et al., 2009).

PAX6 binds as a monomer to relatively long (15-22 bp)

DNA binding sites, and the 14 aa insertion in the paired

domain allows different binding affinity to DNA

sequences between PAX6a and PAX6b (Epstein et al.,

1994a; Epstein et al., 1994b). Through binding to

different DNA sequences via usage of various DNA

binding motifs alone or in combination, PAX6 controls

the expression of various downstream target genes

involved in complex gene regulatory networks for cell

proliferation, adhesion, migration, and neurogenesis

(Schmahl et al., 1993; Caric et al., 1997; Sander et al.,

1997; Sax et al., 1997; Tang et al., 1997; Duncan et al.,

1998; Beimesche et al., 1999; Meech et al., 1999;

Singh et al., 2000; Sivak et al., 2000; Zhou et al., 2000;

Chauhan et al., 2002; Mishra et al., 2002; Skala-

Rubinson et al., 2002; Zhou et al., 2002; Andrews and

Mastick, 2003; Davis et al., 2003; Horie et al., 2003;

Tyas et al., 2003; Cvekl et al., 2004; Grinchuk et al.,

2005; Mayes et al., 2006; Holm et al., 2007; Tuoc and

Stoykova, 2008). Not only reduced, but also increases

level of PAX6 gene dosage also cause defects in

developmental processes that are sensitive to PAX6

dosage, including eye organogenesis and corticogenesis

(Schedl et al., 1996; Berger et al., 2007; Manuel et al.,

2007).

Homology

PAX6 shares homology through the conserved paired

box domain with the other members of the nine PAX

gene family.

Mutations

Germinal

Heterozygous intragenic mutation of PAX6, that causes

loss of function of one copy of the PAX6 gene, is the

cause of aniridia syndrome (Ton et al., 1991; Glaser et

al., 1992; Prosser and van Heyningen, 1998; Robinson

et al., 2008; Hingorani et al., 2009; MRC Human

Genetics Unit) and cerebral malformation, olfactory

dysfunction, absence of the pineal gland and unilateral

polymicrogyria (Sisodiya et al., 2001; Free et al., 2003;

Mitchell et al., 2003; Bamiou et al., 2007a; Bamiou et

al., 2007b).

Page 27: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

PAX6 (paired box 6) Zhou YH

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 647

PAX6 3' deletion also results in aniridia, autism and

mental retardation (Davis et al., 2008).

Implicated in

Brain cancer

Note

The expression level of PAX6 in human glioma cell

lines was shown to be negatively associated with the

degree of tumorigenicity. PAX6 expression level is

lower in glioblastoma compared to the adjacent normal

tissue and to the anaplastic astrocytoma previously

formed in the same patient (Zhou et al., 2003). Ectopic

expression of PAX6 in glioma cell lines suppressed cell

anchorage independent growth, ability to survive under

oxidative stress induced by cell detachment, ability to

invade partially by suppression of MMP2 gene

expression, ability to induce angiogenesis by initiating

a new signaling pathway independent of PI3K/Akt-

HIF1A signaling to suppress VEGFA, and overall

tumor growth after intracranial implantation in

immunocompromised mouse brain (Zhou et al., 2005;

Mayes et al., 2006; Chang et al., 2007; Zhou et al.,

2009). Mutation analysis for PAX6 in gliomas failed to

identify PAX6 mutation in its coding and regulating

regions, suggesting involvement of epigenetic

mechanisms in the silencing of PAX6 in glioma (Pinto

et al., 2007). PAX6 expression is activated in glioma

cell line with re-introduction of a normal ch.10,

suggesting that PAX6 is regulated by a gene(s) on

ch.10 (Zhou et al., 2005).

Prognosis

PAX6 is a factor related to a longer survival prognosis

for astrocytic gliomas (Zhou et al., 2003).

Pancreatic cancer

Note

PAX6 is expressed in pancreatic adenocarcinoma and

is downregulated during induction of terminal

differentiation (Lang et al., 2008). In pancreatic

carcinoma cell lines, PAX6 bind directly to an

enhancer element in the MET promoter and activate the

expression of the MET gene (Mascarenhas et al.,

2009).

Bladder cancer

Note

Methylation of PAX6-promoters is increased in early

bladder cancer and in normal mucosa adjacent to pTa

tumours (Hellwinkel et al., 2008).

Familial adenomatous polyposis (FAP) related carcinoma

Note

PAX6 gene is methylated in FAP-related carcinoma.

Patients with familial adenomatous polyposis (FAP)

have a high risk of developing duodenal carcinomas

(Berkhout et al., 2007).

WAGR syndrome

Note

WAGR syndrome can have aniridia due to deletion of

chromosome 11 including PAX6 (Gronskov et al.,

2001; Chao et al., 2003). However, PAX6 mutation is

only found in aniridia patient, not WAGR syndrome

associated anomalies (Robinson et al., 2008).

References Hill RE, Favor J, Hogan BL, Ton CC, Saunders GF, Hanson IM, Prosser J, Jordan T, Hastie ND, van Heyningen V. Mouse small eye results from mutations in a paired-like homeobox-containing gene. Nature. 1991 Dec 19-26;354(6354):522-5

Ton CC, Hirvonen H, Miwa H, Weil MM, Monaghan P, Jordan T, van Heyningen V, Hastie ND, Meijers-Heijboer H, Drechsler M. Positional cloning and characterization of a paired box- and homeobox-containing gene from the aniridia region. Cell. 1991 Dec 20;67(6):1059-74

Walther C, Gruss P. Pax-6, a murine paired box gene, is expressed in the developing CNS. Development. 1991 Dec;113(4):1435-49

Glaser T, Walton DS, Maas RL. Genomic structure, evolutionary conservation and aniridia mutations in the human PAX6 gene. Nat Genet. 1992 Nov;2(3):232-9

Matsuo T, Osumi-Yamashita N, Noji S, Ohuchi H, Koyama E, Myokai F, Matsuo N, Taniguchi S, Doi H, Iseki S. A mutation in the Pax-6 gene in rat small eye is associated with impaired migration of midbrain crest cells. Nat Genet. 1993 Apr;3(4):299-304

Schmahl W, Knoedlseder M, Favor J, Davidson D. Defects of neuronal migration and the pathogenesis of cortical malformations are associated with Small eye (Sey) in the mouse, a point mutation at the Pax-6-locus. Acta Neuropathol. 1993;86(2):126-35

Epstein J, Cai J, Glaser T, Jepeal L, Maas R. Identification of a Pax paired domain recognition sequence and evidence for DNA-dependent conformational changes. J Biol Chem. 1994 Mar 18;269(11):8355-61

Epstein JA, Glaser T, Cai J, Jepeal L, Walton DS, Maas RL. Two independent and interactive DNA-binding subdomains of the Pax6 paired domain are regulated by alternative splicing. Genes Dev. 1994 Sep 1;8(17):2022-34

Stoykova A, Gruss P. Roles of Pax-genes in developing and adult brain as suggested by expression patterns. J Neurosci. 1994 Mar;14(3 Pt 2):1395-412

Grindley JC, Davidson DR, Hill RE. The role of Pax-6 in eye and nasal development. Development. 1995 May;121(5):1433-42

Halder G, Callaerts P, Gehring WJ. New perspectives on eye evolution. Curr Opin Genet Dev. 1995 Oct;5(5):602-9

Davis JA, Reed RR. Role of Olf-1 and Pax-6 transcription factors in neurodevelopment. J Neurosci. 1996 Aug 15;16(16):5082-94

Quinn JC, West JD, Hill RE. Multiple functions for Pax6 in mouse eye and nasal development. Genes Dev. 1996 Feb 15;10(4):435-46

Schedl A, Ross A, Lee M, Engelkamp D, Rashbass P, van Heyningen V, Hastie ND. Influence of PAX6 gene dosage on development: overexpression causes severe eye abnormalities. Cell. 1996 Jul 12;86(1):71-82

Page 28: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

PAX6 (paired box 6) Zhou YH

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 648

Stoykova A, Fritsch R, Walther C, Gruss P. Forebrain patterning defects in Small eye mutant mice. Development. 1996 Nov;122(11):3453-65

Carić D, Gooday D, Hill RE, McConnell SK, Price DJ. Determination of the migratory capacity of embryonic cortical cells lacking the transcription factor Pax-6. Development. 1997 Dec;124(24):5087-96

Ericson J, Rashbass P, Schedl A, Brenner-Morton S, Kawakami A, van Heyningen V, Jessell TM, Briscoe J. Pax6 controls progenitor cell identity and neuronal fate in response to graded Shh signaling. Cell. 1997 Jul 11;90(1):169-80

Grindley JC, Hargett LK, Hill RE, Ross A, Hogan BL. Disruption of PAX6 function in mice homozygous for the Pax6Sey-1Neu mutation produces abnormalities in the early development and regionalization of the diencephalon. Mech Dev. 1997 Jun;64(1-2):111-26

Mastick GS, Davis NM, Andrew GL, Easter SS Jr. Pax-6 functions in boundary formation and axon guidance in the embryonic mouse forebrain. Development. 1997 May;124(10):1985-97

Osumi N, Hirota A, Ohuchi H, Nakafuku M, Iimura T, Kuratani S, Fujiwara M, Noji S, Eto K. Pax-6 is involved in the specification of hindbrain motor neuron subtype. Development. 1997 Aug;124(15):2961-72

Sander M, Neubüser A, Kalamaras J, Ee HC, Martin GR, German MS. Genetic analysis reveals that PAX6 is required for normal transcription of pancreatic hormone genes and islet development. Genes Dev. 1997 Jul 1;11(13):1662-73

Sax CM, Cvekl A, Piatigorsky J. Transcriptional regulation of the mouse alpha A-crystallin gene: binding of USF to the -7/+5 region. Gene. 1997 Feb 7;185(2):209-16

St-Onge L, Sosa-Pineda B, Chowdhury K, Mansouri A, Gruss P. Pax6 is required for differentiation of glucagon-producing alpha-cells in mouse pancreas. Nature. 1997 May 22;387(6631):406-9

Tang HK, Chao LY, Saunders GF. Functional analysis of paired box missense mutations in the PAX6 gene. Hum Mol Genet. 1997 Mar;6(3):381-6

Warren N, Price DJ. Roles of Pax-6 in murine diencephalic development. Development. 1997 Apr;124(8):1573-82

Xu ZP, Saunders GF. Transcriptional regulation of the human PAX6 gene promoter. J Biol Chem. 1997 Feb 7;272(6):3430-6

Duncan MK, Haynes JI 2nd, Cvekl A, Piatigorsky J. Dual roles for Pax-6: a transcriptional repressor of lens fiber cell-specific beta-crystallin genes. Mol Cell Biol. 1998 Sep;18(9):5579-86

Götz M, Stoykova A, Gruss P. Pax6 controls radial glia differentiation in the cerebral cortex. Neuron. 1998 Nov;21(5):1031-44

Okladnova O, Syagailo YV, Mössner R, Riederer P, Lesch KP. Regulation of PAX-6 gene transcription: alternate promoter usage in human brain. Brain Res Mol Brain Res. 1998 Oct 1;60(2):177-92

Okladnova O, Syagailo YV, Tranitz M, Stöber G, Riederer P, Mössner R, Lesch KP. A promoter-associated polymorphic repeat modulates PAX-6 expression in human brain. Biochem Biophys Res Commun. 1998 Jul 20;248(2):402-5

Prosser J, van Heyningen V. PAX6 mutations reviewed. Hum Mutat. 1998;11(2):93-108

Sun T, Pringle NP, Hardy AP, Richardson WD, Smith HK. Pax6 influences the time and site of origin of glial precursors in the ventral neural tube. Mol Cell Neurosci. 1998 Nov;12(4-5):228-39

Williams SC, Altmann CR, Chow RL, Hemmati-Brivanlou A, Lang RA. A highly conserved lens transcriptional control element from the Pax-6 gene. Mech Dev. 1998 May;73(2):225-9

Xu ZP, Saunders GF. PAX6 intronic sequence targets expression to the spinal cord. Dev Genet. 1998;23(4):259-63

Beimesche S, Neubauer A, Herzig S, Grzeskowiak R, Diedrich T, Cierny I, Scholz D, Alejel T, Knepel W. Tissue-specific transcriptional activity of a pancreatic islet cell-specific enhancer sequence/Pax6-binding site determined in normal adult tissues in vivo using transgenic mice. Mol Endocrinol. 1999 May;13(5):718-28

Bentley CA, Zidehsarai MP, Grindley JC, Parlow AF, Barth-Hall S, Roberts VJ. Pax6 is implicated in murine pituitary endocrine function. Endocrine. 1999 Apr;10(2):171-7

Engelkamp D, Rashbass P, Seawright A, van Heyningen V. Role of Pax6 in development of the cerebellar system. Development. 1999 Aug;126(16):3585-96

Kammandel B, Chowdhury K, Stoykova A, Aparicio S, Brenner S, Gruss P. Distinct cis-essential modules direct the time-space pattern of the Pax6 gene activity. Dev Biol. 1999 Jan 1;205(1):79-97

Kawano H, Fukuda T, Kubo K, Horie M, Uyemura K, Takeuchi K, Osumi N, Eto K, Kawamura K. Pax-6 is required for thalamocortical pathway formation in fetal rats. J Comp Neurol. 1999 May 31;408(2):147-60

Kioussi C, O'Connell S, St-Onge L, Treier M, Gleiberman AS, Gruss P, Rosenfeld MG. Pax6 is essential for establishing ventral-dorsal cell boundaries in pituitary gland development. Proc Natl Acad Sci U S A. 1999 Dec 7;96(25):14378-82

Meech R, Kallunki P, Edelman GM, Jones FS. A binding site for homeodomain and Pax proteins is necessary for L1 cell adhesion molecule gene expression by Pax-6 and bone morphogenetic proteins. Proc Natl Acad Sci U S A. 1999 Mar 2;96(5):2420-5

Plaza S, Hennemann H, Möröy T, Saule S, Dozier C. Evidence that POU factor Brn-3B regulates expression of Pax-6 in neuroretina cells. J Neurobiol. 1999 Nov 15;41(3):349-58

Plaza S, Saule S, Dozier C. High conservation of cis-regulatory elements between quail and human for the Pax-6 gene. Dev Genes Evol. 1999 Mar;209(3):165-73

Terzić J, Saraga-Babić M. Expression pattern of PAX3 and PAX6 genes during human embryogenesis. Int J Dev Biol. 1999 Sep;43(6):501-8

Warren N, Caric D, Pratt T, Clausen JA, Asavaritikrai P, Mason JO, Hill RE, Price DJ. The transcription factor, Pax6, is required for cell proliferation and differentiation in the developing cerebral cortex. Cereb Cortex. 1999 Sep;9(6):627-35

Xu PX, Zhang X, Heaney S, Yoon A, Michelson AM, Maas RL. Regulation of Pax6 expression is conserved between mice and flies. Development. 1999 Jan;126(2):383-95

Bishop KM, Goudreau G, O'Leary DD. Regulation of area identity in the mammalian neocortex by Emx2 and Pax6. Science. 2000 Apr 14;288(5464):344-9

Dohrmann C, Gruss P, Lemaire L. Pax genes and the differentiation of hormone-producing endocrine cells in the pancreas. Mech Dev. 2000 Mar 15;92(1):47-54

Pratt T, Vitalis T, Warren N, Edgar JM, Mason JO, Price DJ. A role for Pax6 in the normal development of dorsal thalamus and its cortical connections. Development. 2000 Dec;127(23):5167-78

Page 29: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

PAX6 (paired box 6) Zhou YH

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 649

Singh S, Stellrecht CM, Tang HK, Saunders GF. Modulation of PAX6 homeodomain function by the paired domain. J Biol Chem. 2000 Jun 9;275(23):17306-13

Sivak JM, Mohan R, Rinehart WB, Xu PX, Maas RL, Fini ME. Pax-6 expression and activity are induced in the reepithelializing cornea and control activity of the transcriptional promoter for matrix metalloproteinase gelatinase B. Dev Biol. 2000 Jun 1;222(1):41-54

Stoykova A, Treichel D, Hallonet M, Gruss P. Pax6 modulates the dorsoventral patterning of the mammalian telencephalon. J Neurosci. 2000 Nov 1;20(21):8042-50

Toresson H, Potter SS, Campbell K. Genetic control of dorsal-ventral identity in the telencephalon: opposing roles for Pax6 and Gsh2. Development. 2000 Oct;127(20):4361-71

van Raamsdonk CD, Tilghman SM. Dosage requirement and allelic expression of PAX6 during lens placode formation. Development. 2000 Dec;127(24):5439-48

Zhou Y, Zheng JB, Gu X, Li W, Saunders GF. A novel Pax-6 binding site in rodent B1 repetitive elements: coevolution between developmental regulation and repeated elements? Gene. 2000 Mar 21;245(2):319-28

Grønskov K, Olsen JH, Sand A, Pedersen W, Carlsen N, Bak Jylling AM, Lyngbye T, Brøndum-Nielsen K, Rosenberg T. Population-based risk estimates of Wilms tumor in sporadic aniridia. A comprehensive mutation screening procedure of PAX6 identifies 80% of mutations in aniridia. Hum Genet. 2001 Jul;109(1):11-8

Kumar JP, Moses K. EGF receptor and Notch signaling act upstream of Eyeless/Pax6 to control eye specification. Cell. 2001 Mar 9;104(5):687-97

Marquardt T, Ashery-Padan R, Andrejewski N, Scardigli R, Guillemot F, Gruss P. Pax6 is required for the multipotent state of retinal progenitor cells. Cell. 2001 Apr 6;105(1):43-55

Sisodiya SM, Free SL, Williamson KA, Mitchell TN, Willis C, Stevens JM, Kendall BE, Shorvon SD, Hanson IM, Moore AT, van Heyningen V. PAX6 haploinsufficiency causes cerebral malformation and olfactory dysfunction in humans. Nat Genet. 2001 Jul;28(3):214-6

Yamasaki T, Kawaji K, Ono K, Bito H, Hirano T, Osumi N, Kengaku M. Pax6 regulates granule cell polarization during parallel fiber formation in the developing cerebellum. Development. 2001 Aug;128(16):3133-44

Yun K, Potter S, Rubenstein JL. Gsh2 and Pax6 play complementary roles in dorsoventral patterning of the mammalian telencephalon. Development. 2001 Jan;128(2):193-205

Zheng JB, Zhou YH, Maity T, Liao WS, Saunders GF. Activation of the human PAX6 gene through the exon 1 enhancer by transcription factors SEF and Sp1. Nucleic Acids Res. 2001 Oct 1;29(19):4070-8

Chauhan BK, Reed NA, Yang Y, Cermák L, Reneker L, Duncan MK, Cvekl A. A comparative cDNA microarray analysis reveals a spectrum of genes regulated by Pax6 in mouse lens. Genes Cells. 2002 Dec;7(12):1267-83

Estivill-Torrus G, Pearson H, van Heyningen V, Price DJ, Rashbass P. Pax6 is required to regulate the cell cycle and the rate of progression from symmetrical to asymmetrical division in mammalian cortical progenitors. Development. 2002 Jan;129(2):455-66

Heins N, Malatesta P, Cecconi F, Nakafuku M, Tucker KL, Hack MA, Chapouton P, Barde YA, Götz M. Glial cells generate neurons: the role of the transcription factor Pax6. Nat Neurosci. 2002 Apr;5(4):308-15

Mishra R, Gorlov IP, Chao LY, Singh S, Saunders GF. PAX6, paired domain influences sequence recognition by the homeodomain. J Biol Chem. 2002 Dec 20;277(51):49488-94

Onuma Y, Takahashi S, Asashima M, Kurata S, Gehring WJ. Conservation of Pax 6 function and upstream activation by Notch signaling in eye development of frogs and flies. Proc Natl Acad Sci U S A. 2002 Feb 19;99(4):2020-5

Pratt T, Quinn JC, Simpson TI, West JD, Mason JO, Price DJ. Disruption of early events in thalamocortical tract formation in mice lacking the transcription factors Pax6 or Foxg1. J Neurosci. 2002 Oct 1;22(19):8523-31

Schinner S, Dellas C, Schroder M, Heinlein CA, Chang C, Fischer J, Knepel W. Repression of glucagon gene transcription by peroxisome proliferator-activated receptor gamma through inhibition of Pax6 transcriptional activity. J Biol Chem. 2002 Jan 18;277(3):1941-8

Simpson TI, Price DJ. Pax6; a pleiotropic player in development. Bioessays. 2002 Nov;24(11):1041-51

Singh S, Mishra R, Arango NA, Deng JM, Behringer RR, Saunders GF. Iris hypoplasia in mice that lack the alternatively spliced Pax6(5a) isoform. Proc Natl Acad Sci U S A. 2002 May 14;99(10):6812-5

Skala-Rubinson H, Vinh J, Labas V, Kahn A, Phan DT. Novel target sequences for Pax-6 in the brain-specific activating regions of the rat aldolase C gene. J Biol Chem. 2002 Dec 6;277(49):47190-6

van Heyningen V, Williamson KA. PAX6 in sensory development. Hum Mol Genet. 2002 May 15;11(10):1161-7

Zhou YH, Zheng JB, Gu X, Saunders GF, Yung WK. Novel PAX6 binding sites in the human genome and the role of repetitive elements in the evolution of gene regulation. Genome Res. 2002 Nov;12(11):1716-22

Andrews GL, Mastick GS. R-cadherin is a Pax6-regulated, growth-promoting cue for pioneer axons. J Neurosci. 2003 Oct 29;23(30):9873-80

Chao LY, Mishra R, Strong LC, Saunders GF. Missense mutations in the DNA-binding region and termination codon in PAX6. Hum Mutat. 2003 Feb;21(2):138-45

Collinson JM, Quinn JC, Hill RE, West JD. The roles of Pax6 in the cornea, retina, and olfactory epithelium of the developing mouse embryo. Dev Biol. 2003 Mar 15;255(2):303-12

Davis J, Duncan MK, Robison WG Jr, Piatigorsky J. Requirement for Pax6 in corneal morphogenesis: a role in adhesion. J Cell Sci. 2003 Jun 1;116(Pt 11):2157-67

Free SL, Mitchell TN, Williamson KA, Churchill AJ, Shorvon SD, Moore AT, van Heyningen V, Sisodiya SM. Quantitative MR image analysis in subjects with defects in the PAX6 gene. Neuroimage. 2003 Dec;20(4):2281-90

Horie M, Sango K, Takeuchi K, Honma S, Osumi N, Kawamura K, Kawano H. Subpial neuronal migration in the medulla oblongata of Pax-6-deficient rats. Eur J Neurosci. 2003 Jan;17(1):49-57

Marsich E, Vetere A, Di Piazza M, Tell G, Paoletti S. The PAX6 gene is activated by the basic helix-loop-helix transcription factor NeuroD/BETA2. Biochem J. 2003 Dec 15;376(Pt 3):707-15

Mitchell TN, Free SL, Williamson KA, Stevens JM, Churchill AJ, Hanson IM, Shorvon SD, Moore AT, van Heyningen V, Sisodiya SM. Polymicrogyria and absence of pineal gland due to PAX6 mutation. Ann Neurol. 2003 May;53(5):658-63

Talamillo A, Quinn JC, Collinson JM, Caric D, Price DJ, West JD, Hill RE. Pax6 regulates regional development and

Page 30: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

PAX6 (paired box 6) Zhou YH

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 650

neuronal migration in the cerebral cortex. Dev Biol. 2003 Mar 1;255(1):151-63

Tyas DA, Pearson H, Rashbass P, Price DJ. Pax6 regulates cell adhesion during cortical development. Cereb Cortex. 2003 Jun;13(6):612-9

Zhang X, Heaney S, Maas RL. Cre-loxp fate-mapping of Pax6 enhancer active retinal and pancreatic progenitors. Genesis. 2003 Jan;35(1):22-30

Zhou YH, Tan F, Hess KR, Yung WK. The expression of PAX6, PTEN, vascular endothelial growth factor, and epidermal growth factor receptor in gliomas: relationship to tumor grade and survival. Clin Cancer Res. 2003 Aug 15;9(9):3369-75

Collinson JM, Chanas SA, Hill RE, West JD. Corneal development, limbal stem cell function, and corneal epithelial cell migration in the Pax6(+/-) mouse. Invest Ophthalmol Vis Sci. 2004 Apr;45(4):1101-8

Cvekl A, Yang Y, Chauhan BK, Cveklova K. Regulation of gene expression by Pax6 in ocular cells: a case of tissue-preferred expression of crystallins in lens. Int J Dev Biol. 2004;48(8-9):829-44

Haubst N, Berger J, Radjendirane V, Graw J, Favor J, Saunders GF, Stoykova A, Götz M. Molecular dissection of Pax6 function: the specific roles of the paired domain and homeodomain in brain development. Development. 2004 Dec;131(24):6131-40

Nomura T, Osumi N. Misrouting of mitral cell progenitors in the Pax6/small eye rat telencephalon. Development. 2004 Feb;131(4):787-96

Schuurmans C, Armant O, Nieto M, Stenman JM, Britz O, Klenin N, Brown C, Langevin LM, Seibt J, Tang H, Cunningham JM, Dyck R, Walsh C, Campbell K, Polleux F, Guillemot F. Sequential phases of cortical specification involve Neurogenin-dependent and -independent pathways. EMBO J. 2004 Jul 21;23(14):2892-902

Gehring WJ. New perspectives on eye development and the evolution of eyes and photoreceptors. J Hered. 2005 May-Jun;96(3):171-84

Grinchuk O, Kozmik Z, Wu X, Tomarev S. The Optimedin gene is a downstream target of Pax6. J Biol Chem. 2005 Oct 21;280(42):35228-37

Hack MA, Saghatelyan A, de Chevigny A, Pfeifer A, Ashery-Padan R, Lledo PM, Götz M. Neuronal fate determinants of adult olfactory bulb neurogenesis. Nat Neurosci. 2005 Jul;8(7):865-72

Kohwi M, Osumi N, Rubenstein JL, Alvarez-Buylla A. Pax6 is required for making specific subpopulations of granule and periglomerular neurons in the olfactory bulb. J Neurosci. 2005 Jul 27;25(30):6997-7003

Li T, Lu L. Epidermal growth factor-induced proliferation requires down-regulation of Pax6 in corneal epithelial cells. J Biol Chem. 2005 Apr 1;280(13):12988-95

Maekawa M, Takashima N, Arai Y, Nomura T, Inokuchi K, Yuasa S, Osumi N. Pax6 is required for production and maintenance of progenitor cells in postnatal hippocampal neurogenesis. Genes Cells. 2005 Oct;10(10):1001-14

Nacher J, Varea E, Blasco-Ibañez JM, Castillo-Gomez E, Crespo C, Martinez-Guijarro FJ, McEwen BS. Expression of the transcription factor Pax 6 in the adult rat dentate gyrus. J Neurosci Res. 2005 Sep 15;81(6):753-61

Pinson J, Mason JO, Simpson TI, Price DJ. Regulation of the Pax6 : Pax6(5a) mRNA ratio in the developing mammalian brain. BMC Dev Biol. 2005 Jul 19;5:13

Zhou YH, Wu X, Tan F, Shi YX, Glass T, Liu TJ, Wathen K, Hess KR, Gumin J, Lang F, Yung WK. PAX6 suppresses growth of human glioblastoma cells. J Neurooncol. 2005 Feb;71(3):223-9

Kim EA, Noh YT, Ryu MJ, Kim HT, Lee SE, Kim CH, Lee C, Kim YH, Choi CY. Phosphorylation and transactivation of Pax6 by homeodomain-interacting protein kinase 2. J Biol Chem. 2006 Mar 17;281(11):7489-97

Kleinjan DA, Seawright A, Mella S, Carr CB, Tyas DA, Simpson TI, Mason JO, Price DJ, van Heyningen V. Long-range downstream enhancers are essential for Pax6 expression. Dev Biol. 2006 Nov 15;299(2):563-81

Li T, Lu Z, Lu L. Pax6 regulation in retinal cells by CCCTC binding factor. Invest Ophthalmol Vis Sci. 2006 Dec;47(12):5218-26

Mayes DA, Hu Y, Teng Y, Siegel E, Wu X, Panda K, Tan F, Yung WK, Zhou YH. PAX6 suppresses the invasiveness of glioblastoma cells and the expression of the matrix metalloproteinase-2 gene. Cancer Res. 2006 Oct 15;66(20):9809-17

Tyas DA, Simpson TI, Carr CB, Kleinjan DA, van Heyningen V, Mason JO, Price DJ. Functional conservation of Pax6 regulatory elements in humans and mice demonstrated with a novel transgenic reporter mouse. BMC Dev Biol. 2006 May 4;6:21

Wu D, Li T, Lu Z, Dai W, Xu M, Lu L. Effect of CTCF-binding motif on regulation of PAX6 transcription. Invest Ophthalmol Vis Sci. 2006 Jun;47(6):2422-9

Bamiou DE, Campbell NG, Musiek FE, Taylor R, Chong WK, Moore A, van Heyningen V, Free S, Sisodiya S, Luxon LM. Auditory and verbal working memory deficits in a child with congenital aniridia due to a PAX6 mutation. Int J Audiol. 2007 Apr;46(4):196-202

Bamiou DE, Free SL, Sisodiya SM, Chong WK, Musiek F, Williamson KA, van Heyningen V, Moore AT, Gadian D, Luxon LM. Auditory interhemispheric transfer deficits, hearing difficulties, and brain magnetic resonance imaging abnormalities in children with congenital aniridia due to PAX6 mutations. Arch Pediatr Adolesc Med. 2007 May;161(5):463-9

Bel-Vialar S, Medevielle F, Pituello F. The on/off of Pax6 controls the tempo of neuronal differentiation in the developing spinal cord. Dev Biol. 2007 May 15;305(2):659-73

Berger J, Berger S, Tuoc TC, D'Amelio M, Cecconi F, Gorski JA, Jones KR, Gruss P, Stoykova A. Conditional activation of Pax6 in the developing cortex of transgenic mice causes progenitor apoptosis. Development. 2007 Apr;134(7):1311-22

Berkhout M, Nagtegaal ID, Cornelissen SJ, Dekkers MM, van de Molengraft FJ, Peters WH, Nagengast FM, van Krieken JH, Jeuken JW. Chromosomal and methylation alterations in sporadic and familial adenomatous polyposis-related duodenal carcinomas. Mod Pathol. 2007 Dec;20(12):1253-62

Chang JY, Hu Y, Siegel E, Stanley L, Zhou YH. PAX6 increases glioma cell susceptibility to detachment and oxidative stress. J Neurooncol. 2007 Aug;84(1):9-19

Duparc RH, Abdouh M, David J, Lépine M, Tétreault N, Bernier G. Pax6 controls the proliferation rate of neuroepithelial progenitors from the mouse optic vesicle. Dev Biol. 2007 Jan 15;301(2):374-87

Grocott T, Frost V, Maillard M, Johansen T, Wheeler GN, Dawes LJ, Wormstone IM, Chantry A. The MH1 domain of Smad3 interacts with Pax6 and represses autoregulation of the Pax6 P1 promoter. Nucleic Acids Res. 2007;35(3):890-901

Holm PC, Mader MT, Haubst N, Wizenmann A, Sigvardsson M, Götz M. Loss- and gain-of-function analyses reveal targets

Page 31: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

PAX6 (paired box 6) Zhou YH

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 651

of Pax6 in the developing mouse telencephalon. Mol Cell Neurosci. 2007 Jan;34(1):99-119

Manuel M, Georgala PA, Carr CB, Chanas S, Kleinjan DA, Martynoga B, Mason JO, Molinek M, Pinson J, Pratt T, Quinn JC, Simpson TI, Tyas DA, van Heyningen V, West JD, Price DJ. Controlled overexpression of Pax6 in vivo negatively autoregulates the Pax6 locus, causing cell-autonomous defects of late cortical progenitor proliferation with little effect on cortical arealization. Development. 2007 Feb;134(3):545-55

Pinto GR, Clara CA, Santos MJ, Almeida JR, Burbano RR, Rey JA, Casartelli C. Mutation analysis of gene PAX6 in human gliomas. Genet Mol Res. 2007 Oct 5;6(4):1019-25

Quinn JC, Molinek M, Martynoga BS, Zaki PA, Faedo A, Bulfone A, Hevner RF, West JD, Price DJ. Pax6 controls cerebral cortical cell number by regulating exit from the cell cycle and specifies cortical cell identity by a cell autonomous mechanism. Dev Biol. 2007 Feb 1;302(1):50-65

Yan Q, Liu WB, Qin J, Liu J, Chen HG, Huang X, Chen L, Sun S, Deng M, Gong L, Li Y, Zhang L, Liu Y, Feng H, Xiao Y, Liu Y, Li DW. Protein phosphatase-1 modulates the function of Pax-6, a transcription factor controlling brain and eye development. J Biol Chem. 2007 May 11;282(19):13954-65

Brill MS, Snapyan M, Wohlfrom H, Ninkovic J, Jawerka M, Mastick GS, Ashery-Padan R, Saghatelyan A, Berninger B, Götz M. A dlx2- and pax6-dependent transcriptional code for periglomerular neuron specification in the adult olfactory bulb. J Neurosci. 2008 Jun 18;28(25):6439-52

Canto-Soler MV, Huang H, Romero MS, Adler R. Transcription factors CTCF and Pax6 are segregated to different cell types during retinal cell differentiation. Dev Dyn. 2008 Mar;237(3):758-67

Davis LK, Meyer KJ, Rudd DS, Librant AL, Epping EA, Sheffield VC, Wassink TH. Pax6 3' deletion results in aniridia, autism and mental retardation. Hum Genet. 2008 May;123(4):371-8

Hellwinkel OJ, Kedia M, Isbarn H, Budäus L, Friedrich MG. Methylation of the TPEF- and PAX6-promoters is increased in early bladder cancer and in normal mucosa adjacent to pTa tumours. BJU Int. 2008 Mar;101(6):753-7

Lang D, Mascarenhas JB, Powell SK, Halegoua J, Nelson M, Ruggeri BA. PAX6 is expressed in pancreatic adenocarcinoma

and is downregulated during induction of terminal differentiation. Mol Carcinog. 2008 Feb;47(2):148-56

Oron-Karni V, Farhy C, Elgart M, Marquardt T, Remizova L, Yaron O, Xie Q, Cvekl A, Ashery-Padan R. Dual requirement for Pax6 in retinal progenitor cells. Development. 2008 Dec;135(24):4037-47

Osumi N, Shinohara H, Numayama-Tsuruta K, Maekawa M. Concise review: Pax6 transcription factor contributes to both embryonic and adult neurogenesis as a multifunctional regulator. Stem Cells. 2008 Jul;26(7):1663-72

Robinson DO, Howarth RJ, Williamson KA, van Heyningen V, Beal SJ, Crolla JA. Genetic analysis of chromosome 11p13 and the PAX6 gene in a series of 125 cases referred with aniridia. Am J Med Genet A. 2008 Mar 1;146A(5):558-69

Tuoc TC, Stoykova A. Er81 is a downstream target of Pax6 in cortical progenitors. BMC Dev Biol. 2008 Feb 28;8:23

Gubert F, Zaverucha-do-Valle C, Pimentel-Coelho PM, Mendez-Otero R, Santiago MF. Radial glia-like cells persist in the adult rat brain. Brain Res. 2009 Mar 3;1258:43-52

Hingorani M, Williamson KA, Moore AT, van Heyningen V. Detailed ophthalmologic evaluation of 43 individuals with PAX6 mutations. Invest Ophthalmol Vis Sci. 2009 Jun;50(6):2581-90

Mascarenhas JB, Young KP, Littlejohn EL, Yoo BK, Salgia R, Lang D. PAX6 is expressed in pancreatic cancer and actively participates in cancer progression through activation of the MET tyrosine kinase receptor gene. J Biol Chem. 2009 Oct 2;284(40):27524-32

Sansom SN, Griffiths DS, Faedo A, Kleinjan DJ, Ruan Y, Smith J, van Heyningen V, Rubenstein JL, Livesey FJ. The level of the transcription factor Pax6 is essential for controlling the balance between neural stem cell self-renewal and neurogenesis. PLoS Genet. 2009 Jun;5(6):e1000511

Zhou YH, Hu Y, Mayes D, Siegel E, Kim JG, Mathews MS, Hsu N, Eskander D, Yu O, Tromberg BJ, Linskey ME. PAX6 suppression of glioma angiogenesis and the expression of vascular endothelial growth factor A. J Neurooncol. 2010 Jan;96(2):191-200

This article should be referenced as such:

Zhou YH. PAX6 (paired box 6). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):645-651.

Page 32: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 652

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Luke B Hesson, Farida Latif

Lowy Cancer Centre and Prince of Wales Clinical School, Faculty of Medicine, University of New South

Wales, NSW2052, Australia (LBH), School of Clinical and Experimental Medicine, College of Medical and

Dental Sciences, Department of Medical and Molecular Genetics, University of Birmingham, Birmingham

B15 2TT, UK (FL)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/RASSF2ID43461ch20p13.html DOI: 10.4267/2042/44798

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: DKFZp781O1747; KIAA0168;

RASFADIN

HGNC (Hugo): RASSF2

Location: 20p13

Local order:

Telomere-PRNP-PRNT-RASSF2-SLC23A2-

Centromere.

Juxtaposed to the PRNP prion locus conserved in the

syntenic bovine region (Choi et al., 2006).

Note: Brief overview

The RASSF family of tumour suppressor genes (TSG)

encode Ras superfamily effector proteins that, amongst

other functions, mediate some of the growth inhibitory

functions of Ras proteins. Several members of this

family are inactivated by promoter DNA

hypermethylation in a broad range of cancers and

inactivation of RASSF2 has been described in a

growing number of tumour types. RASSF2 functions as

a K-Ras adaptor protein and mediates some of the

growth inhibitory properties of K-Ras. RASSF2

regulates apoptosis and cell cycle progression through

interactions with several downstream effectors

including MST1 and MST2.

DNA/RNA

Description

The RASSF2 gene occupies 43,621bp of genomic

DNA (-ve strand). RASSF2A variant 1

[GenBank:NM_014737] contains 12 exons and is

transcribed from a large (1,850bp) 5' CpG island

encompassing the first two non-coding exons. There is

evidence of multiple splice variants and transcription

initiation sites for the RASSF2 gene. Additional

isoforms of RASSF2 include RASSF2A variant 2

[GenBank:CR627436] that is predicted to produce an

identical protein to RASSF2A variant 1, RASSF2B

[GenBank:AY154471] and RASSF2C

[GenBank:AY154472].

A further isoform [GenBank:CR620887] produces a

non-coding mRNA. RASSF2B and RASSF2C are not

associated with CpG islands (figure 1). Akino et al.,

(2005) investigated promoter activity of the region

upstream of the transcription start site of NM_014737

(RASSF2A variant 1) and found promoter activity was

dependent on a CACCC box and SP1 site just upstream

of exon 1. The authors however did not investigate the

CpG island region of the RASSF2 gene, which is

largely located in intron 1 of the NM_014737

transcript.

Page 33: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 653

Figure 1: RASSF2 gene structure. Transcription of the RASSF2A isoforms begins within a CpG island that spans exons 1 and 2 (grey box) at -105 to +1745 bp relative to the transcription start site of NM_014737. RASSF2B transcription begins at exon 1beta in intron 5. Transcription of RASSF2C begins at exon 1gamma in intron 2. RASSF2B and RASSF2C do not have 5' CpG islands or predicted promoter regions.

Protein

Description

As mentioned above multiple isoforms are expressed

from the RASSF2 locus. However, RASSF2A variants

1 and 2, as well as RASSF2C (if expressed at all)

contain identical open reading frames encoding a 326

amino acid protein, whilst RASSF2B mRNA is

predicted to encode a truncated protein of 157 amino

acids but is expressed at extremely low levels in all

tissues analysed. Therefore, the protein is simply

referred to as RASSF2 in the literature. The RASSF2

protein (figure 2) contains C-terminal Ras-association

(RA) and Sav/RASSF/Hpo (SARAH) domains that

define the 'classical' RASSF family (RASSF1,

RASSF2, RASSF3, RASSF4, RASSF5, RASSF6). In

addition RASSF2 contains a central bipartite nuclear

localisation signal (NLS) which has been shown to be

essential for tumour suppressor function (Cooper et al.,

2008). The C-terminus of RASSF2 also contains a

sequence shown to be necessary for nuclear export

(Kumari et al., 2009). Detection of endogenous

RASSF2 protein has been described in a variety of cell

lines using an in-house antibody (Vos et al., 2003) or a

commercially available antibody from Santa Cruz

(Cooper et al., 2009).

Expression

Northern blotting shows RASSF2 mRNA is highly

expressed in many normal tissues including brain,

thymus, spleen, liver, small intestines, placenta, lung

and peripheral blood (Vos et al., 2003). The probe used

for northern blotting did not discriminate between

RASSF2 isoforms.

The coding region of RASSF2 has been cloned from a

brain-specific cDNA library (Hesson et al., 2005).

Currently there has been limited analysis of expression

patterns and distribution of the different RASSF2

isoforms. RASSF2A variants 1 and 2 are both

ubiquitously expressed in a range of normal tissues

including colon, stomach, heart, bone marrow, kidney,

ovary, lung, liver, breast, testis and pancreas

(Maruyama et al., 2008). However, expression of the

RASSF2B and RASSF2C isoforms was virtually

undetectable in a range of normal tissues (Maruyama et

al., 2008; L Hesson and F Latif, unpublished

observations). Expression of the RASSF2 gene is

inactivated by DNA methylation of the 5' CpG island

promoter region in a broad spectrum of cancers (see

below).

Localisation

When over expressed RASSF2 is clearly predominantly

nuclear, as demonstrated by immunofluorescence

(Cooper et al., 2008; Kurnari et al., 2007). Some

evidence suggests that the NLS of RASSF2 is an

integral part of the ability of RASSF2 to act as a

tumour suppressor. The localisation of RASSF2 is cell

context specific. Two independent studies indicate that

phosphorylation of RASSF2 appears to be critical for

relocalisation to the cytoplasm, though the critical

phosphorylation sites remain to be determined. Cooper

Page 34: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 654

Figure 2: RASSF2 transcript and protein structure. RASSF2A [GenBank:NP_055552] is a 326 aa protein containing a central bipartite nuclear localisation signal (NLS), a Ras-association (RA) domain of the RalGDS/AF-6 variety and acidic coiled-coil Sav/RASSF/Hpo (SARAH) domain. RASSF2B [GenBank:AAN59976] is a predicted 157 amino acid protein containing a truncated RA domain. The RASSF2C predicted protein is identical to RASSF2A. The mRNA transcript shown (red bar) represents RASSF2A variant 1 (NM_014737).

Figure 3: RASSF2 is conserved with RASSF paralogues. Schematic representation of the 10 human members of the RASSF family showing the Ras-association, SARAH and predicted diacylglycerol binding domains. The longest isoform for each RASSF gene is represented (GenBank accession numbers: RASSF1A[NP_009113], RASSF2[NP_055552], RASSF3[NP_835463], RASSF4[NP_114412], RASSF5A[NP_872604], RASSF6[NP_958834], RASSF7[NP_003466], RASSF8[BAC98838], RASSF9[NP_005438], RASSF10[NP_001073990]). The RASSF family is subdivided into 'classical' RASSF members and 'N-terminal' RASSF members as indicated. Shown is the percentage amino acid identity of each RASSF members with RASSF2. Protein sequence identity of the 'classical' RASSF members is greatest over the C-terminus. * RASSF10 protein sequence as described in Hesson et al., 2009 (which is N-terminally truncated with respect to GenBank accession number NP_001073990).

et al., (2009) demonstrated that relocalisation of over

expressed RASSF2 from the nucleus to the cytoplasm

is dependent on active MST1 or MST2 and that either

kinase was capable of phosphorylating RASSF2 in

vitro. However, the work of Kumari et al., (2009)

demonstrates that RASSF2 relocalisation is dependent

on the activity of Extracellular signal-Related Kinase 2

(ERK2). Both MSTs and ERK2 can participate in Ras

signalling therefore both studies may be observing the

effects of activation of the same pathway. The presence

of sequences essential for both nuclear import and

export within RASSF2 seems to suggest that the

protein may continuously cycle between cytoplasm and

nucleus in a similar manner to MST1 and MST2 (Lee

and Yonehara, 2002). Given the strong binding of

RASSF2 with MST1 and MST2 (see below) it seems

likely that this would occur in complex with MSTs.

RASSF2 nuclear important may be dependent on

importin-alpha interaction (Kumari et al., 2007), whilst

nuclear export appears to involve the NES (nuclear

export signal)-dependent transport protein CRM-

1/XPO1 (Kumari et al., 2009). What remains to be

determined is the exact conditions under which the

kinetics of nuclear export predominates nuclear import

and vice versa.

Function

Tumour suppressor function of RASSF2

Similar to several other RASSF members RASSF2

suppresses tumour growth when expressed. This has

been demonstrated for colorectal, lung, breast, gastric,

nasopharyngeal and oral squamous cell carcinoma

(OSCC) cell lines in vitro using colony formation,

growth curve and soft agar growth assays (Akino et al.,

2005; Vos et al., 2003; Cooper et al., 2008; Maruyama

et al., 2008; Imai et al., 2008; Zhang et al., 2006).

Page 35: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 655

Furthermore, RASSF2 re-expression in breast tumour

cells inhibits in vivo tumour growth when cells are

subcutaneously injected into severe combined

immunodeficiency (SCID) mice (Cooper et al., 2008).

Several studies demonstrate that these tumour

suppressive properties are likely to arise from the

ability of RASSF2 to regulate apoptosis and cell cycle

progression (Vos et al., 2003; Maruyama et al., 2008;

Imai et al., 2008; Akino et al., 2005).

In breast cancer cells and Cos-7 cells growth

suppression by RASSF2 is dependent on the nuclear

localisation signal (NLS) located at amino acids 151-

167 (Cooper et al., 2008; Kumari et al., 2009), whilst

other reports have indicated that in OSCC and gastric

cancer cells the C-terminal portion of RASSF2

(RASSF2 [163-326]), containing the RA domain, is

critical for tumour suppressive function (Imai et al.,

2008; Maruyama et al., 2008). Interestingly, in OSCC

this C-terminal portion exhibited enhanced growth

suppression relative to full length RASSF2 (Imai et al.,

2008). In fact, RASSF2 [163-326] also disrupts the

NLS yet leaves the sequence required for nuclear

export intact. In a separate study of colorectal cancer

cells both RASSF2 truncations (RASSF2 [1-163] and

RASSF2 [163-326]) exhibited reduced growth

suppression compared to full length RASSF2 (Akino et

al., 2005). Whilst in gastric cancer transfection of

RASSF2 with deletion of the NLS [RASSF2deltaNLS]

actually increased the percentage of apoptotic cells

relative to full length RASSF2 (Maruyama et al.,

2008). These studies indicate the growth suppressive

properties of RASSF2 are likely cell background

specific but more importantly that nuclear import,

nuclear export and the Ras-association domain are

required for correctly regulated RASSF2 growth

suppression.

RASSF2 interaction with Ras

RASSF2 contains a functional RA domain that displays

a strong binding to K-Ras, but only weak binding to H-

Ras (Vos et al., 2003). RASSF2 associates with the K-

Ras effector domain in a GTP-dependent manner thus

displaying the basic properties of a genuine Ras

effector. RASSF2 growth inhibition is enhanced in the

presence of K-RasG12V. Furthermore, siRNA-

mediated knock-down of RASSF2 in K-Ras

transformed cells enhanced anchorage-independent

growth. However, in the absence of K-Ras

transformation knock-down of RASSF2 inhibited

growth (Akino et al., 2005). These data indicate that

RASSF2 mediates some of the growth inhibitory

properties of K-Ras and that inactivation of RASSF2

enhances K-Ras-induced transformation.

RASSF2 interaction with the proapoptotic kinases

MST1 and MST2.How the interaction of RASSF

proteins with Ras results in growth suppression has

been the subject of intense investigation. The

proapoptotic mammalian Serine/Threonine kinases

MST1 and MST2 were identified as RASSF2

interacting partners by yeast two-hybrid (Y2-H)

(Khokhlatchev et al., 2002). RASSF1 and RASSF5

were also identified as MST binding partners as part of

a novel Ras-regulated signalling pathway. Recently the

interaction of RASSF2 and MST1/2 was formally

demonstrated in human cells at the endogenous level.

Interaction occurs between the SARAH domains found

within RASSF2 and MST1/2 (Cooper et al., 2009).

RASSF2 appears to have a distinct role in regulating

MST2 function. Activation of MST2 is followed by a

rapid proteasome-dependent loss of MST2 stability

(that is not associated with MST2 cleavage).

Interestingly, over expression of RASSF2 results in

increased levels of MST2 and provides protection of

MST2 from degradation following its activation. In

agreement with this loss of RASSF2 protein in

colorectal tumours, or in colorectal tumour cell lines in

which RASSF2 levels are decreased by shRNAi, also

leads to decreases in MST2 levels (Cooper et al., 2009).

RASSF2 appears to be a substrate for MST1 and MST2

and co-expression of either kinase with RASSF2

relocalises RASSF2 from the nucleus to the cytoplasm

in a manner dependent on kinase activity (Cooper et al.,

2009). Since MST2 remains in complex with RASSF2

following its activation these data collectively suggest

RASSF2 stabilises active MST2 allowing (or perhaps

even targeting) MST2 substrate phosphorylation. This

RASSF2-mediated stabilisation also appears to be true

for MST1. Thus loss of RASSF2, as is frequently

observed in cancer, leads to loss of MST1 and MST2

leading to a decrease in apoptotic potential. That

RASSF2 appears to be capable of influencing MST

stability so drastically is likely to be due to the

observation that the majority of both MST1 and MST2

are in complex with RASSF2 in at least some cell types

(Cooper et al., 2009). The interaction of RASSF2 with

MST1/2 poses an interesting question with regards to

the regulation of RASSF2 localisation. Both RASSF2

and MST1/2 have been shown to contain sequences

essential for nuclear import and export (Lee and

Yonehara, 2002; Kumari et al., 2009). Both the NLS

and NES sequences within MST1/2 and RASSF2

respectively are located very close to or within the

SARAH domains and neither are canonical NLS/NES

sequences. Mapping of both these sequences were

determined by deletion mapping, which would most

likely also affect RASSF2-MST1/2 interaction thus it

now seems likely that the RASSF2-MST1/2 complex

constantly cycles between the nucleus (by virtue of

RASSF2 NLS) and cytoplasm (by virture of MST1/2

NES) and disruption of the interaction between

RASSF2 and MST1/2 would likely affect the

localisation of both proteins. Also, the fact that

RASSF2 translocation to the cytoplasm is dependent on

ERK2 activity (Kumari and Mahalingam, 2009)

suggests the Ras-MEK-ERK pathway may serve to

phosphorylate MST1/2, which then phosphorylates

RASSF2, translocating it to the cytoplasm and allowing

RASSF2 to interact with Ras (figure 4). That nuclear

RASSF2 is required for full tumour suppressor activity

Page 36: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 656

(Cooper et al., 2008; Kumari et al., 2009) may be

explained by the fact that ERK2 translocates to the

nucleus upon its activation (Khokhlatchev et al., 1998).

Other functions of RASSF2

Other functions and interacting partners of RASSF2 are

extremely likely. Y2-H using RASSF2 as bait

implicates NORE1A and RASSF3 in RASSF2

function, although these have not yet been confirmed in

mammalian cells (Hesson et al., 2005). These

interactions may implicate other RASSF members in

modulating RASSF2 function and suggests a complex

network of cross-talk between signalling pathways

involving RASSF proteins. Also, the exact mechanisms

of apoptotic and cell cycle regulation of RASSF2 have

yet to be completely defined. Microarray analysis of

gene expression before and after exogenous expression

of RASSF2 in gastric and OSCC cancer cell lines

showed RASSF2 downregulates expression of several

inflammatory response genes including the cytokines

IL-8, LCN2, CXCL1, CXCL2, CXCL3, CXCL5 and

CXCL6, CCL20 and CCL21 and genes involved in

immune-cell chemotaxis (Maruyama et al., 2008; Imai

et al., 2008). A possible pathway influenced by

RASSF2 is the NF-kB pathway since over expression

of RASSF2 significantly downregulated NF-kB

transcriptional activity (Maruyama et al., 2008; Imai et

al., 2008). Of note is the recent observation that pigs

experimentally infected with Porcine Circovirus Type 2

(PCV2) show upregulation of several CXCL family

cytokines as well as RASSF2 (Fernandes et al., 2009)

therefore it is likely a role for RASSF2 in regulating

immune response pathways remains to be discovered.

There is also evidence that RASSF2 may regulate the

actin cytoskeleton since re-expression of RASSF2 leads

to loss of stress fibres, cell rounding and the

suppression of RhoGTPase activation (Maruyama et

al., 2008; Akino et al., 2005). Additionally, RASSF2

upregulation appears to be a cellular response to

ionising radiation (Sakamoto-Hojo et al., 2003).

Homology

Human RASSF2 has highly conserved orthologues

across many species (table 1). The main features of the

RASSF2 protein are conserved across these species

including the RA and SARAH domains as well as the

NLS and the sequence important for nuclear export.

RASSF2 is one of 10 members of the Ras-association

domain family (RASSF) comprising RASSF1-10.

RASSF1-6 are termed the 'classical' RASSF family and

contain C-terminal RA and SARAH domains.

Consequently, RASSF1-6 are most similar in sequence

within their C-termini. RASSF7-10 (RASSF7,

RASSF8, RASSF9, RASSF10) represent evolutionarily

conserved but structurally distinct RASSF members

that lack the SARAH domains and contain N-terminal

RA domains. RASSF7-10 are termed the 'N-terminal'

RASSF family. RASSF2 is most similar to

Figure 4: One possible RASSF2 pathway. Recent evidence suggests a RASSF2-MST1/2 complex may continuously cycle through the nucleus and that nuclear localisation of RASSF2 is essential for tumour suppressor function. There are also reports demonstrating that the activity of the kinases ERK2 and MST1/2 is crucial for cytoplasmic relocalisation of RASSF2. Therefore activation of the RASSF2 tumour suppressor pathway may emanate from the nucleus following ERK2 and MST1/2 activation allowing RASSF2 to accumulate in the cytoplasm where it may encounter another interacting partner K-RasGTP.

Page 37: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 657

Table 1: RASSF2 has highly conserved orthologues in several species.

RASSF4 and RASSF6 (figure 3), both of which are

also epigenetically inactivated in cancer, and

participate in K-Ras signalling to inhibit tumour cell

growth and induce apoptosis (Hesson et al., 2009;

Ikeda et al., 2006; Allen et al., 2006; Eckfeld et al.,

2005; Chow et al., 2004).

Mutations

Note

Similar to all other RASSF members, perhaps with the

exception of RASSF1A (Kashuba et al., 2009; Pan et

al., 2005), mutation of RASSF2 is a rare event and to

date no inactivating mutations have been described.

Analysis of ovarian primary tumours failed to identify a

single amino acid changing or truncating point

mutation (Cooper et al., 2008). However, more

thorough investigations of larger cohorts of different

tumour types may be required to determine this

definitively.

Implicated in

Colorectal carcinoma (CRC)

Note

Colorectal carcinoma (CRC) including colon

adenomas.

Prognosis

In early colorectal cancers RASSF2 methylation with

oncogenic activation of either K-Ras, B-Raf or

PIK3CA presented significantly more frequently in

cases of venous invasion (Nosho et al., 2007).

Oncogenesis

Similar to several other RASSF members, RASSF2 is

frequently inactivated by CpG island DNA

hypermethylation (Hesson et al., 2007). RASSF2

inactivation has been most extensively investigated in

colorectal cancer in which inactivation of other RASSF

members is relatively rare. Several studies now

Page 38: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 658

strongly suggest that RASSF2 inactivation is a frequent

and early event in colorectal cancer formation being

present in colon adenomas, particularly those with a

villous component (Kakar et al., 2008; Hesson et al.,

2005; Akino et al., 2005; Harada et al., 2007). The

frequencies of RASSF2 methylation in CRCs vary

between 42% (Akino et al., 2005) and 70% (Hesson et

al., 2005) whilst for adenomas methylation occurs

between 25% (Harada et al., 2007) and 94% (Kakar et

al., 2008). This most likely reflects the differences in

histopathological subtypes and the CpG island region

analysed as well differences in colorectal cancer

aetiology in different populations.

Some reports describe variable frequencies of RASSF2

methylation depending on tumour location within the

colon (Harada et al., 2007). Interestingly, cells from

apparently normal colonic epithelium from patients

with hyperplastic polyposis (HPP) also frequently

demonstrate methylation of several TSGs including

RASSF2 (Minoo et al., 2006). This may indicate an

early role for RASSF2 inactivation in colonic

hyperplasia. RASSF2 methylation was found to be

associated with K-Ras or BRAF mutation (Harada et

al., 2007; Akino et al., 2005) however, in another study

K-Ras mutation and RASSF2 methylation were

mutually exclusive (Hesson et al., 2005). Methylation

of the RASSF2 promoter and loss of expression

occurred in conjunction with loss of histone H3

acetylation, a marker of transcriptional activity.

Reduced RASSF2A expression also correlated with

methylation in primary CRCs (Akino et al., 2005). In

colorectal tumour cell lines re-expression of RASSF2

resulted in inhibition of anchorage-independent growth

in soft agar, which was associated with morphological

changes, cell detachment, disruption of the actin stress

fibre network, decreased Rho activity, increased

apoptosis and inhibition of cell cycle progression.

Deregulation of the actin cytoskeletal network and

morphological changes that result in cellular

detachment may therefore be a mechanism by which

RASSF2 induces anoikis, a form of suspension-

dependent apoptosis.

Non-small cell lung carcinoma (NSCLC)

Oncogenesis

Investigation of NSCLC primary tumours found

methylation of the RASSF2 CpG island in 44%

(22/50). Methylation was found at an equal frequency

in all grades (I/II = 44% (7/16); IIIA = 44% (4/9) and

IV = 44% (10/23)) suggesting RASSF2 became

hypermethylated early in tumour formation and was not

associated with development to higher grades (Cooper

et al., 2008). The incidence of RASSF2 methylation

appears much more frequent in NSCLC than SCLC

(small cell lung cancer) as shown by an earlier study by

Kaira et al., (2007), in which only 18% (4/22) SCLC

but 62% (16/26) NSCLC cell lines demonstrated

RASSF2 methylation with concomitant loss of

RASSF2A expression. RASSF2A expression was

restored following treatment with the DNA

demethylating agent 5-aza-2'deoxycytidine and/or

trichostatin A. In primary NSCLC tumours 31%

(33/106) were methylated and methylation was more

frequent in specimens from non-smokers than from

smokers (45%, 18/40 vs 23%, 15/66 respectively;

p=0.014).

Nasopharyngeal carcinoma

Prognosis

RASSF2 methylation correlated with lymph node

metastasis in nasopharyngeal carcinoma (Zhang et al.,

2006).

Oncogenesis

Fifty one percent (27/53) of primary nasopharyngeal

carcinomas (NPCs) showed cancer-specific RASSF2

methylation, which correlated with loss of RASSF2A

expression in both NPC cell lines and primary tumours

(Zhang et al., 2006). This study also provided evidence

that RASSF2 re-expression suppressed colony

formation ability in NPC cell lines (with concomitant

inhibition of cell cycle progression) and decreased cell

motility and migration as determined by wound healing

assay.

Gastric cancer

Prognosis

Both Maruyama et al., (2008) and Endoh et al., (2005)

found DNA methylation of the region around the

transcription start site of RASSF2A variant 1

significantly correlated with an absence of lymphatic

invasion. Whilst Maruyama et al., (2009) found further

associations with methylation and the absence of

venous invasion or lymph node metastasis, less

advanced stage, presence of EBV infection, the absence

of TP53 mutations and the presence of a CpG-island

methylator phenotype (CIMP).

Oncogenesis

The RASSF2 gene contains a 1.8kb CpG island that

encompasses the first two non-coding exons (figure 1).

The majority of this CpG island was interrogated for

hypermethylation in a series of gastric cancers (Endoh

et al., 2005). The study found varying frequencies of

methylation throughout the CpG island ranging from

29% (23/78) at the region encompassing the

transcription start site, to 79% (62/78) at a region of the

CpG island within intron 1 (though this intronic region

also exhibited a frequency of 60% (47/78) methylation

in corresponding normal gastric epithelia). Methylation

was mostly cancer specific at and around the

transcription start site and silencing of RASSF2A

expression most closely correlated with methylation at

this region (Endoh et al., 2005). Similar findings were

described by Maruyama et al., (2008) who examined

the methylation status of the CpG island regions

encompassing the transcriptional start sites of

RASSF2A variants 1 and 2. Methylation of RASSF2A

variant 1 was detected in 29.5% (23/78), whilst

Page 39: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 659

RASSF2A variant 2 was methylated in 25.6% (23/78)

of gastric cancer cases.

This may be significant since the expression of

RASSF2A variants 1 and 2 appear to be differentially

regulated by DNA methylation from within the same

CpG island indicating multiple promoters and possibly

accounting for the differences in methylation densities

throughout the region. RASSF2A re-expression

inhibited the growth of gastric cancer cell lines as

shown by reduced colony formation ability. This was a

result of inhibition of cell cycle progression and

induction of apoptosis (Maruyama et al., 2008).

Prostate cancer

Prognosis

A prospective study of a large cohort of patients

referred for prostate biopsy determined that detection

of RASSF2 methylation in patient urine shows

promising clinical utility as an early detection

biomarker for prostate cancer (Payne et al., 2009).

Though primary prostate tumour tissues were not

investigated the study nevertheless provided

information independent of the extensively used pre-

existing prostate cancer biomarker PSA (prostate

specific antigen). Detection of RASSF2 methylation

was significantly more frequent in patients with non-

organ-confined prostate cancer (Payne et al., 2009).

Thus RASSF2 methylation may represent an interesting

biomarker for early prostate cancer detection and

predicting invasive potential but requires further

validation.

Breast cancer

Oncogenesis

RASSF2 was frequently hypermethylated in primary

breast cancers (38%, 15/40) and re-expression in breast

cancer cell lines inhibited colony formation ability,

anchorage-independent growth in soft agar and in vivo

tumour formation in SCID mice (Cooper et al., 2008).

RASSF2 growth suppression was dependent on a

functional NLS (located at amino acids 151-167) since

its mutation prevented anchorage-independent growth

inhibition.

Hepatocellular carcinoma (HCC)

Oncogenesis

In an extensive DNA hypermethylation analysis of

cancerous and non-cancerous liver tissues (including

normal liver tissues from non-cancer patients) Nishida

et al., (2008) assessed the methylation status of 19 gene

loci in hepatitis B virus (HBV) and hepatitis C virus

(HCV)-related HCCs. This study found that normal

ageing within liver tissues is associated with a gradual

increase in aberrant methylation and that HCV

infection in particular may accelerate age-related

methylation. RASSF2 methylation was identified as a

cancer-specific event that is completely absent in

normal liver (0/22), infrequent in non-cancerous liver

from HCC patients (2.6%, 2/77), yet frequent in HCV-

related HCC (48%, 21/44) vs virus-negative HCC

(5.6%, 1/18; p=0.0029). Though this suggests loss of

RASSF2 expression may play a role in HCC the

potential importance and clinical implications of these

findings require further investigation.

Oral squamous cell carcinoma (OSCC)

Oncogenesis

Analysis of the expression of RASSF1-6 in OSCC cell

lines identified RASSF2 as the most frequently

downregulated RASSF gene analysed. This loss of

expression was caused by RASSF2 CpG island

methylation, which was found in 26% (12/46) of

primary OSCCs (Imai et al., 2008). In OSCC cell lines

re-expression of RASSF2 inhibited colony formation

ability by inducing apoptosis and inhibiting cell cycle

progression. Investigation of 482 OSCCs identified

RASSF2 methylation in 28% (134/482) cases. The

combination of RASSF1A and RASSF2 methylation

was significantly associated with poor disease-free

survival (p=0.009, Huang et al., 2009). Furthermore,

methylation of RASSF1A and RASSF2 increased in

patients undergoing post-surgical radiotherapy when

compared with surgery-only patients possibly

indicating that hypermethylation of RASSF1A and

RASSF2 is associated with the radioresistance

commonly observed in OSCC patients. It also indicates

the potential of using combined epigenetic and

radiotherapy as an adjuvant to surgery.

Endometrial carcinoma

Oncogenesis

Liao et al., (2008) investigated endometrial carcinomas

for RASSF2 CpG island methylation and found 25/76

(33%) were methylated. RASSF2 methylation was

found more frequently in tumour samples from older

patients. Similar findings were observed in colorectal

carcinomas (Hesson et al., 2005) and in OSCC (Imai et

al., 2008) suggesting loss of RASSF2 expression may

be a gradual age-related process.

Ovarian cancer

Oncogenesis

Although methylation of the RASSF2 promoter is not

an event associated with ovarian cancer (Cooper et al.,

2008) the gene does localise to one of the regions

deleted in ovarian tumour cell lines as indicated by

array-based genomic hybridisation (Lambros et al.,

2005).

Leukaemia

Oncogenesis

Recent evidence suggests that RASSF2 expression may

be downregulated in leukaemias with MLL

rearrangement by overexpression of one or more of the

miR-17-92 polycistronic miRNA oncogene cluster (Li

et al., 2009) that may target a region within the 3'UTR

of the RASSF2 mRNA.

Page 40: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 660

References Khokhlatchev AV, Canagarajah B, Wilsbacher J, Robinson M, Atkinson M, Goldsmith E, Cobb MH. Phosphorylation of the MAP kinase ERK2 promotes its homodimerization and nuclear translocation. Cell. 1998 May 15;93(4):605-15

Khokhlatchev A, Rabizadeh S, Xavier R, Nedwidek M, Chen T, Zhang XF, Seed B, Avruch J. Identification of a novel Ras-regulated proapoptotic pathway. Curr Biol. 2002 Feb 19;12(4):253-65

Lee KK, Yonehara S. Phosphorylation and dimerization regulate nucleocytoplasmic shuttling of mammalian STE20-like kinase (MST). J Biol Chem. 2002 Apr 5;277(14):12351-8

Sakamoto-Hojo ET, Mello SS, Pereira E, Fachin AL, Cardoso RS, Junta CM, Sandrin-Garcia P, Donadi EA, Passos GA. Gene expression profiles in human cells submitted to genotoxic stress. Mutat Res. 2003 Nov;544(2-3):403-13

Vos MD, Ellis CA, Elam C, Ulku AS, Taylor BJ, Clark GJ. RASSF2 is a novel K-Ras-specific effector and potential tumor suppressor. J Biol Chem. 2003 Jul 25;278(30):28045-51

Chow LS, Lo KW, Kwong J, Wong AY, Huang DP. Aberrant methylation of RASSF4/AD037 in nasopharyngeal carcinoma. Oncol Rep. 2004 Oct;12(4):781-7

Eckfeld K, Hesson L, Vos MD, Bieche I, Latif F, Clark GJ. RASSF4/AD037 is a potential ras effector/tumor suppressor of the RASSF family. Cancer Res. 2004 Dec 1;64(23):8688-93

Akino K, Toyota M, Suzuki H, Mita H, Sasaki Y, Ohe-Toyota M, Issa JP, Hinoda Y, Imai K, Tokino T. The Ras effector RASSF2 is a novel tumor-suppressor gene in human colorectal cancer. Gastroenterology. 2005 Jul;129(1):156-69

Endoh M, Tamura G, Honda T, Homma N, Terashima M, Nishizuka S, Motoyama T. RASSF2, a potential tumour suppressor, is silenced by CpG island hypermethylation in gastric cancer. Br J Cancer. 2005 Dec 12;93(12):1395-9

Hesson LB, Wilson R, Morton D, Adams C, Walker M, Maher ER, Latif F. CpG island promoter hypermethylation of a novel Ras-effector gene RASSF2A is an early event in colon carcinogenesis and correlates inversely with K-ras mutations. Oncogene. 2005 Jun 2;24(24):3987-94

Lambros MB, Fiegler H, Jones A, Gorman P, Roylance RR, Carter NP, Tomlinson IP. Analysis of ovarian cancer cell lines using array-based comparative genomic hybridization. J Pathol. 2005 Jan;205(1):29-40

Pan ZG, Kashuba VI, Liu XQ, Shao JY, Zhang RH, Jiang JH, Guo C, Zabarovsky E, Ernberg I, Zeng YX. High frequency somatic mutations in RASSF1A in nasopharyngeal carcinoma. Cancer Biol Ther. 2005 Oct;4(10):1116-22

Choi SH, Kim IC, Kim DS, Kim DW, Chae SH, Choi HH, Choi I, Yeo JS, Song MN, Park HS. Comparative genomic organization of the human and bovine PRNP locus. Genomics. 2006 May;87(5):598-607

Minoo P, Baker K, Goswami R, Chong G, Foulkes WD, Ruszkiewicz AR, Barker M, Buchanan D, Young J, Jass JR. Extensive DNA methylation in normal colorectal mucosa in hyperplastic polyposis. Gut. 2006 Oct;55(10):1467-74

Vos MD, Dallol A, Eckfeld K, Allen NP, Donninger H, Hesson LB, Calvisi D, Latif F, Clark GJ. The RASSF1A tumor suppressor activates Bax via MOAP-1. J Biol Chem. 2006 Feb 24;281(8):4557-63

Allen NP, Donninger H, Vos MD, Eckfeld K, Hesson L, Gordon L, Birrer MJ, Latif F, Clark GJ. RASSF6 is a novel member of the RASSF family of tumor suppressors. Oncogene. 2007 Sep 13;26(42):6203-11

Harada K, Hiraoka S, Kato J, Horii J, Fujita H, Sakaguchi K, Shiratori Y. Genetic and epigenetic alterations of Ras signalling pathway in colorectal neoplasia: analysis based on tumour clinicopathological features. Br J Cancer. 2007 Nov 19;97(10):1425-31

Hesson LB, Cooper WN, Latif F. Evaluation of the 3p21.3 tumour-suppressor gene cluster. Oncogene. 2007 Nov 15;26(52):7283-301

Hesson LB, Cooper WN, Latif F. The role of RASSF1A methylation in cancer. Dis Markers. 2007;23(1-2):73-87

Ikeda M, Hirabayashi S, Fujiwara N, Mori H, Kawata A, Iida J, Bao Y, Sato Y, Iida T, Sugimura H, Hata Y. Ras-association domain family protein 6 induces apoptosis via both caspase-dependent and caspase-independent pathways. Exp Cell Res. 2007 Apr 15;313(7):1484-95

Kaira K, Sunaga N, Tomizawa Y, Yanagitani N, Ishizuka T, Saito R, Nakajima T, Mori M. Epigenetic inactivation of the RAS-effector gene RASSF2 in lung cancers. Int J Oncol. 2007 Jul;31(1):169-73

Kumari G, Singhal PK, Rao MR, Mahalingam S. Nuclear transport of Ras-associated tumor suppressor proteins: different transport receptor binding specificities for arginine-rich nuclear targeting signals. J Mol Biol. 2007 Apr 13;367(5):1294-311

Nosho K, Yamamoto H, Takahashi T, Mikami M, Taniguchi H, Miyamoto N, Adachi Y, Arimura Y, Itoh F, Imai K, Shinomura Y. Genetic and epigenetic profiling in early colorectal tumors and prediction of invasive potential in pT1 (early invasive) colorectal cancers. Carcinogenesis. 2007 Jun;28(6):1364-70

Zhang Z, Sun D, Van do N, Tang A, Hu L, Huang G. Inactivation of RASSF2A by promoter methylation correlates with lymph node metastasis in nasopharyngeal carcinoma. Int J Cancer. 2007 Jan 1;120(1):32-8

Cooper WN, Dickinson RE, Dallol A, Grigorieva EV, Pavlova TV, Hesson LB, Bieche I, Broggini M, Maher ER, Zabarovsky ER, Clark GJ, Latif F. Epigenetic regulation of the ras effector/tumour suppressor RASSF2 in breast and lung cancer. Oncogene. 2008 Mar 13;27(12):1805-11

Imai T, Toyota M, Suzuki H, Akino K, Ogi K, Sogabe Y, Kashima L, Maruyama R, Nojima M, Mita H, Sasaki Y, Itoh F, Imai K, Shinomura Y, Hiratsuka H, Tokino T. Epigenetic inactivation of RASSF2 in oral squamous cell carcinoma. Cancer Sci. 2008 May;99(5):958-66

Kakar S, Deng G, Cun L, Sahai V, Kim YS. CpG island methylation is frequently present in tubulovillous and villous adenomas and correlates with size, site, and villous component. Hum Pathol. 2008 Jan;39(1):30-6

Liao X, Siu MK, Chan KY, Wong ES, Ngan HY, Chan QK, Li AS, Khoo US, Cheung AN. Hypermethylation of RAS effector related genes and DNA methyltransferase 1 expression in endometrial carcinogenesis. Int J Cancer. 2008 Jul 15;123(2):296-302

Maruyama R, Akino K, Toyota M, Suzuki H, Imai T, Ohe-Toyota M, Yamamoto E, Nojima M, Fujikane T, Sasaki Y, Yamashita T, Watanabe Y, Hiratsuka H, Hirata K, Itoh F, Imai K, Shinomura Y, Tokino T. Cytoplasmic RASSF2A is a proapoptotic mediator whose expression is epigenetically silenced in gastric cancer. Carcinogenesis. 2008 Jul;29(7):1312-8

Nishida N, Nagasaka T, Nishimura T, Ikai I, Boland CR, Goel A. Aberrant methylation of multiple tumor suppressor genes in aging liver, chronic hepatitis, and hepatocellular carcinoma. Hepatology. 2008 Mar;47(3):908-18

Page 41: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

RASSF2 (Ras association (RalGDS/AF-6) domain family member 2) Hesson LB, Latif F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 661

Cooper WN, Hesson LB, Matallanas D, Dallol A, von Kriegsheim A, Ward R, Kolch W, Latif F. RASSF2 associates with and stabilizes the proapoptotic kinase MST2. Oncogene. 2009 Aug 20;28(33):2988-98

Fernandes LT, Tomás A, Bensaid A, Pérez-Enciso M, Sibila M, Sánchez A, Segalés J. Exploratory study on the transcriptional profile of pigs subclinically infected with porcine circovirus type 2. Anim Biotechnol. 2009;20(3):96-109

Hesson LB, Dunwell TL, Cooper WN, Catchpoole D, Brini AT, Chiaramonte R, Griffiths M, Chalmers AD, Maher ER, Latif F. The novel RASSF6 and RASSF10 candidate tumour suppressor genes are frequently epigenetically inactivated in childhood leukaemias. Mol Cancer. 2009 Jul 1;8:42

Huang KH, Huang SF, Chen IH, Liao CT, Wang HM, Hsieh LL. Methylation of RASSF1A, RASSF2A, and HIN-1 is associated with poor outcome after radiotherapy, but not surgery, in oral squamous cell carcinoma. Clin Cancer Res. 2009 Jun 15;15(12):4174-80

Kashuba VI, Pavlova TV, Grigorieva EV, Kutsenko A, Yenamandra SP, Li J, Wang F, Protopopov AI, Zabarovska VI, Senchenko V, Haraldson K, Eshchenko T, Kobliakova J, Vorontsova O, Kuzmin I, Braga E, Blinov VM, Kisselev LL, Zeng YX, Ernberg I, Lerman MI, Klein G, Zabarovsky ER. High mutability of the tumor suppressor genes RASSF1 and RBSP3 (CTDSPL) in cancer. PLoS One. 2009 May 29;4(5):e5231

Kumari G, Mahalingam S. Extracellular signal-regulated kinase 2 (ERK-2) mediated phosphorylation regulates nucleo-cytoplasmic shuttling and cell growth control of Ras-associated tumor suppressor protein, RASSF2. Exp Cell Res. 2009 Oct 1;315(16):2775-90

Li Z, Luo RT, Mi S, Sun M, Chen P, Bao J, Neilly MB, Jayathilaka N, Johnson DS, Wang L, Lavau C, Zhang Y, Tseng C, Zhang X, Wang J, Yu J, Yang H, Wang SM, Rowley JD, Chen J, Thirman MJ. Consistent deregulation of gene expression between human and murine MLL rearrangement leukemias. Cancer Res. 2009 Feb 1;69(3):1109-16

Payne SR, Serth J, Schostak M, Kamradt J, Strauss A, Thelen P, Model F, Day JK, Liebenberg V, Morotti A, Yamamura S, Lograsso J, Sledziewski A, Semjonow A. DNA methylation biomarkers of prostate cancer: confirmation of candidates and evidence urine is the most sensitive body fluid for non-invasive detection. Prostate. 2009 Sep 1;69(12):1257-69

This article should be referenced as such:

Hesson LB, Latif F. RASSF2 (Ras association (RalGDS/AF-6) domain family member 2). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):652-661.

Page 42: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 662

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

SEMA3B (sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3B) Munmi Bhattacharyya, Ranjan Tamuli

Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati-781 039, Assam, India

(MB, RT)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/SEMA3BID42252ch3p21.html DOI: 10.4267/2042/44799

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: FLJ34863; LUCA-1; SEMA5; SEMAA;

SemA; semaV

HGNC (Hugo): SEMA3B

Location: 3p21.31

DNA/RNA

Note

SEMA3B was first discovered as a secreted member of

the semaphorin/collapsing family (contains a highly

conserved semaphorin domain) and it has a role in

axonal guidance.

Description

DNA size 11.53kb; mRNA size 9534 bp 18 exons.

Protein

Description

749 amino acids; region 1-24 (24) is a signal peptide,

30-513 (484) is the sema domain and 573-659 (87) is

the Ig-like C2-type domain.

Isoforms: two isoforms have been identified.

- Isoform 1 (identifier: Q13214-1): this isoform has

been chosen as the 'canonical' sequence.

- Isoform 2 (identifier: Q13214-2): the sequence of this

isoform differs from the 'canonical' sequence, amino

acid residues from 332-332 are missing.

Expression

It is expressed abundantly, but expressed differentially

in neural and non-neural tissues.

Localisation

Secreted.

Function

SEMA3B belongs to the semaphorin/collapsing group

of family (contains a highly conserved 749 amino acid

semaphoring domain at NH2-terminal). SEMA3B

involves in diverse processes such as immune

modulation, organogenesis, neuronal apoptosis and

drug resistance. SEM3B also plays a critical role in

axonal guidance during neuronal development.

SEMA3B can act as a tumour suppressor by inducing

apoptosis either by its expression in tumour cells or

when applied as a soluble ligand. SEM3B induced

apoptosis is associated with increase in cytochrome c

release and caspase-3 cleavage, as well as increased

phosphorylation of several proapoptotic proteins,

including glycogen synthase kinase-3beta, FKHR and

Page 43: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

SEMA3B (sema domain, immunoglobulin domain (Ig), short basic domain, secreted, Bhattacharyya M, Tamuli R (semaphorin) 3B)

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 663

MDM-2. The common method of inactivation of

SEMA3B is by allelic loss and gene inactivation via

promoter methylation and consequently, expression

level of SEM3B is reduced in tumor cells.

Homology

The percent identity below represents identity of

SEMA3B over an aligned region in UniGene.

Pan troglodytes 98.54%, Bos taurus 90.24%, Rattus

norvegicus 89.19%, Canis lupus familiaris 88.72%,

Mus musculus 88.65%.

Mutations

Note

A missense mutation in SEMA3B is reported in

African-American and Latino-American population.

Implicated in

Gallbladder carcinoma (GBC)

Note

SEMA3B believes to play a role in gallbladder

carcinoma (GBC), which is a highly malignant

neoplasm in the Chilean females. A very high

frequency (46/50, 92%) of abnormal promoter

methylation that causes epigenetic inactivation of

SEMA3B and the loss of heterozygosity at 3p21.3

(14/32, 44%) region (that contains SEMA3B gene) was

detected among the Chilean females with GBC.

Therefore, SEMA3B gene alterations may play a role

in GBC pathogenesis via a two-hit mechanism,

including allelic loss and abnormal promoter

methylation.

Nasopharyngeal carcinoma

Note

SEMA3B is associated with nasopharyngeal carcinoma

(NPC), as evident from both loss of heterozygosity

analysis and functional studies. 21 primary NPC tumors

and 2 NPC cell lines (CNE2 and SUNE1) screened for

mutations by PCR-sequencing and two missense

polymorphisms including Thr415Ile and lle242Met

were found in SEMA3B. For the Thr415Ile

polymorphism, the Ile allele type which leads to

SEMA3B function defects was predominant in NPC

with the allele frequency of 64% (27/42). SEMA3B

mRNA is expressed in non-neoplastic nasopharyngeal

epithelia, but found absent or down-regulated in 76%

(16/21) of primary NPC tumors. Thus, high frequency

of SEMA3B expression alterations suggests that the

inactivation of this gene was strongly associated with

NPC.

Neuroblastoma

Note

In neuroblastoma, significantly higher percentage of

methylated CpG sites in the SEMA3B promoter was

detected in tumors exhibiting 3p loss (95%), relative to

tumors without loss (52%), suggesting a two-hit

mechanism of allele inactivation. Additionally, low

levels of SEMA3B expression were also seen in tumors

with unmethylated SEMA3B promoters (n = 4).

However, SEMA3B was upregulated in the SK-N-BE

neuroblastoma cell line following induction of

differentiation with retinoic acid and interestingly,

higher levels of SEMA3B expression was found in

differentiated tumors with favorable histopathology (n

= 19) than in tumors with unfavorable histology (n =

22). The association of SEMA3B expression with

neuroblastoma differentiation suggests that this TSG

may play a role in neuroblastoma pathobiology and

SEMA3B expression profile suggests that

transcriptional regulation of this locus is complex.

Colorectal carcinoma

Disease

SEMA3B was also found frequently downregulated in

colorectal cancer, which suggests that SEMA3B is

involved in the suppression of colon tumor growth.

However, the molecular mechanism through which

SEM3B suppresses colorectal cancer is not clear.

Breast cancer

Disease

Expression of SEMA3B induces apoptosis in breast

cancer cells. SEMA3B induces apoptosis through the

neuropilin-1 (Np-1) receptor by inactivating the Akt

signaling pathway.

Ovarian cancer

Note

Decreased expression of SEMA3B and loss of

heterozygosity (LOH) at SEMA gene loci also account

for ovarian cancer progression. Patients with a high

vascular endothelial growth factor/SEMA

(VEGF/SEMA) ratio showed poor survival than those

with a low VEGF/SEMA ratio.

Lung cancer

Note

A single nucleotide alteration in the SEMA3B leads to

amino acid substitution T415I and this variant protein

has a reduced ability to act as a tumour suppressor. Thr

to Ile substitution alters the structure of protein by

altering its conformation and affects binding of

SEMA3B with neuropilin receptors-1 (NRP-1) and

NRP-2. The variant Ile allele occurs at an allele

frequency of 0.18 in African-American and 0.39 in

Latino-American population.

References Püschel AW, Adams RH, Betz H. Murine semaphorin D/collapsin is a member of a diverse gene family and creates domains inhibitory for axonal extension. Neuron. 1995 May;14(5):941-8

Sekido Y, Bader S, Latif F, Chen JY, Duh FM, Wei MH, Albanesi JP, Lee CC, Lerman MI, Minna JD. Human

Page 44: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

SEMA3B (sema domain, immunoglobulin domain (Ig), short basic domain, secreted, Bhattacharyya M, Tamuli R (semaphorin) 3B)

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 664

semaphorins A(V) and IV reside in the 3p21.3 small cell lung cancer deletion region and demonstrate distinct expression patterns. Proc Natl Acad Sci U S A. 1996 Apr 30;93(9):4120-5

Lerman MI, Minna JD. The 630-kb lung cancer homozygous deletion region on human chromosome 3p21.3: identification and evaluation of the resident candidate tumor suppressor genes. The International Lung Cancer Chromosome 3p21.3 Tumor Suppressor Gene Consortium. Cancer Res. 2000 Nov 1;60(21):6116-33

Tse C, Xiang RH, Bracht T, Naylor SL. Human Semaphorin 3B (SEMA3B) located at chromosome 3p21.3 suppresses tumor formation in an adenocarcinoma cell line. Cancer Res. 2002 Jan 15;62(2):542-6

Kuroki T, Trapasso F, Yendamuri S, Matsuyama A, Alder H, Williams NN, Kaiser LR, Croce CM. Allelic loss on chromosome 3p21.3 and promoter hypermethylation of semaphorin 3B in non-small cell lung cancer. Cancer Res. 2003 Jun 15;63(12):3352-5

Liu XQ, Sun M, Chen HK, Li JX, Pan ZG, Long QX, Wang XZ, Zeng YX. [Mutation and expression of SEMA3B and SEMA3F gene in nasopharyngeal carcinoma]. Ai Zheng. 2003 Jan;22(1):16-20

Castro-Rivera E, Ran S, Thorpe P, Minna JD. Semaphorin 3B (SEMA3B) induces apoptosis in lung and breast cancer, whereas VEGF165 antagonizes this effect. Proc Natl Acad Sci U S A. 2004 Aug 3;101(31):11432-7

Marsit CJ, Wiencke JK, Liu M, Kelsey KT. The race associated allele of Semaphorin 3B (SEMA3B) T415I and its role in lung cancer in African-Americans and Latino-Americans. Carcinogenesis. 2005 Aug;26(8):1446-9

Osada R, Horiuchi A, Kikuchi N, Ohira S, Ota M, Katsuyama Y, Konishi I. Expression of semaphorins, vascular endothelial

growth factor, and their common receptor neuropilins and alleic loss of semaphorin locus in epithelial ovarian neoplasms: increased ratio of vascular endothelial growth factor to semaphorin is a poor prognostic factor in ovarian carcinomas. Hum Pathol. 2006 Nov;37(11):1414-25

Nair PN, McArdle L, Cornell J, Cohn SL, Stallings RL. High-resolution analysis of 3p deletion in neuroblastoma and differential methylation of the SEMA3B tumor suppressor gene. Cancer Genet Cytogenet. 2007 Apr 15;174(2):100-10

Riquelme E, Tang M, Baez S, Diaz A, Pruyas M, Wistuba II, Corvalan A. Frequent epigenetic inactivation of chromosome 3p candidate tumor suppressor genes in gallbladder carcinoma. Cancer Lett. 2007 May 18;250(1):100-6

Castro-Rivera E, Ran S, Brekken RA, Minna JD. Semaphorin 3B inhibits the phosphatidylinositol 3-kinase/Akt pathway through neuropilin-1 in lung and breast cancer cells. Cancer Res. 2008 Oct 15;68(20):8295-303

Rolny C, Capparuccia L, Casazza A, Mazzone M, Vallario A, Cignetti A, Medico E, Carmeliet P, Comoglio PM, Tamagnone L. The tumor suppressor semaphorin 3B triggers a prometastatic program mediated by interleukin 8 and the tumor microenvironment. J Exp Med. 2008 May 12;205(5):1155-71

Pronina IV, Loginov VI, Prasolov VS, Klimov EA, Khodyrev DS, Kazubskaia TP, Gar'kavtseva RF, Sulimova GE, Braga EA. [Alteration of SEMA3B gene expression levels in epithelial tumors]. Mol Biol (Mosk). 2009 May-Jun;43(3):439-45

This article should be referenced as such:

Bhattacharyya M, Tamuli R. SEMA3B (sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3B). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):662-664.

Page 45: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 665

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15) Gui-Li Yang, Jian-Wei Qi, Zhi-Song Zhang, Lu-Yuan Li

Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA (LYL);

College of Pharmacy and College of Life Sciences, Nankai University, 94 Wei Jin Road, 300071 Tianjin,

China (LYL, GLY, JWQ, ZSZ)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/TNFSF15ID42638ch9q32.html DOI: 10.4267/2042/44800

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: MGC129934; MGC129935; TL1;

TL1A; VEGI; VEGI192A

HGNC (Hugo): TNFSF15

Location: 9q32

Local order: TNFSF15 gene at 9q32, near the CD30L

gene at 9q33.

DNA/RNA

Description

The human VEGI gene spans about 17 kb and consists

of four exons.

Transcription

The size of VEGI mRNA is approximately 6.5 kb.

Boxes with roman numerals above represent exons and horizontal lines represent intronic sequence. The putative transcription start site is indicated by a double arrowhead. R denotes the 5' untranslated sequence unique to each respective transcript, and stippled boxe represents the common 3' untranslated region.

Figure A. All three isoforms.

Page 46: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15) Yang GL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 666

Figure B. A ribbon diagram of the TL1A trimer. (Jin et al. BBRC 364:1, 2007).

It is unusual for a human gene of 6.5 kb to contain only

a small open reading frame of 522 nucleotides.

Multiple VEGI transcripts generated by the use of

cryptic splice sites and alternate exons.

Pseudogene

Not known.

Protein

Description

Hydrophobicity analysis of VEGI predicts a 13 amino

acid hydrophobic region that follows the amino

terminal segment of 12 amino acids, suggesting a

structure characteristic of a type II transmembrane

protein, with residues 26-174 constituting an

extracellular domain analogous to domains found in

other TNF family members.

VEGI isoforms exhibit a carboxyl terminal domain of

151 amino acid residues, which is encoded by part of

the fourth exon, termed IVb. The initially characterized

VEGI isoform, designated VEGI-174, is encoded by

the fourth exon (parts IVa and IVb) alone, which

includes both the putative transmembrane domain and

the conserved extracellular domain. There are two

additional endothelial-specific transcripts of 7.5 and 2.0

kb, which encode peptides of 251 (VEGI-251) and 192

(VEGI-192) residues, respectively. The VEGI-251 and

-192 isoforms differ in their amino terminal regions,

but share the conserved 151-amino acid residue

carboxy terminal domain. VEGI-251 possesses a

putative secretory signal peptide and its overexpression

causes apoptosis of endothelial cells and inhibition of

tumor growth.

Expression

VEGI is specifically expressed in endothelial cells.

Analysis of total RNA preparations from many cell

lines and primary cell cultures by Northern blot

analysis confirmed the specificity of VEGI expression,

with only HUVEC and human venous endothelial cells

demonstrating detectable levels of expression. Using

multiple tissue Northern blots, the VEGI transcript was

found in many adult human tissues, including placenta,

lung, skeletal muscle, kidney, pancreas, spleen,

prostate, small intestine, and colon, suggesting that the

gene product may play a role in the function of a

normal vasculature.The failure to detect the transcripts

of this new gene in some of the human tissues probably

is due to relatively small proportion of endothelial cells

in these tissues. Using isoform-specific probes, we

have determined that the distribution profiles of VEGI

isoforms in human organs and tissues appear to be

different. The 7.5 kb transcript encoding VEGI-251

was expressed at high levels in the placenta, kidney,

lung and liver, whereas the 2 kb transcript

corresponding to VEGI-174 was observed in liver,

kidney, skeletal muscle and heart. VEGI-174 mRNA

was more abundant in heart, skeletal muscle, pancreas,

adrenal gland, and liver, while VEGI-251 was more

abundant in fetal kidney and fetal lung. Overlapping

expression of VEGI-251 and VEGI-174 mRNA was

detected in prostate, salivary gland and placenta,

whereas VEGI-192 mRNA was not readily detected by

Northern blot. These expression patterns suggest the

possibility of tissue or developmentally specific

functions for VEGI isoforms.

Page 47: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15) Yang GL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 667

Amino acid sequence alignment of three VEGI isoforms. The putative hydrophobic regions of VEGI-251 and VEGI-174 are underlined. Asterisk denotes the start of shared sequences for all three isoforms.

Alternatively, this expression pattern also supports the

view that one VEGI isoform is the functional cytokine,

while the others act in regulatory roles to modulate the

activity of the active isoform. In this case, it is possible

that the non-functional isoforms do not exist at the

protein level. VEGI isoform expression has also been

examined in cultured cells by RNase protection assay.

All three known VEGI isoforms were detected in

human endothelial cells, including coronary artery

endothelial (HCAE), HUVE cells, and human

microvascular endothelial (HMVE) cells. Very low

levels are sometimes detected in adult bovine aortic

endothelial (ABAE) cells. Little VEGI expression was

detectable in human coronary artery smooth muscle

(CASM) and mouse endothelioma bEND.3 cells. More

than one isoform is detectable simultaneously, with

VEGI-251 being the most abundant. The expression of

this protein is inducible by TNF and IL-1 alpha, but not

by gamma-interferon.

Localisation

Endothelial cells and monocytes. However, VEGI was

not expressed in either B or T cells.

Function

VEGI is an endogenous inhibitor of angiogenesis

produced largely by vascular endothelial cells and

exerts a specific inhibitory activity on the proliferation

of endothelial cells. VEGI enforces growth arrest of

endothelial cells in G0 and early G1 phases of the cell

cycle but induces apoptosis in proliferating endothelial

cells. The MAPKs p38 and jun N-terminal kinase

(JNK) are required for VEGI-mediated endothelial

inhibition. Engineered overexpression of secreted

VEGI by cancer cells or systemic administration of

recombinant VEGI to tumor-bearing mice inhibits

tumor growth in numerous tumor models. Recent

studies show that VEGI helps modulate the immune

system by activating T cells and stimulating dendritic

cell maturation, suggesting that VEGI is directly

involved in modulating the interaction between the

endothelium and the immune system. Recombinant

VEGI has an inhibitory activity on mouse bone

marrow-derived EPCs in culture, preventing their

differentiation toward endothelial cells.

Interaction of TL1A with DR3 promotes T cell

expansion during an immune response (Migone et al.,

2002).

Homology

VEGI exhibits 20-30% sequence homology to human

TNF-alpha, TNF-beta, and the Fas ligand, similar to

that among other TNF family members.

Implicated in

Colon carcinoma

Note

Local production of a secreted form of VEGI via gene

transfer caused complete suppression of the growth of

MC-38 murine colon cancers in syngeneic

C57BL/6mice. Histological examination showed

marked reduction of vascularization in MC-38 tumors

that expressed soluble but not membrane-bound VEGI

or were transfected with control vector. The

conditioned media from soluble VEGI-expressing cells

showed marked inhibitory effect on in vitro

proliferation of adult bovine aortic endothelial cells.

Page 48: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15) Yang GL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 668

Breast cancer

Note

The anticancer potential of VEGI was examined in a

breast cancer xenograft tumor model in which the

cancer cells were co-injected with Chinese hamster

ovary cells overexpressing a secreted form of the

protein. The co-injection resulted in potent inhibition of

xenograft tumor growth. Our findings are consistent

with the view that VEGI is an endothelial cell-specific

negative regulator of angiogenesis.

Mucosal vaccine adjuvant

Note

Kayamuro et al., (2009) reported that TL1A induced

the strongest immune response and augmented OVA-

specific IgG and IgA responses in serum and mucosal

compartments, respectively. The OVA-specific

immune response of TL1A was characterized by high

levels of serum IgG1 and increased production of IL-4

and IL-5 from splenocytes of immunized mice,

suggesting that TL1A might induce Th2-type

responses. These findings indicate that TL1A has the

most potential as a mucosal adjuvant among the TNFS

cytokines.

Inflammatory bowel disease

Note

Bamias et al., (2003) provided evidence that the novel

cytokine TL1A may play an important role in a Th1-

mediated disease such as Crohn's disease. Takedatsu et

al., (2008) revealed that TL1A is an important

modulator in the development of chronic mucosal

inflammation by enhancing T(H)1 and T(H)17 effector

functions. The central role of TL1A represents an

attractive, novel therapeutic target for the treatment of

Crohn's disease patients.

Inflammatory arthritis

Note

Bull et al., (2008) demonstrated that the DR3-TL1A

pathway regulates joint destruction in two murine

models of arthritis and represents a potential novel

target for therapeutic intervention in inflammatory joint

disease. Bamias et al., (2008) concluded that TL1A

may serve as an inflammatory marker in rheumatoid

arthritis. Interactions between TL1A and its receptors

may be important in the pathogenesis of rheumatoid

arthritis.

Renal inflammation and injury

Note

Al-Lamki et al., (2008) suggested that TL1A may

contribute to renal inflammation and injury through

DR3-mediated activation of NF-kappaB and caspase-3,

respectively, but that an unidentified receptor may

mediate the NF-kappaB-independent induction of

TNFR2 in tubular epithelial cells.

References Tan KB, Harrop J, Reddy M, Young P, Terrett J, Emery J, Moore G, Truneh A. Characterization of a novel TNF-like ligand and recently described TNF ligand and TNF receptor superfamily genes and their constitutive and inducible expression in hematopoietic and non-hematopoietic cells. Gene. 1997 Dec 19;204(1-2):35-46

Yue TL, Ni J, Romanic AM, Gu JL, Keller P, Wang C, Kumar S, Yu GL, Hart TK, Wang X, Xia Z, DeWolf WE Jr, Feuerstein GZ. TL1, a novel tumor necrosis factor-like cytokine, induces apoptosis in endothelial cells. Involvement of activation of stress protein kinases (stress-activated protein kinase and p38 mitogen-activated protein kinase) and caspase-3-like protease. J Biol Chem. 1999 Jan 15;274(3):1479-86

Zhai Y, Ni J, Jiang GW, Lu J, Xing L, Lincoln C, Carter KC, Janat F, Kozak D, Xu S, Rojas L, Aggarwal BB, Ruben S, Li LY, Gentz R, Yu GL. VEGI, a novel cytokine of the tumor necrosis factor family, is an angiogenesis inhibitor that suppresses the growth of colon carcinomas in vivo. FASEB J. 1999 Jan;13(1):181-9

Zhai Y, Yu J, Iruela-Arispe L, Huang WQ, Wang Z, Hayes AJ, Lu J, Jiang G, Rojas L, Lippman ME, Ni J, Yu GL, Li LY. Inhibition of angiogenesis and breast cancer xenograft tumor growth by VEGI, a novel cytokine of the TNF superfamily. Int J Cancer. 1999 Jul 2;82(1):131-6

Yu J, Tian S, Metheny-Barlow L, Chew LJ, Hayes AJ, Pan H, Yu GL, Li LY. Modulation of endothelial cell growth arrest and apoptosis by vascular endothelial growth inhibitor. Circ Res. 2001 Dec 7;89(12):1161-7

Chew LJ, Pan H, Yu J, Tian S, Huang WQ, Zhang JY, Pang S, Li LY. A novel secreted splice variant of vascular endothelial cell growth inhibitor. FASEB J. 2002 May;16(7):742-4

Migone TS, Zhang J, Luo X, Zhuang L, Chen C, Hu B, Hong JS, Perry JW, Chen SF, Zhou JX, Cho YH, Ullrich S, Kanakaraj P, Carrell J, Boyd E, Olsen HS, Hu G, Pukac L, Liu D, Ni J, Kim S, Gentz R, Feng P, Moore PA, Ruben SM, Wei P. TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell costimulator. Immunity. 2002 Mar;16(3):479-92

Bamias G, Martin C 3rd, Marini M, Hoang S, Mishina M, Ross WG, Sachedina MA, Friel CM, Mize J, Bickston SJ, Pizarro TT, Wei P, Cominelli F. Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. J Immunol. 2003 Nov 1;171(9):4868-74

Zilberberg L, Shinkaruk S, Lequin O, Rousseau B, Hagedorn M, Costa F, Caronzolo D, Balke M, Canron X, Convert O, Laïn G, Gionnet K, Goncalvès M, Bayle M, Bello L, Chassaing G, Deleris G, Bikfalvi A. Structure and inhibitory effects on angiogenesis and tumor development of a new vascular endothelial growth inhibitor. J Biol Chem. 2003 Sep 12;278(37):35564-73

Asahara T, Kawamoto A. Endothelial progenitor cells for postnatal vasculogenesis. Am J Physiol Cell Physiol. 2004 Sep;287(3):C572-9

Hou W, Medynski D, Wu S, Lin X, Li LY. VEGI-192, a new isoform of TNFSF15, specifically eliminates tumor vascular endothelial cells and suppresses tumor growth. Clin Cancer Res. 2005 Aug 1;11(15):5595-602

Bamias G, Mishina M, Nyce M, Ross WG, Kollias G, Rivera-Nieves J, Pizarro TT, Cominelli F. Role of TL1A and its receptor DR3 in two models of chronic murine ileitis. Proc Natl Acad Sci U S A. 2006 May 30;103(22):8441-6

Page 49: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15) Yang GL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 669

Metheny-Barlow LJ, Li LY. Vascular endothelial growth inhibitor (VEGI), an endogenous negative regulator of angiogenesis. Semin Ophthalmol. 2006 Jan-Mar;21(1):49-58

Jin T, Kim S, Guo F, Howard A, Zhang YZ. Purification and crystallization of recombinant human TNF-like ligand TL1A. Cytokine. 2007 Nov;40(2):115-22

Tian F, Grimaldo S, Fujita M, Cutts J, Vujanovic NL, Li LY. The endothelial cell-produced antiangiogenic cytokine vascular endothelial growth inhibitor induces dendritic cell maturation. J Immunol. 2007 Sep 15;179(6):3742-51

Al-Lamki RS, Wang J, Tolkovsky AM, Bradley JA, Griffin JL, Thiru S, Wang EC, Bolton E, Min W, Moore P, Pober JS, Bradley JR. TL1A both promotes and protects from renal inflammation and injury. J Am Soc Nephrol. 2008 May;19(5):953-60

Bamias G, Siakavellas SI, Stamatelopoulos KS, Chryssochoou E, Papamichael C, Sfikakis PP. Circulating levels of TNF-like cytokine 1A (TL1A) and its decoy receptor 3 (DcR3) in rheumatoid arthritis. Clin Immunol. 2008 Nov;129(2):249-55

Bull MJ, Williams AS, Mecklenburgh Z, Calder CJ, Twohig JP, Elford C, Evans BA, Rowley TF, Slebioda TJ, Taraban VY, Al-Shamkhani A, Wang EC. The Death Receptor 3-TNF-like protein 1A pathway drives adverse bone pathology in inflammatory arthritis. J Exp Med. 2008 Oct 27;205(11):2457-64

Cai J, Wei R, Cheng J. Preparation and characterization of a novel chimeric protein VEGI-CTT in Escherichia coli. J Biomed Biotechnol. 2008;2008:564969

Gao D, Nolan DJ, Mellick AS, Bambino K, McDonnell K, Mittal V. Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science. 2008 Jan 11;319(5860):195-8

Takedatsu H, Michelsen KS, Wei B, Landers CJ, Thomas LS, Dhall D, Braun J, Targan SR. TL1A (TNFSF15) regulates the development of chronic colitis by modulating both T-helper 1 and T-helper 17 activation. Gastroenterology. 2008 Aug;135(2):552-67

Kayamuro H, Yoshioka Y, Abe Y, Katayama K, Yoshida T, Yamashita K, Yoshikawa T, Hiroi T, Itoh N, Kawai Y, Mayumi T, Kamada H, Tsunoda S, Tsutsumi Y. TNF superfamily member, TL1A, is a potential mucosal vaccine adjuvant. Biochem Biophys Res Commun. 2009 Jul 3;384(3):296-300

Tian F, Liang PH, Li LY. Inhibition of endothelial progenitor cell differentiation by VEGI. Blood. 2009 May 21;113(21):5352-60

This article should be referenced as such:

Yang GL, Qi JW, Zhang ZS, Li LY. TNFSF15 (tumor necrosis factor (ligand) superfamily, member 15). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):665-669.

Page 50: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 670

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

BAP1 (BRCA1 associated protein-1 (ubiquitin carboxy-terminal hydrolase)) Frédéric Guénard, Francine Durocher

Cancer Genomics Laboratory, Oncology and Molecular Endocrinology Research Centre, CRCHUL, CHUQ

and Laval University, Québec, G1V 4G2, Canada (FG, FD)

Published in Atlas Database: September 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/BAP1ID755ch3p21.html DOI: 10.4267/2042/44801

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: DKFZp686N04275; FLJ35406;

FLJ37180; HUCEP-13; KIAA0272; UCHL2; hucep-6

HGNC (Hugo): BAP1

Location : 3p21.1

DNA/RNA

Description

The gene spans 9.0 kb and is composed of 17 exons.

Transcription

Transcription start is 115 bp upstream of first ATG of

the BAP1 ORF.

Pseudogene

No pseudogene reported.

Protein

Description

Human BAP1 is 729 amino acids with a molecular

weight of 90 kDa. The amino-terminal 240 amino acids

show homology to ubiquitin C-terminal hydrolases

(UCH).

BAP1 also contains a region of extreme acidity (amino

acids 396 to 408), multiple potential phosphorylation

sites and N-linked glycosylation sites. The C-terminal

region contains two putative nuclear localization

signals.

BAP1 binds to the RING finger domain of BRCA1

through its carboxyl-terminal region (594-657 amino

acids). Domain comprised by residues 182-365 of

BAP1 interacts with the RING finger domain of

BARD1. Interaction of BAP1 with HCF-1 (host cell

factor 1; HCFC1) is dependent on the NHNY sequence

resembling the HCF-binding motif (HBM).

Expression

BAP1 is expressed in a variety of human adult tissues.

High expression was detected in testis, placenta and

ovary, with varying levels detected in other tissues.

Expression of BAP1 in normal human breast tissue was

also detected.

Analysis conducted in mice revealed that Bap1

expression is up-regulated in the breast during puberty,

pregnancy and as a result of parity.

Structure of BAP1. BAP1 is a 729 aa protein. UCH, Ubiquitin C-terminal hydrolase; HBM, HCF-binding motif (NHNY sequence); NLS, Nuclear localization signal.

Page 51: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

BAP1 (BRCA1 associated protein-1 (ubiquitin carboxy-terminal hydrolase)) Guénard F, Durocher F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 671

BAP1 mRNA level is significantly increased in

MCF10a cell line following genistein treatment, an

isoflavone found in soya and proposed to prevent breast

cancer.

Localisation

BAP1 is a nuclear-localized ubiquitin carboxy-terminal

hydrolase.

Function

BAP1 enhances BRCA1-mediated inhibition of breast

cancer cell growth and may serve as a

regulator/effector of BRCA1 growth

control/differentiation pathways. BAP1 interacts with

HCF-1, a transcriptional cofactor found in a number of

important regulatory complexes. Bap1 may help to

control cell proliferation by regulating HCF-1 protein

levels and by associating with genes involved in the

G1-S transition.

The BRCA1/BARD1 complex possess a dual E3

ubiquitin ligase activity, promotes its own

ubiquitination and targets other proteins. Although

BAP1 associates with BRCA1, it does not appear to

function in the deubiquitylation of the BRCA1/BARD1

complex. BAP1 inhibits the E3 ligase activity of

BRCA1/BARD1 by binding the RING finger domain

of BARD1 and possesses deubiquitination activity

toward ubiquitin chains catalyzed by BRCA1/BARD1.

BAP1 and BRCA1/BARD1 may coordinately regulate

ubiquitination during the DNA damage response and

the cell cycle, BAP1 being phosphorylated by ATM

and ATR in response to DNA damage and BAP1

inhibition causing S-phase retardation.

It was also proposed that specific regions and UCH

activity of BAP1 play an essential role in TCR.

Homology

The amino-terminal 240 amino acids show significant

homology to a class of thiol proteases, designated

UCH, which are implicated in the proteolytic

processing of ubiquitin.

Mutations

Note

The mutation of a residue predicted to disrupt the

helical nature of the extreme C-terminal region of

BAP1 abolishes the BAP1/BRCA1 interaction.

BAP1 can suppress tumorigenicity of lung cancer cells

in athymic nude mice.

Deubiquitinating activity and nuclear localization are

both required for BAP1-mediated tumor suppression.

Moreover, BAP1-mediated growth suppression is

independent of wild-type BRCA1.

Squamous-cell carcinomas and large-cell

undifferentiated carcinomas showed LOH for a 3p21-

22 locus.

Large rearrangements, deletions, and missense

mutations of the BAP1 locus have been found in lung

and sporadic breast tumors and in lung cancer cell

lines.

Implicated in

Breast cancer

Note

A study conducted on high-risk breast cancer families

from the French population revealed that the BAP1

gene does not appear to be commonly involved in high-

risk breast cancer predisposition. These results were

thereafter confirmed in a larger study conducted on

families with high risk of breast cancer from the French

Canadian population. These studies do not rule out the

possibility that BAP1 alleles might be associated with

moderate or low breast cancer risk.

Selected variations of the BAP1 gene were also

excluded as low penetrance risk alleles in sporadic

breast cancer carried from the Spanish population.

Medulloblastoma

Note

Medulloblastoma is a highly malignant tumor of the

cerebellum. This disease with poor prognosis occurs

mostly in children. A screen of cDNA libraries with

autologous sera to identify antigen-specific immune

responses associated with this agressive tumor type

pointed to the BAP1 gene as a possible target of

immune response.

Schizophrenia

Note

The BAP1 gene was excluded as a promizing candidate

gene for schizophrenia in a fine mapping association

study carried out on chromosome 3p, one of the regions

showing strong evidence of linkage with schizophrenia.

References Buchhagen DL, Qiu L, Etkind P. Homozygous deletion, rearrangement and hypermethylation implicate chromosome region 3p14.3-3p21.3 in sporadic breast-cancer development. Int J Cancer. 1994 May 15;57(4):473-9

Jensen DE, Proctor M, Marquis ST, Gardner HP, Ha SI, Chodosh LA, Ishov AM, Tommerup N, Vissing H, Sekido Y, Minna J, Borodovsky A, Schultz DC, Wilkinson KD, Maul GG, Barlev N, Berger SL, Prendergast GC, Rauscher FJ 3rd. BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene. 1998 Mar 5;16(9):1097-112

Jensen DE, Rauscher FJ 3rd. BAP1, a candidate tumor suppressor protein that interacts with BRCA1. Ann N Y Acad Sci. 1999;886:191-4

Hu JJ, Mohrenweiser HW, Bell DA, Leadon SA, Miller MS. Symposium overview: genetic polymorphisms in DNA repair and cancer risk. Toxicol Appl Pharmacol. 2002 Nov 15;185(1):64-73

Mallery DL, Vandenberg CJ, Hiom K. Activation of the E3 ligase function of the BRCA1/BARD1 complex by polyubiquitin chains. EMBO J. 2002 Dec 16;21(24):6755-62

Page 52: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

BAP1 (BRCA1 associated protein-1 (ubiquitin carboxy-terminal hydrolase)) Guénard F, Durocher F

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 672

Behrends U, Schneider I, Rössler S, Frauenknecht H, Golbeck A, Lechner B, Eigenstetter G, Zobywalski C, Müller-Weihrich S, Graubner U, Schmid I, Sackerer D, Späth M, Goetz C, Prantl F, Asmuss HP, Bise K, Mautner J. Novel tumor antigens identified by autologous antibody screening of childhood medulloblastoma cDNA libraries. Int J Cancer. 2003 Aug 20;106(2):244-51

Coupier I, Cousin PY, Hughes D, Legoix-Né P, Trehin A, Sinilnikova OM, Stoppa-Lyonnet D. BAP1 and breast cancer risk. Fam Cancer. 2005;4(4):273-7

Caëtano B, Le Corre L, Chalabi N, Delort L, Bignon YJ, Bernard-Gallon DJ. Soya phytonutrients act on a panel of genes implicated with BRCA1 and BRCA2 oncosuppressors in human breast cell lines. Br J Nutr. 2006 Feb;95(2):406-13

Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER 3rd, Hurov KE, Luo J, Bakalarski CE, Zhao Z, Solimini N, Lerenthal Y, Shiloh Y, Gygi SP, Elledge SJ. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 2007 May 25;316(5828):1160-6

Ventii KH, Devi NS, Friedrich KL, Chernova TA, Tighiouart M, Van Meir EG, Wilkinson KD. BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer Res. 2008 Sep 1;68(17):6953-62

Guénard F, Labrie Y, Ouellette G, Beauparlant CJ, Durocher F. Genetic sequence variations of BRCA1-interacting genes AURKA, BAP1, BARD1 and DHX9 in French Canadian families with high risk of breast cancer. J Hum Genet. 2009 Mar;54(3):152-61

Misaghi S, Ottosen S, Izrael-Tomasevic A, Arnott D, Lamkanfi M, Lee J, Liu J, O'Rourke K, Dixit VM, Wilson AC. Association of C-terminal ubiquitin hydrolase BRCA1-associated protein 1 with cell cycle regulator host cell factor 1. Mol Cell Biol. 2009 Apr;29(8):2181-92

Nishikawa H, Wu W, Koike A, Kojima R, Gomi H, Fukuda M, Ohta T. BRCA1-associated protein 1 interferes with BRCA1/BARD1 RING heterodimer activity. Cancer Res. 2009 Jan 1;69(1):111-9

Vega A, Salas A, Milne RL, Carracedo B, Ribas G, Ruibal A, de León AC, González-Hernández A, Benítez J, Carracedo A. Evaluating new candidate SNPs as low penetrance risk factors in sporadic breast cancer: a two-stage Spanish case-control study. Gynecol Oncol. 2009 Jan;112(1):210-4

So HC, Fong PY, Chen RY, Hui TC, Ng MY, Cherny SS, Mak WW, Cheung EF, Chan RC, Chen EY, Li T, Sham PC. Identification of neuroglycan C and interacting partners as potential susceptibility genes for schizophrenia in a Southern Chinese population. Am J Med Genet B Neuropsychiatr Genet. 2010 Jan 5;153B(1):103-13

This article should be referenced as such:

Guénard F, Durocher F. BAP1 (BRCA1 associated protein-1 (ubiquitin carboxy-terminal hydrolase)). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):670-672.

Page 53: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 673

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

CDA (Cytidine Deaminase) Yoshiro Saito

Division of Medicinal Safety Science, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku,

Tokyo 158-8501, Japan (YS)

Published in Atlas Database: September 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/CDAID998ch1p36.html DOI: 10.4267/2042/44802

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: CDD

HGNC (Hugo): CDA

Location: 1p36.12

Note: CDA catalyzes hydrolytic deamination of

cytidine and deoxycytidine into uridine and

deoxyuridine, respectively.

DNA/RNA

Description

The human CDA spans approximately 30 kB and

consists of 4 exons. No splice variant was reported.

Transcription

The full length CDA mRNA is 985 bp with an open

reading frame of 441 bp.

Pseudogene

No pseudogene was reported.

Protein

Note

X-ray crystal structures of CDA from Yeast (1R5T)

and Bacillus Subtilis (1JTK, 1UX0, 1UX1 and 1UWZ)

are publicized in the PDB.

Description

The human CDA protein consists of 146 amino acids

and has a molecular weight of 16,184. This is a soluble

cytoplasmic protein and it is involved in pyrimidine

salvaging.

Expression

Although the protein expression profile in tissues has

not been revealed, its mRNA expression determined by

Nothern blotting was observed in high levels in liver

and placenta, low in lung and kidney, but not in heart,

brain and muscle (Laliberte and Momparler, 1994).

High CDA activity was reported in liver and spleen,

and moderate in lung, kidney, large intestine mucosa

and colon mucosa (Ho, 1973).

Page 54: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CDA (Cytidine Deaminase) Saito Y

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 674

Localisation

This protein is localized in cytoplasm.

Function

CDA catalyzes hydrolytic deamination of cytidine and

deoxycytidine into uridine and deoxyuridine,

respectively. This protein also inactivate

chemotherapeutic nucleoside analogs 2,2-

difluorodeoxycytidine (gemcitabine) and cytosine

arabinoside (cytarabine, Ara-C).

Mutations

Germinal

Two nonsynonymous genetic varitions, 79A>C

(Lys27Gln) and 208G>A (Ala70Thr), have been found

in the human CDA gene (Yue et al., 2003). Ethnic

differences in the minor allele frequencies of these

variations have been reported. The 79A>C (Lys27Gln)

was found at 0.30-0.36 frequencies in Caucasians, at

0.20-0.21 in Japanese and at 0.04-0.10 in Africans

(Ueno et al., 2007). In contrast, the 208G>A

(Ala70Thr) was found at 0.13 in Africans and 0.04 in

Japanese, but not in Caucasians. Interestingly, the

208G>A (Ala70Thr) has not been detected in African-

Americans. The mutant protein with 70Thr was

reported to have remarkably reduced activities in vitro

(Yue et al., 2003) and in vivo (Sugiyama et al., 2007).

On the other hand, controvertial results on the effects of

activities have been obtained for 79A>C (Lys27Gln).

The recombinant enzyme with Gln27 retained its

catalytic activities for cytidine and ara-C as substrates

(Yue et al., 2003), while showing reduced activity with

increased Km value in the case of gemcitabine (Gilbert

et al., 2006). However, the minor allele of this SNP was

reported to be associated with higher enzymatic

activities for gemcitbine based on tests using lysates of

red blood cells taken from Caucasian cancer patients

(Giovannetti et al., 2008; Tibaldi et al., 2008). In line

with this, the minor allele was associated with

decreased response, shorter time to progression and

overall survival, and lower frequencies of grade 3 and 4

neutropenia in Caucasian non-small cell lung cancer

patients treated with gemcitabine and cisplatin (Tibaldi

et al., 2008).

Implicated in

Adverse reactions by anti-cancer drugs

Note

CDA is involved in the metabolic inactivation of anti-

cancer drug gemcitabine and cytosine arabinoside (ara-

C). CDA polymorphisms 208G>A (Ala70Thr) has been

associated with adverse reactions including neutropenia

by gemcitabine. Reduced clearance of gemcitabine and

plasma CDA activities significantly depended on the

number of minor allele 208A (70Thr) in 256 Japanese

patients with cancer (Sugiyama et al., 2007). This

polymorphism was also associated with increased

incidences of grade 3/4 neutropenia in the patients

coadministered with other anti-cancer drugs (Sugiyama

et al., 2007). Notably, one patient with homozygous

208A (70Thr) showed severe hematologic and

nonhematologic toxicities during chemotherapy with

gemcitabine and cisplatin, and had 1/5 value of

gemcitabine clearance and 12% of plasma CDA

activity compared to those of the patients without CDA

nonsynonymous polymorphisms (Yonemori et al.,

2005, Sugiyama et al., 2007). Among the other panels

of Japanese pancreatic cancer patients, three patients

encountered life-threatening toxicities after

chemotherapies including gemcitabine (Ueno et al.,

2009). Two of them had homozygous CDA 208A

(70Thr), and showed extremely low plasma CDA

activity and gemcitabine clearance. Together with the

previous one patient, homozygous 208A (70Thr) was

suggested to be a key factor causing gemcitabine-

induced severe adverse reactions in the Japanese (Ueno

et al., 2009). With regard to another nonsynonymous

polymorphism, the minor allele of CDA 79A>C

(Lys27Gln) was associated with decreased response,

shorter time to progression and overall survival, and

lower frequencies of grade 3 and 4 neutropenia in

Caucasian non-small cell lung cancer patients treated

with gemcitabine and cisplatin (Tibaldi et al., 2008).

Homozygous 79C (27Gln) was also associated with

increased postinduction treatment-related motality with

ara-C in patients with acute myeloid leukemia (Bhatla

et al., 2008).

Acute myeloid leukemia

Disease

CDA genetic polymorphisms (79A>C, Lys27Gln;

208G>A, Ala70Thr; 435T>C, silent) were not

associated with susceptibility to acute myeloid

leukemia in Chinese children (Yue et al., 2007).

Colorectal cancer

Note

Combination of the five gene expression levels (CDA,

MGC20553, BANK1, BCNP1 and MS4A1) in

peripheral white blood cells could be used as a

biomarker for diagnosis of colorectal cancer (Han et al.,

2008).

References Ho DH. Distribution of kinase and deaminase of 1-beta-D-arabinofuranosylcytosine in tissues of man and mouse. Cancer Res. 1973 Nov;33(11):2816-20

Laliberté J, Momparler RL. Human cytidine deaminase: purification of enzyme, cloning, and expression of its complementary DNA. Cancer Res. 1994 Oct 15;54(20):5401-7

Yue L, Saikawa Y, Ota K, Tanaka M, Nishimura R, Uehara T, Maeba H, Ito T, Sasaki T, Koizumi S. A functional single-nucleotide polymorphism in the human cytidine deaminase gene contributing to ara-C sensitivity. Pharmacogenetics. 2003 Jan;13(1):29-38

Page 55: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CDA (Cytidine Deaminase) Saito Y

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 675

Yonemori K, Ueno H, Okusaka T, Yamamoto N, Ikeda M, Saijo N, Yoshida T, Ishii H, Furuse J, Sugiyama E, Kim SR, Kikura-Hanajiri R, Hasegawa R, Saito Y, Ozawa S, Kaniwa N, Sawada J. Severe drug toxicity associated with a single-nucleotide polymorphism of the cytidine deaminase gene in a Japanese cancer patient treated with gemcitabine plus cisplatin. Clin Cancer Res. 2005 Apr 1;11(7):2620-4

Gilbert JA, Salavaggione OE, Ji Y, Pelleymounter LL, Eckloff BW, Wieben ED, Ames MM, Weinshilboum RM. Gemcitabine pharmacogenomics: cytidine deaminase and deoxycytidylate deaminase gene resequencing and functional genomics. Clin Cancer Res. 2006 Mar 15;12(6):1794-803

Sugiyama E, Kaniwa N, Kim SR, Kikura-Hanajiri R, Hasegawa R, Maekawa K, Saito Y, Ozawa S, Sawada J, Kamatani N, Furuse J, Ishii H, Yoshida T, Ueno H, Okusaka T, Saijo N. Pharmacokinetics of gemcitabine in Japanese cancer patients: the impact of a cytidine deaminase polymorphism. J Clin Oncol. 2007 Jan 1;25(1):32-42

Ueno H, Kiyosawa K, Kaniwa N. Pharmacogenomics of gemcitabine: can genetic studies lead to tailor-made therapy? Br J Cancer. 2007 Jul 16;97(2):145-51

Yue LJ, Chen XW, Li CR, Li CG, Shi HS, Zhang M. [Single-nucleotide polymorphisms of the cytidine deaminase gene in childhood with acute leukemia and normal Chinese children]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2007 Dec;24(6):699-702

Giovannetti E, Laan AC, Vasile E, Tibaldi C, Nannizzi S, Ricciardi S, Falcone A, Danesi R, Peters GJ. Correlation between cytidine deaminase genotype and gemcitabine

deamination in blood samples. Nucleosides Nucleotides Nucleic Acids. 2008 Jun;27(6):720-5

Han M, Liew CT, Zhang HW, Chao S, Zheng R, Yip KT, Song ZY, Li HM, Geng XP, Zhu LX, Lin JJ, Marshall KW, Liew CC. Novel blood-based, five-gene biomarker set for the detection of colorectal cancer. Clin Cancer Res. 2008 Jan 15;14(2):455-60

Tibaldi C, Giovannetti E, Vasile E, Mey V, Laan AC, Nannizzi S, Di Marsico R, Antonuzzo A, Orlandini C, Ricciardi S, Del Tacca M, Peters GJ, Falcone A, Danesi R. Correlation of CDA, ERCC1, and XPD polymorphisms with response and survival in gemcitabine/cisplatin-treated advanced non-small cell lung cancer patients. Clin Cancer Res. 2008 Mar 15;14(6):1797-803

Bhatla D, Gerbing RB, Alonzo TA, Conner H, Ross JA, Meshinchi S, Zhai X, Zamzow T, Mehta PA, Geiger H, Perentesis J, Davies SM. Cytidine deaminase genotype and toxicity of cytosine arabinoside therapy in children with acute myeloid leukemia. Br J Haematol. 2009 Feb;144(3):388-94

Ueno H, Kaniwa N, Okusaka T, Ikeda M, Morizane C, Kondo S, Sugiyama E, Kim SR, Hasegawa R, Saito Y, Yoshida T, Saijo N, Sawada J. Homozygous CDA*3 is a major cause of life-threatening toxicities in gemcitabine-treated Japanese cancer patients. Br J Cancer. 2009 Mar 24;100(6):870-3

This article should be referenced as such:

Saito Y. CDA (Cytidine Deaminase). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):673-675.

Page 56: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 676

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

CKS1B (CDC28 protein kinase regulatory subunit 1B) Yongyou Zhang

Case Western Reserve University, 2103 Cornell Rd, WRB-3101, Cleveland, Ohio 44106, USA (YZ)

Published in Atlas Database: September 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/CKS1BID40092ch1q21.html DOI: 10.4267/2042/44803

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: CKS-1; CKS1; PNAS-143; PNAS-16;

PNAS-18; ckshs1

HGNC (Hugo): CKS1B

Location: 1q21.3

DNA/RNA

Genomic organization of the CKS1B gene.

Description

Three exons, spans approximately 4.61 kb of genomic

DNA in the centromere-to-telomere orientation. The

translation initiation codon ATG is located in exon 1,

and the stop codon in exon 3.

Transcription

mRNA of approximately 1.8 kb. There are two

transcript variants for CKS1B gene. The variant 2 uses

a different splice site at the 3' end of the first exon

compared to variant 1. There is no evidence that variant

2 encodes a protein.

Pseudogene

4 processed, non-expressed, pseudogenes in human

genome.

Protein

Description

The open reading frame encodes a 79 amino acid

protein, with an estimated molecular weight of

approximately 9660 Da.

The side chains of residues of CKS1b in the binding sites for Cdk2, Skp2 and phosphorylated substrate are shown, with residues in the Cdk2-binding site in green, in the phosphate-binding site in blue, and the Skp2-binding site in red. (The structure figure modified from the origin paper: Markus A. Seeliger et al, Role of Conformational Heterogeneity in Domain Swapping and Adapter Function of the Cks Proteins. J. Biol. Chem., Vol. 280, Issue 34, 30448-30459, August 26, 2005).

Page 57: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CKS1B (CDC28 protein kinase regulatory subunit 1B) Zhang Y

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 677

Expression

Basical level expression in all mammalian cell and

aberrant expression in cancer cell.

Localisation

Cytoplasm and nucleus.

Function

CKS1B protein binds to the catalytic subunit of the

cyclin dependent kinases and is essential for their

biological function of cell cycle control.

Schematic of the regulation of cell cycle by the CKS1B. CKS1B associates with the p27kip1-Cdk/cyclin complex, induces the formation of the p27kip1-SCF ubiquitin ligase complex, triggers degradation of p27kip1, and signals cells to undergo the G1/S transition by releasing and activating the Cdk/cyclin A/E complexes. CycE/A, cyclin E or Cyclin A; Ub, ubiquitin.

Homology

The CKS1B proteins are evolutionary conserved.

Mammalian cells express two well-conserved

members, like the human CKS1B and CKS2 proteins.

The CKSB1B protein is highly conserved across

species.

Implicated in

Cancer

Note

The expression of CKS1B is elevaled in multiple

cancer, including breast cancer, lymphoma, myeloma,

colon cancer, prostate cancer, lung cancer, renal

carcinoma, oesophageal squamous cell carcinoma,

salivary cancer, serous ovarian cancer, bladder cancer,

urothelial carcinoma et al.

Prognosis

Overexpression of CKS1B is associated with poor

prognosis in multiple cancer, including myeloma,

breast cancer, lymphoma, renal carcinoma, ovarian

cancer et al.

Oncogenesis

Amplification and overexpression of CKS1B were

strongly associated with lymph node metastasis and

poor prognosis in breast, salivary cancer and

oesophageal squamous cell carcinoma. Generally,

CKS1B is an essential factor in facilitating Skp2-

dependent degradation of p27. In breast cancer cell,

overexpression of CKS1B may inhibit the apoptosis

through the MEK-Erk pathway. All of these suggest

that CKS1B alterations may have a significant

biological role in the tumorigenesis in different tissue

and the novel therapeutic strategy for cancer through

inhibiting the CKS1B activity. Therefore, disruption of

Skp2-CKS1B assembly or down-regulation of CKS1B

expression may be used for cancer therapy.

References Richardson HE, Stueland CS, Thomas J, Russell P, Reed SI. Human cDNAs encoding homologs of the small p34Cdc28/Cdc2-associated protein of Saccharomyces cerevisiae and Schizosaccharomyces pombe. Genes Dev. 1990 Aug;4(8):1332-44

Bourne Y, Watson MH, Hickey MJ, Holmes W, Rocque W, Reed SI, Tainer JA. Crystal structure and mutational analysis of the human CDK2 kinase complex with cell cycle-regulatory protein CksHs1. Cell. 1996 Mar 22;84(6):863-74

Demetrick DJ, Zhang H, Beach DH. Chromosomal mapping of the human genes CKS1 to 8q21 and CKS2 to 9q22. Cytogenet Cell Genet. 1996;73(3):250-4

Spruck C, Strohmaier H, Watson M, Smith AP, Ryan A, Krek TW, Reed SI. A CDK-independent function of mammalian Cks1: targeting of SCF(Skp2) to the CDK inhibitor p27Kip1. Mol Cell. 2001 Mar;7(3):639-50

Morris MC, Kaiser P, Rudyak S, Baskerville C, Watson MH, Reed SI. Cks1-dependent proteasome recruitment and activation of CDC20 transcription in budding yeast. Nature. 2003 Jun 26;423(6943):1009-13

Bashir T, Dorrello NV, Amador V, Guardavaccaro D, Pagano M. Control of the SCF(Skp2-Cks1) ubiquitin ligase by the APC/C(Cdh1) ubiquitin ligase. Nature. 2004 Mar 11;428(6979):190-3

Kitajima S, Kudo Y, Ogawa I, Bashir T, Kitagawa M, Miyauchi M, Pagano M, Takata T. Role of Cks1 overexpression in oral squamous cell carcinomas: cooperation with Skp2 in promoting p27 degradation. Am J Pathol. 2004 Dec;165(6):2147-55

Zhang Y, Lin Y, Bowles C, Wang F. Direct cell cycle regulation by the fibroblast growth factor receptor (FGFR) kinase through phosphorylation-dependent release of Cks1 from FGFR substrate 2. J Biol Chem. 2004 Dec 31;279(53):55348-54

Shapira M, Ben-Izhak O, Linn S, Futerman B, Minkov I, Hershko DD. The prognostic impact of the ubiquitin ligase subunits Skp2 and Cks1 in colorectal carcinoma. Cancer. 2005 Apr 1;103(7):1336-46

Slotky M, Shapira M, Ben-Izhak O, Linn S, Futerman B, Tsalic M, Hershko DD. The expression of the ubiquitin ligase subunit Cks1 in human breast cancer. Breast Cancer Res. 2005;7(5):R737-44

Ouellet V, Guyot MC, Le Page C, Filali-Mouhim A, Lussier C, Tonin PN, Provencher DM, Mes-Masson AM. Tissue array analysis of expression microarray candidates identifies markers associated with tumor grade and outcome in serous epithelial ovarian cancer. Int J Cancer. 2006 Aug 1;119(3):599-607

Kawakami K, Enokida H, Tachiwada T, Nishiyama K, Seki N, Nakagawa M. Increased SKP2 and CKS1 gene expression contributes to the progression of human urothelial carcinoma. J Urol. 2007 Jul;178(1):301-7

Westbrook L, Manuvakhova M, Kern FG, Estes NR 2nd, Ramanathan HN, Thottassery JV. Cks1 regulates cdk1

Page 58: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

CKS1B (CDC28 protein kinase regulatory subunit 1B) Zhang Y

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 678

expression: a novel role during mitotic entry in breast cancer cells. Cancer Res. 2007 Dec 1;67(23):11393-401

Krishnan A, Hariharan R, Nair SA, Pillai MR. Fluoxetine mediates G0/G1 arrest by inducing functional inhibition of cyclin dependent kinase subunit (CKS)1. Biochem Pharmacol. 2008 May 15;75(10):1924-34

Lan Y, Zhang Y, Wang J, Lin C, Ittmann MM, Wang F. Aberrant expression of Cks1 and Cks2 contributes to prostate tumorigenesis by promoting proliferation and inhibiting programmed cell death. Int J Cancer. 2008 Aug 1;123(3):543-51

Liu Z, Fu Q, Lv J, Wang F, Ding K. Prognostic implication of p27Kip1, Skp2 and Cks1 expression in renal cell carcinoma: a tissue microarray study. J Exp Clin Cancer Res. 2008 Oct 15;27:51

Martinsson-Ahlzén HS, Liberal V, Grünenfelder B, Chaves SR, Spruck CH, Reed SI. Cyclin-dependent kinase-associated proteins Cks1 and Cks2 are essential during early embryogenesis and for cell cycle progression in somatic cells. Mol Cell Biol. 2008 Sep;28(18):5698-709

Nagler RM, Ben-Izhak O, Ostrovsky D, Golz A, Hershko DD. The expression and prognostic significance of Cks1 in salivary cancer. Cancer Invest. 2009 Jun;27(5):512-20

Wang XC, Tian J, Tian LL, Wu HL, Meng AM, Ma TH, Xiao J, Xiao XL, Li CH. Role of Cks1 amplification and overexpression in breast cancer. Biochem Biophys Res Commun. 2009 Feb 20;379(4):1107-13

Calvisi DF, Pinna F, Ladu S, Muroni MR, Frau M, Demartis I, Tomasi ML, Sini M, Simile MM, Seddaiu MA, Feo F, Pascale RM. The degradation of cell cycle regulators by SKP2/CKS1 ubiquitin ligase is genetically controlled in rodent liver cancer and contributes to determine the susceptibility to the disease. Int J Cancer. 2010 Mar 1;126(5):1275-81

This article should be referenced as such:

Zhang Y. CKS1B (CDC28 protein kinase regulatory subunit 1B). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):676-678.

Page 59: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 679

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

COL16A1 (collagen, type XVI, alpha 1) Susanne Grässel, Sabine Ratzinger

Orthopaedic Surgery, University of Regensburg, Abt. Experimentelle Orthopadie, ZMB im BioPark 1, Josef-

Engert-Strasse 9, D-93053 Regensburg, Germany (SG); Centre for Medical Biotechnology, BioPark 1,

Regensburg, Germany (SR)

Published in Atlas Database: September 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/COL16A1ID44542ch1p35.html DOI: 10.4267/2042/44804

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: 447AA; FP1572

HGNC (Hugo): COL16A1

Location: 1p35.2

DNA/RNA

Description

In 1992 the cDNA sequence of COL16A1 has been

discovered in a screening for collagen-like sequences in

cDNA banks of a human fibroblast cell line and human

placenta tissue. Two laboratories published

independently the human COL16A1 cDNA sequence

(Pan et al., 1992; Yamaguchi et al., 1992). The coding

sequence of authentic collagen type XVI comprises of

1604 amino acids including a 21 amino acid signal

peptide, whereas the recombinant version of collagen

type XVI contains 1597 amino acids (Kassner et al.,

2004). The nucleotide sequences published by Pan et

al., and Yamaguchi et al., were completed by Kassner

et al., with respect to a missing codon for one amino

acid (Kassner et al., 2004). Two predicted

imperfections in the collagenous region could not be

confirmed (unpublished data).

Transcription

The cDNA of 5.4 kb comprises a 4809 bp coding

sequence, framed by non-translated parts, including a

425 bp 3'-non-coding sequence which contains

polyadenylating signals. COL16A1 has been localized

to chromosome 1, 1p35-p34 (Pan et al., 1992). No

splice variants have been described up to

now. COL16A1 gene expression and transcription

varies in various phases of cell growth in cultured skin

fibroblasts. Gene expression was increased in

stationary phases (G0/G1) of cell growth when cell

proliferation was inhibited by serum deprivation or

suspension arrest (Tajima et al., 2000). Transcription

activity of the COL16A1 gene appears to be

mechanosensitive. It is downregulated in HCS2/8

human chondrosarcoma cells after application of

continuous hydrostatic pressure (Sironen et al., 2002).

Pseudogene

No pseudogenes are described up to now.

Protein

Description

Collagen type XVI, by structural analogy a member of

the FACIT - (fibril-associated collagens with

interrupted triple helices) family of collagens, contains

10 collagenous (COL) domains interspersed with 11

non-collagenous (NC) regions (Fig. 1A-C). It is a

homotrimeric molecule of about 210 kDa for each

native alpha1 chain. 32 cysteine residues, which are

almost all located in the non-collagenous domains at

the junction to the preceding collagenous regions,

contribute to a high thermal stability of the homotrimer

in form of disulfide bonds (Pan et al., 1992; Yamaguchi

et al., 1992). The prominent non-collagenous NC11

domain consists mainly of a 200-residue motif referred

to as proline-arginine-rich protein (PARP) in several

other collagen types or as tsp-1 in thrombospondin

(Fig. 1A).

Page 60: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 680

Figure 1: Collagen type XVI, domain and molecular structure. Collagen type XVI affinity- purified from culture medium of over expressing HEK 293 EBNA cells elutes in full- length chains and proteolytically processed fragments with following molecular weight as (213 kDa) (A), (182 kDa) (B) and (78 kDa) (C). One alpha 1 chain of intact collagen XVI consists of 10 collagenous domains (COL1-COL10) and 11 non-collagenous domains (NC1-NC11). Atomic Force Microscopy (AFM) of affinity-purified recombinant full-length collagen type XVI trimers allows measurement of molecular size (D). Rotary shadowed TEM image of purified recombinant full-length collagen type XVI trimers corroborates the AFM data (E). Length of intact collagen XVI comes to about 240 nm. COL = collagenous domains, NC = non-collagenous domains.

Page 61: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 681

It has been recombinantly expressed and used for

generation of specific anti-NC11 antibodies (Tillet et

al., 1995). 24% of the total number of proline residues

and 48% of the total number of lysine residues were

hydroxylated in recombinant collagen type XVI.

Because only lysine and proline in the Y position of X-

Y-Gly amino acid triplets and additionally some lysine

residues in the Y position of X-Y-Ser and X-Y-Ala in

collagens are subject to hydroxylation, the amino acid

sequence of authentic human collagen type XVI would

imply that 54% of the available prolines and a

maximum of 92% of the available lysines could

potentially be hydroxylated in recombinant collagen

type XVI. Two of the three potential N-glycosylation

sites reside in the N-terminal NC11 region and are

glycosylated. One has been assigned to the NC1

domain whereas here, no evidence has been found for

the attachment of a glycosaminoglycan chain (Kassner

et al., 2004).

Atomic force images of individual trimeric molecules

exhibited a total length of 168 ± 3 nm including the N-

terminal NC11 globular domains and the flexible

threadlike tail comprising all collagenous regions plus

the remaining non-collagenous domains. The height of

the NC11 domain constituted 0.94 ± 0.06 nm and the

radius at half height (r0.5h) was calculated as 9.48 ± 0.47

nm. The threadlike section of the remaining molecule

C-terminal of the NC11 domain appeared to be a thin

flat structure with a height between 0.5 and 0.7 nm.

Notably, at a distance of 94.8 ± 4.6 nm from the NC11

terminus the molecular measurements increased either

in height or diameter. The extension of this section

contributed with 73.1 ± 1.6 nm to the total length of the

protein (Fig. 1D). Rotary shadowing images of purified

recombinant collagen type XVI exhibited extended rod-

like molecules with a globular domain at one end,

probably constituting the large N-terminal NC11

domain (Fig. 1E). The shape of the molecules revealed

highly flexible regions, in some molecules even two or

more kinks. The length of the molecules varied

between 100-240 nm, with the majority being close to

150 nm.

The N-terminal half of human fibrillin-1/fibrillin-2

binds dose-dependent to collagen type XVI at low salt

concentrations of 50-100 mM NaCl, while interaction

between collagen type XVI and the C-terminal half of

fibrillin-1/-2 under these conditions were considerably

lower. These results indicate that monomeric fibrillin-

1/-2 can interact with collagen type XVI with low

affinity. Both, fibrillin-1 and -2 did not interact with the

recombinant NC11 domain. Soluble recombinant

fibronectin interacted strongly with collagen type XVI

at 150 mM NaCl

with half maximal binding at about 12 µg/ml (about 55

nM fibronectin), indicating that fibronectin can bind to

collagen XVI with high affinity (Kassner et al., 2004).

Collagen XVI co-localizes with alpha2 integrin at the

dermal epidermal junction (DEJ) (Fig. 2A-C) and with

alpha1 integrin and around fat cells in subdermal layers

(Fig. 2D-F). Cells bearing the integrins alpha1beta1

and alpha2beta1 attach and spread on recombinant

collagen type XVI. Collagen type XVI induces the

recruitment of these integrins into focal adhesion

plaques, a principal step in integrin signaling. In cell-

free binding assays, collagen type XVI is more avidly

bound by alpha1beta1 integrin than by alpha2beta1

integrin. Both integrins interact with collagen type XVI

via the A-domain of their alpha-subunits. A tryptic

collagen type XVI fragment comprising the

collagenous domains 1-3 is recognized by alpha1beta1

integrin. Electron microscopy of complexes of

alpha1beta1 integrin with this tryptic collagen XVI

fragment or with full-length collagen type XVI

revealed a unique alpha1beta1 integrin binding site

within collagen type XVI located in the COL 1-3

domains (Eble et al., 2006).

Expression

Collagen type XVI is expressed in various cells and

tissues. It is synthezised by dermal fibroblasts, smooth

muscle cells (Grassel et al., 1996), dermal dendrocytes

and dendritic cells in the skin (Akagi et al., 2002),

articular and costal chondrocytes (Kassner et al., 2003),

endometrial stromal cells (Tierney et al., 2003), basal

dermal and oral keratinocytes (Grassel et al., 1999),

bone marrow derived mesenchymal stem cells (Grassel

et al., 2009), neurons from the dorsal root ganglion

(Hubert et al., 2007), glioblastoma/astrocytoma cells

(Senner et al., 2008) and intestinal myofibroblasts

(Ratzinger et al., Matrix Biol., in revision). Collagen

type XVI is further expressed in the limbal

stem/progenitor niche which comprises clusters of cells

in the basal epithelium. There it is associated with the

corneal-limbal transition zone (Schlotzer-Schrehardt et

al., 2007). During early mouse development, collagen

type XVI occurs in many tissues and is co-distributed

with the major fibrillar collagens. In particular, it is

strongly expressed in differentiating chondrocytes and

dermal fibroblasts, smooth muscle cells of the heart and

dorsal root neural fibers, whereas in adult mice no

signal appears in brain tissue. Additional expression is

found in the cortical areas of the kidney and ovaries

(Lai et al., 1996). In adults it is found in skin, cartilage,

gastrointestinal tract and glioma tissues.

Page 62: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 682

Figure 2: Distribution of collagen XVI and integrins alpha1beta1 and alpha2beta1 in adult murine skin. Cryosections from murine skin were stained with purified anti-collagen XVI-antibodies (red fluorescence, A, B, D, E), and with mAb JA221 directed against the integrin alpha2 subunit (green fluorescence, A, C) and with mAb AGF-1 directed against the integrin alpha1 subunit (green fluorescence, D, F), Panels A and D show combined staining for collagen XVI and the integrin subunits. The same sections are shown either in panel B, E stained for collagen XVI together with DAPI, or in panel C, F stained for integrin subunits and DAPI. E = epidermis, F = fatty tissue, D = dermis, DEJ = dermal epidermal junction zone. All pictures were taken at 400 x magnification.

Figure 3: Ultrastructural localization of collagen XVI in fibrillar extracts from skin and cartilage by immunogold electron microscopy. Double labeling of collagen XVI (18 nm gold particles, light arrow) and fibrillin-1 (12 nm gold particles, dark arrow) demonstrate co-localization on "bead on the string" microfibrils with collagen XVI bound at one distinct bead of the microfibrils. The large D-periodically banded collagen I fibrils lack collagen XVI labeling (light arrowhead). Bar = 0.12 µm. (A). In cartilage a special subpopulation of thin D-periodically banded cartilage fibrils were labeled with collagen XVI (black arrow, 18 nm gold particles), other fibril populations lack collagen XVI association, Bar = 0.09 µm. (B).

Page 63: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 683

Localisation

Extracellular matrix of tissues. For skin and cartilage

tissue its suprastructure is known. It is associated to the

fibrillin containing microfibrillar apparatus in the

dermal-epidermal junction zone in skin and to collagen

II containing D-banded cartilage fibrils in costal

cartilage (Grassel et al., 1999; Kassner et al., 2003). It

is deposited pericellular around fibroblasts and smooth

muscle cells (Grassel et al., 1996) and in the territorial

region of chondrocytes (Kassner et al., 2003).

Function

Morphogenesis and assembly of distinct

suprastructures in different tissues, i.e. microfibrillar

apparatus in the dermis and fibrillar networks in

various connective tissues (Fig. 3A, B). Presumably, it

is an adaptor protein such as collagen type IX and

connects and organizes large fibrillar networks and thus

regulates integrity and stability of ECMs (Eble et al.,

2006; Grassel et al., 1999; Kassner et al., 2003). It is a

substrate for adhesion and invasion of tumor cells, i.e.

glioblastomas (Senner et al., 2008) and regeneration of

connective tissues after neural injury (Hubert et al.,

2007).

Homology

Based on conserved structural features with other

FACIT - collagens, namely: collagen type IX, collagen

type XII, collagen type XIV, collagen type XIX,

collagen type XX, collagen type XXI and collagen type

XXII. These structural features are: the presence of two

highly conserved cysteine residues separated by four

amino acids at the NC1-COL1 junctions and the

existence of two G-X-Y triplet imperfections within the

COL2 domain. A succession of triple-helical domains

connected by short non collagneous domains and the

presence of a large N-terminal domain that always

exhibits a TSPN subdomain next to the collagenous

domain. Besides from these common criteria, the

FACITs display remarkable divergence in the size and

composition of their N-terminal domains and in the

number of their collagenous domains (Ricard-Blum et

al., 2005).

Mutations

Note

There are no reports about mutations in the COL16A1

gene published as yet.

Germinal

None yet described.

Somatic

None yet described.

Implicated in

Glioblastoma tumorigenesis

Note

The progression of glioblastoma growth is

characterized by diffuse invasion of tumor cells into the

brain tissue. COL16A1 was upregulated on mRNA

level in glioblastoma tissues compared to normal cortex

(Fig. 4). Collagen type XVI protein was detected in

glioblastoma tissue and was secreted by glioblastoma

cell lines. A siRNA mediated knockdown of collagen

type XVI resulted in decreased cell adhesion of

glioblastoma cell lines whereas adhesion was

augmented on culture surfaces coated with recombinant

collagen type XVI. However, the migration potential of

glioblastoma cells on collagen type XVI remained

unaffected. Collagen type XVI appears to play a

supportive role for tumour specific remodelling of

extracellular matrix indicated through de novo

expression by glioblastoma cells (Senner et al., 2008).

Disease

Gliomas are the most frequent intrinsic brain tumors

and comprise astrocytic gliomas (grades II, III, IV)

including fibrillary astrocytoma (WHO grade II),

anaplastic astrocytoma (WHO grade III), and

glioblastoma (WHO grade IV). They are characterized

by diffuse invasion of tumour cells into the brain

parenchyma. The fatal outcome of this disease results

from single-tumour cells that have already invaded

distant brain regions at the time of diagnosis.

Glioblastoma behave highly invasive which cause the

high morbidity and mortality rates of these tumours

(Claes et al., 2007; Louis et al., 2007).

Prognosis

The 5-year survival rate of glioblastoma (WHO grade

IV) is 3%.

Neuronal development and regeneration

Note

In the nervous system, low collagen type XVI

expression was reported in the brain, however, in spinal

root fibres high gene expression levels were detected

during development (Lai et al., 1996). A SAGE banks

analysis showed an induction of Col16A1 gene

expression during development and after nerve injury

in dorsal root ganglia (DRG) of mice which contain the

cell bodies of neurons (Mechaly et al., 2006). Their

axons transmit sensory information from the periphery

to the central nervous system. During development and

regeneration, neurites require extracellular matrix for

growth and guidance (Hari et al., 2004),

Page 64: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 684

Figure 4: Immunohistochemical staining of collagen type XVI and collagen type IV in brain sections. Immunofluorescence staining on cryo-sections for collagen XVI (red, white arrows) and collagen IV (green, light blue arrow heads) reveals an expression of collagen XVI around blood vessels, however not in the parenchyma of normal brain (A), whereas in glioblastoma (B) and pilocystic astrocytoma (C) collagen XVI is highly expressed throughout the tumour tissue.

however, the composition of the ECM is yet unknown.

In cell culture, satellite cells express collagen type XVI

indicating secretion and deposition by neuronal and

glia cells. Collagen type XVI participates in final steps

of DRG structural and functional maturation. So far,

collagen type XVI is the only FACIT collagen, whose

expression is regulated by nerve injury, taking

presumably part in remodelling events like

inflammation, cell proliferation, and neuronal death

(Hubert et al., 2007).

Fibrotic skin diseases

Note

In skin, COL16A1 transcripts were detected in cultured

dermal fibroblasts and keratinocytes (Pan et al., 1992).

Gene expression in fibroblasts varied according to the

horizontal layers in skin. Fibroblasts explanted from the

upper dermis displayed higher COL16A1 gene

expression than those from the middle and lower

dermis. In cultured skin fibroblasts an increase of

COL16A1 mRNA level was observed in stationary

phases of the cell cycle (non-adherent and confluent

phases) (Tajima et al., 2000). In localized scleroderma

and in systemic scleroderma COL16A1 gene

expression was upregulated 2.3 fold and 3.6 fold,

respectively, compared with keloid and normal controls

(Akagi et al., 1999).

Disease

Systemic and localized scleroderma are characterized

by systemic and localized deposition of highly

overproduced collagens in the skin. This collagen

accumulation is a result of overproduction of collagens

type I, II, and VI (Graves et al., 1983; Krieg et al.,

1985).

Crohn's disease

Note

Collagen type XVI is produced by myofibroblasts in

the normal intestine and its synthesis is increased in the

inflamed bowel wall (Fig. 5). Collagen type XVI

promotes cell spreading, formation and maturation of

focal adhesion contacts. Myofibroblasts develop

increased numbers of focal adhesion contacts on

collagen type XVI with increased recruitment of alpha1

integrin into the focal adhesions at the tip of the cells.

Focal adhesions on myofibroblasts from inflamed colon

tissue also display an increase in length on collagen

type XVI compared to collagen type I. As a result,

larger forces can be transmitted which then promote

and augment contraction of the ECM and ultimately

result in elevated stricture formation. Increased cell

spreading on collagen type XVI presumably adds to the

maintenance of cells in the inflamed intestinal regions

and thus promotes fibrotic responses of the

Page 65: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 685

tissue and prolongs further disturbances of the delicate

hoemostasis between cells and surrounding ECM

(Ratzinger et al., Matrix Biol., in revision).

Disease

Crohn's disease is characterized by chronic

inflammation of the gastrointestinal tract, accompanied

by other systemic abnormalities. Inflammatory lesions

progress to intestinal fibrotic processes. A

pathologically overshooting healing response to

inflammation-induced disintegration of mucosal tissue

leads to excessive tissue repair.

An altered cytoarchitecture of the bowel wall with

disruption of the muscularis mucosa, thickening of the

muscularis propria, and deposition of collagens

contributes to the inflammation process (Burke et al.,

2007). Fibrillar and non-fibrillar collagens are up-

regulated in CD (type I, II, IV, V, VI) (Graham et al.,

1988; Matthes et al., 1992; Pucilowska et al.,

2000; Stallmach et al., 1992). Mesenchymal cells like

fibroblasts, myofibroblasts and smooth-muscle cells are

the main producers of extracellular matrix components

and play an important part in tissue growth and

development (Powell et al., 1999; Simon-Assmann et

al., 1995). Myofibroblasts are considered as central

player in tissue repair contributing to fibrosis, stricture

formation and stenosis by reconstituting a collagen-rich

extracellular matrix (ECM) and promoting wound

closure by contraction (Pucilowska et al., 2000;

Tomasek et al., 2002). Myofibroblasts motility, their

ability to contract wounds and the production of ECM

is altered in chronic inflammation. Normal wound

healing would terminate the contractile and

synthesizing activity of myofibroblasts by apoptotic

reduction of the cell number (Desmouliere et al., 1995).

Page 66: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 686

Figure 5: Morphological distribution of collagen type XVI in health and disease. Morphological distribution of collagen XVI (green fluorescence) and alpha-smooth muscle actin (red fluorescence) is demonstrated for the bowel wall of healthy tissue (A-D) and CD tissue (E-H). Arrow heads indicate positive staining for collagen XVI. A negative control is displayed as inlet in D. c: crypts, m: muscle layer, s: submucosa, bv: blood vessel, e: erythrocytes, ep: epithelial cells, lp: lamina propria.

References Graves PN, Weiss IK, Perlish JS, Fleischmajer R. Increased procollagen mRNA levels in scleroderma skin fibroblasts. J Invest Dermatol. 1983 Feb;80(2):130-2

Krieg T, Perlish JS, Mauch C, Fleischmajer R. Collagen synthesis by scleroderma fibroblasts. Ann N Y Acad Sci. 1985;460:375-86

Graham MF, Diegelmann RF, Elson CO, Lindblad WJ, Gotschalk N, Gay S, Gay R. Collagen content and types in the intestinal strictures of Crohn's disease. Gastroenterology. 1988 Feb;94(2):257-65

Matthes H, Herbst H, Schuppan D, Stallmach A, Milani S, Stein H, Riecken EO. Cellular localization of procollagen gene transcripts in inflammatory bowel diseases. Gastroenterology. 1992 Feb;102(2):431-42

Pan TC, Zhang RZ, Mattei MG, Timpl R, Chu ML. Cloning and chromosomal location of human alpha 1(XVI) collagen. Proc Natl Acad Sci U S A. 1992 Jul 15;89(14):6565-9

Stallmach A, Schuppan D, Riese HH, Matthes H, Riecken EO. Increased collagen type III synthesis by fibroblasts isolated from strictures of patients with Crohn's disease. Gastroenterology. 1992 Jun;102(6):1920-9

Yamaguchi N, Kimura S, McBride OW, Hori H, Yamada Y, Kanamori T, Yamakoshi H, Nagai Y. Molecular cloning and partial characterization of a novel collagen chain, alpha 1(XVI), consisting of repetitive collagenous domains and cysteine-containing non-collagenous segments. J Biochem. 1992 Dec;112(6):856-63

Desmoulière A, Redard M, Darby I, Gabbiani G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. Am J Pathol. 1995 Jan;146(1):56-66

Simon-Assmann P, Kedinger M, De Arcangelis A, Rousseau V, Simo P. Extracellular matrix components in intestinal development. Experientia. 1995 Sep 29;51(9-10):883-900

Tillet E, Mann K, Nischt R, Pan TC, Chu ML, Timpl R. Recombinant analysis of human alpha 1 (XVI) collagen. Evidence for processing of the N-terminal globular domain. Eur J Biochem. 1995 Feb 15;228(1):160-8

Grässel S, Timpl R, Tan EM, Chu ML. Biosynthesis and processing of type XVI collagen in human fibroblasts and smooth muscle cells. Eur J Biochem. 1996 Dec 15;242(3):576-84

Lai CH, Chu ML. Tissue distribution and developmental expression of type XVI collagen in the mouse. Tissue Cell. 1996 Apr;28(2):155-64

Akagi A, Tajima S, Ishibashi A, Yamaguchi N, Nagai Y. Expression of type XVI collagen in human skin fibroblasts: enhanced expression in fibrotic skin diseases. J Invest Dermatol. 1999 Aug;113(2):246-50

Grässel S, Unsöld C, Schäcke H, Bruckner-Tuderman L, Bruckner P. Collagen XVI is expressed by human dermal fibroblasts and keratinocytes and is associated with the microfibrillar apparatus in the upper papillary dermis. Matrix Biol. 1999 Jun;18(3):309-17

Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB. Myofibroblasts. II. Intestinal subepithelial myofibroblasts. Am J Physiol. 1999 Aug;277(2 Pt 1):C183-201

Pucilowska JB, Williams KL, Lund PK. Fibrogenesis. IV. Fibrosis and inflammatory bowel disease: cellular mediators and animal models. Am J Physiol Gastrointest Liver Physiol. 2000 Oct;279(4):G653-9

Tajima S, Akagi A, Tanaka N, Ishibashi A, Kawada A, Yamaguchi N. Expression of type XVI collagen in cultured skin fibroblasts is related to cell growth arrest. FEBS Lett. 2000 Mar 3;469(1):1-4

Akagi A, Tajima S, Ishibashi A, Matsubara Y, Takehana M, Kobayashi S, Yamaguchi N. Type XVI collagen is expressed in factor XIIIa+ monocyte-derived dermal dendrocytes and constitutes a potential substrate for factor XIIIa. J Invest Dermatol. 2002 Feb;118(2):267-74

Sironen RK, Karjalainen HM, Törrönen K, Elo MA, Kaarniranta K, Takigawa M, Helminen HJ, Lammi MJ. High pressure effects on cellular expression profile and mRNA stability. A cDNA array analysis. Biorheology. 2002;39(1-2):111-7

Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002 May;3(5):349-63

Kassner A, Hansen U, Miosge N, Reinhardt DP, Aigner T, Bruckner-Tuderman L, Bruckner P, Grässel S. Discrete

Page 67: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COL16A1 (collagen, type XVI, alpha 1) Grässel S, Ratzinger S

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 687

integration of collagen XVI into tissue-specific collagen fibrils or beaded microfibrils. Matrix Biol. 2003 Apr;22(2):131-43

Tierney EP, Tulac S, Huang ST, Giudice LC. Activation of the protein kinase A pathway in human endometrial stromal cells reveals sequential categorical gene regulation. Physiol Genomics. 2003 Dec 16;16(1):47-66

Hari A, Djohar B, Skutella T, Montazeri S. Neurotrophins and extracellular matrix molecules modulate sensory axon outgrowth. Int J Dev Neurosci. 2004 Apr;22(2):113-7

Kassner A, Tiedemann K, Notbohm H, Ludwig T, Mörgelin M, Reinhardt DP, Chu ML, Bruckner P, Grässel S. Molecular structure and interaction of recombinant human type XVI collagen. J Mol Biol. 2004 Jun 11;339(4):835-53

Ricard-Blum S, Ruggiero F. The collagen superfamily: from the extracellular matrix to the cell membrane. Pathol Biol (Paris). 2005 Sep;53(7):430-42

Eble JA, Kassner A, Niland S, Mörgelin M, Grifka J, Grässel S. Collagen XVI harbors an integrin alpha1 beta1 recognition site in its C-terminal domains. J Biol Chem. 2006 Sep 1;281(35):25745-56

Méchaly I, Bourane S, Piquemal D, Al-Jumaily M, Ventéo S, Puech S, Scamps F, Valmier J, Carroll P. Gene profiling during development and after a peripheral nerve traumatism reveals genes specifically induced by injury in dorsal root ganglia. Mol Cell Neurosci. 2006 Jul;32(3):217-29

Burke JP, Mulsow JJ, O'Keane C, Docherty NG, Watson RW, O'Connell PR. Fibrogenesis in Crohn's disease. Am J Gastroenterol. 2007 Feb;102(2):439-48

Claes A, Idema AJ, Wesseling P. Diffuse glioma growth: a guerilla war. Acta Neuropathol. 2007 Nov;114(5):443-58

Hubert T, Grimal S, Ratzinger S, Mechaly I, Grassel S, Fichard-Carroll A. Collagen XVI is a neural component of the developing and regenerating dorsal root ganglia extracellular matrix. Matrix Biol. 2007 Apr;26(3):206-10

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007 Aug;114(2):97-109

Schlötzer-Schrehardt U, Dietrich T, Saito K, Sorokin L, Sasaki T, Paulsson M, Kruse FE. Characterization of extracellular matrix components in the limbal epithelial stem cell compartment. Exp Eye Res. 2007 Dec;85(6):845-60

Senner V, Ratzinger S, Mertsch S, Grässel S, Paulus W. Collagen XVI expression is upregulated in glioblastomas and promotes tumor cell adhesion. FEBS Lett. 2008 Oct 15;582(23-24):3293-300

Grässel S, Ahmed N, Göttl C, Grifka J. Gene and protein expression profile of naive and osteo-chondrogenically differentiated rat bone marrow-derived mesenchymal progenitor cells. Int J Mol Med. 2009 Jun;23(6):745-55

This article should be referenced as such:

Grässel S, Ratzinger S. COL16A1 (collagen, type XVI, alpha 1). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):679-687.

Page 68: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 688

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

COPS2 (COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)) Susanne Jennek, Florian Kraft, Aria Baniahmad

Institute of Human Genetics and Anthropology, Jena University Hospital, Kollegiengasse 10, 07743 Jena,

Germany (SJ, FK, AB)

Published in Atlas Database: September 2009

Online updated version : http://AtlasGeneticsOncology.org/Genes/COPS2ID47362ch15q21.html DOI: 10.4267/2042/44805

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: ALIEN; CSN2; SGN2; TRIP15

HGNC (Hugo): COPS2

Location: 15q21.1

Note:

The beta casein is also abbreviated as CSN2.

DNA/RNA

Transcription

The promoter region of CSN2 contains 4 NF-kB

binding sites. Binding to these sites activates the

transcription of CSN2 gene. Deletion of the C terminus

of NF-kB abrogates the ability to induce CSN2 gene

expression (Wu et al., 2009).

Furthermore, CSN2/Alien gene expression in vivo is

activated by thyroid hormone receptor (TR) and thyroid

hormone suggesting a regulatory feedback mechanism

between TR and CSN2/Alien expression (Tenbaum et

al., 2003).

The CSN2 gene is localized on chromosome 15q21.2 (top panel). A homologous sequence is located on chromosome 9q33.2 (not shown).The gene structure highlights intron/exon arrangement, whereas red boxes display coding sequence (CDS) and grey the untranslated regions. The lower panel exhibits the CSN2/Alien cDNA. The green boxes indicate the 13 exons and the black line below illustrates the coding sequences CDS. The grey box in isoform 2 highlights the additional inserted 21bp stretch which is specific for this splice variant. Differential functions of these isoforms are not yet known. The italic numbers illustrate the base pairs.

Page 69: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COPS2 (COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)) Jennek S, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 689

Schematic structure of the CSN2 protein (modified according to Akiyama et al., 2003). The N-terminal region contains a region for interaction with DAX-1/NiF3l1 (aa 1-275). The central part of the protein includes the nuclear localization signal (NLS), a leucine zipper (LZ) domain and a corepressor region (CR) which contains an I/LXXI/VI motif. The C-terminal region contains a PCI domain, might be used for interaction between CSN subunits (reviewed in: Wei and Deng, 2003). The numbers represent the amino acids (aa).

Protein

Description

Three isoforms are known:

CSN2 short/Alien a: 305 amino acids (aa); 36 kDa

protein.

CSN2 long 1/Alien b1: 443 aa; 51.6 kDa protein.

CSN2 long 2/Alien b2: 450 aa; 52.4 kDa protein.

CSN2 activity can be regulated through

phosphorylation and dephosphorylation (Kapelari et al.,

2000).

Expression

Mouse Csn2 is widely expressed in embryonic, fetal

and adult tissues (Schaefer et al., 1998). Several mRNA

levels have been described in mice: 1,8 kb ; 2,2 kb ; 4

kb and 6 kb (Schaefer et al.,1998; Altincicek et al.,

2000; Tenbaum et al., 2003).

Localisation

CSN2 is localized in both the cytoplasm and the

nucleus, predominantly being localized in the nucleus

(Schaefer et al., 1998; Dressel et al., 1999; Tenbaum et

al., 2003).

Function

CSN2 short/Alien a acts as a corepressor for nuclear

hormone receptors (NHR). Originally in mammalians,

Alien was identified as an interacting protein of the

thyroid hormone receptor (TR) in a ligand-sensitive

manner (Lee et al., 1995). Moreover, Alien enhances

TR-mediated gene silencing through its autonomous

silencing function (Dressel et al., 1999). Additionally,

the vitamin D receptor (VDR), the androgen receptor

(AR) and the orphan receptor DAX-1 can also interact

with Alien. Functionally Alien enhances gene silencing

mediated by these nuclear receptors (Altincicek et al.,

2000; Polly et al., 2000; Moehren et al., 2007).

Notably, Alien seems to lack interaction with retinoid

X receptor (RXR), retinoid acid receptor (RAR),

estrogen receptor (ER), glucocorticoid receptor (GR)

and germ cell nuclear factor (GCNF) (Dressel et al.,

1999; Fuhrmann et al., 2000; reviewed in: Papaioannou

et al., 2007).

Also, corepression function was identified by CSN2

short/Alien a for transcription factors involved in cell

cycle regulation and DNA repair such as several

members of the E2F transcription factor family (Escher

et al., 2007; reviewed in: Papaioannou et al., 2007).

Alien is recruited to the E2F1 gene promoter repressing

endogenous E2F1 gene expression in vivo. The data

also suggest that Alien inhibits transactivation of E2F1,

a positive regulator of cell cycle progression. In line

with this, Alien represses cell cycle progression.

Remarkably, the inhibition of E2F1-mediated

transactivation is independent of retinoblastoma protein

pRB (Tenbaum et al., 2007). pRB represses E2F1

transcriptional activation. It is not yet known whether

Alien is able to substitute pRB function during cell

cycle progression (Tenbaum et al., 2007). Furthermore,

a direct interaction between Alien and pRB is detected.

Interestingly, a pRB-mutant lacking silencing function

also lacks interaction with CSN2 short/Alien (Escher et

al., 2007).

In addition, Alien interacts with the highly conserved

chromatin associated tumor suppressor proteins

Inhibitor of growth 1 (ING1b) and 2 (ING2) in vivo

and both p33ING1b and p33ING2 are known to induce

premature cellular senescence. It is shown that p33ING

proteins enhance Alien-mediated gene silencing

(Fegers et al., 2007).

The recruitment of HDAC-activity is one mechanism

by which Alien realizes its corepression functions

(Dressel et al., 1999). However, it is suggested that

Alien exhibits both HDAC-dependent and -independent

options for gene repression (reviewed in: Papaioannou

et al., 2007). Moreover, CSN2 short/Alien a interacts

with nucleosome assembly protein 1 (NAP1) in vivo

and in vitro regulating its activity through enhancing

NAP1-mediated nucleosome assembly on DNA and

thereby leading to gene repression (Eckey et al., 2007).

The CSN2 long/Alien b isoform is an essential part of

the COP9 signalosome (CSN) complex which is highly

conserved in eukaryotes and consists of eight subunits

(reviewed in: Wie et al., 2008). The CSN complex

plays a central role in the regulation of degradation of

multiple proteins through the ability to de-neddylate

cullin, which enables the association of cullin with

CAND1, a negative regulator of the cullin-based E3

ubiquitin ligases (reviewed in: von Arnim, 2003;

reviewed in: Wolf et al., 2003; Chamovitz, 2009; Wu et

al., 2009). A role for CSN2 long/Alien b is suggested

by the interaction between CSN2 and subunits of the

26S proteasome was already shown (Huang et al.,

2005). The promoter region of the Csn2 gene contains

NF-kB binding sites like other CSN subunits.

Page 70: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COPS2 (COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)) Jennek S, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 690

Accordingly, these members of the CSN complex are

regulated by NF-kB. Snail, a transcription factor, which

is a part of the TGF-b pathway and is involved in

inflammatory-triggered migration, invasiveness and

metastasis of tumor cells, is stabilized by the induction

of the CSN complex via NF-kB (Wu et al., 2009).

There are also COP9 subcomplexes with yet unknown

functions (reviewed in: Wei et al, 2008).

Interaction of COP9 via CSN2 with p53 in tumors can

raise the stability of p53, the most important protein

involving in a variety of essential tumor suppressive

functions and induction of cellular senescence. But in

contrast to Snail, the lower turnover does not lead to an

increase in transcription activity and therefore neither

to an increased p21 expression nor to cell cycle arrest

(Leal et al., 2008).

Moreover, CSN2 protein interacts physically with the

anaphase-promoting complex (APC/C), a major

regulator of the cell cycle and affects specifically its

stability (Kob et al., 2008).

Homology

CSN2 is a highly conserved protein from humans to

Drosophila (Dressel et al., 1999). CSN2 has

homologies in any multicellular organism including

plants. It is over 60% identical between animal and

plant counterparts (Wei and Deng, 2003;

Schwechheimer, 2004).

Originally, the name Alien was given to a gene in the

Drosophila genome with an unknown function

(Goubeaud et al., 1996). It shares high homologies with

Thyroid hormone receptor-interacting protein 15

(TRIP15), a mammalian protein (Lee et al., 1995;

Dressel et al., 1999).

Mutations

Note

So far natural occurring point mutations of CSN2 in

association with cancer and other disease were not yet

described.

Implicated in

Human tumors

Oncogenesis

Aberrant expression of CNS2/Alien seems to be

associated with human tumors. CSN2 expression is lost

in several human tumors. In thyroid tumors the loss of

CSN2 is at least 50% (Leal et al., 2008). Moreover,

high percentages of reduction (15-30%) of CSN2

mRNA level were observed in tumors of pancreas,

breast, ovary, kidney, uterus und rectum (Leal et al.,

2008).

It was shown by quantitative analysis that the CSN2

expression is reduced up to 50% in tumors of prostate,

lung and colon (Leal et al., 2008).

References Lee JW, Choi HS, Gyuris J, Brent R, Moore DD. Two classes of proteins dependent on either the presence or absence of thyroid hormone for interaction with the thyroid hormone receptor. Mol Endocrinol. 1995 Feb;9(2):243-54

Goubeaud A, Knirr S, Renkawitz-Pohl R, Paululat A. The Drosophila gene alien is expressed in the muscle attachment sites during embryogenesis and encodes a protein highly conserved between plants, Drosophila and vertebrates. Mech Dev. 1996 Jun;57(1):59-68

Dressel U, Thormeyer D, Altincicek B, Paululat A, Eggert M, Schneider S, Tenbaum SP, Renkawitz R, Baniahmad A. Alien, a highly conserved protein with characteristics of a corepressor for members of the nuclear hormone receptor superfamily. Mol Cell Biol. 1999 May;19(5):3383-94

Altincicek B, Tenbaum SP, Dressel U, Thormeyer D, Renkawitz R, Baniahmad A. Interaction of the corepressor Alien with DAX-1 is abrogated by mutations of DAX-1 involved in adrenal hypoplasia congenita. J Biol Chem. 2000 Mar 17;275(11):7662-7

Kapelari B, Bech-Otschir D, Hegerl R, Schade R, Dumdey R, Dubiel W. Electron microscopy and subunit-subunit interaction studies reveal a first architecture of COP9 signalosome. J Mol Biol. 2000 Jul 28;300(5):1169-78

Polly P, Herdick M, Moehren U, Baniahmad A, Heinzel T, Carlberg C. VDR-Alien: a novel, DNA-selective vitamin D(3) receptor-corepressor partnership. FASEB J. 2000 Jul;14(10):1455-63

Fuhrmann G, Chung AC, Jackson KJ, Hummelke G, Baniahmad A, Sutter J, Sylvester I, Schöler HR, Cooney AJ. Mouse germline restriction of Oct4 expression by germ cell nuclear factor. Dev Cell. 2001 Sep;1(3):377-87

Akiyama H, Fujisawa N, Tashiro Y, Takanabe N, Sugiyama A, Tashiro F. The role of transcriptional corepressor Nif3l1 in early stage of neural differentiation via cooperation with Trip15/CSN2. J Biol Chem. 2003 Mar 21;278(12):10752-62

Tenbaum SP, Juenemann S, Schlitt T, Bernal J, Renkawitz R, Muñoz A, Baniahmad A. Alien/CSN2 gene expression is regulated by thyroid hormone in rat brain. Dev Biol. 2003 Feb 1;254(1):149-60

von Arnim AG. On again-off again: COP9 signalosome turns the key on protein degradation. Curr Opin Plant Biol. 2003 Dec;6(6):520-9

Wei N, Deng XW. The COP9 signalosome. Annu Rev Cell Dev Biol. 2003;19:261-86

Wolf DA, Zhou C, Wee S. The COP9 signalosome: an assembly and maintenance platform for cullin ubiquitin ligases? Nat Cell Biol. 2003 Dec;5(12):1029-33

Schwechheimer C. The COP9 signalosome (CSN): an evolutionary conserved proteolysis regulator in eukaryotic development. Biochim Biophys Acta. 2004 Nov 29;1695(1-3):45-54

Huang X, Hetfeld BK, Seifert U, Kähne T, Kloetzel PM, Naumann M, Bech-Otschir D, Dubiel W. Consequences of COP9 signalosome and 26S proteasome interaction. FEBS J. 2005 Aug;272(15):3909-17

Eckey M, Hong W, Papaioannou M, Baniahmad A. The nucleosome assembly activity of NAP1 is enhanced by Alien. Mol Cell Biol. 2007 May;27(10):3557-68

Page 71: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

COPS2 (COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)) Jennek S, et al.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 691

Escher N, Kob R, Tenbaum SP, Eisold M, Baniahmad A, von Eggeling F, Melle C. Various members of the E2F transcription factor family interact in vivo with the corepressor alien. J Proteome Res. 2007 Mar;6(3):1158-64

Fegers I, Kob R, Eckey M, Schmidt O, Goeman F, Papaioannou M, Escher N, von Eggeling F, Melle C, Baniahmad A. The tumor suppressors p33ING1 and p33ING2 interact with alien in vivo and enhance alien-mediated gene silencing. J Proteome Res. 2007 Nov;6(11):4182-8

Papaioannou M, Melle C, Baniahmad A. The coregulator Alien. Nucl Recept Signal. 2007 Nov 30;5:e008

Tenbaum SP, Papaioannou M, Reeb CA, Goeman F, Escher N, Kob R, von Eggeling F, Melle C, Baniahmad A. Alien inhibits E2F1 gene expression and cell proliferation. Biochim Biophys Acta. 2007 Sep;1773(9):1447-54

Leal JF, Fominaya J, Cascón A, Guijarro MV, Blanco-Aparicio C, Lleonart M, Castro ME, Ramon Y Cajal S, Robledo M,

Beach DH, Carnero A. Cellular senescence bypass screen identifies new putative tumor suppressor genes. Oncogene. 2008 Mar 27;27(14):1961-70

Chamovitz DA. Revisiting the COP9 signalosome as a transcriptional regulator. EMBO Rep. 2009 Apr;10(4):352-8

Kob R, Kelm J, Posorski N, Baniahmad A, von Eggeling F, Melle C. Regulation of the anaphase-promoting complex by the COP9 signalosome. Cell Cycle. 2009 Jul 1;8(13):2041-9

Wu Y, Deng J, Rychahou PG, Qiu S, Evers BM, Zhou BP. Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell. 2009 May 5;15(5):416-28

This article should be referenced as such:

Jennek S, Kraft F, Baniahmad A. COPS2 (COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):688-691.

Page 72: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 692

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(3;6)(q27;p21) Jean-Loup Huret

Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France

(JLH)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t0306q27p21ID2155.html DOI: 10.4267/2042/44806

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Note: In only two cases were the hybrid genes

uncovered: a PIM1/BCL6 was found in one case

(Yoshida et al., 1999), and a SFRS3/BCL6 in another

case (Chen et al., 2001). These cases will also be

described separately.

Clinics and pathology

Disease

Non Hodgkin lymphoma (NHL).

Epidemiology

Seven cases available: 3 cases of follicular lymphoma

(FL), 3 cases of diffuse large B-cell lymphoma

(DLBCL), and one NHL not otherwise specified. Very

few data available: 4 male / 2 female patients, aged 49-

79 years (Ohno et al., 1994; Miura et al., 1996; Yoshida

et al., 1999; Chen et al., 2001; Varga et al., 2001;

Keller et al., 2006).

Cytogenetics

Cytogenetics morphological

The t(3;6) was the sole anomaly in one case (a FL

case), accompanied with +X in a subclone and +21 in

another in the other FL case, found within a complex

karyotype in the 4 remainind cases with cytogenetic

data. Other additional anomalies of note were: del(7q),

t(14;18)(q32;q21) (2 cases each) and t(1;14)(q21;q32),

del(1q), del(5q) (1 case each).

Genes involved and proteins

BCL6

Location

3q27

Protein

706 amino acids; composed of a NH2-term BTB/POZ

domain (amino acids 1-130 (32-99 according to Swiss-

Prot)) which mediates homodimerization and protein-

protein interactions with other corepressors (including

HDAC1 and NCOR2/SMRT) to constitute a large

repressing complex, another transcription repression

domain (191-386), PEST sequences (300-417) with a

KKYK motif (375-379), and six zinc finger at the C-

term (518-541, 546-568, 574-596, 602-624, 630-652,

658-681), responsible for sequence specific DNA

binding. Transcription repressor; recognizes the

consensus sequence: TTCCT(A/C)GAA (Albagli-

Curiel, 2003).

References Ohno H, Kerckaert JP, Bastard C, Fukuhara S. Heterogeneity in B-cell neoplasms associated with rearrangement of the LAZ3 gene on chromosome band 3q27. Jpn J Cancer Res. 1994 Jun;85(6):592-600

Miura I, Ohshima A, Takahashi N, Hashimoto K, Nimura T, Utsumi S, Saito M, Miki T, Hirosawa S, Miura AB. A new non-random chromosomal translocation t(3;6)(q27;p21.3) associated with BCL6 rearrangement in two patients with non-Hodgkin's lymphoma. Int J Hematol. 1996 Oct;64(3-4):249-56

Yoshida S, Kaneita Y, Aoki Y, Seto M, Mori S, Moriyama M. Identification of heterologous translocation partner genes fused to the BCL6 gene in diffuse large B-cell lymphomas: 5'-RACE and LA - PCR analyses of biopsy samples. Oncogene. 1999 Dec 23;18(56):7994-9

Chen W, Itoyama T, Chaganti RS. Splicing factor SRP20 is a novel partner of BCL6 in a t(3;6)(q27;p21) translocation in transformed follicular lymphoma. Genes Chromosomes Cancer. 2001 Nov;32(3):281-4

Varga AE, Dobrovic A, Webb GC, Hutchinson R. Clustering of 1p36 breakpoints distal to 1p36.2 in hematological malignancies. Cancer Genet Cytogenet. 2001 Feb;125(1):78-9

Albagli-Curiel O. Ambivalent role of BCL6 in cell survival and transformation. Oncogene. 2003 Jan 30;22(4):507-16

Page 73: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

t(3;6)(q27;p21) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 693

Keller CE, Nandula S, Vakiani E, Alobeid B, Murty VV, Bhagat G. Intrachromosomal rearrangement of chromosome 3q27: an under recognized mechanism of BCL6 translocation in B-cell non-Hodgkin lymphoma. Hum Pathol. 2006 Aug;37(8):1093-9

This article should be referenced as such:

Huret JL. t(3;6)(q27;p21). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):692-693.

Page 74: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 694

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(3;6)(q27;p21) PIM1/BCL6 Jean-Loup Huret

Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France

(JLH)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t0306q27p21ID2088.html DOI: 10.4267/2042/44807

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Note: t(3;6)(q27;p21) has been described in a few cases

were PIM1/BCL6 rearrangement has not been

ascertained, or were another hybrid gene has been

uncovered.

Clinics and pathology

Disease

Non Hodgkin lymphoma (NHL).

Epidemiology

Only one case available: a case of diffuse large B-cell

lymphoma (DLBCL) (Yoshida et al., 1999).

Genes involved and proteins

BCL6

Location

3q27

Protein

706 amino acids; composed of a NH2-term BTB/POZ

domain (amino acids 1-130 (32-99 according to Swiss-

Prot)) which mediates homodimerization and protein-

protein interactions with other corepressors (including

HDAC1 and NCOR2/SMRT) to constitute a large

repressing complex, another transcription repression

domain (191-386), PEST sequences (300-417) with a

KKYK motif (375-379), and six zinc finger at the C-

term (518-541, 546-568, 574-596, 602-624, 630-652,

658-681), responsible for sequence specific DNA

binding.

Transcription repressor; recognizes the consensus

sequence: TTCCT(A/C)GAA (Albagli-Curiel, 2003).

PIM1

Location

6p21.2

Protein

404 amino acids; serine/threonine-protein kinase;

regulated by hematopoietic cytokine receptors; synergy

with c-MYC in cell proliferation and in apoptosis

induction through an enhancement of the activation of

caspase-3-like proteases; Cdc25A (cell cycle

phosphatase) is a substrate for Pim-1.

Result of the chromosomal anomaly

Hybrid gene

Description

5' PIM1 - 3' BCL6, but also 5' BCL6 - 3' PIM1;

breakpoint in BCL6 between exon 1 and 2.

References Yoshida S, Kaneita Y, Aoki Y, Seto M, Mori S, Moriyama M. Identification of heterologous translocation partner genes fused to the BCL6 gene in diffuse large B-cell lymphomas: 5'-RACE and LA - PCR analyses of biopsy samples. Oncogene. 1999 Dec 23;18(56):7994-9

Albagli-Curiel O. Ambivalent role of BCL6 in cell survival and transformation. Oncogene. 2003 Jan 30;22(4):507-16

This article should be referenced as such:

Huret JL. t(3;6)(q27;p21) PIM1/BCL6. Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):694.

Page 75: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 695

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(3;6)(q27;p21) SFRS3/BCL6 Jean-Loup Huret

Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France

(JLH)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t0306q27p21ID1336.html DOI: 10.4267/2042/44808

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Note: t(3;6)(q27;p21) has been described in a few cases

where SFRS3/BCL6 rearrangement has not been

ascertained, or where another hybrid gene has been

uncovered.

Clinics and pathology

Disease

Non Hodgkin lymphoma (NHL).

Epidemiology

Only one case available: a case of follicular lymphoma

(FL) (Chen et al., 2001).

Cytogenetics

Cytogenetics morphological

The t(3;6) was accompanied with del(1q), del(7q),

t(14;18)(q32;q21) and other anomalies.

Genes involved and proteins

BCL6

Location

3q27

Protein

706 amino acids; composed of a NH2-term BTB/POZ

domain (amino acids 1-130 (32-99 according to Swiss-

Prot)) which mediates homodimerization and protein-

protein interactions

with other corepressors (including HDAC1 and

NCOR2/SMRT) to constitute a large repressing

complex, another transcription repression domain (191-

386), PEST sequences (300-417) with a KKYK motif

(375-379), and six zinc finger at the C-term (518-541,

546-568, 574-596, 602-624, 630-652, 658-681),

responsible for sequence specific DNA binding.

Transcription repressor; recognizes the consensus

sequence: TTCCT(A/C)GAA (Albagli-Curiel, 2003).

SFRS3

Location

6p21.3

Protein

164 amino acids (alternate splicing 124 aa); SFRS3 is a

member of the serine- and arginine-rich (SR) protein

family; comprise a RNA recognition motifs (RRM)

(amino acids 10-83) and an SR domain (aa 86-164)

according to Swiss-Prot; role in splicing of mRNA

precursors; promote the export of some cellular

mRNAs; possible role in cell cycle.

References Chen W, Itoyama T, Chaganti RS. Splicing factor SRP20 is a novel partner of BCL6 in a t(3;6)(q27;p21) translocation in transformed follicular lymphoma. Genes Chromosomes Cancer. 2001 Nov;32(3):281-4

Albagli-Curiel O. Ambivalent role of BCL6 in cell survival and transformation. Oncogene. 2003 Jan 30;22(4):507-16

This article should be referenced as such:

Huret JL. t(3;6)(q27;p21) SFRS3/BCL6. Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):695.

Page 76: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 696

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(8;20)(p11;q13) Marie-Joëlle Mozziconacci, Christine Pérot

Institut Paoli-Calmettes, 232 Bd de Sainte-Marguerite, 13009 Marseille, France (MJM); Hôpital Saint-

Antoine, 184 rue du Faubourg Saint-Antoine, 75012 Paris, France (CP)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t820p11q13ID1507.html DOI: 10.4267/2042/44809

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Top. GTG and R-banded partial karyotypes. Bottom. FISH of metaphase chromosomes of t(8;20)(p11;q13) with digoxigenin-labeled RP11-313J18 (MYST3 at 8p11) and biotinylated RP11-1151C1 (5' and main part of NCOA3 region at 20q13) and RP11-122N8 (3' part of NCOA3 and SULF2 region at 20q13). Fused RP11-313J18 / RP11-1151C1+RP11-122N8 (red/green) signals are observed on der(8) and der(20) chromosomes. Courtesy of Christine Pérot and Marie-Joëlle Mozziconacci.

Page 77: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

t(8;20)(p11;q13) Mozziconacci MJ, Pérot C

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 697

Clinics and pathology

Disease

Acute myeloid leukemia AML-M5 without features of

erythrophagocytosis.

Note

Only one case reported (a 75-year old woman). t(8;20)

as a sole abnormality.

Clinics

Splenomegaly, DIVC. Hyperleucocytosis, anemia and

thrombopenia.

Treatment

Hydroxyurea and low-dose cytosine-arabinoside.

Evolution

Death 2 months after diagnosis. No remission obtained.

Genes involved and proteins

MYST3 (MYST histone acetyltransferase (monocytic leukemia) 3)

Location

8p11

Note

MYST3 is a histone acetyltransferase (HAT) belonging

to the MYST family of HATs, that includes proteins

involved in cell cycle regulation, chromatin remodeling

and dosage compensation. MYST3 plays an important

role during hematopoiesis with his transcriptional

coregulator activity.

DNA/RNA

Breakpoint in intron 17.

Protein

MYST3 contains a LAP (Leukemia associated protein)

zinc finger domain, a HAT domain (Histone

acetyltransferase) and a acidic domain. 2004 amino

acids; 225 kDa.

NCOA3 (Nuclear Receptor Coactivator 3)

Location

20q13.1

Note

NCOA3 is a transcriptional coactivator that interacts

with nuclear hormone receptors and with other

transcription factors including TP52, NfkB and ER81.

It has intrinsic histone acetyltransferase activity and

recruits CREB Binding Protein (CBP)/p300 co-

integrators into multisubunit coactivator complexes.

DNA/RNA

Breakpoint in exon 13 (Δ45 bp).

Protein

Member of the p160/steroid receptor coactivator

family. 1424 amino acids; 155 kDa (130 kDa encoded

by isoform b).

Result of the chromosomal anomaly

Hybrid gene

Note

Both MYST3-NCOA3 and NCOA3-MYST3 are

expressed. Only the MYST3-NCOA3 fusion transcript

has an open reading frame that may generate a

functional chimeric protein.

Fusion protein

Note

The CREB-interacting domain in NCOA3 (1046-

1092aa) is conserved in the putative MYST3-NCOA3

fusion protein (at positions 1246-1292aa).

References Esteyries S, Perot C, Adelaide J, Imbert M, Lagarde A, Pautas C, Olschwang S, Birnbaum D, Chaffanet M, Mozziconacci MJ. NCOA3, a new fusion partner for MOZ/MYST3 in M5 acute myeloid leukemia. Leukemia. 2008 Mar;22(3):663-5

This article should be referenced as such:

Mozziconacci MJ, Pérot C. t(8;20)(p11;q13). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):696-697.

Page 78: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Solid Tumour Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 698

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma DunFa Peng, Wael El-Rifai

Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA (DP, WER)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Tumors/BarrettsEsophagID5591.html DOI: 10.4267/2042/44810

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Note: Barrett's adenocarcinoma is one of malignancies

with the most rapid increase in incidence during past

decades in the Western countries. It is defined as

adenocarcinoma of the lower esophagus and

gastroesophageal junction associated with Barrett's

esophagus. Barrett's esophagus is the only known

precursor for Barrett's adenocarcinoma through

Barrett's dysplasia (also called metaplasia-dysplasia-

adenocarcinoma sequence).

Clinics and pathology

Disease

Barrett's Esophagus

Note

Barrett's esophagus is defined as the normal esophageal

squamous epithelium that is replaced by intestinalized

metaplastic columnar epithelia.

Phenotype / cell stem origin

The progenitor cell from which Barrett's oesophagus

develops is still unclear. Progenitor cells resident in the

submucosal glands or the interbasal layer of the

epithelium, bone-marrow-derived stem cells, or

transdifferentiated squamous cells are included in the

candidate cells.

Etiology

Gastroesophageal reflux disease (GERD) is considered

the major risk factor for Barrett's esophagus. About 1 in

10 patients with GERD are found to have Barrett's

esophagus. GERD generates reactive oxygen species

that produce oxidative stress and subsequent oxidative

DNA damage. Some DNA damage may cause DNA

mutations that accumulate and cause tumor formation.

Epidemiology

Barrett's esophagus is more common in men than in

women, with a male:female ratio of about 2:1. The risk

factors of Barrett's esophagus include Age (increasing

with age), Race (more common in Caucasians),

smoking (not clear), alcohol consumption (not clear),

gastroesophageal reflux disease (GERD, major factor),

and obesity. On the other hand, some controversial

reports indicate that H. pylori infection and the virulent

cagA strains in particular, may protect against the

development of Barrett's oesophagus and progression

to adenocarcinoma.

Clinics

Heartburn is the most common symptom of GERD and

Barrett's esophagus.

Page 79: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma Peng D, El-Rifai W

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 699

Normal esophagus is covered by squamous epithelia (A). However, in Barrett's esophagus (B), the squamous epithelia are replaced by intestinalized metaplastic columnar epithelia.

Disease

Barrett's dysplasia

Note

Barrett's dysplasia is defined morphologically as

unequivocal neoplastic epithelium that remains

confined within the basement membrane of the

epithelium from which it developed. In patients with

Barrett's esophagus, dysplasia is graded as either low or

high, depending on its cytological and architectural

features.

Epidemiology

Most Barrett's esophagus never progress to dysplasia

and carcinoma. It has been reported that the male

gender, longstanding gastroesophageal reflux disease,

hiatal hernia size, and segment length are strongly

associated with Barrett's dysplasia. On the other hand,

successful antireflux surgery protects the Barrett's

mucosa from developing high-grade dysplasia and

esophageal adenocarcinoma.

Disease

Barrett's adenocarcinoma

Note

Adenocarcinoma of lower esophagus and

gastroesophageal junction associated with Barrett's

esophagus through metaplasia-dysplasia-

adenocarcinoma sequence.

Classification: Tumor classification is based on UICC

TNM classification for esophageal cancers.

Etiology

Barrett's esophagus with dysplasia

Epidemiology

Barrett's esophagus is the only known precursor for

Barrett's adenocarcinoma. The patients with Barrett's

esophagus have 20 folds more risk for developing

esophageal adenocarcinoma. However, only 1-5% of

Barrett's esophagus progress to Barrett's

adenocarcinoma. Although longstanding

gastroesophageal reflux disease, hiatal hernia size,

Page 80: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma Peng D, El-Rifai W

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 700

A representative image of a Barrett's adenocarcinoma with moderate to poor differentiation. Atypic tumor cells form quite irregular tubules and some form solid cord.

and segment length are strongly associated with

adenocarcinoma, Barrett's esophagus with dysplasia is

likely the true precursor for developing to

adenocarcinoma.

Clinics

Heartburn is the most common symptom of GERD and

Barrett's esophagus. As for Barrett's adenocarcinoma, it

shares the symptoms with other esophageal type

cancers, such as difficulty swallowing, unexplained

weight loss, pain in the throat or mid-chest, etc.

Pathology

There is no difference in the term of histology of

Barrett's adenocarcinoma from that in the stomach and

colon. It can be graded into well-, moderately- and

poorly-differentiated adenocarcinoma based on their

cytologic and architectural atypia. The Lauren

classification for gastric cancer has also been used by

some pathologists and physicians to divide into either

intestinal or diffuse histological type.

Treatment

Esophagectomy is still the most common primary

treatment. Other treatment modalities include

chemotherapy, radiation therapy, stents, photodynamic

therapy, and endoscopic therapy with an Nd:YAG

laser. Combined modality therapy (i.e., chemotherapy

plus surgery or chemotherapy and radiation therapy

plus surgery) is under clinical evaluation.

Prognosis

The prognosis of Barrett's adenocarcinoma depends on

the stage at diagnosis, treatment and the patients'

general condition. The overall 5-year survival rate in

patients amenable to definitive treatment ranges from 5

to 30%.

Cytogenetics

Note

The chromosomal alterations most frequently identified

in Barrett's adenocarcinoma by CGH were: gains in 8q

(80%), 20q (60%); 2p, 7p and 10q (47% each), 6p

(37%), 15q (33%), and 17q (30%). Losses were

observed predominantly in the 4q (50%); 5q and 9p

(43% each), 18q (40%), 7q (33%), and 14q (30%).

Genes involved and proteins

Note

There are many genes that have been reported to be

genetically and/or epigenetically dysregulated, such as

gene mutation, amplification, and LOH and DNA

methylation that involved cell cycle control, apoptosis,

cell adhesion and antioxidative stress, etc. Some

representative genes are described below.

ERBB2 (HER2/neu)

Location

17q21

Note

ERBB2 protein over-expression and/or DNA

amplification have been reported in 10-70% of

esophageal adenocarcinomas. The present literature

data suggest that ERBB2 overexpression may be a late

event in the dysplasia-carcinoma sequence, as it

occurred predominantly in high-grade dysplasia and

adenocarcinomas.

Page 81: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma Peng D, El-Rifai W

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 701

CGH analysis of a case of Barrett's esophageal adenocarcinoma. Tumor DNA was labeled with FITC (Green) and reference DNA was labeled with TRITC (red). The hybridizations were analyzed using an Olympus fluorescence microscope and the ISIS digital image analysis system (Metasystems GmbH, Altlussheim, Germany) based on integrated high-sensitivity monochrome CCD camera and automated CGH analysis software. The green colon indicates areas of DNA gains whereas the red color indicates DNA losses in the tumor sample.

It has been reported that ERBB2 over-

expression/amplification in carcinoma correlated

significantly with tumor invasion, lymph node

metastasis, and poor prognosis in patients with Barrett's

related adenocarcinoma.

Protein

The ERBB2 (also called HER2 or NEU) gene encodes

an integral type I protein of 185 kDa, 1255 amino

acids, with a cysteine-rich extracellular ligand-binding

domain, a transmembrane domain and an intracellular

region endowed with a tyrosine kinase activity.

C-MYC

Location

8q24

Note

Frequent high-level chromosomal amplification of

8q21 has been reported in Barrett's adenocarcinoma

and c-myc is the potential target gene for this

amplification.

It has been reported that amplification of c-myc was

detected in 25% of high-grade dysplasia and 44% of

adenocarcinomas, but in none of Barrett's metaplasia

and low-grade dysplasia.

Protein

DNA binding protein with 439 amino acids and 48 kDa

(p64); 454 amino acids (p67, 15 additional amino acids

in N-term), contains a transactivation domain, an acidic

domain, a nuclear localization signal, a basic domain, a

helix-loop-helix motif, and a leucin zipper.

CDX1

Location

5q33.1

Note

CDX1 is predominantly expressed in the small intestine

and colon, but not in normal esophageal squamous

epithelia or gastric epithelia.

CDX1 was over-expressed in Barrett's esophagus and

adenocarcinomas, most likely through promoter DNA

hypomethylation.

Protein

Member of the caudal-related homeobox transcription

factor gene family. The encoded protein regulates

intestine-specific gene expression and differentiation of

intestine.

CDX2

Location

13q12.2

Note

CDX2 is predominantly expressed in the small intestine

and colon, but not in normal esophageal squamous

epithelia or gastric epithelia.

CDX2 expression is observed in the intestinal

metastasis area.

Page 82: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma Peng D, El-Rifai W

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 702

Protein

Member of the caudal-related homeobox transcription

factor gene family.

The encoded protein regulates intestine-specific gene

expression and differentiation of intestine. It is

suggested that CDX2 is a "master switch" gene whose

normal expression determines the proximal and distal

specialization of the gut in embryogenesis.

CDKN2A (p16)

Location

9p21.3

Note

Tumor suppressor gene controls the G1/S transition of

the cell cycle. Inactivation of CDKN2A is among the

most common genetic/epigenetic alterations through

Barrett's carcinogenesis and is an early event. LOH,

promoter hypermethylation, or sequence mutations

have been reported in over 85% of Barrett's esophagus

that were associated with p16 inactivation.

DNA / RNA

7288 bp.

Exon Count: 3.

Protein

Tumor suppressor protein having 156 aa, functions as

an inhibitor of CDK4 kinase in cell cycle G1 control.

TP53

Location

17p13

Note

Loss of p53 occurs through either LOH, sequence

mutation, or both. Loss of p53 has been reported in

Barrett's esophagus and likely correlates with

progression to adenocarcinoma, as patients with LOH

in TP53 are 16 times more likely to progress to

adenocarcinoma than patients without TP53 LOH.

DNA / RNA

Exon Count: 11.

Protein

A tumor suppressor protein essential in cell cycle

regulation and in DNA damage repair. p53

transcriptionally regulates multiple genes functioning

as an inhibitor of cell growth and proliferation and

inducer of apoptosis. Loss of TP53 functions promotes

tumor progression, most likely by preventing cell cycle

arrest, suppressing apoptosis and permitting genetic

instability for subsequent genetic alterations.

CDKN1B (p27)

Location

12p13

Note

It has been reported that in 83% of Barrett's

adenocarcinomas, p27 protein was down-regulated by

an immunohistochemical study. And for p27 to arrest

cell cycle, it must localize in the nucleus. However, in

approximately 50% of high-grade dysplasia, p27 was

observed in cytoplasmic localization, which renders it

inactive.

Protein

A tumor suppressor protein essential in cell cycle

regulation, a CDK inhibitor.

GPX3

Location

5q33.1

Note

GPX3 is one of the glutathione peroxidase family

members, which functions in the detoxification of

hydrogen peroxide. Frequent GPX3 gene promoter

hypermethylation has recently been demonstrated in

Barrett's adenocarcinomas and its precancerous lesions,

Barrett's esophagus and dysplasia, and was

significantly associated with gene down-regulation. It

is noted that GPX3 has been recently reported as a

potential tumor suppressor in prostatic carcinomas.

Protein

GPX3 is a secretory protein.

GPX7

Location

1p32.3

Note

GPX7 is one of the glutathione peroxidase family

members which function in the detoxification of

hydrogen peroxide. Unlike other glutathione peroxidase

family members, GPX7 incorporates cysteine instead

of selenocysteine in the conserved catalytic motif.

Frequent GPX7 gene promoter hypermethylation has

recently been demonstrated in Barrett's

adenocarcinomas and its precancerous lesion, Barrett's

dysplasia, and was significantly associated with gene

down-regulation. Recent research indicates that GPX7

may have dual functions, antioxidative activity and

tumor suppressor function in Barrett's adenocarcinoma.

DNA / RNA

Genomic Size: 6679.

Exon Count: 3.

Coding Exon Count: 3.

MGMT

Location

10q26.3

Note

It is recently reported that hypermethylation was

detected in 78.9% of esophageal adenocarcinomas, in

100% of Barrett's intraepithelial neoplasia, in 88.9% of

Barrett's metaplasia, in only 21.4% of normal

esophageal mucosa samples (P<0.001), and correlated

significantly with the down-regulation of MGMT

transcripts (P=0.048) and protein expression (P=0.02).

The decrease of protein expression was significantly

Page 83: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma Peng D, El-Rifai W

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 703

correlated with progressed stage of disease, lymph node

invasion and tumor size.

DNA / RNA

Genomic Size: 299903.

Exon Count: 5.

Coding Exon Count: 4.

Protein

O6-methylguanine-DNA methyltransferase is involved

in the cellular defense against the biological effects of

O6-methylguanine (O6-MeG) in DNA. It repairs

alkylated guanine in DNA by stoichiometrically

transferring the alkyl group at the O6 position to a

cysteine residue in the enzyme.

APC

Location

5q21

Note

LOH of 5q has been reported as a higher occurrence in

high-grade dysplasia and adenocarcinomas of the

esophagus. Hypermethylation of the APC gene has

been found in 68-100% of Barrett's adenocarcinomas

and approximately 50% of Barrett's esophagus. More

importantly, hypermethylation of APC with other genes

such as p16, strongly predicts progression to high-grade

dysplasia or cancer in patients with BE. Absence of p16

and APC hypermethylation is associated with a benign

course.

DNA / RNA

Genomic Size: 138719.

Exon Count: 16.

Coding Exon Count: 15.

Protein

Adenomatous polyposis coli protein which possesses

tumor suppressor functions, works as an antagonist of

the Wnt signaling pathway. APC binding to beta

catenin leads to ubiquitin-mediated beta catenin

destruction; loss of APC function increases

transcription of beta catenin targets, such as C-MYC

and Cyclin D. It is also involved in other processes

including cell migration and adhesion, transcriptional

activation, and apoptosis. Germline defects in this gene

cause familial adenomatous polyposis (FAP), an

autosomal dominant pre-malignant disease that usually

progresses to malignancy. Disease-associated

mutations tend to be clustered in a small region

designated the mutation cluster region (MCR) and

result in a truncated protein product.

References BARRETT NR. The lower esophagus lined by columnar epithelium. Surgery. 1957 Jun;41(6):881-94

El-Rifai W, Frierson HF Jr, Moskaluk CA, Harper JC, Petroni GR, Bissonette EA, Jones DR, Knuutila S, Powell SM. Genetic differences between adenocarcinomas arising in Barrett's esophagus and gastric mucosa. Gastroenterology. 2001 Sep;121(3):592-8

Bian YS, Osterheld MC, Fontolliet C, Bosman FT, Benhattar J. p16 inactivation by methylation of the CDKN2A promoter occurs early during neoplastic progression in Barrett's esophagus. Gastroenterology. 2002 Apr;122(4):1113-21

Koppert LB, Wijnhoven BP, van Dekken H, Tilanus HW, Dinjens WN. The molecular biology of esophageal adenocarcinoma. J Surg Oncol. 2005 Dec 1;92(3):169-90

Lee OJ, Schneider-Stock R, McChesney PA, Kuester D, Roessner A, Vieth M, Moskaluk CA, El-Rifai W. Hypermethylation and loss of expression of glutathione peroxidase-3 in Barrett's tumorigenesis. Neoplasia. 2005 Sep;7(9):854-61

Oberg S, Wenner J, Johansson J, Walther B, Willén R. Barrett esophagus: risk factors for progression to dysplasia and adenocarcinoma. Ann Surg. 2005 Jul;242(1):49-54

Wong A, Fitzgerald RC. Epidemiologic risk factors for Barrett's esophagus and associated adenocarcinoma. Clin Gastroenterol Hepatol. 2005 Jan;3(1):1-10

Fitzgerald RC. Molecular basis of Barrett's oesophagus and oesophageal adenocarcinoma. Gut. 2006 Dec;55(12):1810-20

Maley CC. Multistage carcinogenesis in Barrett's esophagus. Cancer Lett. 2007 Jan 8;245(1-2):22-32

Razvi MH, Peng D, Dar AA, Powell SM, Frierson HF Jr, Moskaluk CA, Washington K, El-Rifai W. Transcriptional oncogenomic hot spots in Barrett's adenocarcinomas: serial analysis of gene expression. Genes Chromosomes Cancer. 2007 Oct;46(10):914-28

Kuester D, El-Rifai W, Peng D, Ruemmele P, Kroeckel I, Peters B, Moskaluk CA, Stolte M, Mönkemüller K, Meyer F, Schulz HU, Hartmann A, Roessner A, Schneider-Stock R. Silencing of MGMT expression by promoter hypermethylation in the metaplasia-dysplasia-carcinoma sequence of Barrett's esophagus. Cancer Lett. 2009 Mar 8;275(1):117-26

Peng DF, Razvi M, Chen H, Washington K, Roessner A, Schneider-Stock R, El-Rifai W. DNA hypermethylation regulates the expression of members of the Mu-class glutathione S-transferases and glutathione peroxidases in Barrett's adenocarcinoma. Gut. 2009 Jan;58(1):5-15

This article should be referenced as such:

Peng D, El-Rifai W. Esophagus: Barrett's esophagus, dysplasia and adenocarcinoma. Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):698-703.

Page 84: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Solid Tumour Section Review

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 704

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Head and neck: Retinoblastoma Hayyam Kiratli, Berçin Tarlan

Ocular Oncology Service, Department of Ophthalmology, Hacettepe University School of Medicine, Ankara,

Turkey (HK, BT)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Tumors/RetinoblastomID5008.html DOI: 10.4267/2042/44811

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Alias

Retinal glioma; Fungus hematodes

Note

Retinoblastoma is a malignant primary intraocular

tumor predominantly encountered in infancy and early

childhood. Another striking definition of

retinoblastoma is that it is a childhood cancer that can

be completely cured with radiotherapy alone. This

tumor develops from the retina and by the age of 5

years, 90% of cases are diagnosed.

Classification

Note

Retinoblastoma is evaluated on genetic basis, laterality

and focality. Approximately 85% of

retinoblastomas are sporadic and 15% of cases are

familial. Unilateral tumors account for 60% of cases

and bilateral involvement is seen in 40% of patients.

Eighty-five percent of unilateral tumors have somatic

mutations and 15% have germinal mutations.

Conversely, 90% of bilateral tumors have germinal

mutations and 10 have postzygotic somatic mutations.

When both eyes are involved multifocality is the rule.

There is an average of five tumors per eye in bilateral

cases. Trilateral retinoblastoma is a rare and often

lethal condition in which there is an undifferentiated

neuroectodermal tumor in the pineal gland or in

para/supra sellar region of the midbrain. Ninety percent

of pinealoblastomas develop in patients with bilateral

disease. Unilateral cases have a 0.5% risk of

developing trilateral retinoblastoma.

Group I a. Solitary tumor, less than 4 disc diameters in size, at or behind the equator.

b. Multiple tumors, none over 4 disc diameters in size, all at or behind the equator.

Group II a. Solitary tumor, 4 to 10 disc diameters in size, at or behind the equator.

b. Multiple tumors, 4 to 10 disc diameters in size, behind the equator.

Group III a. Any lesion anterior to the equator.

b. Solitary tumors larger than 10 disc diameters behind the equator.

Group IV a. Multiple tumors some larger than 10 disc diameters.

b. Any lesion extending anteriorly to the ora serrata.

Group V a. Massive tumors involving half of the retina.

b. Vitreous seeding.

Table 1. Reese-Ellsworth classification for intraocular retinoblastoma.

Group A All tumors ≤ 3mm, confined to the retina, at least 3 mm from the foveola, 1.5 mm from the disc. No

Page 85: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Head and neck: Retinoblastoma Kiratli H, Tarlan B

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 705

vitreous or subretinal seeding. (Very low risk).

Group B Discrete retinal tumor of any size and location without vitreous or subretinal seeding. Subretinal fluid

extending ≤ 5mm from the tumor base is allowed. (Low risk).

Group C Discrete retinal tumors of any size and location with focal vitreous or subretinal seeding treatable with

brachytherapy. One quadrant of subretinal fluid is allowed. (Moderate risk).

Group D Diffuse vitreous or subretinal seeding associated with massive, nondiscrete endophytic or exophytic

tumor. > 1 quadrant of retinal detachment. (High risk).

Group E

Eyes destroyed anatomically and functionally by: neovascular glaucoma, intraocular hemorrhage, tumor

in the anterior vitreous, tumor touching the lens, aseptic orbital cellulitis, diffuse type retinoblastoma,

phthisis bulbi. (Very high risk).

Table 2. International Classification of Retinoblastoma (Murphree).

Clinics and pathology

Note

Leukocoria (white reflex from the pupillary aperture)

(60%) and strabismus (20%) are the two major

presenting signs of retinoblastoma (see Figure 1A).

Rarely patients may present with buphthalmus, pseudo-

hypopyon, hyphema, vitreous hemorrhage, and pseudo-

orbital cellulitis. A typical retinoblastoma is a round or

oval shaped, variably vascularized pink mass. It may

sometimes appear chalky white because of

calcification. Endophytic retinoblastoma grows into the

vitreous cavity and accounts for 60% of typical

retinoblastomas (see Figure 1B).

Exophytic tumors (39%) grow under the subretinal

space and cause retinal detachment. Rarely the tumor

may show diffuse growth pattern (1%) in which there is

no detectable mass but a sheet-like dissemination of the

malignant cells within the retina. This latter type of

presentation usually occurs in older children. Currently,

the Reese-Ellsworth (RE) and International

Classification of Retinoblastoma (ICRB) systems are

used in staging the disease, which is very important in

treatment planning (see tables above).

Phenotype / cell stem origin

The cell of origin of retinoblastoma is a topic of hot

debate. In simple terms, the cell of origin is the cell in

which the tumor suppression activity of pRB is first

required. This may not necessarily be the cell in which

loss of RB1 gene occurs.

Several models and hypotheses exist on the cell of

origin. It was long believed that a retinal multipotent

cell committed to cone differentiation was the cell of

origin. Recent studies found strong expressions of

minimicrosome maintenance protein 2 (MCM2)

and ABCG2 (a casette-binding transmembrane protein

that confers drug resistance) on retinoblastoma cells

that favors a neural cancer stem cell origin. Also tumor

cells were found to express CD44 (hyaluronate

receptor), PROX1 and syntaxin 1A (retinal progenitor

markers), CD90 (retinal ganglion cell marker) and

CD133 (photoreceptor cell marker) all supporting the

cancer stem cell theory. Against the stem cell theory,

some studies showed expression of mature neural cell

markers including MAP2, NSE, synaptophysin and

opsin suggesting mature neural and amacrine cells as

the source of retinoblastoma. Others observed that a

fully differentiated retinal horizontal cell could reenter

the cell cycle and could form tumor foci.

Etiology

Inactivation of both wild type alleles of the

retinoblastoma susceptibility gene (RB1) is postulated

to cause the development of retinoblastoma.

Epidemiology

Retinoblastoma accounts for 4% of all pediatric cancers

and the cumulative incidence is 1/18000-30000 live

births per year regardless of sex, race or geographic

predilection. Each year, 5000 to 8000 new cases of

retinoblastoma are diagnosed on the global scale.

Retinoblastoma is responsible for 1% of all deaths

below 15 years of age. The incidence of hereditary

retinoblastoma is relatively constant in various parts of

the world. However, there seems to be increased

incidence of non-hereditary sporadic retinoblastoma in

underdeveloped parts of the world. This was partly

related to the widely common human papilloma virus

infections in those areas. Advanced paternal age is also

associated with more sporadic gene mutations.

Page 86: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Head and neck: Retinoblastoma Kiratli H, Tarlan B

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 706

Figure 1 A. Left leukocoria in a 2 year-old child. B. A typical macular endophytic retinoblastoma with another small tumor nasal to the optic disc. C. A Flexner-Wintersteiner rosette with a clear lumen at the centre of the figure. D. Fleurettes within a well-differentiated retinoblastoma.

Pathology

Retinoblastoma is composed of small, round densely

packed cells with large hyperchromic nuclei and

basophilic cytoplasms. There may be vast areas of

necrosis and calcification because of rapid tumor

growth and insufficient blood supply. Various degrees

of photoreceptor differentiation are evidenced by

typical cellular arrangements. Flexner-Wintersteiner

rosettes are aggregates of cuboidal or columnar cells

around a central lumen, considered as an aborted

attempt to form photoreceptors (see Figure 1C).

A more advanced step towards photoreceptor formation

is the fleurette type rosettes. These are formed by tumor

cells with eosinophilic cellular extensions arranged in a

semicircular fashion with bulb-like endings. It is

believed that red and green cones participate in the

formation of these rosettes whereas blue cones tend to

form structures called bacillettes.

Histopathological demonstration of tumor cells 1 mm

beyond the lamina cribrosa, scleral invasion, clumps of

cells within more than 50% of choroidal thickness are

established risk factors for extraocular dissemination of

retinoblastoma. Increased intraocular pressure, iris

neovascularisation and buphthalmus are risk factors for

optic nerve invasion (see Figure 1D).

Treatment

Several options exist depending on the stage and

laterality of the tumor.

1. Chemotherapy: Systemic chemotherapy has

become the most commonly used method worldwide

within the past 10 years for almost all intraocular

retinoblastomas. The rationale is to shrink the tumor

(chemoreduction) so that subsequent local

consolidation treatments are used to further destroy the

tumor and thus avoid enucleation or external beam

radiotherapy.

In general, eyes having a potential of a useful vision

but containing large tumors untreatable with local

methods, children under the age of one year, and

advanced bilateral cases are eligible for

chemoreduction. Current protocols include vincristine,

carboplatin and etoposide or teniposide. A successful

outcome can be obtained in 100% of

Page 87: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Head and neck: Retinoblastoma Kiratli H, Tarlan B

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 707

Figure 2 A. RE IIIb or ICRB group C retinoblastoma before chemotherapy. B. The same eye shown above following 9 cycles of VEC chemoreduction protocol. C. Multiple small tumors easily treatable with TTT. D. Atrophic scars following TTT. E. Osteogenic sarcoma of the maxillary sinus 11 years after external beam radiotherapy.

group A, 93% in group B, 90% in group C, and 47% in

group D eyes. Most tumors regress more than 50%

within 3-4 weeks. The most important complication of

chemoreduction therapy is recurrence of the tumor,

which is more common in macular tumors. Also, new

ocular tumors may develop while under systemic

chemotherapy. Transient myelosuppression, cytopenia

and neutropenia occur in 100% of patients. The

Page 88: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Head and neck: Retinoblastoma Kiratli H, Tarlan B

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 708

development of secondary non-ocular cancers

following chemotherapy is an unresolved issue.

Preliminary studies suggest an increased incidence of

AML particularly in patients who had received

teniposide, which acts on chromosome 11q35. (see

Figure 2A and 2B).

2. Local chemotherapy: Local administration of

chemotherapeutic agents is in use to deliver higher

concentrations of the drug into the eye and avoid

systemic toxicity and side effects. Large molecules can

easily pass the sclera regardless of lipophilicity.

Injection of carboplatin into the subtenon space is

effective against localized mild amounts of vitreous

seedings but this effect is transient and rarely curative.

Additionally, carboplatin is rapidly cleared from the

vitreous limiting its effects. To overcome this

inconvenience, a sustained delivery system of

carboplatin from fibrin sealants is developed. Recently,

supraselective intra-arterial infusion of melphelan into

the ophthalmic artery resulted in satisfactory tumor

regression in eyes that would otherwise have to be

enucleated.

3. Enucleation: This time-honored surgical treatment

is indicated for most of group E or RE V eyes where

there is no prospect of vision. Eyes with elevated

intraocular pressure, rubeosis iridis, tumor in the

anterior chamber, buphthalmus and evidence for optic

nerve involvement need to be enucleated. Failure of

prior chemotherapy and radiotherapy are other

indications for enucleation. In general, enucleation

becomes necessary in 75% of unilateral cases because

of the advanced stage at the time of diagnosis.

Likewise, enucleation of at least one eye (the worst

eye) becomes unavoidable in 60% of bilateral cases.

Bilateral enucleation may be performed in 1% of cases.

4. Cryotherapy: Rapid freezing of the tumor to -90°C

damages the vascular endothelia causing platelet plugs

to form thrombosis and induces tumor ischemia. In

addition, intracellular ice crystal formation causes

rupture of the cellular membranes. All tumors with less

than 5 mm basal diameter and few vitreous seedings

close to the tumor can successfully be treated with

cryotherapy.

5. Brachytherapy: Iodine-125 and Ruthenium-106

radioactive plaques are widely used to treat solitary

tumors having 6-15 mm basal diameters and less than 9

mm thickness. The trend is to prescribe 4000-4500 cGy

radiation to the tumor apex. To overall success rate is

90% but there is a tumor recurrence rate of 12% at one

year. Radiation induced retinopathy and optic

neuropathy are the most common complications.

6. Transpupillary thermotherapy (TTT): 810 nm

infrared diode laser is used for this treatment. Tumors

smaller than 3 mm of basal diameter without vitreous

seedings can be reliably treated with TTT either

primarily or following chemoreduction. Because of

technical difficulties, peripheral tumors are avoided.

The power is usually set at 200-1000 mW and 1.2 to 3

mm spot sizes are used for 1 minute each, aiming

directly the tumor. The result is a flat and atrophic scar.

There is an overall 86% success rate with

complications including focal iris atrophy, lens

opacities, optic disc atrophy, retinal tractions, vascular

occlusions, and retinal hemorrhages. (see Figure 2C

and 2D).

7. External Beam Radiotherapy (EBRT): This

modality continues to be very effective in selected

patients despite fears for secondary cancers. Eyes with

multifocal tumors not treatable by other local

techniques, macular tumors where other methods may

ultimately destroy the central vision, and advanced

bilateral disease are good candidates for EBRT. Also,

EBRT can be performed after failure of other methods

as a salvage therapy, in patients with extraocular orbital

tumor invasion or tumor at the surgical margin of the

resected optic nerve. The target tumor receives 4200-

4600 cGy radiation in 180-200 cGy fractionated doses

daily. Local tumor control rates vary between 50% to

88% depending on the stage of the disease. If vitreous

seedings are present the success rate of EBRT is only

17%. The most significant concern with the use of

EBRT is the development of second non-ocular and

periocular cancers particularly in survivors of

hereditary retinoblastoma. There is a 400-600 fold

increase in the risk of developing second cancers in

hereditary retinoblastoma if treated with EBRT and this

risk is further multiplied by 8 if the treatment is given

below the age of 1 year. Second malignant tumors

develop in 4.4% of patients during the first 10 years, in

18.3% within 20 years and in 26.1% after 30 years. The

most common second cancers include osteogenic

sarcoma, leiomyosarcoma, pinealoblastoma, skin

melanoma, Hodgkin's lymphoma, lung and breast

carcinomas. (see Figure 2E).

8. Gene Therapy: The preliminary results of

intravitreal injection of adenovirus carrying the coding

sequence of thymidine kinase followed by ganciclovir

injection appear promising. In human subjects, this

treatment decreased vitreous seedings but main tumors

remained intact.

9. Experimental Therapies:

- COX-2, which is expressed in retinoblastoma, is a

prostaglandin synthetase promoting angiogenesis,

suppressing apoptosis and increasing tumor

invasiveness. The role of COX-2 inhibitors is

investigated in retinoblastoma.

- Oxidative stress, which is high in retinoblastoma,

upregulates aA-crystallins, member of heath shock

proteins, helping tumor cells to escape apoptosis. Anti-

aA-crystallin therapy is thought to have a potential to

limit tumor growth in retinoblastoma.

- Retinoblastoma cells can produce VEGF and basic

fibroblast growth factor both of which induce

angiogenesis. The initial enthusiasm on anti-angiogenic

drugs vanished because it was found that these drugs

were active against immature vasculature found in the

periphery of the tumor. Vessels that are more central

had pericyte components and thus became mature no

Page 89: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Head and neck: Retinoblastoma Kiratli H, Tarlan B

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 709

more dependant on angiogenic stimuli. This seriously

limits the effects of anti-angiogenic agents.

- Arsenic trioxide has been shown to have effect on

retinoblastoma cells by generating reactive oxygen

species which oxidize lipids in the mitochondria

membranes. This results in cytochrome C release and

activation of the caspase system leading to apoptosis.

- Retinoblastoma contains many hypoxic areas where

cellular proliferation is slower compared to areas close

to blood vessels. These slow proliferating cells usually

do not respond to available chemotherapeutic drugs. 2-

deoxy-D-glucose (2-DG), a glycolytic inhibitor, holds

promise against these non-responding cell populations.

Prognosis

With increased awareness and early diagnosis coupled

with the current diagnostic techniques and management

options, 99% of children with intraocular

retinoblastoma survive the disease and 90% of patients

retain useful vision in at least one eye. The prognosis is

still dismal if there is orbital extension of the tumor or

distant hematogenous metastasis.

Cytogenetics

Note

Retinoblastoma develops because of inactivation of

both alleles of the RB1 tumor suppressor gene after two

successive mutations (M1 and M2). In patients with

hereditary (germinal) retinoblastoma, the germline

contains an inactivated RB1 allele that is also present in

all cells of the individual. The tumor develops when the

other allele is lost (M2) in a retinal cell. The result is

bilateral and multifocal tumors. In non-hereditary

cases, both M1 and M2 occur in a single retinal cell

thus producing unilateral and unifocal disease.

Genes involved and proteins

Note

The RB1 is the first gene to be discovered to have

tumor suppression function. In most cases, the first

allele is lost because of a point mutation (M1). Most

mutations are non-sense which produces a premature

stop codon and a resultant non functional protein. Loss

of heterozygosity (M2) is responsible for the loss of the

second allele in 60% of tumors. It is now recognized

that M1 and M2 are not sufficient to drive the cell into

malignant transformation and that other genomic

changes (M3-MX) are necessary. Recent studies

identified gains and amplifications at 1q32 (MDM4 and

KIF14 genes) and 6p22 (E2F3 and DEK genes) as M3

and M4 respectively. Less frequent but important

genomic changes include 16q22 loss (CDH11 and

RBL2 genes) and 2p24 gains (MYCN and DDX1

genes).

RB1

Location

13q14

Note

The retinoblastoma gene RB1 is localized on

chromosome 13q14. In only 3% of tumors,

karyotypically visible large deletions can be

demonstrated in this location.

RB1 spans 180 kb and is composed of 27 exons. This

gene encodes a 4.8 kb mRNA and the protein is a 110

kD nuclear phosphoprotein (pRB) containing 928

aminoacids. This protein has anti-oncogenic function,

induces differentiation and blocks the anti-apoptotic

properties of MDM2. The tumor suppressor function is

through E2F at the cell cycle checkpoint between G1

and S-phase entry. The protein has many pockets which

bind several molecules the most important being E2F

transcription factors.

When pRB is in normal hypophosphorylated state,

E2F1 is bound and the cell cannot enter the S-phase.

When pRB is phosphorylated or other competing

molecules for the pockets including SV40 virus,

papillomavirus or adenovirus oncoproteins bind, E2F1

is released and the cascade for uncontrolled cellular

proliferation proceeds.

References Abramson DH, Schefler AC. Update on retinoblastoma. Retina. 2004 Dec;24(6):828-48

Abramson DH. Retinoblastoma in the 20th century: past success and future challenges the Weisenfeld lecture. Invest Ophthalmol Vis Sci. 2005 Aug;46(8):2683-91

Shields JA, Shields CL, Meadows AT. Chemoreduction in the management of retinoblastoma. Am J Ophthalmol. 2005 Sep;140(3):505-6

Shields CL, Mashayekhi A, Au AK, Czyz C, Leahey A, Meadows AT, Shields JA. The International Classification of Retinoblastoma predicts chemoreduction success. Ophthalmology. 2006 Dec;113(12):2276-80

Kim JW, Abramson DH, Dunkel IJ. Current management strategies for intraocular retinoblastoma. Drugs. 2007;67(15):2173-85

Schefler AC, Abramson DH. Retinoblastoma: what is new in 2007-2008. Curr Opin Ophthalmol. 2008 Nov;19(6):526-34

Shields CL, Ramasubramanian A, Thangappan A, Hartzell K, Leahey A, Meadows AT, Shields JA. Chemoreduction for group E retinoblastoma: comparison of chemoreduction alone versus chemoreduction plus low-dose external radiotherapy in 76 eyes. Ophthalmology. 2009 Mar;116(3):544-551.e1

This article should be referenced as such:

Kiratli H, Tarlan B. Head and neck: Retinoblastoma. Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):704-709.

Page 90: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Case Report Section Paper co-edited with the European LeukemiaNet

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 710

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(1;21)(p32;q22) as a non-random abnormality in AML M4 Lena Reindl, Claudia Haferlach

MLL, Munich Leukemia Laboratory, Max-Lebsche-Platz 31, Germany (LR, CH)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Reports/0121ReindlID100041.html DOI: 10.4267/2042/44812

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Clinics

Age and sex

63 years old female patient.

Previous history

No preleukemia; no previous malignancy; no inborn

condition of note.

Organomegaly

No hepatomegaly , no splenomegaly , no enlarged

lymph nodes, no central nervous system involvement.

Blood WBC : 3.980X 10

9/l

HB : 7.9g/dl

Platelets : 64.000X 109/l

Blasts : 48,5%

Cyto-Pathology Classification

Cytology

(FAB) AML M4.

Immunophenotype

Hypercellular bone marrow showed a myelomonocytic

blast population. 49.5% blasts were detected in total

bone marrow. 30% of the cells were clearly EST

positive.

Futhermore POX was positive, no ringsiderobalsts

were found and erythropoiesis showed dysplasia.

Myelomonocytic cells with MPO+ (48%), CD13+

(17%), CD33+ (63%), CD14 (19%) and CD64 (37%).

Diagnosis

AML M4

Survival

Date of diagnosis: 06-2008

Treatment: None

Complete remission : no

Treatment related death : no

Relapse : no

Status : Lost

Karyotype

Sample: bone marrow

Culture time: 24 - 48h

Banding: GAG.

Results: 46,XX,t(1;21)(p32;q22)[15/15].

Other molecular cytogenetics technics:

FISH with commercial AML1 probe (Abbott) and

whole chromosome painting with WCP#1 and

WCP#21 (MetaSystems).

Other molecular cytogenetics results:

40% of cells with AML1-split.

Other Molecular Studies

Technics: PCR

Results: Tandem duplication of MLL gene (MLL-PTD

positive).

Page 91: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

t(1;21)(p32;q22) as a non-random abnormality in AML M4 Reindl L, Haferlach C

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 711

Partial GTG-banding karyotype showing t(1;21)(p32;q22).

FISH and whole chromosome painting of the same metaphase with t(1;21)(p32;q22); Left picture: AML1 probe on metaphase; Right picture: whole chromosome painting, WCP#1 green, WCP#21 red.

Comments Only two cases with t(1;21)(p32;q22) were described

so far in literature. The first reported case is a 25-year-

old male with an acute myelomonoblastic leukemia

(M4 by FAB subtype) (Cherry et al., 2001). The second

patient, a 29-year-old Japanese male, showed a acute

myelogenous leukemia M4 with NUP98-HOXA9

fusion detected by PCR at the initial diagnosis. In

relapse he acquired additional to the NUP98-HOXA9

fusion a t(1;22)(p32;q22) (Aoki et al., 2008). The here

reported case is a 63-year-old female with an acute

myeloid leukemia (M4 by FAB subtype). So far the

cases have the same morphology in common.

Correlations to age or sex cannot be determined yet.

Call for Collaborations

Lena Reindl

MLL, Munich Leukemia Laboratory,

Max-Lebsche-Platz 31, Germany

[email protected]

References Cherry AM, Bangs CD, Jones P, Hall S, Natkunam Y. A unique AML1 (CBF2A) rearrangement, t(1;21)(p32;q22), observed in a patient with acute myelomonocytic leukemia. Cancer Genet Cytogenet. 2001 Sep;129(2):155-60

La Starza R, Trubia M, Crescenzi B, Matteucci C, Negrini M, Martelli MF, Pelicci PG, Mecucci C. Human homeobox gene HOXC13 is the partner of NUP98 in adult acute myeloid leukemia with t(11;12)(p15;q13). Genes Chromosomes Cancer. 2003 Apr;36(4):420-3

Dal Cin P, Yee AJ, Dey B.. A de novo AML with a t(1;21)(p36;q22) in an elderly patient. Atlas Genet Cytogenet Oncol Haematol. March 2007 URL : http://AtlasGeneticsOncology.org/Reports/0121DalCinID100021.html .

Aoki T, Miyamoto T, Yoshida S, Yamamoto A, Yamauchi T, Yoshimoto G, Mori Y, Kamezaki K, Iwasaki H, Takenaka K, Harada N, Nagafuji K, Teshima T, Akashi K. Additional acquisition of t(1;21)(p32;q22) in a patient relapsing with acute myelogenous leukemia with NUP98-HOXA9. Int J Hematol. 2008 Dec;88(5):571-4

This article should be referenced as such:

Reindl L, Haferlach C. t(1;21)(p32;q22) as a non-random abnormality in AML M4. Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):710-711.

Page 92: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

Case Report Section Paper co-edited with the European LeukemiaNet

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 712

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(3;7)(q26;q21) as a secondary abnormality in MDS RAEB-2 Lena Reindl, Claudia Haferlach

MLL, Munich Leukemia Laboratory, Max-Lebsche-Platz 31, Germany (LR, CH)

Published in Atlas Database: August 2009

Online updated version : http://AtlasGeneticsOncology.org/Reports/0307ReindlID100042.html DOI: 10.4267/2042/44813

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2010 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Clinics

Age and sex

72 years old male patient.

Previous history

No preleukemia; no previous malignancy; no inborn

condition of note.

Organomegaly

No hepatomegaly , no splenomegaly , no enlarged

lymph nodes , no central nervous system involvement.

Blood WBC : 12.700X 10

9/l

HB : 11.9g/dl

Platelets : 70.000X 109/l

Blasts : 15%

Bone marrow : 15 (The hypercellular bone marrow

shows 15% blasts and multilineage dyplasias.

Granulopoesis shows significant dysplasia and

dyplasias were also found in the decreased

erythropoiesis and thrombopoiesis. No Auer rods were

detected.)

Cyto-Pathology Classification

Cytology

MDS RAEB-2

Immunophenotype

12% myeloid blasts CD33+, CD117+; 31%

hypogranulated granuloyctes; 11% monocytes CD56+.

Survival

Date of diagnosis: 07-2008

Status: Lost

Karyotype

Sample : bone marrow

Culture time : 24 - 72h

Banding : GAG.

Results : 46,XY,del(20)(q11) [2]/46,XY,idem,

t(3;7)(q26;q21)[14].

Other molecular cytogenetics technics:

FISH with commercial EVI/3q26 probe (Kreatech).

Other molecular cytogenetics results:

65% cells with EVI-rearrangement.

Partial GTG-karyotype showing t(3;7)(q26;q21).

Page 93: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

t(3;7)(q26;q21) as a secondary abnormality in MDS RAEB-2 Reindl L, Haferlach C

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 713

EVI1 break-apart probe on metaphase chromosomes, one green signal left on derivatives chromosome 3 and one yellow signal is located on derivative chromosome 7.

Whole chromosome painting, WCP#3 yellow, WCP#7 red and WCP#20 green.

Comments 5 cases with t(3;7)(q26;q21) were described so far in

literature. 4 cases showed chronic myeloid leukemia

with t(3;7)(q26;q21) as a additional aberration to

t(9;22)(q34;q11) (Storlazzi et al., 2004; Henzan et al.,

2004; Bobadilla et al., 2007; Tien et al., 1989). One

case suffered from acute myeloid leukemia (NOS) and

had a trisomy 13 as additional aberration to

t(3;7)(q26;q21) (Madrigal et al., 2006). The here

described case - a 72-year-old male - had a MDS

RAEB-2 (FAB) and the t(3;7)(q26;q21) was found as

additional aberration to a 20q-deletion. So in this case

for the first time a t(3;7)(q26;q21) was found in MDS

and the translocation appears also as secondary

aberration as seen in 4 cases with CML before.

Call for Collaborations

Lena Reindl

MLL, Munich Leukemia Laboratory,

Max-Lebsche-Platz 31,

81377 Munich, Germany

[email protected]

References Tien HF, Chuang SM, Wang CH, Lee FY, Chien SH, Chen YC, Shen MC, Liu CH. Chromosomal characteristics of Ph-positive chronic myelogenous leukemia in transformation. A study of 23 Chinese patients in Taiwan. Cancer Genet Cytogenet. 1989 May;39(1):89-97

Henzan H, Yoshimoto G, Okeda A, Nagasaki Y, Hirano G, Takase K, Tanimoto T, Miyamoto T, Fukuda T, Nagafuji K, Harada M. Myeloid/natural killer cell blast crisis representing an additional translocation, t(3;7)(q26;q21) in Philadelphia-positive chronic myelogenous leukemia. Ann Hematol. 2004 Dec;83(12):784-8

Storlazzi CT, Anelli L, Albano F, Zagaria A, Ventura M, Rocchi M, Panagopoulos I, Pannunzio A, Ottaviani E, Liso V, Specchia G. A novel chromosomal translocation t(3;7)(q26;q21) in myeloid leukemia resulting in overexpression of EVI1. Ann Hematol. 2004 Feb;83(2):78-83

Madrigal I, Carrió A, Gómez C, Rozman M, Esteve J, Nomdedeu B, Campo E, Costa D. Fluorescence in situ hybridization studies using BAC clones of the EVI1 locus in hematological malignancies with 3q rearrangements. Cancer Genet Cytogenet. 2006 Oct 15;170(2):115-20

Bobadilla D, Enriquez EL, Alvarez G, Gaytan P, Smith D, Slovak ML. An interphase fluorescence in situ hybridisation assay for the detection of 3q26.2/EVI1 rearrangements in myeloid malignancies. Br J Haematol. 2007 Mar;136(6):806-13

This article should be referenced as such:

Reindl L, Haferlach C. t(3;7)(q26;q21) as a secondary abnormality in MDS RAEB-2. Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7):712-713.

Page 94: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

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

Page 95: Atlas of Genetics and Cytogenetics in Oncology and Haematologydocuments.irevues.inist.fr/bitstream/handle/2042/45791/vol_14_7... · The PDF version of the Atlas of Genetics and Cytogenetics

t(3;7)(q26;q21) as a secondary abnormality in MDS RAEB-2 Reindl L, Haferlach C

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(7) 715